A Selective and Cell-Permeable Mitochondrial Calcium Uniporter (MCU) Inhibitor Preserves Mitochondrial Bioenergetics after Hypoxia/Reoxygenation InjuryClick to copy article linkArticle link copied!
- Joshua J. WoodsJoshua J. WoodsRobert F. Smith School for Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United StatesDepartment of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United StatesMore by Joshua J. Woods
- Neeharika NemaniNeeharika NemaniDepartment of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United StatesCenter for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United StatesMore by Neeharika Nemani
- Santhanam ShanmughapriyaSanthanam ShanmughapriyaDepartment of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United StatesCenter for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United StatesMore by Santhanam Shanmughapriya
- Akshay KumarAkshay KumarDepartment of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United StatesCenter for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United StatesMore by Akshay Kumar
- MengQi ZhangMengQi ZhangDepartment of Physiology and Pharmacology, Western University, London, Ontario N6A 5C1, CanadaMore by MengQi Zhang
- Sarah R. NathanSarah R. NathanDepartment of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United StatesMore by Sarah R. Nathan
- Manfred ThomasManfred ThomasDepartment of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United StatesCenter for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United StatesMore by Manfred Thomas
- Edmund CarvalhoEdmund CarvalhoDepartment of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United StatesCenter for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United StatesMore by Edmund Carvalho
- Karthik RamachandranKarthik RamachandranDepartment of Medicine/Nephrology, Institute for Precision Medicine and Health, University of Texas Health San Antonio, San Antonio, Texas 78229, United StatesMore by Karthik Ramachandran
- Subramanya SrikantanSubramanya SrikantanDepartment of Medicine/Nephrology, Institute for Precision Medicine and Health, University of Texas Health San Antonio, San Antonio, Texas 78229, United StatesMore by Subramanya Srikantan
- Peter B. StathopulosPeter B. StathopulosDepartment of Physiology and Pharmacology, Western University, London, Ontario N6A 5C1, CanadaMore by Peter B. Stathopulos
- Justin J. Wilson*Justin J. Wilson*E-mail: [email protected]Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United StatesMore by Justin J. Wilson
- Muniswamy Madesh*Muniswamy Madesh*E-mail: [email protected]Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United StatesCenter for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United StatesDepartment of Medicine/Nephrology, Institute for Precision Medicine and Health, University of Texas Health San Antonio, San Antonio, Texas 78229, United StatesMore by Muniswamy Madesh
Abstract
Mitochondrial Ca2+ (mCa2+) uptake mediated by the mitochondrial calcium uniporter (MCU) plays a critical role in signal transduction, bioenergetics, and cell death, and its dysregulation is linked to several human diseases. In this study, we report a new ruthenium complex Ru265 that is cell-permeable, minimally toxic, and highly potent with respect to MCU inhibition. Cells treated with Ru265 show inhibited MCU activity without any effect on cytosolic Ca2+ dynamics and mitochondrial membrane potential (ΔΨm). Dose-dependent studies reveal that Ru265 is more potent than the currently employed MCU inhibitor Ru360. Site-directed mutagenesis of Cys97 in the N-terminal domain of human MCU ablates the inhibitory activity of Ru265, suggesting that this matrix-residing domain is its target site. Additionally, Ru265 prevented hypoxia/reoxygenation injury and subsequent mitochondrial dysfunction, demonstrating that this new inhibitor is a valuable tool for studying the functional role of the MCU in intact biological models.
Synopsis
A new, cell-permeable inhibitor of mitochondrial calcium uptake, called Ru265, is reported, and its application in biological systems is described.
Introduction
Results
Synthesis and Characterization
Mitochondrial Ca2+ Uptake Inhibition and Cell Permeability
Exploring the Mechanism of MCU Inhibition
Protection from Ca2+-Induced PTP Opening and Hypoxia/Reoxygenation Injury
Discussion
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acscentsci.8b00773.
Experimental details, complex characterization data, cell viability curves, cell uptake, UV–vis spectra, and dose–response data for Ca2+ uptake inhibition (PDF)
X-ray crystal data for C-2 and C-3 (CIF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
This research in the laboratory of M. Madesh was funded by the National Institutes of Health (R01GM109882, R01HL086699, R01HL142673, R01HL119306). Research in the laboratory of J. J. Wilson was supported by the National Science Foundation (NSF) (CHE-1750295). S. Shanmughapriya is supported by the NIH K99/R00 grant (1 K99 HL138268-01). E. Carvalho and N. Nemani are supported by the AHA fellowships (18POST33990217 and 17PRE33660720). J. J. Woods is supported by the NSF-GRFP (DGE-1650441). This work made use of the Cornell NMR facility, which is funded in part by the NSF (CHE-1531632). We thank Reggie Jacob for helpful comments on the manuscript and Samantha Davalos for assistance in preparing the artwork associated with this manuscript.
References
This article references 94 other publications.
- 1Berridge, M. J.; Bootman, M. D.; Roderick, H. L. Calcium Signalling: Dynamics, Homeostasis and Remodelling. Nat. Rev. Mol. Cell Biol. 2003, 4, 517– 529, DOI: 10.1038/nrm1155Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXltVWmsr8%253D&md5=da79c7368cc92aeb53f243858e1fcd70Calcium: Calcium signaling: dynamics, homeostasis and remodelingBerridge, Michael J.; Bootman, Martin D.; Roderick, H. LlewelynNature Reviews Molecular Cell Biology (2003), 4 (7), 517-529CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. Ca2+ is a highly versatile intracellular signal that operates over a wide temporal range to regulate many different cellular processes. An extensive Ca2+-signaling toolkit is used to assemble signaling systems with very different spatial and temporal dynamics. Rapid highly localized Ca2+ spikes regulate fast responses, whereas slower responses are controlled by repetitive global Ca2+ transients or intracellular Ca2+ waves. Ca2+ plays a direct role in controlling the expression patterns of its signaling systems that are constantly being remodelled in both health and disease.
- 2Soboloff, J.; Rothberg, B. S.; Madesh, M.; Gill, D. L. STIM Proteins: Dynamic Calcium Signal Transducers. Nat. Rev. Mol. Cell Biol. 2012, 13, 549– 565, DOI: 10.1038/nrm3414Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xht1Ghsb7N&md5=b26a6baa9b336d6f750ec2447c143fc7STIM proteins: dynamic calcium signal transducersSoboloff, Jonathan; Rothberg, Brad S.; Madesh, Muniswamy; Gill, Donald L.Nature Reviews Molecular Cell Biology (2012), 13 (9), 549-565CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. Stromal interaction mol. (STIM) proteins function in cells as dynamic coordinators of cellular calcium (Ca2+) signals. Spanning the endoplasmic reticulum (ER) membrane, they sense tiny changes in the levels of Ca2+ stored within the ER lumen. As ER Ca2+ is released to generate primary Ca2+ signals, STIM proteins undergo an intricate activation reaction and rapidly translocate into junctions formed between the ER and the plasma membrane. There, STIM proteins tether and activate the highly Ca2+-selective Orai channels to mediate finely controlled Ca2+ signals and to homeostatically balance cellular Ca2+. Details are emerging on the remarkable organization within these STIM-induced junctional microdomains and the identification of new regulators and alternative target proteins for STIM.
- 3Clapham, D. E. Calcium Signaling. Cell 2007, 131, 1047– 1058, DOI: 10.1016/j.cell.2007.11.028Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXksFGgsA%253D%253D&md5=268945d7ca1ec7a455f69cb72272be85Calcium signalingClapham, David E.Cell (Cambridge, MA, United States) (2007), 131 (6), 1047-1058CODEN: CELLB5; ISSN:0092-8674. (Cell Press)A review. Ca2+ ions impact nearly every aspect of cellular life. Here, the author examines the principles of Ca2+ signaling, from changes in protein conformations driven by Ca2+ to the mechanisms that control Ca2+ levels in the cytoplasm and organelles. Also discussed is the highly localized nature of Ca2+-mediated signal transduction and its specific role in excitability, exocytosis, motility, apoptosis, and transcription.
- 4Hogan, P. G.; Lewis, R. S.; Rao, A. Molecular Basis of Calcium Signaling in Lymphocytes: STIM and ORAI. Annu. Rev. Immunol. 2010, 28, 491– 533, DOI: 10.1146/annurev.immunol.021908.132550Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXlsVSmsL8%253D&md5=11db4a7bf473601794aa7e49e0de68d8Molecular basis of calcium signaling in lymphocytes: STIM and ORAIHogan, Patrick G.; Lewis, Richard S.; Rao, AnjanaAnnual Review of Immunology (2010), 28 (), 491-533CODEN: ARIMDU; ISSN:0732-0582. (Annual Reviews Inc.)A review. Ca2+ entry into cells of the peripheral immune system occurs through highly Ca2+-selective channels known as CRAC (calcium release-activated calcium) channels. CRAC channels are a very well-characterized example of store-operated Ca2+ channels, so designated because they open when the endoplasmic reticulum (ER) Ca2+ store becomes depleted. Physiol., Ca2+ is released from the ER lumen into the cytoplasm when activated receptors couple to phospholipase C and trigger prodn. of the second messenger inositol 1,4,5-trisphosphate (IP3). IP3 binds to IP3 receptors in the ER membrane and activates Ca2+ release. The proteins STIM and ORAI were discovered through limited and genome-wide RNAi screens, resp., performed in Drosophila cells and focused on identifying modulators of store-operated Ca2+ entry. STIM1 and STIM2 sense the depletion of ER Ca2+ stores, whereas ORAI1 is a pore subunit of the CRAC channel. In this review, we discuss selected aspects of Ca2+ signaling in cells of the immune system, focusing on the roles of STIM and ORAI proteins in store-operated Ca2+ entry.
- 5Kirichok, Y.; Krapivinsky, G.; Clapham, D. E. The Mitochondrial Calcium Uniporter is a Highly Selective Ion Channel. Nature 2004, 427, 360– 364, DOI: 10.1038/nature02246Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXltFCgtw%253D%253D&md5=37aefce568d7ac7225de675619283847The mitochondrial calcium uniporter is a highly selective ion channelKirichok, Yuriy; Krapivinsky, Grigory; Clapham, David E.Nature (London, United Kingdom) (2004), 427 (6972), 360-364CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)During intracellular Ca2+ signaling mitochondria accumulate significant amts. of Ca2+ from the cytosol. Mitochondrial Ca2+ uptake controls the rate of energy prodn., shapes the amplitude and spatio-temporal patterns of intracellular Ca2+ signals, and is instrumental to cell death. This Ca2+ uptake is undertaken by the mitochondrial Ca2+ uniporter (MCU) located in the organelle's inner membrane. The uniporter passes Ca2+ down the electrochem. gradient maintained across this membrane without direct coupling to ATP hydrolysis or transport of other ions. Carriers are characterized by turnover nos. that are typically 1,000-fold lower than ion channels, and until now it has been unclear whether the MCU is a carrier or a channel. By patch-clamping the inner mitochondrial membrane, we identified a previously unknown Ca2+-selective ion channel sensitive to inhibitors of mitochondrial Ca2+ uptake. Our data indicate that this unique channel binds Ca2+ with extremely high affinity (dissocn. const. ≤2 nM), enabling high Ca2+ selectivity despite relatively low cytoplasmic Ca2+ concns. The channel is inwardly rectifying, making it esp. effective for Ca2+ uptake into energized mitochondria. Thus, we conclude that the properties of the current mediated by this novel channel are those of the MCU.
- 6Kamer, K. J.; Mootha, V. K. The Molecular Era of the Mitochondrial Calcium Uniporter. Nat. Rev. Mol. Cell Biol. 2015, 16, 545– 553, DOI: 10.1038/nrm4039Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtlKntrbO&md5=8f0222b7cb0d6693adf957a3cd9ba983The molecular era of the mitochondrial calcium uniporterKamer, Kimberli J.; Mootha, Vamsi K.Nature Reviews Molecular Cell Biology (2015), 16 (9), 545-553CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. The mitochondrial calcium uniporter is an evolutionarily conserved calcium channel, and its biophys. properties and relevance to cell death, bioenergetics and signalling have been investigated for decades. However, the genes encoding this channel have only recently been discovered, opening up a new 'mol. era' in the study of its biol. We now know that the uniporter is not a single protein but rather a macromol. complex consisting of pore-forming and regulatory subunits. We review recent studies that harnessed the power of mol. biol. and genetics to characterize the mechanism of action of the uniporter, its evolution and its contribution to physiol. and human disease.
- 7Rizzuto, R.; De Stefani, D.; Raffaello, A.; Mammucari, C. Mitochondria as Sensors and Regulators of Calcium Signalling. Nat. Rev. Mol. Cell Biol. 2012, 13, 566– 578, DOI: 10.1038/nrm3412Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtFWms77O&md5=4b6f8ac747922757957102c8fa1420daMitochondria as sensors and regulators of calcium signallingRizzuto, Rosario; De Stefani, Diego; Raffaello, Anna; Mammucari, CristinaNature Reviews Molecular Cell Biology (2012), 13 (9), 566-578CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. During the past two decades, calcium (Ca2+) accumulation in energized mitochondria has emerged as a biol. process of utmost physiol. relevance. Mitochondrial Ca2+ uptake was shown to control intracellular Ca2+ signalling, cell metab., cell survival and other cell-type specific functions by buffering cytosolic Ca2+ levels and regulating mitochondrial effectors. Recently, the identity of mitochondrial Ca2+ transporters has been revealed, opening new perspectives for investigation and mol. intervention.
- 8Nemani, N.; Shanmughapriya, S.; Madesh, M. Molecular Regulation of MCU: Implications in Physiology and Disease. Cell Calcium 2018, 74, 86– 93, DOI: 10.1016/j.ceca.2018.06.006Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXht1CntLvE&md5=c0b9d850bd8055add666550c2ade0867Molecular regulation of MCU: Implications in physiology and diseaseNemani, Neeharika; Shanmughapriya, Santhanam; Madesh, MuniswamyCell Calcium (2018), 74 (), 86-93CODEN: CECADV; ISSN:0143-4160. (Elsevier Ltd.)Ca2+ flux across the inner mitochondrial membrane (IMM) regulates cellular bioenergetics, intra-cellular cytoplasmic Ca2+ signals, and various cell death pathways. Ca2+ entry into the mitochondria occurs due to the highly neg. membrane potential (ΔΨm) through a selective inward rectifying MCU channel. In addn. to being regulated by various mitochondrial matrix resident proteins such as MICUs, MCUb, MCUR1 and EMRE, the channel is transcriptionally regulated by upstream Ca2+ cascade, post transnational modification and by divalent cations. The mode of regulation either inhibits or enhances MCU channel activity and thus regulates mitochondrial metab. and cell fate.
- 9Baughman, J. M.; Perocchi, F.; Girgis, H. S.; Plovanich, M.; Belcher-Timme, C. A.; Sancak, Y.; Bao, X. R.; Strittmatter, L.; Goldberger, O.; Bogorad, R. L.; Koteliansky, V.; Mootha, V. K. Integrative Genomics Identifies MCU as an Essential Component of the Mitochondrial Calcium Uniporter. Nature 2011, 476, 341– 345, DOI: 10.1038/nature10234Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXnslSktLo%253D&md5=14aa9d448ee67a2d3cc473ccbcabe822Integrative genomics identifies MCU as an essential component of the mitochondrial calcium uniporterBaughman, Joshua M.; Perocchi, Fabiana; Girgis, Hany S.; Plovanich, Molly; Belcher-Timme, Casey A.; Sancak, Yasemin; Bao, X. Robert; Strittmatter, Laura; Goldberger, Olga; Bogorad, Roman L.; Koteliansky, Victor; Mootha, Vamsi K.Nature (London, United Kingdom) (2011), 476 (7360), 341-345CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Mitochondria from diverse organisms are capable of transporting large amts. of Ca2+ via a ruthenium-red-sensitive, membrane-potential-dependent mechanism called the uniporter. Although the uniporter's biophys. properties have been studied extensively, its mol. compn. remains elusive. We recently used comparative proteomics to identify MICU1 (also known as CBARA1), an EF-hand-contg. protein that serves as a putative regulator of the uniporter. Here, we use whole-genome phylogenetic profiling, genome-wide RNA co-expression anal. and organelle-wide protein coexpression anal. to predict proteins functionally related to MICU1. All three methods converge on a novel predicted transmembrane protein, CCDC109A, that we now call "mitochondrial calcium uniporter" (MCU). MCU forms oligomers in the mitochondrial inner membrane, phys. interacts with MICU1, and resides within a large mol. wt. complex. Silencing MCU in cultured cells or in vivo in mouse liver severely abrogates mitochondrial Ca2+ uptake, whereas mitochondrial respiration and membrane potential remain fully intact. MCU has two predicted transmembrane helixes, which are sepd. by a highly conserved linker facing the intermembrane space. Acidic residues in this linker are required for its full activity. However, an S259A point mutation retains function but confers resistance to Ru360, the most potent inhibitor of the uniporter. Our genomic, physiol., biochem. and pharmacol. data firmly establish MCU as an essential component of the mitochondrial Ca2+ uniporter.
- 10De Stefani, D.; Raffaello, A.; Teardo, E.; Szabò, I.; Rizzuto, R. A Forty-Kilodalton Protein of the Inner Membrane is the Mitochondrial Calcium Uniporter. Nature 2011, 476, 336– 340, DOI: 10.1038/nature10230Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXnslSksLg%253D&md5=8c86db6f4556b498cddb9d24a97de832A forty-kilodalton protein of the inner membrane is the mitochondrial calcium uniporterDe Stefani, Diego; Raffaello, Anna; Teardo, Enrico; Szabo, Ildiko; Rizzuto, RosarioNature (London, United Kingdom) (2011), 476 (7360), 336-340CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Mitochondrial Ca2+ homeostasis has a key role in the regulation of aerobic metab. and cell survival, but the mol. identity of the Ca2+ channel, the mitochondrial calcium uniporter, is still unknown. Here we have identified in silico a protein (named MCU) that shares tissue distribution with MICU1 (also known as CBARA1), a recently characterized uniporter regulator, is present in organisms in which mitochondrial Ca2+ uptake was demonstrated and whose sequence includes two transmembrane domains. Short interfering RNA (siRNA) silencing of MCU in HeLa cells markedly reduced mitochondrial Ca2+ uptake. MCU overexpression doubled the matrix Ca2+ concn. increase evoked by inositol 1,4,5-trisphosphate-generating agonists, thus significantly buffering the cytosolic elevation. The purified MCU protein showed channel activity in planar lipid bilayers, with electrophysiol. properties and inhibitor sensitivity of the uniporter. A mutant MCU, in which two neg. charged residues of the putative pore-forming region were replaced, had no channel activity and reduced agonist-dependent matrix Ca2+ concn. transients when overexpressed in HeLa cells. Overall, these data demonstrate that the 40-kDa protein identified is the channel responsible for ruthenium-red-sensitive mitochondrial Ca2+ uptake, thus providing a mol. basis for this process of utmost physiol. and pathol. relevance.
- 11Mallilankaraman, K.; Doonan, P.; Cárdenas, C.; Chandramoorthy, H. C.; Müller, M.; Miller, R.; Hoffman, N. E.; Gandhirajan, R. K.; Molgó, J.; Birnbaum, M. J.; Rothberg, B. S.; Mak, D. O. D.; Foskett, J. K.; Madesh, M. MICU1 is an Essential Gatekeeper for MCU-Mediated Mitochondrial Ca2+ Uptake That Regulates Cell Survival. Cell 2012, 151, 630– 644, DOI: 10.1016/j.cell.2012.10.011Google ScholarThere is no corresponding record for this reference.
- 12Sancak, Y.; Markhard, A. L.; Kitami, T.; Kovacs-Bogdan, E.; Kamer, K. J.; Udeshi, N. D.; Carr, S. A.; Chaudhuri, D.; Clapham, D. E.; Li, A. A.; Calvo, S. E.; Goldberger, O.; Mootha, V. K. EMRE is an Essential Component of the Mitochondrial Calcium Uniporter Complex. Science 2013, 342, 1379– 1382, DOI: 10.1126/science.1242993Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvV2lsLjM&md5=1f9f02e65f9b735064db194245903737EMRE Is an Essential Component of the Mitochondrial Calcium Uniporter ComplexSancak, Yasemin; Markhard, Andrew L.; Kitami, Toshimori; Kovacs-Bogdan, Erika; Kamer, Kimberli J.; Udeshi, Namrata D.; Carr, Steven A.; Chaudhuri, Dipayan; Clapham, David E.; Li, Andrew A.; Calvo, Sarah E.; Goldberger, Olga; Mootha, Vamsi K.Science (Washington, DC, United States) (2013), 342 (6164), 1379-1382CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The mitochondrial uniporter is a highly selective calcium channel in the organelle's inner membrane. Its mol. components include the EF-hand-contg. calcium-binding proteins mitochondrial calcium uptake 1 (MICU1) and MICU2 and the pore-forming subunit mitochondrial calcium uniporter (MCU). We sought to achieve a full mol. characterization of the uniporter holocomplex (uniplex). Quant. mass spectrometry of affinity-purified uniplex recovered MICU1 and MICU2, MCU and its paralog MCUb, and a previously uncharacterized 10-kilodalton metazoan-specific single transmembrane domain protein designated EMRE ("essential MCU regulator"). In its absence, uniporter channel activity was lost despite intact MCU expression and oligomerization. EMRE was required for the interaction of MCU with MICU1 and MICU2. Hence, EMRE is essential for in vivo uniporter current and addnl. bridges the calcium-sensing role of MICU1 and MICU2 with the calcium-conducting role of MCU.
- 13Perocchi, F.; Gohil, V. M.; Girgis, H. S.; Bao, X. R.; McCombs, J. E.; Palmer, A. E.; Mootha, V. K. MICU1 Encodes a Mitochondrial EF Hand Protein Required for Ca2+ uptake. Nature 2010, 467, 291– 296, DOI: 10.1038/nature09358Google ScholarThere is no corresponding record for this reference.
- 14Plovanich, M.; Bogorad, R. L.; Sancak, Y.; Kamer, K. J.; Strittmatter, L.; Li, A. A.; Girgis, H. S.; Kuchimanchi, S.; De Groot, J.; Speciner, L.; Taneja, N.; OShea, J.; Koteliansky, V.; Mootha, V. K. MICU2, a Paralog of MICU1, Resides within the Mitochondrial Uniporter Complex to Regulate Calcium Handling. PLoS One 2013, 8, e55785, DOI: 10.1371/journal.pone.0055785Google ScholarThere is no corresponding record for this reference.
- 15Csordás, G.; Golenár, T.; Seifert, E. L.; Kamer, K. J.; Sancak, Y.; Perocchi, F.; Moffat, C.; Weaver, D.; De la Fuente, S.; Bogorad, R.; Koteliansky, V.; Adijanto, J.; Mootha, V. K.; Hajnóczky, G. MICU1 Controls Both the Threshold and Cooperative Activation of the Mitochondrial Ca2+ Uniporter. Cell Metab. 2013, 17, 976– 987, DOI: 10.1016/j.cmet.2013.04.020Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXptV2ks7o%253D&md5=1c83a13cd426d596daed8be670b8318cMICU1 Controls Both the Threshold and Cooperative Activation of the Mitochondrial Ca2+ UniporterCsordas, Gyorgy; Golenar, Tunde; Seifert, Erin L.; Kamer, Kimberli J.; Sancak, Yasemin; Perocchi, Fabiana; Moffat, Cynthia; Weaver, David; Perez, Sergio de la Fuente; Bogorad, Roman; Koteliansky, Victor; Adijanto, Jeffrey; Mootha, Vamsi K.; Hajnoczky, GyorgyCell Metabolism (2013), 17 (6), 976-987CODEN: CMEEB5; ISSN:1550-4131. (Elsevier Inc.)Mitochondrial Ca2+ uptake via the uniporter is central to cell metab., signaling, and survival. Recent studies identified MCU as the uniporter's likely pore and MICU1, an EF-hand protein, as its crit. regulator. How this complex decodes dynamic cytoplasmic [Ca2+] ([Ca2+]c) signals, to tune out small [Ca2+]c increases yet permit pulse transmission, remains unknown. We report that loss of MICU1 in mouse liver and cultured cells causes mitochondrial Ca2+ accumulation during small [Ca2+]c elevations but an attenuated response to agonist-induced [Ca2+]c pulses. The latter reflects loss of pos. cooperativity, likely via the EF-hands. MICU1 faces the intermembrane space and responds to [Ca2+]c changes. Prolonged MICU1 loss leads to an adaptive increase in matrix Ca2+ binding, yet cells show impaired oxidative metab. and sensitization to Ca2+ overload. Collectively, the data indicate that MICU1 senses the [Ca2+]c to establish the uniporter's threshold and gain, thereby allowing mitochondria to properly decode different inputs.
- 16Mallilankaraman, K.; Cárdenas, C.; Doonan, P. J.; Chandramoorthy, H. C.; Irrinki, K. M.; Golenár, T.; Csordás, G.; Madireddi, P.; Yang, J.; Müller, M.; Miller, R.; Kolesar, J. E.; Molgó, J.; Kaufman, B.; Hajnóczky, G.; Foskett, J. K.; Madesh, M. MCUR1 is an Essential Component of Mitochondrial Ca2+ Uptake that Regulates Cellular Metabolism. Nat. Cell Biol. 2012, 14, 1336– 1343, DOI: 10.1038/ncb2622Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhslagsbzO&md5=43a3b38905a4498473229cdf34d3dd1cMCUR1 is an essential component of mitochondrial Ca2+ uptake that regulates cellular metabolismMallilankaraman, Karthik; Cardenas, Cesar; Doonan, Patrick J.; Chandramoorthy, Harish C.; Irrinki, Krishna M.; Golenar, Tuende; Csordas, Gyoergy; Madireddi, Priyanka; Yang, Jun; Mueller, Marioly; Miller, Russell; Kolesar, Jill E.; Molgo, Jordi; Kaufman, Brett; Hajnoczky, Gyoergy; Foskett, J. Kevin; Madesh, MuniswamyNature Cell Biology (2012), 14 (12), 1336-1343CODEN: NCBIFN; ISSN:1465-7392. (Nature Publishing Group)Ca2+ flux across the mitochondrial inner membrane regulates bioenergetics, cytoplasmic Ca2+ signals and activation of cell death pathways. Mitochondrial Ca2+ uptake occurs at regions of close apposition with intracellular Ca2+ release sites, driven by the inner membrane voltage generated by oxidative phosphorylation and mediated by a Ca2+ selective ion channel (MiCa; ref. ) called the uniporter whose complete mol. identity remains unknown. Mitochondrial calcium uniporter (MCU) was recently identified as the likely ion-conducting pore. In addn., MICU1 was identified as a mitochondrial regulator of uniporter-mediated Ca2+ uptake in HeLa cells. Here we identified CCDC90A, hereafter referred to as MCUR1 (mitochondrial calcium uniporter regulator 1), an integral membrane protein required for MCU-dependent mitochondrial Ca2+ uptake. MCUR1 binds to MCU and regulates ruthenium-red-sensitive MCU-dependent Ca2+ uptake. MCUR1 knockdown does not alter MCU localization, but abrogates Ca2+ uptake by energized mitochondria in intact and permeabilized cells. Ablation of MCUR1 disrupts oxidative phosphorylation, lowers cellular ATP and activates AMP kinase-dependent pro-survival autophagy. Thus, MCUR1 is a crit. component of a mitochondrial uniporter channel complex required for mitochondrial Ca2+ uptake and maintenance of normal cellular bioenergetics.
- 17Patron, M.; Checchetto, V.; Raffaello, A.; Teardo, E.; VecellioReane, D.; Mantoan, M.; Granatiero, V.; Szabò, I.; DeStefani, D.; Rizzuto, R. MICU1 and MICU2 Finely Tune the Mitochondrial Ca2+ Uniporter by Exerting Opposite Effects on MCU Activity. Mol. Cell 2014, 53, 726– 737, DOI: 10.1016/j.molcel.2014.01.013Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXivFCitr0%253D&md5=56712d0f63ab7ad878a37055a9ae18c3MICU1 and MICU2 Finely Tune the Mitochondrial Ca2+ Uniporter by Exerting Opposite Effects on MCU ActivityPatron, Maria; Checchetto, Vanessa; Raffaello, Anna; Teardo, Enrico; Vecellio Reane, Denis; Mantoan, Maura; Granatiero, Veronica; Szabo, Ildiko; De Stefani, Diego; Rizzuto, RosarioMolecular Cell (2014), 53 (5), 726-737CODEN: MOCEFL; ISSN:1097-2765. (Elsevier Inc.)Mitochondrial calcium accumulation was recently shown to depend on a complex composed of an inner-membrane channel (MCU and MCUb) and regulatory subunits (MICU1, MCUR1, and EMRE). A fundamental property of MCU is low activity at resting cytosolic Ca2+ concns., preventing deleterious Ca2+ cycling and organelle overload. Here we demonstrate that these properties are ensured by a regulatory heterodimer composed of two proteins with opposite effects - MICU1 and MICU2 - which in both purified lipid bilayers and in intact cells stimulate and inhibit MCU activity, resp. Both MICU1 and MICU2 are regulated by calcium through their EF-hand domains, thus accounting for the sigmoidal response of MCU to [Ca2+] in situ and allowing tight physiol. control. At low [Ca2+], the dominant effect of MICU2 largely shuts down MCU activity; at higher [Ca2+], the stimulatory effect of MICU1 allows the prompt response of mitochondria to Ca2+ signals generated in the cytoplasm.
- 18Oxenoid, K.; Dong, Y.; Cao, C.; Cui, T.; Sancak, Y.; Markhard, A. L.; Grabarek, Z.; Kong, L.; Liu, Z.; Ouyang, B.; Cong, Y.; Mootha, V. K.; Chou, J. J. Architecture of the Mitochondrial Calcium Uniporter. Nature 2016, 533, 269– 273, DOI: 10.1038/nature17656Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XntVCksrk%253D&md5=0a977baa9b661326236a9914eccf760cArchitecture of the mitochondrial calcium uniporterOxenoid, Kirill; Dong, Ying; Cao, Chan; Cui, Tanxing; Sancak, Yasemin; Markhard, Andrew L.; Grabarek, Zenon; Kong, Liangliang; Liu, Zhijun; Ouyang, Bo; Cong, Yao; Mootha, Vamsi K.; Chou, James J.Nature (London, United Kingdom) (2016), 533 (7602), 269-273CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Mitochondria from many eukaryotic clades take up large amts. of Ca2+ via an inner membrane transporter called the mitochondrial calcium uniporter (MCU). Transport by MCU is membrane potential-dependent and sensitive to ruthenium red or its deriv., Ru360. Electrophysiol. studies have shown that MCU is an ion channel with remarkably high conductance and selectivity. Ca2+ entry into mitochondria is also known to activate the tricarboxylic acid cycle and seems to be crucial for matching the prodn. of ATP in mitochondria with its cytosolic demand. MCU is the pore-forming and Ca2+-conducting subunit of the uniporter holocomplex, but its primary sequence does not resemble any Ca2+ channel studied to date. Here, the authors report the structure of the pore domain of MCU from Caenorhabditis elegans, detd. using NMR spectroscopy and electron microscopy (EM). MCU is a homo-oligomer in which the 2nd transmembrane helix forms a hydrophilic pore across the membrane. The channel assembly represented a new soln. of ion channel architecture, and was stabilized by a coiled-coil motif protruding into the mitochondrial matrix. The crit. DXXE motif formed the pore entrance, which featured 2 carboxylate rings; based on the ring dimensions and functional mutagenesis, these rings appeared to form the selectivity filter. To the authors' knowledge, this is one of the largest membrane protein structures characterized by NMR, and provides a structural blueprint for understanding the function of this channel.
- 19Bick, A. G.; Calvo, S. E.; Mootha, V. K. Evolutionary Diversity of the Mitochondrial Calcium Uniporter. Science 2012, 336, 886, DOI: 10.1126/science.1214977Google ScholarThere is no corresponding record for this reference.
- 20Baradaran, R.; Wang, C.; Siliciano, A. F.; Long, S. B. Cryo-EM Structures of Fungal and Metazoan Mitochondrial Calcium Uniporters. Nature 2018, 559, 580– 584, DOI: 10.1038/s41586-018-0331-8Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlWjsbfL&md5=742571dbb000126c2013abefe5963fa6Cryo-EM structures of fungal and metazoan mitochondrial calcium uniportersBaradaran, Rozbeh; Wang, Chongyuan; Siliciano, Andrew Francis; Long, Stephen BarstowNature (London, United Kingdom) (2018), 559 (7715), 580-584CODEN: NATUAS; ISSN:0028-0836. (Nature Research)The mitochondrial calcium uniporter (MCU) is a highly selective calcium channel and a major route of calcium entry into mitochondria. How the channel catalyzes ion permeation and achieves ion selectivity are not well understood, partly because MCU is thought to have a distinct architecture in comparison to other cellular channels. Here we report cryo-electron microscopy reconstructions of MCU channels from zebrafish and Cyphellophora europaea at 8.5 Å and 3.2 Å resolns., resp. In contrast to a previous report of pentameric stoichiometry for MCU, both channels are tetramers. The at. model of C. europaea MCU shows that a conserved WDXXEP signature sequence forms the selectivity filter, in which calcium ions are arranged in single file. Coiled-coil legs connect the pore to N-terminal domains in the mitochondrial matrix. In C. europaea MCU, the N-terminal domains assemble as a dimer of dimers; in zebrafish MCU, they form an asym. crescent. The structures define principles that underlie ion permeation and calcium selectivity in this unusual channel.
- 21Fan, C.; Fan, M.; Orlando, B. J.; Fastman, N. M.; Zhang, J.; Xu, Y.; Chambers, M. G.; Xu, X.; Perry, K.; Liao, M.; Feng, L. X-Ray and Cryo-EM Structures of the Mitochondrial Calcium Uniporter. Nature 2018, 559, 575– 579, DOI: 10.1038/s41586-018-0330-9Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlWjtr7M&md5=2293cca6a52861da2fc998e1c00af7d5X-ray and cryo-EM structures of the mitochondrial calcium uniporterFan, Chao; Fan, Minrui; Orlando, Benjamin J.; Fastman, Nathan M.; Zhang, Jinru; Xu, Yan; Chambers, Melissa G.; Xu, Xiaofang; Perry, Kay; Liao, Maofu; Feng, LiangNature (London, United Kingdom) (2018), 559 (7715), 575-579CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Mitochondrial Ca2+ uptake is crit. for regulating ATP prodn., intracellular Ca2+ signaling, and cell death. This uptake is mediated by a highly selective Ca2+ channel called the mitochondrial calcium uniporter (MCU). Here, we detd. the structures of the pore-forming MCU proteins from 2 fungi (Fusarium graminearum and Metarhizium acridum) by x-ray crystallog. and single-particle cryo-electron microscopy (cryo-EM). The stoichiometry, overall architecture, and individual subunit structure differed markedly from those described in the recent NMR structure of Caenorhabditis elegans MCU. We obsd. a dimer-of-dimer architecture across species and chem. environments, which was corroborated by biochem. expts. Structural analyses and functional characterization uncovered the roles of key residues in the pore. These results revealed a new ion channel architecture, provide insights into Ca2+ coordination, selectivity, and conduction, and established a structural framework for understanding the mechanism of MCU function.
- 22Nguyen, N. X.; Armache, J. P.; Lee, C.; Yang, Y.; Zeng, W.; Mootha, V. K.; Cheng, Y.; Bai, X.; Jiang, Y. Cryo-EM Structure of a Fungal Mitochondrial Calcium Uniporter. Nature 2018, 559, 570– 574, DOI: 10.1038/s41586-018-0333-6Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlWjtr7N&md5=43d5c4d30e00ba77b51bdeeba408cf6cCryo-EM structure of a fungal mitochondrial calcium uniporterNguyen, Nam X.; Armache, Jean-Paul; Lee, Changkeun; Yang, Yi; Zeng, Weizhong; Mootha, Vamsi K.; Cheng, Yifan; Bai, Xiao-chen; Jiang, YouxingNature (London, United Kingdom) (2018), 559 (7715), 570-574CODEN: NATUAS; ISSN:0028-0836. (Nature Research)The mitochondrial calcium uniporter (MCU) is a highly selective calcium channel localized to the inner mitochondrial membrane. Here, we describe the structure of an MCU ortholog from the fungus Neosartorya fischeri (NfMCU) detd. to 3.8 Å resoln. by phase-plate cryo-electron microscopy. The channel is a homotetramer with two-fold symmetry in its amino-terminal domain (NTD) that adopts a similar structure to that of human MCU. The NTD assembles as a dimer of dimers to form a tetrameric ring that connects to the transmembrane domain through an elongated coiled-coil domain. The ion-conducting pore domain maintains four-fold symmetry, with the selectivity filter positioned at the start of the pore-forming TM2 helix. The aspartate and glutamate sidechains of the conserved DIME motif are oriented towards the central axis and sepd. by one helical turn. The structure of NfMCU offers insights into channel assembly, selective calcium permeation, and inhibitor binding.
- 23Yoo, J.; Wu, M.; Yin, Y.; Herzik, M. A.; Lander, G. C.; Lee, S.-Y. Cryo-EM Structure of a Mitochondrial Calcium Uniporter. Science 2018, 361, 506– 511, DOI: 10.1126/science.aar4056Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsVahtr%252FE&md5=2072ca08fe5036a5b3749a4073a7e902Cryo-EM structure of a mitochondrial calcium uniporterYoo, Jiho; Wu, Mengyu; Yin, Ying; Herzik, Mark A., Jr; Lander, Gabriel C.; Lee, Seok-YongScience (Washington, DC, United States) (2018), 361 (6401), 506-511CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Calcium transport plays an important role in regulating mitochondrial physiol. and pathophysiol. The mitochondrial calcium uniporter (MCU) is a calcium-selective ion channel that is the primary mediator for calcium uptake into the mitochondrial matrix. Here, we present the cryo-electron microscopy structure of the full-length MCU from Neurospora crassa to an overall resoln. of ∼3.7 angstroms. Our structure reveals a tetrameric architecture, with the sol. and transmembrane domains adopting different sym. arrangements within the channel. The conserved W-D-Φ-Φ-E-P-V-T-Y sequence motif of MCU pore forms a selectivity filter comprising two acidic rings sepd. by one helical turn along the central axis of the channel pore. The structure combined with mutagenesis gives insight into the basis of calcium recognition.
- 24Hoffman, N. E.; Chandramoorthy, H. C.; Shamugapriya, S.; Zhang, X.; Rajan, S.; Mallilankaraman, K.; Gandhirajan, R. K.; Vagnozzi, R. J.; Ferrer, L. M.; Sreekrishnanilayam, K.; Natarajaseenivasan, K.; Vallem, S.; Force, T.; Choi, E. T.; Cheung, J. Y.; Madesh, M. MICU1 Motifs Define Mitochondrial Calcium Uniporter Binding and Activity. Cell Rep. 2013, 5, 1576– 1588, DOI: 10.1016/j.celrep.2013.11.026Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvFOhtrbK&md5=7333ca5d17d6d2e112a240a4d3949d80MICU1 Motifs Define Mitochondrial Calcium Uniporter Binding and ActivityHoffman, Nicholas E.; Chandramoorthy, Harish C.; Shamugapriya, Santhanam; Zhang, Xueqian; Rajan, Sudarsan; Mallilankaraman, Karthik; Gandhirajan, Rajesh Kumar; Vagnozzi, Ronald J.; Ferrer, Lucas M.; Sreekrishnanilayam, Krishnalatha; Natarajaseenivasan, Kalimuthusamy; Vallem, Sandhya; Force, Thomas; Choi, Eric T.; Cheung, Joseph Y.; Madesh, MuniswamyCell Reports (2013), 5 (6), 1576-1588CODEN: CREED8; ISSN:2211-1247. (Cell Press)Resting mitochondrial matrix Ca2+ is maintained through a mitochondrial calcium uptake 1 (MICU1)-established threshold inhibition of mitochondrial calcium uniporter (MCU) activity. It is not known how MICU1 interacts with MCU to establish this Ca2+ threshold for mitochondrial Ca2+ uptake and MCU activity. Here, we show that MICU1 localizes to the mitochondrial matrix side of the inner mitochondrial membrane and MICU1/MCU binding is detd. by a MICU1 N-terminal polybasic domain and two interacting coiled-coil domains of MCU. Further investigation reveals that MICU1 forms homo-oligomers, and this oligomerization is independent of the polybasic region. However, the polybasic region confers MICU1 oligomeric binding to MCU and controls mitochondrial Ca2+ current (IMCU). Moreover, MICU1 EF hands regulate MCU channel activity, but do not det. MCU binding. Loss of MICU1 promotes MCU activation leading to oxidative burden and a halt to cell migration. These studies establish a mol. mechanism for MICU1 control of MCU-mediated mitochondrial Ca2+ accumulation, and dysregulation of this mechanism probably enhances vascular dysfunction.
- 25Petrungaro, C.; Zimmermann, K. M.; Küttner, V.; Fischer, M.; Dengjel, J.; Bogeski, I.; Riemer, J. The Ca2+-Dependent Release of the Mia40-Induced MICU1-MICU2 Dimer from MCU Regulates Mitochondrial Ca2+ Uptake. Cell Metab. 2015, 22, 721– 733, DOI: 10.1016/j.cmet.2015.08.019Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFaqsr%252FI&md5=92341726e998629476fc842d3c950665The Ca2+-Dependent Release of the Mia40-Induced MICU1-MICU2 Dimer from MCU Regulates Mitochondrial Ca2+ UptakePetrungaro, Carmelina; Zimmermann, Katharina M.; Kuettner, Victoria; Fischer, Manuel; Dengjel, Joern; Bogeski, Ivan; Riemer, JanCell Metabolism (2015), 22 (4), 721-733CODEN: CMEEB5; ISSN:1550-4131. (Elsevier Inc.)The essential oxidoreductase Mia40/CHCHD4 mediates disulfide bond formation and protein folding in the mitochondrial intermembrane space. Here, we investigated the interactome of Mia40 thereby revealing links between thiol-oxidn. and apoptosis, energy metab., and Ca2+ signaling. Among the interaction partners of Mia40 is MICU1-the regulator of the mitochondrial Ca2+ uniporter (MCU), which transfers Ca2+ across the inner membrane. We examd. the biogenesis of MICU1 and find that Mia40 introduces an intermol. disulfide bond that links MICU1 and its inhibitory paralog MICU2 in a heterodimer. Absence of this disulfide bond results in increased receptor-induced mitochondrial Ca2+ uptake. In the presence of the disulfide bond, MICU1-MICU2 heterodimer binding to MCU is controlled by Ca2+ levels: the dimer assocs. with MCU at low levels of Ca2+ and dissocs. upon high Ca2+ concns. Our findings support a model in which mitochondrial Ca2+ uptake is regulated by a Ca2+-dependent remodeling of the uniporter complex.
- 26Lee, Y.; Min, C. K.; Kim, T. G.; Song, H. K.; Lim, Y.; Kim, D.; Shin, K.; Kang, M.; Kang, J. Y.; Youn, H.-S.; Lee, J.-G.; An, J. Y.; Park, K. R.; Lim, J. J.; Kim, J. H.; Kim, J. H.; Park, Z. Y.; Kim, Y.-S.; Wang, J.; Kim, D. H.; Eom, S. H. Structure and Function of the N-Terminal Domain of the Human Mitochondrial Calcium Uniporter. EMBO Rep. 2015, 16, 1318– 1333, DOI: 10.15252/embr.201540436Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsVGntbrJ&md5=4b530d1c88f106b0ed8a5e2103c46e9dStructure and function of the N-terminal domain of the human mitochondrial calcium uniporterLee, Youngjin; Min, Choon Kee; Kim, Tae Gyun; Song, Hong Ki; Lim, Yunki; Kim, Dongwook; Shin, Kahee; Kang, Moonkyung; Kang, Jung Youn; Youn, Hyung-Seop; Lee, Jung-Gyu; An, Jun Yop; Park, Kyoung Ryoung; Lim, Jia Jia; Kim, Ji Hun; Kim, Ji Hye; Park, Zee Yong; Kim, Yeon-Soo; Wang, Jimin; Kim, Do Han; Eom, Soo HyunEMBO Reports (2015), 16 (10), 1318-1333CODEN: ERMEAX; ISSN:1469-221X. (Wiley-VCH Verlag GmbH & Co. KGaA)The mitochondrial calcium uniporter (MCU) is responsible for mitochondrial calcium uptake and homeostasis. It is also a target for the regulation of cellular anti-/pro-apoptosis and necrosis by several oncogenes and tumor suppressors. Herein, we report the crystal structure of the MCU N-terminal domain (NTD) at a resoln. of 1.50 Å in a novel fold and the S92A MCU mutant at 2.75 Å resoln.; the residue S92 is a predicted CaMKII phosphorylation site. The assembly of the mitochondrial calcium uniporter complex (uniplex) and the interaction with the MCU regulators such as the mitochondrial calcium uptake-1 and mitochondrial calcium uptake-2 proteins (MICU1 and MICU2) are not affected by the deletion of MCU NTD. However, the expression of the S92A mutant or a NTD deletion mutant failed to restore mitochondrial Ca2+ uptake in a stable MCU knockdown HeLa cell line and exerted dominant-neg. effects in the wild-type MCU-expressing cell line. These results suggest that the NTD of MCU is essential for the modulation of MCU function, although it does not affect the uniplex formation.
- 27Lee, S. K.; Shanmughapriya, S.; Mok, M. C. Y.; Dong, Z.; Tomar, D.; Carvalho, E.; Rajan, S.; Junop, M. S.; Madesh, M.; Stathopulos, P. B. Structural Insights into Mitochondrial Calcium Uniporter Regulation by Divalent Cations. Cell Chem. Biol. 2016, 23, 1157– 1169, DOI: 10.1016/j.chembiol.2016.07.012Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsVWmtLbL&md5=f414a4d19f961b5c3b040955beb83c6bStructural Insights into Mitochondrial Calcium Uniporter Regulation by Divalent CationsLee, Samuel K.; Shanmughapriya, Santhanam; Mok, Mac C. Y.; Dong, Zhiwei; Tomar, Dhanendra; Carvalho, Edmund; Rajan, Sudarsan; Junop, Murray S.; Madesh, Muniswamy; Stathopulos, Peter B.Cell Chemical Biology (2016), 23 (9), 1157-1169CODEN: CCBEBM; ISSN:2451-9448. (Cell Press)Calcium (Ca2+) flux into the matrix is tightly controlled by the mitochondrial Ca2+ uniporter (MCU) due to vital roles in cell death and bioenergetics. However, the precise at. mechanisms of MCU regulation remain unclear. Here, we solved the crystal structure of the N-terminal matrix domain of human MCU, revealing a β-grasp-like fold with a cluster of neg. charged residues that interacts with divalent cations. Binding of Ca2+ or Mg2+ destabilizes and shifts the self-assocn. equil. of the domain toward monomer. Mutational disruption of the acidic face weakens oligomerization of the isolated matrix domain and full-length human protein similar to cation binding and markedly decreases MCU activity. Moreover, mitochondrial Mg2+ loading or blockade of mitochondrial Ca2+ extrusion suppresses MCU Ca2+-uptake rates. Collectively, our data reveal that the β-grasp-like matrix region harbors an MCU-regulating acidic patch that inhibits human MCU activity in response to Mg2+ and Ca2+ binding.
- 28Tomar, D.; Dong, Z.; Shanmughapriya, S.; Koch, D. A.; Thomas, T.; Hoffman, N. E.; Timbalia, S. A.; Goldman, S. J.; Breves, S. L.; Corbally, D. P.; Nemani, N.; Fairweather, J. P.; Cutri, A. R.; Zhang, X.; Song, J.; Jaña, F.; Huang, J.; Barrero, C.; Rabinowitz, J. E.; Luongo, T. S.; Schumacher, S. M.; Rockman, M. E.; Dietrich, A.; Merali, S.; Caplan, J.; Stathopulos, P. B.; Ahima, R. S.; Cheung, J. Y.; Houser, S. R.; Koch, W. J.; Patel, V.; Gohil, V. M.; Elrod, J. W.; Rajan, S.; Madesh, M. MCUR1 is a Scaffold Factor for the MCU Complex Function and Promotes Mitochondrial Bioenergetics. Cell Rep. 2016, 15, 1673– 1685, DOI: 10.1016/j.celrep.2016.04.050Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XnvV2rs7Y%253D&md5=e38a25d3096ac38e15b8a34e3a61f6d7MCUR1 Is a Scaffold Factor for the MCU Complex Function and Promotes Mitochondrial BioenergeticsTomar, Dhanendra; Dong, Zhiwei; Shanmughapriya, Santhanam; Koch, Diana A.; Thomas, Toby; Hoffman, Nicholas E.; Timbalia, Shrishiv A.; Goldman, Samuel J.; Breves, Sarah L.; Corbally, Daniel P.; Nemani, Neeharika; Fairweather, Joseph P.; Cutri, Allison R.; Zhang, Xueqian; Song, Jianliang; Jana, Fabian; Huang, Jianhe; Barrero, Carlos; Rabinowitz, Joseph E.; Luongo, Timothy S.; Schumacher, Sarah M.; Rockman, Michael E.; Dietrich, Alexander; Merali, Salim; Caplan, Jeffrey; Stathopulos, Peter; Ahima, Rexford S.; Cheung, Joseph Y.; Houser, Steven R.; Koch, Walter J.; Patel, Vickas; Gohil, Vishal M.; Elrod, John W.; Rajan, Sudarsan; Madesh, MuniswamyCell Reports (2016), 15 (8), 1673-1685CODEN: CREED8; ISSN:2211-1247. (Cell Press)Mitochondrial Ca2+ Uniporter (MCU)-dependent mitochondrial Ca2+ uptake is the primary mechanism for increasing matrix Ca2+ in most cell types. However, a limited understanding of the MCU complex assembly impedes the comprehension of the precise mechanisms underlying MCU activity. Here, we report that mouse cardiomyocytes and endothelial cells lacking MCU regulator 1 (MCUR1) have severely impaired [Ca2+]m uptake and IMCU current. MCUR1 binds to MCU and EMRE and function as a scaffold factor. Our protein binding analyses identified the minimal, highly conserved regions of coiled-coil domain of both MCU and MCUR1 that are necessary for heterooligomeric complex formation. Loss of MCUR1 perturbed MCU heterooligomeric complex and functions as a scaffold factor for the assembly of MCU complex. Vascular endothelial deletion of MCU and MCUR1 impaired mitochondrial bioenergetics, cell proliferation, and migration but elicited autophagy. These studies establish the existence of a MCU complex that assembles at the mitochondrial integral membrane and regulates Ca2+-dependent mitochondrial metab.
- 29Gunter, T. E.; Gunter, K. K.; Sheu, S.-S.; Gavin, C. E. Mitochondrial Calcium Transport: Physiological and Pathological Relevance. Am. J. Physiol. 1994, 267, C313– 319, DOI: 10.1152/ajpcell.1994.267.2.C313Google ScholarThere is no corresponding record for this reference.
- 30Hajnóczky, G.; Robb-Gaspers, L. D.; Seitz, M. B.; Thomas, A. P. Decoding of Cytosolic Calcium Oscillations in the Mitochondria. Cell 1995, 82, 415– 424, DOI: 10.1016/0092-8674(95)90430-1Google ScholarThere is no corresponding record for this reference.
- 31Denton, R.; McCormack, J. G. Ca2+ as a Second Messenger Within Mitochondria Of The Heart And Other Tissues. Annu. Rev. Physiol. 1990, 52, 451– 466, DOI: 10.1146/annurev.ph.52.030190.002315Google ScholarThere is no corresponding record for this reference.
- 32Santo-Domingo, J.; Demaurex, N. Calcium Uptake Mechanisms of Mitochondria. Biochim. Biophys. Acta, Bioenerg. 2010, 1797, 907– 912, DOI: 10.1016/j.bbabio.2010.01.005Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXnvV2qtr4%253D&md5=b7a5831a8d832ac99c2a3854f647b76aCalcium uptake mechanisms of mitochondriaSanto-Domingo, Jaime; Demaurex, NicolasBiochimica et Biophysica Acta, Bioenergetics (2010), 1797 (6-7), 907-912CODEN: BBBEB4; ISSN:0005-2728. (Elsevier B. V.)A review. The ability of mitochondria to capture Ca2+ ions has important functional implications for cells, because mitochondria shape cellular Ca2+ signals by acting as a Ca2+ buffer and respond to Ca2+ elevations either by increasing the cell energy supply or by triggering the cell death program of apoptosis. A mitochondrial Ca2+ channel known as the uniporter drives the rapid and massive entry of Ca2+ ions into mitochondria. The uniporter operates at high, micromolar cytosolic Ca2+ concns. that are only reached transiently in cells, near Ca2+ release channels. Mitochondria can also take up Ca2+ at low, nanomolar concns., but this high affinity mode of Ca2+ uptake is not well characterized. Recently, the leucine zipper-EF hand-contg. transmembrane region (Letm1) was proposed to be an electrogenic 1:1 mitochondrial Ca2+/H+ antiporter that drives the uptake of Ca2+ into mitochondria at nanomolar cytosolic Ca2+ concns. Here, the authors review the properties of the Ca2+ import systems of mitochondria and discuss how Ca2+ uptake via an electrogenic 1:1 Ca2+/H+ antiport challenges the current thinking of the mitochondrial Ca2+ uptake mechanism.
- 33Bernardi, P. Mitochondrial Transport of Cations: Channels, Exchangers, and Permeability Transition. Physiol. Rev. 1999, 79, 1127– 1155, DOI: 10.1152/physrev.1999.79.4.1127Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXmvVymtLw%253D&md5=594fe4b9901aee10ba77bf8164865822Mitochondrial transport of cations: channels, exchangers, and permeability transitionBernardi, PaoloPhysiological Reviews (1999), 79 (4), 1127-1155CODEN: PHREA7; ISSN:0031-9333. (American Physiological Society)A review with 372 refs. This review provides a selective history of how studies of mitochondrial cation transport (K+, Na+, Ca2+) developed in relation to the major themes of research in bioenergetics. It then covers in some detail specific transport pathways for these cations, and it introduces and discusses open problems about their nature and physiol. function, particularly in relation to vol. regulation and Ca2+ homeostasis. The review should provide the basic elements needed to understand both earlier mitochondrial literature and current problems assocd. with mitochondrial transport of cations and hopefully will foster new interest in the mol. definition of mitochondrial cation channels and exchangers as well as their roles in cell physiol.
- 34Shanmughapriya, S.; Rajan, S.; Hoffman, N. E.; Higgins, A. M.; Tomar, D.; Nemani, N.; Hines, K. J.; Smith, D. J.; Eguchi, A.; Vallem, S.; Shaikh, F.; Cheung, M.; Leonard, N. J.; Stolakis, R. S.; Wolfers, M. P.; Ibetti, J.; Chuprun, J. K.; Jog, N. R.; Houser, S. R.; Koch, W. J.; Elrod, J. W.; Madesh, M. SPG7 is an Essential and Conserved Component of the Mitochondrial Permeability Transition Pore. Mol. Cell 2015, 60, 47– 62, DOI: 10.1016/j.molcel.2015.08.009Google ScholarThere is no corresponding record for this reference.
- 35Orrenius, S.; Zhivotovsky, B.; Nicotera, P. Regulation of Cell Death: The Calcium-Apoptosis Link. Nat. Rev. Mol. Cell Biol. 2003, 4, 552– 565, DOI: 10.1038/nrm1150Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXltVWnu7g%253D&md5=6efb194177308e68f3d9e3545d7095f6Calcium: Regulation of cell death: the calcium-apoptosis linkOrrenius, Sten; Zhivotovsky, Boris; Nicotera, PierluigiNature Reviews Molecular Cell Biology (2003), 4 (7), 552-565CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review and discussion. To live or to die. This crucial question eloquently reflects the dual role of Ca2+ in living organisms, as a survival factor or as a ruthless killer. It has long been known that Ca2+ signals govern a host of vital cell functions and so are necessary for cell survival. However, more recently it has become clear that cellular Ca2+ overload, or perturbation of intracellular Ca2+ compartmentalization, can cause cytotoxicity and trigger either apoptotic or necrotic cell death.
- 36De Stefani, D.; Rizzuto, R.; Pozzan, T. Enjoy the Trip: Calcium in Mitochondria Back and Forth. Annu. Rev. Biochem. 2016, 85, 161– 192, DOI: 10.1146/annurev-biochem-060614-034216Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XntlKgsbs%253D&md5=5ce87bb458bf96a84be012e840b27580Enjoy the Trip: Calcium in Mitochondria Back and ForthDe Stefani, Diego; Rizzuto, Rosario; Pozzan, TullioAnnual Review of Biochemistry (2016), 85 (), 161-192CODEN: ARBOAW; ISSN:0066-4154. (Annual Reviews)A review. In the last 5 years, most of the mols. that control mitochondrial Ca2+ homeostasis have been finally identified. Mitochondrial Ca2+ uptake is mediated by the Mitochondrial Calcium Uniporter (MCU) complex, a macromol. structure that guarantees Ca2+ accumulation inside mitochondrial matrix upon increases in cytosolic Ca2+. Conversely, Ca2+ release is under the control of the Na+/Ca2+ exchanger, encoded by the NCLX gene, and of a H+/Ca2+ antiporter, whose identity is still debated. The low affinity of the MCU complex, coupled to the activity of the efflux systems, protects cells from continuous futile cycles of Ca2+ across the inner mitochondrial membrane and consequent massive energy dissipation. In this review, we discuss the basic principles that govern mitochondrial Ca2+ homeostasis and the methods used to investigate the dynamics of Ca2+ concn. within the organelles. We discuss the functional and structural role of the different mols. involved in mitochondrial Ca2+ handling and their pathophysiol. role.
- 37Marchi, S.; Pinton, P. The Mitochondrial Calcium Uniporter Complex: Molecular Components, Structure and Physiopathological Implications. J. Physiol. 2014, 592, 829– 839, DOI: 10.1113/jphysiol.2013.268235Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXjtlOkuro%253D&md5=5f8c78976edf6e35e484a9a40e107784The mitochondrial calcium uniporter complex: molecular components, structure and physiopathological implicationsMarchi, Saverio; Pinton, PaoloJournal of Physiology (Oxford, United Kingdom) (2014), 592 (5), 829-839CODEN: JPHYA7; ISSN:0022-3751. (Wiley-Blackwell)A review. Although it has long been known that mitochondria take up Ca2+, the mol. identities of the channels and transporters involved in this process were revealed only recently. Here, we discuss the recent work that has led to the characterization of the mitochondrial calcium uniporter complex, which includes the channel-forming subunit MCU (mitochondrial calcium uniporter) and its regulators MICU1, MICU2, MCUb, EMRE, MCUR1 and miR-25. We review not only the biochem. identities and structures of the proteins required for mitochondrial Ca2+ uptake but also their implications in different physiopathol. contexts.
- 38Luongo, T. S.; Lambert, J. P.; Yuan, A.; Zhang, X.; Gross, P.; Song, J.; Shanmughapriya, S.; Gao, E.; Jain, M.; Houser, S. R.; Koch, W. J.; Cheung, J. Y.; Madesh, M.; Elrod, J. W. The Mitochondrial Calcium Uniporter Matches Energetic Supply with Cardiac Workload during Stress and Modulates Permeability Transition. Cell Rep. 2015, 12, 23– 34, DOI: 10.1016/j.celrep.2015.06.017Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtV2itbbP&md5=35f1feedc69b60ede6620f6722a35fbcThe Mitochondrial Calcium Uniporter Matches Energetic Supply with Cardiac Workload during Stress and Modulates Permeability TransitionLuongo, Timothy S.; Lambert, Jonathan P.; Yuan, Ancai; Zhang, Xueqian; Gross, Polina; Song, Jianliang; Shanmughapriya, Santhanam; Gao, Erhe; Jain, Mohit; Houser, Steven R.; Koch, Walter J.; Cheung, Joseph Y.; Madesh, Muniswamy; Elrod, John W.Cell Reports (2015), 12 (1), 23-34CODEN: CREED8; ISSN:2211-1247. (Cell Press)Cardiac contractility is mediated by a variable flux in intracellular calcium (Ca2+), thought to be integrated into mitochondria via the mitochondrial calcium uniporter (MCU) channel to match energetic demand. Here, we examine a conditional, cardiomyocyte-specific, mutant mouse lacking Mcu, the pore-forming subunit of the MCU channel, in adulthood. Mcu-/- mice display no overt baseline phenotype and are protected against mCa2+ overload in an in vivo myocardial ischemia-reperfusion injury model by preventing the activation of the mitochondrial permeability transition pore, decreasing infarct size, and preserving cardiac function. In addn., we find that Mcu-/- mice lack contractile responsiveness to acute β-adrenergic receptor stimulation and in parallel are unable to activate mitochondrial dehydrogenases and display reduced bioenergetic reserve capacity. These results support the hypothesis that MCU may be dispensable for homeostatic cardiac function but required to modulate Ca2+-dependent metab. during acute stress.
- 39Kwong, J. Q.; Lu, X.; Correll, R. N.; Schwanekamp, J. A.; Vagnozzi, R. J.; Sargent, M. A.; York, A. J.; Zhang, J.; Bers, D. M.; Molkentin, J. D. The Mitochondrial Calcium Uniporter Selectively Matches Metabolic Output to Acute Contractile Stress in the Heart. Cell Rep. 2015, 12, 15– 22, DOI: 10.1016/j.celrep.2015.06.002Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtV2itbnM&md5=18e145fd5c845366c5ef24b91b079451The Mitochondrial Calcium Uniporter Selectively Matches Metabolic Output to Acute Contractile Stress in the HeartKwong, Jennifer Q.; Lu, Xiyuan; Correll, Robert N.; Schwanekamp, Jennifer A.; Vagnozzi, Ronald J.; Sargent, Michelle A.; York, Allen J.; Zhang, Jianyi; Bers, Donald M.; Molkentin, Jeffery D.Cell Reports (2015), 12 (1), 15-22CODEN: CREED8; ISSN:2211-1247. (Cell Press)In the heart, augmented Ca2+ fluxing drives contractility and ATP generation through mitochondrial Ca2+ loading. Pathol. mitochondrial Ca2+ overload with ischemic injury triggers mitochondrial permeability transition pore (MPTP) opening and cardiomyocyte death. Mitochondrial Ca2+ uptake is primarily mediated by the mitochondrial Ca2+ uniporter (MCU). Here, we generated mice with adult and cardiomyocyte-specific deletion of Mcu, which produced mitochondria refractory to acute Ca2+ uptake, with impaired ATP prodn., and inhibited MPTP opening upon acute Ca2+ challenge. Mice lacking Mcu in the adult heart were also protected from acute ischemia-reperfusion injury. However, resting/basal mitochondrial Ca2+ levels were normal in hearts of Mcu-deleted mice, and mitochondria lacking MCU eventually loaded with Ca2+ after stress stimulation. Indeed, Mcu-deleted mice were unable to immediately sprint on a treadmill unless warmed up for 30 min. Hence, MCU is a dedicated regulator of short-term mitochondrial Ca2+ loading underlying a "fight-or-flight" response that acutely matches cardiac workload with ATP prodn.
- 40Medvedeva, Y. V.; Weiss, J. H.; Weiss, J. H.; Medvedeva, Y. V. Intramitochondrial Zn2+ Accumulation via the Ca2+ Uniporter Contributes to Acute Ischemic Neurodegeneration. Neurobiol. Dis. 2014, 68, 137– 144, DOI: 10.1016/j.nbd.2014.04.011Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtVWrtLfK&md5=695cd0a52d3fab9addb4805e0da8af60Intramitochondrial Zn2 + accumulation via the Ca2 + uniporter contributes to acute ischemic neurodegenerationMedvedeva, Yuliya V.; Weiss, John H.Neurobiology of Disease (2014), 68 (), 137-144CODEN: NUDIEM; ISSN:0969-9961. (Elsevier Inc.)Ca2 + and Zn2 + have both been implicated in the induction of acute ischemic neurodegeneration. We recently examd. changes in intracellular Zn2 + and Ca2 + in CA1 pyramidal neurons subjected to oxygen glucose deprivation (OGD), and found that Zn2 + rises precede and contribute to the onset of terminal Ca2 + rises ("Ca2 + deregulation"), which are causatively linked to a lethal loss of membrane integrity. The present study seeks to examine the specific role of intramitochondrial Zn2 + accumulation in ischemic injury, using blockers of the mitochondrial Ca2 + uniporter (MCU), through which both Zn2 + and Ca2 + appear able to enter the mitochondrial matrix. In physiol. extracellular Ca2 +, treatment with the MCU blocker, Ruthenium Red (RR), accelerated the Ca2 + deregulation, most likely by disrupting mitochondrial Ca2 + buffering and thus accelerating the lethal cytosolic Ca2 + overload. However, when intracellular Ca2 + overload was slowed, either by adding blockers of major Ca2 + entry channels or by lowering the concn. of Ca2 + in the extracellular buffer, Ca2 + deregulation was delayed, and under these conditions either Zn2 + chelation or MCU blockade resulted in similar further delays of the Ca2 + deregulation. In parallel studies using the reactive oxygen species (ROS) indicator, hydroethidine, lowering Ca2 + surprisingly accelerated OGD induced ROS generation, and in these low Ca2 + conditions, either Zn2 + chelation or MCU block slowed the ROS generation. These studies suggest that, during acute ischemia, Zn2 + entry into mitochondria via the MCU induces mitochondrial dysfunction (including ROS generation) that occurs upstream of, and contributes to the terminal Ca2 + deregulation.
- 41Giorgi, C.; Agnoletto, C.; Bononi, A.; Bonora, M.; De Marchi, E.; Marchi, S.; Missiroli, S.; Patergnani, S.; Poletti, F.; Rimessi, A.; Suski, J. M.; Wieckowski, M. R.; Pinton, P. Mitochondrial Calcium Homeostasis as Potential Target for Mitochondrial Medicine. Mitochondrion 2012, 12, 77– 85, DOI: 10.1016/j.mito.2011.07.004Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhvVOltrc%253D&md5=32b483cd42cd9ee7c1f0db4187eb0bceMitochondrial calcium homeostasis as potential target for mitochondrial medicineGiorgi, Carlotta; Agnoletto, Chiara; Bononi, Angela; Bonora, Massimo; De Marchi, Elena; Marchi, Saverio; Missiroli, Sonia; Patergnani, Simone; Poletti, Federica; Rimessi, Alessandro; Suski, Jan M.; Wieckowski, Mariusz R.; Pinton, PaoloMitochondrion (2012), 12 (1), 77-85CODEN: MITOCN; ISSN:1567-7249. (Elsevier B.V.)A review. Mitochondria are crucial in different intracellular pathways of signal transduction. Mitochondria are capable of decoding a variety of extracellular stimuli into markedly different intracellular actions, ranging from energy prodn. to cell death. The fine modulation of mitochondrial calcium (Ca2+) homeostasis plays a fundamental role in many of the processes involving this organelle. When mitochondrial Ca2+ homeostasis is compromised, different pathol. conditions can occur, depending on the cell type involved. Recent data have shed light on the mol. identity of the main proteins involved in the handling of mitochondrial Ca2+ traffic, opening fascinating and ambitious new avenues for mitochondria-based pharmacol. strategies.
- 42Arduino, D. M.; Wettmarshausen, J.; Vais, H.; Navas-Navarro, P.; Cheng, Y.; Leimpek, A.; Ma, Z.; Delrio-Lorenzo, A.; Giordano, A.; Garcia-Perez, C.; Médard, G.; Kuster, B.; García-Sancho, J.; Mokranjac, D.; Foskett, J. K.; Alonso, M. T.; Perocchi, F. Systematic Identification of MCU Modulators by Orthogonal Interspecies Chemical Screening. Mol. Cell 2017, 67, 711– 723, DOI: 10.1016/j.molcel.2017.07.019Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtlKmsbfO&md5=f9cfb524e127ab132d85c3cc49161dd4Systematic Identification of MCU Modulators by Orthogonal Interspecies Chemical ScreeningArduino, Daniela M.; Wettmarshausen, Jennifer; Vais, Horia; Navas-Navarro, Paloma; Cheng, Yiming; Leimpek, Anja; Ma, Zhongming; Delrio-Lorenzo, Alba; Giordano, Andrea; Garcia-Perez, Cecilia; Medard, Guillaume; Kuster, Bernhard; Garcia-Sancho, Javier; Mokranjac, Dejana; Foskett, J. Kevin; Alonso, M. Teresa; Perocchi, FabianaMolecular Cell (2017), 67 (4), 711-723.e7CODEN: MOCEFL; ISSN:1097-2765. (Elsevier Inc.)The mitochondrial calcium uniporter complex is essential for calcium (Ca2+) uptake into mitochondria of all mammalian tissues, where it regulates bioenergetics, cell death, and Ca2+ signal transduction. Despite its involvement in several human diseases, we currently lack pharmacol. agents for targeting uniporter activity. Here we introduce a high-throughput assay that selects for human MCU-specific small-mol. modulators in primary drug screens. Using isolated yeast mitochondria, reconstituted with human MCU, its essential regulator EMRE, and aequorin, and exploiting a D-lactate- and mannitol/sucrose-based bioenergetic shunt that greatly minimizes false-pos. hits, we identify mitoxantrone out of more than 600 clin. approved drugs as a direct selective inhibitor of human MCU. We validate mitoxantrone in orthogonal mammalian cell-based assays, demonstrating that our screening approach is an effective and robust tool for MCU-specific drug discovery and, more generally, for the identification of compds. that target mitochondrial functions.
- 43Kon, N.; Murakoshi, M.; Isobe, A.; Kagechika, K.; Miyoshi, N.; Nagayama, T. DS16570511 is a Small-Molecule Inhibitor of the Mitochondrial Calcium Uniporter. Cell Death Discov. 2017, 3, 17045, DOI: 10.1038/cddiscovery.2017.45Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFygtLzI&md5=351f1655a4841af9ee62ee64e691e85bDS16570511 is a small-molecule inhibitor of the mitochondrial calcium uniporterKon, Naohiro; Murakoshi, Michiko; Isobe, Aya; Kagechika, Katsuji; Miyoshi, Naoki; Nagayama, TakahiroCell Death Discovery (2017), 3 (), 17045CODEN: CDDEB5; ISSN:2058-7716. (Nature Publishing Group)In cardiac myocytes, regulation of mitochondrial Ca2+ is important for cellular signaling and cardiac contraction. Ca2+ entry into the mitochondria is mediated by a highly selective Ca2+ channel called the mitochondrial calcium uniporter, which consists of a pore-forming subunit MCU and regulatory subunits such as MICU1. Although pharmacol. regulation of the mitochondrial Ca2+ influx is a promising approach to controlling the cellular functions, a cell-permeable and specific inhibitor of the mitochondrial calcium uniporter has not yet been developed. Here, we identify a novel cell-permeable inhibitor of the uniporter by a high-throughput screening of 120 000 small-mol. compds. In our study, DS16570511 dose-dependently inhibited serum-induced mitochondrial Ca2+ influx in HEK293A cells with an IC50 of 7 μM. DS16570511 inhibited Ca2+ uptake of isolated mitochondria from human cells, rat heart and pig heart. Overexpression of hMCU or hMICU1 in HEK293A cells increased mitochondrial Ca2+ influx, and the increases were completely suppressed by the pretreatment with DS16570511. DS16570511 also blocks mitochondrial Ca2+ overload in a Langendorff perfused beating rat heart. Interestingly, DS16570511 increased cardiac contractility without affecting heart rate in the perfused heart. These results show that DS16570511 is a novel cell-permeable inhibitor of the mitochondrial calcium uniporter and applicable for control of the cardiac functions.
- 44Thu, V. T.; Kim, H. K.; Long, L. T.; Lee, S. R.; Hanh, T. M.; Ko, T. H.; Heo, H. J.; Kim, N.; Kim, S. H.; Ko, K. S.; Rhee, B. D.; Han, J. NecroX-5 Prevents Hypoxia/Reoxygenation Injury by Inhibiting the Mitochondrial Calcium Uniporter. Cardiovasc. Res. 2012, 94, 342– 350, DOI: 10.1093/cvr/cvs122Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XmtVegsrc%253D&md5=356fbd417f7ef089927c33aebdafa7c2NecroX-5 prevents hypoxia/reoxygenation injury by inhibiting the mitochondrial calcium uniporterThu, Vu Thi; Kim, Hyoung-Kyu; Long, Le Thanh; Lee, Sung-Ryul; Hanh, Tran My; Ko, Tae Hee; Heo, Hye-Jin; Kim, Nari; Kim, Soon Ha; Ko, Kyung Soo; Rhee, Byoung Doo; Han, JinCardiovascular Research (2012), 94 (2), 342-350CODEN: CVREAU; ISSN:0008-6363. (Oxford University Press)Aims: Preservation of mitochondrial function is essential to limit myocardial damage in ischemic heart disease. We examd. the protective effects and mechanism of a new compd., NecroX-5, on rat heart mitochondria in a hypoxia/reoxygenation (HR) model. Methods and results: NecroX-5 reduced mitochondrial oxidative stress, prevented the collapse in mitochondrial membrane potential, improved mitochondrial oxygen consumption, and suppressed mitochondrial Ca2+ overload during reoxygenation in an in vitro rat heart HR model. Furthermore, NecroX-5 reduced the ouabain- or histamine-induced increase in mitochondrial Ca2+. Conclusion: These findings suggest that NecroX-5 may act as a mitochondrial Ca2+ uniporter inhibitor to protect cardiac mitochondria against HR damage.
- 45Antonenko, Y. N.; Rokitskaya, T. I.; Cooper, A. J. L.; Krasnikov, B. F. Minocycline Chelates Ca2+, Binds to Membranes, and Depolarizes Mitochondria by Formation of Ca2+-Dependent Ion Channels. J. Bioenerg. Biomembr. 2010, 42, 151– 163, DOI: 10.1007/s10863-010-9271-1Google ScholarThere is no corresponding record for this reference.
- 46Santo-Domingo, J.; Vay, L.; Hernández-Sanmiguel, E.; Lobatón, C. D.; Moreno, A.; Montero, M.; Alvarez, J. The Plasma Membrane Na+/Ca2+ Exchange Inhibitor KB-R7943 is Also a Potent Inhibitor of the Mitochondrial Ca2+ Uniporter. Br. J. Pharmacol. 2007, 151, 647– 654, DOI: 10.1038/sj.bjp.0707260Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXntFemsbk%253D&md5=09d94d943bd1e5e1000a4ec7d367fc8aThe plasma membrane Na+/Ca2+ exchange inhibitor KB-R7943 is also a potent inhibitor of the mitochondrial Ca2+ uniporterSanto-Domingo, J.; Vay, L.; Hernandez-SanMiguel, E.; Lobaton, C. D.; Moreno, A.; Montero, M.; Alvarez, J.British Journal of Pharmacology (2007), 151 (5), 647-654CODEN: BJPCBM; ISSN:0007-1188. (Nature Publishing Group)Background and purpose: The thiourea deriv. KB-R7943, originally developed as inhibitor of the plasma membrane Na+/Ca2+ exchanger, has been shown to protect against myocardial ischemia-reperfusion injury. We have studied here its effects on mitochondrial Ca2+ fluxes. [Ca2+] in cytosol, mitochondria and endoplasmic reticulum (ER), and mitochondrial membrane potential were monitored using both luminescent (targeted aequorins) and fluorescent (fura-2, tetramethylrhodamine Et ester) probes in HeLa cells. KB-R7943 was also a potent inhibitor of the mitochondrial Ca2+ uniporter (MCU). In permeabilized HeLa cells, KB-R7943 inhibited mitochondrial Ca2+ uptake with a Ki of 5.5±1.3 μM (mean±S.D.). In intact cells, 10μM KB-R7943 reduced by 80% the mitochondrial [Ca2+] peak induced by histamine. KB-R7943 did not modify the mitochondrial membrane potential and had no effect on the mitochondrial Na+/Ca2+ exchanger. KB-R7943 inhibited histamine-induced ER-Ca2+ release in intact cells, but not in cells loaded with a Ca2+-chelator to damp cytosolic [Ca2+] changes. Therefore, inhibition of ER-Ca2+-release by KB-R7943 was probably due to the increased feedback Ca2+-inhibition of inositol 1,4,5-trisphosphate receptors after MCU block. This mechanism also explains why KB-R7943 reversibly blocked histamine-induced cytosolic [Ca2+] oscillations in the same range of concns. required to inhibit MCU. Inhibition of MCU by KB-R7943 may contribute to its cardioprotective activity by preventing mitochondrial Ca2+-overload during ischemia-reperfusion. In addn., the effects of KB-R7943 on Ca2+ homeostasis provide new evidence for the role of mitochondria modulating Ca2+-release and regenerative Ca2+-oscillations. Search for permeable and selective MCU inhibitors may yield useful pharmacol. tools in the future.
- 47Kapuscinski, J.; Darzynkiewicz, Z. Interactions of Antitumor Agents Ametantrone and Mitoxantrone (Novatrone) with Double-Stranded DNA. Biochem. Pharmacol. 1985, 34, 4203– 4213, DOI: 10.1016/0006-2952(85)90275-8Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL28XotFKitA%253D%253D&md5=f6f28eedd99ec102a125650d13f86b24Interactions of antitumor agents Ametantrone and mitoxantrone (Novatrone) with double-stranded DNAKapuscinski, Jan; Darzynkiewicz, ZbigniewBiochemical Pharmacology (1985), 34 (24), 4203-13CODEN: BCPCA6; ISSN:0006-2952.Interactions of mitoxantrone and ametantrone with natural and synthetic nucleic acids in aq. medium [0.15 NaCl, 5 mM 4-(2-hydroxyethyl)-1-piperazine-ethanesulfonic acid (Hepes), pH 7.0. 25%] were studied using computer-aided spectrophotometric techniques. Absorption spectra of the drugs in monomeric and dimeric form and their complexes with DNAs at low drug/phosphate ratios (D/P) were established. The latter were red-shifted and had lower amplitude as compared with the spectra of the free ligand's monomer; the change is consistent with the already well-established intercalative mode of drug-nucleic acid interaction. Drug-DNA equil. were studied using the McGhee-von Hippel model of noncooperative ligand-polymer interaction, with the correction for dimerization of drugs. Although mitoxantrone was 2 orders of magnitude more potent an antitumor drug than Ametantrone, the intrinsic assocn. consts. (Ki) of both drugs were of similar magnitude. Also, no significant DNA-base specificity for either of the drugs (measured as Ki value for various homopolymers) was obsd. Therefore, no correlation was apparent between the intercalative mode of binding to DNA, regardless of base compn., and the pharmacol. activity of these drugs. At higher D/P ratios, a secondary mode of binding was detected by both spectroscopy and light-scattering measurement. Homopolymer-pairs and polymers contg. only dI and dC were esp. susceptible to this secondary type of binding. The possibility that this secondary type of binding may be responsible for the antitumor properties of the drugs is considered.
- 48Mazerski, J.; Martelli, S.; Borowski, E. The Geometry of Intercalation Complex of Antitumor Mitoxantrone and Ametantrone with DNA: Molecular Dynamics Simulations. Acta Biochim. Polym. 1998, 45, 1– 11Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXkt1egsbg%253D&md5=84573969f77849b9405683e28bd32d27The geometry of intercalation complex of antitumor mitoxantrone and ametantrone with DNA: molecular dynamics simulationsMazerski, Jan; Martelli, Sante; Borowski, EdwardActa Biochimica Polonica (1998), 45 (1), 1-11CODEN: ABPLAF; ISSN:0001-527X. (Polish Biochemical Society)Intercalative binding of the antitumor drugs ametantrone and mitoxantrone to the dodecamer duplex d(CGCGAGCTCGCG)2 was studied by applying mol. dynamics in water with the GROMOS 87 force field. A no. of reasonable binding orientations were tested by short pre-simulations. It was shown that in energetically favorable orientation the anthraquinone chromophore is perpendicular to the direction of inter-base hydrogen bonds. Helically shaped side-chains of the drugs fit to the minor groove. The best orientation obtained in pre-simulations was applied in the main simulations. Small but significant differences were found between structures of inter-calation complexes of the two drugs with the dodecamer duplex, the mitoxantrone complex possessing more favorable energy. The mol. nature of interactions responsible for those differences has been discussed.
- 49Boland, M. P.; Fitzgerald, K. A.; O’Neill, L. A. J. Topoisomerase II Is Required for Mitoxantrone to Signal Nuclear Factor ΚB Activation in HL60 Cells. J. Biol. Chem. 2000, 275, 25231– 25238, DOI: 10.1074/jbc.275.33.25231Google ScholarThere is no corresponding record for this reference.
- 50Smith, P. J.; Morgan, S. A.; Fox, M. E.; Watson, J. V. Mitoxantrone-DNA Binding and the Induction of Topoisomerase II Associated DNA Damage in Multi-Drug Resistant Small Cell Lung Cancer Cells. Biochem. Pharmacol. 1990, 40, 2069– 2078, DOI: 10.1016/0006-2952(90)90237-FGoogle ScholarThere is no corresponding record for this reference.
- 51Nägele, H.; Castel, M. A.; Deutsch, O.; Wagner, F. M.; Reichenspurner, H. Heart Transplantation in a Patient with Multiple Sclerosis and Mitoxantrone-Induced Cardiomyopathy. J. Heart Lung Transplant. 2004, 23, 641– 643, DOI: 10.1016/S1053-2498(03)00307-3Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2c3jslKitA%253D%253D&md5=ce71e137ff3f9fbce1c00a9c4fdc31d4Heart transplantation in a patient with multiple sclerosis and mitoxantrone-induced cardiomyopathyNagele H; Castel M A; Deutsch O; Wagner F M; Reichenspurner HThe Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation (2004), 23 (5), 641-3 ISSN:1053-2498.We describe a 30-year-old man with end-stage heart failure after therapy with mitoxantrone for multiple sclerosis. A successful orthotopic heart transplantation was performed when intensified medical therapy failed to improve the patient's hemodynamics. In spite of the severe underlying disease he did well on dual immunosuppression with methylprednisone and cyclosporine. Neurologic symptoms remained stable throughout the procedure and, after 2 months, he resumed preoperative ambulatory status. Eight years after the operation, the patient is now in New York Heart Association (NYHA) Class I status. Using canes, he is able to walk short distances. Repeated urinary tract infections caused by Escherichia coli became a problem, but have been controlled by long-term oral antibiotic prophylaxis with trimethoprim.
- 52Goebel, M.; Kaplan, E. Anthracycline-Induced Cardiotoxicity – A Review. Oncol. Res. Treat. 2004, 15, 198– 204, DOI: 10.1159/000217360Google ScholarThere is no corresponding record for this reference.
- 53Ying, W.-L.; Emerson, J.; Clarke, M. J.; Sanadi, D. R. Inhibition of Mitochondrial Calcium Ion Transport by an Oxo-Bridged Dinuclear Ruthenium Ammine Complex. Biochemistry 1991, 30, 4949– 4952, DOI: 10.1021/bi00234a016Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXit1eksrk%253D&md5=dcf10f1224e51c3dce2d31eadc77dbb4Inhibition of mitochondrial calcium ion transport by an oxo-bridged dinuclear ruthenium ammine complexYing, Wen Long; Emerson, Jeffrey; Clarke, Michael J.; Sanadi, D. RaoBiochemistry (1991), 30 (20), 4949-52CODEN: BICHAW; ISSN:0006-2960.Ruthenium red is a well-known and effective inhibitor of the mitochondria Ca2+ uniporter; however, K.C. Reed and F. Bygrave (1974) tentatively attributed this inhibition to a colorless impurity present in com. samples of ruthenium red (RR). This component has now been isolated and a deriv., (μ-O)[(HCO2)(NH3)4Ru]2Cl3, structurally characterized. The active species in soln. appears to be the sym. oxo-bridged ion, [X(NH3)4Ru-O-Ru(NH3)4X]3+, where X = Cl- or OH-. Its absorption spectrum shows a max. at 360 nm. The dinuclear ruthenium ammine complex inhibits Ca2+-stimulated respiration of rat liver mitochondria with an I50 of 3.5 pmol/mg of protein compared to the value of 60 pmol of RR/mg of protein. The inhibition by the dinuclear compd. is noncompetitive with Ca2+. Respiration-linked swelling of mitochondria induced by Cd2+ also responds similarly to both the dinuclear complex and RR. A close correlation was obsd. between binding to mitochondria as monitored with 103Ru-labeled dinuclear complex and inhibition of Ca2+ transport. A Scatchard plot yielded ests. of max. specific binding and dissocn. const. of 7.5 pmol/mg of protein and 1.3 nM, resp. The inhibitor has the characteristics of a satisfactory affinity ligand for purifn. of the uniporter.
- 54Matlib, M. A.; Zhou, Z.; Knight, S.; Ahmed, S.; Choi, K. M.; Krause-Bauer, J.; Phillips, R.; Altschuld, R.; Katsube, Y.; Sperelakis, N.; Bers, D. M. Oxygen-Bridged Dinuclear Ruthenium Amine Complex Specifically Inhibits Ca2+ Uptake into Mitochondria in Vitro and in Situ in Single Cardiac Myocytes. J. Biol. Chem. 1998, 273, 10223– 10231, DOI: 10.1074/jbc.273.17.10223Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXivFKnsbc%253D&md5=53f8261b91276b659d0038814914ec86Oxygen-bridged dinuclear ruthenium amine complex specifically inhibits Ca2+ uptake into mitochondria in vitro and in situ in single cardiac myocytesMatlib, Mohammed A.; Zhou, Zhuan; Knight, Selena; Ahmed, Saadia; Choi, Kin M.; Krause-Bauer, Jeanette; Phillips, Ronald; Altschuld, Ruth; Katsube, Yasuhiro; Sperelakis, Nicholas; Bers, Donald M.Journal of Biological Chemistry (1998), 273 (17), 10223-10231CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Ruthenium red is a well known inhibitor of Ca2+ uptake into mitochondria in vitro. However, its utility as an inhibitor of Ca2+ uptake into mitochondria in vivo or in situ in intact cells is limited because of its inhibitory effects on sarcoplasmic reticulum Ca2+ release channel and other cellular processes. We have synthesized a ruthenium deriv. and found it to be an oxygen-bridged dinuclear ruthenium amine complex. It has the same chem. structure as Ru360 reported previously (Emerson, J., Clarke, M. J., Ying, W-L., and Sanadi, D. R. (1993) J. Am. Chem. Soc. 115, 11799-11805). Ru360 has been shown to be a potent inhibitor of Ca2+-stimulated respiration of liver mitochondria in vitro. However, the specificity of Ru360 on Ca2+ uptake into mitochondria in vitro or in intact cells has not been detd. The present study reports in detail the potency, the effectiveness, and the mechanism of inhibition of mitochondrial Ca2+ uptake by Ru360 and its specificity in vitro in isolated mitochondria and in situ in isolated cardiac myocytes. Ru360 was more potent (IC50 = 0.184 nM) than ruthenium red (IC50 = 6.85 nM) in inhibiting Ca2+ uptake into mitochondria. 103Ru360 was found to bind to isolated mitochondria with high affinity (Kd = 0.34 nM, Bmax = 80 fmol/mg of mitochondrial protein). The IC50 of 103Ru360 for the inhibition of Ca2+ uptake into mitochondria was also 0.2 nM, indicating that satn. of a specific binding site is responsible for the inhibition of Ca2+ uptake. Ru360, as high as 10 μM, produced no effect on sarcoplasmic reticulum Ca2+ uptake or release, sarcolemmal Na+/Ca2+ exchange, actomyosin ATPase activity, L-type Ca2+ channel current, cytosolic Ca2+ transients, or cell shortening. 103Ru360 was taken up by isolated myocytes in a time-dependent biphasic manner. Ru360 (10 μM) applied outside intact voltage-clamped ventricular myocytes prevented Ca2+ uptake into mitochondria in situ where the cells were progressively loaded with Ca2+ via sarcolemmal Na+/Ca2+ exchange by depolarization to +110 mV. We conclude that Ru360 specifically blocks Ca2+ uptake into mitochondria and can be used in intact cells.
- 55Emerson, J.; Clarke, M. J.; Ying, W. L.; Sanadi, D. R. The Component of “Ruthenium Red” Responsible for Inhibition of Mitochondrial Calcium Ion Transport. Spectra, Electrochemistry, and Aquation Kinetics. Crystal Structure of μ-O-[(HCO)2(NH3)4Ru]2Cl3. J. Am. Chem. Soc. 1993, 115, 11799– 11805, DOI: 10.1021/ja00078a019Google ScholarThere is no corresponding record for this reference.
- 56Nathan, S. R.; Pino, N. W.; Arduino, D. M.; Perocchi, F.; MacMillan, S. N.; Wilson, J. J. Synthetic Methods for the Preparation of a Functional Analogue of Ru360, a Potent Inhibitor of Mitochondrial Calcium Uptake. Inorg. Chem. 2017, 56, 3123– 3126, DOI: 10.1021/acs.inorgchem.6b03108Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjsVCgtL8%253D&md5=bbd2e540a1cb9f4a4358d96d11817f55Synthetic Methods for the Preparation of a Functional Analogue of Ru360, a Potent Inhibitor of Mitochondrial Calcium UptakeNathan, Sarah R.; Pino, Nicholas W.; Arduino, Daniela M.; Perocchi, Fabiana; MacMillan, Samantha N.; Wilson, Justin J.Inorganic Chemistry (2017), 56 (6), 3123-3126CODEN: INOCAJ; ISSN:0020-1669. (American Chemical Society)The mixed-valent oxo-bridged Ru complex [(HCO2)(NH3)4Ru(μ-O)Ru(NH3)4(O2CH)]3+, known as Ru360, is a selective inhibitor of mitochondrial Ca uptake. Although this compd. is useful for studying the role of mitochondrial Ca in biol. processes, its widespread availability is limited because of challenges in purifn. and characterization. Here, the authors describe the authors' studies of three different synthetic methods for the prepn. of a functional analog of this valuable compd. This analog, isolated from the authors' procedures, exhibits potent mitochondrial Ca uptake inhibitory properties in permeabilized HeLa cells and in isolated mitochondria.
- 57Nathan, S. R.; Wilson, J. J. Synthesis and Evaluation of a Ruthenium-Based Mitochondrial Calcium Uptake Inhibitor. J. Visualized Exp. 2017, 128, e56527, DOI: 10.3791/56527Google ScholarThere is no corresponding record for this reference.
- 58Cao, C.; Wang, S.; Cui, T.; Su, X.-C.; Chou, J. J. Ion and Inhibitor Binding of the Double-Ring Ion Selectivity Filter of the Mitochondrial Calcium Uniporter. Proc. Natl. Acad. Sci. U. S. A. 2017, 114, E2846– E2851, DOI: 10.1073/pnas.1620316114Google Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXks1Smtr0%253D&md5=c3ec4bd6337c0234eab5aed8e354c2f4Ion and inhibitor binding of the double-ring ion selectivity filter of the mitochondrial calcium uniporterCao, Chan; Wang, Shuqing; Cui, Tanxing; Su, Xun-Cheng; Chou, James J.Proceedings of the National Academy of Sciences of the United States of America (2017), 114 (14), E2846-E2851CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The mitochondrial Ca2+ uniporter (MCU) is a holocomplex consisting of the Ca2+-conducting channel and several accessory and regulatory components. A previous electrophysiol. study found that the uniporter had high Ca2+ selectivity and conductance and this depended critically on the conserved amino acid sequence motif, DXXE (Asp-X-X-Glu) of MCU. A recent NMR structure of the MCU channel from Caenorhabditis elegans revealed that the DXXE formed 2 parallel carboxylate rings at the channel entrance that seem to serve as the ion selectivity filter, although direct ion interaction of this structural motif has not been addressed. Here, we used a paramagnetic probe, Mn2+, to investigate ion and inhibitor binding of this putative selectivity filter. The paramagnetic NMR data showed that mutants with a single carboxylate ring, NXXE (Asn-X-X-Glu) and DXXQ (Asp-X-X-Gln), each could bind Mn2+ specifically, whereas in the wild type the 2 rings bound Mn2+ cooperatively, resulting in ∼1000-fold higher apparent affinity. Ca2+ could specifically displace bound Mn2+ at the DXXE site in the channel. Furthermore, titrating the sample with the known channel inhibitor, ruthenium 360 (Ru360), could displace Mn2+ binding from the solvent-accessible Asp site but not the inner Glu site. The NMR titrn. data, together with structural anal. of the DXXE motif and mol. dynamics simulations, indicated that the double carboxylate rings at the apex of the MCU pore constitute the ion selectivity filter and that Ru360 directly blocks ion entry into the filter by binding to the outer carboxylate ring.
- 59Gushchin, A. L.; Laricheva, Y. A.; Abramov, P. A.; Sokolov, M. N.; Abramov, P. A. Synthesis and Electrochemical Properties of [RuIV2O(PhCN)4Cl6]. Inorg. Chem. Commun. 2018, 95, 1387– 7003, DOI: 10.1016/j.inoche.2018.07.033Google ScholarThere is no corresponding record for this reference.
- 60Urgiles, J.; Nathan, S. R.; MacMillan, S. N.; Wilson, J. J. Dinuclear Nitrido-Bridged Ruthenium Complexes Bearing Diimine Ligands. Dalton Trans. 2017, 46, 14256– 14263, DOI: 10.1039/C7DT03085AGoogle Scholar60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhs1alt7%252FN&md5=e404175cf715aa29108493160fb5edd4Dinuclear nitrido-bridged ruthenium complexes bearing diimine ligandsUrgiles, Julie; Nathan, Sarah R.; MacMillan, Samantha N.; Wilson, Justin J.Dalton Transactions (2017), 46 (41), 14256-14263CODEN: DTARAF; ISSN:1477-9226. (Royal Society of Chemistry)Reactions of K3[Ru2NCl8(H2O)2] with 2,2'-bipyridine (bpy), 4,4'-dimethyl-2,2'-bipyridine (dmbpy), and 4,4'-dimethoxy-2,2'-bipyridine (dmobpy) yielded the nitrido-bridged dinuclear complexes [Ru2N(L)2Cl5(DMF)] where L = bpy (1), dmbpy (2), and dmobpy (3). The crystal structures of these complexes reveal a linear Ru-N-Ru moiety with each Ru center bearing a bidentate diimine ligand. The complexes were further characterized by NMR, IR, and UV-visible spectroscopic methods and cyclic voltammetry. Because the compds. bear some structural similarities with the mitochondrial Ca uptake inhibitor Ru360, the ability of these complexes to act in this capacity was evaluated. 1-3 All fail to block mitochondrial Ca uptake, revealing new facets of the structure-activity relations for Ru-based mitochondrial Ca uptake inhibitors.
- 61Dunitz, J. D.; Orgel, L. E. Application of Molecular-Orbital Theory to Some Binuclear Coordination Compounds. J. Chem. Soc. 1953, 2594– 2596, DOI: 10.1039/jr9530002594Google ScholarThere is no corresponding record for this reference.
- 62Cleare, M. J.; Griffith, W. P. Binuclear Nitrido-Complexes of Ruthenium. Chem. Commun. 1968, 21, 1302, DOI: 10.1039/c19680001302Google ScholarThere is no corresponding record for this reference.
- 63Griffith, W. P.; McManus, N. T.; Skapski, A. C. X-Ray Crystal Structure of [Ru2N(ethylenediamine)5]Cl5· H2O; a Novel Complex Containing Both Nitrido and Ethylenediamine Bridges. J. Chem. Soc., Chem. Commun. 1984, 7, 434, DOI: 10.1039/c39840000434Google ScholarThere is no corresponding record for this reference.
- 64Ng, H.-Y.; Cheung, W.-M.; Kwan Huang, E.; Wong, K.-L.; Sung, H. H.-Y.; Williams, I. D.; Leung, W.-H. Ruthenium Chalcogenonitrosyl and Bridged Nitrido Complexes Containing Chelating Sulfur and Oxygen Ligands. Dalton Trans. 2015, 44, 18459– 18468, DOI: 10.1039/C5DT02513CGoogle Scholar64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFCqs7%252FO&md5=3ff794cd2a4f5027e01d33b5b91a0f09Ruthenium chalcogenonitrosyl and bridged nitrido complexes containing chelating sulfur and oxygen ligandsNg, Ho-Yuen; Cheung, Wai-Man; Kwan Huang, Enrique; Wong, Kang-Long; Sung, Herman H.-Y.; Williams, Ian D.; Leung, Wa-HungDalton Transactions (2015), 44 (42), 18459-18468CODEN: DTARAF; ISSN:1477-9226. (Royal Society of Chemistry)Ruthenium thio- and seleno-nitrosyl complexes contg. chelating S and O ligands were synthesized and their de-chalcogenation reactions were studied. The reaction of mer-[Ru(N)Cl3(AsPh3)2] with elemental S and Se in THF at reflux afforded the chalcogenonitrosyl complexes mer-[Ru(NX)Cl3(AsPh3)2] [X = S (1), Se (2)]. Treatment of 1 with KN(R2PS)2 afforded trans-[Ru(NS)Cl{N(R2PS)2}2] [R = Ph (3), Pri (4), But (5)]. Alternatively, the thionitrosyl complex 5 was obtained from [Bu4N][Ru(N)Cl4] and KN(But2PS)2, presumably via S atom transfer from [N(But2PS)2]- to the nitride. Reactions of 1 and 2 with NaLOEt (LOEt- = [Co(η5-C5H5){P(O)(LOEt)2}3]-) gave [Ru(NX)LOEtCl2] (X = S (8), Se (9)). Treatment of [Bu4N][Ru(N)Cl4] with KN(R2PS)2 produced RuIV-RuIV μ-nitrido complexes [Ru2(μ-N){N(R2PS)2}4Cl] [R = Ph (6), Pri (7)]. Reactions of 3 and 9 with PPh3 afforded 6 and [Ru(NPPh3)LOEtCl2], resp. The desulfurization of 5 with [Ni(cod)2] (cod = 1,5-cyclooctadiene) gave the mixed valance RuIII-RuIV μ-nitrido complex [Ru2(μ-N){N(But2PS)2}4] (10) that was oxidized by [Cp2Fe](PF6) to give the RuIV-RuIV complex [Ru2(μ-N){N(But2PS)2}4](PF6) ([10]PF6). The crystal structures of 1, 2, 3, 7, 9 and 10 were detd.
- 65Ciechanowicz, M.; Skapski, A. C. Crystal Structure of Potassium μ-Nitrido-Bis[Aquotetrachlororuthenate(IV)]. J. Chem. Soc. A 1971, 84, 1792– 1794, DOI: 10.1039/J19710001792Google ScholarThere is no corresponding record for this reference.
- 66Freshney, R. I. Culture of Animal Cells, 5th ed.; John Wiley & Sons, Inc.: Hoboken, NJ, 2005.Google ScholarThere is no corresponding record for this reference.
- 67Shanmughapriya, S.; Rajan, S.; Hoffman, N. E.; Zhang, X.; Guo, S.; Kolesar, J. E.; Hines, K. J.; Ragheb, J.; Jog, N. R.; Caricchio, R.; Baba, Y.; Zhou, Y.; Kaufman, B. A.; Cheung, J. Y.; Kurosaki, T.; Gill, D. L.; Madesh, M. Ca2+ Signals Regulate Mitochondrial Metabolism by Stimulating CREB-Mediated Expression of the Mitochondrial Ca2+ Uniporter Gene MCU. Sci. Signaling 2015, 8, ra23, DOI: 10.1126/scisignal.2005673Google ScholarThere is no corresponding record for this reference.
- 68Reers, M.; Smith, T. W.; Chen, L. B. J-aggregate formation of a carbocyanine as a quantitative fluorescent indicator of membrane potential. Biochemistry 1991, 30, 4480– 4486, DOI: 10.1021/bi00232a015Google Scholar68https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXitVart7o%253D&md5=2c2befb752f3baa12d4fc1255f925448J-aggregate formation of a carbocyanine as a quantitative fluorescent indicator of membrane potentialReers, Martin; Smith, Thomas W.; Chen, Lan BoBiochemistry (1991), 30 (18), 4480-6CODEN: BICHAW; ISSN:0006-2960.The spectral properties of a novel membrane potential sensitive probe (JC-1) were characterized in aq. buffers and in isolated cardiac mitochondria. JC-1 is a carbocyanine with a delocalized pos. charge. It formed under favorable conditions a concn.-dependent fluorescent nematic phase consisting of J-aggregates. When excited at 490 nm, the monomers exhibited an emission max. at 527 nm and J-aggregates at 590 nm. Increasing concns. of JC-1 above a certain concn. caused a linear rise in the J-aggregate fluorescence, while the monomer fluorescence remained const. The membrane potential of energized mitochondria (neg. inside) promoted a directional uptake of JC-1 into the matrix, also with subsequent formation of J-aggregates. The J-aggregate fluorescence was sensitive to transient membrane potential changes induced by ADP and to metabolic inhibitors of oxidative phosphorylation. The J-aggregate fluorescence was pH independent within the physiol. pH range of 7.15-8.0 and could be linearly calibrated with valinomycin-induced K+ diffusion potentials. The advantage of JC-1 over rhodamines and other carbocyanines is that its color altered reversibly from green to red with increasing membrane potentials. This can be exploited for imaging live mitochondria on the stage of a microscope.
- 69King, A. P.; Gellineau, H. A.; Ahn, J. E.; MacMillan, S. N.; Wilson, J. J. Bis(Thiosemicarbazone) Complexes of Cobalt(III). Synthesis, Characterization, and Anticancer Potential. Inorg. Chem. 2017, 56, 6609– 6623, DOI: 10.1021/acs.inorgchem.7b00710Google Scholar69https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXns1yku7o%253D&md5=d4eeafefb534c53f21a1ef80a7657172Bis(thiosemicarbazone) Complexes of Cobalt(III). Synthesis, Characterization, and Anticancer PotentialKing, A. Paden; Gellineau, Hendryck A.; Ahn, Jung-Eun; MacMillan, Samantha N.; Wilson, Justin J.Inorganic Chemistry (2017), 56 (11), 6609-6623CODEN: INOCAJ; ISSN:0020-1669. (American Chemical Society)Nine bis(thiosemicarbazone) (BTSC) cobalt(III) complexes of the general formula [Co(BTSC)(L)2]NO3 were synthesized, where BTSC = diacetyl bis(thiosemicarbazone) (ATS), pyruvaldehyde bis(thiosemicarbazone) (PTS), or glyoxal bis(thiosemicarbazone) (GTS) and L = ammonia, imidazole (Im), or benzylamine (BnA). These compds. were characterized by multinuclear NMR spectroscopy, mass spectrometry, cyclic voltammetry, and x-ray crystallog. Their stability in phosphate-buffered saline was investigated and is highly dependent on the nature of the axial ligand, L. These studies revealed that complex stability is primarily dictated by the axial ligand following the sequence NH3 > Im > BnA. The cellular uptake and cytotoxicity in cancer cells were also detd. Both the cellular uptake and cytotoxicity were significantly affected by the nature of the equatorial BTSC. Complexes of ATS were taken up much more effectively than those of PTS and GTS. The cytotoxicity of the complexes was correlated to that of the free ligand. Cell uptake and cytotoxicity were also detd. under hypoxic conditions. Only minor differences in the hypoxia activity and uptake were obsd. Treatment of the cancer cells with the copper-depleting agent tetrathiomolybdate decreased the cytotoxic potency of the complexes, indicating that they may operate via a copper-dependent mechanism. These results provide a structure-activity relation for this class of compds., which may be applied for the rational design of new cobalt(III) anticancer agents.
- 70Egger, A. E.; Rappel, C.; Jakupec, M. A.; Hartinger, C. G.; Heffeter, P.; Keppler, B. K. Development of an Experimental Protocol for Uptake Studies of Metal Compounds in Adherent Tumor Cells. J. Anal. At. Spectrom. 2009, 24, 51– 61, DOI: 10.1039/B810481FGoogle Scholar70https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsFWisr%252FK&md5=e1f3d2c5048697874728de374d85aa21Development of an experimental protocol for uptake studies of metal compounds in adherent tumor cellsEgger, Alexander E.; Rappel, Christina; Jakupec, Michael A.; Hartinger, Christian G.; Heffeter, Petra; Keppler, Bernhard K.Journal of Analytical Atomic Spectrometry (2009), 24 (1), 51-61CODEN: JASPE2; ISSN:0267-9477. (Royal Society of Chemistry)Cellular uptake is being widely investigated in the context of diverse biol. activities of metal compds. on the cellular level. However, the applied techniques differ considerably, and a validated methodol. is not at hand. Therefore, we have varied numerous aspects of sample prepn. of the human colon carcinoma cell line SW480 exposed in vitro to the tumor-inhibiting metal complexes cisplatin and indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (KP1019) prior to anal. with ICP-MS, and the results were found to be tremendously influenced by adsorption to the culture dishes. Adsorption to culture plates increases linearly with the concn. of KP1019, depends on the protein content of the medium, the duration of contact to protein-contg. medium prior to drug addn. and the hydrophilicity/lipophilicity of the compd. For varying degrees of cell confluence, adsorption of Ru hardly differs from cell-free expts. Desorption from the plates contributes to total Ru detected in dependence on the cell harvesting method. Desorption kinetics for lysis in HNO3 and tetramethylammonium hydroxide (TMAH) are comparable, but TMAH is a more potent desorbant. Sample storage conditions prior to anal. influence significantly the recovery of analyte. Protocols using cell lysis in the culture plate without proper corrections run the risk of producing artifacts resulting from metal adsorption/desorption to an extent comparable with the actual cellular content. However, exptl. protocols reported in the literature frequently do not contain information whether adsorption or blank correction were performed and should be regarded with caution, esp. if lysis was performed directly in the culture dishes.
- 71Komor, A. C.; Schneider, C. J.; Weidmann, A. G.; Barton, J. K. Cell-Selective Biological Activity of Rhodium Metalloinsertors Correlates with Subcellular Localization. J. Am. Chem. Soc. 2012, 134, 19223– 19233, DOI: 10.1021/ja3090687Google Scholar71https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhs1CksrrI&md5=ac877e082357f44187d45af67896047aCell-Selective Biological Activity of Rhodium Metalloinsertors Correlates with Subcellular LocalizationKomor, Alexis C.; Schneider, Curtis J.; Weidmann, Alyson G.; Barton, Jacqueline K.Journal of the American Chemical Society (2012), 134 (46), 19223-19233CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Deficiencies in the mismatch repair (MMR) pathway are assocd. with several types of cancers, as well as resistance to commonly used chemotherapeutics. Rhodium metalloinsertors have been found to bind DNA mismatches with high affinity and specificity in vitro, and also exhibit cell-selective cytotoxicity, targeting MMR-deficient cells over MMR-proficient cells. Ten distinct metalloinsertors with varying lipophilicities have been synthesized and their mismatch binding affinities and biol. activities detd. Although DNA photocleavage expts. demonstrate that their binding affinities are quite similar, their cell-selective antiproliferative and cytotoxic activities vary significantly. Inductively coupled plasma mass spectrometry (ICP-MS) expts. have uncovered a relation between the subcellular distribution of these metalloinsertors and their biol. activities. Specifically, we find that all of our metalloinsertors localize in the nucleus at sufficient concns. for binding to DNA mismatches. However, the metalloinsertors with high rhodium localization in the mitochondria show toxicity that is not selective for MMR-deficient cells, whereas metalloinsertors with less mitochondrial rhodium show activity that is highly selective for MMR-deficient vs. proficient cells. This work supports the notion that specific targeting of the metalloinsertors to nuclear DNA gives rise to their cell-selective cytotoxic and antiproliferative activities. The selectivity in cellular targeting depends upon binding to mismatches in genomic DNA.
- 72Boyle, K. M.; Barton, J. K. A Family of Rhodium Complexes with Selective Toxicity toward Mismatch Repair-Deficient Cancers. J. Am. Chem. Soc. 2018, 140, 5612– 5624, DOI: 10.1021/jacs.8b02271Google Scholar72https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXntFWit7k%253D&md5=019869099e85f1d376b51720aebcb7feA Family of Rhodium Complexes with Selective Toxicity toward Mismatch Repair-Deficient CancersBoyle, Kelsey M.; Barton, Jacqueline K.Journal of the American Chemical Society (2018), 140 (16), 5612-5624CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Rhodium metalloinsertors are a unique set of metal complexes that bind specifically to DNA base pair mismatches in vitro and kill mismatch repair (MMR)-deficient cells at lower concns. than their MMR-proficient counterparts. A family of metalloinsertors contg. rhodium-oxygen ligand coordination, termed "Rh-O" metalloinsertors, has been prepd. and shown to have a significant increase in both overall potency and selectivity toward MMR-deficient cells regardless of structural changes in the ancillary ligands. Here we describe DNA-binding and cellular studies with the second generation of Rh-O metalloinsertors in which an ancillary ligand is varied in both steric bulk and lipophilicity. These complexes, of the form [Rh(L)(chrysi)(PPO)]2+, all include the O-contg. PPO ligand (PPO = 2-(pyridine-2-yl)propan-2-ol) and the arom. inserting ligand chrysi (5,6-chrysene quinone diimine) but differ in the identity of their ancillary ligand L, where L is a phenanthroline or bipyridyl deriv. The Rh-O metalloinsertors in this family all show micromolar binding affinities for a 29-mer DNA hairpin contg. a single CC mismatch. The complexes display comparable lipophilic tendencies and pKa values of 8.1-9.1 for dissocn. of an imine proton on the chrysi ligand. In cellular proliferation and cytotoxicity assays with MMR-deficient cells (HCT116O) and MMR-proficient cells (HCT116N), the complexes contg. the phenanthroline-derived ligands show highly selective cytotoxic preference for the MMR-deficient cells at nanomolar concns. Using mass spectral analyses, it is shown that the complexes are taken into cells through a passive mechanism and exhibit low accumulation in mitochondria, an off-target organelle that, when targeted by parent metalloinsertors, can lead to nonselective cytotoxicity. Overall, these Rh-O metalloinsertors have distinct and improved behavior compared to previous generations of parent metalloinsertors, making them ideal candidates for further therapeutic assessment.
- 73Ahmad, K. A.; Iskandar, K. B.; Hirpara, J. L.; Clement, M. V.; Pervaiz, S. Hydrogen Peroxide-Mediated Cytosolic Acidification is a Signal for Mitochondrial Translocation of Bax during Drug-Induced Apoptosis of Tumor Cells. Cancer Res. 2004, 64, 7867– 7878, DOI: 10.1158/0008-5472.CAN-04-0648Google ScholarThere is no corresponding record for this reference.
- 74Dong, Z.; Shanmughapriya, S.; Tomar, D.; Siddiqui, N.; Lynch, S.; Nemani, N.; Breves, S. L.; Zhang, X.; Tripathi, A.; Palaniappan, P.; Riitano, M. F.; Worth, A. M.; Seelam, A.; Carvalho, E.; Subbiah, R.; Jaña, F.; Soboloff, J.; Peng, Y.; Cheung, J. Y.; Joseph, S. K.; Caplan, J.; Rajan, S.; Stathopulos, P. B.; Madesh, M. Mitochondrial Ca2+ Uniporter is a Mitochondrial Luminal Redox Sensor That Augments MCU Channel Activity. Mol. Cell 2017, 65, 1014– 1028, DOI: 10.1016/j.molcel.2017.01.032Google Scholar74https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjvVWqu74%253D&md5=31aae22ab2cd06621296a3b43a7898fdMitochondrial Ca2+ Uniporter Is a Mitochondrial Luminal Redox Sensor that Augments MCU Channel ActivityDong, Zhiwei; Shanmughapriya, Santhanam; Tomar, Dhanendra; Siddiqui, Naveed; Lynch, Solomon; Nemani, Neeharika; Breves, Sarah L.; Zhang, Xueqian; Tripathi, Aparna; Palaniappan, Palaniappan; Riitano, Massimo F.; Worth, Alison M.; Seelam, Ajay; Carvalho, Edmund; Subbiah, Ramasamy; Jana, Fabian; Soboloff, Jonathan; Peng, Yizhi; Cheung, Joseph Y.; Joseph, Suresh K.; Caplan, Jeffrey; Rajan, Sudarsan; Stathopulos, Peter B.; Madesh, MuniswamyMolecular Cell (2017), 65 (6), 1014-1028.e7CODEN: MOCEFL; ISSN:1097-2765. (Elsevier Inc.)Ca2+ dynamics and oxidative signaling are fundamental mechanisms for mitochondrial bioenergetics and cell function. The MCU complex is the major pathway by which these signals are integrated in mitochondria. Whether and how these coactive elements interact with MCU have not been established. As an approach toward understanding the regulation of MCU channel by oxidative milieu, we adapted inflammatory and hypoxia models. We identified the conserved cysteine 97 (Cys-97) to be the only reactive thiol in human MCU that undergoes S-glutathionylation. Furthermore, biochem., structural, and superresoln. imaging anal. revealed that MCU oxidn. promotes MCU higher order oligomer formation. Both oxidn. and mutation of MCU Cys-97 exhibited persistent MCU channel activity with higher [Ca2+]m uptake rate, elevated mROS, and enhanced [Ca2+]m overload-induced cell death. In contrast, these effects were largely independent of MCU interaction with its regulators. These findings reveal a distinct functional role for Cys-97 in ROS sensing and regulation of MCU activity.
- 75Zhao, Y.; Araki, S.; Wu, J.; Teramoto, T.; Chang, Y.-F.; Nakano, M.; Abdelfattah, A. S.; Fujiwara, M.; Ishihara, T.; Nagai, T.; Campbell, R. E. An Expanded Palette of Genetically Encoded Ca2+ Indicators. Science 2011, 333, 1888– 1891, DOI: 10.1126/science.1208592Google Scholar75https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXht1aitr%252FK&md5=192ab4166c581df59b11b6373f689847An Expanded Palette of Genetically Encoded Ca2+ IndicatorsZhao, Yongxin; Araki, Satoko; Wu, Jiahui; Teramoto, Takayuki; Chang, Yu-Fen; Nakano, Masahiro; Abdelfattah, Ahmed S.; Fujiwara, Manabi; Ishihara, Takeshi; Nagai, Takeharu; Campbell, Robert E.Science (Washington, DC, United States) (2011), 333 (6051), 1888-1891CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Engineered fluorescent protein (FP) chimeras that modulate their fluorescence in response to changes in calcium ion (Ca2+) concn. are powerful tools for visualizing intracellular signaling activity. However, despite a decade of availability, the palette of single FP-based Ca2+ indicators has remained limited to a single green hue. The authors have expanded this palette by developing blue, improved green, and red intensiometric indicators, as well as an emission ratiometric indicator with an 11,000% ratio change. This series enables improved single-color Ca2+ imaging in neurons and transgenic Caenorhabditis elegans. In HeLa cells, Ca2+ was imaged in three subcellular compartments, and, in conjunction with a cyan FP-yellow FP-based indicator, Ca2+ and ATP were simultaneously imaged. This palette of indicators paints the way to a colorful new era of Ca2+ imaging.
- 76Nakai, J.; Ohkura, M.; Imoto, K. A High Signal-to-Noise Ca2+ Probe Composed of a Single Green Fluorescent Protein. Nat. Biotechnol. 2001, 19, 137– 141, DOI: 10.1038/84397Google Scholar76https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXhtVaitrw%253D&md5=7d7985a03973490fbf3c5e509ea2b94cA high signal-to-noise Ca2+ probe composed of a single green fluorescent proteinNakai, Junichi; Ohkura, Masamichi; Imoto, KeijiNature Biotechnology (2001), 19 (2), 137-141CODEN: NABIF9; ISSN:1087-0156. (Nature America Inc.)Recently, several groups have developed green fluorescent protein (GFP)-based Ca2+ probes. When applied in cells, however, these probes are difficult to use because of a low signal-to-noise ratio. Here we report the development of a high-affinity Ca2+ probe composed of a single GFP (named G-CaMP). G-CaMP showed an apparent Kd for Ca2+ of 235 μM. Assocn. kinetics of Ca2+ binding were faster at higher Ca2+ concns., with time consts. decreasing from 230 ms at 0.2 μM Ca2+ to 2.5 ms at 1 μM Ca2+. Dissocn. kinetics (τ -200 ms) are independent of Ca2+ concns. In HEK-293 cells and mouse myotubes expressing G-CaMP, large fluorescent changes were obsd. in response to application of drugs or elec. stimulations. G-CaMP will be a useful tool for visualizing intracellular Ca2+ in living cells. Mutational anal., together with previous structural information, suggests the residues that may alter the fluorescence of GFP.
- 77Chiesi, M.; Schwaller, R.; Eichenberger, K. Structural Dependency of the Inhibitory Action of Benzodiazepines and Related Compounds on the Mitochondrial Na+-Ca2+ Exchanger. Biochem. Pharmacol. 1988, 37, 4399– 4403, DOI: 10.1016/0006-2952(88)90623-5Google ScholarThere is no corresponding record for this reference.
- 78Luongo, T. S.; Lambert, J. P.; Gross, P.; Nwokedi, M.; Lombardi, A. A.; Shanmughapriya, S.; Carpenter, A. C.; Kolmetzky, D.; Gao, E.; van Berlo, J. H.; Tsai, E. J.; Molkentin, J. D.; Chen, X.; Madesh, M.; Houser, S. R.; Elrod, J. W. The Mitochondrial Na+/Ca2+ Exchanger is Essential for Ca2+ Homeostasis and Viability. Nature 2017, 545, 93– 97, DOI: 10.1038/nature22082Google Scholar78https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXmslehs74%253D&md5=0a3311e8284f11453ef09956541e9a0fThe mitochondrial Na+/Ca2+ exchanger is essential for Ca2+ homeostasis and viabilityLuongo, Timothy S.; Lambert, Jonathan P.; Gross, Polina; Nwokedi, Mary; Lombardi, Alyssa A.; Shanmughapriya, Santhanam; Carpenter, April C.; Kolmetzky, Devin; Gao, Erhe; van Berlo, Jop H.; Tsai, Emily J.; Molkentin, Jeffery D.; Chen, Xiongwen; Madesh, Muniswamy; Houser, Steven R.; Elrod, John W.Nature (London, United Kingdom) (2017), 545 (7652), 93-97CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Mitochondrial calcium (mCa2+) has a central role in both metabolic regulation and cell death signaling, however its role in homeostatic function and disease is controversial. Slc8b1 encodes the mitochondrial Na+/Ca2+ exchanger (NCLX), which is proposed to be the primary mechanism for mCa2+ extrusion in excitable cells. Here we show that tamoxifen-induced deletion of Slc8b1 in adult mouse hearts causes sudden death, with less than 13% of affected mice surviving after 14 days. Lethality correlated with severe myocardial dysfunction and fulminant heart failure. Mechanistically, cardiac pathol. was attributed to mCa2+ overload driving increased generation of superoxide and necrotic cell death, which was rescued by genetic inhibition of mitochondrial permeability transition pore activation. Corroborating these findings, overexpression of NCLX in the mouse heart by conditional transgenesis had the beneficial effect of augmenting mCa2+ clearance, preventing permeability transition and protecting against ischemia-induced cardiomyocyte necrosis and heart failure. These results demonstrate the essential nature of mCa2+ efflux in cellular function and suggest that augmenting mCa2+ efflux may be a viable therapeutic strategy in disease.
- 79Storey, N. M.; Lambert, D. G. Mitochondrial Pharmacology Turns Its Sights on the Ca2+ Uniporter. Cell Death Discov. 2017, 3, 17064, DOI: 10.1038/cddiscovery.2017.64Google Scholar79https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1M%252FjtVGrtw%253D%253D&md5=412a9c4b52c621ffff75e034314a4cf7Mitochondrial pharmacology turns its sights on the Ca(2+) uniporterStorey Nina M; Lambert David GCell death discovery (2017), 3 (), 17064 ISSN:2058-7716.There is no expanded citation for this reference.
- 80Sebag, S. C.; Koval, O. M.; Paschke, J. D.; Winters, C. J.; Comellas, A. P.; Grumbach, I. M. Inhibition of the Mitochondrial Calcium Uniporter Prevents IL-13 and Allergen-Mediated Airway Epithelial Apoptosis and Loss of Barrier Function. Exp. Cell Res. 2018, 362, 400– 411, DOI: 10.1016/j.yexcr.2017.12.003Google Scholar80https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhvF2js73K&md5=4f3e72238fc7003961f65a2cf19e7e3bInhibition of the mitochondrial calcium uniporter prevents IL-13 and allergen-mediated airway epithelial apoptosis and loss of barrier functionSebag, Sara C.; Koval, Olha M.; Paschke, John D.; Winters, Christopher J.; Comellas, Alejandro P.; Grumbach, Isabella M.Experimental Cell Research (2018), 362 (2), 400-411CODEN: ECREAL; ISSN:0014-4827. (Elsevier B.V.)Mitochondria are increasingly recognized as key mediators of acute cellular stress responses in asthma. However, the distinct roles of regulators of mitochondrial physiol. on allergic asthma phenotypes are currently unknown. The mitochondrial Ca2+ uniporter (MCU) resides in the inner mitochondrial membrane and controls mitochondrial Ca2+ uptake into the mitochondrial matrix. To understand the function of MCU in models of allergic asthma, in vitro and in vivo studies were performed using models of functional deficiency or knockout of MCU. In primary human respiratory epithelial cells, MCU inhibition abrogated mitochondrial Ca2+ uptake and reactive oxygen species (ROS) prodn., preserved the mitochondrial membrane potential and protected from apoptosis in response to the pleiotropic Th2 cytokine IL-13. Consequently, epithelial barrier function was maintained with MCU inhibition. Similarly, the endothelial barrier was preserved in respiratory epithelium isolated from MCU-/- mice after exposure to IL-13. In the ovalbumin-model of allergic airway disease, MCU deficiency resulted in decreased apoptosis within the large airway epithelial cells. Concordantly, expression of the tight junction protein ZO-1 was preserved, indicative of maintenance of epithelial barrier function. These data implicate mitochondrial Ca2+ uptake through MCU as a key controller of epithelial cell viability in acute allergic asthma.
- 81Xie, N.; Wu, C.; Wang, C.; Cheng, X.; Zhang, L.; Zhang, H.; Lian, Y. Inhibition of the Mitochondrial Calcium Uniporter Inhibits Aβ-Induced Apoptosis by Reducing Reactive Oxygen Species-Mediated Endoplasmic Reticulum Stress in Cultured Microglia. Brain Res. 2017, 1676, 100– 106, DOI: 10.1016/j.brainres.2017.08.035Google Scholar81https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsFGht77E&md5=bcfa374163b91a61f96a1e42056ea510Inhibition of the mitochondrial calcium uniporter inhibits Aβ-induced apoptosis by reducing reactive oxygen species-mediated endoplasmic reticulum stress in cultured microgliaXie, Nanchang; Wu, Chuanjie; Wang, Cui; Cheng, Xuan; Zhang, Lu; Zhang, Haifeng; Lian, YajunBrain Research (2017), 1676 (), 100-106CODEN: BRREAP; ISSN:0006-8993. (Elsevier B.V.)Amyloid-beta (Aβ) has been shown to induce microglial apoptosis, which is itself sensitive to disturbed mitochondrial calcium (Ca2+) homeostasis. The mitochondrial calcium uniporter (MCU) plays an important regulatory role in mitochondrial Ca2+ homeostasis, but its role in Aβ-induced microglia apoptosis is unknown. In this study, we found increased mitochondrial Ca2+ concn. in Aβ-treated primary microglia and BV-2 cells; also, the MCU inhibitor Ru360 significantly attenuated Aβ-induced microglial apoptosis, whereas the MCU activator spermine augmented it. In addn., Ru360 significantly attenuated Aβ-induced mitochondrial reactive oxygen species (ROS) prodn., as well as endoplasmic reticulum (ER) stress characterized by glucose-regulated protein 78 (GRP78) and C/-EBP homologous protein (CHOP) expression. Spermine, however, exerted the opposite effects on mitochondrial ROS prodn. and ER stress. We also found that mitochondria-targeted antioxidant (Mito-TEMPO) treatment decreased GRP78 and CHOP expression in Aβ-treated microglia. Moreover, blocking endogenous CHOP expression using a CHOP small interfering RNA (siRNA) attenuated Aβ-induced cell death. Altogether, our data suggested that (1) inhibition of MCU exerts a neuroprotective effect on Aβ-induced microglia apoptosis, and (2) that the underlying mechanism may be related to reducing mitochondrial ROS-mediated ER stress.
- 82Alessio, E. Thirty Years of the Drug Candidate NAMI-A and the Myths in the Field of Ruthenium Anticancer Compounds: A Personal Perspective. Eur. J. Inorg. Chem. 2017, 2017, 1549– 1560, DOI: 10.1002/ejic.201600986Google Scholar82https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFKru7bK&md5=ca48413b7dd02a63edf5049a9a5c7377Thirty Years of the Drug Candidate NAMI-A and the Myths in the Field of Ruthenium Anticancer Compounds: A Personal PerspectiveAlessio, EnzoEuropean Journal of Inorganic Chemistry (2017), 2017 (12), 1549-1560CODEN: EJICFO; ISSN:1434-1948. (Wiley-VCH Verlag GmbH & Co. KGaA)As anticipated in the title, this contribution is basically divided into two, strictly connected, parts. The first is a personal overview of the ruthenium drug candidate NAMI-A, almost 30 years after its synthesis and the discovery of its unprecedented antimetastatic properties in animal models at nontoxic dosages. The sections relating to the chem. and biol. behavior of the complex, and the hypotheses on its mechanism(s) of action, are kept to a min., whereas more space is devoted to discussion of the results of the clin. investigations. The second part deals in detail with a no. of undemonstrated misconceptions (or myths) that, over the years, have thrived around NAMI-A and other ruthenium drug candidates, thus neg. affecting the whole field of Ru anticancer drugs.
- 83Wachter, E.; Heidary, D. K.; Howerton, B. S.; Parkin, S.; Glazer, E. C. Light-Activated Ruthenium Complexes Photobind DNA and are Cytotoxic in the Photodynamic Therapy Window. Chem. Commun. 2012, 48, 9649– 9651, DOI: 10.1039/c2cc33359gGoogle Scholar83https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xht12qsrjJ&md5=dcab2483021e3f4d03850e162ce23167Light-activated ruthenium complexes photobind DNA and are cytotoxic in the photodynamic therapy windowWachter, Erin; Heidary, David K.; Howerton, Brock S.; Parkin, Sean; Glazer, Edith C.Chemical Communications (Cambridge, United Kingdom) (2012), 48 (77), 9649-9651CODEN: CHCOFS; ISSN:1359-7345. (Royal Society of Chemistry)Incorporation of biquinoline ligands into Ru(ii) polypyridyl complexes produces light-activated systems that eject a ligand and photobind DNA after irradn. with visible and near-IR light. Structural anal. shows that distortion facilitates the photochem., and gel shift and cytotoxicity studies prove the compds. act as anti-cancer photodynamic therapy (PDT) agents in the tissue penetrant region.
- 84Han Ang, W.; Dyson, P. J. Classical and Non-Classical Ruthenium-Based Anticancer Drugs: Towards Targeted Chemotherapy. Eur. J. Inorg. Chem. 2006, 2006, 4003– 4018, DOI: 10.1002/ejic.200600723Google ScholarThere is no corresponding record for this reference.
- 85Süss-Fink, G. Areneruthenium Complexes as Anticancer Agents. Dalton Trans. 2010, 39, 1673– 1688, DOI: 10.1039/B916860PGoogle Scholar85https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtlCqtr4%253D&md5=2758ad6804f77d33612401411fe7c116Arene ruthenium complexes as anticancer agentsSuess-Fink, GeorgDalton Transactions (2010), 39 (7), 1673-1688CODEN: DTARAF; ISSN:1477-9226. (Royal Society of Chemistry)A review. Neutral or cationic arene ruthenium complexes providing both hydrophilic as well as hydrophobic properties due to the robustness of the ruthenium-arene unit hold a high potential for the development of metal-based anticancer drugs. Mononuclear arene ruthenium complexes contg. P- or N-donor ligands or N,N-, N,O- or O,O-chelating ligands, dinuclear arene ruthenium systems with adjustable org. linkers, trinuclear arene ruthenium clusters contg. an oxo cap, tetranuclear arene ruthenium porphyrin derivs. that are photoactive, as well as hexanuclear ruthenium cages that are either empty or filled with other mols. have been shown to be active against a variety of cancer cells.
- 86Wang, F.; Chen, H.; Parsons, S.; Oswald, I. D. H.; Davidson, J. E.; Sadler, P. J. Kinetics of Aquation and Anation of Ruthenium(II) Arene Anticancer Complexes, Acidity and X-Ray Structures of Aqua Adducts. Chem. - Eur. J. 2003, 9, 5810– 5820, DOI: 10.1002/chem.200304724Google Scholar86https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhvVGk&md5=7d156972deaa9b35381da9013f931e2dKinetics of aquation and anation of ruthenium(II) arene anticancer complexes, acidity and X-ray structures of aqua adductsWang, Fuyi; Chen, Haimei; Parsons, Simon; Oswald, Iain D. H.; Davidson, James E.; Sadler, Peter J.Chemistry - A European Journal (2003), 9 (23), 5810-5820CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)The aqua adducts of the anticancer complexes [(η6-X)Ru(en)Cl] [PF6] (X = biphenyl (Bip) 1, X = 5,8,9,10-tetrahydroanthracene (THA) 2, X = 9,10-dihydroanthracene (DHA) 3; en = ethylenediamime) were sepd. by HPLC and characterized by mass spectrometry as the products of hydrolysis in H2O. The x-ray structures of the aqua complexes [(η6-X)Ru(en)Y] [PF6]n, X = Bip, Y = 0.5H2O/0.5OH, n = 1.5 (4), X = THA, Y = 0.5H2O/0.5OH, n = 1.5 (5A), X = THA, Y = H2O, n = 2 (5B), and X = DHA, Y = H2O, n = 2(6), are reported. In complex 4 there is a large propeller twist of 45° of the pendant Ph ring with respect to the coordinated Ph ring. Although the THA ligand in 5A and 5B is relatively flat, the DHA ring system in 6 is markedly bent (hinge bend ∼35°) as in the chloro complex 3 (41°). The rates of aquation of 1-3 detd. by UV/visible spectroscopy at various ionic strengths and temps. (1.23-2.59 × 10-3s-1 at 298 K, I = 0.1M) are >20× faster than that of cisplatin. The reverse, anation reactions were very rapid on addn. of 100 mM NaCl (a similar concn. to that in blood plasma). The aquation and anation reactions were about two times faster for the DHA and THA complexes compared to the biphenyl complex. The hydrolysis reactions appear to occur by an associative pathway. The pKa values of the aqua adducts were detd. by 1H NMR spectroscopy as 7.71 for 4, 8.01 for 5 and 7.89 for 6. At physiol.-relevant concns. (0.5-5 μM) and temp. (310 K), the complexes will exist in blood plasma as >89% chloro complex, whereas in the cell nucleus significant amts. (45-65%) of the more reactive aqua adducts would be formed together with smaller amts. of the hydroxo complexes (9-25%, pH 7.4, [Cl-] = 4 mM).
- 87Hartinger, C. G.; Jakupec, M. A.; Zorbas-Seifried, S.; Groessl, M.; Egger, A.; Berger, W.; Zorbas, H.; Dyson, P. J.; Keppler, B. K. KP1019, A New Redox-Active Anticancer Agent - Preclinical Development and Results of a Clinical Phase I Study in Tumor Patients. Chem. Biodiversity 2008, 5, 2140– 2155, DOI: 10.1002/cbdv.200890195Google Scholar87https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsVWktLvO&md5=2d209d437e1c5fefb72827d3eade21d7KP1019, a new redox-active anticancer agent - preclinical development and results of a clinical phase I study in tumor patientsHartinger, Christian G.; Jakupec, Michael A.; Zorbas-Seifried, Stefanie; Groessl, Michael; Egger, Alexander; Berger, Walter; Zorbas, Haralabos; Dyson, Paul J.; Keppler, Bernhard K.Chemistry & Biodiversity (2008), 5 (10), 2140-2155CODEN: CBHIAM; ISSN:1612-1872. (Verlag Helvetica Chimica Acta)A review. The promising drug candidate indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (KP1019) is the second Ru-based anticancer agent to enter clin. trials. In this review, which is an update of a paper from 2006, the exptl. evidence for the proposed mode of action of this coordination compd. is discussed, including transport into the cell via the transferrin cycle and activation by redn. The results of the early clin. development of KP1019 are summarized in which 5 out of 6 evaluated patients experienced disease stabilization with no severe side effects.
- 88Peacock, A. F. A.; Habtemariam, A.; Fernández, R.; Walland, V.; Fabbiani, F. P. A.; Parsons, S.; Aird, R. E.; Jodrell, D. I.; Sadler, P. J. Tuning the Reactivity of Osmium(II) and Ruthenium(II) Arene Complexes under Physiological Conditions. J. Am. Chem. Soc. 2006, 128, 1739– 1748, DOI: 10.1021/ja055886rGoogle Scholar88https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XjvV2ktQ%253D%253D&md5=f93f88f8995cbf36730b9c69a816dfcaTuning the Reactivity of Osmium(II) and Ruthenium(II) Arene Complexes under Physiological ConditionsPeacock, Anna F. A.; Habtemariam, Abraha; Fernandez, Rafael; Walland, Victoria; Fabbiani, Francesca P. A.; Parsons, Simon; Aird, Rhona E.; Jodrell, Duncan I.; Sadler, Peter J.Journal of the American Chemical Society (2006), 128 (5), 1739-1748CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The OsII arene ethylenediamine (en) complexes [(η6-biphenyl)Os(en)Cl][Z], Z = BPh4 (4) and BF4 (5), are inactive toward A2780 ovarian cancer cells despite 4 being isostructural with an active RuII analog, 4R. Hydrolysis of 5 occurred 40 times more slowly than 4R. The aqua adduct 5A has a low pKa (6.3) compared to that of [(η6-biphenyl)Ru(en)(OH2)]2+ (7.7) and is therefore largely in the hydroxo form at physiol. pH. The rate and extent of reaction of 5 with 9-ethylguanine were also less than those of 4R. The authors replaced the neutral en ligand by anionic acetylacetonate (acac). The complexes [(η6-arene)Os(acac)Cl], arene = biphenyl (6), benzene (7), and p-cymene (8), adopt piano-stool structures similar to those of the RuII analogs and form weak dimers through intermol. (arene)C-H···O(acac) H-bonds. Remarkably, these OsII acac complexes undergo rapid hydrolysis to produce not only the aqua adduct, [(η6-arene)Os(acac)(OH2)]+, but also the hydroxo-bridged dimer, [(η6-arene)Os(μ2-OH)3Os(η6-arene)]+. The pKa values for the aqua adducts 6A, 7A, and 8A (7.1, 7.3, and 7.6, resp.) are lower than that for [(η6-p-cymene)Ru(acac)(OH2)]+ (9.4). Complex 8A rapidly forms adducts with 9-ethylguanine and adenosine, but not with cytidine or thymidine. Despite their reactivity toward nucleobases, complexes 6-8 were inactive toward A549 lung cancer cells. This is attributable to rapid hydrolysis and formation of unreactive hydroxo-bridged dimers which, surprisingly, were the only species present in aq. soln. at biol. relevant concns. Hence, the choice of chelating ligand in OsII (and RuII) arene complexes can have a dramatic effect on hydrolysis behavior and nucleobase binding and provides a means of tuning the reactivity and the potential for discovery of anticancer complexes.
- 89Mühlgassner, G.; Bartel, C.; Schmid, W. F.; Jakupec, M. A.; Arion, V. B.; Keppler, B. K. Biological Activity of Ruthenium and Osmium Arene Complexes with Modified Paullones in Human Cancer Cells. J. Inorg. Biochem. 2012, 116, 180– 187, DOI: 10.1016/j.jinorgbio.2012.06.003Google Scholar89https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsFKnsbzN&md5=331ebaa32b9ab9aa12ad2bfe5a1051e7Biological activity of ruthenium and osmium arene complexes with modified paullones in human cancer cellsMuehlgassner, Gerhard; Bartel, Caroline; Schmid, Wolfgang F.; Jakupec, Michael A.; Arion, Vladimir B.; Keppler, Bernhard K.Journal of Inorganic Biochemistry (2012), 116 (), 180-187CODEN: JIBIDJ; ISSN:0162-0134. (Elsevier)In an attempt to combine the ability of indolobenzazepines (paullones) to inhibit cyclin-dependent kinases (Cdks) and that of platinum-group metal ions to interact with proteins and DNA, ruthenium(II) and osmium(II) arene complexes with paullones were prepd., expecting synergies and an increase of soly. of paullones. Complexes with the general formula [MIICl(η6-p-cymene)L]Cl, where M = Ru (1, 3) or Os (2, 4), and L = L1 (1, 2) or L2 (3, 4), L1 = N-(9-bromo-7,12-dihydroindolo[3,2-d][1]-benzazepin-6(5H)-yliden-N'-(2-hydroxybenzylidene)azine and L2 = N-(9-bromo-7,12-dihydroindolo[3,2-d][1]benzazepin-6-yl)-N'-[3-hydroxy-5-(hydroxymethyl)-2-methylpyridin-4-yl-methylene]azinium chloride (L2*HCl), were now investigated regarding cytotoxicity and accumulation in cancer cells, impact on the cell cycle, capacity of inhibiting DNA synthesis and inducing apoptosis as well as their ability to inhibit Cdk activity. The MTT (3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide) assay yielded IC50 values in the nanomolar to low micromolar range. In accordance with cytotoxicity data, the BrdU assay showed that 1 is the most and 4 the least effective of these compds. regarding inhibition of DNA synthesis. Effects on the cell cycle are minor, although concn.-dependent inhibition of Cdk2/cyclin E activity was obsd. in cell-free expts. Induction of apoptosis is most pronounced for complex 1, accompanied by a low fraction of necrotic cells, as obsd. by annexin V-fluorescein isothiocyanate/propidium iodide staining and flow cytometric anal.
- 90Lameijer, L. N.; Ernst, D.; Hopkins, S. L.; Meijer, M. S.; Askes, S. H. C.; Le Dévédec, S. E.; Bonnet, S. A Red-Light-Activated Ruthenium-Caged NAMPT Inhibitor Remains Phototoxic in Hypoxic Cancer Cells. Angew. Chem., Int. Ed. 2017, 56, 11549– 11553, DOI: 10.1002/anie.201703890Google Scholar90https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtlSmu77L&md5=b5ab704916712872f33ed4d67838e380A Red-Light-Activated Ruthenium-Caged NAMPT Inhibitor Remains Phototoxic in Hypoxic Cancer CellsLameijer, Lucien N.; Ernst, Daniel; Hopkins, Samantha L.; Meijer, Michael S.; Askes, Sven H. C.; Le Devedec, Sylvia E.; Bonnet, SylvestreAngewandte Chemie, International Edition (2017), 56 (38), 11549-11553CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)We describe two water-sol. ruthenium complexes, [1]Cl2 and [2]Cl2, that photodissociate to release a cytotoxic nicotinamide phosphoribosyltransferase (NAMPT) inhibitor with a low dose (21 J cm-2) of red light in an oxygen-independent manner. Using a specific NAMPT activity assay, up to an 18-fold increase in inhibition potency was measured upon red-light activation of [2]Cl2, while [1]Cl2 was thermally unstable. For the first time, the dark and red-light-induced cytotoxicity of these photocaged compds. could be tested under hypoxia (1 % O2). In skin (A431) and lung (A549) cancer cells, a 3- to 4-fold increase in cytotoxicity was found upon red-light irradn. for [2]Cl2, whether the cells were cultured and irradiated with 1 % or 21 % O2. These results demonstrate the potential of photoactivated chemotherapy for hypoxic cancer cells, in which classical photodynamic therapy, which relies on oxygen activation, is poorly efficient.
- 91Dutta, B.; Scolaro, C.; Scopelliti, R.; Dyson, P. J.; Severin, K. Importance of the π-Ligand: Remarkable Effect of the Cyclopentadienyl Ring on the Cytotoxicity of Ruthenium PTA Compounds. Organometallics 2008, 27, 1355– 1357, DOI: 10.1021/om800025aGoogle Scholar91https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXis1ektLg%253D&md5=ffaf9ea19304cb2698d4ba3aeeefa6c8Importance of the π-Ligand: Remarkable Effect of the Cyclopentadienyl Ring on the Cytotoxicity of Ruthenium PTA CompoundsDutta, Barnali; Scolaro, Claudine; Scopelliti, Rosario; Dyson, Paul J.; Severin, KayOrganometallics (2008), 27 (7), 1355-1357CODEN: ORGND7; ISSN:0276-7333. (American Chemical Society)The water-sol. complexes [(Cp'OR)RuCl(PTA)2] (Cp'OR = η5-1-alkoxy-2,4-di-tert-butyl-3-neopentylcyclopentadienyl; R = Me, Et; PTA = 1,3,5-triaza-7-phosphaadamantane) are considerably more cytotoxic (∼2 orders of magnitude) than the cyclopentadienyl analog [CpRuCl(PTA)2] (i.e., IC50 = 4-10 vs. >1000 μM, depending on the cell line). The structure of [(Cp'OMe)RuCl(PTA)2] is reported, together with that of the precursor [(Cp'OEt)Ru(μ-Cl)]2.
- 92Allardyce, C. S.; Dyson, P. J. Metal-Based Drugs that Break the Rules. Dalton Trans. 2016, 45, 3201– 3209, DOI: 10.1039/C5DT03919CGoogle Scholar92https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFymurs%253D&md5=75a2d446dd76210a55096041a529ec31Metal-based drugs that break the rulesAllardyce, Claire S.; Dyson, Paul J.Dalton Transactions (2016), 45 (8), 3201-3209CODEN: DTARAF; ISSN:1477-9226. (Royal Society of Chemistry)Cisplatin and other platinum compds. have had a huge impact in the treatment of cancers and are applied in the majority of anticancer chemotherapeutic regimens. The success of these compds. has biased the approaches used to discover new metal-based anticancer drugs. In this perspective we highlight compds. that are apparently incompatible with the more classical (platinum-derived) concepts employed in the development of metal-based anticancer drugs, with respect to both compd. design and the approaches used to validate their utility. Possible design approaches for the future are also suggested.
- 93Kilpin, K. J.; Dyson, P. J. Enzyme Inhibition by Metal Complexes: Concepts, Strategies and Applications. Chem. Sci. 2013, 4, 1410– 1419, DOI: 10.1039/c3sc22349cGoogle Scholar93https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXjtlyms7c%253D&md5=4619c55c66ea098947112400a47a4c65Enzyme inhibition by metal complexes: concepts, strategies and applicationsKilpin, Kelly J.; Dyson, Paul J.Chemical Science (2013), 4 (4), 1410-1419CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)A review. Metal complexes are increasingly being used to inhibit enzymes. The reasons for this increased interest arise from the special features that metal complexes offer, e.g. the facile construction of 3D architectures that tightly fill enzyme active sites increasing selectivity and the possibility of facile coordination to protein residues that enhances enzyme inhibition. In this review we classify the main modes of enzyme inhibition by metal-based complexes and correlate the enzyme inhibition activity to macroscopic properties such as anticancer activity.
- 94Barry, N. P. E.; Sadler, P. J. Exploration of the Medical Periodic Table: Towards New Targets. Chem. Commun. 2013, 49, 5106– 5131, DOI: 10.1039/c3cc41143eGoogle Scholar94https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXnsFaru7c%253D&md5=b91db5563e59a43e7f9d05860a03b3b0Exploration of the medical periodic table: towards new targetsBarry, Nicolas P. E.; Sadler, Peter J.Chemical Communications (Cambridge, United Kingdom) (2013), 49 (45), 5106-5131CODEN: CHCOFS; ISSN:1359-7345. (Royal Society of Chemistry)A review. Metallodrugs offer potential for unique mechanisms of drug action based on the choice of the metal, its oxidn. state, the types and no. of coordinated ligands and the coordination geometry. We discuss recent progress in identifying new target sites and elucidating the mechanisms of action of anti-cancer, anti-bacterial, anti-viral, anti-parasitic, anti-inflammatory, and anti-neurodegenerative agents, as well as in the design of metal-based diagnostic agents. Progress in identifying and defining target sites has been accelerated recently by advances in proteomics, genomics and metal speciation anal. Examples of metal compds. and chelating agents (enzyme inhibitors) currently in clin. use, clin. trials or preclin. development are highlighted.
Cited By
This article is cited by 118 publications.
- Peng Wang, Xian Zheng, Ronghuan Du, Jinghan Xu, Jing Li, Huaqi Zhang, Xi Liang, Hui Liang. Astaxanthin Protects against Alcoholic Liver Injury via Regulating Mitochondrial Redox Balance and Calcium Homeostasis. Journal of Agricultural and Food Chemistry 2023, 71
(49)
, 19531-19550. https://doi.org/10.1021/acs.jafc.3c05529
- Nicholas P. Bigham, Justin J. Wilson. Metal Coordination Complexes as Therapeutic Agents for Ischemia-Reperfusion Injury. Journal of the American Chemical Society 2023, 145
(17)
, 9389-9409. https://doi.org/10.1021/jacs.3c01984
- Nicholas P. Bigham, Zhouyang Huang, Jesse Spivey, Joshua J. Woods, Samantha N. MacMillan, Justin J. Wilson. Carboxylate-Capped Analogues of Ru265 Are MCU Inhibitor Prodrugs. Inorganic Chemistry 2022, 61
(43)
, 17299-17312. https://doi.org/10.1021/acs.inorgchem.2c02930
- Jakub Cervinka, Alberto Gobbo, Lorenzo Biancalana, Lenka Markova, Vojtech Novohradsky, Massimo Guelfi, Stefano Zacchini, Jana Kasparkova, Viktor Brabec, Fabio Marchetti. Ruthenium(II)–Tris-pyrazolylmethane Complexes Inhibit Cancer Cell Growth by Disrupting Mitochondrial Calcium Homeostasis. Journal of Medicinal Chemistry 2022, 65
(15)
, 10567-10587. https://doi.org/10.1021/acs.jmedchem.2c00722
- Hiromitsu Sasaki, Ichiro Nakagawa, Takanori Furuta, Shohei Yokoyama, Yudai Morisaki, Yasuhiko Saito, Hiroyuki Nakase. Mitochondrial Calcium Uniporter (MCU) is Involved in an Ischemic Postconditioning Effect Against Ischemic Reperfusion Brain Injury in Mice. Cellular and Molecular Neurobiology 2024, 44
(1)
https://doi.org/10.1007/s10571-024-01464-7
- Esther Densu Agyapong, Gaia Pedriali, Daniela Ramaccini, Esmaa Bouhamida, Elena Tremoli, Carlotta Giorgi, Paolo Pinton, Giampaolo Morciano. Calcium signaling from sarcoplasmic reticulum and mitochondria contact sites in acute myocardial infarction. Journal of Translational Medicine 2024, 22
(1)
https://doi.org/10.1186/s12967-024-05240-5
- João L. Alves, Patrícia M. Reis, Rosa M. Quinta-Ferreira, M. Emília Quinta-Ferreira, Carlos M. Matias. Changes in reactive oxygen species and autofluorescence under hypoxia at the hippocampal CA3 area: Role of calcium and zinc influxes. Neurochemistry International 2024, 180 , 105882. https://doi.org/10.1016/j.neuint.2024.105882
- Anagha Inguva Sheth, Mark J. Althoff, Hunter Tolison, Krysta Engel, Maria L. Amaya, Anna E. Krug, Tracy N. Young, Mohammad Minhajuddin, Shanshan Pei, Sweta B. Patel, Amanda Winters, Regan Miller, Ian T. Shelton, Jonathan St-Germain, Tianyi Ling, Courtney L. Jones, Brian Raught, Austin E. Gillen, Monica Ransom, Sarah Staggs, Clayton A. Smith, Daniel A. Pollyea, Brett M. Stevens, Craig T. Jordan. Targeting Acute Myeloid Leukemia Stem Cells through Perturbation of Mitochondrial Calcium. Cancer Discovery 2024, 14
(10)
, 1922-1939. https://doi.org/10.1158/2159-8290.CD-23-1145
- J. P. Jose Merlin, Anine Crous, Heidi Abrahamse. Combining Photodynamic Therapy and Targeted Drug Delivery Systems: Enhancing Mitochondrial Toxicity for Improved Cancer Outcomes. International Journal of Molecular Sciences 2024, 25
(19)
, 10796. https://doi.org/10.3390/ijms251910796
- An Xie, Gyeoung-Jin Kang, Eun Ji Kim, Hong Liu, Feng Feng, Samuel C. Dudley. c-Src Is Responsible for Mitochondria-Mediated Arrhythmic Risk in Ischemic Cardiomyopathy. Circulation: Arrhythmia and Electrophysiology 2024, 17
(10)
https://doi.org/10.1161/CIRCEP.124.013054
- Jin Guo, Yukun Wang, Chunxia Shi, Danmei Zhang, Qingqi Zhang, Luwen Wang, Zuojiong Gong. Mitochondrial calcium uniporter complex: Unveiling the interplay between its regulators and calcium homeostasis. Cellular Signalling 2024, 121 , 111284. https://doi.org/10.1016/j.cellsig.2024.111284
- Nicholas P. Bigham, Robyn J. Novorolsky, Keana R. Davis, Haipei Zou, Samantha N. MacMillan, Michael J. Stevenson, George S. Robertson, Justin J. Wilson. Supramolecular delivery of dinuclear ruthenium and osmium MCU inhibitors. Inorganic Chemistry Frontiers 2024, 11
(16)
, 5064-5079. https://doi.org/10.1039/D4QI01102C
- Tulanisa Kadier, Yi-guo Zhang, Yi-xin Jing, Zi-yi Weng, Shi-shi Liao, Jie Luo, Ke Ding, Chen Cao, Rong Chen, Qing-tao Meng. MCU inhibition protects against intestinal ischemia‒reperfusion by inhibiting Drp1-dependent mitochondrial fission. Free Radical Biology and Medicine 2024, 221 , 111-124. https://doi.org/10.1016/j.freeradbiomed.2024.05.024
- Matthew K. Kirchner, Ferdinand Althammer, Elba Campos-Lira, Juliana Montanez, Javier E. Stern. Endoplasmic Reticulum and Mitochondrial Calcium Handling Dynamically Shape Slow Afterhyperpolarizations in Vasopressin Magnocellular Neurons. The Journal of Neuroscience 2024, 44
(30)
, e0003242024. https://doi.org/10.1523/JNEUROSCI.0003-24.2024
- Robert Valencia, Joshua W. Kranrod, Liye Fang, Amro M. Soliman, Brandon Azer, Xavier Clemente‐Casares, John M. Seubert. Linoleic acid‐derived diol 12,13‐
DiHOME
enhances
NLRP3
inflammasome activation in macrophages. The FASEB Journal 2024, 38
(13)
https://doi.org/10.1096/fj.202301640RR
- Danielle M. Colussi, Peter B. Stathopulos. The mitochondrial calcium uniporter: Balancing tumourigenic and anti‐tumourigenic responses. The Journal of Physiology 2024, 602
(14)
, 3315-3339. https://doi.org/10.1113/JP285515
- Umar Amjid, Ubair Aziz, Uzma Habib, Ishrat Jabeen. Biological regulatory network analysis for targeting the mitochondrial calcium uniporter (MCU) mediated calcium (Ca
2+
) transport in neurodegenerative disorders. Cell Biochemistry and Function 2024, 42
(5)
https://doi.org/10.1002/cbf.4082
- Yunkyung Eom, Sung Rae Kim, Yeong-Kyeong Kim, Sung Hoon Lee. Mitochondrial Calcium Waves by Electrical Stimulation in Cultured Hippocampal Neurons. Molecular Neurobiology 2024, 61
(6)
, 3477-3489. https://doi.org/10.1007/s12035-023-03795-w
- Ya Hou, Fuhan Fan, Na Xie, Yi Zhang, Xiaobo Wang, Xianli Meng. Rhodiola crenulata alleviates hypobaric hypoxia-induced brain injury by maintaining BBB integrity and balancing energy metabolism dysfunction. Phytomedicine 2024, 128 , 155529. https://doi.org/10.1016/j.phymed.2024.155529
- Peng Xu, Sarpras Swain, Robyn J. Novorolsky, Esperanza Garcia, Zhouyang Huang, Terrance P. Snutch, Justin J. Wilson, George S. Robertson, Robert B. Renden. The mitochondrial calcium uniporter inhibitor Ru265 increases neuronal excitability and reduces neurotransmission via off‐target effects. British Journal of Pharmacology 2024, https://doi.org/10.1111/bph.16425
- Thiruvelselvan Ponnusamy, Prema Velusamy, Santhanam Shanmughapriya. Mrs2-mediated mitochondrial magnesium uptake is essential for the regulation of MCU-mediated mitochondrial Ca2+ uptake and viability. Mitochondrion 2024, 76 , 101877. https://doi.org/10.1016/j.mito.2024.101877
- Tyler L. Stevens, Henry M. Cohen, Joanne F. Garbincius, John W. Elrod. Mitochondrial calcium uniporter channel gatekeeping in cardiovascular disease. Nature Cardiovascular Research 2024, 3
(5)
, 500-514. https://doi.org/10.1038/s44161-024-00463-7
- Alissa Lance‐Byrne, Brent Lindquist‐Kleissler, Timothy C. Johnstone. Chemical Structure Elucidation in the Development of Inorganic Drugs: Evidence from Ru‐, Au‐, As‐, and Sb‐based Medicines. European Journal of Inorganic Chemistry 2024, 27
(11)
https://doi.org/10.1002/ejic.202300717
- Sasha R. Weller, John E. Burnell, Brandon M. Aho, Bright Obeng, Emily L. Ledue, Juyoung K. Shim, Samuel T. Hess, Julie A. Gosse. Antimicrobial cetylpyridinium chloride causes functional inhibition of mitochondria as potently as canonical mitotoxicants, nanostructural disruption of mitochondria, and mitochondrial Ca2+ efflux in living rodent and primary human cells. Food and Chemical Toxicology 2024, 186 , 114547. https://doi.org/10.1016/j.fct.2024.114547
- Renjia Zhong, Michael T. Rua, Lan Wei‐LaPierre. Targeting mitochondrial Ca
2+
uptake for the treatment of amyotrophic lateral sclerosis. The Journal of Physiology 2024, 602
(8)
, 1519-1549. https://doi.org/10.1113/JP284143
- Abhishek Singh, Satish Kumar, Tusar Kanta Acharya, Shamit Kumar, Saurabh Chawla, Chandan Goswami, Luna Goswami. Modulation of calcium-influx by carboxymethyl tamarind‑gold nanoparticles promotes biomineralization for tissue regeneration. International Journal of Biological Macromolecules 2024, 264 , 130605. https://doi.org/10.1016/j.ijbiomac.2024.130605
- Anqi Xu, Yixuan Wang, Dongliu Luo, Yu Xia, Hua Xue, Haidong Yao, Shu Li. By regulating the IP3R/GRP75/VDAC1 complex to restore mitochondrial dynamic balance, selenomethionine reduces lipopolysaccharide‐induced neuronal apoptosis. Journal of Cellular Physiology 2024, 239
(4)
https://doi.org/10.1002/jcp.31190
- Rachel L Doser, Kaz M Knight, Ennis W Deihl, Frederic J Hoerndli. Activity-dependent mitochondrial ROS signaling regulates recruitment of glutamate receptors to synapses. eLife 2024, 13 https://doi.org/10.7554/eLife.92376
- Jin Wang, Jinyong Jiang, Haoliang Hu, Linxi Chen. MCU complex: Exploring emerging targets and mechanisms of mitochondrial physiology and pathology. Journal of Advanced Research 2024, 2021 https://doi.org/10.1016/j.jare.2024.02.013
- Kamila S. Nebesnaya, Albert R. Makhmudov, Khondamir R. Rustamov, Nigina S.H. Rakhmatullina, Sarvinoz I. Rustamova, Ulugbek Z. Mirkhodjaev, Oksana S. Charishnikova, Ravshan Z. Sabirov, Artyom Y. Baev. Inorganic polyphosphate regulates functions of thymocytes via activation of P2X purinoreceptors. Biochimica et Biophysica Acta (BBA) - General Subjects 2024, 1868
(1)
, 130523. https://doi.org/10.1016/j.bbagen.2023.130523
- Erna Mitaishvili, Hanna Feinsod, Zachary David, Jessica Shpigel, Chelsea Fernandez, Moira Sauane, Columba de la Parra. The Molecular Mechanisms behind Advanced Breast Cancer Metabolism: Warburg Effect, OXPHOS, and Calcium. Frontiers in Bioscience-Landmark 2024, 29
(3)
https://doi.org/10.31083/j.fbl2903099
- Alejandro Marmolejo-Garza, Inge E. Krabbendam, Minh Danh Anh Luu, Famke Brouwer, Marina Trombetta-Lima, Osman Unal, Shane J. O’Connor, Naďa Majerníková, Carolina R. S. Elzinga, Cristina Mammucari, Martina Schmidt, Muniswamy Madesh, Erik Boddeke, Amalia M. Dolga. Negative modulation of mitochondrial calcium uniporter complex protects neurons against ferroptosis. Cell Death & Disease 2023, 14
(11)
https://doi.org/10.1038/s41419-023-06290-1
- Xavier R. Chapa-Dubocq, Keishla M. Rodríguez-Graciani, Nelson Escobales, Sabzali Javadov. Mitochondrial Volume Regulation and Swelling Mechanisms in Cardiomyocytes. Antioxidants 2023, 12
(8)
, 1517. https://doi.org/10.3390/antiox12081517
- Robyn J. Novorolsky, Gracious D. S. Kasheke, Antoine Hakim, Marianna Foldvari, Gabriel G. Dorighello, Israel Sekler, Vidyasagar Vuligonda, Martin E. Sanders, Robert B. Renden, Justin J. Wilson, George S. Robertson. Preserving and enhancing mitochondrial function after stroke to protect and repair the neurovascular unit: novel opportunities for nanoparticle-based drug delivery. Frontiers in Cellular Neuroscience 2023, 17 https://doi.org/10.3389/fncel.2023.1226630
- Preetam Kishore, Amelie C. T. Collinet, Bianca J. J. M. Brundel. Prevention of Atrial Fibrillation: Putting Proteostasis Derailment Back on Track. Journal of Clinical Medicine 2023, 12
(13)
, 4352. https://doi.org/10.3390/jcm12134352
- Siva Prasad Panda, Adarsh Kesharwani. Micronutrients/miRs/ATP networking in mitochondria: Clinical intervention with ferroptosis, cuproptosis, and calcium burden. Mitochondrion 2023, 71 , 1-16. https://doi.org/10.1016/j.mito.2023.05.003
- Zhouyang Huang, Justin J. Wilson. Structure‐Activity Relationships of Metal‐Based Inhibitors of the Mitochondrial Calcium Uniporter. ChemMedChem 2023, 18
(12)
https://doi.org/10.1002/cmdc.202300106
- Yavuz F. Yazicioglu, Eros Marin, Ciaran Sandhu, Silvia Galiani, Iwan G. A. Raza, Mohammad Ali, Barbara Kronsteiner, Ewoud B. Compeer, Moustafa Attar, Susanna J. Dunachie, Michael L. Dustin, Alexander J. Clarke. Dynamic mitochondrial transcription and translation in B cells control germinal center entry and lymphomagenesis. Nature Immunology 2023, 24
(6)
, 991-1006. https://doi.org/10.1038/s41590-023-01484-3
- Agnese De Mario, Donato D'Angelo, Giuseppe Zanotti, Anna Raffaello, Cristina Mammucari. The mitochondrial calcium uniporter complex–A play in five acts. Cell Calcium 2023, 112 , 102720. https://doi.org/10.1016/j.ceca.2023.102720
- Macarena Rodríguez-Prados, Kai-Ting Huang, Katalin Márta, Melanie Paillard, György Csordás, Suresh K. Joseph, György Hajnóczky. MICU1 controls the sensitivity of the mitochondrial Ca2+ uniporter to activators and inhibitors. Cell Chemical Biology 2023, 30
(6)
, 606-617.e4. https://doi.org/10.1016/j.chembiol.2023.05.002
- Ildiko Szabo, Adam Szewczyk. Mitochondrial Ion Channels. Annual Review of Biophysics 2023, 52
(1)
, 229-254. https://doi.org/10.1146/annurev-biophys-092622-094853
- Saeid Bagheri‐Mohammadi, Mohammad Farjami, Ali Jaafari Suha, Seyed Mohsen Aghaei Zarch, Sajad Najafi, Ali Esmaeili. The mitochondrial calcium signaling, regulation, and cellular functions: A novel target for therapeutic medicine in neurological disorders. Journal of Cellular Biochemistry 2023, 124
(5)
, 635-655. https://doi.org/10.1002/jcb.30414
- Qingxi Zhang, Yin Huang, Anbiao Wu, Qingrui Duan, Peikun He, Haifeng Huang, Yuyuan Gao, Kun Nie, Qicai Liu, Lijuan Wang. Calcium/calmodulin-dependent serine protein kinase exacerbates mitochondrial calcium uniporter-related mitochondrial calcium overload by phosphorylating α-synuclein in Parkinson’s disease. The International Journal of Biochemistry & Cell Biology 2023, 157 , 106385. https://doi.org/10.1016/j.biocel.2023.106385
- Omar Lozano, Patricio Marcos, Felipe de Jesús Salazar‐Ramirez, Anay F. Lázaro‐Alfaro, Luis Sobrevia, Gerardo García‐Rivas. Targeting the mitochondrial Ca
2+
uniporter complex in cardiovascular disease. Acta Physiologica 2023, 237
(4)
https://doi.org/10.1111/apha.13946
- Yun Haeng Lee, Myeong Uk Kuk, Moon Kyoung So, Eun Seon Song, Haneur Lee, Soon Kil Ahn, Hyung Wook Kwon, Joon Tae Park, Sang Chul Park. Targeting Mitochondrial Oxidative Stress as a Strategy to Treat Aging and Age-Related Diseases. Antioxidants 2023, 12
(4)
, 934. https://doi.org/10.3390/antiox12040934
- Nicholas P. Bigham, Justin J. Wilson. Investigation of Cobalt(III) Cage Complexes as Inhibitors of the Mitochondrial Calcium Uniporter. European Journal of Inorganic Chemistry 2023, 26
(9)
https://doi.org/10.1002/ejic.202200735
- Zhouyang Huang, Samantha N. MacMillan, Justin J. Wilson. A Fluorogenic Inhibitor of the Mitochondrial Calcium Uniporter. Angewandte Chemie 2023, 135
(6)
https://doi.org/10.1002/ange.202214920
- Zhouyang Huang, Samantha N. MacMillan, Justin J. Wilson. A Fluorogenic Inhibitor of the Mitochondrial Calcium Uniporter. Angewandte Chemie International Edition 2023, 62
(6)
https://doi.org/10.1002/anie.202214920
- Zhouyang Huang, Jesse A. Spivey, Samantha N. MacMillan, Justin J. Wilson. A ferrocene-containing analogue of the MCU inhibitor Ru265 with increased cell permeability. Inorganic Chemistry Frontiers 2023, 10
(2)
, 591-599. https://doi.org/10.1039/D2QI02183H
- Joshua J. Woods, Robyn J. Novorolsky, Nicholas P. Bigham, George S. Robertson, Justin J. Wilson. Dinuclear nitrido-bridged osmium complexes inhibit the mitochondrial calcium uniporter and protect cortical neurons against lethal oxygen–glucose deprivation. RSC Chemical Biology 2023, 4
(1)
, 84-93. https://doi.org/10.1039/D2CB00189F
- Danielle M. Colussi, Peter B. Stathopulos. From passage to inhibition: Uncovering the structural and physiological inhibitory mechanisms of
MCUb
in mitochondrial calcium regulation. The FASEB Journal 2023, 37
(1)
https://doi.org/10.1096/fj.202201080R
- Weiwei Zhang, Bo Liu, Yazhou Wang, Hengli Zhang, Lang He, Pan Wang, Mingqing Dong. Mitochondrial dysfunction in pulmonary arterial hypertension. Frontiers in Physiology 2022, 13 https://doi.org/10.3389/fphys.2022.1079989
- Christopher J. Groten, Brian A. MacVicar. Mitochondrial Ca2+ uptake by the MCU facilitates pyramidal neuron excitability and metabolism during action potential firing. Communications Biology 2022, 5
(1)
https://doi.org/10.1038/s42003-022-03848-1
- Nikita Arnst, Nelly Redolfi, Annamaria Lia, Martina Bedetta, Elisa Greotti, Paola Pizzo. Mitochondrial Ca2+ Signaling and Bioenergetics in Alzheimer’s Disease. Biomedicines 2022, 10
(12)
, 3025. https://doi.org/10.3390/biomedicines10123025
- Keerti Mishra, Min Luo. Mitochondrial Channels and Their Role in Cardioprotection. 2022https://doi.org/10.5772/intechopen.101127
- Shashank Kumar Maurya, Suchi Gupta, Amrita Bakshi, Harpreet Kaur, Arushi Jain, Sabyasachi Senapati, Meghraj Singh Baghel. Targeting mitochondria in the regulation of neurodegenerative diseases: A comprehensive review. Journal of Neuroscience Research 2022, 100
(10)
, 1845-1861. https://doi.org/10.1002/jnr.25110
- Li Zhang, Felicia Dietsche, Bruno Seitaj, Liliana Rojas-Charry, Nadina Latchman, Dhanendra Tomar, Rob CI Wüst, Alexander Nickel, Katrin BM Frauenknecht, Benedikt Schoser, Sven Schumann, Michael J Schmeisser, Johannes vom Berg, Thorsten Buch, Stefanie Finger, Philip Wenzel, Christoph Maack, John W Elrod, Jan B Parys, Geert Bultynck, Axel Methner. TMBIM5 loss of function alters mitochondrial matrix ion homeostasis and causes a skeletal myopathy. Life Science Alliance 2022, 5
(10)
, e202201478. https://doi.org/10.26508/lsa.202201478
- Alejandro Marmolejo-Garza, Tiago Medeiros-Furquim, Ramya Rao, Bart J.L. Eggen, Erik Boddeke, Amalia M. Dolga. Transcriptomic and epigenomic landscapes of Alzheimer's disease evidence mitochondrial-related pathways. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research 2022, 1869
(10)
, 119326. https://doi.org/10.1016/j.bbamcr.2022.119326
- Karima Ait-Aissa, Olha M. Koval, Nathanial R. Lindsey, Isabella M. Grumbach. Mitochondrial Ca
2+
Uptake Drives Endothelial Injury By Radiation Therapy. Arteriosclerosis, Thrombosis, and Vascular Biology 2022, 42
(9)
, 1121-1136. https://doi.org/10.1161/ATVBAHA.122.317869
- Celio Damacena de Angelis, Benney T. Endoni, Daniel Nuno, Kathryn Lamping, Johannes Ledolter, Olha M. Koval, Isabella M. Grumbach. Sex‐Specific Differences in Endothelial Function Are Driven by Divergent Mitochondrial Ca
2+
Handling. Journal of the American Heart Association 2022, 11
(13)
https://doi.org/10.1161/JAHA.121.023912
- Zhanchen Dong, Jianyu Wang, Tianming Qiu, Jialu Wu, Yu An, Xiaoxia Shi, Xiance Sun, Liping Jiang, Xiaofang Liu, Guang Yang, Jun Cao, Xiaofeng Yao. Perfluorooctane sulfonate induces mitochondrial calcium overload and early hepatic insulin resistance via autophagy/detyrosinated alpha-tubulin-regulated IP3R2-VDAC1-MICU1 interaction. Science of The Total Environment 2022, 825 , 153933. https://doi.org/10.1016/j.scitotenv.2022.153933
- Joanne F. Garbincius, John W. Elrod. Mitochondrial calcium exchange in physiology and disease. Physiological Reviews 2022, 102
(2)
, 893-992. https://doi.org/10.1152/physrev.00041.2020
- Giampaolo Morciano, Alessandro Rimessi, Simone Patergnani, Veronica A.M. Vitto, Alberto Danese, Asrat Kahsay, Laura Palumbo, Massimo Bonora, Mariusz R. Wieckowski, Carlotta Giorgi, Paolo Pinton. Calcium dysregulation in heart diseases: Targeting calcium channels to achieve a correct calcium homeostasis. Pharmacological Research 2022, 177 , 106119. https://doi.org/10.1016/j.phrs.2022.106119
- Dale S. George, Sandra Hackelberg, Nirupa D. Jayaraj, Dongjun Ren, Seby L. Edassery, Craig A. Rathwell, Rachel E. Miller, Anne-Marie Malfait, Jeffrey N. Savas, Richard J. Miller, Daniela M. Menichella. Mitochondrial calcium uniporter deletion prevents painful diabetic neuropathy by restoring mitochondrial morphology and dynamics. Pain 2022, 163
(3)
, 560-578. https://doi.org/10.1097/j.pain.0000000000002391
- Joshua J. Woods, Jesse A. Spivey, Justin J. Wilson. A [
1
H,
15
N] Heteronuclear Single Quantum Coherence NMR Study of the Solution Reactivity of the Ruthenium‐Based Mitochondrial Calcium Uniporter Inhibitor Ru265. European Journal of Inorganic Chemistry 2022, 2022
(6)
https://doi.org/10.1002/ejic.202100995
- Anu Antony, Neville Ng, Antonio Lauto, Jens R. Coorssen, Simon J. Myers. Calcium-Mediated Calpain Activation and Microtubule Dissociation in Cell Model of Hereditary Sensory Neuropathy Type-1 Expressing V144D
SPTLC1
Mutation. DNA and Cell Biology 2022, 41
(2)
, 225-234. https://doi.org/10.1089/dna.2021.0816
- Veronica Costiniti, Guilherme H. S. Bomfim, Maria Neginskaya, Ga‐Yeon Son, Erna Mitaishvili, Marta Giacomello, Evgeny Pavlov, Rodrigo S. Lacruz. Mitochondria modulate ameloblast Ca
2+
signaling. The FASEB Journal 2022, 36
(2)
https://doi.org/10.1096/fj.202100602R
- Anna Maria Krstic, Amelia Sally Power, Marie-Louise Ward. Visualization of Dynamic Mitochondrial Calcium Fluxes in Isolated Cardiomyocytes. Frontiers in Physiology 2022, 12 https://doi.org/10.3389/fphys.2021.808798
- Zhanchen Dong, Xiaofeng Yao. Insight of the role of mitochondrial calcium homeostasis in hepatic insulin resistance. Mitochondrion 2022, 62 , 128-138. https://doi.org/10.1016/j.mito.2021.11.007
- Andrey Khaitin. Calcium in Neuronal and Glial Response to Axotomy. International Journal of Molecular Sciences 2021, 22
(24)
, 13344. https://doi.org/10.3390/ijms222413344
- Neelanjan Vishnu, Justin Wilson, Muniswamy Madesh. Emergence of repurposed drugs as modulators of MCU channel for clinical therapeutics. Cell Calcium 2021, 99 , 102456. https://doi.org/10.1016/j.ceca.2021.102456
- Yan-Cheng Shen, Yan-Jhih Shen, Wen-Sen Lee, Michael Yu-Chih Chen, Wei-Chia Tu, Kun-Ta Yang. Two Benzene Rings with a Boron Atom Comprise the Core Structure of 2-APB Responsible for the Anti-Oxidative and Protective Effect on the Ischemia/Reperfusion-Induced Rat Heart Injury. Antioxidants 2021, 10
(11)
, 1667. https://doi.org/10.3390/antiox10111667
- Yun Haeng Lee, Ji Yun Park, Haneur Lee, Eun Seon Song, Myeong Uk Kuk, Junghyun Joo, Sekyung Oh, Hyung Wook Kwon, Joon Tae Park, Sang Chul Park. Targeting Mitochondrial Metabolism as a Strategy to Treat Senescence. Cells 2021, 10
(11)
, 3003. https://doi.org/10.3390/cells10113003
- Suman Saurav, Jyoti Tanwar, Kriti Ahuja, Rajender K. Motiani. Dysregulation of host cell calcium signaling during viral infections: Emerging paradigm with high clinical relevance. Molecular Aspects of Medicine 2021, 81 , 101004. https://doi.org/10.1016/j.mam.2021.101004
- Alicia G. Gómez-Valadés, Macarena Pozo, Luis Varela, Mehdi Boutagouga Boudjadja, Sara Ramírez, Iñigo Chivite, Elena Eyre, Roberta Haddad-Tóvolli, Arnaud Obri, Maria Milà-Guasch, Jordi Altirriba, Marc Schneeberger, Mónica Imbernón, Angela R. Garcia-Rendueles, Pau Gama-Perez, Jonathan Rojo-Ruiz, Bence Rácz, Maria Teresa Alonso, Ramon Gomis, Antonio Zorzano, Giuseppe D’Agostino, Clara V. Alvarez, Rubén Nogueiras, Pablo M. Garcia-Roves, Tamas L. Horvath, Marc Claret. Mitochondrial cristae-remodeling protein OPA1 in POMC neurons couples Ca2+ homeostasis with adipose tissue lipolysis. Cell Metabolism 2021, 33
(9)
, 1820-1835.e9. https://doi.org/10.1016/j.cmet.2021.07.008
- Shanna Hamilton, Radmila Terentyeva, Richard T. Clements, Andriy E. Belevych, Dmitry Terentyev. Sarcoplasmic reticulum-mitochondria communication; implications for cardiac arrhythmia. Journal of Molecular and Cellular Cardiology 2021, 156 , 105-113. https://doi.org/10.1016/j.yjmcc.2021.04.002
- Alberto Danese, Sara Leo, Alessandro Rimessi, Mariusz R. Wieckowski, Francesco Fiorica, Carlotta Giorgi, Paolo Pinton. Cell death as a result of calcium signaling modulation: A cancer-centric prospective. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research 2021, 1868
(8)
, 119061. https://doi.org/10.1016/j.bbamcr.2021.119061
- Joshua J. Woods, Madison X. Rodriguez, Chen-Wei Tsai, Ming-Feng Tsai, Justin J. Wilson. Cobalt amine complexes and Ru265 interact with the DIME region of the mitochondrial calcium uniporter. Chemical Communications 2021, 57
(50)
, 6161-6164. https://doi.org/10.1039/D1CC01623G
- Taylor E. Huntington, Rahul Srinivasan. Astrocytic mitochondria in adult mouse brain slices show spontaneous calcium influx events with unique properties. Cell Calcium 2021, 96 , 102383. https://doi.org/10.1016/j.ceca.2021.102383
- Agnese De Mario, Anna Tosatto, Julia Marie Hill, Janos Kriston-Vizi, Robin Ketteler, Denis Vecellio Reane, Gino Cortopassi, Gyorgy Szabadkai, Rosario Rizzuto, Cristina Mammucari. Identification and functional validation of FDA-approved positive and negative modulators of the mitochondrial calcium uniporter. Cell Reports 2021, 35
(12)
, 109275. https://doi.org/10.1016/j.celrep.2021.109275
- Ildiko Szabo, Mario Zoratti, Lucia Biasutto. Targeting mitochondrial ion channels for cancer therapy. Redox Biology 2021, 42 , 101846. https://doi.org/10.1016/j.redox.2020.101846
- Lorenzo Modesti, Alberto Danese, Veronica Angela Maria Vitto, Daniela Ramaccini, Gianluca Aguiari, Roberta Gafà, Giovanni Lanza, Carlotta Giorgi, Paolo Pinton. Mitochondrial Ca2+ Signaling in Health, Disease and Therapy. Cells 2021, 10
(6)
, 1317. https://doi.org/10.3390/cells10061317
- Konstantin N. Belosludtsev, Rinat R. Sharipov, Dmitry P. Boyarkin, Natalia V. Belosludtseva, Mikhail V. Dubinin, Irina A. Krasilnikova, Zanda V. Bakaeva, Arina E. Zgodova, Vsevolod G. Pinelis, Alexander M. Surin. The effect of DS16570511, a new inhibitor of mitochondrial calcium uniporter, on calcium homeostasis, metabolism, and functional state of cultured cortical neurons and isolated brain mitochondria. Biochimica et Biophysica Acta (BBA) - General Subjects 2021, 1865
(5)
, 129847. https://doi.org/10.1016/j.bbagen.2021.129847
- Ian de Ridder, Martijn Kerkhofs, Santhini Pulikkal Veettil, Wim Dehaen, Geert Bultynck. Cancer cell death strategies by targeting Bcl-2's BH4 domain. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research 2021, 1868
(5)
, 118983. https://doi.org/10.1016/j.bbamcr.2021.118983
- Alessandro Rimessi, Veronica A. M. Vitto, Simone Patergnani, Paolo Pinton. Update on Calcium Signaling in Cystic Fibrosis Lung Disease. Frontiers in Pharmacology 2021, 12 https://doi.org/10.3389/fphar.2021.581645
- Martijn Kerkhofs, Rita La Rovere, Kirsten Welkenhuysen, Ann Janssens, Peter Vandenberghe, Muniswamy Madesh, Jan B. Parys, Geert Bultynck. BIRD-2, a BH4-domain-targeting peptide of Bcl-2, provokes Bax/Bak-independent cell death in B-cell cancers through mitochondrial Ca2+-dependent mPTP opening. Cell Calcium 2021, 94 , 102333. https://doi.org/10.1016/j.ceca.2020.102333
- Md Imam Faizan, Tanveer Ahmad. Altered mitochondrial calcium handling and cell death by necroptosis: An emerging paradigm. Mitochondrion 2021, 57 , 47-62. https://doi.org/10.1016/j.mito.2020.12.004
- Michael D. Wetzel, Kristen Stanley, Soumya Maity, Muniswamy Madesh, Jean C. Bopassa, Alaa S. Awad. Homoarginine ameliorates diabetic nephropathy independent of nitric oxide synthase‐3. Physiological Reports 2021, 9
(5)
https://doi.org/10.14814/phy2.14766
- Elizabeth Murphy, Charles Steenbergen. Regulation of Mitochondrial Ca
2+
Uptake. Annual Review of Physiology 2021, 83
(1)
, 107-126. https://doi.org/10.1146/annurev-physiol-031920-092419
- Katalin Márta, Prottoy Hasan, Macarena Rodríguez-Prados, Melanie Paillard, György Hajnóczky. Pharmacological inhibition of the mitochondrial Ca2+ uniporter: Relevance for pathophysiology and human therapy. Journal of Molecular and Cellular Cardiology 2021, 151 , 135-144. https://doi.org/10.1016/j.yjmcc.2020.09.014
- Riccardo Filadi, Elisa Greotti. The yin and yang of mitochondrial Ca2+ signaling in cell physiology and pathology. Cell Calcium 2021, 93 , 102321. https://doi.org/10.1016/j.ceca.2020.102321
- Bhargavi Duvvuri, Christian Lood. Mitochondrial Calcification. Immunometabolism 2021, 3
(1)
https://doi.org/10.20900/immunometab20210008
- Yang Liu, Mingpeng Jin, Yaya Wang, Jianjun Zhu, Rui Tan, Jing Zhao, Xiaoying Ji, Chao Jin, Yongfeng Jia, Tingting Ren, Jinliang Xing. MCU-induced mitochondrial calcium uptake promotes mitochondrial biogenesis and colorectal cancer growth. Signal Transduction and Targeted Therapy 2020, 5
(1)
https://doi.org/10.1038/s41392-020-0155-5
- Mayara S. Bertolini, Roberto Docampo. Different Sensitivity of Control and MICU1- and MICU2-Ablated Trypanosoma cruzi Mitochondrial Calcium Uniporter Complex to Ruthenium-Based Inhibitors. International Journal of Molecular Sciences 2020, 21
(23)
, 9316. https://doi.org/10.3390/ijms21239316
- Archita Ray, Ashish Jaiswal, Joytri Dutta, Sabita Singh, Ulaganathan Mabalirajan. A looming role of mitochondrial calcium in dictating the lung epithelial integrity and pathophysiology of lung diseases. Mitochondrion 2020, 55 , 111-121. https://doi.org/10.1016/j.mito.2020.09.004
- Qun Guan, Le‐Le Zhou, Fan‐Hong Lv, Wen‐Yan Li, Yan‐An Li, Yu‐Bin Dong. A Glycosylated Covalent Organic Framework Equipped with BODIPY and CaCO
3
for Synergistic Tumor Therapy. Angewandte Chemie International Edition 2020, 59
(41)
, 18042-18047. https://doi.org/10.1002/anie.202008055
- Qun Guan, Le‐Le Zhou, Fan‐Hong Lv, Wen‐Yan Li, Yan‐An Li, Yu‐Bin Dong. A Glycosylated Covalent Organic Framework Equipped with BODIPY and CaCO
3
for Synergistic Tumor Therapy. Angewandte Chemie 2020, 132
(41)
, 18198-18203. https://doi.org/10.1002/ange.202008055
- Michael D. Wetzel, Kristen Stanley, Wei Wei Wang, Soumya Maity, Muniswamy Madesh, W. Brian Reeves, Alaa S. Awad. Selective inhibition of arginase-2 in endothelial cells but not proximal tubules reduces renal fibrosis. JCI Insight 2020, 5
(19)
https://doi.org/10.1172/jci.insight.142187
- Simone Patergnani, Veronica A.M. Vitto, Paolo Pinton, Alessandro Rimessi. Mitochondrial Stress Responses and “Mito-Inflammation” in Cystic Fibrosis. Frontiers in Pharmacology 2020, 11 https://doi.org/10.3389/fphar.2020.581114
- Sonia Missiroli, Mariasole Perrone, Ilaria Genovese, Paolo Pinton, Carlotta Giorgi. Cancer metabolism and mitochondria: Finding novel mechanisms to fight tumours. eBioMedicine 2020, 59 , 102943. https://doi.org/10.1016/j.ebiom.2020.102943
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 94 other publications.
- 1Berridge, M. J.; Bootman, M. D.; Roderick, H. L. Calcium Signalling: Dynamics, Homeostasis and Remodelling. Nat. Rev. Mol. Cell Biol. 2003, 4, 517– 529, DOI: 10.1038/nrm11551https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXltVWmsr8%253D&md5=da79c7368cc92aeb53f243858e1fcd70Calcium: Calcium signaling: dynamics, homeostasis and remodelingBerridge, Michael J.; Bootman, Martin D.; Roderick, H. LlewelynNature Reviews Molecular Cell Biology (2003), 4 (7), 517-529CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. Ca2+ is a highly versatile intracellular signal that operates over a wide temporal range to regulate many different cellular processes. An extensive Ca2+-signaling toolkit is used to assemble signaling systems with very different spatial and temporal dynamics. Rapid highly localized Ca2+ spikes regulate fast responses, whereas slower responses are controlled by repetitive global Ca2+ transients or intracellular Ca2+ waves. Ca2+ plays a direct role in controlling the expression patterns of its signaling systems that are constantly being remodelled in both health and disease.
- 2Soboloff, J.; Rothberg, B. S.; Madesh, M.; Gill, D. L. STIM Proteins: Dynamic Calcium Signal Transducers. Nat. Rev. Mol. Cell Biol. 2012, 13, 549– 565, DOI: 10.1038/nrm34142https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xht1Ghsb7N&md5=b26a6baa9b336d6f750ec2447c143fc7STIM proteins: dynamic calcium signal transducersSoboloff, Jonathan; Rothberg, Brad S.; Madesh, Muniswamy; Gill, Donald L.Nature Reviews Molecular Cell Biology (2012), 13 (9), 549-565CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. Stromal interaction mol. (STIM) proteins function in cells as dynamic coordinators of cellular calcium (Ca2+) signals. Spanning the endoplasmic reticulum (ER) membrane, they sense tiny changes in the levels of Ca2+ stored within the ER lumen. As ER Ca2+ is released to generate primary Ca2+ signals, STIM proteins undergo an intricate activation reaction and rapidly translocate into junctions formed between the ER and the plasma membrane. There, STIM proteins tether and activate the highly Ca2+-selective Orai channels to mediate finely controlled Ca2+ signals and to homeostatically balance cellular Ca2+. Details are emerging on the remarkable organization within these STIM-induced junctional microdomains and the identification of new regulators and alternative target proteins for STIM.
- 3Clapham, D. E. Calcium Signaling. Cell 2007, 131, 1047– 1058, DOI: 10.1016/j.cell.2007.11.0283https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXksFGgsA%253D%253D&md5=268945d7ca1ec7a455f69cb72272be85Calcium signalingClapham, David E.Cell (Cambridge, MA, United States) (2007), 131 (6), 1047-1058CODEN: CELLB5; ISSN:0092-8674. (Cell Press)A review. Ca2+ ions impact nearly every aspect of cellular life. Here, the author examines the principles of Ca2+ signaling, from changes in protein conformations driven by Ca2+ to the mechanisms that control Ca2+ levels in the cytoplasm and organelles. Also discussed is the highly localized nature of Ca2+-mediated signal transduction and its specific role in excitability, exocytosis, motility, apoptosis, and transcription.
- 4Hogan, P. G.; Lewis, R. S.; Rao, A. Molecular Basis of Calcium Signaling in Lymphocytes: STIM and ORAI. Annu. Rev. Immunol. 2010, 28, 491– 533, DOI: 10.1146/annurev.immunol.021908.1325504https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXlsVSmsL8%253D&md5=11db4a7bf473601794aa7e49e0de68d8Molecular basis of calcium signaling in lymphocytes: STIM and ORAIHogan, Patrick G.; Lewis, Richard S.; Rao, AnjanaAnnual Review of Immunology (2010), 28 (), 491-533CODEN: ARIMDU; ISSN:0732-0582. (Annual Reviews Inc.)A review. Ca2+ entry into cells of the peripheral immune system occurs through highly Ca2+-selective channels known as CRAC (calcium release-activated calcium) channels. CRAC channels are a very well-characterized example of store-operated Ca2+ channels, so designated because they open when the endoplasmic reticulum (ER) Ca2+ store becomes depleted. Physiol., Ca2+ is released from the ER lumen into the cytoplasm when activated receptors couple to phospholipase C and trigger prodn. of the second messenger inositol 1,4,5-trisphosphate (IP3). IP3 binds to IP3 receptors in the ER membrane and activates Ca2+ release. The proteins STIM and ORAI were discovered through limited and genome-wide RNAi screens, resp., performed in Drosophila cells and focused on identifying modulators of store-operated Ca2+ entry. STIM1 and STIM2 sense the depletion of ER Ca2+ stores, whereas ORAI1 is a pore subunit of the CRAC channel. In this review, we discuss selected aspects of Ca2+ signaling in cells of the immune system, focusing on the roles of STIM and ORAI proteins in store-operated Ca2+ entry.
- 5Kirichok, Y.; Krapivinsky, G.; Clapham, D. E. The Mitochondrial Calcium Uniporter is a Highly Selective Ion Channel. Nature 2004, 427, 360– 364, DOI: 10.1038/nature022465https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXltFCgtw%253D%253D&md5=37aefce568d7ac7225de675619283847The mitochondrial calcium uniporter is a highly selective ion channelKirichok, Yuriy; Krapivinsky, Grigory; Clapham, David E.Nature (London, United Kingdom) (2004), 427 (6972), 360-364CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)During intracellular Ca2+ signaling mitochondria accumulate significant amts. of Ca2+ from the cytosol. Mitochondrial Ca2+ uptake controls the rate of energy prodn., shapes the amplitude and spatio-temporal patterns of intracellular Ca2+ signals, and is instrumental to cell death. This Ca2+ uptake is undertaken by the mitochondrial Ca2+ uniporter (MCU) located in the organelle's inner membrane. The uniporter passes Ca2+ down the electrochem. gradient maintained across this membrane without direct coupling to ATP hydrolysis or transport of other ions. Carriers are characterized by turnover nos. that are typically 1,000-fold lower than ion channels, and until now it has been unclear whether the MCU is a carrier or a channel. By patch-clamping the inner mitochondrial membrane, we identified a previously unknown Ca2+-selective ion channel sensitive to inhibitors of mitochondrial Ca2+ uptake. Our data indicate that this unique channel binds Ca2+ with extremely high affinity (dissocn. const. ≤2 nM), enabling high Ca2+ selectivity despite relatively low cytoplasmic Ca2+ concns. The channel is inwardly rectifying, making it esp. effective for Ca2+ uptake into energized mitochondria. Thus, we conclude that the properties of the current mediated by this novel channel are those of the MCU.
- 6Kamer, K. J.; Mootha, V. K. The Molecular Era of the Mitochondrial Calcium Uniporter. Nat. Rev. Mol. Cell Biol. 2015, 16, 545– 553, DOI: 10.1038/nrm40396https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtlKntrbO&md5=8f0222b7cb0d6693adf957a3cd9ba983The molecular era of the mitochondrial calcium uniporterKamer, Kimberli J.; Mootha, Vamsi K.Nature Reviews Molecular Cell Biology (2015), 16 (9), 545-553CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. The mitochondrial calcium uniporter is an evolutionarily conserved calcium channel, and its biophys. properties and relevance to cell death, bioenergetics and signalling have been investigated for decades. However, the genes encoding this channel have only recently been discovered, opening up a new 'mol. era' in the study of its biol. We now know that the uniporter is not a single protein but rather a macromol. complex consisting of pore-forming and regulatory subunits. We review recent studies that harnessed the power of mol. biol. and genetics to characterize the mechanism of action of the uniporter, its evolution and its contribution to physiol. and human disease.
- 7Rizzuto, R.; De Stefani, D.; Raffaello, A.; Mammucari, C. Mitochondria as Sensors and Regulators of Calcium Signalling. Nat. Rev. Mol. Cell Biol. 2012, 13, 566– 578, DOI: 10.1038/nrm34127https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtFWms77O&md5=4b6f8ac747922757957102c8fa1420daMitochondria as sensors and regulators of calcium signallingRizzuto, Rosario; De Stefani, Diego; Raffaello, Anna; Mammucari, CristinaNature Reviews Molecular Cell Biology (2012), 13 (9), 566-578CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. During the past two decades, calcium (Ca2+) accumulation in energized mitochondria has emerged as a biol. process of utmost physiol. relevance. Mitochondrial Ca2+ uptake was shown to control intracellular Ca2+ signalling, cell metab., cell survival and other cell-type specific functions by buffering cytosolic Ca2+ levels and regulating mitochondrial effectors. Recently, the identity of mitochondrial Ca2+ transporters has been revealed, opening new perspectives for investigation and mol. intervention.
- 8Nemani, N.; Shanmughapriya, S.; Madesh, M. Molecular Regulation of MCU: Implications in Physiology and Disease. Cell Calcium 2018, 74, 86– 93, DOI: 10.1016/j.ceca.2018.06.0068https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXht1CntLvE&md5=c0b9d850bd8055add666550c2ade0867Molecular regulation of MCU: Implications in physiology and diseaseNemani, Neeharika; Shanmughapriya, Santhanam; Madesh, MuniswamyCell Calcium (2018), 74 (), 86-93CODEN: CECADV; ISSN:0143-4160. (Elsevier Ltd.)Ca2+ flux across the inner mitochondrial membrane (IMM) regulates cellular bioenergetics, intra-cellular cytoplasmic Ca2+ signals, and various cell death pathways. Ca2+ entry into the mitochondria occurs due to the highly neg. membrane potential (ΔΨm) through a selective inward rectifying MCU channel. In addn. to being regulated by various mitochondrial matrix resident proteins such as MICUs, MCUb, MCUR1 and EMRE, the channel is transcriptionally regulated by upstream Ca2+ cascade, post transnational modification and by divalent cations. The mode of regulation either inhibits or enhances MCU channel activity and thus regulates mitochondrial metab. and cell fate.
- 9Baughman, J. M.; Perocchi, F.; Girgis, H. S.; Plovanich, M.; Belcher-Timme, C. A.; Sancak, Y.; Bao, X. R.; Strittmatter, L.; Goldberger, O.; Bogorad, R. L.; Koteliansky, V.; Mootha, V. K. Integrative Genomics Identifies MCU as an Essential Component of the Mitochondrial Calcium Uniporter. Nature 2011, 476, 341– 345, DOI: 10.1038/nature102349https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXnslSktLo%253D&md5=14aa9d448ee67a2d3cc473ccbcabe822Integrative genomics identifies MCU as an essential component of the mitochondrial calcium uniporterBaughman, Joshua M.; Perocchi, Fabiana; Girgis, Hany S.; Plovanich, Molly; Belcher-Timme, Casey A.; Sancak, Yasemin; Bao, X. Robert; Strittmatter, Laura; Goldberger, Olga; Bogorad, Roman L.; Koteliansky, Victor; Mootha, Vamsi K.Nature (London, United Kingdom) (2011), 476 (7360), 341-345CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Mitochondria from diverse organisms are capable of transporting large amts. of Ca2+ via a ruthenium-red-sensitive, membrane-potential-dependent mechanism called the uniporter. Although the uniporter's biophys. properties have been studied extensively, its mol. compn. remains elusive. We recently used comparative proteomics to identify MICU1 (also known as CBARA1), an EF-hand-contg. protein that serves as a putative regulator of the uniporter. Here, we use whole-genome phylogenetic profiling, genome-wide RNA co-expression anal. and organelle-wide protein coexpression anal. to predict proteins functionally related to MICU1. All three methods converge on a novel predicted transmembrane protein, CCDC109A, that we now call "mitochondrial calcium uniporter" (MCU). MCU forms oligomers in the mitochondrial inner membrane, phys. interacts with MICU1, and resides within a large mol. wt. complex. Silencing MCU in cultured cells or in vivo in mouse liver severely abrogates mitochondrial Ca2+ uptake, whereas mitochondrial respiration and membrane potential remain fully intact. MCU has two predicted transmembrane helixes, which are sepd. by a highly conserved linker facing the intermembrane space. Acidic residues in this linker are required for its full activity. However, an S259A point mutation retains function but confers resistance to Ru360, the most potent inhibitor of the uniporter. Our genomic, physiol., biochem. and pharmacol. data firmly establish MCU as an essential component of the mitochondrial Ca2+ uniporter.
- 10De Stefani, D.; Raffaello, A.; Teardo, E.; Szabò, I.; Rizzuto, R. A Forty-Kilodalton Protein of the Inner Membrane is the Mitochondrial Calcium Uniporter. Nature 2011, 476, 336– 340, DOI: 10.1038/nature1023010https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXnslSksLg%253D&md5=8c86db6f4556b498cddb9d24a97de832A forty-kilodalton protein of the inner membrane is the mitochondrial calcium uniporterDe Stefani, Diego; Raffaello, Anna; Teardo, Enrico; Szabo, Ildiko; Rizzuto, RosarioNature (London, United Kingdom) (2011), 476 (7360), 336-340CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Mitochondrial Ca2+ homeostasis has a key role in the regulation of aerobic metab. and cell survival, but the mol. identity of the Ca2+ channel, the mitochondrial calcium uniporter, is still unknown. Here we have identified in silico a protein (named MCU) that shares tissue distribution with MICU1 (also known as CBARA1), a recently characterized uniporter regulator, is present in organisms in which mitochondrial Ca2+ uptake was demonstrated and whose sequence includes two transmembrane domains. Short interfering RNA (siRNA) silencing of MCU in HeLa cells markedly reduced mitochondrial Ca2+ uptake. MCU overexpression doubled the matrix Ca2+ concn. increase evoked by inositol 1,4,5-trisphosphate-generating agonists, thus significantly buffering the cytosolic elevation. The purified MCU protein showed channel activity in planar lipid bilayers, with electrophysiol. properties and inhibitor sensitivity of the uniporter. A mutant MCU, in which two neg. charged residues of the putative pore-forming region were replaced, had no channel activity and reduced agonist-dependent matrix Ca2+ concn. transients when overexpressed in HeLa cells. Overall, these data demonstrate that the 40-kDa protein identified is the channel responsible for ruthenium-red-sensitive mitochondrial Ca2+ uptake, thus providing a mol. basis for this process of utmost physiol. and pathol. relevance.
- 11Mallilankaraman, K.; Doonan, P.; Cárdenas, C.; Chandramoorthy, H. C.; Müller, M.; Miller, R.; Hoffman, N. E.; Gandhirajan, R. K.; Molgó, J.; Birnbaum, M. J.; Rothberg, B. S.; Mak, D. O. D.; Foskett, J. K.; Madesh, M. MICU1 is an Essential Gatekeeper for MCU-Mediated Mitochondrial Ca2+ Uptake That Regulates Cell Survival. Cell 2012, 151, 630– 644, DOI: 10.1016/j.cell.2012.10.011There is no corresponding record for this reference.
- 12Sancak, Y.; Markhard, A. L.; Kitami, T.; Kovacs-Bogdan, E.; Kamer, K. J.; Udeshi, N. D.; Carr, S. A.; Chaudhuri, D.; Clapham, D. E.; Li, A. A.; Calvo, S. E.; Goldberger, O.; Mootha, V. K. EMRE is an Essential Component of the Mitochondrial Calcium Uniporter Complex. Science 2013, 342, 1379– 1382, DOI: 10.1126/science.124299312https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvV2lsLjM&md5=1f9f02e65f9b735064db194245903737EMRE Is an Essential Component of the Mitochondrial Calcium Uniporter ComplexSancak, Yasemin; Markhard, Andrew L.; Kitami, Toshimori; Kovacs-Bogdan, Erika; Kamer, Kimberli J.; Udeshi, Namrata D.; Carr, Steven A.; Chaudhuri, Dipayan; Clapham, David E.; Li, Andrew A.; Calvo, Sarah E.; Goldberger, Olga; Mootha, Vamsi K.Science (Washington, DC, United States) (2013), 342 (6164), 1379-1382CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The mitochondrial uniporter is a highly selective calcium channel in the organelle's inner membrane. Its mol. components include the EF-hand-contg. calcium-binding proteins mitochondrial calcium uptake 1 (MICU1) and MICU2 and the pore-forming subunit mitochondrial calcium uniporter (MCU). We sought to achieve a full mol. characterization of the uniporter holocomplex (uniplex). Quant. mass spectrometry of affinity-purified uniplex recovered MICU1 and MICU2, MCU and its paralog MCUb, and a previously uncharacterized 10-kilodalton metazoan-specific single transmembrane domain protein designated EMRE ("essential MCU regulator"). In its absence, uniporter channel activity was lost despite intact MCU expression and oligomerization. EMRE was required for the interaction of MCU with MICU1 and MICU2. Hence, EMRE is essential for in vivo uniporter current and addnl. bridges the calcium-sensing role of MICU1 and MICU2 with the calcium-conducting role of MCU.
- 13Perocchi, F.; Gohil, V. M.; Girgis, H. S.; Bao, X. R.; McCombs, J. E.; Palmer, A. E.; Mootha, V. K. MICU1 Encodes a Mitochondrial EF Hand Protein Required for Ca2+ uptake. Nature 2010, 467, 291– 296, DOI: 10.1038/nature09358There is no corresponding record for this reference.
- 14Plovanich, M.; Bogorad, R. L.; Sancak, Y.; Kamer, K. J.; Strittmatter, L.; Li, A. A.; Girgis, H. S.; Kuchimanchi, S.; De Groot, J.; Speciner, L.; Taneja, N.; OShea, J.; Koteliansky, V.; Mootha, V. K. MICU2, a Paralog of MICU1, Resides within the Mitochondrial Uniporter Complex to Regulate Calcium Handling. PLoS One 2013, 8, e55785, DOI: 10.1371/journal.pone.0055785There is no corresponding record for this reference.
- 15Csordás, G.; Golenár, T.; Seifert, E. L.; Kamer, K. J.; Sancak, Y.; Perocchi, F.; Moffat, C.; Weaver, D.; De la Fuente, S.; Bogorad, R.; Koteliansky, V.; Adijanto, J.; Mootha, V. K.; Hajnóczky, G. MICU1 Controls Both the Threshold and Cooperative Activation of the Mitochondrial Ca2+ Uniporter. Cell Metab. 2013, 17, 976– 987, DOI: 10.1016/j.cmet.2013.04.02015https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXptV2ks7o%253D&md5=1c83a13cd426d596daed8be670b8318cMICU1 Controls Both the Threshold and Cooperative Activation of the Mitochondrial Ca2+ UniporterCsordas, Gyorgy; Golenar, Tunde; Seifert, Erin L.; Kamer, Kimberli J.; Sancak, Yasemin; Perocchi, Fabiana; Moffat, Cynthia; Weaver, David; Perez, Sergio de la Fuente; Bogorad, Roman; Koteliansky, Victor; Adijanto, Jeffrey; Mootha, Vamsi K.; Hajnoczky, GyorgyCell Metabolism (2013), 17 (6), 976-987CODEN: CMEEB5; ISSN:1550-4131. (Elsevier Inc.)Mitochondrial Ca2+ uptake via the uniporter is central to cell metab., signaling, and survival. Recent studies identified MCU as the uniporter's likely pore and MICU1, an EF-hand protein, as its crit. regulator. How this complex decodes dynamic cytoplasmic [Ca2+] ([Ca2+]c) signals, to tune out small [Ca2+]c increases yet permit pulse transmission, remains unknown. We report that loss of MICU1 in mouse liver and cultured cells causes mitochondrial Ca2+ accumulation during small [Ca2+]c elevations but an attenuated response to agonist-induced [Ca2+]c pulses. The latter reflects loss of pos. cooperativity, likely via the EF-hands. MICU1 faces the intermembrane space and responds to [Ca2+]c changes. Prolonged MICU1 loss leads to an adaptive increase in matrix Ca2+ binding, yet cells show impaired oxidative metab. and sensitization to Ca2+ overload. Collectively, the data indicate that MICU1 senses the [Ca2+]c to establish the uniporter's threshold and gain, thereby allowing mitochondria to properly decode different inputs.
- 16Mallilankaraman, K.; Cárdenas, C.; Doonan, P. J.; Chandramoorthy, H. C.; Irrinki, K. M.; Golenár, T.; Csordás, G.; Madireddi, P.; Yang, J.; Müller, M.; Miller, R.; Kolesar, J. E.; Molgó, J.; Kaufman, B.; Hajnóczky, G.; Foskett, J. K.; Madesh, M. MCUR1 is an Essential Component of Mitochondrial Ca2+ Uptake that Regulates Cellular Metabolism. Nat. Cell Biol. 2012, 14, 1336– 1343, DOI: 10.1038/ncb262216https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhslagsbzO&md5=43a3b38905a4498473229cdf34d3dd1cMCUR1 is an essential component of mitochondrial Ca2+ uptake that regulates cellular metabolismMallilankaraman, Karthik; Cardenas, Cesar; Doonan, Patrick J.; Chandramoorthy, Harish C.; Irrinki, Krishna M.; Golenar, Tuende; Csordas, Gyoergy; Madireddi, Priyanka; Yang, Jun; Mueller, Marioly; Miller, Russell; Kolesar, Jill E.; Molgo, Jordi; Kaufman, Brett; Hajnoczky, Gyoergy; Foskett, J. Kevin; Madesh, MuniswamyNature Cell Biology (2012), 14 (12), 1336-1343CODEN: NCBIFN; ISSN:1465-7392. (Nature Publishing Group)Ca2+ flux across the mitochondrial inner membrane regulates bioenergetics, cytoplasmic Ca2+ signals and activation of cell death pathways. Mitochondrial Ca2+ uptake occurs at regions of close apposition with intracellular Ca2+ release sites, driven by the inner membrane voltage generated by oxidative phosphorylation and mediated by a Ca2+ selective ion channel (MiCa; ref. ) called the uniporter whose complete mol. identity remains unknown. Mitochondrial calcium uniporter (MCU) was recently identified as the likely ion-conducting pore. In addn., MICU1 was identified as a mitochondrial regulator of uniporter-mediated Ca2+ uptake in HeLa cells. Here we identified CCDC90A, hereafter referred to as MCUR1 (mitochondrial calcium uniporter regulator 1), an integral membrane protein required for MCU-dependent mitochondrial Ca2+ uptake. MCUR1 binds to MCU and regulates ruthenium-red-sensitive MCU-dependent Ca2+ uptake. MCUR1 knockdown does not alter MCU localization, but abrogates Ca2+ uptake by energized mitochondria in intact and permeabilized cells. Ablation of MCUR1 disrupts oxidative phosphorylation, lowers cellular ATP and activates AMP kinase-dependent pro-survival autophagy. Thus, MCUR1 is a crit. component of a mitochondrial uniporter channel complex required for mitochondrial Ca2+ uptake and maintenance of normal cellular bioenergetics.
- 17Patron, M.; Checchetto, V.; Raffaello, A.; Teardo, E.; VecellioReane, D.; Mantoan, M.; Granatiero, V.; Szabò, I.; DeStefani, D.; Rizzuto, R. MICU1 and MICU2 Finely Tune the Mitochondrial Ca2+ Uniporter by Exerting Opposite Effects on MCU Activity. Mol. Cell 2014, 53, 726– 737, DOI: 10.1016/j.molcel.2014.01.01317https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXivFCitr0%253D&md5=56712d0f63ab7ad878a37055a9ae18c3MICU1 and MICU2 Finely Tune the Mitochondrial Ca2+ Uniporter by Exerting Opposite Effects on MCU ActivityPatron, Maria; Checchetto, Vanessa; Raffaello, Anna; Teardo, Enrico; Vecellio Reane, Denis; Mantoan, Maura; Granatiero, Veronica; Szabo, Ildiko; De Stefani, Diego; Rizzuto, RosarioMolecular Cell (2014), 53 (5), 726-737CODEN: MOCEFL; ISSN:1097-2765. (Elsevier Inc.)Mitochondrial calcium accumulation was recently shown to depend on a complex composed of an inner-membrane channel (MCU and MCUb) and regulatory subunits (MICU1, MCUR1, and EMRE). A fundamental property of MCU is low activity at resting cytosolic Ca2+ concns., preventing deleterious Ca2+ cycling and organelle overload. Here we demonstrate that these properties are ensured by a regulatory heterodimer composed of two proteins with opposite effects - MICU1 and MICU2 - which in both purified lipid bilayers and in intact cells stimulate and inhibit MCU activity, resp. Both MICU1 and MICU2 are regulated by calcium through their EF-hand domains, thus accounting for the sigmoidal response of MCU to [Ca2+] in situ and allowing tight physiol. control. At low [Ca2+], the dominant effect of MICU2 largely shuts down MCU activity; at higher [Ca2+], the stimulatory effect of MICU1 allows the prompt response of mitochondria to Ca2+ signals generated in the cytoplasm.
- 18Oxenoid, K.; Dong, Y.; Cao, C.; Cui, T.; Sancak, Y.; Markhard, A. L.; Grabarek, Z.; Kong, L.; Liu, Z.; Ouyang, B.; Cong, Y.; Mootha, V. K.; Chou, J. J. Architecture of the Mitochondrial Calcium Uniporter. Nature 2016, 533, 269– 273, DOI: 10.1038/nature1765618https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XntVCksrk%253D&md5=0a977baa9b661326236a9914eccf760cArchitecture of the mitochondrial calcium uniporterOxenoid, Kirill; Dong, Ying; Cao, Chan; Cui, Tanxing; Sancak, Yasemin; Markhard, Andrew L.; Grabarek, Zenon; Kong, Liangliang; Liu, Zhijun; Ouyang, Bo; Cong, Yao; Mootha, Vamsi K.; Chou, James J.Nature (London, United Kingdom) (2016), 533 (7602), 269-273CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Mitochondria from many eukaryotic clades take up large amts. of Ca2+ via an inner membrane transporter called the mitochondrial calcium uniporter (MCU). Transport by MCU is membrane potential-dependent and sensitive to ruthenium red or its deriv., Ru360. Electrophysiol. studies have shown that MCU is an ion channel with remarkably high conductance and selectivity. Ca2+ entry into mitochondria is also known to activate the tricarboxylic acid cycle and seems to be crucial for matching the prodn. of ATP in mitochondria with its cytosolic demand. MCU is the pore-forming and Ca2+-conducting subunit of the uniporter holocomplex, but its primary sequence does not resemble any Ca2+ channel studied to date. Here, the authors report the structure of the pore domain of MCU from Caenorhabditis elegans, detd. using NMR spectroscopy and electron microscopy (EM). MCU is a homo-oligomer in which the 2nd transmembrane helix forms a hydrophilic pore across the membrane. The channel assembly represented a new soln. of ion channel architecture, and was stabilized by a coiled-coil motif protruding into the mitochondrial matrix. The crit. DXXE motif formed the pore entrance, which featured 2 carboxylate rings; based on the ring dimensions and functional mutagenesis, these rings appeared to form the selectivity filter. To the authors' knowledge, this is one of the largest membrane protein structures characterized by NMR, and provides a structural blueprint for understanding the function of this channel.
- 19Bick, A. G.; Calvo, S. E.; Mootha, V. K. Evolutionary Diversity of the Mitochondrial Calcium Uniporter. Science 2012, 336, 886, DOI: 10.1126/science.1214977There is no corresponding record for this reference.
- 20Baradaran, R.; Wang, C.; Siliciano, A. F.; Long, S. B. Cryo-EM Structures of Fungal and Metazoan Mitochondrial Calcium Uniporters. Nature 2018, 559, 580– 584, DOI: 10.1038/s41586-018-0331-820https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlWjsbfL&md5=742571dbb000126c2013abefe5963fa6Cryo-EM structures of fungal and metazoan mitochondrial calcium uniportersBaradaran, Rozbeh; Wang, Chongyuan; Siliciano, Andrew Francis; Long, Stephen BarstowNature (London, United Kingdom) (2018), 559 (7715), 580-584CODEN: NATUAS; ISSN:0028-0836. (Nature Research)The mitochondrial calcium uniporter (MCU) is a highly selective calcium channel and a major route of calcium entry into mitochondria. How the channel catalyzes ion permeation and achieves ion selectivity are not well understood, partly because MCU is thought to have a distinct architecture in comparison to other cellular channels. Here we report cryo-electron microscopy reconstructions of MCU channels from zebrafish and Cyphellophora europaea at 8.5 Å and 3.2 Å resolns., resp. In contrast to a previous report of pentameric stoichiometry for MCU, both channels are tetramers. The at. model of C. europaea MCU shows that a conserved WDXXEP signature sequence forms the selectivity filter, in which calcium ions are arranged in single file. Coiled-coil legs connect the pore to N-terminal domains in the mitochondrial matrix. In C. europaea MCU, the N-terminal domains assemble as a dimer of dimers; in zebrafish MCU, they form an asym. crescent. The structures define principles that underlie ion permeation and calcium selectivity in this unusual channel.
- 21Fan, C.; Fan, M.; Orlando, B. J.; Fastman, N. M.; Zhang, J.; Xu, Y.; Chambers, M. G.; Xu, X.; Perry, K.; Liao, M.; Feng, L. X-Ray and Cryo-EM Structures of the Mitochondrial Calcium Uniporter. Nature 2018, 559, 575– 579, DOI: 10.1038/s41586-018-0330-921https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlWjtr7M&md5=2293cca6a52861da2fc998e1c00af7d5X-ray and cryo-EM structures of the mitochondrial calcium uniporterFan, Chao; Fan, Minrui; Orlando, Benjamin J.; Fastman, Nathan M.; Zhang, Jinru; Xu, Yan; Chambers, Melissa G.; Xu, Xiaofang; Perry, Kay; Liao, Maofu; Feng, LiangNature (London, United Kingdom) (2018), 559 (7715), 575-579CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Mitochondrial Ca2+ uptake is crit. for regulating ATP prodn., intracellular Ca2+ signaling, and cell death. This uptake is mediated by a highly selective Ca2+ channel called the mitochondrial calcium uniporter (MCU). Here, we detd. the structures of the pore-forming MCU proteins from 2 fungi (Fusarium graminearum and Metarhizium acridum) by x-ray crystallog. and single-particle cryo-electron microscopy (cryo-EM). The stoichiometry, overall architecture, and individual subunit structure differed markedly from those described in the recent NMR structure of Caenorhabditis elegans MCU. We obsd. a dimer-of-dimer architecture across species and chem. environments, which was corroborated by biochem. expts. Structural analyses and functional characterization uncovered the roles of key residues in the pore. These results revealed a new ion channel architecture, provide insights into Ca2+ coordination, selectivity, and conduction, and established a structural framework for understanding the mechanism of MCU function.
- 22Nguyen, N. X.; Armache, J. P.; Lee, C.; Yang, Y.; Zeng, W.; Mootha, V. K.; Cheng, Y.; Bai, X.; Jiang, Y. Cryo-EM Structure of a Fungal Mitochondrial Calcium Uniporter. Nature 2018, 559, 570– 574, DOI: 10.1038/s41586-018-0333-622https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlWjtr7N&md5=43d5c4d30e00ba77b51bdeeba408cf6cCryo-EM structure of a fungal mitochondrial calcium uniporterNguyen, Nam X.; Armache, Jean-Paul; Lee, Changkeun; Yang, Yi; Zeng, Weizhong; Mootha, Vamsi K.; Cheng, Yifan; Bai, Xiao-chen; Jiang, YouxingNature (London, United Kingdom) (2018), 559 (7715), 570-574CODEN: NATUAS; ISSN:0028-0836. (Nature Research)The mitochondrial calcium uniporter (MCU) is a highly selective calcium channel localized to the inner mitochondrial membrane. Here, we describe the structure of an MCU ortholog from the fungus Neosartorya fischeri (NfMCU) detd. to 3.8 Å resoln. by phase-plate cryo-electron microscopy. The channel is a homotetramer with two-fold symmetry in its amino-terminal domain (NTD) that adopts a similar structure to that of human MCU. The NTD assembles as a dimer of dimers to form a tetrameric ring that connects to the transmembrane domain through an elongated coiled-coil domain. The ion-conducting pore domain maintains four-fold symmetry, with the selectivity filter positioned at the start of the pore-forming TM2 helix. The aspartate and glutamate sidechains of the conserved DIME motif are oriented towards the central axis and sepd. by one helical turn. The structure of NfMCU offers insights into channel assembly, selective calcium permeation, and inhibitor binding.
- 23Yoo, J.; Wu, M.; Yin, Y.; Herzik, M. A.; Lander, G. C.; Lee, S.-Y. Cryo-EM Structure of a Mitochondrial Calcium Uniporter. Science 2018, 361, 506– 511, DOI: 10.1126/science.aar405623https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsVahtr%252FE&md5=2072ca08fe5036a5b3749a4073a7e902Cryo-EM structure of a mitochondrial calcium uniporterYoo, Jiho; Wu, Mengyu; Yin, Ying; Herzik, Mark A., Jr; Lander, Gabriel C.; Lee, Seok-YongScience (Washington, DC, United States) (2018), 361 (6401), 506-511CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Calcium transport plays an important role in regulating mitochondrial physiol. and pathophysiol. The mitochondrial calcium uniporter (MCU) is a calcium-selective ion channel that is the primary mediator for calcium uptake into the mitochondrial matrix. Here, we present the cryo-electron microscopy structure of the full-length MCU from Neurospora crassa to an overall resoln. of ∼3.7 angstroms. Our structure reveals a tetrameric architecture, with the sol. and transmembrane domains adopting different sym. arrangements within the channel. The conserved W-D-Φ-Φ-E-P-V-T-Y sequence motif of MCU pore forms a selectivity filter comprising two acidic rings sepd. by one helical turn along the central axis of the channel pore. The structure combined with mutagenesis gives insight into the basis of calcium recognition.
- 24Hoffman, N. E.; Chandramoorthy, H. C.; Shamugapriya, S.; Zhang, X.; Rajan, S.; Mallilankaraman, K.; Gandhirajan, R. K.; Vagnozzi, R. J.; Ferrer, L. M.; Sreekrishnanilayam, K.; Natarajaseenivasan, K.; Vallem, S.; Force, T.; Choi, E. T.; Cheung, J. Y.; Madesh, M. MICU1 Motifs Define Mitochondrial Calcium Uniporter Binding and Activity. Cell Rep. 2013, 5, 1576– 1588, DOI: 10.1016/j.celrep.2013.11.02624https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvFOhtrbK&md5=7333ca5d17d6d2e112a240a4d3949d80MICU1 Motifs Define Mitochondrial Calcium Uniporter Binding and ActivityHoffman, Nicholas E.; Chandramoorthy, Harish C.; Shamugapriya, Santhanam; Zhang, Xueqian; Rajan, Sudarsan; Mallilankaraman, Karthik; Gandhirajan, Rajesh Kumar; Vagnozzi, Ronald J.; Ferrer, Lucas M.; Sreekrishnanilayam, Krishnalatha; Natarajaseenivasan, Kalimuthusamy; Vallem, Sandhya; Force, Thomas; Choi, Eric T.; Cheung, Joseph Y.; Madesh, MuniswamyCell Reports (2013), 5 (6), 1576-1588CODEN: CREED8; ISSN:2211-1247. (Cell Press)Resting mitochondrial matrix Ca2+ is maintained through a mitochondrial calcium uptake 1 (MICU1)-established threshold inhibition of mitochondrial calcium uniporter (MCU) activity. It is not known how MICU1 interacts with MCU to establish this Ca2+ threshold for mitochondrial Ca2+ uptake and MCU activity. Here, we show that MICU1 localizes to the mitochondrial matrix side of the inner mitochondrial membrane and MICU1/MCU binding is detd. by a MICU1 N-terminal polybasic domain and two interacting coiled-coil domains of MCU. Further investigation reveals that MICU1 forms homo-oligomers, and this oligomerization is independent of the polybasic region. However, the polybasic region confers MICU1 oligomeric binding to MCU and controls mitochondrial Ca2+ current (IMCU). Moreover, MICU1 EF hands regulate MCU channel activity, but do not det. MCU binding. Loss of MICU1 promotes MCU activation leading to oxidative burden and a halt to cell migration. These studies establish a mol. mechanism for MICU1 control of MCU-mediated mitochondrial Ca2+ accumulation, and dysregulation of this mechanism probably enhances vascular dysfunction.
- 25Petrungaro, C.; Zimmermann, K. M.; Küttner, V.; Fischer, M.; Dengjel, J.; Bogeski, I.; Riemer, J. The Ca2+-Dependent Release of the Mia40-Induced MICU1-MICU2 Dimer from MCU Regulates Mitochondrial Ca2+ Uptake. Cell Metab. 2015, 22, 721– 733, DOI: 10.1016/j.cmet.2015.08.01925https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFaqsr%252FI&md5=92341726e998629476fc842d3c950665The Ca2+-Dependent Release of the Mia40-Induced MICU1-MICU2 Dimer from MCU Regulates Mitochondrial Ca2+ UptakePetrungaro, Carmelina; Zimmermann, Katharina M.; Kuettner, Victoria; Fischer, Manuel; Dengjel, Joern; Bogeski, Ivan; Riemer, JanCell Metabolism (2015), 22 (4), 721-733CODEN: CMEEB5; ISSN:1550-4131. (Elsevier Inc.)The essential oxidoreductase Mia40/CHCHD4 mediates disulfide bond formation and protein folding in the mitochondrial intermembrane space. Here, we investigated the interactome of Mia40 thereby revealing links between thiol-oxidn. and apoptosis, energy metab., and Ca2+ signaling. Among the interaction partners of Mia40 is MICU1-the regulator of the mitochondrial Ca2+ uniporter (MCU), which transfers Ca2+ across the inner membrane. We examd. the biogenesis of MICU1 and find that Mia40 introduces an intermol. disulfide bond that links MICU1 and its inhibitory paralog MICU2 in a heterodimer. Absence of this disulfide bond results in increased receptor-induced mitochondrial Ca2+ uptake. In the presence of the disulfide bond, MICU1-MICU2 heterodimer binding to MCU is controlled by Ca2+ levels: the dimer assocs. with MCU at low levels of Ca2+ and dissocs. upon high Ca2+ concns. Our findings support a model in which mitochondrial Ca2+ uptake is regulated by a Ca2+-dependent remodeling of the uniporter complex.
- 26Lee, Y.; Min, C. K.; Kim, T. G.; Song, H. K.; Lim, Y.; Kim, D.; Shin, K.; Kang, M.; Kang, J. Y.; Youn, H.-S.; Lee, J.-G.; An, J. Y.; Park, K. R.; Lim, J. J.; Kim, J. H.; Kim, J. H.; Park, Z. Y.; Kim, Y.-S.; Wang, J.; Kim, D. H.; Eom, S. H. Structure and Function of the N-Terminal Domain of the Human Mitochondrial Calcium Uniporter. EMBO Rep. 2015, 16, 1318– 1333, DOI: 10.15252/embr.20154043626https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsVGntbrJ&md5=4b530d1c88f106b0ed8a5e2103c46e9dStructure and function of the N-terminal domain of the human mitochondrial calcium uniporterLee, Youngjin; Min, Choon Kee; Kim, Tae Gyun; Song, Hong Ki; Lim, Yunki; Kim, Dongwook; Shin, Kahee; Kang, Moonkyung; Kang, Jung Youn; Youn, Hyung-Seop; Lee, Jung-Gyu; An, Jun Yop; Park, Kyoung Ryoung; Lim, Jia Jia; Kim, Ji Hun; Kim, Ji Hye; Park, Zee Yong; Kim, Yeon-Soo; Wang, Jimin; Kim, Do Han; Eom, Soo HyunEMBO Reports (2015), 16 (10), 1318-1333CODEN: ERMEAX; ISSN:1469-221X. (Wiley-VCH Verlag GmbH & Co. KGaA)The mitochondrial calcium uniporter (MCU) is responsible for mitochondrial calcium uptake and homeostasis. It is also a target for the regulation of cellular anti-/pro-apoptosis and necrosis by several oncogenes and tumor suppressors. Herein, we report the crystal structure of the MCU N-terminal domain (NTD) at a resoln. of 1.50 Å in a novel fold and the S92A MCU mutant at 2.75 Å resoln.; the residue S92 is a predicted CaMKII phosphorylation site. The assembly of the mitochondrial calcium uniporter complex (uniplex) and the interaction with the MCU regulators such as the mitochondrial calcium uptake-1 and mitochondrial calcium uptake-2 proteins (MICU1 and MICU2) are not affected by the deletion of MCU NTD. However, the expression of the S92A mutant or a NTD deletion mutant failed to restore mitochondrial Ca2+ uptake in a stable MCU knockdown HeLa cell line and exerted dominant-neg. effects in the wild-type MCU-expressing cell line. These results suggest that the NTD of MCU is essential for the modulation of MCU function, although it does not affect the uniplex formation.
- 27Lee, S. K.; Shanmughapriya, S.; Mok, M. C. Y.; Dong, Z.; Tomar, D.; Carvalho, E.; Rajan, S.; Junop, M. S.; Madesh, M.; Stathopulos, P. B. Structural Insights into Mitochondrial Calcium Uniporter Regulation by Divalent Cations. Cell Chem. Biol. 2016, 23, 1157– 1169, DOI: 10.1016/j.chembiol.2016.07.01227https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsVWmtLbL&md5=f414a4d19f961b5c3b040955beb83c6bStructural Insights into Mitochondrial Calcium Uniporter Regulation by Divalent CationsLee, Samuel K.; Shanmughapriya, Santhanam; Mok, Mac C. Y.; Dong, Zhiwei; Tomar, Dhanendra; Carvalho, Edmund; Rajan, Sudarsan; Junop, Murray S.; Madesh, Muniswamy; Stathopulos, Peter B.Cell Chemical Biology (2016), 23 (9), 1157-1169CODEN: CCBEBM; ISSN:2451-9448. (Cell Press)Calcium (Ca2+) flux into the matrix is tightly controlled by the mitochondrial Ca2+ uniporter (MCU) due to vital roles in cell death and bioenergetics. However, the precise at. mechanisms of MCU regulation remain unclear. Here, we solved the crystal structure of the N-terminal matrix domain of human MCU, revealing a β-grasp-like fold with a cluster of neg. charged residues that interacts with divalent cations. Binding of Ca2+ or Mg2+ destabilizes and shifts the self-assocn. equil. of the domain toward monomer. Mutational disruption of the acidic face weakens oligomerization of the isolated matrix domain and full-length human protein similar to cation binding and markedly decreases MCU activity. Moreover, mitochondrial Mg2+ loading or blockade of mitochondrial Ca2+ extrusion suppresses MCU Ca2+-uptake rates. Collectively, our data reveal that the β-grasp-like matrix region harbors an MCU-regulating acidic patch that inhibits human MCU activity in response to Mg2+ and Ca2+ binding.
- 28Tomar, D.; Dong, Z.; Shanmughapriya, S.; Koch, D. A.; Thomas, T.; Hoffman, N. E.; Timbalia, S. A.; Goldman, S. J.; Breves, S. L.; Corbally, D. P.; Nemani, N.; Fairweather, J. P.; Cutri, A. R.; Zhang, X.; Song, J.; Jaña, F.; Huang, J.; Barrero, C.; Rabinowitz, J. E.; Luongo, T. S.; Schumacher, S. M.; Rockman, M. E.; Dietrich, A.; Merali, S.; Caplan, J.; Stathopulos, P. B.; Ahima, R. S.; Cheung, J. Y.; Houser, S. R.; Koch, W. J.; Patel, V.; Gohil, V. M.; Elrod, J. W.; Rajan, S.; Madesh, M. MCUR1 is a Scaffold Factor for the MCU Complex Function and Promotes Mitochondrial Bioenergetics. Cell Rep. 2016, 15, 1673– 1685, DOI: 10.1016/j.celrep.2016.04.05028https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XnvV2rs7Y%253D&md5=e38a25d3096ac38e15b8a34e3a61f6d7MCUR1 Is a Scaffold Factor for the MCU Complex Function and Promotes Mitochondrial BioenergeticsTomar, Dhanendra; Dong, Zhiwei; Shanmughapriya, Santhanam; Koch, Diana A.; Thomas, Toby; Hoffman, Nicholas E.; Timbalia, Shrishiv A.; Goldman, Samuel J.; Breves, Sarah L.; Corbally, Daniel P.; Nemani, Neeharika; Fairweather, Joseph P.; Cutri, Allison R.; Zhang, Xueqian; Song, Jianliang; Jana, Fabian; Huang, Jianhe; Barrero, Carlos; Rabinowitz, Joseph E.; Luongo, Timothy S.; Schumacher, Sarah M.; Rockman, Michael E.; Dietrich, Alexander; Merali, Salim; Caplan, Jeffrey; Stathopulos, Peter; Ahima, Rexford S.; Cheung, Joseph Y.; Houser, Steven R.; Koch, Walter J.; Patel, Vickas; Gohil, Vishal M.; Elrod, John W.; Rajan, Sudarsan; Madesh, MuniswamyCell Reports (2016), 15 (8), 1673-1685CODEN: CREED8; ISSN:2211-1247. (Cell Press)Mitochondrial Ca2+ Uniporter (MCU)-dependent mitochondrial Ca2+ uptake is the primary mechanism for increasing matrix Ca2+ in most cell types. However, a limited understanding of the MCU complex assembly impedes the comprehension of the precise mechanisms underlying MCU activity. Here, we report that mouse cardiomyocytes and endothelial cells lacking MCU regulator 1 (MCUR1) have severely impaired [Ca2+]m uptake and IMCU current. MCUR1 binds to MCU and EMRE and function as a scaffold factor. Our protein binding analyses identified the minimal, highly conserved regions of coiled-coil domain of both MCU and MCUR1 that are necessary for heterooligomeric complex formation. Loss of MCUR1 perturbed MCU heterooligomeric complex and functions as a scaffold factor for the assembly of MCU complex. Vascular endothelial deletion of MCU and MCUR1 impaired mitochondrial bioenergetics, cell proliferation, and migration but elicited autophagy. These studies establish the existence of a MCU complex that assembles at the mitochondrial integral membrane and regulates Ca2+-dependent mitochondrial metab.
- 29Gunter, T. E.; Gunter, K. K.; Sheu, S.-S.; Gavin, C. E. Mitochondrial Calcium Transport: Physiological and Pathological Relevance. Am. J. Physiol. 1994, 267, C313– 319, DOI: 10.1152/ajpcell.1994.267.2.C313There is no corresponding record for this reference.
- 30Hajnóczky, G.; Robb-Gaspers, L. D.; Seitz, M. B.; Thomas, A. P. Decoding of Cytosolic Calcium Oscillations in the Mitochondria. Cell 1995, 82, 415– 424, DOI: 10.1016/0092-8674(95)90430-1There is no corresponding record for this reference.
- 31Denton, R.; McCormack, J. G. Ca2+ as a Second Messenger Within Mitochondria Of The Heart And Other Tissues. Annu. Rev. Physiol. 1990, 52, 451– 466, DOI: 10.1146/annurev.ph.52.030190.002315There is no corresponding record for this reference.
- 32Santo-Domingo, J.; Demaurex, N. Calcium Uptake Mechanisms of Mitochondria. Biochim. Biophys. Acta, Bioenerg. 2010, 1797, 907– 912, DOI: 10.1016/j.bbabio.2010.01.00532https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXnvV2qtr4%253D&md5=b7a5831a8d832ac99c2a3854f647b76aCalcium uptake mechanisms of mitochondriaSanto-Domingo, Jaime; Demaurex, NicolasBiochimica et Biophysica Acta, Bioenergetics (2010), 1797 (6-7), 907-912CODEN: BBBEB4; ISSN:0005-2728. (Elsevier B. V.)A review. The ability of mitochondria to capture Ca2+ ions has important functional implications for cells, because mitochondria shape cellular Ca2+ signals by acting as a Ca2+ buffer and respond to Ca2+ elevations either by increasing the cell energy supply or by triggering the cell death program of apoptosis. A mitochondrial Ca2+ channel known as the uniporter drives the rapid and massive entry of Ca2+ ions into mitochondria. The uniporter operates at high, micromolar cytosolic Ca2+ concns. that are only reached transiently in cells, near Ca2+ release channels. Mitochondria can also take up Ca2+ at low, nanomolar concns., but this high affinity mode of Ca2+ uptake is not well characterized. Recently, the leucine zipper-EF hand-contg. transmembrane region (Letm1) was proposed to be an electrogenic 1:1 mitochondrial Ca2+/H+ antiporter that drives the uptake of Ca2+ into mitochondria at nanomolar cytosolic Ca2+ concns. Here, the authors review the properties of the Ca2+ import systems of mitochondria and discuss how Ca2+ uptake via an electrogenic 1:1 Ca2+/H+ antiport challenges the current thinking of the mitochondrial Ca2+ uptake mechanism.
- 33Bernardi, P. Mitochondrial Transport of Cations: Channels, Exchangers, and Permeability Transition. Physiol. Rev. 1999, 79, 1127– 1155, DOI: 10.1152/physrev.1999.79.4.112733https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXmvVymtLw%253D&md5=594fe4b9901aee10ba77bf8164865822Mitochondrial transport of cations: channels, exchangers, and permeability transitionBernardi, PaoloPhysiological Reviews (1999), 79 (4), 1127-1155CODEN: PHREA7; ISSN:0031-9333. (American Physiological Society)A review with 372 refs. This review provides a selective history of how studies of mitochondrial cation transport (K+, Na+, Ca2+) developed in relation to the major themes of research in bioenergetics. It then covers in some detail specific transport pathways for these cations, and it introduces and discusses open problems about their nature and physiol. function, particularly in relation to vol. regulation and Ca2+ homeostasis. The review should provide the basic elements needed to understand both earlier mitochondrial literature and current problems assocd. with mitochondrial transport of cations and hopefully will foster new interest in the mol. definition of mitochondrial cation channels and exchangers as well as their roles in cell physiol.
- 34Shanmughapriya, S.; Rajan, S.; Hoffman, N. E.; Higgins, A. M.; Tomar, D.; Nemani, N.; Hines, K. J.; Smith, D. J.; Eguchi, A.; Vallem, S.; Shaikh, F.; Cheung, M.; Leonard, N. J.; Stolakis, R. S.; Wolfers, M. P.; Ibetti, J.; Chuprun, J. K.; Jog, N. R.; Houser, S. R.; Koch, W. J.; Elrod, J. W.; Madesh, M. SPG7 is an Essential and Conserved Component of the Mitochondrial Permeability Transition Pore. Mol. Cell 2015, 60, 47– 62, DOI: 10.1016/j.molcel.2015.08.009There is no corresponding record for this reference.
- 35Orrenius, S.; Zhivotovsky, B.; Nicotera, P. Regulation of Cell Death: The Calcium-Apoptosis Link. Nat. Rev. Mol. Cell Biol. 2003, 4, 552– 565, DOI: 10.1038/nrm115035https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXltVWnu7g%253D&md5=6efb194177308e68f3d9e3545d7095f6Calcium: Regulation of cell death: the calcium-apoptosis linkOrrenius, Sten; Zhivotovsky, Boris; Nicotera, PierluigiNature Reviews Molecular Cell Biology (2003), 4 (7), 552-565CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review and discussion. To live or to die. This crucial question eloquently reflects the dual role of Ca2+ in living organisms, as a survival factor or as a ruthless killer. It has long been known that Ca2+ signals govern a host of vital cell functions and so are necessary for cell survival. However, more recently it has become clear that cellular Ca2+ overload, or perturbation of intracellular Ca2+ compartmentalization, can cause cytotoxicity and trigger either apoptotic or necrotic cell death.
- 36De Stefani, D.; Rizzuto, R.; Pozzan, T. Enjoy the Trip: Calcium in Mitochondria Back and Forth. Annu. Rev. Biochem. 2016, 85, 161– 192, DOI: 10.1146/annurev-biochem-060614-03421636https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XntlKgsbs%253D&md5=5ce87bb458bf96a84be012e840b27580Enjoy the Trip: Calcium in Mitochondria Back and ForthDe Stefani, Diego; Rizzuto, Rosario; Pozzan, TullioAnnual Review of Biochemistry (2016), 85 (), 161-192CODEN: ARBOAW; ISSN:0066-4154. (Annual Reviews)A review. In the last 5 years, most of the mols. that control mitochondrial Ca2+ homeostasis have been finally identified. Mitochondrial Ca2+ uptake is mediated by the Mitochondrial Calcium Uniporter (MCU) complex, a macromol. structure that guarantees Ca2+ accumulation inside mitochondrial matrix upon increases in cytosolic Ca2+. Conversely, Ca2+ release is under the control of the Na+/Ca2+ exchanger, encoded by the NCLX gene, and of a H+/Ca2+ antiporter, whose identity is still debated. The low affinity of the MCU complex, coupled to the activity of the efflux systems, protects cells from continuous futile cycles of Ca2+ across the inner mitochondrial membrane and consequent massive energy dissipation. In this review, we discuss the basic principles that govern mitochondrial Ca2+ homeostasis and the methods used to investigate the dynamics of Ca2+ concn. within the organelles. We discuss the functional and structural role of the different mols. involved in mitochondrial Ca2+ handling and their pathophysiol. role.
- 37Marchi, S.; Pinton, P. The Mitochondrial Calcium Uniporter Complex: Molecular Components, Structure and Physiopathological Implications. J. Physiol. 2014, 592, 829– 839, DOI: 10.1113/jphysiol.2013.26823537https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXjtlOkuro%253D&md5=5f8c78976edf6e35e484a9a40e107784The mitochondrial calcium uniporter complex: molecular components, structure and physiopathological implicationsMarchi, Saverio; Pinton, PaoloJournal of Physiology (Oxford, United Kingdom) (2014), 592 (5), 829-839CODEN: JPHYA7; ISSN:0022-3751. (Wiley-Blackwell)A review. Although it has long been known that mitochondria take up Ca2+, the mol. identities of the channels and transporters involved in this process were revealed only recently. Here, we discuss the recent work that has led to the characterization of the mitochondrial calcium uniporter complex, which includes the channel-forming subunit MCU (mitochondrial calcium uniporter) and its regulators MICU1, MICU2, MCUb, EMRE, MCUR1 and miR-25. We review not only the biochem. identities and structures of the proteins required for mitochondrial Ca2+ uptake but also their implications in different physiopathol. contexts.
- 38Luongo, T. S.; Lambert, J. P.; Yuan, A.; Zhang, X.; Gross, P.; Song, J.; Shanmughapriya, S.; Gao, E.; Jain, M.; Houser, S. R.; Koch, W. J.; Cheung, J. Y.; Madesh, M.; Elrod, J. W. The Mitochondrial Calcium Uniporter Matches Energetic Supply with Cardiac Workload during Stress and Modulates Permeability Transition. Cell Rep. 2015, 12, 23– 34, DOI: 10.1016/j.celrep.2015.06.01738https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtV2itbbP&md5=35f1feedc69b60ede6620f6722a35fbcThe Mitochondrial Calcium Uniporter Matches Energetic Supply with Cardiac Workload during Stress and Modulates Permeability TransitionLuongo, Timothy S.; Lambert, Jonathan P.; Yuan, Ancai; Zhang, Xueqian; Gross, Polina; Song, Jianliang; Shanmughapriya, Santhanam; Gao, Erhe; Jain, Mohit; Houser, Steven R.; Koch, Walter J.; Cheung, Joseph Y.; Madesh, Muniswamy; Elrod, John W.Cell Reports (2015), 12 (1), 23-34CODEN: CREED8; ISSN:2211-1247. (Cell Press)Cardiac contractility is mediated by a variable flux in intracellular calcium (Ca2+), thought to be integrated into mitochondria via the mitochondrial calcium uniporter (MCU) channel to match energetic demand. Here, we examine a conditional, cardiomyocyte-specific, mutant mouse lacking Mcu, the pore-forming subunit of the MCU channel, in adulthood. Mcu-/- mice display no overt baseline phenotype and are protected against mCa2+ overload in an in vivo myocardial ischemia-reperfusion injury model by preventing the activation of the mitochondrial permeability transition pore, decreasing infarct size, and preserving cardiac function. In addn., we find that Mcu-/- mice lack contractile responsiveness to acute β-adrenergic receptor stimulation and in parallel are unable to activate mitochondrial dehydrogenases and display reduced bioenergetic reserve capacity. These results support the hypothesis that MCU may be dispensable for homeostatic cardiac function but required to modulate Ca2+-dependent metab. during acute stress.
- 39Kwong, J. Q.; Lu, X.; Correll, R. N.; Schwanekamp, J. A.; Vagnozzi, R. J.; Sargent, M. A.; York, A. J.; Zhang, J.; Bers, D. M.; Molkentin, J. D. The Mitochondrial Calcium Uniporter Selectively Matches Metabolic Output to Acute Contractile Stress in the Heart. Cell Rep. 2015, 12, 15– 22, DOI: 10.1016/j.celrep.2015.06.00239https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtV2itbnM&md5=18e145fd5c845366c5ef24b91b079451The Mitochondrial Calcium Uniporter Selectively Matches Metabolic Output to Acute Contractile Stress in the HeartKwong, Jennifer Q.; Lu, Xiyuan; Correll, Robert N.; Schwanekamp, Jennifer A.; Vagnozzi, Ronald J.; Sargent, Michelle A.; York, Allen J.; Zhang, Jianyi; Bers, Donald M.; Molkentin, Jeffery D.Cell Reports (2015), 12 (1), 15-22CODEN: CREED8; ISSN:2211-1247. (Cell Press)In the heart, augmented Ca2+ fluxing drives contractility and ATP generation through mitochondrial Ca2+ loading. Pathol. mitochondrial Ca2+ overload with ischemic injury triggers mitochondrial permeability transition pore (MPTP) opening and cardiomyocyte death. Mitochondrial Ca2+ uptake is primarily mediated by the mitochondrial Ca2+ uniporter (MCU). Here, we generated mice with adult and cardiomyocyte-specific deletion of Mcu, which produced mitochondria refractory to acute Ca2+ uptake, with impaired ATP prodn., and inhibited MPTP opening upon acute Ca2+ challenge. Mice lacking Mcu in the adult heart were also protected from acute ischemia-reperfusion injury. However, resting/basal mitochondrial Ca2+ levels were normal in hearts of Mcu-deleted mice, and mitochondria lacking MCU eventually loaded with Ca2+ after stress stimulation. Indeed, Mcu-deleted mice were unable to immediately sprint on a treadmill unless warmed up for 30 min. Hence, MCU is a dedicated regulator of short-term mitochondrial Ca2+ loading underlying a "fight-or-flight" response that acutely matches cardiac workload with ATP prodn.
- 40Medvedeva, Y. V.; Weiss, J. H.; Weiss, J. H.; Medvedeva, Y. V. Intramitochondrial Zn2+ Accumulation via the Ca2+ Uniporter Contributes to Acute Ischemic Neurodegeneration. Neurobiol. Dis. 2014, 68, 137– 144, DOI: 10.1016/j.nbd.2014.04.01140https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtVWrtLfK&md5=695cd0a52d3fab9addb4805e0da8af60Intramitochondrial Zn2 + accumulation via the Ca2 + uniporter contributes to acute ischemic neurodegenerationMedvedeva, Yuliya V.; Weiss, John H.Neurobiology of Disease (2014), 68 (), 137-144CODEN: NUDIEM; ISSN:0969-9961. (Elsevier Inc.)Ca2 + and Zn2 + have both been implicated in the induction of acute ischemic neurodegeneration. We recently examd. changes in intracellular Zn2 + and Ca2 + in CA1 pyramidal neurons subjected to oxygen glucose deprivation (OGD), and found that Zn2 + rises precede and contribute to the onset of terminal Ca2 + rises ("Ca2 + deregulation"), which are causatively linked to a lethal loss of membrane integrity. The present study seeks to examine the specific role of intramitochondrial Zn2 + accumulation in ischemic injury, using blockers of the mitochondrial Ca2 + uniporter (MCU), through which both Zn2 + and Ca2 + appear able to enter the mitochondrial matrix. In physiol. extracellular Ca2 +, treatment with the MCU blocker, Ruthenium Red (RR), accelerated the Ca2 + deregulation, most likely by disrupting mitochondrial Ca2 + buffering and thus accelerating the lethal cytosolic Ca2 + overload. However, when intracellular Ca2 + overload was slowed, either by adding blockers of major Ca2 + entry channels or by lowering the concn. of Ca2 + in the extracellular buffer, Ca2 + deregulation was delayed, and under these conditions either Zn2 + chelation or MCU blockade resulted in similar further delays of the Ca2 + deregulation. In parallel studies using the reactive oxygen species (ROS) indicator, hydroethidine, lowering Ca2 + surprisingly accelerated OGD induced ROS generation, and in these low Ca2 + conditions, either Zn2 + chelation or MCU block slowed the ROS generation. These studies suggest that, during acute ischemia, Zn2 + entry into mitochondria via the MCU induces mitochondrial dysfunction (including ROS generation) that occurs upstream of, and contributes to the terminal Ca2 + deregulation.
- 41Giorgi, C.; Agnoletto, C.; Bononi, A.; Bonora, M.; De Marchi, E.; Marchi, S.; Missiroli, S.; Patergnani, S.; Poletti, F.; Rimessi, A.; Suski, J. M.; Wieckowski, M. R.; Pinton, P. Mitochondrial Calcium Homeostasis as Potential Target for Mitochondrial Medicine. Mitochondrion 2012, 12, 77– 85, DOI: 10.1016/j.mito.2011.07.00441https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhvVOltrc%253D&md5=32b483cd42cd9ee7c1f0db4187eb0bceMitochondrial calcium homeostasis as potential target for mitochondrial medicineGiorgi, Carlotta; Agnoletto, Chiara; Bononi, Angela; Bonora, Massimo; De Marchi, Elena; Marchi, Saverio; Missiroli, Sonia; Patergnani, Simone; Poletti, Federica; Rimessi, Alessandro; Suski, Jan M.; Wieckowski, Mariusz R.; Pinton, PaoloMitochondrion (2012), 12 (1), 77-85CODEN: MITOCN; ISSN:1567-7249. (Elsevier B.V.)A review. Mitochondria are crucial in different intracellular pathways of signal transduction. Mitochondria are capable of decoding a variety of extracellular stimuli into markedly different intracellular actions, ranging from energy prodn. to cell death. The fine modulation of mitochondrial calcium (Ca2+) homeostasis plays a fundamental role in many of the processes involving this organelle. When mitochondrial Ca2+ homeostasis is compromised, different pathol. conditions can occur, depending on the cell type involved. Recent data have shed light on the mol. identity of the main proteins involved in the handling of mitochondrial Ca2+ traffic, opening fascinating and ambitious new avenues for mitochondria-based pharmacol. strategies.
- 42Arduino, D. M.; Wettmarshausen, J.; Vais, H.; Navas-Navarro, P.; Cheng, Y.; Leimpek, A.; Ma, Z.; Delrio-Lorenzo, A.; Giordano, A.; Garcia-Perez, C.; Médard, G.; Kuster, B.; García-Sancho, J.; Mokranjac, D.; Foskett, J. K.; Alonso, M. T.; Perocchi, F. Systematic Identification of MCU Modulators by Orthogonal Interspecies Chemical Screening. Mol. Cell 2017, 67, 711– 723, DOI: 10.1016/j.molcel.2017.07.01942https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtlKmsbfO&md5=f9cfb524e127ab132d85c3cc49161dd4Systematic Identification of MCU Modulators by Orthogonal Interspecies Chemical ScreeningArduino, Daniela M.; Wettmarshausen, Jennifer; Vais, Horia; Navas-Navarro, Paloma; Cheng, Yiming; Leimpek, Anja; Ma, Zhongming; Delrio-Lorenzo, Alba; Giordano, Andrea; Garcia-Perez, Cecilia; Medard, Guillaume; Kuster, Bernhard; Garcia-Sancho, Javier; Mokranjac, Dejana; Foskett, J. Kevin; Alonso, M. Teresa; Perocchi, FabianaMolecular Cell (2017), 67 (4), 711-723.e7CODEN: MOCEFL; ISSN:1097-2765. (Elsevier Inc.)The mitochondrial calcium uniporter complex is essential for calcium (Ca2+) uptake into mitochondria of all mammalian tissues, where it regulates bioenergetics, cell death, and Ca2+ signal transduction. Despite its involvement in several human diseases, we currently lack pharmacol. agents for targeting uniporter activity. Here we introduce a high-throughput assay that selects for human MCU-specific small-mol. modulators in primary drug screens. Using isolated yeast mitochondria, reconstituted with human MCU, its essential regulator EMRE, and aequorin, and exploiting a D-lactate- and mannitol/sucrose-based bioenergetic shunt that greatly minimizes false-pos. hits, we identify mitoxantrone out of more than 600 clin. approved drugs as a direct selective inhibitor of human MCU. We validate mitoxantrone in orthogonal mammalian cell-based assays, demonstrating that our screening approach is an effective and robust tool for MCU-specific drug discovery and, more generally, for the identification of compds. that target mitochondrial functions.
- 43Kon, N.; Murakoshi, M.; Isobe, A.; Kagechika, K.; Miyoshi, N.; Nagayama, T. DS16570511 is a Small-Molecule Inhibitor of the Mitochondrial Calcium Uniporter. Cell Death Discov. 2017, 3, 17045, DOI: 10.1038/cddiscovery.2017.4543https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFygtLzI&md5=351f1655a4841af9ee62ee64e691e85bDS16570511 is a small-molecule inhibitor of the mitochondrial calcium uniporterKon, Naohiro; Murakoshi, Michiko; Isobe, Aya; Kagechika, Katsuji; Miyoshi, Naoki; Nagayama, TakahiroCell Death Discovery (2017), 3 (), 17045CODEN: CDDEB5; ISSN:2058-7716. (Nature Publishing Group)In cardiac myocytes, regulation of mitochondrial Ca2+ is important for cellular signaling and cardiac contraction. Ca2+ entry into the mitochondria is mediated by a highly selective Ca2+ channel called the mitochondrial calcium uniporter, which consists of a pore-forming subunit MCU and regulatory subunits such as MICU1. Although pharmacol. regulation of the mitochondrial Ca2+ influx is a promising approach to controlling the cellular functions, a cell-permeable and specific inhibitor of the mitochondrial calcium uniporter has not yet been developed. Here, we identify a novel cell-permeable inhibitor of the uniporter by a high-throughput screening of 120 000 small-mol. compds. In our study, DS16570511 dose-dependently inhibited serum-induced mitochondrial Ca2+ influx in HEK293A cells with an IC50 of 7 μM. DS16570511 inhibited Ca2+ uptake of isolated mitochondria from human cells, rat heart and pig heart. Overexpression of hMCU or hMICU1 in HEK293A cells increased mitochondrial Ca2+ influx, and the increases were completely suppressed by the pretreatment with DS16570511. DS16570511 also blocks mitochondrial Ca2+ overload in a Langendorff perfused beating rat heart. Interestingly, DS16570511 increased cardiac contractility without affecting heart rate in the perfused heart. These results show that DS16570511 is a novel cell-permeable inhibitor of the mitochondrial calcium uniporter and applicable for control of the cardiac functions.
- 44Thu, V. T.; Kim, H. K.; Long, L. T.; Lee, S. R.; Hanh, T. M.; Ko, T. H.; Heo, H. J.; Kim, N.; Kim, S. H.; Ko, K. S.; Rhee, B. D.; Han, J. NecroX-5 Prevents Hypoxia/Reoxygenation Injury by Inhibiting the Mitochondrial Calcium Uniporter. Cardiovasc. Res. 2012, 94, 342– 350, DOI: 10.1093/cvr/cvs12244https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XmtVegsrc%253D&md5=356fbd417f7ef089927c33aebdafa7c2NecroX-5 prevents hypoxia/reoxygenation injury by inhibiting the mitochondrial calcium uniporterThu, Vu Thi; Kim, Hyoung-Kyu; Long, Le Thanh; Lee, Sung-Ryul; Hanh, Tran My; Ko, Tae Hee; Heo, Hye-Jin; Kim, Nari; Kim, Soon Ha; Ko, Kyung Soo; Rhee, Byoung Doo; Han, JinCardiovascular Research (2012), 94 (2), 342-350CODEN: CVREAU; ISSN:0008-6363. (Oxford University Press)Aims: Preservation of mitochondrial function is essential to limit myocardial damage in ischemic heart disease. We examd. the protective effects and mechanism of a new compd., NecroX-5, on rat heart mitochondria in a hypoxia/reoxygenation (HR) model. Methods and results: NecroX-5 reduced mitochondrial oxidative stress, prevented the collapse in mitochondrial membrane potential, improved mitochondrial oxygen consumption, and suppressed mitochondrial Ca2+ overload during reoxygenation in an in vitro rat heart HR model. Furthermore, NecroX-5 reduced the ouabain- or histamine-induced increase in mitochondrial Ca2+. Conclusion: These findings suggest that NecroX-5 may act as a mitochondrial Ca2+ uniporter inhibitor to protect cardiac mitochondria against HR damage.
- 45Antonenko, Y. N.; Rokitskaya, T. I.; Cooper, A. J. L.; Krasnikov, B. F. Minocycline Chelates Ca2+, Binds to Membranes, and Depolarizes Mitochondria by Formation of Ca2+-Dependent Ion Channels. J. Bioenerg. Biomembr. 2010, 42, 151– 163, DOI: 10.1007/s10863-010-9271-1There is no corresponding record for this reference.
- 46Santo-Domingo, J.; Vay, L.; Hernández-Sanmiguel, E.; Lobatón, C. D.; Moreno, A.; Montero, M.; Alvarez, J. The Plasma Membrane Na+/Ca2+ Exchange Inhibitor KB-R7943 is Also a Potent Inhibitor of the Mitochondrial Ca2+ Uniporter. Br. J. Pharmacol. 2007, 151, 647– 654, DOI: 10.1038/sj.bjp.070726046https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXntFemsbk%253D&md5=09d94d943bd1e5e1000a4ec7d367fc8aThe plasma membrane Na+/Ca2+ exchange inhibitor KB-R7943 is also a potent inhibitor of the mitochondrial Ca2+ uniporterSanto-Domingo, J.; Vay, L.; Hernandez-SanMiguel, E.; Lobaton, C. D.; Moreno, A.; Montero, M.; Alvarez, J.British Journal of Pharmacology (2007), 151 (5), 647-654CODEN: BJPCBM; ISSN:0007-1188. (Nature Publishing Group)Background and purpose: The thiourea deriv. KB-R7943, originally developed as inhibitor of the plasma membrane Na+/Ca2+ exchanger, has been shown to protect against myocardial ischemia-reperfusion injury. We have studied here its effects on mitochondrial Ca2+ fluxes. [Ca2+] in cytosol, mitochondria and endoplasmic reticulum (ER), and mitochondrial membrane potential were monitored using both luminescent (targeted aequorins) and fluorescent (fura-2, tetramethylrhodamine Et ester) probes in HeLa cells. KB-R7943 was also a potent inhibitor of the mitochondrial Ca2+ uniporter (MCU). In permeabilized HeLa cells, KB-R7943 inhibited mitochondrial Ca2+ uptake with a Ki of 5.5±1.3 μM (mean±S.D.). In intact cells, 10μM KB-R7943 reduced by 80% the mitochondrial [Ca2+] peak induced by histamine. KB-R7943 did not modify the mitochondrial membrane potential and had no effect on the mitochondrial Na+/Ca2+ exchanger. KB-R7943 inhibited histamine-induced ER-Ca2+ release in intact cells, but not in cells loaded with a Ca2+-chelator to damp cytosolic [Ca2+] changes. Therefore, inhibition of ER-Ca2+-release by KB-R7943 was probably due to the increased feedback Ca2+-inhibition of inositol 1,4,5-trisphosphate receptors after MCU block. This mechanism also explains why KB-R7943 reversibly blocked histamine-induced cytosolic [Ca2+] oscillations in the same range of concns. required to inhibit MCU. Inhibition of MCU by KB-R7943 may contribute to its cardioprotective activity by preventing mitochondrial Ca2+-overload during ischemia-reperfusion. In addn., the effects of KB-R7943 on Ca2+ homeostasis provide new evidence for the role of mitochondria modulating Ca2+-release and regenerative Ca2+-oscillations. Search for permeable and selective MCU inhibitors may yield useful pharmacol. tools in the future.
- 47Kapuscinski, J.; Darzynkiewicz, Z. Interactions of Antitumor Agents Ametantrone and Mitoxantrone (Novatrone) with Double-Stranded DNA. Biochem. Pharmacol. 1985, 34, 4203– 4213, DOI: 10.1016/0006-2952(85)90275-847https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL28XotFKitA%253D%253D&md5=f6f28eedd99ec102a125650d13f86b24Interactions of antitumor agents Ametantrone and mitoxantrone (Novatrone) with double-stranded DNAKapuscinski, Jan; Darzynkiewicz, ZbigniewBiochemical Pharmacology (1985), 34 (24), 4203-13CODEN: BCPCA6; ISSN:0006-2952.Interactions of mitoxantrone and ametantrone with natural and synthetic nucleic acids in aq. medium [0.15 NaCl, 5 mM 4-(2-hydroxyethyl)-1-piperazine-ethanesulfonic acid (Hepes), pH 7.0. 25%] were studied using computer-aided spectrophotometric techniques. Absorption spectra of the drugs in monomeric and dimeric form and their complexes with DNAs at low drug/phosphate ratios (D/P) were established. The latter were red-shifted and had lower amplitude as compared with the spectra of the free ligand's monomer; the change is consistent with the already well-established intercalative mode of drug-nucleic acid interaction. Drug-DNA equil. were studied using the McGhee-von Hippel model of noncooperative ligand-polymer interaction, with the correction for dimerization of drugs. Although mitoxantrone was 2 orders of magnitude more potent an antitumor drug than Ametantrone, the intrinsic assocn. consts. (Ki) of both drugs were of similar magnitude. Also, no significant DNA-base specificity for either of the drugs (measured as Ki value for various homopolymers) was obsd. Therefore, no correlation was apparent between the intercalative mode of binding to DNA, regardless of base compn., and the pharmacol. activity of these drugs. At higher D/P ratios, a secondary mode of binding was detected by both spectroscopy and light-scattering measurement. Homopolymer-pairs and polymers contg. only dI and dC were esp. susceptible to this secondary type of binding. The possibility that this secondary type of binding may be responsible for the antitumor properties of the drugs is considered.
- 48Mazerski, J.; Martelli, S.; Borowski, E. The Geometry of Intercalation Complex of Antitumor Mitoxantrone and Ametantrone with DNA: Molecular Dynamics Simulations. Acta Biochim. Polym. 1998, 45, 1– 1148https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXkt1egsbg%253D&md5=84573969f77849b9405683e28bd32d27The geometry of intercalation complex of antitumor mitoxantrone and ametantrone with DNA: molecular dynamics simulationsMazerski, Jan; Martelli, Sante; Borowski, EdwardActa Biochimica Polonica (1998), 45 (1), 1-11CODEN: ABPLAF; ISSN:0001-527X. (Polish Biochemical Society)Intercalative binding of the antitumor drugs ametantrone and mitoxantrone to the dodecamer duplex d(CGCGAGCTCGCG)2 was studied by applying mol. dynamics in water with the GROMOS 87 force field. A no. of reasonable binding orientations were tested by short pre-simulations. It was shown that in energetically favorable orientation the anthraquinone chromophore is perpendicular to the direction of inter-base hydrogen bonds. Helically shaped side-chains of the drugs fit to the minor groove. The best orientation obtained in pre-simulations was applied in the main simulations. Small but significant differences were found between structures of inter-calation complexes of the two drugs with the dodecamer duplex, the mitoxantrone complex possessing more favorable energy. The mol. nature of interactions responsible for those differences has been discussed.
- 49Boland, M. P.; Fitzgerald, K. A.; O’Neill, L. A. J. Topoisomerase II Is Required for Mitoxantrone to Signal Nuclear Factor ΚB Activation in HL60 Cells. J. Biol. Chem. 2000, 275, 25231– 25238, DOI: 10.1074/jbc.275.33.25231There is no corresponding record for this reference.
- 50Smith, P. J.; Morgan, S. A.; Fox, M. E.; Watson, J. V. Mitoxantrone-DNA Binding and the Induction of Topoisomerase II Associated DNA Damage in Multi-Drug Resistant Small Cell Lung Cancer Cells. Biochem. Pharmacol. 1990, 40, 2069– 2078, DOI: 10.1016/0006-2952(90)90237-FThere is no corresponding record for this reference.
- 51Nägele, H.; Castel, M. A.; Deutsch, O.; Wagner, F. M.; Reichenspurner, H. Heart Transplantation in a Patient with Multiple Sclerosis and Mitoxantrone-Induced Cardiomyopathy. J. Heart Lung Transplant. 2004, 23, 641– 643, DOI: 10.1016/S1053-2498(03)00307-351https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2c3jslKitA%253D%253D&md5=ce71e137ff3f9fbce1c00a9c4fdc31d4Heart transplantation in a patient with multiple sclerosis and mitoxantrone-induced cardiomyopathyNagele H; Castel M A; Deutsch O; Wagner F M; Reichenspurner HThe Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation (2004), 23 (5), 641-3 ISSN:1053-2498.We describe a 30-year-old man with end-stage heart failure after therapy with mitoxantrone for multiple sclerosis. A successful orthotopic heart transplantation was performed when intensified medical therapy failed to improve the patient's hemodynamics. In spite of the severe underlying disease he did well on dual immunosuppression with methylprednisone and cyclosporine. Neurologic symptoms remained stable throughout the procedure and, after 2 months, he resumed preoperative ambulatory status. Eight years after the operation, the patient is now in New York Heart Association (NYHA) Class I status. Using canes, he is able to walk short distances. Repeated urinary tract infections caused by Escherichia coli became a problem, but have been controlled by long-term oral antibiotic prophylaxis with trimethoprim.
- 52Goebel, M.; Kaplan, E. Anthracycline-Induced Cardiotoxicity – A Review. Oncol. Res. Treat. 2004, 15, 198– 204, DOI: 10.1159/000217360There is no corresponding record for this reference.
- 53Ying, W.-L.; Emerson, J.; Clarke, M. J.; Sanadi, D. R. Inhibition of Mitochondrial Calcium Ion Transport by an Oxo-Bridged Dinuclear Ruthenium Ammine Complex. Biochemistry 1991, 30, 4949– 4952, DOI: 10.1021/bi00234a01653https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXit1eksrk%253D&md5=dcf10f1224e51c3dce2d31eadc77dbb4Inhibition of mitochondrial calcium ion transport by an oxo-bridged dinuclear ruthenium ammine complexYing, Wen Long; Emerson, Jeffrey; Clarke, Michael J.; Sanadi, D. RaoBiochemistry (1991), 30 (20), 4949-52CODEN: BICHAW; ISSN:0006-2960.Ruthenium red is a well-known and effective inhibitor of the mitochondria Ca2+ uniporter; however, K.C. Reed and F. Bygrave (1974) tentatively attributed this inhibition to a colorless impurity present in com. samples of ruthenium red (RR). This component has now been isolated and a deriv., (μ-O)[(HCO2)(NH3)4Ru]2Cl3, structurally characterized. The active species in soln. appears to be the sym. oxo-bridged ion, [X(NH3)4Ru-O-Ru(NH3)4X]3+, where X = Cl- or OH-. Its absorption spectrum shows a max. at 360 nm. The dinuclear ruthenium ammine complex inhibits Ca2+-stimulated respiration of rat liver mitochondria with an I50 of 3.5 pmol/mg of protein compared to the value of 60 pmol of RR/mg of protein. The inhibition by the dinuclear compd. is noncompetitive with Ca2+. Respiration-linked swelling of mitochondria induced by Cd2+ also responds similarly to both the dinuclear complex and RR. A close correlation was obsd. between binding to mitochondria as monitored with 103Ru-labeled dinuclear complex and inhibition of Ca2+ transport. A Scatchard plot yielded ests. of max. specific binding and dissocn. const. of 7.5 pmol/mg of protein and 1.3 nM, resp. The inhibitor has the characteristics of a satisfactory affinity ligand for purifn. of the uniporter.
- 54Matlib, M. A.; Zhou, Z.; Knight, S.; Ahmed, S.; Choi, K. M.; Krause-Bauer, J.; Phillips, R.; Altschuld, R.; Katsube, Y.; Sperelakis, N.; Bers, D. M. Oxygen-Bridged Dinuclear Ruthenium Amine Complex Specifically Inhibits Ca2+ Uptake into Mitochondria in Vitro and in Situ in Single Cardiac Myocytes. J. Biol. Chem. 1998, 273, 10223– 10231, DOI: 10.1074/jbc.273.17.1022354https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXivFKnsbc%253D&md5=53f8261b91276b659d0038814914ec86Oxygen-bridged dinuclear ruthenium amine complex specifically inhibits Ca2+ uptake into mitochondria in vitro and in situ in single cardiac myocytesMatlib, Mohammed A.; Zhou, Zhuan; Knight, Selena; Ahmed, Saadia; Choi, Kin M.; Krause-Bauer, Jeanette; Phillips, Ronald; Altschuld, Ruth; Katsube, Yasuhiro; Sperelakis, Nicholas; Bers, Donald M.Journal of Biological Chemistry (1998), 273 (17), 10223-10231CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Ruthenium red is a well known inhibitor of Ca2+ uptake into mitochondria in vitro. However, its utility as an inhibitor of Ca2+ uptake into mitochondria in vivo or in situ in intact cells is limited because of its inhibitory effects on sarcoplasmic reticulum Ca2+ release channel and other cellular processes. We have synthesized a ruthenium deriv. and found it to be an oxygen-bridged dinuclear ruthenium amine complex. It has the same chem. structure as Ru360 reported previously (Emerson, J., Clarke, M. J., Ying, W-L., and Sanadi, D. R. (1993) J. Am. Chem. Soc. 115, 11799-11805). Ru360 has been shown to be a potent inhibitor of Ca2+-stimulated respiration of liver mitochondria in vitro. However, the specificity of Ru360 on Ca2+ uptake into mitochondria in vitro or in intact cells has not been detd. The present study reports in detail the potency, the effectiveness, and the mechanism of inhibition of mitochondrial Ca2+ uptake by Ru360 and its specificity in vitro in isolated mitochondria and in situ in isolated cardiac myocytes. Ru360 was more potent (IC50 = 0.184 nM) than ruthenium red (IC50 = 6.85 nM) in inhibiting Ca2+ uptake into mitochondria. 103Ru360 was found to bind to isolated mitochondria with high affinity (Kd = 0.34 nM, Bmax = 80 fmol/mg of mitochondrial protein). The IC50 of 103Ru360 for the inhibition of Ca2+ uptake into mitochondria was also 0.2 nM, indicating that satn. of a specific binding site is responsible for the inhibition of Ca2+ uptake. Ru360, as high as 10 μM, produced no effect on sarcoplasmic reticulum Ca2+ uptake or release, sarcolemmal Na+/Ca2+ exchange, actomyosin ATPase activity, L-type Ca2+ channel current, cytosolic Ca2+ transients, or cell shortening. 103Ru360 was taken up by isolated myocytes in a time-dependent biphasic manner. Ru360 (10 μM) applied outside intact voltage-clamped ventricular myocytes prevented Ca2+ uptake into mitochondria in situ where the cells were progressively loaded with Ca2+ via sarcolemmal Na+/Ca2+ exchange by depolarization to +110 mV. We conclude that Ru360 specifically blocks Ca2+ uptake into mitochondria and can be used in intact cells.
- 55Emerson, J.; Clarke, M. J.; Ying, W. L.; Sanadi, D. R. The Component of “Ruthenium Red” Responsible for Inhibition of Mitochondrial Calcium Ion Transport. Spectra, Electrochemistry, and Aquation Kinetics. Crystal Structure of μ-O-[(HCO)2(NH3)4Ru]2Cl3. J. Am. Chem. Soc. 1993, 115, 11799– 11805, DOI: 10.1021/ja00078a019There is no corresponding record for this reference.
- 56Nathan, S. R.; Pino, N. W.; Arduino, D. M.; Perocchi, F.; MacMillan, S. N.; Wilson, J. J. Synthetic Methods for the Preparation of a Functional Analogue of Ru360, a Potent Inhibitor of Mitochondrial Calcium Uptake. Inorg. Chem. 2017, 56, 3123– 3126, DOI: 10.1021/acs.inorgchem.6b0310856https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjsVCgtL8%253D&md5=bbd2e540a1cb9f4a4358d96d11817f55Synthetic Methods for the Preparation of a Functional Analogue of Ru360, a Potent Inhibitor of Mitochondrial Calcium UptakeNathan, Sarah R.; Pino, Nicholas W.; Arduino, Daniela M.; Perocchi, Fabiana; MacMillan, Samantha N.; Wilson, Justin J.Inorganic Chemistry (2017), 56 (6), 3123-3126CODEN: INOCAJ; ISSN:0020-1669. (American Chemical Society)The mixed-valent oxo-bridged Ru complex [(HCO2)(NH3)4Ru(μ-O)Ru(NH3)4(O2CH)]3+, known as Ru360, is a selective inhibitor of mitochondrial Ca uptake. Although this compd. is useful for studying the role of mitochondrial Ca in biol. processes, its widespread availability is limited because of challenges in purifn. and characterization. Here, the authors describe the authors' studies of three different synthetic methods for the prepn. of a functional analog of this valuable compd. This analog, isolated from the authors' procedures, exhibits potent mitochondrial Ca uptake inhibitory properties in permeabilized HeLa cells and in isolated mitochondria.
- 57Nathan, S. R.; Wilson, J. J. Synthesis and Evaluation of a Ruthenium-Based Mitochondrial Calcium Uptake Inhibitor. J. Visualized Exp. 2017, 128, e56527, DOI: 10.3791/56527There is no corresponding record for this reference.
- 58Cao, C.; Wang, S.; Cui, T.; Su, X.-C.; Chou, J. J. Ion and Inhibitor Binding of the Double-Ring Ion Selectivity Filter of the Mitochondrial Calcium Uniporter. Proc. Natl. Acad. Sci. U. S. A. 2017, 114, E2846– E2851, DOI: 10.1073/pnas.162031611458https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXks1Smtr0%253D&md5=c3ec4bd6337c0234eab5aed8e354c2f4Ion and inhibitor binding of the double-ring ion selectivity filter of the mitochondrial calcium uniporterCao, Chan; Wang, Shuqing; Cui, Tanxing; Su, Xun-Cheng; Chou, James J.Proceedings of the National Academy of Sciences of the United States of America (2017), 114 (14), E2846-E2851CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The mitochondrial Ca2+ uniporter (MCU) is a holocomplex consisting of the Ca2+-conducting channel and several accessory and regulatory components. A previous electrophysiol. study found that the uniporter had high Ca2+ selectivity and conductance and this depended critically on the conserved amino acid sequence motif, DXXE (Asp-X-X-Glu) of MCU. A recent NMR structure of the MCU channel from Caenorhabditis elegans revealed that the DXXE formed 2 parallel carboxylate rings at the channel entrance that seem to serve as the ion selectivity filter, although direct ion interaction of this structural motif has not been addressed. Here, we used a paramagnetic probe, Mn2+, to investigate ion and inhibitor binding of this putative selectivity filter. The paramagnetic NMR data showed that mutants with a single carboxylate ring, NXXE (Asn-X-X-Glu) and DXXQ (Asp-X-X-Gln), each could bind Mn2+ specifically, whereas in the wild type the 2 rings bound Mn2+ cooperatively, resulting in ∼1000-fold higher apparent affinity. Ca2+ could specifically displace bound Mn2+ at the DXXE site in the channel. Furthermore, titrating the sample with the known channel inhibitor, ruthenium 360 (Ru360), could displace Mn2+ binding from the solvent-accessible Asp site but not the inner Glu site. The NMR titrn. data, together with structural anal. of the DXXE motif and mol. dynamics simulations, indicated that the double carboxylate rings at the apex of the MCU pore constitute the ion selectivity filter and that Ru360 directly blocks ion entry into the filter by binding to the outer carboxylate ring.
- 59Gushchin, A. L.; Laricheva, Y. A.; Abramov, P. A.; Sokolov, M. N.; Abramov, P. A. Synthesis and Electrochemical Properties of [RuIV2O(PhCN)4Cl6]. Inorg. Chem. Commun. 2018, 95, 1387– 7003, DOI: 10.1016/j.inoche.2018.07.033There is no corresponding record for this reference.
- 60Urgiles, J.; Nathan, S. R.; MacMillan, S. N.; Wilson, J. J. Dinuclear Nitrido-Bridged Ruthenium Complexes Bearing Diimine Ligands. Dalton Trans. 2017, 46, 14256– 14263, DOI: 10.1039/C7DT03085A60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhs1alt7%252FN&md5=e404175cf715aa29108493160fb5edd4Dinuclear nitrido-bridged ruthenium complexes bearing diimine ligandsUrgiles, Julie; Nathan, Sarah R.; MacMillan, Samantha N.; Wilson, Justin J.Dalton Transactions (2017), 46 (41), 14256-14263CODEN: DTARAF; ISSN:1477-9226. (Royal Society of Chemistry)Reactions of K3[Ru2NCl8(H2O)2] with 2,2'-bipyridine (bpy), 4,4'-dimethyl-2,2'-bipyridine (dmbpy), and 4,4'-dimethoxy-2,2'-bipyridine (dmobpy) yielded the nitrido-bridged dinuclear complexes [Ru2N(L)2Cl5(DMF)] where L = bpy (1), dmbpy (2), and dmobpy (3). The crystal structures of these complexes reveal a linear Ru-N-Ru moiety with each Ru center bearing a bidentate diimine ligand. The complexes were further characterized by NMR, IR, and UV-visible spectroscopic methods and cyclic voltammetry. Because the compds. bear some structural similarities with the mitochondrial Ca uptake inhibitor Ru360, the ability of these complexes to act in this capacity was evaluated. 1-3 All fail to block mitochondrial Ca uptake, revealing new facets of the structure-activity relations for Ru-based mitochondrial Ca uptake inhibitors.
- 61Dunitz, J. D.; Orgel, L. E. Application of Molecular-Orbital Theory to Some Binuclear Coordination Compounds. J. Chem. Soc. 1953, 2594– 2596, DOI: 10.1039/jr9530002594There is no corresponding record for this reference.
- 62Cleare, M. J.; Griffith, W. P. Binuclear Nitrido-Complexes of Ruthenium. Chem. Commun. 1968, 21, 1302, DOI: 10.1039/c19680001302There is no corresponding record for this reference.
- 63Griffith, W. P.; McManus, N. T.; Skapski, A. C. X-Ray Crystal Structure of [Ru2N(ethylenediamine)5]Cl5· H2O; a Novel Complex Containing Both Nitrido and Ethylenediamine Bridges. J. Chem. Soc., Chem. Commun. 1984, 7, 434, DOI: 10.1039/c39840000434There is no corresponding record for this reference.
- 64Ng, H.-Y.; Cheung, W.-M.; Kwan Huang, E.; Wong, K.-L.; Sung, H. H.-Y.; Williams, I. D.; Leung, W.-H. Ruthenium Chalcogenonitrosyl and Bridged Nitrido Complexes Containing Chelating Sulfur and Oxygen Ligands. Dalton Trans. 2015, 44, 18459– 18468, DOI: 10.1039/C5DT02513C64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFCqs7%252FO&md5=3ff794cd2a4f5027e01d33b5b91a0f09Ruthenium chalcogenonitrosyl and bridged nitrido complexes containing chelating sulfur and oxygen ligandsNg, Ho-Yuen; Cheung, Wai-Man; Kwan Huang, Enrique; Wong, Kang-Long; Sung, Herman H.-Y.; Williams, Ian D.; Leung, Wa-HungDalton Transactions (2015), 44 (42), 18459-18468CODEN: DTARAF; ISSN:1477-9226. (Royal Society of Chemistry)Ruthenium thio- and seleno-nitrosyl complexes contg. chelating S and O ligands were synthesized and their de-chalcogenation reactions were studied. The reaction of mer-[Ru(N)Cl3(AsPh3)2] with elemental S and Se in THF at reflux afforded the chalcogenonitrosyl complexes mer-[Ru(NX)Cl3(AsPh3)2] [X = S (1), Se (2)]. Treatment of 1 with KN(R2PS)2 afforded trans-[Ru(NS)Cl{N(R2PS)2}2] [R = Ph (3), Pri (4), But (5)]. Alternatively, the thionitrosyl complex 5 was obtained from [Bu4N][Ru(N)Cl4] and KN(But2PS)2, presumably via S atom transfer from [N(But2PS)2]- to the nitride. Reactions of 1 and 2 with NaLOEt (LOEt- = [Co(η5-C5H5){P(O)(LOEt)2}3]-) gave [Ru(NX)LOEtCl2] (X = S (8), Se (9)). Treatment of [Bu4N][Ru(N)Cl4] with KN(R2PS)2 produced RuIV-RuIV μ-nitrido complexes [Ru2(μ-N){N(R2PS)2}4Cl] [R = Ph (6), Pri (7)]. Reactions of 3 and 9 with PPh3 afforded 6 and [Ru(NPPh3)LOEtCl2], resp. The desulfurization of 5 with [Ni(cod)2] (cod = 1,5-cyclooctadiene) gave the mixed valance RuIII-RuIV μ-nitrido complex [Ru2(μ-N){N(But2PS)2}4] (10) that was oxidized by [Cp2Fe](PF6) to give the RuIV-RuIV complex [Ru2(μ-N){N(But2PS)2}4](PF6) ([10]PF6). The crystal structures of 1, 2, 3, 7, 9 and 10 were detd.
- 65Ciechanowicz, M.; Skapski, A. C. Crystal Structure of Potassium μ-Nitrido-Bis[Aquotetrachlororuthenate(IV)]. J. Chem. Soc. A 1971, 84, 1792– 1794, DOI: 10.1039/J19710001792There is no corresponding record for this reference.
- 66Freshney, R. I. Culture of Animal Cells, 5th ed.; John Wiley & Sons, Inc.: Hoboken, NJ, 2005.There is no corresponding record for this reference.
- 67Shanmughapriya, S.; Rajan, S.; Hoffman, N. E.; Zhang, X.; Guo, S.; Kolesar, J. E.; Hines, K. J.; Ragheb, J.; Jog, N. R.; Caricchio, R.; Baba, Y.; Zhou, Y.; Kaufman, B. A.; Cheung, J. Y.; Kurosaki, T.; Gill, D. L.; Madesh, M. Ca2+ Signals Regulate Mitochondrial Metabolism by Stimulating CREB-Mediated Expression of the Mitochondrial Ca2+ Uniporter Gene MCU. Sci. Signaling 2015, 8, ra23, DOI: 10.1126/scisignal.2005673There is no corresponding record for this reference.
- 68Reers, M.; Smith, T. W.; Chen, L. B. J-aggregate formation of a carbocyanine as a quantitative fluorescent indicator of membrane potential. Biochemistry 1991, 30, 4480– 4486, DOI: 10.1021/bi00232a01568https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXitVart7o%253D&md5=2c2befb752f3baa12d4fc1255f925448J-aggregate formation of a carbocyanine as a quantitative fluorescent indicator of membrane potentialReers, Martin; Smith, Thomas W.; Chen, Lan BoBiochemistry (1991), 30 (18), 4480-6CODEN: BICHAW; ISSN:0006-2960.The spectral properties of a novel membrane potential sensitive probe (JC-1) were characterized in aq. buffers and in isolated cardiac mitochondria. JC-1 is a carbocyanine with a delocalized pos. charge. It formed under favorable conditions a concn.-dependent fluorescent nematic phase consisting of J-aggregates. When excited at 490 nm, the monomers exhibited an emission max. at 527 nm and J-aggregates at 590 nm. Increasing concns. of JC-1 above a certain concn. caused a linear rise in the J-aggregate fluorescence, while the monomer fluorescence remained const. The membrane potential of energized mitochondria (neg. inside) promoted a directional uptake of JC-1 into the matrix, also with subsequent formation of J-aggregates. The J-aggregate fluorescence was sensitive to transient membrane potential changes induced by ADP and to metabolic inhibitors of oxidative phosphorylation. The J-aggregate fluorescence was pH independent within the physiol. pH range of 7.15-8.0 and could be linearly calibrated with valinomycin-induced K+ diffusion potentials. The advantage of JC-1 over rhodamines and other carbocyanines is that its color altered reversibly from green to red with increasing membrane potentials. This can be exploited for imaging live mitochondria on the stage of a microscope.
- 69King, A. P.; Gellineau, H. A.; Ahn, J. E.; MacMillan, S. N.; Wilson, J. J. Bis(Thiosemicarbazone) Complexes of Cobalt(III). Synthesis, Characterization, and Anticancer Potential. Inorg. Chem. 2017, 56, 6609– 6623, DOI: 10.1021/acs.inorgchem.7b0071069https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXns1yku7o%253D&md5=d4eeafefb534c53f21a1ef80a7657172Bis(thiosemicarbazone) Complexes of Cobalt(III). Synthesis, Characterization, and Anticancer PotentialKing, A. Paden; Gellineau, Hendryck A.; Ahn, Jung-Eun; MacMillan, Samantha N.; Wilson, Justin J.Inorganic Chemistry (2017), 56 (11), 6609-6623CODEN: INOCAJ; ISSN:0020-1669. (American Chemical Society)Nine bis(thiosemicarbazone) (BTSC) cobalt(III) complexes of the general formula [Co(BTSC)(L)2]NO3 were synthesized, where BTSC = diacetyl bis(thiosemicarbazone) (ATS), pyruvaldehyde bis(thiosemicarbazone) (PTS), or glyoxal bis(thiosemicarbazone) (GTS) and L = ammonia, imidazole (Im), or benzylamine (BnA). These compds. were characterized by multinuclear NMR spectroscopy, mass spectrometry, cyclic voltammetry, and x-ray crystallog. Their stability in phosphate-buffered saline was investigated and is highly dependent on the nature of the axial ligand, L. These studies revealed that complex stability is primarily dictated by the axial ligand following the sequence NH3 > Im > BnA. The cellular uptake and cytotoxicity in cancer cells were also detd. Both the cellular uptake and cytotoxicity were significantly affected by the nature of the equatorial BTSC. Complexes of ATS were taken up much more effectively than those of PTS and GTS. The cytotoxicity of the complexes was correlated to that of the free ligand. Cell uptake and cytotoxicity were also detd. under hypoxic conditions. Only minor differences in the hypoxia activity and uptake were obsd. Treatment of the cancer cells with the copper-depleting agent tetrathiomolybdate decreased the cytotoxic potency of the complexes, indicating that they may operate via a copper-dependent mechanism. These results provide a structure-activity relation for this class of compds., which may be applied for the rational design of new cobalt(III) anticancer agents.
- 70Egger, A. E.; Rappel, C.; Jakupec, M. A.; Hartinger, C. G.; Heffeter, P.; Keppler, B. K. Development of an Experimental Protocol for Uptake Studies of Metal Compounds in Adherent Tumor Cells. J. Anal. At. Spectrom. 2009, 24, 51– 61, DOI: 10.1039/B810481F70https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsFWisr%252FK&md5=e1f3d2c5048697874728de374d85aa21Development of an experimental protocol for uptake studies of metal compounds in adherent tumor cellsEgger, Alexander E.; Rappel, Christina; Jakupec, Michael A.; Hartinger, Christian G.; Heffeter, Petra; Keppler, Bernhard K.Journal of Analytical Atomic Spectrometry (2009), 24 (1), 51-61CODEN: JASPE2; ISSN:0267-9477. (Royal Society of Chemistry)Cellular uptake is being widely investigated in the context of diverse biol. activities of metal compds. on the cellular level. However, the applied techniques differ considerably, and a validated methodol. is not at hand. Therefore, we have varied numerous aspects of sample prepn. of the human colon carcinoma cell line SW480 exposed in vitro to the tumor-inhibiting metal complexes cisplatin and indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (KP1019) prior to anal. with ICP-MS, and the results were found to be tremendously influenced by adsorption to the culture dishes. Adsorption to culture plates increases linearly with the concn. of KP1019, depends on the protein content of the medium, the duration of contact to protein-contg. medium prior to drug addn. and the hydrophilicity/lipophilicity of the compd. For varying degrees of cell confluence, adsorption of Ru hardly differs from cell-free expts. Desorption from the plates contributes to total Ru detected in dependence on the cell harvesting method. Desorption kinetics for lysis in HNO3 and tetramethylammonium hydroxide (TMAH) are comparable, but TMAH is a more potent desorbant. Sample storage conditions prior to anal. influence significantly the recovery of analyte. Protocols using cell lysis in the culture plate without proper corrections run the risk of producing artifacts resulting from metal adsorption/desorption to an extent comparable with the actual cellular content. However, exptl. protocols reported in the literature frequently do not contain information whether adsorption or blank correction were performed and should be regarded with caution, esp. if lysis was performed directly in the culture dishes.
- 71Komor, A. C.; Schneider, C. J.; Weidmann, A. G.; Barton, J. K. Cell-Selective Biological Activity of Rhodium Metalloinsertors Correlates with Subcellular Localization. J. Am. Chem. Soc. 2012, 134, 19223– 19233, DOI: 10.1021/ja309068771https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhs1CksrrI&md5=ac877e082357f44187d45af67896047aCell-Selective Biological Activity of Rhodium Metalloinsertors Correlates with Subcellular LocalizationKomor, Alexis C.; Schneider, Curtis J.; Weidmann, Alyson G.; Barton, Jacqueline K.Journal of the American Chemical Society (2012), 134 (46), 19223-19233CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Deficiencies in the mismatch repair (MMR) pathway are assocd. with several types of cancers, as well as resistance to commonly used chemotherapeutics. Rhodium metalloinsertors have been found to bind DNA mismatches with high affinity and specificity in vitro, and also exhibit cell-selective cytotoxicity, targeting MMR-deficient cells over MMR-proficient cells. Ten distinct metalloinsertors with varying lipophilicities have been synthesized and their mismatch binding affinities and biol. activities detd. Although DNA photocleavage expts. demonstrate that their binding affinities are quite similar, their cell-selective antiproliferative and cytotoxic activities vary significantly. Inductively coupled plasma mass spectrometry (ICP-MS) expts. have uncovered a relation between the subcellular distribution of these metalloinsertors and their biol. activities. Specifically, we find that all of our metalloinsertors localize in the nucleus at sufficient concns. for binding to DNA mismatches. However, the metalloinsertors with high rhodium localization in the mitochondria show toxicity that is not selective for MMR-deficient cells, whereas metalloinsertors with less mitochondrial rhodium show activity that is highly selective for MMR-deficient vs. proficient cells. This work supports the notion that specific targeting of the metalloinsertors to nuclear DNA gives rise to their cell-selective cytotoxic and antiproliferative activities. The selectivity in cellular targeting depends upon binding to mismatches in genomic DNA.
- 72Boyle, K. M.; Barton, J. K. A Family of Rhodium Complexes with Selective Toxicity toward Mismatch Repair-Deficient Cancers. J. Am. Chem. Soc. 2018, 140, 5612– 5624, DOI: 10.1021/jacs.8b0227172https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXntFWit7k%253D&md5=019869099e85f1d376b51720aebcb7feA Family of Rhodium Complexes with Selective Toxicity toward Mismatch Repair-Deficient CancersBoyle, Kelsey M.; Barton, Jacqueline K.Journal of the American Chemical Society (2018), 140 (16), 5612-5624CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Rhodium metalloinsertors are a unique set of metal complexes that bind specifically to DNA base pair mismatches in vitro and kill mismatch repair (MMR)-deficient cells at lower concns. than their MMR-proficient counterparts. A family of metalloinsertors contg. rhodium-oxygen ligand coordination, termed "Rh-O" metalloinsertors, has been prepd. and shown to have a significant increase in both overall potency and selectivity toward MMR-deficient cells regardless of structural changes in the ancillary ligands. Here we describe DNA-binding and cellular studies with the second generation of Rh-O metalloinsertors in which an ancillary ligand is varied in both steric bulk and lipophilicity. These complexes, of the form [Rh(L)(chrysi)(PPO)]2+, all include the O-contg. PPO ligand (PPO = 2-(pyridine-2-yl)propan-2-ol) and the arom. inserting ligand chrysi (5,6-chrysene quinone diimine) but differ in the identity of their ancillary ligand L, where L is a phenanthroline or bipyridyl deriv. The Rh-O metalloinsertors in this family all show micromolar binding affinities for a 29-mer DNA hairpin contg. a single CC mismatch. The complexes display comparable lipophilic tendencies and pKa values of 8.1-9.1 for dissocn. of an imine proton on the chrysi ligand. In cellular proliferation and cytotoxicity assays with MMR-deficient cells (HCT116O) and MMR-proficient cells (HCT116N), the complexes contg. the phenanthroline-derived ligands show highly selective cytotoxic preference for the MMR-deficient cells at nanomolar concns. Using mass spectral analyses, it is shown that the complexes are taken into cells through a passive mechanism and exhibit low accumulation in mitochondria, an off-target organelle that, when targeted by parent metalloinsertors, can lead to nonselective cytotoxicity. Overall, these Rh-O metalloinsertors have distinct and improved behavior compared to previous generations of parent metalloinsertors, making them ideal candidates for further therapeutic assessment.
- 73Ahmad, K. A.; Iskandar, K. B.; Hirpara, J. L.; Clement, M. V.; Pervaiz, S. Hydrogen Peroxide-Mediated Cytosolic Acidification is a Signal for Mitochondrial Translocation of Bax during Drug-Induced Apoptosis of Tumor Cells. Cancer Res. 2004, 64, 7867– 7878, DOI: 10.1158/0008-5472.CAN-04-0648There is no corresponding record for this reference.
- 74Dong, Z.; Shanmughapriya, S.; Tomar, D.; Siddiqui, N.; Lynch, S.; Nemani, N.; Breves, S. L.; Zhang, X.; Tripathi, A.; Palaniappan, P.; Riitano, M. F.; Worth, A. M.; Seelam, A.; Carvalho, E.; Subbiah, R.; Jaña, F.; Soboloff, J.; Peng, Y.; Cheung, J. Y.; Joseph, S. K.; Caplan, J.; Rajan, S.; Stathopulos, P. B.; Madesh, M. Mitochondrial Ca2+ Uniporter is a Mitochondrial Luminal Redox Sensor That Augments MCU Channel Activity. Mol. Cell 2017, 65, 1014– 1028, DOI: 10.1016/j.molcel.2017.01.03274https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjvVWqu74%253D&md5=31aae22ab2cd06621296a3b43a7898fdMitochondrial Ca2+ Uniporter Is a Mitochondrial Luminal Redox Sensor that Augments MCU Channel ActivityDong, Zhiwei; Shanmughapriya, Santhanam; Tomar, Dhanendra; Siddiqui, Naveed; Lynch, Solomon; Nemani, Neeharika; Breves, Sarah L.; Zhang, Xueqian; Tripathi, Aparna; Palaniappan, Palaniappan; Riitano, Massimo F.; Worth, Alison M.; Seelam, Ajay; Carvalho, Edmund; Subbiah, Ramasamy; Jana, Fabian; Soboloff, Jonathan; Peng, Yizhi; Cheung, Joseph Y.; Joseph, Suresh K.; Caplan, Jeffrey; Rajan, Sudarsan; Stathopulos, Peter B.; Madesh, MuniswamyMolecular Cell (2017), 65 (6), 1014-1028.e7CODEN: MOCEFL; ISSN:1097-2765. (Elsevier Inc.)Ca2+ dynamics and oxidative signaling are fundamental mechanisms for mitochondrial bioenergetics and cell function. The MCU complex is the major pathway by which these signals are integrated in mitochondria. Whether and how these coactive elements interact with MCU have not been established. As an approach toward understanding the regulation of MCU channel by oxidative milieu, we adapted inflammatory and hypoxia models. We identified the conserved cysteine 97 (Cys-97) to be the only reactive thiol in human MCU that undergoes S-glutathionylation. Furthermore, biochem., structural, and superresoln. imaging anal. revealed that MCU oxidn. promotes MCU higher order oligomer formation. Both oxidn. and mutation of MCU Cys-97 exhibited persistent MCU channel activity with higher [Ca2+]m uptake rate, elevated mROS, and enhanced [Ca2+]m overload-induced cell death. In contrast, these effects were largely independent of MCU interaction with its regulators. These findings reveal a distinct functional role for Cys-97 in ROS sensing and regulation of MCU activity.
- 75Zhao, Y.; Araki, S.; Wu, J.; Teramoto, T.; Chang, Y.-F.; Nakano, M.; Abdelfattah, A. S.; Fujiwara, M.; Ishihara, T.; Nagai, T.; Campbell, R. E. An Expanded Palette of Genetically Encoded Ca2+ Indicators. Science 2011, 333, 1888– 1891, DOI: 10.1126/science.120859275https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXht1aitr%252FK&md5=192ab4166c581df59b11b6373f689847An Expanded Palette of Genetically Encoded Ca2+ IndicatorsZhao, Yongxin; Araki, Satoko; Wu, Jiahui; Teramoto, Takayuki; Chang, Yu-Fen; Nakano, Masahiro; Abdelfattah, Ahmed S.; Fujiwara, Manabi; Ishihara, Takeshi; Nagai, Takeharu; Campbell, Robert E.Science (Washington, DC, United States) (2011), 333 (6051), 1888-1891CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Engineered fluorescent protein (FP) chimeras that modulate their fluorescence in response to changes in calcium ion (Ca2+) concn. are powerful tools for visualizing intracellular signaling activity. However, despite a decade of availability, the palette of single FP-based Ca2+ indicators has remained limited to a single green hue. The authors have expanded this palette by developing blue, improved green, and red intensiometric indicators, as well as an emission ratiometric indicator with an 11,000% ratio change. This series enables improved single-color Ca2+ imaging in neurons and transgenic Caenorhabditis elegans. In HeLa cells, Ca2+ was imaged in three subcellular compartments, and, in conjunction with a cyan FP-yellow FP-based indicator, Ca2+ and ATP were simultaneously imaged. This palette of indicators paints the way to a colorful new era of Ca2+ imaging.
- 76Nakai, J.; Ohkura, M.; Imoto, K. A High Signal-to-Noise Ca2+ Probe Composed of a Single Green Fluorescent Protein. Nat. Biotechnol. 2001, 19, 137– 141, DOI: 10.1038/8439776https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXhtVaitrw%253D&md5=7d7985a03973490fbf3c5e509ea2b94cA high signal-to-noise Ca2+ probe composed of a single green fluorescent proteinNakai, Junichi; Ohkura, Masamichi; Imoto, KeijiNature Biotechnology (2001), 19 (2), 137-141CODEN: NABIF9; ISSN:1087-0156. (Nature America Inc.)Recently, several groups have developed green fluorescent protein (GFP)-based Ca2+ probes. When applied in cells, however, these probes are difficult to use because of a low signal-to-noise ratio. Here we report the development of a high-affinity Ca2+ probe composed of a single GFP (named G-CaMP). G-CaMP showed an apparent Kd for Ca2+ of 235 μM. Assocn. kinetics of Ca2+ binding were faster at higher Ca2+ concns., with time consts. decreasing from 230 ms at 0.2 μM Ca2+ to 2.5 ms at 1 μM Ca2+. Dissocn. kinetics (τ -200 ms) are independent of Ca2+ concns. In HEK-293 cells and mouse myotubes expressing G-CaMP, large fluorescent changes were obsd. in response to application of drugs or elec. stimulations. G-CaMP will be a useful tool for visualizing intracellular Ca2+ in living cells. Mutational anal., together with previous structural information, suggests the residues that may alter the fluorescence of GFP.
- 77Chiesi, M.; Schwaller, R.; Eichenberger, K. Structural Dependency of the Inhibitory Action of Benzodiazepines and Related Compounds on the Mitochondrial Na+-Ca2+ Exchanger. Biochem. Pharmacol. 1988, 37, 4399– 4403, DOI: 10.1016/0006-2952(88)90623-5There is no corresponding record for this reference.
- 78Luongo, T. S.; Lambert, J. P.; Gross, P.; Nwokedi, M.; Lombardi, A. A.; Shanmughapriya, S.; Carpenter, A. C.; Kolmetzky, D.; Gao, E.; van Berlo, J. H.; Tsai, E. J.; Molkentin, J. D.; Chen, X.; Madesh, M.; Houser, S. R.; Elrod, J. W. The Mitochondrial Na+/Ca2+ Exchanger is Essential for Ca2+ Homeostasis and Viability. Nature 2017, 545, 93– 97, DOI: 10.1038/nature2208278https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXmslehs74%253D&md5=0a3311e8284f11453ef09956541e9a0fThe mitochondrial Na+/Ca2+ exchanger is essential for Ca2+ homeostasis and viabilityLuongo, Timothy S.; Lambert, Jonathan P.; Gross, Polina; Nwokedi, Mary; Lombardi, Alyssa A.; Shanmughapriya, Santhanam; Carpenter, April C.; Kolmetzky, Devin; Gao, Erhe; van Berlo, Jop H.; Tsai, Emily J.; Molkentin, Jeffery D.; Chen, Xiongwen; Madesh, Muniswamy; Houser, Steven R.; Elrod, John W.Nature (London, United Kingdom) (2017), 545 (7652), 93-97CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Mitochondrial calcium (mCa2+) has a central role in both metabolic regulation and cell death signaling, however its role in homeostatic function and disease is controversial. Slc8b1 encodes the mitochondrial Na+/Ca2+ exchanger (NCLX), which is proposed to be the primary mechanism for mCa2+ extrusion in excitable cells. Here we show that tamoxifen-induced deletion of Slc8b1 in adult mouse hearts causes sudden death, with less than 13% of affected mice surviving after 14 days. Lethality correlated with severe myocardial dysfunction and fulminant heart failure. Mechanistically, cardiac pathol. was attributed to mCa2+ overload driving increased generation of superoxide and necrotic cell death, which was rescued by genetic inhibition of mitochondrial permeability transition pore activation. Corroborating these findings, overexpression of NCLX in the mouse heart by conditional transgenesis had the beneficial effect of augmenting mCa2+ clearance, preventing permeability transition and protecting against ischemia-induced cardiomyocyte necrosis and heart failure. These results demonstrate the essential nature of mCa2+ efflux in cellular function and suggest that augmenting mCa2+ efflux may be a viable therapeutic strategy in disease.
- 79Storey, N. M.; Lambert, D. G. Mitochondrial Pharmacology Turns Its Sights on the Ca2+ Uniporter. Cell Death Discov. 2017, 3, 17064, DOI: 10.1038/cddiscovery.2017.6479https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1M%252FjtVGrtw%253D%253D&md5=412a9c4b52c621ffff75e034314a4cf7Mitochondrial pharmacology turns its sights on the Ca(2+) uniporterStorey Nina M; Lambert David GCell death discovery (2017), 3 (), 17064 ISSN:2058-7716.There is no expanded citation for this reference.
- 80Sebag, S. C.; Koval, O. M.; Paschke, J. D.; Winters, C. J.; Comellas, A. P.; Grumbach, I. M. Inhibition of the Mitochondrial Calcium Uniporter Prevents IL-13 and Allergen-Mediated Airway Epithelial Apoptosis and Loss of Barrier Function. Exp. Cell Res. 2018, 362, 400– 411, DOI: 10.1016/j.yexcr.2017.12.00380https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhvF2js73K&md5=4f3e72238fc7003961f65a2cf19e7e3bInhibition of the mitochondrial calcium uniporter prevents IL-13 and allergen-mediated airway epithelial apoptosis and loss of barrier functionSebag, Sara C.; Koval, Olha M.; Paschke, John D.; Winters, Christopher J.; Comellas, Alejandro P.; Grumbach, Isabella M.Experimental Cell Research (2018), 362 (2), 400-411CODEN: ECREAL; ISSN:0014-4827. (Elsevier B.V.)Mitochondria are increasingly recognized as key mediators of acute cellular stress responses in asthma. However, the distinct roles of regulators of mitochondrial physiol. on allergic asthma phenotypes are currently unknown. The mitochondrial Ca2+ uniporter (MCU) resides in the inner mitochondrial membrane and controls mitochondrial Ca2+ uptake into the mitochondrial matrix. To understand the function of MCU in models of allergic asthma, in vitro and in vivo studies were performed using models of functional deficiency or knockout of MCU. In primary human respiratory epithelial cells, MCU inhibition abrogated mitochondrial Ca2+ uptake and reactive oxygen species (ROS) prodn., preserved the mitochondrial membrane potential and protected from apoptosis in response to the pleiotropic Th2 cytokine IL-13. Consequently, epithelial barrier function was maintained with MCU inhibition. Similarly, the endothelial barrier was preserved in respiratory epithelium isolated from MCU-/- mice after exposure to IL-13. In the ovalbumin-model of allergic airway disease, MCU deficiency resulted in decreased apoptosis within the large airway epithelial cells. Concordantly, expression of the tight junction protein ZO-1 was preserved, indicative of maintenance of epithelial barrier function. These data implicate mitochondrial Ca2+ uptake through MCU as a key controller of epithelial cell viability in acute allergic asthma.
- 81Xie, N.; Wu, C.; Wang, C.; Cheng, X.; Zhang, L.; Zhang, H.; Lian, Y. Inhibition of the Mitochondrial Calcium Uniporter Inhibits Aβ-Induced Apoptosis by Reducing Reactive Oxygen Species-Mediated Endoplasmic Reticulum Stress in Cultured Microglia. Brain Res. 2017, 1676, 100– 106, DOI: 10.1016/j.brainres.2017.08.03581https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsFGht77E&md5=bcfa374163b91a61f96a1e42056ea510Inhibition of the mitochondrial calcium uniporter inhibits Aβ-induced apoptosis by reducing reactive oxygen species-mediated endoplasmic reticulum stress in cultured microgliaXie, Nanchang; Wu, Chuanjie; Wang, Cui; Cheng, Xuan; Zhang, Lu; Zhang, Haifeng; Lian, YajunBrain Research (2017), 1676 (), 100-106CODEN: BRREAP; ISSN:0006-8993. (Elsevier B.V.)Amyloid-beta (Aβ) has been shown to induce microglial apoptosis, which is itself sensitive to disturbed mitochondrial calcium (Ca2+) homeostasis. The mitochondrial calcium uniporter (MCU) plays an important regulatory role in mitochondrial Ca2+ homeostasis, but its role in Aβ-induced microglia apoptosis is unknown. In this study, we found increased mitochondrial Ca2+ concn. in Aβ-treated primary microglia and BV-2 cells; also, the MCU inhibitor Ru360 significantly attenuated Aβ-induced microglial apoptosis, whereas the MCU activator spermine augmented it. In addn., Ru360 significantly attenuated Aβ-induced mitochondrial reactive oxygen species (ROS) prodn., as well as endoplasmic reticulum (ER) stress characterized by glucose-regulated protein 78 (GRP78) and C/-EBP homologous protein (CHOP) expression. Spermine, however, exerted the opposite effects on mitochondrial ROS prodn. and ER stress. We also found that mitochondria-targeted antioxidant (Mito-TEMPO) treatment decreased GRP78 and CHOP expression in Aβ-treated microglia. Moreover, blocking endogenous CHOP expression using a CHOP small interfering RNA (siRNA) attenuated Aβ-induced cell death. Altogether, our data suggested that (1) inhibition of MCU exerts a neuroprotective effect on Aβ-induced microglia apoptosis, and (2) that the underlying mechanism may be related to reducing mitochondrial ROS-mediated ER stress.
- 82Alessio, E. Thirty Years of the Drug Candidate NAMI-A and the Myths in the Field of Ruthenium Anticancer Compounds: A Personal Perspective. Eur. J. Inorg. Chem. 2017, 2017, 1549– 1560, DOI: 10.1002/ejic.20160098682https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFKru7bK&md5=ca48413b7dd02a63edf5049a9a5c7377Thirty Years of the Drug Candidate NAMI-A and the Myths in the Field of Ruthenium Anticancer Compounds: A Personal PerspectiveAlessio, EnzoEuropean Journal of Inorganic Chemistry (2017), 2017 (12), 1549-1560CODEN: EJICFO; ISSN:1434-1948. (Wiley-VCH Verlag GmbH & Co. KGaA)As anticipated in the title, this contribution is basically divided into two, strictly connected, parts. The first is a personal overview of the ruthenium drug candidate NAMI-A, almost 30 years after its synthesis and the discovery of its unprecedented antimetastatic properties in animal models at nontoxic dosages. The sections relating to the chem. and biol. behavior of the complex, and the hypotheses on its mechanism(s) of action, are kept to a min., whereas more space is devoted to discussion of the results of the clin. investigations. The second part deals in detail with a no. of undemonstrated misconceptions (or myths) that, over the years, have thrived around NAMI-A and other ruthenium drug candidates, thus neg. affecting the whole field of Ru anticancer drugs.
- 83Wachter, E.; Heidary, D. K.; Howerton, B. S.; Parkin, S.; Glazer, E. C. Light-Activated Ruthenium Complexes Photobind DNA and are Cytotoxic in the Photodynamic Therapy Window. Chem. Commun. 2012, 48, 9649– 9651, DOI: 10.1039/c2cc33359g83https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xht12qsrjJ&md5=dcab2483021e3f4d03850e162ce23167Light-activated ruthenium complexes photobind DNA and are cytotoxic in the photodynamic therapy windowWachter, Erin; Heidary, David K.; Howerton, Brock S.; Parkin, Sean; Glazer, Edith C.Chemical Communications (Cambridge, United Kingdom) (2012), 48 (77), 9649-9651CODEN: CHCOFS; ISSN:1359-7345. (Royal Society of Chemistry)Incorporation of biquinoline ligands into Ru(ii) polypyridyl complexes produces light-activated systems that eject a ligand and photobind DNA after irradn. with visible and near-IR light. Structural anal. shows that distortion facilitates the photochem., and gel shift and cytotoxicity studies prove the compds. act as anti-cancer photodynamic therapy (PDT) agents in the tissue penetrant region.
- 84Han Ang, W.; Dyson, P. J. Classical and Non-Classical Ruthenium-Based Anticancer Drugs: Towards Targeted Chemotherapy. Eur. J. Inorg. Chem. 2006, 2006, 4003– 4018, DOI: 10.1002/ejic.200600723There is no corresponding record for this reference.
- 85Süss-Fink, G. Areneruthenium Complexes as Anticancer Agents. Dalton Trans. 2010, 39, 1673– 1688, DOI: 10.1039/B916860P85https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtlCqtr4%253D&md5=2758ad6804f77d33612401411fe7c116Arene ruthenium complexes as anticancer agentsSuess-Fink, GeorgDalton Transactions (2010), 39 (7), 1673-1688CODEN: DTARAF; ISSN:1477-9226. (Royal Society of Chemistry)A review. Neutral or cationic arene ruthenium complexes providing both hydrophilic as well as hydrophobic properties due to the robustness of the ruthenium-arene unit hold a high potential for the development of metal-based anticancer drugs. Mononuclear arene ruthenium complexes contg. P- or N-donor ligands or N,N-, N,O- or O,O-chelating ligands, dinuclear arene ruthenium systems with adjustable org. linkers, trinuclear arene ruthenium clusters contg. an oxo cap, tetranuclear arene ruthenium porphyrin derivs. that are photoactive, as well as hexanuclear ruthenium cages that are either empty or filled with other mols. have been shown to be active against a variety of cancer cells.
- 86Wang, F.; Chen, H.; Parsons, S.; Oswald, I. D. H.; Davidson, J. E.; Sadler, P. J. Kinetics of Aquation and Anation of Ruthenium(II) Arene Anticancer Complexes, Acidity and X-Ray Structures of Aqua Adducts. Chem. - Eur. J. 2003, 9, 5810– 5820, DOI: 10.1002/chem.20030472486https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhvVGk&md5=7d156972deaa9b35381da9013f931e2dKinetics of aquation and anation of ruthenium(II) arene anticancer complexes, acidity and X-ray structures of aqua adductsWang, Fuyi; Chen, Haimei; Parsons, Simon; Oswald, Iain D. H.; Davidson, James E.; Sadler, Peter J.Chemistry - A European Journal (2003), 9 (23), 5810-5820CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)The aqua adducts of the anticancer complexes [(η6-X)Ru(en)Cl] [PF6] (X = biphenyl (Bip) 1, X = 5,8,9,10-tetrahydroanthracene (THA) 2, X = 9,10-dihydroanthracene (DHA) 3; en = ethylenediamime) were sepd. by HPLC and characterized by mass spectrometry as the products of hydrolysis in H2O. The x-ray structures of the aqua complexes [(η6-X)Ru(en)Y] [PF6]n, X = Bip, Y = 0.5H2O/0.5OH, n = 1.5 (4), X = THA, Y = 0.5H2O/0.5OH, n = 1.5 (5A), X = THA, Y = H2O, n = 2 (5B), and X = DHA, Y = H2O, n = 2(6), are reported. In complex 4 there is a large propeller twist of 45° of the pendant Ph ring with respect to the coordinated Ph ring. Although the THA ligand in 5A and 5B is relatively flat, the DHA ring system in 6 is markedly bent (hinge bend ∼35°) as in the chloro complex 3 (41°). The rates of aquation of 1-3 detd. by UV/visible spectroscopy at various ionic strengths and temps. (1.23-2.59 × 10-3s-1 at 298 K, I = 0.1M) are >20× faster than that of cisplatin. The reverse, anation reactions were very rapid on addn. of 100 mM NaCl (a similar concn. to that in blood plasma). The aquation and anation reactions were about two times faster for the DHA and THA complexes compared to the biphenyl complex. The hydrolysis reactions appear to occur by an associative pathway. The pKa values of the aqua adducts were detd. by 1H NMR spectroscopy as 7.71 for 4, 8.01 for 5 and 7.89 for 6. At physiol.-relevant concns. (0.5-5 μM) and temp. (310 K), the complexes will exist in blood plasma as >89% chloro complex, whereas in the cell nucleus significant amts. (45-65%) of the more reactive aqua adducts would be formed together with smaller amts. of the hydroxo complexes (9-25%, pH 7.4, [Cl-] = 4 mM).
- 87Hartinger, C. G.; Jakupec, M. A.; Zorbas-Seifried, S.; Groessl, M.; Egger, A.; Berger, W.; Zorbas, H.; Dyson, P. J.; Keppler, B. K. KP1019, A New Redox-Active Anticancer Agent - Preclinical Development and Results of a Clinical Phase I Study in Tumor Patients. Chem. Biodiversity 2008, 5, 2140– 2155, DOI: 10.1002/cbdv.20089019587https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsVWktLvO&md5=2d209d437e1c5fefb72827d3eade21d7KP1019, a new redox-active anticancer agent - preclinical development and results of a clinical phase I study in tumor patientsHartinger, Christian G.; Jakupec, Michael A.; Zorbas-Seifried, Stefanie; Groessl, Michael; Egger, Alexander; Berger, Walter; Zorbas, Haralabos; Dyson, Paul J.; Keppler, Bernhard K.Chemistry & Biodiversity (2008), 5 (10), 2140-2155CODEN: CBHIAM; ISSN:1612-1872. (Verlag Helvetica Chimica Acta)A review. The promising drug candidate indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (KP1019) is the second Ru-based anticancer agent to enter clin. trials. In this review, which is an update of a paper from 2006, the exptl. evidence for the proposed mode of action of this coordination compd. is discussed, including transport into the cell via the transferrin cycle and activation by redn. The results of the early clin. development of KP1019 are summarized in which 5 out of 6 evaluated patients experienced disease stabilization with no severe side effects.
- 88Peacock, A. F. A.; Habtemariam, A.; Fernández, R.; Walland, V.; Fabbiani, F. P. A.; Parsons, S.; Aird, R. E.; Jodrell, D. I.; Sadler, P. J. Tuning the Reactivity of Osmium(II) and Ruthenium(II) Arene Complexes under Physiological Conditions. J. Am. Chem. Soc. 2006, 128, 1739– 1748, DOI: 10.1021/ja055886r88https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XjvV2ktQ%253D%253D&md5=f93f88f8995cbf36730b9c69a816dfcaTuning the Reactivity of Osmium(II) and Ruthenium(II) Arene Complexes under Physiological ConditionsPeacock, Anna F. A.; Habtemariam, Abraha; Fernandez, Rafael; Walland, Victoria; Fabbiani, Francesca P. A.; Parsons, Simon; Aird, Rhona E.; Jodrell, Duncan I.; Sadler, Peter J.Journal of the American Chemical Society (2006), 128 (5), 1739-1748CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The OsII arene ethylenediamine (en) complexes [(η6-biphenyl)Os(en)Cl][Z], Z = BPh4 (4) and BF4 (5), are inactive toward A2780 ovarian cancer cells despite 4 being isostructural with an active RuII analog, 4R. Hydrolysis of 5 occurred 40 times more slowly than 4R. The aqua adduct 5A has a low pKa (6.3) compared to that of [(η6-biphenyl)Ru(en)(OH2)]2+ (7.7) and is therefore largely in the hydroxo form at physiol. pH. The rate and extent of reaction of 5 with 9-ethylguanine were also less than those of 4R. The authors replaced the neutral en ligand by anionic acetylacetonate (acac). The complexes [(η6-arene)Os(acac)Cl], arene = biphenyl (6), benzene (7), and p-cymene (8), adopt piano-stool structures similar to those of the RuII analogs and form weak dimers through intermol. (arene)C-H···O(acac) H-bonds. Remarkably, these OsII acac complexes undergo rapid hydrolysis to produce not only the aqua adduct, [(η6-arene)Os(acac)(OH2)]+, but also the hydroxo-bridged dimer, [(η6-arene)Os(μ2-OH)3Os(η6-arene)]+. The pKa values for the aqua adducts 6A, 7A, and 8A (7.1, 7.3, and 7.6, resp.) are lower than that for [(η6-p-cymene)Ru(acac)(OH2)]+ (9.4). Complex 8A rapidly forms adducts with 9-ethylguanine and adenosine, but not with cytidine or thymidine. Despite their reactivity toward nucleobases, complexes 6-8 were inactive toward A549 lung cancer cells. This is attributable to rapid hydrolysis and formation of unreactive hydroxo-bridged dimers which, surprisingly, were the only species present in aq. soln. at biol. relevant concns. Hence, the choice of chelating ligand in OsII (and RuII) arene complexes can have a dramatic effect on hydrolysis behavior and nucleobase binding and provides a means of tuning the reactivity and the potential for discovery of anticancer complexes.
- 89Mühlgassner, G.; Bartel, C.; Schmid, W. F.; Jakupec, M. A.; Arion, V. B.; Keppler, B. K. Biological Activity of Ruthenium and Osmium Arene Complexes with Modified Paullones in Human Cancer Cells. J. Inorg. Biochem. 2012, 116, 180– 187, DOI: 10.1016/j.jinorgbio.2012.06.00389https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsFKnsbzN&md5=331ebaa32b9ab9aa12ad2bfe5a1051e7Biological activity of ruthenium and osmium arene complexes with modified paullones in human cancer cellsMuehlgassner, Gerhard; Bartel, Caroline; Schmid, Wolfgang F.; Jakupec, Michael A.; Arion, Vladimir B.; Keppler, Bernhard K.Journal of Inorganic Biochemistry (2012), 116 (), 180-187CODEN: JIBIDJ; ISSN:0162-0134. (Elsevier)In an attempt to combine the ability of indolobenzazepines (paullones) to inhibit cyclin-dependent kinases (Cdks) and that of platinum-group metal ions to interact with proteins and DNA, ruthenium(II) and osmium(II) arene complexes with paullones were prepd., expecting synergies and an increase of soly. of paullones. Complexes with the general formula [MIICl(η6-p-cymene)L]Cl, where M = Ru (1, 3) or Os (2, 4), and L = L1 (1, 2) or L2 (3, 4), L1 = N-(9-bromo-7,12-dihydroindolo[3,2-d][1]-benzazepin-6(5H)-yliden-N'-(2-hydroxybenzylidene)azine and L2 = N-(9-bromo-7,12-dihydroindolo[3,2-d][1]benzazepin-6-yl)-N'-[3-hydroxy-5-(hydroxymethyl)-2-methylpyridin-4-yl-methylene]azinium chloride (L2*HCl), were now investigated regarding cytotoxicity and accumulation in cancer cells, impact on the cell cycle, capacity of inhibiting DNA synthesis and inducing apoptosis as well as their ability to inhibit Cdk activity. The MTT (3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide) assay yielded IC50 values in the nanomolar to low micromolar range. In accordance with cytotoxicity data, the BrdU assay showed that 1 is the most and 4 the least effective of these compds. regarding inhibition of DNA synthesis. Effects on the cell cycle are minor, although concn.-dependent inhibition of Cdk2/cyclin E activity was obsd. in cell-free expts. Induction of apoptosis is most pronounced for complex 1, accompanied by a low fraction of necrotic cells, as obsd. by annexin V-fluorescein isothiocyanate/propidium iodide staining and flow cytometric anal.
- 90Lameijer, L. N.; Ernst, D.; Hopkins, S. L.; Meijer, M. S.; Askes, S. H. C.; Le Dévédec, S. E.; Bonnet, S. A Red-Light-Activated Ruthenium-Caged NAMPT Inhibitor Remains Phototoxic in Hypoxic Cancer Cells. Angew. Chem., Int. Ed. 2017, 56, 11549– 11553, DOI: 10.1002/anie.20170389090https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtlSmu77L&md5=b5ab704916712872f33ed4d67838e380A Red-Light-Activated Ruthenium-Caged NAMPT Inhibitor Remains Phototoxic in Hypoxic Cancer CellsLameijer, Lucien N.; Ernst, Daniel; Hopkins, Samantha L.; Meijer, Michael S.; Askes, Sven H. C.; Le Devedec, Sylvia E.; Bonnet, SylvestreAngewandte Chemie, International Edition (2017), 56 (38), 11549-11553CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)We describe two water-sol. ruthenium complexes, [1]Cl2 and [2]Cl2, that photodissociate to release a cytotoxic nicotinamide phosphoribosyltransferase (NAMPT) inhibitor with a low dose (21 J cm-2) of red light in an oxygen-independent manner. Using a specific NAMPT activity assay, up to an 18-fold increase in inhibition potency was measured upon red-light activation of [2]Cl2, while [1]Cl2 was thermally unstable. For the first time, the dark and red-light-induced cytotoxicity of these photocaged compds. could be tested under hypoxia (1 % O2). In skin (A431) and lung (A549) cancer cells, a 3- to 4-fold increase in cytotoxicity was found upon red-light irradn. for [2]Cl2, whether the cells were cultured and irradiated with 1 % or 21 % O2. These results demonstrate the potential of photoactivated chemotherapy for hypoxic cancer cells, in which classical photodynamic therapy, which relies on oxygen activation, is poorly efficient.
- 91Dutta, B.; Scolaro, C.; Scopelliti, R.; Dyson, P. J.; Severin, K. Importance of the π-Ligand: Remarkable Effect of the Cyclopentadienyl Ring on the Cytotoxicity of Ruthenium PTA Compounds. Organometallics 2008, 27, 1355– 1357, DOI: 10.1021/om800025a91https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXis1ektLg%253D&md5=ffaf9ea19304cb2698d4ba3aeeefa6c8Importance of the π-Ligand: Remarkable Effect of the Cyclopentadienyl Ring on the Cytotoxicity of Ruthenium PTA CompoundsDutta, Barnali; Scolaro, Claudine; Scopelliti, Rosario; Dyson, Paul J.; Severin, KayOrganometallics (2008), 27 (7), 1355-1357CODEN: ORGND7; ISSN:0276-7333. (American Chemical Society)The water-sol. complexes [(Cp'OR)RuCl(PTA)2] (Cp'OR = η5-1-alkoxy-2,4-di-tert-butyl-3-neopentylcyclopentadienyl; R = Me, Et; PTA = 1,3,5-triaza-7-phosphaadamantane) are considerably more cytotoxic (∼2 orders of magnitude) than the cyclopentadienyl analog [CpRuCl(PTA)2] (i.e., IC50 = 4-10 vs. >1000 μM, depending on the cell line). The structure of [(Cp'OMe)RuCl(PTA)2] is reported, together with that of the precursor [(Cp'OEt)Ru(μ-Cl)]2.
- 92Allardyce, C. S.; Dyson, P. J. Metal-Based Drugs that Break the Rules. Dalton Trans. 2016, 45, 3201– 3209, DOI: 10.1039/C5DT03919C92https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFymurs%253D&md5=75a2d446dd76210a55096041a529ec31Metal-based drugs that break the rulesAllardyce, Claire S.; Dyson, Paul J.Dalton Transactions (2016), 45 (8), 3201-3209CODEN: DTARAF; ISSN:1477-9226. (Royal Society of Chemistry)Cisplatin and other platinum compds. have had a huge impact in the treatment of cancers and are applied in the majority of anticancer chemotherapeutic regimens. The success of these compds. has biased the approaches used to discover new metal-based anticancer drugs. In this perspective we highlight compds. that are apparently incompatible with the more classical (platinum-derived) concepts employed in the development of metal-based anticancer drugs, with respect to both compd. design and the approaches used to validate their utility. Possible design approaches for the future are also suggested.
- 93Kilpin, K. J.; Dyson, P. J. Enzyme Inhibition by Metal Complexes: Concepts, Strategies and Applications. Chem. Sci. 2013, 4, 1410– 1419, DOI: 10.1039/c3sc22349c93https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXjtlyms7c%253D&md5=4619c55c66ea098947112400a47a4c65Enzyme inhibition by metal complexes: concepts, strategies and applicationsKilpin, Kelly J.; Dyson, Paul J.Chemical Science (2013), 4 (4), 1410-1419CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)A review. Metal complexes are increasingly being used to inhibit enzymes. The reasons for this increased interest arise from the special features that metal complexes offer, e.g. the facile construction of 3D architectures that tightly fill enzyme active sites increasing selectivity and the possibility of facile coordination to protein residues that enhances enzyme inhibition. In this review we classify the main modes of enzyme inhibition by metal-based complexes and correlate the enzyme inhibition activity to macroscopic properties such as anticancer activity.
- 94Barry, N. P. E.; Sadler, P. J. Exploration of the Medical Periodic Table: Towards New Targets. Chem. Commun. 2013, 49, 5106– 5131, DOI: 10.1039/c3cc41143e94https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXnsFaru7c%253D&md5=b91db5563e59a43e7f9d05860a03b3b0Exploration of the medical periodic table: towards new targetsBarry, Nicolas P. E.; Sadler, Peter J.Chemical Communications (Cambridge, United Kingdom) (2013), 49 (45), 5106-5131CODEN: CHCOFS; ISSN:1359-7345. (Royal Society of Chemistry)A review. Metallodrugs offer potential for unique mechanisms of drug action based on the choice of the metal, its oxidn. state, the types and no. of coordinated ligands and the coordination geometry. We discuss recent progress in identifying new target sites and elucidating the mechanisms of action of anti-cancer, anti-bacterial, anti-viral, anti-parasitic, anti-inflammatory, and anti-neurodegenerative agents, as well as in the design of metal-based diagnostic agents. Progress in identifying and defining target sites has been accelerated recently by advances in proteomics, genomics and metal speciation anal. Examples of metal compds. and chelating agents (enzyme inhibitors) currently in clin. use, clin. trials or preclin. development are highlighted.
Supporting Information
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acscentsci.8b00773.
Experimental details, complex characterization data, cell viability curves, cell uptake, UV–vis spectra, and dose–response data for Ca2+ uptake inhibition (PDF)
X-ray crystal data for C-2 and C-3 (CIF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.