Natural Compound from Olive Oil Inhibits S100A9 Amyloid Formation and Cytotoxicity: Implications for Preventing Alzheimer’s DiseaseClick to copy article linkArticle link copied!
- Manuela LeriManuela LeriDepartment of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, ItalyDepartment of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, 50139 Florence, ItalyMore by Manuela Leri
- Himanshu ChaudharyHimanshu ChaudharyDepartment of Medical Biochemistry and Biophysics, Umeå University, 90187 Umeå, SwedenMore by Himanshu Chaudhary
- Igor A. IashchishynIgor A. IashchishynDepartment of Medical Biochemistry and Biophysics, Umeå University, 90187 Umeå, SwedenMore by Igor A. Iashchishyn
- Jonathan PansieriJonathan PansieriDepartment of Medical Biochemistry and Biophysics, Umeå University, 90187 Umeå, SwedenMore by Jonathan Pansieri
- Željko M. SvedružićŽeljko M. SvedružićDepartment of Biotechnology, University of Rijeka, HR 51000 Rijeka, CroatiaMore by Željko M. Svedružić
- Silvia Gómez AlcaldeSilvia Gómez AlcaldeDepartment of Medical Biochemistry and Biophysics, Umeå University, 90187 Umeå, SwedenMore by Silvia Gómez Alcalde
- Greta MusteikyteGreta MusteikyteInstitute of Biotechnology, Life Sciences Center, Vilnius University, LT-10257 Vilnius, LithuaniaMore by Greta Musteikyte
- Vytautas SmirnovasVytautas SmirnovasInstitute of Biotechnology, Life Sciences Center, Vilnius University, LT-10257 Vilnius, LithuaniaMore by Vytautas Smirnovas
- Massimo StefaniMassimo StefaniDepartment of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, ItalyMore by Massimo Stefani
- Monica Bucciantini*Monica Bucciantini*Email: [email protected]Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, ItalyMore by Monica Bucciantini
- Ludmilla A. Morozova-Roche*Ludmilla A. Morozova-Roche*Phone: +46736205283, +46907865283. Email: [email protected]Department of Medical Biochemistry and Biophysics, Umeå University, 90187 Umeå, SwedenMore by Ludmilla A. Morozova-Roche
Abstract
Polyphenolic compounds in the Mediterranean diet have received increasing attention due to their protective properties in amyloid neurodegenerative and many other diseases. Here, we have demonstrated for the first time that polyphenol oleuropein aglycone (OleA), which is the most abundant compound in olive oil, has multiple potencies for the inhibition of amyloid self-assembly of pro-inflammatory protein S100A9 and the mitigation of the damaging effect of its amyloids on neuroblastoma SH-SY5Y cells. OleA directly interacts with both native and fibrillar S100A9 as shown by intrinsic fluorescence and molecular dynamic simulation. OleA prevents S100A9 amyloid oligomerization as shown using amyloid oligomer-specific antibodies and cross-β-sheet formation detected by circular dichroism. It decreases the length of amyloid fibrils measured by atomic force microscopy (AFM) as well as reduces the effective rate of amyloid growth and the overall amyloid load as derived from the kinetic analysis of amyloid formation. OleA disintegrates already preformed fibrils of S100A9, converting them into nonfibrillar and nontoxic aggregates as revealed by amyloid thioflavin-T dye binding, AFM, and cytotoxicity assays. At the cellular level, OleA targets S100A9 amyloids already at the membranes as shown by immunofluorescence and fluorescence resonance energy transfer, significantly reducing the amyloid accumulation in GM1 ganglioside containing membrane rafts. OleA increases overall cell viability when neuroblastoma cells are subjected to the amyloid load and alleviates amyloid-induced intracellular rise of reactive oxidative species and free Ca2+. Since S100A9 is both a pro-inflammatory and amyloidogenic protein, OleA may effectively mitigate the pathological consequences of the S100A9-dependent amyloid-neuroinflammatory cascade as well as provide protection from neurodegeneration, if used within the Mediterranean diet as a potential preventive measure.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Note Added after ASAP Publication
This paper was published on May 12, 2021, with an author’s name misspelled. The corrected version was reposted on May 13, 2021.
Introduction
Results and Discussion
Inhibiting S100A9 Amyloid Aggregation by OleA
Interaction of OleA with Native and Amyloid S100A9 followed by Intrinsic Fluorescence
MD Simulation of the OleA Interaction with S100A9
OleA Alleviates S100A9 Induced Cytotoxicity Measured by the MTT Assay
OleA Mitigates the Perturbations in Reactive Oxidative Species (ROS) and Intracellular Ca2+ Levels Induced by S100A9 Amyloids
Effect of S100A9 Amyloids on Differentiated SH-SY5Y Neuroblastoma Cells
OleA Interferes with S100A9 Amyloid Binding to Cell Membrane Rafts Studied by Immunofluorescence and Fluorescence Resonance Energy Transfer (FRET)
Concluding Remarks
Methods
Amyloid Fibril Formation
Preparation of OleA
Dissolving OleA
ThT Fluorescence Assay
AFM Imaging
Dot Blot Analysis
Titration of Native and Aggregated S100A9 by OleA Using Intrinsic Fluorescence
Titration of S100A9 Amyloids by OleA Monitored by ThT Fluorescence
Fitting the Kinetics of S100A9 Amyloid Formation in the Absence and Presence of OleA
Fitting the Titration of S100A9 by OleA Monitored by Intrinsic Fluorescence
Far-UV CD
MD
Molecular Docking Studies
All-Atom Molecular Dynamics Studies
Neuroblastoma Cell Culture
Differentiating Neuroblastoma Cells
MTT Viability Assay
Intracellular ROS Levels
Cytosolic Levels of Free Calcium Measured by Confocal Imaging
Immunofluorescence Confocal Imaging of S100A9 Amyloid Interactions with Cell Membranes
Statistical Analysis
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acschemneuro.0c00828.
Time dependences of ThT fluorescence during OleA incubation; interaction of S100A9 amyloids with A11 amyloid oligomer specific antibodies upon increasing OleA concentration; cell viability and intracellular free Ca2+ level in SH-SY5Y cells affected by S100A9 amyloids; preformed S100A9 fibrils treated with OleA during various times; cytotoxicity of S100A9 fibrils to undifferentiated and differentiated SH-SY5Y cells (PDF)
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
We thank Professor Rakez Kayed, University of Texas, USA, for the gift of amyloid specific antibodies. We acknowledge the Biochemical Imaging Center at Umea University and the National Microscopy Infrastructure (NMI) (VR-RFI 2016-00968) for the use of the microscope. Research is supported by the Swedish Medical Research Council (2019-01232), Forskningsstrategiska medel, Medical Faculty, Umea University and Insamlingsstiftelsen, Umeå for LAM-R, and Research Council of Lithuania (No. S-SEN-20-3) for V.S. and ANCC-COOP/Airalzh ONLUS (Reg. No. 0043966.30-10-359 2014-u) through University of Florence (D.R.595/2016) for M.L. High-performance computing at the University of Rijeka is supported by the European Fund for Regional Development (ERDF) and by the Ministry of Science, Education and Sports of the Republic of Croatia under the project number RC.2.2.06-000.
AD | Alzheimer’s disease |
AFM | atomic force miscroscopy |
CD | circular dichroism |
FRET | fluorescence resonance energy transfer |
MD | molecular dynamics |
OleA | oleuropein aglycone |
PD | Parkinson’s disease |
ROS | reactive oxidative species |
RMSD | root-mean-square deviation |
ThT | thioflavin-T |
References
This article references 69 other publications.
- 1Akiyama, H., Barger, S., Barnum, S., Bradt, B., Bauer, J., Cole, G. M., Cooper, N. R., Eikelenboom, P., Emmerling, M., Fiebich, B. L. (2000) Inflammation and Alzheimer’s disease. Neurobiol. Aging 21 (3), 383– 421, DOI: 10.1016/S0197-4580(00)00124-XGoogle Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXktVSmt74%253D&md5=409a0ca2920ef2cba12474cb7d05f4aaInflammation and Alzheimer's diseaseAkiyama, H.; Barger, S.; Barnum, S.; Bradt, B.; Bauer, J.; Cole, G. M.; Cooper, N. R.; Eikelenboom, P.; Emmerling, M.; Fiebich, B. L.; Finch, C. E.; Frautschy, S.; Griffin, W. S. T.; Hampel, H.; Hull, M.; Landreth, G.; Lue, L.-F.; Mrak, R.; Mackenzie, I. R.; McGeer, P. L.; O'Banion, M. K.; Pachter, J.; Pasinetti, G.; Plata-Salaman, C.; Rogers, J.; Rydel, R.; Shen, Y.; Streit, W.; Strohmeyer, R.; Tooyoma, I.; Van Muiswinkel, F. L.; Veerhuis, R.; Walker, D.; Webster, S.; Wegrzyniak, B.; Wenk, G.; Wyss-Coray, T.Neurobiology of Aging (2000), 21 (3), 383-421CODEN: NEAGDO; ISSN:0197-4580. (Elsevier Science Inc.)A review, with 636 refs. Inflammation clearly occurs in pathol. vulnerable regions of the Alzheimer's disease (AD) brain, and it does so with the full complexity of local peripheral inflammatory responses. In the periphery, degenerating tissue and the deposition of highly insol. abnormal materials are classical stimulants of inflammation. Likewise, in the AD brain damaged neurons and neurites and highly insol. amyloid β peptide deposits and neurofibrillary tangles provide obvious stimuli for inflammation. Because these stimuli are discrete, microlocalized, and present from early preclin. to terminal stages of AD, local upregulation of complement, cytokines, acute-phase reactants, and other inflammatory mediators is also discrete, microlocalized, and chronic. Cumulated over many years, direct and bystander damage from AD inflammatory mechanisms is likely to exacerbate the very pathogenic processes that gave rise to it. Thus, animal models and clin. studies, although still in their infancy, strongly suggest that AD inflammation contributes to AD pathogenesis. By better understanding AD inflammatory and immunoregulatory processes, it should be possible to develop anti-inflammatory approaches that may not cure AD but will likely help slow the progression or delay the onset of this devastating disorder.
- 2Hardy, J. and Selkoe, D. J. (2002) The Amyloid Hypothesis of Alzheimer’ s Disease: Progress and Problems on the Road to Therapeutics. Science 297 (5580), 353, DOI: 10.1126/science.1072994Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38Xls1Cju7s%253D&md5=f7b8db1bc6f13e85f887b73f3042e86eThe amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeuticsHardy, John; Selkoe, Dennis J.Science (Washington, DC, United States) (2002), 297 (5580), 353-356CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)A review. It has been more than 10 yr since it was first proposed that the neurodegeneration in Alzheimer's disease (AD) may be caused by deposition of amyloid β-peptide (Aβ) in plaques in brain tissue. According to the amyloid hypothesis, accumulation of Aβ in the brain is the primary influence driving AD pathogenesis. The rest of the disease process, including formation of neurofibrillary tangles contg. tau protein, is proposed to result from an imbalance between Aβ prodn. and Aβ clearance.
- 3Zhu, S., Wang, J., Zhang, Y., He, J., Kong, J., Wang, J.-F., and Li, X.-M. (2017) The Role of Neuroinflammation and Amyloid in Cognitive Impairment in an APP/PS1 Transgenic Mouse Model of Alzheimer’s Disease. CNS Neurosci. Ther. 23 (4), 310– 320, DOI: 10.1111/cns.12677Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXksFaktr8%253D&md5=34feee5613ff847c8e2615f444238bdcThe role of neuroinflammation and amyloid in cognitive impairment in an APP/PS1 transgenic mouse model of Alzheimer's diseaseZhu, Shenghua; Wang, Junhui; Zhang, Yanbo; He, Jue; Kong, Jiming; Wang, Jun-Feng; Li, Xin-MinCNS Neuroscience & Therapeutics (2017), 23 (4), 310-320CODEN: CNTNAB; ISSN:1755-5930. (Wiley-Blackwell)Aims : Both amyloid deposition and neuroinflammation appear in the early course of Alzheimer's disease (AD). However, the progression of neuroinflammation and its relationship with amyloid deposition and behavioral changes have not been fully elucidated. A better understanding the role of neuroinflammation in AD might extend our current knowledge to therapeutic intervention possibilities. Methods : This study systematically characterized changes in behavioral abnormalities in APP/PS1 transgenic mice. Brain pathol. measures were performed in post-mortem brain tissues of mice from 2 to 22 mo. Results : APP/PS1 mice exhibited significant memory deficits from 5 mo old, which were aggravated at the later stage of life. However, the degree of memory impairments reached a plateau at 12 mo. An early appearance of amyloid plaques was at 3 mo with a linear increase throughout the disease course. CD11b-pos. microglia and glial fibrillary acidic protein-(GFAP) pos. astrocytes were first detected at 3 mo with a close assocn. with amyloid plaques. Yet, the rate of changes in glial activation slowed down from 12 mo despite the steady increase in Aβ. Conclusion : These findings provided evidence that neuroinflammation might be involved in the development and progression of cognitive deficits in APP/PS1 mice, suggesting novel intervention and prevention strategies for AD.
- 4Demuro, A., Mina, E., Kayed, R., Milton, S. C., Parker, I., and Glabe, C. G. (2005) Calcium Dysregulation and Membrane Disruption as a Ubiquitous Neurotoxic Mechanism of Soluble Amyloid Oligomers. J. Biol. Chem. 280 (17), 17294– 17300, DOI: 10.1074/jbc.M500997200Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXjsFOguro%253D&md5=bbecbaadfc5f74d42bd09f2cddcf5763Calcium Dysregulation and Membrane Disruption as a Ubiquitous Neurotoxic Mechanism of Soluble Amyloid OligomersDemuro, Angelo; Mina, Erene; Kayed, Rakez; Milton, Saskia C.; Parker, Ian; Glabe, Charles G.Journal of Biological Chemistry (2005), 280 (17), 17294-17300CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Increasing evidence suggests that amyloid peptides assocd. with a variety of degenerative diseases induce neurotoxicity in their intermediate oligomeric state, rather than as monomers or fibrils. To test this hypothesis and investigate the possible involvement of Ca2+ signaling disruptions in amyloid-induced cytotoxicity, the authors made homogeneous prepns. of disease-related amyloids (Aβ, prion, islet amyloid polypeptide, polyglutamine, and lysozyme) in various aggregation states and tested their actions on fluo-3-loaded SH-SY5Y cells. Application of oligomeric forms of all amyloids tested (0.6-6 μg ml-1) rapidly (∼5 s) elevated intracellular Ca2+, whereas equiv. amts. of monomers and fibrils did not. Ca2+ signals evoked by Aβ42 oligomers persisted after depletion of intracellular Ca2+ stores, and small signals remained in Ca2+-free medium, indicating contributions from both extracellular and intracellular Ca2+ sources. The increased membrane permeability to Ca2+ cannot be attributed to activation of endogenous Ca2+ channels, because responses were unaffected by the potent Ca2+-channel blocker cobalt (20 μm). Instead, observations that Aβ42 and other oligomers caused rapid cellular leakage of anionic fluorescent dyes point to a generalized increase in membrane permeability. The resulting unregulated flux of ions and mols. may provide a common mechanism for oligomer-mediated toxicity in many amyloidogenic diseases, with dysregulation of Ca2+ ions playing a crucial role because of their strong trans-membrane concn. gradient and involvement in cell dysfunction and death.
- 5Perry, V. H., Nicoll, J. A. R., and Holmes, C. (2010) Microglia in Neurodegenerative Disease. Nat. Rev. Neurol. 6 (4), 193– 201, DOI: 10.1038/nrneurol.2010.17Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3c3kslyntw%253D%253D&md5=efb215a8a737c924905e7aa04a5b6706Microglia in neurodegenerative diseasePerry V Hugh; Nicoll James A R; Holmes CliveNature reviews. Neurology (2010), 6 (4), 193-201 ISSN:.Microglia, the resident macrophages of the CNS, are exquisitely sensitive to brain injury and disease, altering their morphology and phenotype to adopt a so-called activated state in response to pathophysiological brain insults. Morphologically activated microglia, like other tissue macrophages, exist as many different phenotypes, depending on the nature of the tissue injury. Microglial responsiveness to injury suggests that these cells have the potential to act as diagnostic markers of disease onset or progression, and could contribute to the outcome of neurodegenerative diseases. The persistence of activated microglia long after acute injury and in chronic disease suggests that these cells have an innate immune memory of tissue injury and degeneration. Microglial phenotype is also modified by systemic infection or inflammation. Evidence from some preclinical models shows that systemic manipulations can ameliorate disease progression, although data from other models indicates that systemic inflammation exacerbates disease progression. Systemic inflammation is associated with a decline in function in patients with chronic neurodegenerative disease, both acutely and in the long term. The fact that diseases with a chronic systemic inflammatory component are risk factors for Alzheimer disease implies that crosstalk occurs between systemic inflammation and microglia in the CNS.
- 6Paradisi, S., Sacchetti, B., Balduzzi, M., Gaudi, S., and Malchiodi-Albedi, F. (2004) Astrocyte Modulation of In Vitro β-Amyloid Neurotoxicity. Glia 46 (3), 252– 260, DOI: 10.1002/glia.20005Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2c7lsVerug%253D%253D&md5=d8fce7b7e08c280b10ed9f8b444a8936Astrocyte modulation of in vitro beta-amyloid neurotoxicityParadisi Silvia; Sacchetti Benedetto; Balduzzi Maria; Gaudi Simona; Malchiodi-Albedi FiorellaGlia (2004), 46 (3), 252-60 ISSN:0894-1491.In Alzheimer's disease brain, beta-amyloid (Abeta) deposition is accompanied by astrocyte activation, whose role in the pathogenesis of the disease is still unclear. To explore the subject, we compared Abeta neurotoxicity in pure hippocampal cultures and neuronal-astrocytic cocultures, where astrocytes conditioned neurons but were not in contact with them or Abeta. In the presence of astrocytes, neurons were protected from Abeta neurotoxicity. Neuritic dystrophy was reduced, synapses were partially preserved, and apoptosis was contrasted. The protection disappeared when astrocytes were also treated with Abeta, suggesting that Abeta-astrocyte interaction is deleterious for neurons. This was supported by comparing Abeta neurotoxicity in pure neurons and neurons grown on astrocytes. In this case, where astrocytes were also in contact with Abeta, neuritic damage was enhanced and expression of synaptic vesicle proteins decreased. Our results suggest that astrocytes can protect neurons from Abeta neurotoxicity, but when they interact with Abeta, the protection is undermined and neurotoxicity enhanced.
- 7Eikelenboom, P., Hoozemans, J. J., Veerhuis, R., van Exel, E., Rozemuller, A. J., and van Gool, W. A. (2012) Whether, When and How Chronic Inflammation Increases the Risk of Developing Late-onset Alzheimer’s Disease. Alzheimer's Res. Ther. 4 (3), 15– 15, DOI: 10.1186/alzrt118Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xpt1ajurw%253D&md5=a2a92116ef2fb0e70170b35ee945713cWhether, when and how chronic inflammation increases the risk of developing late-onset Alzheimer's diseaseEikelenboom, Piet; Hoozemans, Jeroen J. M.; Veerhuis, Rob; van Exel, Eric; Rozemuller, Annemieke J. M.; van Gool, Willem A.Alzheimer's Research & Therapy (2012), 4 (3), 15CODEN: ARTLCD; ISSN:1758-9193. (BioMed Central Ltd.)A review. Neuropathol. studies have revealed the presence of a broad variety of inflammation-related proteins (complement factors, acute-phase proteins, pro-inflammatory cytokines) in Alzheimer's disease (AD) brains. These constituents of innate immunity are involved in several crucial pathogenic events of the underlying pathol. cascade in AD and recent studies have shown that innate immunity is involved in the etiol. of late-onset AD. Genome-wide assocn. studies have demonstrated gene loci that are linked to the complement system. Neuropathol. and exptl. studies indicate that fibrillar amyloid-β (Aβ) can activate the innate immunity-related CD14 and Toll-like receptor signaling pathways of glial cells for pro-inflammatory cytokine prodn. The prodn. capacity of this pathway is under genetic control and offspring with a parental history of late-onset AD have a higher prodn. capacity for pro-inflammatory cytokines. The activation of microglia by fibrillar Aβ deposits in the early preclin. stages of AD can make the brain susceptible later on for a second immune challenge leading to enhanced prodn. of pro-inflammatory cytokines. An example of a second immune challenge could be systemic inflammation in patients with preclin. AD. Prospective epidemiol. studies show that elevated serum levels of acute phase reactants can be considered as a risk factor for AD. Clin. studies suggest that peripheral inflammation increases the risk of dementia, esp. in patients with preexistent cognitive impairment and accelerates further deterioration in demented patients. The view that peripheral inflammation can increase the risk of dementia in older people provides scope for prevention.
- 8Lim, S., Chun, Y., Lee, J. S., and Lee, S.-J. (2016) Neuroinflammation in Synucleinopathies. Brain Pathol. 26 (3), 404– 409, DOI: 10.1111/bpa.12371Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC28jmslyjsw%253D%253D&md5=0db4eb9a45b73151df154d7ce22d55a9Neuroinflammation in SynucleinopathiesLim Somin; Chun Yewon; Lee Jun Sung; Lee Seung-JaeBrain pathology (Zurich, Switzerland) (2016), 26 (3), 404-9 ISSN:.The causes of most neurodegenerative diseases are attributed to multiple genetic and environmental factors interacting with one another. Above all, inflammation in the nervous system has been implicated in many neurodegenerative diseases. Still, the roles of neuroinflammation in disease mechanisms and the triggers of inflammatory responses in disease-inflicted brain tissues seem to remain unclear. This review will examine previous studies that had been done from genetic, pathological and epidemiological perspectives. These studies assess the involvement of neuroinflammation in synucleinopathies, a group of neurodegenerative diseases that are characterized by deposition of α-synuclein aggregates such as Parkinson's disease, dementia with Lewy bodies and multiple system atrophy. The review will also discuss the role of α-synuclein aggregates in triggering inflammatory responses from glial cells. It is expected that a precise assessment of the roles and mechanisms of neuroinflammation in neurodegenerative diseases will pave the way for the development of disease-modifying drugs.
- 9Wang, C., Iashchishyn, I. A., Pansieri, J., Nystrom, S., Klementieva, O., Kara, J., Horvath, I., Moskalenko, R., Rofougaran, R., Gouras, G. (2018) S100A9-Driven Amyloid-Neuroinflammatory Cascade in Traumatic Brain Injury as a Precursor State for Alzheimer’s Disease. Sci. Rep. 8, 12836, DOI: 10.1038/s41598-018-31141-xGoogle Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3c3itlOitw%253D%253D&md5=f4623ac4ea93ef3ab98aa20a3c70f5cfS100A9-Driven Amyloid-Neuroinflammatory Cascade in Traumatic Brain Injury as a Precursor State for Alzheimer's DiseaseWang Chao; Iashchishyn Igor A; Pansieri Jonathan; Kara John; Horvath Istvan; Moskalenko Roman; Rofougaran Reza; Morozova-Roche Ludmilla A; Iashchishyn Igor A; Nystrom Sofie; Klementieva Oxana; Gouras Gunnar; Moskalenko Roman; Kovacs Gabor G; Shankar S KScientific reports (2018), 8 (1), 12836 ISSN:.Pro-inflammatory and amyloidogenic S100A9 protein is an important contributor to Alzheimer's disease (AD) pathology. Traumatic brain injury (TBI) is viewed as a precursor state for AD. Here we have shown that S100A9-driven amyloid-neuroinflammatory cascade was initiated in TBI and may serve as a mechanistic link between TBI and AD. By analyzing the TBI and AD human brain tissues, we demonstrated that in post-TBI tissues S100A9, produced by neurons and microglia, becomes drastically abundant compared to Aβ and contributes to both precursor-plaque formation and intracellular amyloid oligomerization. Conditions implicated in TBI, such as elevated S100A9 concentration, acidification and fever, provide strong positive feedback for S100A9 nucleation-dependent amyloid formation and delay in its proteinase clearance. Consequently, both intracellular and extracellular S100A9 oligomerization correlated with TBI secondary neuronal loss. Common morphology of TBI and AD plaques indicated their similar initiation around multiple aggregation centers. Importantly, in AD and TBI we found S100A9 plaques without Aβ. S100A9 and Aβ plaque pathology was significantly advanced in AD cases with TBI history at earlier age, signifying TBI as a risk factor. These new findings highlight the detrimental consequences of prolonged post-TBI neuroinflammation, which can sustain S100A9-driven amyloid-neurodegenerative cascade as a specific mechanism leading to AD development.
- 10Horvath, I., Iashchishyn, I. A., Moskalenko, R. A., Wang, C., Warmlander, S. K. T. S., Wallin, C., Graslund, A., Kovacs, G. G., and Morozova-Roche, L. A. (2018) Co-aggregation of Pro-inflammatory S100A9 with Alpha-Synuclein in Parkinson’s Disease: Ex Vivo and In Vitro Studies. J. Neuroinflammation 15, 172, DOI: 10.1186/s12974-018-1210-9Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisFWgurrK&md5=713ff8fce99cbbd976b097ec98515dbcCo-aggregation of pro-inflammatory S100A9 with α-synuclein in Parkinson's disease: ex vivo and in vitro studiesHorvath, Istvan; Iashchishyn, Igor A.; Moskalenko, Roman A.; Wang, Chao; Waermlaender, Sebastian K. T. S.; Wallin, Cecilia; Graeslund, Astrid; Kovacs, Gabor G.; Morozova-Roche, Ludmilla A.Journal of Neuroinflammation (2018), 15 (), 172/1-172/16CODEN: JNOEB3; ISSN:1742-2094. (BioMed Central Ltd.)Background: Chronic neuroinflammation is a hallmark of Parkinson's disease (PD) pathophysiol., assocd. with increased levels of pro-inflammatory factors in PD brain tissues. The pro-inflammatory mediator and highly amyloidogenic protein S100A9 is involved in the amyloid-neuroinflammatory cascade in Alzheimer's disease. This is the first report on the co-aggregation of α-synuclein (α-syn) and S100A9 both in vitro and ex vivo in PD brain. Methods: Single and sequential immunohistochem., immunofluorescence, scanning electron and at. force (AFM) microscopies were used to analyze the ex vivo PD brain tissues for S100A9 and α-syn location and aggregation. In vitro studies revealing S100A9 and α-syn interaction and co-aggregation were conducted by NMR, CD, Thioflavin-T fluorescence, AFM, and surface plasmon resonance methods. Results: Co-localized and co-aggregated S100A9 and α-syn were found in 20% Lewy bodies and 77% neuronal cells in the substantia nigra; both proteins were also obsd. in Lewy bodies in PD frontal lobe (Braak stages 4-6). Lewy bodies were characterized by ca. 10-23 μm outer diam., with S100A9 and α-syn being co-localized in the same lamellar structures. S100A9 was also detected in neurons and blood vessels of the aged patients without PD, but in much lesser extent. In vitro S100A9 and α-syn were shown to interact with each other via the α-syn C-terminus with an apparent dissocn. const. of ca. 5 μM. Their co-aggregation occurred significantly faster and led to formation of larger amyloid aggregates than the self-assembly of individual proteins. S100A9 amyloid oligomers were more toxic than those of α-syn, while co-aggregation of both proteins mitigated the cytotoxicity of S100A9 oligomers. Conclusions: We suggest that sustained neuroinflammation promoting the spread of amyloidogenic S100A9 in the brain tissues may trigger the amyloid cascade involving α-syn and S100A9 and leading to PD, similar to the effect of S100A9 and Aβ co-aggregation in Alzheimer's disease. The finding of S100A9 involvement in PD may open a new avenue for therapeutic interventions targeting S100A9 and preventing its amyloid self-assembly in affected brain tissues.
- 11Wang, C., Klechikov, A. G., Gharibyan, A. L., Wärmländer, S. K. T. S., Jarvet, J., Zhao, L., Jia, X., Narayana, V. K., Shankar, S. K., Olofsson, A. (2014) The Role of Pro-inflammatory S100A9 in Alzheimer’s Disease Amyloid-neuroinflammatory Cascade. Acta Neuropathol. 127 (4), 507– 522, DOI: 10.1007/s00401-013-1208-4Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslymu7bF&md5=af7f01a5c3f6f24fc24aa907b72e9e43The role of pro-inflammatory S100A9 in Alzheimer's disease amyloid-neuroinflammatory cascadeWang, Chao; Klechikov, Alexey G.; Gharibyan, Anna L.; Waermlaender, Sebastian K. T. S.; Jarvet, Juri; Zhao, Lina; Jia, Xueen; Shankar, S. K.; Olofsson, Anders; Braennstroem, Thomas; Mu, Yuguang; Graeslund, Astrid; Morozova-Roche, Ludmilla A.Acta Neuropathologica (2014), 127 (4), 507-522CODEN: ANPTAL; ISSN:0001-6322. (Springer)Pro-inflammatory S100A9 protein is increasingly recognized as an important contributor to inflammation-related neurodegeneration. Here, we provide insights into S100A9 specific mechanisms of action in Alzheimer's disease (AD). Due to its inherent amyloidogenicity S100A9 contributes to amyloid plaque formation together with Aβ. In traumatic brain injury (TBI) S100A9 itself rapidly forms amyloid plaques, which were reactive with oligomer-specific antibodies, but not with Aβ and amyloid fibrillar antibodies. They may serve as precursor-plaques for AD, implicating TBI as an AD risk factor. S100A9 was obsd. in some hippocampal and cortical neurons in TBI, AD and non-demented aging. In vitro S100A9 forms neurotoxic linear and annular amyloids resembling Aβ protofilaments. S100A9 amyloid cytotoxicity and native S100A9 pro-inflammatory signaling can be mitigated by its co-aggregation with Aβ, which results in a variety of micron-scale amyloid complexes. NMR and mol. docking demonstrated transient interactions between native S100A9 and Aβ. Thus, abundantly present in AD brain pro-inflammatory S100A9, possessing also intrinsic amyloidogenic properties and ability to modulate Aβ aggregation, can serve as a link between the AD amyloid and neuroinflammatory cascades and as a prospective therapeutic target.
- 12Pruenster, M., Vogl, T., Roth, J., and Sperandio, M. (2016) S100A8/A9: From Basic Science to Clinical Application. Pharmacol. Ther. 167, 120– 131, DOI: 10.1016/j.pharmthera.2016.07.015Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtlWjurzJ&md5=99ce16a70a32600c9f393ae6bfef9b90S100A8/A9: From basic science to clinical applicationPruenster, Monika; Vogl, Thomas; Roth, Johannes; Sperandio, MarkusPharmacology & Therapeutics (2016), 167 (), 120-131CODEN: PHTHDT; ISSN:0163-7258. (Elsevier)Neutrophils and monocytes belong to the first line of immune defense cells and are recruited to sites of inflammation during infection or sterile injury. Both cells contain huge amts. of the heterodimeric protein S100A8/A9 in their cytoplasm. S100A8/A9 belongs to the Ca2 + binding S100 protein family and has recently gained a lot of interest as a crit. alarmin modulating the inflammatory response after its release (extracellular S100A8/A9) from neutrophils and monocytes. Extracellular S100A8/A9 interacts with the pattern recognition receptors Toll-like receptor 4 (TLR4) and Receptor for Advanced Glycation Endproducts (RAGE) promoting cell activation and recruitment. Besides its biol. function, S100A8/A9 (also known as myeloid related protein 8/14, MRP8/14) was identified as interesting biomarker to monitor disease activity in chronic inflammatory disorders including inflammatory bowel disease and rheumatoid arthritis. Furthermore, S100A8/A9 has been tested successfully in pre-clin. imaging studies to localize sites of infection or sterile injury. Finally, recent evidence using small mol. inhibitors for S100A8/A9 also suggests that blocking S100A8/A9 activity exerts beneficial effects on disease activity in animal models of autoimmune diseases including multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis and inflammatory bowel disease. This review will provide a comprehensive and detailed overview into the structure and biol. function of S100A8/A9 and also will give an outlook in terms of diagnostic and therapeutic applications targeting S100A8/A9.
- 13Markowitz, J. and Carson, W. E., 3rd. (2013) Review of S100A9 Biology and Its Role in Cancer. Biochim. Biophys. Acta, Rev. Cancer 1835 (1), 100– 109, DOI: 10.1016/j.bbcan.2012.10.003Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhvVKqurzJ&md5=b417186ba1010bf09ea0e350e2acd708Review of S100A9 biology and its role in cancerMarkowitz, Joseph; Carson, William E.Biochimica et Biophysica Acta, Reviews on Cancer (2013), 1835 (1), 100-109CODEN: BBACEU; ISSN:0304-419X. (Elsevier B.V.)A review. S100A9 is a calcium binding protein with multiple ligands and post-translation modifications that is involved in inflammatory events and the initial development of the cancer cell through to the development of metastatic disease. This review has a threefold purpose: 1) describe the S100A9 structural elements important for its biol. activity, 2) describe the S100A9 biol. in the context of the immune system, and 3) illustrate the role of S100A9 in the development of malignancy via interactions with the immune system and other cellular processes.
- 14Schluesener, H. J., Kremsner, P. G., and Meyermann, R. (1998) Widespread Expression of MRP8 and MRP14 in Human Cerebral Malaria by Microglial Cells. Acta Neuropathol. 96 (6), 575– 580, DOI: 10.1007/s004010050938Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXmvFyitLo%253D&md5=4db5ed68a417b831c80894978e554c30Widespread expression of MRP8 and MRP14 in human cerebral malaria by microglial cellsSchluesener, H. J.; Kremsner, Peter G.; Meyermann, RichardActa Neuropathologica (1998), 96 (6), 575-580CODEN: ANPTAL; ISSN:0001-6322. (Springer-Verlag)Human cerebral malaria (CM) is an often fatal infection. The cascades of signaling events resulting in tissue trauma and coma are only slowly becoming unraveled. Here the authors report that microglial cells, sensitive cellular sensors of threats to the central nervous system, in CM express the myeloid-related proteins MRP8 (S100A8) and MRP14 (S100A9), Ca2+-binding sensor proteins of activated monocytes. Surprisingly, microglial activation was widespread throughout the brain in white and gray matter and not limited to areas of petechial bleedings or sequestration of infected erythrocytes. Further, apoptosis/necrosis is prominent in CM; not only leukocytes appeared apoptotic, neurons also appeared damaged and DNA fragmentation was revealed by in situ nick translation. Thus, a prominent feature of human CM is activation of microglia, and anal. of these reactive microglia might further promote the authors' understanding of CM pathol. and guide development of future therapeutic intervention of the local reactive processes.
- 15Postler, E., Lehr, A., Schluesener, H., and Meyermann, R. (1997) Expression of the S-100 proteins MRP-8 and −14 in Ischemic Brain Lesions. Glia 19 (1), 27– 34, DOI: 10.1002/(SICI)1098-1136(199701)19:1<27::AID-GLIA3>3.0.CO;2-7Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK2s7ktlGmtg%253D%253D&md5=b269af3ced945688d66f310da4d3e5dcExpression of the S-100 proteins MRP-8 and -14 in ischemic brain lesionsPostler E; Lehr A; Schluesener H; Meyermann RGlia (1997), 19 (1), 27-34 ISSN:0894-1491.So far, microglial activation in cerebral ischemia has only been studied in different animal models. We have investigated the activation of microglial cells in human cerebral ischemia. As a marker for the activation of these "brain macrophages," we have used the macrophage inhibitor factor-related-proteins MRP-8 and MRP-14, which belong to the calcium binding S-100 protein family. The proteins can be detected on microglial cells in bacterial encephalitis and Alzheimer's disease but have so far not been studied in non-inflammatory diseases, in which microglial activation also occurs. Antibodies against MRP-8 and -14 detected ramified microglial cells within the first 3 days after cerebral infarction. Labeled cells were found selectively in the periinfarctional area. To support the notion that these cells belong to the locally activated resident microglial population, we studied their proliferation rate by staining the Ki-67 antigen with the antibody MIB-1. Double-labeling clearly showed that in the early phase of cerebral infarction microglial cells in the periinfarctional area express MRP-8 and -14 and also proliferate. Surprisingly, MRPs are expressed no longer than 3 days post infarction. This indicates that the activation of the resident microglia is an early step of tissue reaction after cerebral infarction. Additionally, we found evidence that microglial cells contribute to the population of phagocytes only during the first 3 days post infarction. The majority of lipid phagocytes found in the later stages are obviously recruited from the blood-borne macrophage pool.
- 16Nagareddy, P. R., Murphy, A. J., Stirzaker, R. A., Hu, Y., Yu, S., Miller, R. G., Ramkhelawon, B., Distel, E., Westerterp, M., Huang, L.-S. (2013) Hyperglycemia Promotes Myelopoiesis and Impairs the Resolution of Atherosclerosis. Cell Metab. 17 (5), 695– 708, DOI: 10.1016/j.cmet.2013.04.001Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXnsVyqs7Y%253D&md5=ff11d96d88b7b22191ca54f90233b5d4Hyperglycemia Promotes Myelopoiesis and Impairs the Resolution of AtherosclerosisNagareddy, Prabhakara R.; Murphy, Andrew J.; Stirzaker, Roslynn A.; Hu, Yunying; Yu, Shiquing; Miller, Rachel G.; Ramkhelawon, Bhama; Distel, Emilie; Westerterp, Marit; Huang, Li-Shin; Schmidt, Ann Marie; Orchard, Trevor J.; Fisher, Edward A.; Tall, Alan R.; Goldberg, Ira J.Cell Metabolism (2013), 17 (5), 695-708CODEN: CMEEB5; ISSN:1550-4131. (Elsevier Inc.)Diabetes is a major risk factor for atherosclerosis. Although atherosclerosis is initiated by deposition of cholesterol-rich lipoproteins in the artery wall, the entry of inflammatory leukocytes into lesions fuels disease progression and impairs resoln. We show that diabetic mice have increased nos. of circulating neutrophils and Ly6-Chi monocytes, reflecting hyperglycemia-induced proliferation and expansion of bone marrow myeloid progenitors and release of monocytes into the circulation. Increased neutrophil prodn. of S100A8/S100A9, and its subsequent interaction with the receptor for advanced glycation end products on common myeloid progenitor cells, leads to enhanced myelopoiesis. Treatment of hyperglycemia reduces monocytosis, entry of monocytes into atherosclerotic lesions, and promotes regression. In patients with type 1 diabetes, plasma S100A8/S100A9 levels correlate with leukocyte counts and coronary artery disease. Thus, hyperglycemia drives myelopoiesis and promotes atherogenesis in diabetes.
- 17Ma, L.-P., Haugen, E., Ikemoto, M., Fujita, M., Terasaki, F., and Fu, M. (2012) S100A8/A9 Complex as a New Biomarker in Prediction of Mortality in Elderly Patients with Severe Heart Failure. Int. J. Cardiol. 155 (1), 26– 32, DOI: 10.1016/j.ijcard.2011.01.082Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC383gsFyiuw%253D%253D&md5=9e31f29e08d75738e968ef7fbe218022S100A8/A9 complex as a new biomarker in prediction of mortality in elderly patients with severe heart failureMa Li-Ping; Haugen Espen; Ikemoto Masaki; Fujita Masatoshi; Terasaki Fumio; Fu MichaelInternational journal of cardiology (2012), 155 (1), 26-32 ISSN:.BACKGROUND: S100A8/A9 complex is a new inflammation-related protein and has a positive correlation with C-reaction protein level. However its role in chronic heart failure (CHF) remains unclear. METHODS AND RESULTS: Circulating levels of S100A8/A9 complex and other biomarkers (IL-6, IL-8, TNF-α, and BNP) were measured in CHF (n = 54) and hypertensive without CHF (n = 31) as well as healthy subjects (n = 23), with follow up to 1480 days. During follow-up, cumulative mortality rate for CHF patients was 63%. Plasma levels of S100A8/A9 complex, IL-6, IL-8 and TNF-α were significantly higher in CHF than the hypertensive patients and healthy subjects. A significant positive correlation was found between S100A8/A9 complex and IL-6 and IL-8 respectively. Cox regression analysis showed that IL-6 and IL-8 were predictors for mortality for 6 months, and S100A8/A9 complex, IL-6, IL-8 and age were predictors for mortality for one year whereas BNP, TNF-α, IL-6 and IL-8 remained predictors for mortality for two years. A combination of S100A8/A9 complex and IL-6 provided powerful predictive value in mortality for both 6 and 12 months. CONCLUSIONS: S100A8/A9 complex is a useful biomarker as a predictor for one year mortality and its combination with IL-6 is able to provide additive prognostic information in this vulnerable heart failure population in the elderly.
- 18Swindell, W. R., Johnston, A., Xing, X., Little, A., Robichaud, P., Voorhees, J. J., Fisher, G., and Gudjonsson, J. E. (2013) Robust Shifts in S100A9 Expression with Aging: A Novel Mechanism for Chronic Inflammation. Sci. Rep. 3 (1), 1215, DOI: 10.1038/srep01215Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXpvFWjt74%253D&md5=8ff1b1f10a81db6c7d8496626fc2c72bRobust shifts in S100a9 expression with aging: a novel mechanism for chronic inflammationSwindell, William R.; Johnston, Andrew; Xing, Xianying; Little, Andrew; Robichaud, Patrick; Voorhees, John J.; Fisher, Gary; Gudjonsson, Johann E.Scientific Reports (2013), 3 (), 1215, 13 pp.CODEN: SRCEC3; ISSN:2045-2322. (Nature Publishing Group)The S100a8 and S100a9 genes encode a pro-inflammatory protein (calgranulin) that has been implicated in multiple diseases. However, involvement of S100a8/a9 in the basic mechanisms of intrinsic aging has not been established. In this study, we show that shifts in the abundance of S100a8 and S100a9 mRNA are a robust feature of aging in mammalian tissues, involving a range of cell types including the central nervous system. To identify transcription factors that control S100a9 expression, we performed a large-scale transcriptome anal. of 62 mouse and human cell types. We identified cell type-specific trends, as well as robust assocns. linking S100a9 coexpression to elevated frequency of ETS family motifs and in particular, to motifs recognized by the transcription factor SPI/PU.1. Sparse occurrence of SATB1 motifs was also a strong predictor of S100a9 coexpression. These findings offer support for a novel mechanism by which a SPI1/PU.1-S100a9 axis sustains chronic inflammation during aging.
- 19Iashchishyn, I. A., Sulskis, D., Nguyen Ngoc, M., Smirnovas, V., and Morozova-Roche, L. A. (2017) Finke–Watzky Two-Step Nucleation–Autocatalysis Model of S100A9 Amyloid Formation: Protein Misfolding as “Nucleation” Event. ACS Chem. Neurosci. 8 (10), 2152– 2158, DOI: 10.1021/acschemneuro.7b00251Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXht1Cltb3P&md5=9e6270edc1a9e7e2d0277ab483d7f14fFinke-Watzky Two-Step Nucleation-Autocatalysis Model of S100A9 Amyloid Formation: Protein Misfolding as "Nucleation" EventIashchishyn, Igor A.; Sulskis, Darius; Nguyen Ngoc, Mai; Smirnovas, Vytautas; Morozova-Roche, Ludmilla A.ACS Chemical Neuroscience (2017), 8 (10), 2152-2158CODEN: ACNCDM; ISSN:1948-7193. (American Chemical Society)Quant. kinetic anal. is crit. for understanding amyloid mechanisms. Here, we demonstrate the application of the generic Finke-Watzky (F-W; 1997, 2008) 2-step nucleation-autocatalytic growth model to concn.-dependent amyloid kinetics of proinflammatory α-helical S100A9 protein/calgranulin B at pH 7.4 and temps. of 37 and 42°. The model is based on 2 pseudo-elementary reaction steps applied without further anal. constraints and its treatment of S100A9 amyloid self-assembly demonstrates that initial misfolding and β-sheet formation, defined as a "nucleation" step, spontaneously takes place within individual S100A9 mols. at a higher rate than the subsequent fibrillar growth. The latter, described as an autocatalytic process, will proceed if misfolded amyloid-prone S100A9 is populated on a macroscopic time scale. Short lengths of S100A9 fibrils were consistent with the F-W model. The anal. of fibrillar length distribution by the Bekker-Doering model demonstrated independently that such distribution was solely detd. by slow fibril growth and that there was no fragmentation or secondary pathways decreasing fibrillar length.
- 20Yanamandra, K., Alexeyev, O., Zamotin, V., Srivastava, V., Shchukarev, A., Brorsson, A.-C., Tartaglia, G. G., Vogl, T., Kayed, R., Wingsle, G. (2009) Amyloid Formation by the Pro-inflammatory S100A8/A9 Proteins in the Ageing Prostate. PLoS One 4 (5), e5562– e5562, DOI: 10.1371/journal.pone.0005562Google ScholarThere is no corresponding record for this reference.
- 21Eremenko, E., Ben-Zvi, A., Morozova-Roche, L. A., and Raveh, D. (2013) Aggregation of Human S100A8 and S100A9 Amyloidogenic Proteins Perturbs Proteostasis in a Yeast Model. PLoS One 8 (3), e58218 DOI: 10.1371/journal.pone.0058218Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXktlWjsrg%253D&md5=adb676af9433824aad66ff794da89062Aggregation of human S100A8 and S100A9 amyloidogenic proteins perturbs proteostasis in a yeast modelEremenko, Ekaterina; Ben-Zvi, Anat; Morozova-Roche, Ludmilla A.; Raveh, DinaPLoS One (2013), 8 (3), e58218CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)Amyloid aggregates of the calcium-binding EF-hand proteins S100A8 and S100A9 have been found in the corpora amylacea of patients with prostate cancer and may play a role in carcinogenesis. Here, the authors present a novel model system using the yeast Saccharomyces cerevisiae to study human S100A8 and S100A9 aggregation and toxicity. They found that S100A8, S100A9 and S100A8/9 cotransfomants form SDS-resistant non-toxic aggregates in yeast cells. Using fluorescently tagged proteins, the authors showed that S100A8 and S100A9 accumulate in foci. After prolonged induction, S100A8 foci localized to the cell vacuole, whereas the S100A9 foci remained in the cytoplasm when present alone, but entered the vacuole in cotransformants. Biochem. anal. of the proteins indicated that S100A8 and S100A9 alone or coexpressed together form amyloid-like aggregates in yeast. Expression of S100A8 and S100A9 in wild-type yeast did not affect cell viability, but these proteins were toxic when expressed on a background of unrelated metastable temp.-sensitive mutant proteins, Cdc53-1p, Cdc34-2p, Srp1-31p and Sec27-1p. This finding suggests that the expression and aggregation of S100A8 and S100A9 may limit the capacity of the cellular proteostasis machinery. To test this hypothesis, the authors screened a set of chaperone deletion mutants and found that reducing the levels of the heat-shock proteins Hsp104p and Hsp70p was sufficient to induce S100A8 and S100A9 toxicity. This result indicates that the chaperone activity of the Hsp104/Hsp70 bichaperone system in wild type cells is sufficient to reduce S100A8 and S100A9 amyloid toxicity and preserve cellular proteostasis. Expression of human S100A8 and S100A9 in yeast thus provides a novel model system for the study of the interaction of amyloid deposits with the proteostasis machinery.
- 22Horvath, I., Jia, X., Johansson, P., Wang, C., Moskalenko, R., Steinau, A., Forsgren, L., Wågberg, T., Svensson, J., Zetterberg, H., and Morozova-Roche, L. A. (2016) Pro-inflammatory S100A9 Protein as a Robust Biomarker Differentiating Early Stages of Cognitive Impairment in Alzheimer’s Disease. ACS Chem. Neurosci. 7 (1), 34– 39, DOI: 10.1021/acschemneuro.5b00265Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhslyktbjM&md5=9a2362b63b01d23b69f4dd2b4e6c5c88Pro-inflammatory S100A9 Protein as a Robust Biomarker Differentiating Early Stages of Cognitive Impairment in Alzheimer's DiseaseHorvath, Istvan; Jia, Xueen; Johansson, Per; Wang, Chao; Moskalenko, Roman; Steinau, Andreas; Forsgren, Lars; Waagberg, Thomas; Svensson, Johan; Zetterberg, Henrik; Morozova-Roche, Ludmilla A.ACS Chemical Neuroscience (2016), 7 (1), 34-39CODEN: ACNCDM; ISSN:1948-7193. (American Chemical Society)Pro-inflammatory protein S100A9 was established as a biomarker of dementia progression and compared with others such as Aβ1-42 and tau-proteins. CSF samples from 104 stringently diagnosed individuals divided into five subgroups were analyzed, including nondemented controls, stable mild cognitive impairment (SMCI), mild cognitive impairment due to Alzheimer's disease (MCI-AD), Alzheimer's disease (AD), and vascular dementia (VaD) patients. ELISA, dot-blotting, and electrochem. impedance spectroscopy were used as research methods. The S100A9 and Aβ1-42 levels correlated with each other: their CSF content decreased already at the SMCI stage and declined further under MCI-AD, AD, and VaD conditions. Immunohistochem. anal. also revealed involvement of both Aβ1-42 and S100A9 in the amyloid-neuroinflammatory cascade already during SMCI. Tau proteins were not yet altered in SMCI; however their contents increased during MCI-AD and AD, diagnosing later dementia stages. Thus, four biomarkers together, reflecting different underlying pathol. causes, can accurately differentiate dementia progression and also distinguish AD from VaD.
- 23Stefani, M. and Rigacci, S. (2014) Beneficial Properties of Natural Phenols: Highlight on Protection Against Pathological Conditions Associated with Amyloid Aggregation. BioFactors 40 (5), 482– 493, DOI: 10.1002/biof.1171Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvVWnsLfM&md5=6fe575d1bd7e30a74c54978a209a92f9Beneficial properties of natural phenols: Highlight on protection against pathological conditions associated with amyloid aggregationStefani, Massimo; Rigacci, StefaniaBioFactors (2014), 40 (5), 482-493CODEN: BIFAEU; ISSN:1872-8081. (Wiley-Blackwell)A review. Mediterranean and Asian diets are currently considered as the most healthy traditional feeding habits effective against risk of age-assocd., particularly cardiovascular and neurodegenerative, diseases. A common feature of these two regimens is the abundance of foods and beverages of plant origin (green tea, extra virgin olive oil, red wine, spices, berries, and arom. herbs) that are considered responsible for the obsd. beneficial effects. Epidemiol. data suggest that the phenolic component remarkably enriched in these foods plays an important role in reducing the incidence of amyloid diseases, pathol. conditions assocd. to tissue deposition of toxic protein aggregates responsible for progressive functional deterioration. Great effort is being spent to provide knowledge on the effects of several natural phenols in this context, moving from the test tube to animal models and, more slowly, to the patient's bed. An emerging feature that makes these mols. increasingly attractive for amyloid disease prevention and therapy is their wide spectrum of activity: recent pieces of evidence suggest that they can inhibit the prodn. of amyloidogenic peptides from precursors, increase antioxidant enzyme activity, activate autophagy and reduce inflammation. Our concept should than shift from considering natural phenols simply as antioxidants or, at the best, as amyloid aggregation inhibitors, to describing them as potentially multitargeting drugs. A main concern is the low bioavailability of such compds. and efforts aimed at improving it are underway, with encapsulation strategies being the most promising ones. © 2014 BioFactors, 40(5):482-493, 2014.
- 24Leri, M., Scuto, M., Ontario, M. L., Calabrese, V., Calabrese, E. J., Bucciantini, M., and Stefani, M. (2020) Healthy Effects of Plant Polyphenols: Molecular Mechanisms. Int. J. Mol. Sci. 21 (4), 1250, DOI: 10.3390/ijms21041250Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhslGmt7%252FN&md5=8bfe06c440f984df718a7db0c12e74e0Healthy effects of plant polyphenols molecular mechanismsLeri, Manuela; Scuto, Maria; Ontario, Maria Laura; Calabrese, Vittorio; Calabrese, Edward J.; Bucciantini, Monica; Stefani, MassimoInternational Journal of Molecular Sciences (2020), 21 (4), 1250CODEN: IJMCFK; ISSN:1422-0067. (MDPI AG)The increasing extension in life expectancy of human beings in developed countries is accompanied by a progressively greater rate of degenerative diseases assocd. with lifestyle and aging, most of which are still waiting for effective, not merely symptomatic, therapies. Accordingly, at present, the recommendations aimed at reducing the prevalence of these conditions in the population are limited to a safer lifestyle including phys./mental exercise, a reduced caloric intake, and a proper diet in a convivial environment. The claimed health benefits of the Mediterranean and Asian diets have been confirmed in many clin. trials and epidemiol. surveys. These diets are characterized by several features, including low meat consumption, the intake of oils instead of fats as lipid sources, moderate amts. of red wine, and significant amts. of fresh fruit and vegetables. In particular, the latter have attracted popular and scientific attention for their content, though in reduced amts., of a no. of mols. increasingly investigated for their healthy properties. Among the latter, plant polyphenols have raised remarkable interest in the scientific community; in fact, several clin. trials have confirmed that many health benefits of the Mediterranean/Asian diets can be traced back to the presence of significant amts. of these mols., even though, in some cases, contradictory results have been reported, which highlights the need for further investigation. In light of the results of these trials, recent research has sought to provide information on the biochem., mol., epigenetic, and cell biol. modifications by plant polyphenols in cell, organismal, animal, and human models of cancer, metabolic, and neurodegenerative pathologies, notably Alzheimer's and Parkinson disease. The findings reported in the last decade are starting to help to decipher the complex relations between plant polyphenols and cell homeostatic systems including metabolic and redox equil., proteostasis, and the inflammatory response, establishing an increasingly solid mol. basis for the healthy effects of these mols. Taken together, the data currently available, though still incomplete, are providing a rationale for the possible use of natural polyphenols, or their mol. scaffolds, as nutraceuticals to contrast aging and to combat many assocd. pathologies.
- 25Rigacci, S., Guidotti, V., Bucciantini, M., Parri, M., Nediani, C., Cerbai, E., Stefani, M., and Berti, A. (2010) Oleuropein Aglycon Prevents Cytotoxic Amyloid Aggregation of Human Amylin. J. Nutr. Biochem. 21 (8), 726– 735, DOI: 10.1016/j.jnutbio.2009.04.010Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXptVamt78%253D&md5=8571339f9624289049a490f0e25d965aOleuropein aglycon prevents cytotoxic amyloid aggregation of human amylinRigacci, Stefania; Guidotti, Valentina; Bucciantini, Monica; Parri, Matteo; Nediani, Chiara; Cerbai, Elisabetta; Stefani, Massimo; Berti, AndreaJournal of Nutritional Biochemistry (2010), 21 (8), 726-735CODEN: JNBIEL; ISSN:0955-2863. (Elsevier)Pancreatic amyloid deposits of amylin are a hallmark of Type II diabetes and considerable evidence indicates that amylin oligomers are cytotoxic to β-cells. Many efforts are presently spent to find out naturally occurring mols., or to design synthetic ones, able to hinder amylin aggregation or to protect cells against aggregate cytotoxicity. In this context, a protective effect of some polyphenols against amyloid cytotoxicity was reported. Actually dietary polyphenols are endowed with multiple health benefits, and extra virgin olive oil is attracting increasing interest as a source of these substances. Here, we investigated the effects on amylin aggregation and cytotoxicity of the secoiridoid oleuropein aglycon, the main phenolic component of extra virgin olive oil. We found that oleuropein, when present during the aggregation of amylin, consistently prevented its cytotoxicity to RIN-5F pancreatic β-cells, as detd. by the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide test and caspase-3 activity assay. A lack of interaction with the cell membrane of amylin aggregates grown in the presence of oleuropein was shown by fluorescence microscopy and synthetic lipid vesicle permeabilization. Moreover, our ThT assay, CD anal. and electron microscopy images suggested that oleuropein interferes with amylin aggregation, resulting in a different path skipping the formation of toxic pre-fibrillar aggregates. These results provide a mol. basis for some of the benefits potentially coming from extra virgin olive oil consumption and pave the way to further studies on the possible pharmacol. use of oleuropein to prevent or to slow down the progression of type II diabetes.
- 26Leri, M., Nosi, D., Natalello, A., Porcari, R., Ramazzotti, M., Chiti, F., Bellotti, V., Doglia, S. M., Stefani, M., and Bucciantini, M. (2016) The Polyphenol Oleuropein Aglycone Hinders the Growth of Toxic Transthyretin Amyloid Assemblies. J. Nutr. Biochem. 30, 153– 166, DOI: 10.1016/j.jnutbio.2015.12.009Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xis1Gis7g%253D&md5=df648a9675770f47caa514e982d62264The polyphenol Oleuropein aglycone hinders the growth of toxic transthyretin amyloid assembliesLeri, Manuela; Nosi, Daniele; Natalello, Antonino; Porcari, Riccardo; Ramazzotti, Matteo; Chiti, Fabrizio; Bellotti, Vittorio; Doglia, Silvia Maria; Stefani, Massimo; Bucciantini, MonicaJournal of Nutritional Biochemistry (2016), 30 (), 153-166CODEN: JNBIEL; ISSN:0955-2863. (Elsevier)Transthyretin (TTR) is involved in a subset of familial or sporadic amyloid diseases including senile systemic amyloidosis (SSA), familial amyloid polyneuropathy and cardiomyopathy (FAP/FAC) for which no effective therapy has been found yet. These conditions are characterized by extracellular deposits primarily found in the heart parenchyma and in peripheral nerves whose main component are amyloid fibrils, presently considered the main culprits of cell sufferance. The latter are polymeric assemblies grown from misfolded TTR, either wt or carrying one out of many identified mutations. The recent introduction in the clin. practice of synthetic TTR-stabilizing mols. that reduce protein aggregation provides the rationale to search natural effective mols. able to interfere with TTR amyloid aggregation by hindering the appearance of toxic species or by favoring the growth of harmless aggregates. Here we carried out an in depth biophys. and morphol. study on the mol. features of the aggregation of wt- and L55P-TTR involved in SSA or FAP/FAC, resp., and on the interference with fibril aggregation, stability and toxicity to cardiac HL-1 cells to demonstrate the ability of Oleuropein aglycon (OleA), the main phenolic component of the extra virgin olive oil. We describe the mol. basis of such interference and the resulting redn. of TTR amyloid aggregate cytotoxicity. Our data offer the possibility to validate and optimize the use of OleA or its mol. scaffold to rationally design promising drugs against TTR-related pathologies that could enter a clin. exptl. phase.
- 27Rigacci, S., Guidotti, V., Bucciantini, M., Nichino, D., Relini, A., Berti, A., and Stefani, M. (2011) Aβ(1–42) Aggregates into Non-Toxic Amyloid Assemblies in the Presence of the Natural Polyphenol Oleuropein Aglycon. Curr. Alzheimer Res. 8 (8), 841– 852, DOI: 10.2174/156720511798192682Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs1ShtrvN&md5=4280c13b299c367bc10a8e52145a8ffcAβ(1-42) aggregates into non-toxic amyloid assemblies in the presence of the natural polyphenol oleuropein aglyconRigacci, Stefania; Guidotti, Valentina; Bucciantini, Monica; Nichino, Daniela; Relini, Annalisa; Berti, Andrea; Stefani, MassimoCurrent Alzheimer Research (2011), 8 (8), 841-852CODEN: CARUBY; ISSN:1567-2050. (Bentham Science Publishers Ltd.)Amyloid aggregation starts with the initial misfolding of peptide/protein precursors, with subsequent structural rearrangement into oligomers and protofibrils; the latter eventually organize into fibrils with shared basic structural features, found deposited in amyloid diseases. Mounting evidence indicates early oligomers as the most toxic amyloid species; accordingly, the search of inhibitors of their growth is considered a promising target to prevent amyloid toxicity. We recently showed that oleuropein aglycon, a polyphenol abundant in the extra virgin olive oil, interferes with the aggregation of amylin (involved in type-2 diabetes), eliminating its cytotoxicity. Here we report that oleuropein aglycon also hinders amyloid aggregation of Aβ(1-42) and its cytotoxicity, suggesting a general effect of such polyphenol. In particular, by a wide panel of different spectroscopic, immunol., cell viability and imaging techniques we provide a more detailed description of Aβ(1-42) structural modifications arising in the presence of the inhibitor and the resulting cytotoxicity. We here report that the polyphenol eliminates the appearance of early toxic oligomers favoring the formation of stable harmless protofibrils, structurally different from the typical Aβ(1-42) fibrils. We also show that oleuropein aglycon is maximally effective when is present at the beginning of the aggregation process; furthermore, when added to pre-formed fibrils, it does not induce the release of toxic oligomers but, rather, neutralizes any residual toxicity possibly arising from the residual presence of traces of sol. oligomers and other toxic aggregates. The possible use of this polyphenol as anti-aggregation mol. is discussed in the light of these data.
- 28Palazzi, L., Bruzzone, E., Bisello, G., Leri, M., Stefani, M., Bucciantini, M., and Polverino de Laureto, P. (2018) Oleuropein Aglycone Stabilizes the Monomeric α-Synuclein and Favours the Growth of Non-Toxic Aggregates. Sci. Rep. 8 (1), 8337, DOI: 10.1038/s41598-018-26645-5Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MbhsFemtg%253D%253D&md5=e9bed4333ac2bf282a12dc0f4994ee47Oleuropein aglycone stabilizes the monomeric α-synuclein and favours the growth of non-toxic aggregatesPalazzi Luana; Bisello Giovanni; Polverino de Laureto Patrizia; Bruzzone Elena; Leri Manuela; Stefani Massimo; Bucciantini Monica; Leri ManuelaScientific reports (2018), 8 (1), 8337 ISSN:.α-synuclein plays a key role in the pathogenesis of Parkinson's disease (PD); its deposits are found as amyloid fibrils in Lewy bodies and Lewy neurites, the histopathological hallmarks of PD. Amyloid fibrillation is a progressive polymerization path starting from peptide/protein misfolding and proceeding through the transient growth of oligomeric intermediates widely considered as the most toxic species. Consequently, a promising approach of intervention against PD might be preventing α-synuclein build-up, misfolding and aggregation. A possible strategy involves the use of small molecules able to slow down the aggregation process or to alter oligomer conformation favouring the growth of non-pathogenic species. Here, we show that oleuropein aglycone (OleA), the main olive oil polyphenol, exhibits anti-amyloidogenic power in vitro by interacting with, and stabilizing, α-synuclein monomers thus hampering the growth of on-pathway oligomers and favouring the growth of stable and harmless aggregates with no tendency to evolve into other cytotoxic amyloids. We investigated the molecular basis of such interference by both biophysical techniques and limited proteolysis; aggregate morphology was monitored by electron microscopy. We also found that OleA reduces the cytotoxicity of α-synuclein aggregates by hindering their binding to cell membrane components and preventing the resulting oxidative damage to cells.
- 29Leri, M., Natalello, A., Bruzzone, E., Stefani, M., and Bucciantini, M. (2019) Oleuropein Aglycone and Hydroxytyrosol Interfere Differently with Toxic Aβ1–42 Aggregation. Food Chem. Toxicol. 129, 1– 12, DOI: 10.1016/j.fct.2019.04.015Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXnvF2ht7s%253D&md5=3863c374b1298e87ce4fb3add2a6419fOleuropein aglycone and hydroxytyrosol interfere differently with toxic Aβ1-42 aggregationLeri, Manuela; Natalello, Antonino; Bruzzone, Elena; Stefani, Massimo; Bucciantini, MonicaFood and Chemical Toxicology (2019), 129 (), 1-12CODEN: FCTOD7; ISSN:0278-6915. (Elsevier Ltd.)Oleuropein aglycon (OleA), the most abundant polyphenol in extra virgin olive oil (EVOO), and Hydroxythyrosol (HT), the OleA main metabolite, have attracted our interest due to their multitarget effects, including the interference with amyloid aggregation path. However, the mechanistic details of their anti-amyloid effect are not known yet. We report here a broad biophys. approach and cell biol. techniques that enabled us to characterize the different mol. mechanisms by which OleA and HT modulate the Aβ1-42 fibrillation, a main histopathol. feature of Alzheimer's disease (AD). In particular, OleA prevents the growth of toxic Aβ1-42 oligomers and blocks their successive growth into mature fibrils following its interaction with the peptide N-terminus, while HT speeds up harmless fibril formation. Our data demonstrate that, by stabilizing oligomers and fibrils, both polyphenols reduce their seeding activity and aggregate/membrane interaction on human neuroblastoma SH-SY5Y cells. These findings highlight the great potential of EVOO polyphenols and offer the possibility to validate and to optimize their use for possible AD prevention and therapy.
- 30Palazzi, L., Leri, M., Cesaro, S., Stefani, M., Bucciantini, M., and Polverino de Laureto, P. (2020) Insight into the Molecular Mechanism Underlying the Inhibition of α-Synuclein Aggregation by Hydroxytyrosol. Biochem. Pharmacol. 173, 113722, DOI: 10.1016/j.bcp.2019.113722Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXit12ktL7F&md5=a4445287929f44b1f225758d549bf706Insight into the molecular mechanism underlying the inhibition of α-synuclein aggregation by hydroxytyrosolPalazzi, Luana; Leri, Manuela; Cesaro, Samuele; Stefani, Massimo; Bucciantini, Monica; Polverino de Laureto, PatriziaBiochemical Pharmacology (Amsterdam, Netherlands) (2020), 173 (), 113722CODEN: BCPCA6; ISSN:0006-2952. (Elsevier B.V.)Parkinson's disease (PD) is the second most prevalent neurodegenerative disease in the elderly people. To date, drugs able to reverse the disease are not available; the gold std. is levodopa that only relieves clin. symptoms, yet with severe side effects after prolonged administration. Many efforts are underway to find alternative targets for PD prevention or treatment, the most promising being α-synuclein (Syn). Recently, we reported that oleuropein aglycon (OleA) interferes with amyloid aggregation of Syn both stabilizing its monomeric state and inducing the formation of harmless, off-pathway oligomers. This study is focused at describing the interaction between Syn and hydroxytyrosol (HT), the phenolic moiety and main metabolite of OleA, and the interferences with Syn aggregation by using biophys. and biol. techniques. Our results show that HT dose-dependently inhibits Syn aggregation and that covalent and non-covalent binding mediate HT-Syn interaction. HT does not modify the natively unfolded structure of Syn, rather, it stabilizes specific regions of the mol. leading to inhibition of protein fibrillation. Cellular assays showed that HT reduces the toxicity of Syn aggregates. Moreover, Syn aggregates interaction with the cell membrane, an important factor for prion-like properties of Syn on-pathway oligomers, was reduced in cells exposed to Syn aggregates grown in the presence of HT.
- 31Daccache, A., Lion, C., Sibille, N., Gerard, M., Slomianny, C., Lippens, G., and Cotelle, P. (2011) Oleuropein and Derivatives From Olives as Tau Aggregation Inhibitors. Neurochem. Int. 58 (6), 700– 707, DOI: 10.1016/j.neuint.2011.02.010Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXksFGmtLw%253D&md5=94c9cf92c95514c8dc8b024f96d53ce2Oleuropein and derivatives from olives as Tau aggregation inhibitorsDaccache, Anthony; Lion, Cedric; Sibille, Nathalie; Gerard, Melanie; Slomianny, Christian; Lippens, Guy; Cotelle, PhilippeNeurochemistry International (2011), 58 (6), 700-707CODEN: NEUIDS; ISSN:0197-0186. (Elsevier Ltd.)Tau isoforms constitute a family of microtubule-assocd. proteins that are mainly expressed in neurons of the central nervous system. They promote the assembly of tubulin monomers into microtubules and modulate their stability, thus playing a key structural role in the distal portion of axons. In Alzheimer's disease and related tauopathies, Tau aggregation into fibrillary tangles contributes to intraneuronal and glial lesions. We report herein the ability of three natural phenolic derivs. obtained from olives and derived food products to prevent such Tau fibrillization in vitro, namely hydroxytyrosol, oleuropein, and oleuropein aglycon. The latter was found to be more active than the ref. Tau aggregation inhibitor methylene blue on both wild-type and P301L Tau proteins, inhibiting fibrillization at low micromolar concns. These findings might provide further exptl. support for the beneficial nutritional properties of olives and olive oil as well as a chem. scaffold for the development of new drugs aiming at neurodegenerative tauopathies.
- 32Leri, M., Oropesa-Nuñez, R., Canale, C., Raimondi, S., Giorgetti, S., Bruzzone, E., Bellotti, V., Stefani, M., and Bucciantini, M. (2018) Oleuropein Aglycone: A Polyphenol with Different Targets Against Amyloid Toxicity. Biochim. Biophys. Acta, Gen. Subj. 1862 (6), 1432– 1442, DOI: 10.1016/j.bbagen.2018.03.023Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXmsVKitL4%253D&md5=e3762313d1252ea3bf5f2cd7a8361f18Oleuropein aglycone: A polyphenol with different targets against amyloid toxicityLeri, Manuela; Oropesa-Nunez, Reiner; Canale, Claudio; Raimondi, Sara; Giorgetti, Sofia; Bruzzone, Elena; Bellotti, Vittorio; Stefani, Massimo; Bucciantini, MonicaBiochimica et Biophysica Acta, General Subjects (2018), 1862 (6), 1432-1442CODEN: BBGSB3; ISSN:0304-4165. (Elsevier B.V.)Many data highlight the benefits of the Mediterranean diet and its main lipid component, extra-virgin olive oil (EVOO). EVOO contains many phenolic compds. that have been found effective against several aging- and lifestyle-related diseases, including neurodegeneration. Oleuropein, a phenolic secoiroid glycoside, is the main polyphenol in the olive oil. It has been reported that the aglycon form of Oleuropein (OleA) interferes in vitro and in vivo with amyloid aggregation of a no. of proteins/peptides involved in amyloid, particularly neurodegenerative, diseases avoiding the growth of toxic oligomers and displaying protection against cognitive deterioration. In this study, we carried out a cellular and biophys. study on the relationships between the effects of OleA on the aggregation and cell interactions of the D76N β2-microglobulin (D76N b2m) variant assocd. with a familial form of systemic amyloidosis with progressive bowel dysfunction and extensive visceral amyloid deposits. Our results indicate that OleA protection against D76N b2m cytotoxicity results from (i) a modification of the conformational and biophys. properties of its amyloid fibrils; (ii) a modification of the cell bilayer surface properties of exposed cells. This study reveals that OleA remodels not only D76N b2m aggregates but also the cell membrane interfering with the misfolded proteins-cell membrane assocn., in most cases an early event triggering amyloid-mediated cytotoxicity. The data provided in the present article focus on OleA protection, featuring this polyphenol as a promising plant mol. useful against amyloid diseases.
- 33Rigacci, S., Miceli, C., Nediani, C., Berti, A., Cascella, R., Pantano, D., Nardiello, P., Luccarini, I., Casamenti, F., and Stefani, M. (2015) Oleuropein Aglycone Induces Autophagy via the AMPK/mTOR Signalling Pathway: A Mechanistic Insight. Oncotarget 6 (34), 35344– 35357, DOI: 10.18632/oncotarget.6119Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC28zitlGmuw%253D%253D&md5=4ed55c7e2bd9c10917d22d132d6a7752Oleuropein aglycone induces autophagy via the AMPK/mTOR signalling pathway: a mechanistic insightRigacci Stefania; Miceli Caterina; Nediani Chiara; Berti Andrea; Cascella Roberta; Stefani Massimo; Pantano Daniela; Nardiello Pamela; Luccarini Ilaria; Casamenti FiorellaOncotarget (2015), 6 (34), 35344-57 ISSN:.The healthy effects of plant polyphenols, some of which characterize the so-called Mediterranean diet, have been shown to arise from epigenetic and biological modifications resulting, among others, in autophagy stimulation. Our previous work highlighted the beneficial effects of oleuropein aglycone (OLE), the main polyphenol found in the extra virgin olive oil, against neurodegeneration both in cultured cells and in model organisms, focusing, in particular, autophagy activation. In this study we investigated more in depth the molecular and cellular mechanisms of autophagy induction by OLE using cultured neuroblastoma cells and an OLE-fed mouse model of amylod beta (Aβ) deposition. We found that OLE triggers autophagy in cultured cells through the Ca2+-CAMKKβ-AMPK axis. In particular, in these cells OLE induces a rapid release of Ca2+ from the SR stores which, in turn, activates CAMKKβ, with subsequent phosphorylation and activation of AMPK. The link between AMPK activation and mTOR inhibition was shown in the OLE-fed animal model in which we found that decreased phospho-mTOR immunoreactivity and phosphorylated mTOR substrate p70 S6K levels match enhanced phospho-AMPK levels, supporting the idea that autophagy activation by OLE proceeds through mTOR inhibition. Our results agree with those reported for other plant polyphenols, suggesting a shared molecular mechanism underlying the healthy effects of these substances against ageing, neurodegeneration, cancer, diabetes and other diseases implying autophagy dysfunction.
- 34Grossi, C., Rigacci, S., Ambrosini, S., Ed Dami, T., Luccarini, I., Traini, C., Failli, P., Berti, A., Casamenti, F., and Stefani, M. (2013) The Polyphenol Oleuropein Aglycone Protects TgCRND8Mice against Aß Plaque Pathology. PLoS One 8 (8), e71702 DOI: 10.1371/journal.pone.0071702Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtlamsrfI&md5=0cabef823648e2452389ed35a0320d17The polyphenol oleuropein aglycone protects TgCRND8 mice against Aβ plaque pathologyGrossi, Cristina; Rigacci, Stefania; Ambrosini, Stefano; Ed Dami, Teresa; Luccarini, Ilaria; Traini, Chiara; Failli, Paola; Berti, Andrea; Casamenti, Fiorella; Stefani, MassimoPLoS One (2013), 8 (8), e71702CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)The claimed beneficial effects of the Mediterranean diet include prevention of several age-related dysfunctions including neurodegenerative diseases and Alzheimer-like pathol. These effects have been related to the protection against cognitive decline assocd. with aging and disease by a no. of polyphenols found in red wine and extra virgin olive oil. The double transgenic TgCRND8 mice (overexpressing the Swedish and Indiana mutations in the human amyloid precursor protein), aged 1.5 and 4, and age-matched wild type control mice were used to examine in vivo the effects of 8 wk dietary supplementation of oleuropein aglycon (50 mg/kg of diet), the main polyphenol found in extra virgin olive oil. We report here that dietary supplementation of oleuropein aglycon strongly improves the cognitive performance of young/middle-aged TgCRND8 mice, a model of amyloid-β deposition, respect to age-matched littermates with un-supplemented diet. Immunofluorescence anal. of cerebral tissue in oleuropein aglycon-fed transgenic mice showed remarkably reduced β-amyloid levels and plaque deposits, which appeared less compact and "fluffy"; moreover, microglia migration to the plaques for phagocytosis and a remarkable redn. of the astrocyte reaction were evident. Finally, oleuropein aglycon-fed mice brain displayed an astonishingly intense autophagic reaction, as shown by the increase of autophagic markers expression and of lysosomal activity. Data obtained with cultured cells confirmed the latter evidence, suggesting mTOR regulation by oleuropein aglycon. Our results support, and provide mechanistic insights into, the beneficial effects against Alzheimer-assocd. neurodegeneration of a polyphenol enriched in the extra virgin olive oil, a major component of the Mediterranean diet.
- 35Luccarini, I., Pantano, D., Nardiello, P., Cavone, L., Lapucci, A., Miceli, C., Nediani, C., Berti, A., Stefani, M., and Casamenti, F. (2016) The Polyphenol Oleuropein Aglycone Modulates the PARP1-SIRT1 Interplay: An In Vitro and In Vivo Study. J. Alzheimer's Dis. 54 (2), 737– 750, DOI: 10.3233/JAD-160471Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsVyisrrN&md5=41ee6b88a89ab8643d3fa9cf6f5a723fThe Polyphenol Oleuropein Aglycone Modulates the PARP1-SIRT1 Interplay: An In Vitro and In Vivo StudyLuccarini, Ilaria; Pantano, Daniela; Nardiello, Pamela; Cavone, Leonardo; Lapucci, Andrea; Miceli, Caterina; Nediani, Chiara; Berti, Andrea; Stefani, Massimo; Casamenti, Fiorella; Cassano, TommasoJournal of Alzheimer's Disease (2016), 54 (2), 737-750CODEN: JADIF9; ISSN:1387-2877. (IOS Press)Poly(ADP-ribose) polymerase-1 (PARP1) activation contributes to the cascade of events initiated by amyloid-β (Aβ) peptide eventually leading to cell death in Alzheimer's disease brain. A significant accumulation of PAR polymers and increase of PARP1 expression were detected in the cortex at the early (3.5 mo) and intermediate (6 mo) stage of Aβ deposition in the TgCRND8 mouse model. Our previous data highlighted the beneficial effects of oleuropein aglycon (OLE), the main polyphenol found in the olive oil, against neurodegeneration both in cultured cells and in model organisms. Here we found that 8-wk OLE treatment (50mg/kg of diet) to 6-mo-old TgCRND8 mice rescued to control values PARP1 activation and the levels of its product, PAR. In N2a neuroblastoma cells, PARP1 activation and PAR formation upon exposure to N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) were abolished by pretreatment for 24h with either OLE (100μM) or PARP inhibitors. A significant redn. of the NAD+ content, compared to controls, was found in N2a cells exposed to MNNG (100μM) for 90min; the latter was slightly attenuated by cell treatment for 24h with PJ-34 or with OLE. In vitro and in vivo, the OLE-induced redn. of PARP1 activation was paralleled by the overexpression of Sirtuin1 (SIRT1), and, in vivo, by a decrease of NF-κB and the pro-apoptotic marker p53. In N2a cells, we also found that OLE potentiates the MNNG-induced increase of Beclin1 levels. In conclusion, our data show that OLE treatment counteracts neuronal damage through modulation of the PARP1-SIRT1 interplay.
- 36LeVine, H. (1995) Thioflavine T Interaction with Amyloid β-sheet Structures. Amyloid 2 (1), 1– 6, DOI: 10.3109/13506129509031881Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXlsFyhs7g%253D&md5=4e7b589b31c163fd215a025a16664d49Thioflavine T interaction with amyloid β-sheet structuresLeVine, Harry IIIAmyloid (1995), 2 (1), 1-6CODEN: AIJIET; ISSN:1350-6129.Thioflavine T (ThT) interacts in tissue sections with amyloid deposits comprised of a variety of protein species giving a characteristic fluorescent complex. Binding of the dye to amyloid fibrils in suspension generates an amyloid-specific fluorescent signal. This interaction with amyloid fibrils formed from different polypeptides and proteins contg. antiparallel β-pleated sheet secondary structure is selective, occurring strongly with amyloid fibrils formed from insulin, transthyretin, polyglycine(I), Aβ(1-40), and weakly with β2-microglobulin. No fluorescence changes were seen with β-sheet fibrillar poly-L-lysine, islet amyloid peptide (20-29), or poly-L-serine. Native forms of transthyretin, insulin, β2-microglobulin, poly-L-lysine, Aβ(1-40), or several proteins contg. high percentages of β-sheet were also unreactive. The affinity of the amyloid fibrils for ThT varied: (apparent Kd's 0.033 - 10 μM; Amyloid A protein < insulin < ApoAII < polyglycine < transthyretin < Aβ(1-40)). The spectral changes induced by the different amyloid fibrils are qual. the same regardless of the pKd of the interaction with ThT or the identity of the amyloid fibrils, suggesting that the quaternary and secondary, but not the primary structure of the amyloid fibrils are important in forming the amyloid fibril-specific ThT species. Thus, Thioflavine T provides a useful tool for the investigation of amyloidogenesis in a variety of systems.
- 37Pansieri, J., Ostojić, L., Iashchishyn, I. A., Magzoub, M., Wallin, C., Wärmländer, S. K. T. S., Gräslund, A., Nguyen Ngoc, M., Smirnovas, V., Svedružić, Ž. (2019) Pro-Inflammatory S100A9 Protein Aggregation Promoted by NCAM1 Peptide Constructs. ACS Chem. Biol. 14 (7), 1410– 1417, DOI: 10.1021/acschembio.9b00394Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtV2lsLjM&md5=994b31d58e7e135544a71406e5148697Pro-inflammatory S100A9 protein aggregation promoted by NCAM1 peptide constructsPansieri, Jonathan; Ostojic, Lucija; Iashchishyn, Igor A.; Magzoub, Mazin; Wallin, Cecilia; Waermlaender, Sebastian K. T. S.; Graeslund, Astrid; Nguyen Ngoc, Mai; Smirnovas, Vytautas; Svedruzic, Zeljko; Morozova-Roche, Ludmilla A.ACS Chemical Biology (2019), 14 (7), 1410-1417CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Amyloid cascade and neuroinflammation are hallmarks of neurodegenerative diseases, and pro-inflammatory S100A9 protein is central to both of them. Here, we have shown that NCAM1 peptide constructs carrying polycationic sequences derived from Aβ peptide (KKLVFF) and PrP protein (KKRPKP) significantly promote the S100A9 amyloid self-assembly in a concn.-dependent manner by making transient interactions with individual S100A9 mols., perturbing its native structure and acting as catalysts. Since the individual mol. misfolding is a rate-limiting step in S100A9 amyloid aggregation, the effects of the NCAM1 construct on the native S100A9 are so crit. for its amyloid self-assembly. S100A9 rapid self-assembly into large aggregated clumps may prevent its amyloid tissue propagation, and by modulating S100A9 aggregation as a part of the amyloid cascade, the whole process may be effectively tuned.
- 38Ferrone, F. (1999) Analysis of Protein Aggregation Kinetics. In Amyloid, Prions, and Other Protein Aggregates, Methods in Enzymology, Vol. 309, pp 256– 274, Academic Press, New York. DOI: 10.1016/S0076-6879(99)09019-9 .Google ScholarThere is no corresponding record for this reference.
- 39Pansieri, J., Iashchishyn, I. A., Fakhouri, H., Ostojić, L., Malisauskas, M., Musteikyte, G., Smirnovas, V., Schneider, M. M., Scheidt, T., Xu, C. K. (2020) Templating S100A9 Amyloids on Aβ Fibrillar Surfaces Revealed by Charge Detection Mass Spectrometry, Microscopy, Kinetic and Microfluidic Analyses. Chem. Sci. 11 (27), 7031– 7039, DOI: 10.1039/C9SC05905AGoogle Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtFOqtrjM&md5=870909e223e6273b417714be73311647Templating S100A9 amyloids on Aβ fibrillar surfaces revealed by charge detection mass spectrometry, microscopy, kinetic and microfluidic analysesPansieri, Jonathan; Iashchishyn, Igor A.; Fakhouri, Hussein; Ostojic, Lucija; Malisauskas, Mantas; Musteikyte, Greta; Smirnovas, Vytautas; Schneider, Matthias M.; Scheidt, Tom; Xu, Catherine K.; Meisl, Georg; Knowles, Tuomas P. J.; Gazit, Ehud; Antoine, Rodolphe; Morozova-Roche, Ludmilla A.Chemical Science (2020), 11 (27), 7031-7039CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)The mechanism of amyloid co-aggregation and its nucleation process are not fully understood in spite of extensive studies. Deciphering the interactions between proinflammatory S100A9 protein and Aβ42 peptide in Alzheimer's disease is fundamental since inflammation plays a central role in the disease onset. Here we use innovative charge detection mass spectrometry (CDMS) together with biophys. techniques to provide mechanistic insight into the co-aggregation process and differentiate amyloid complexes at a single particle level. Combination of mass and charge distributions of amyloids together with reconstruction of the differences between them and detailed microscopy reveals that co-aggregation involves templating of S100A9 fibrils on the surface of Aβ42 amyloids. Kinetic anal. further corroborates that the surfaces available for the Aβ42 secondary nucleation are diminished due to the coating by S100A9 amyloids, while the binding of S100A9 to Aβ42 fibrils is validated by a microfluidic assay. We demonstrate that synergy between CDMS, microscopy, kinetic and microfluidic analyses opens new directions in interdisciplinary research.
- 40Lambert de Malezieu, M., Ferron, S., Sauvager, A., Courtel, P., Ramassamy, C., Tomasi, S., and Abasq, M.-L. (2019) UV-Vis Spectroelectrochemistry of Oleuropein, Tyrosol, and p-Coumaric Acid Individually and in an Equimolar Combination. Differences in LC-ESI-MS2 Profiles of Oxidation Products and Their Neuroprotective Properties. Biomolecules 9 (12), 802, DOI: 10.3390/biom9120802Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXisVOisbzF&md5=bce247f0495156061750c7fcec67e4e5UV-vis spectroelectrochemistry of oleuropein, tyrosol, and p-coumaric acid individually and in an equimolar combination. Differences in LC-ESI-MS2 profiles of oxidation products and their neuroprotective propertiesLambert de Malezieu, Morgane; Ferron, Solenn; Sauvager, Aurelie; Courtel, Patricia; Ramassamy, Charles; Tomasi, Sophie; Abasq, Marie-LaurenceBiomolecules (2019), 9 (12), 802CODEN: BIOMHC; ISSN:2218-273X. (MDPI AG)Major phenolic compds. from olive oil (ArOH-EVOO), oleuropein (Ole), tyrosol (Tyr), and p-coumaric acid (p-Cou), are known for their antioxidant and neuroprotective properties. We previously demonstrated that their combination could potentiate their antioxidant activity in vitro and in cellulo. To further our knowledge of their electron-transfer properties, Ole, Tyr, and p-Cou underwent a spectroelectrochem. study, performed either individually or in equimolar mixts. Two mixts. (Mix and Mix-seq) were prepd. in order to det. whether distinct mols. could arise from their simultaneous or sequential oxidn. The comparison of Liq. Chromatog.-Electrospray Ionization-Tandem Mass Spectrometry (LC-ESI-MS2) profiles highlighted the presence of specific oxidized products found in the mixes. We hypothesized that they derived from the dimerization between Tyr and Ole or p-Cou, which have reacted either in their native or oxidized forms. Moreover, Ole regenerates when the Mix undergoes oxidn. Our study also showed significant neuroprotection by oxidized Ole and oxidized Mix against H2O2 toxicity on SK-N-SH cells, after 24 h of treatment with very low concns. (1 and 5 nM). This suggests the putative relevant role of oxidized Ole products to protect or delay neuronal death.
- 41Chang, C. C., Khan, I., Tsai, K. L., Li, H., Yang, L. W., Chou, R. H., and Yu, C. (2016) Blocking the Interaction between S100A9 and RAGE V Domain using CHAPS Molecule: A novel route to Drug Development Against Cell Proliferation. Biochim. Biophys. Acta, Proteins Proteomics 1864 (11), 1558– 69, DOI: 10.1016/j.bbapap.2016.08.008Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtleitrrJ&md5=edf7f6363b5d98376e5294ef68a8fe16Blocking the interaction between S100A9 and RAGE V domain using CHAPS molecule: A novel route to drug development against cell proliferationChang, Chin-Chi; Khan, Imran; Tsai, Kun-Lin; Li, Hongchun; Yang, Lee-Wei; Chou, Ruey-Hwang; Yu, ChinBiochimica et Biophysica Acta, Proteins and Proteomics (2016), 1864 (11), 1558-1569CODEN: BBAPBW; ISSN:1570-9639. (Elsevier B.V.)Human S100A9 (Calgranulin B) is a Ca2+-binding protein, from the S100 family, that often presents as a homodimer in myeloid cells. It becomes an important mediator during inflammation once calcium binds to its EF-hand motifs. Human RAGE protein (receptor for advanced glycation end products) is one of the target-proteins. RAGE binds to a hydrophobic surface on S100A9. Interactions between these proteins trigger signal transduction cascades, promoting cell growth, proliferation, and tumorigenesis. Here, we present the soln. structure of mutant S100A9 (C3S) homodimer, detd. by multi-dimensional NMR expts. We further characterize the soln. interactions between mS100A9 and the RAGE V domain via NMR spectroscopy. CHAPS is a zwitterionic and non-denaturing mol. widely used for protein solubilizing and stabilization. We found out that CHAPS and RAGE V domain would interact with mS100A9 by using 1H-15N HSQC NMR titrns. Therefore, using the HADDOCK program, we superimpose two binary complex models mS100A9-RAGE V domain and mS100A9-CHAPS and demonstrate that CHAPS mols. could play a crucial role in blocking the interaction between mS100A9 and the RAGE V domain. WST-1 assay results also support the conclusion that CHAPS inhibits the bioactivity of mS100A9. This report will help to inform new drug development against cell proliferation.
- 42Ferrer, I., Blanco, R., Carmona, M., Puig, B., Ribera, R., Rey, M. J., and Ribalta, T. (2001) Prion Protein Expression in Senile Plaques in Alzheimer’s Disease. Acta Neuropathol. 101 (1), 49– 56, DOI: 10.1007/s004010000271Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXhtValurs%253D&md5=ad313fb97a3424f0f9f87f3af05b2e20Prion protein expression in senile plaques in Alzheimer's diseaseFerrer, I.; Blanco, R.; Carmona, M.; Puig, B.; Ribera, R.; Rey, M. J.; Ribalta, T.Acta Neuropathologica (2001), 101 (1), 49-56CODEN: ANPTAL; ISSN:0001-6322. (Springer-Verlag)Prion protein (PrPC) is a glycolipid-anchored cell membrane sialoglycoprotein that localizes in presynaptic membranes. Since synapses are vulnerable to Alzheimer's disease (AD), the present study examines PrPC expression in senile plaques, one of the major structural abnormalities in AD, by single- and double-labeling immunohistochem. Punctate PrPC immunoreactivity is found in diffuse plaques, whereas isolated large coarse PrPC-pos. granules reminiscent of dystrophic neurites are obsd. in neuritic plaques. Finally, PrPC deposition also occurs as dense filamentous and amorphous ppts. in amyloid cores of senile plaques, but not in the walls of blood vessels with amyloid angiopathy. In contrast to PrPC, βA4-amyloid immunoreactivity is preserved and even enhanced following incubation of the tissue sections with proteinase K prior to immunohistochem., thus indicating no PrPC and βA4-amyloid cross-reactivity in dense amyloid cores of senile plaques. Punctate PrPC deposition in diffuse plaques is similar to that of synaptophysin, a synaptic vesicle-assocd. protein, as already reported in other studies. Immunopptn., electrophoresis and Western blot studies have shown that synaptophysin, amyloid precursor protein (APP) and βA4 do not co-ppt. with PrP. These results suggest that synaptophysin, APP and βA4 are likely not bound to PrP. PrPC accumulation in βA4-amyloid dense cores may be the consequence of the release of PrP into the extracellular space. Whether PrPC accumulation in the extracellular space is the result of impaired endocytosis and subsequent hydrolysis in the endosomal compartment, in contrast to normal degrdn. of PrPC, resulting from or occurring in parallel to abnormal APP degrdn., deserves further study.
- 43Vogl, T., Eisenblätter, M., Völler, T., Zenker, S., Hermann, S., van Lent, P., Faust, A., Geyer, C., Petersen, B., Roebrock, K. (2014) Alarmin S100A8/S100A9 as a Biomarker for Molecular Imaging of Local Inflammatory Activity. Nat. Commun. 5 (1), 4593, DOI: 10.1038/ncomms5593Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitVaksLfN&md5=12a921b769ee55ecc080d53474332119Alarmin S100A8/S100A9 as a biomarker for molecular imaging of local inflammatory activityVogl, Thomas; Eisenblaetter, Michel; Voeller, Tom; Zenker, Stefanie; Hermann, Sven; van Lent, Peter; Faust, Andreas; Geyer, Christiane; Petersen, Beatrix; Roebrock, Kirsten; Schaefers, Michael; Bremer, Christoph; Roth, JohannesNature Communications (2014), 5 (), 4593CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)Inflammation has a key role in the pathogenesis of various human diseases. The early detection, localization and monitoring of inflammation are crucial for tailoring individual therapies. However, reliable biomarkers to detect local inflammatory activities and to predict disease outcome are still missing. Alarmins, which are locally released during cellular stress, are early amplifiers of inflammation. Here, using optical mol. imaging, we demonstrate that the alarmin S100A8/S100A9 serves as a sensitive local and systemic marker for the detection of even sub-clin. disease activity in inflammatory and immunol. processes like irritative and allergic contact dermatitis. In a model of collagen-induced arthritis, we use S100A8/S100A9 imaging to predict the development of disease activity. Furthermore, S100A8/S100A9 can act as a very early and sensitive biomarker in exptl. leishmaniasis for phagocyte activation linked to an effective Th1-response. In conclusion, the alarmin S100A8/S100A9 is a valuable and sensitive mol. target for novel imaging approaches to monitor clin. relevant inflammatory disorders on a mol. level.
- 44Bucciantini, M., Calloni, G., Chiti, F., Formigli, L., Nosi, D., Dobson, C. M., and Stefani, M. (2004) Prefibrillar Amyloid Protein Aggregates Share Common Features of Cytotoxicity. J. Biol. Chem. 279 (30), 31374– 31382, DOI: 10.1074/jbc.M400348200Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXlslOrs7g%253D&md5=f0ab3f11b00cc2b8b23acb99069545e6Prefibrillar Amyloid Protein Aggregates Share Common Features of CytotoxicityBucciantini, Monica; Calloni, Giulia; Chiti, Fabrizio; Formigli, Lucia; Nosi, Daniele; Dobson, Christopher M.; Stefani, MassimoJournal of Biological Chemistry (2004), 279 (30), 31374-31382CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)The intracellular free Ca2+ concn. and redox status of murine fibroblasts exposed to prefibrillar aggregates of the HypF N-terminal domain have been investigated in vitro and in vivo using a range of fluorescent probes. Aggregate entrance into the cytoplasm is followed by an early rise of reactive oxygen species and free Ca2+ levels and eventually by cell death. Such changes correlate directly with the viability of the cells and are not obsd. when cell are cultured in the presence of reducing agents or in Ca2+-free media. In addn., moderate cell stress following exposure to the aggregates was found to be fully reversible. The results show that the cytotoxicity of prefibrillar aggregates of HypF-N, a protein not assocd. with clin. disease, has the same fundamental origin as that produced by similar types of aggregates of proteins linked with specific amyloidoses. These findings suggest that misfolded proteinaceous aggregates stimulate generic cellular responses as a result of the exposure of regions of the structure (such as hydrophobic residues and the polypeptide main chain) that are buried in the normally folded proteins. They also support the idea that a higher no. of degenerative pathologies than previously known might be considered as protein deposition diseases.
- 45Novitskaya, V., Bocharova, O. V., Bronstein, I., and Baskakov, I. V. (2006) Amyloid Fibrils of Mammalian Prion Protein Are Highly Toxic to Cultured Cells and Primary Neurons. J. Biol. Chem. 281 (19), 13828– 13836, DOI: 10.1074/jbc.M511174200Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xkt1ymurY%253D&md5=7dd9e8722bc1997f4d260444248ddd58Amyloid Fibrils of Mammalian Prion Protein Are Highly Toxic to Cultured Cells and Primary NeuronsNovitskaya, Vera; Bocharova, Olga V.; Bronstein, Igor; Baskakov, Ilia V.Journal of Biological Chemistry (2006), 281 (19), 13828-13836CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)A growing body of evidence indicates that small, sol. oligomeric species generated from a variety of proteins and peptides rather than mature amyloid fibrils are inherently highly cytotoxic. Here, we show for the first time that mature amyloid fibrils produced from full-length recombinant mammalian prion protein (rPrP) were highly toxic to cultured cells and primary hippocampal and cerebellar neurons. Fibrils induced apoptotic cell death in a time- and dose-dependent manner. The toxic effect of fibrils was comparable with that exhibited by sol. small β-oligomers generated from the same protein. Fibrils prepd. from insulin were not toxic, suggesting that the toxic effect was not solely due to the highly polymeric nature of the fibrillar form. The cell death caused by rPrP fibrils or β-oligomers was substantially reduced when expression of endogenous PrPC was down-regulated by small interfering RNAs. In opposition to the β-oligomer and amyloid fibrils of rPrP, the monomeric α-helical form of rPrP stimulated neurite out-growth and survival of neurons. These studies illustrated that both sol. β-oligomer and amyloid fibrils of the prion protein are intrinsically toxic and confirmed that endogenously expressed PrPC is required for mediating the toxicity of abnormally folded external PrP aggregates.
- 46Canale, C., Oropesa-Nuñez, R., Diaspro, A., and Dante, S. (2018) Amyloid and Membrane Complexity: The Toxic Interplay Revealed by AFM. Semin. Cell Dev. Biol. 73, 82– 94, DOI: 10.1016/j.semcdb.2017.08.046Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsFSmsL3J&md5=912b964f5577766e290127a998df02e5Amyloid and membrane complexity: The toxic interplay revealed by AFMCanale, Claudio; Oropesa-Nunez, Reinier; Diaspro, Alberto; Dante, SilviaSeminars in Cell & Developmental Biology (2018), 73 (), 82-94CODEN: SCDBFX; ISSN:1084-9521. (Elsevier Ltd.)A review. Lipid membranes play a fundamental role in the pathol. development of protein misfolding diseases. Several pieces of evidence suggest that the lipid membrane could act as a catalytic surface for protein aggregation. Furthermore, a leading theory indicates the interaction between the cell membrane and misfolded oligomer species as the responsible for cytotoxicity, hence, for neurodegeneration in disorders such as Alzheimer's and Parkinson's disease. The definition of the mechanisms that drive the interaction between pathol. protein aggregates and plasma membrane is fundamental for the development of effective therapies for a large class of diseases. Atomic force microscopy (AFM) has been employed to study how amyloid aggregates affect the cell physiol. properties. Considerable efforts were spent to characterize the interaction with model systems, i.e., planar supported lipid bilayers, but some works also addressed the problem directly on living cells. Here, an overview of the main works involving the use of the AFM on both model system and living cells will be provided. Different kind of approaches will be presented, as well as the main results derived from the AFM anal.
- 47Walsh, P., Vanderlee, G., Yau, J., Campeau, J., Sim, V. L., Yip, C. M., and Sharpe, S. (2014) The Mechanism of Membrane Disruption by Cytotoxic Amyloid Oligomers Formed by Prion Protein (106–126) is Dependent on Bilayer Composition. J. Biol. Chem. 289 (15), 10419– 10430, DOI: 10.1074/jbc.M113.515866Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXmtlCnsbo%253D&md5=6e3cdf9ca126d6309eff637155f6ada0The Mechanism of Membrane Disruption by Cytotoxic Amyloid Oligomers Formed by Prion Protein(106-126) Is Dependent on Bilayer CompositionWalsh, Patrick; Vanderlee, Gillian; Yau, Jason; Campeau, Jody; Sim, Valerie L.; Yip, Christopher M.; Sharpe, SimonJournal of Biological Chemistry (2014), 289 (15), 10419-10430CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)The formation of fibrillar aggregates has long been assocd. with neurodegenerative disorders such as Alzheimer and Parkinson diseases. Although fibrils are still considered important to the pathol. of these disorders, it is now widely understood that smaller amyloid oligomers are the toxic entities along the misfolding pathway. One characteristic shared by the majority of amyloid oligomers is the ability to disrupt membranes, a commonality proposed to be responsible for their toxicity, although the mechanisms linking this to cell death are poorly understood. Here, we describe the phys. basis for the cytotoxicity of oligomers formed by the prion protein (PrP)-derived amyloid peptide PrP(106-126). We show that oligomers of this peptide kill several mammalian cells lines, as well as mouse cerebellar organotypic cultures, and we also show that they exhibit antimicrobial activity. Phys. perturbation of model membranes mimicking bacterial or mammalian cells was investigated using at. force microscopy, polarized total internal reflection fluorescence microscopy, and NMR spectroscopy. Disruption of anionic membranes proceeds through a carpet or detergent model as proposed for other antimicrobial peptides. By contrast, when added to zwitterionic membranes contg. cholesterol-rich ordered domains, PrP(106-126) oligomers induce a loss of domain sepn. and decreased membrane disorder. Loss of raft-like domains may lead to activation of apoptotic pathways, resulting in cell death. This work sheds new light on the phys. mechanisms of amyloid cytotoxicity and is the first to clearly show membrane type-specific modes of action for a cytotoxic peptide.
- 48Leri, M., Bemporad, F., Oropesa-Nuñez, R., Canale, C., Calamai, M., Nosi, D., Ramazzotti, M., Giorgetti, S., Pavone, F. S., Bellotti, V. (2016) Molecular Insights into Cell Toxicity of a Novel Familial Amyloidogenic Variant of β2-microglobulin. J. Cell. Mol. Med. 20 (8), 1443– 1456, DOI: 10.1111/jcmm.12833Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xht1SmtL%252FI&md5=569e255a9a3564a7f0a53904b708a902Molecular insights into cell toxicity of a novel familial amyloidogenic variant of β2-microglobulinLeri, Manuela; Bemporad, Francesco; Oropesa-Nunez, Reinier; Canale, Claudio; Calamai, Martino; Nosi, Daniele; Ramazzotti, Matteo; Giorgetti, Sofia; Pavone, Francesco S.; Bellotti, Vittorio; Stefani, Massimo; Bucciantini, MonicaJournal of Cellular and Molecular Medicine (2016), 20 (8), 1443-1456CODEN: JCMMC9; ISSN:1582-4934. (Wiley-Blackwell)The first genetic variant of β2-microglobulin (b2M) assocd. with a familial form of systemic amyloidosis has been recently described. The mutated protein, carrying a substitution of Asp at position 76 with an Asn (D76N b2M), exhibits a strongly enhanced amyloidogenic tendency to aggregate with respect to the wild-type protein. In this study, we characterized the D76N b2M aggregation path and performed an unprecedented anal. of the biochem. mechanisms underlying aggregate cytotoxicity. We showed that, contrarily to what expected from other amyloid studies, early aggregates of the mutant are not the most toxic species, despite their higher surface hydrophobicity. By modulating ganglioside GM1 content in cell membrane or synthetic lipid bilayers, we confirmed the pivotal role of this lipid as aggregate recruiter favoring their cytotoxicity. We finally obsd. that the aggregates bind to the cell membrane inducing an alteration of its elasticity (with possible functional unbalance and cytotoxicity) in GM1-enriched domains only, thus establishing a link between aggregate-membrane contact and cell damage.
- 49Cicerale, S., Conlan, X. A., Sinclair, A. J., and Keast, R. S. J. (2008) Chemistry and Health of Olive Oil Phenolics. Crit. Rev. Food Sci. Nutr. 49 (3), 218– 236, DOI: 10.1080/10408390701856223Google ScholarThere is no corresponding record for this reference.
- 50Bucciantini, M., Nosi, D., Forzan, M., Russo, E., Calamai, M., Pieri, L., Formigli, L., Quercioli, F., Soria, S., Pavone, F. (2012) Toxic Effects of Amyloid Fibrils on Cell Membranes: The Importance of Ganglioside GM1. FASEB J. 26 (2), 818– 831, DOI: 10.1096/fj.11-189381Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XitFOnt7o%253D&md5=3e235091b37469527df80656e9941281Toxic effects of amyloid fibrils on cell membranes: the importance of ganglioside GM1Bucciantini, Monica; Nosi, Daniele; Forzan, Mario; Russo, Edda; Calamai, Martino; Pieri, Laura; Formigli, Lucia; Quercioli, Franco; Soria, Silvia; Pavone, Francesco; Savistchenko, Jimmy; Melki, Ronald; Stefani, MassimoFASEB Journal (2012), 26 (2), 818-831, 10.1096/fj.11-189381CODEN: FAJOEC; ISSN:0892-6638. (Federation of American Societies for Experimental Biology)The interaction of amyloid aggregates with the cell plasma membrane is currently considered among the basic mechanisms of neuronal dysfunction in amyloid neurodegeneration. We used amyloid oligomers and fibrils grown from the yeast prion Sup35p, responsible for the specific prion trait [PSI+], to investigate how membrane lipids modulate fibril interaction with the membranes of cultured H-END cells and cytotoxicity. Sup35p shares no homol. with endogenous mammalian polypeptide chains. Thus, the generic toxicity of amyloids and the mol. events underlying cell degeneration can be investigated without interference with analogous polypeptides encoded by the cell genome. Sup35 fibrils bound to the cell membrane without increasing its permeability to Ca2+. Fibril binding resulted in structural reorganization and aggregation of membrane rafts, with GM1 clustering and alteration of its mobility. Sup35 fibril binding was affected by GM1 or its sialic acid moiety, but not by cholesterol membrane content, with complete inhibition after treatment with fumonisin B1 or neuraminidase. Finally, cell impairment resulted from caspase-8 activation after Fas receptor translocation on fibril binding to the plasma membrane. Our observations suggest that amyloid fibrils induce abnormal accumulation and overstabilization of raft domains in the cell membrane and provide a reasonable, although not unique, mechanistic and mol. explanation for fibril toxicity.
- 51Calamai, M. and Pavone, F. S. (2013) Partitioning and Confinement of GM1 Ganglioside induced by Amyloid Aggregates. FEBS Lett. 587 (9), 1385– 1391, DOI: 10.1016/j.febslet.2013.03.014Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXltFCktL0%253D&md5=8a6120f6badbd0c18f63fd5ec3a1a42ePartitioning and confinement of GM1 ganglioside induced by amyloid aggregatesCalamai, Martino; Pavone, Francesco S.FEBS Letters (2013), 587 (9), 1385-1391CODEN: FEBLAL; ISSN:0014-5793. (Elsevier B.V.)Growing evidence shows that GM1 ganglioside is involved in amyloid deposition and toxicity. By means of real-time single particle tracking, we show that amyloid oligomers and aggregates formed by Aβ1-42 and amylin, two peptides assocd., resp., with the development of Alzheimer's disease and type II diabetes, interact with GM1 and decrease dramatically its lateral diffusion on the plasma membrane of living neuroblastoma cells. The confinement of GM1, a constituent of membrane rafts involved in neuroprotection, at the level of both types of amyloid aggregates can interfere with cell signaling pathways and contribute to the loss of neuroprotection.
- 52Pellistri, F., Bucciantini, M., Relini, A., Nosi, D., Gliozzi, A., Robello, M., and Stefani, M. (2008) Nonspecific Interaction of Prefibrillar Amyloid Aggregates with Glutamatergic Receptors Results in Ca2+ Increase in Primary Neuronal Cells. J. Biol. Chem. 283 (44), 29950– 29960, DOI: 10.1074/jbc.M803992200Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXht1ynt7bM&md5=3450bdbfbcc79be0a3bd6efcfa9cb12cNonspecific Interaction of Prefibrillar Amyloid Aggregates with Glutamatergic Receptors Results in Ca2+ Increase in Primary Neuronal CellsPellistri, Francesca; Bucciantini, Monica; Relini, Annalisa; Nosi, Daniele; Gliozzi, Alessandra; Robello, Mauro; Stefani, MassimoJournal of Biological Chemistry (2008), 283 (44), 29950-29960CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)It is widely reported that the Ca2+ increase following nonspecific cell membrane permeabilization is among the earliest biochem. modifications in cells exposed to toxic amyloid aggregates. However, more recently receptors with Ca2+ channel activity such as α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA), N-Me D-aspartate (NMDA), ryanodine, and inositol 1,4,5-trisphosphate receptors have been proposed as mediators of the Ca2+ increase in neuronal cells challenged with β-amyloid peptides. We previously showed that prefibrillar aggregates of proteins not assocd. with amyloid diseases are toxic to exposed cells similarly to comparable aggregates of disease-assocd. proteins. In particular, prefibrillar aggregates of the prokaryotic HypF-N were shown to be toxic to different cultured cell lines by eliciting Ca2+ and reactive oxygen species increases. This study was aimed at assessing whether NMDA and AMPA receptor activations could be considered a generic feature of cell interaction with amyloid aggregates rather than a specific effect of some aggregated protein. Therefore, we investigated whether NMDA and AMPA receptors were involved in the Ca2+ increase following exposure of rat cerebellar granule cells to HypF-N prefibrillar aggregates. We found that the intracellular Ca2+ increase was assocd. with the early activation of NMDA and AMPA receptors, although some nonspecific membrane permeabilization was also obsd. at longer times of exposure. This result matched a significant co-localization of the aggregates with both receptors on the plasma membrane. Our data support the possibility that glutamatergic channels are generic sites of interaction with the cell membrane of prefibrillar aggregates of different peptides and proteins as well as the key structures responsible for the resulting early membrane permeabilization to Ca2+.
- 53Salazar, S. V. and Strittmatter, S. M. (2017) Cellular Prion Protein as a Receptor for Amyloid-β Oligomers in Alzheimer’s Disease. Biochem. Biophys. Res. Commun. 483 (4), 1143– 1147, DOI: 10.1016/j.bbrc.2016.09.062Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFCrsbfO&md5=ca66bd363a8321200220fab42dd8a539Cellular prion protein as a receptor for amyloid-β oligomers in Alzheimer's diseaseSalazar, Santiago V.; Strittmatter, Stephen M.Biochemical and Biophysical Research Communications (2017), 483 (4), 1143-1147CODEN: BBRCA9; ISSN:0006-291X. (Elsevier B.V.)Sol. oligomers of amyloid-beta (Aβo) are implicated by biochem. and genetic evidence as a trigger for Alzheimer's disease (AD) pathophysiol. A key step is Aβo interaction with the neuronal surface to initiate a cascade of altered signal transduction leading to synaptic dysfunction and damage. This review discusses neuronal cell surface mols. with high affinity selectively for oligomeric disease-assocd. states of Aβ, with a particular focus on the role of cellular prion protein (PrPC) in this process. Addnl. receptors may contribute to mediation of Aβo action, but PrPC appears to play a primary role in a no. of systems. The specificity of binding, the genetic necessity in mouse models of disease and downstream signaling pathways are considered. Signal transduction downstream of Aβo complexes with PrPC involves metabotropic glutamate receptor 5 (mGluR5), Fyn kinase and Pyk2 kinase, with deleterious effects on synaptic transmission and maintenance. Current data support the hypothesis that a substantial portion of Aβo toxicity in AD is mediated after initial interaction with PrPC on the neuronal surface. As such, the interaction of Aβo with PrPC is a potential therapeutic intervention site for AD.
- 54Haas, L. T. and Strittmatter, S. M. (2016) Oligomers of Amyloid β Prevent Physiological Activation of the Cellular Prion Protein-Metabotropic Glutamate Receptor 5 Complex by Glutamate in Alzheimer Disease. J. Biol. Chem. 291 (33), 17112– 17121, DOI: 10.1074/jbc.M116.720664Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslaktL3P&md5=b338b288603f6edc0d4b6ed1f615df82Oligomers of amyloid β prevent physiological activation of the cellular prion protein-metabotropic glutamate receptor 5 complex by glutamate in Alzheimer diseaseHaas, Laura T.; Strittmatter, Stephen M.Journal of Biological Chemistry (2016), 291 (33), 17112-17121CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)The dysfunction and loss of synapses in Alzheimer disease are central to dementia symptoms. We have recently demonstrated that pathol. Amyloid β oligomer (Aβo) regulates the assocn. between intracellular protein mediators and the synaptic receptor complex composed of cellular prion protein (PrPC) and metabotropic glutamate receptor 5 (mGluR5). Here we sought to det. whether Aβo alters the physiol. signaling of the PrPC-mGluR5 complex upon glutamate activation. We provide evidence that acute exposure to Aβo as well as chronic expression of familial Alzheimer disease mutant transgenes in model mice prevents protein-protein interaction changes of the complex induced by the glutamate analog 3,5-dihydroxyphenylglycine. We further show that 3,5-dihydroxyphenylglycine triggers the phosphorylation and activation of protein tyrosine kinase 2-β (PTK2B, also referred to as Pyk2) and of calcium/calmodulin-dependent protein kinase II in wild-type brain slices but not in Alzheimer disease transgenic brain slices or wild-type slices incubated with Aβo. This study further distinguishes two sep. Aβo-dependent signaling cascades, one dependent on extracellular Ca2+ and Fyn kinase activation and the other dependent on the release of Ca2+ from intracellular stores. Thus, Aβo triggers multiple distinct PrPC-mGluR5-dependent events implicated in neurodegeneration and dementia. We propose that targeting the PrPC-mGluR5 complex will reverse aberrant Aβo-triggered states of the complex to allow physiol. fluctuations of glutamate signaling.
- 55Abedini, A., Cao, P., Plesner, A., Zhang, J., He, M., Derk, J., Patil, S. A., Rosario, R., Lonier, J., Song, F. (2018) RAGE Binds Preamyloid IAPP Intermediates and Mediates Pancreatic β Cell Proteotoxicity. J. Clin. Invest. 128 (2), 682– 698, DOI: 10.1172/JCI85210Google Scholar55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MvhtVKntA%253D%253D&md5=e8bdc41817d48f49d22e4f4b3695b129RAGE binds preamyloid IAPP intermediates and mediates pancreatic β cell proteotoxicityAbedini Andisheh; Zhang Jinghua; He Meilun; Derk Julia; Patil Sachi A; Rosario Rosa; Lonier Jacqueline; Song Fei; Schmidt Ann Marie; Cao Ping; Raleigh Daniel P; Plesner Annette; Koh Hyunwook; Li HuilinThe Journal of clinical investigation (2018), 128 (2), 682-698 ISSN:.Islet amyloidosis is characterized by the aberrant accumulation of islet amyloid polypeptide (IAPP) in pancreatic islets, resulting in β cell toxicity, which exacerbates type 2 diabetes and islet transplant failure. It is not fully clear how IAPP induces cellular stress or how IAPP-induced toxicity can be prevented or treated. We recently defined the properties of toxic IAPP species. Here, we have identified a receptor-mediated mechanism of islet amyloidosis-induced proteotoxicity. In human diabetic pancreas and in cellular and mouse models of islet amyloidosis, increased expression of the receptor for advanced glycation endproducts (RAGE) correlated with human IAPP-induced (h-IAPP-induced) β cell and islet inflammation, toxicity, and apoptosis. RAGE selectively bound toxic intermediates, but not nontoxic forms of h-IAPP, including amyloid fibrils. The isolated extracellular ligand-binding domains of soluble RAGE (sRAGE) blocked both h-IAPP toxicity and amyloid formation. Inhibition of the interaction between h-IAPP and RAGE by sRAGE, RAGE-blocking antibodies, or genetic RAGE deletion protected pancreatic islets, β cells, and smooth muscle cells from h-IAPP-induced inflammation and metabolic dysfunction. sRAGE-treated h-IAPP Tg mice were protected from amyloid deposition, loss of β cell area, β cell inflammation, stress, apoptosis, and glucose intolerance. These findings establish RAGE as a mediator of IAPP-induced toxicity and suggest that targeting the IAPP/RAGE axis is a potential strategy to mitigate this source of β cell dysfunction in metabolic disease.
- 56Vogl, T., Leukert, N., Barczyk, K., Strupat, K., and Roth, J. (2006) Biophysical Characterization of S100A8 and S100A9 in the Absence and Presence of Bivalent Cations. Biochim. Biophys. Acta, Mol. Cell Res. 1763 (11), 1298– 1306, DOI: 10.1016/j.bbamcr.2006.08.028Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xht1CnsLbK&md5=f4bc53c5e2577488e442d1034458f067Biophysical characterization of S100A8 and S100A9 in the absence and presence of bivalent cationsVogl, Thomas; Leukert, Nadja; Barczyk, Katarzyna; Strupat, Kerstin; Roth, JohannesBiochimica et Biophysica Acta, Molecular Cell Research (2006), 1763 (11), 1298-1306CODEN: BBAMCO; ISSN:0167-4889. (Elsevier Ltd.)S100A8 and S100A9 are two proinflammatory mols. belonging to the S100 family of calcium-binding proteins. Common to all S100 proteins, S100A8 and S100A9 form non-covalently assocd. complexes which have been shown to exhibit different functional properties. Besides dimerization, recent research is focused on the importance of higher oligomeric structures of S100 proteins induced by bivalent cations. While S100A8/S100A9-heterodimers are formed in the absence of calcium, tetramerization is strictly calcium-dependent. Heterodimer formation is not a simple process and our biophys. analyses (FRET, ESI-MS) demonstrate that simply mixing both subunits is not sufficient to induce complex formation. Steps of denaturation/renaturation are necessary for the recombinant complex to show identical biophys. properties as S100A8/S100A9 obtained from granulocytes. In addn. to calcium, both proteins are able to bind zinc with high affinity. Here, we demonstrate for the first time by different biophys. methods (MALDI-MS, ESI-MS, fluorescence spectroscopy) that zinc-binding, like calcium, induces (S100A8/S100A9)2-tetramers. Using mass spectrometric investigations, we demonstrate that zinc triggers the formation of (S100A8/S100A9)2-tetramers by zinc-specific binding sites rather than by interactions with calcium-specific EF-hands. The zinc-induced tetramer is structurally very similar to the calcium-induced tetramer. Thus, like calcium, zinc acts as a regulatory factor in S100A8/S100A9-dependent signaling pathways.
- 57Wang, C., Iashchishyn, I. A., Kara, J., Foderà, V., Vetri, V., Sancataldo, G., Marklund, M., and Morozova-Roche, L. A. (2019) Proinflammatory and Amyloidogenic S100A9 Induced by Traumatic Brain Injury in Mouse Model. Neurosci. Lett. 699, 199– 205, DOI: 10.1016/j.neulet.2019.02.012Google Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXjtFOkurY%253D&md5=00ce57ce6592952a11a2f6a928d0cd73Proinflammatory and amyloidogenic S100A9 induced by traumatic brain injury in mouse modelWang, Chao; Iashchishyn, Igor A.; Kara, John; Fodera, Vito; Vetri, Valeria; Sancataldo, Giuseppe; Marklund, Niklas; Morozova-Roche, Ludmilla A.Neuroscience Letters (2019), 699 (), 199-205CODEN: NELED5; ISSN:0304-3940. (Elsevier Ireland Ltd.)Traumatic brain injury (TBI) represents a significant risk factor for development of neurodegenerative diseases such as Alzheimer's and Parkinson's. The S100A9-driven amyloid-neuroinflammatory cascade occurring during primary and secondary TBI events can serve as a mechanistic link between TBI and Alzheimer's as demonstrated recently in the human brain tissues. Here by using immunohistochem. in the controlled cortical impact TBI mouse model we have found pro-inflammatory S100A9 in the brain tissues of all mice on the first and third post-TBI days, while 70% of mice did not show any S100A9 presence on seventh post-TBI day similar to controls. This indicates that defensive mechanisms effectively cleared S100A9 in these mouse brain tissues during post-TBI recovery. By using sequential immunohistochem. we have shown that S100A9 was produced by both neuronal and microglial cells. However, Aβ peptide deposits characteristic for Alzheimer's disease were not detected in any post-TBI animals. On the first and third post-TBI days S100A9 was found to aggregate intracellularly into amyloid oligomers, similar to what was previously obsd. in human TBI tissues. Complementary, by using Rayleigh scatting, intrinsic fluorescence and at. force microscopy we demonstrated that in vitro S100A9 self-assembles into amyloid oligomers within minutes. Its amyloid aggregation is highly dependent on changes of environmental conditions such as variation of calcium levels, pH, temp. and redn./oxidn., which might be relevant to perturbation of cellular and tissues homeostasis under TBI. Present results demonstrate that S100A9 induction mechanisms in TBI are similar in mice and humans, emphasizing that S100A9 is an important marker of brain injury and therefore can be a potential therapeutic target.
- 58Kayed, R., Canto, I., Breydo, L., Rasool, S., Lukacsovich, T., Wu, J., Albay, R., Pensalfini, A., Yeung, S., Head, E. (2010) Conformation Dependent Monoclonal Antibodies Distinguish Different Replicating Strains or Conformers of Prefibrillar Aβ Oligomers. Mol. Neurodegener. 5 (1), 57, DOI: 10.1186/1750-1326-5-57Google Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhs1ams7fE&md5=54b7c4aca24f41e66a682f2a78efc8e0Conformation dependent monoclonal antibodies distinguish different replicating strains or conformers of prefibrillar Aβ oligomersKayed, Rakez; Canto, Isabel; Breydo, Leonid; Rasool, Suhail; Lukacsovich, Tamas; Wu, Jessica; Albay, Ricardo, III; Pensalfini, Anna; Yeung, Stephen; Head, Elizabeth; Marsh, J. Lawrence; Glabe, CharlesMolecular Neurodegeneration (2010), 5 (), 57CODEN: MNOEAZ; ISSN:1750-1326. (BioMed Central Ltd.)Background: Age-related neurodegenerative diseases share a no. of important pathol. features, such as accumulation of misfolded proteins as amyloid oligomers and fibrils. Recent evidence suggests that sol. amyloid oligomers and not the insol. amyloid fibrils may represent the primary pathol. species of protein aggregates. Results: We have produced several monoclonal antibodies that specifically recognize prefibrillar oligomers and do not recognize amyloid fibrils, monomer or natively folded proteins. Like the polyclonal antisera, the individual monoclonals recognize generic epitopes that do not depend on a specific linear amino acid sequence, but they display distinct preferences for different subsets of prefibrillar oligomers. Immunol. anal. of a no. of different prefibrillar Aβ oligomer prepns. show that structural polymorphisms exist in Aβ prefibrillar oligomers that can be distinguished on the basis of their reactivity with monoclonal antibodies. Western blot anal. demonstrates that the conformers defined by the monoclonal antibodies have distinct size distributions, indicating that oligomer structure varies with size. The different conformational types of Aβ prefibrillar oligomers can serve as they serve as templates for monomer addn., indicating that they seed the conversion of Aβ monomer into more prefibrillar oligomers of the same type. Conclusions: These results indicate that distinct structural variants or conformers of prefibrillar Aβ oligomers exist that are capable of seeding their own replication. These conformers may be analogous to different strains of prions.
- 59Eilers, P. H. C. (2003) A Perfect Smoother. Anal. Chem. 75 (14), 3631– 3636, DOI: 10.1021/ac034173tGoogle Scholar59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXktFGqsLw%253D&md5=eba4aed2ebbf3e48bd80049d8e0ce165A Perfect SmootherEilers, Paul H. C.Analytical Chemistry (2003), 75 (14), 3631-3636CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)The A. Savitzky and M. Golay (1964) filter has several disadvantages. A very attractive alternative is a smoother based on penalized least squares, extending ideas presented by E. T. Whittaker (1923). This smoother is extremely fast, gives continuous control over smoothness, interpolates automatically, and allows fast leave-one-out cross-validation. It can be programmed in a few lines of Matlab code. Theory, implementation, and applications are presented.
- 60Wasylewski, Z., Koloczek, H., and Wasniowska, A. (1988) Fluorescence-Quenching-Resolved Spectroscopy of Proteins. Eur. J. Biochem. 172 (3), 719– 724, DOI: 10.1111/j.1432-1033.1988.tb13948.xGoogle Scholar60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1cXhsVKmt70%253D&md5=bb0cf515871a8a98d9b958cd066dde9eFluorescence-quenching-resolved spectroscopy of proteinsWasylewski, Zygmunt; Koloczek, Henryk; Wasniowska, AlicjaEuropean Journal of Biochemistry (1988), 172 (3), 719-24CODEN: EJBCAI; ISSN:0014-2956.A new procedure is described for using fluorescence-quenching data of tryptophan residues in proteins to resolve their fluorescence emission spectra. In this concept, the Stern-Volmer quenching plot is detd. at each particular emission wavelength and iterative nonlinear least-squares fitting procedure allowed to resolve the steady-state emission spectra into components. The resolved components, attributed to each of tryptophan residue, can be characterized by different accessibility to the quencher. The ability to resolve fluorescence emission spectra can be improved by using different kinds of efficient quenchers, which can selectively quench the emission of exposed or both exposed and buried fluorophores. The method was used to decomp. emission fluorescence spectra in 2-tryptophan-contg. proteins: horse liver dehydrogenase, sperm whale apomyoglobin, and metalloprotease from Staphylococcus aureus. The resolved spectra of alc. dehydrogenase and metalloprotease are in excellent agreement with those previously obtained by single-photon counting or phase methods. The method presented here is tech. simple and does not require expensive instrumentation.
- 61Pettersen, E. F., Goddard, T. D., Huang, C. C., Couch, G. S., Greenblatt, D. M., Meng, E. C., and Ferrin, T. E. (2004) UCSF Chimera-AVisualization Sytem for Exploratory Research and Analysis Vesrion. J. Comput. Chem. 25, 1605– 1612, DOI: 10.1002/jcc.20084Google Scholar61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXmvVOhsbs%253D&md5=944b175f440c1ff323705987cf937ee7UCSF Chimera-A visualization system for exploratory research and analysisPettersen, Eric F.; Goddard, Thomas D.; Huang, Conrad C.; Couch, Gregory S.; Greenblatt, Daniel M.; Meng, Elaine C.; Ferrin, Thomas E.Journal of Computational Chemistry (2004), 25 (13), 1605-1612CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)The design, implementation, and capabilities of an extensible visualization system, UCSF Chimera, are discussed. Chimera is segmented into a core that provides basic services and visualization, and extensions that provide most higher level functionality. This architecture ensures that the extension mechanism satisfies the demands of outside developers who wish to incorporate new features. Two unusual extensions are presented: Multiscale, which adds the ability to visualize large-scale mol. assemblies such as viral coats, and Collab., which allows researchers to share a Chimera session interactively despite being at sep. locales. Other extensions include Multalign Viewer, for showing multiple sequence alignments and assocd. structures; ViewDock, for screening docked ligand orientations; Movie, for replaying mol. dynamics trajectories; and Vol. Viewer, for display and anal. of volumetric data. A discussion of the usage of Chimera in real-world situations is given, along with anticipated future directions. Chimera includes full user documentation, is free to academic and nonprofit users, and is available for Microsoft Windows, Linux, Apple Mac OS X, SGI IRIX, and HP Tru64 Unix from http://www.cgl.ucsf.edu/chimera/.
- 62Miletić, V., Odorčić, I., Nikolić, P., and Svedružić, Ž. M. (2017) In Silico Design of the First DNA-Independent Mechanism-Based Inhibitor of Mammalian DNA Methyltransferase Dnmt1. PLoS One 12, e0174410 DOI: 10.1371/journal.pone.0174410Google Scholar62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsValsrbL&md5=e52a25b05645fb00f338b89d0193afa6In silico design of the first DNA-independent mechanism-based inhibitor of mammalian DNA methyltransferase Dnmt1Miletic, Vedran; Odorcic, Ivica; Nikolic, Patrik; Svedruzic, Zeljko M.PLoS One (2017), 12 (4), e0174410/1-e0174410/21CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)We use our earlier exptl. studies of the catalytic mechanism of DNA methyltransferases to prep. in silico a family of novel mechanism-based inhibitors of human Dnmt1. Highly specific inhibitors of DNA methylation can be used for anal. of human epigenome and for the creation of iPS cells. We describe a set of adenosyl-1-methyl-pyrimidin-2-one derivs. as novel mechanism based inhibitors of mammalian DNA methyltransferase Dnmt1. The inhibitors have been designed to bind simultaneously in the active site and the cofactor site and thus act as transition- state analogs. Mol. dynamics studies showed that the lead compd. can form between 6 to 9 binding interactions with Dnmt1. QM/MM anal. showed that the upon binding to Dnmt1 the inhibitor can form a covalent adduct with active site Cys1226 and thus act as a mechanism-based suicide-inhibitor. The inhibitor can target DNA-bond and DNAfree form of Dnmt1, however the suicide-inhibition step is more likely to happen when DNA is bound to Dnmt1. The validity of presented anal. is described in detail using 69 modifications in the lead compd. structure. In total 18 of the presented 69 modifications can be used to prep. a family of highly specific inhibitors that can differentiate even between closely related enzymes such as Dnmt1 and Dnmt3a DNA methyltransferases. Presented results can be used for prepn. of some highly specific and potent inhibitors of mammalian DNA methylation with specific pharmacol. properties.
- 63Ruiz-Carmona, S., Alvarez-Garcia, D., Foloppe, N., Garmendia-Doval, A. B., Juhos, S., Schmidtke, P., Barril, X., Hubbard, R. E., and Morley, S. D. (2014) rDock: a Fast, Versatile and Open Source Program for Docking Ligands to Proteins and Nucleic Acids. PLoS Comput. Biol. 10, e1003571 DOI: 10.1371/journal.pcbi.1003571Google Scholar63https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhsVGlsL%252FO&md5=e4cb786d6567fdc7f2a46f64955a9992rDock: a fast, versatile and open source program for docking ligands to proteins and nucleic acidsRuiz-Carmona, Sergio; Alvarez-Garcia, Daniel; Foloppe, Nicolas; Garmendia-Doval, A. Beatriz; Juhos, Szilveszter; Schmidtke, Peter; Barril, Xavier; Hubbard, Roderick E.; Morley, S. DavidPLoS Computational Biology (2014), 10 (4), e1003571/1-e1003571/7, 7 pp.CODEN: PCBLBG; ISSN:1553-7358. (Public Library of Science)Identification of chem. compds. with specific biol. activities is an important step in both chem. biol. and drug discovery. When the structure of the intended target is available, one approach is to use mol. docking programs to assess the chem. complementarity of small mols. with the target; such calcns. provide a qual. measure of affinity that can be used in virtual screening (VS) to rank order a list of compds. according to their potential to be active. rDock is a mol. docking program developed at Vernalis for high-throughput VS (HTVS) applications. Evolved from RiboDock, the program can be used against proteins and nucleic acids, is designed to be computationally very efficient and allows the user to incorporate addnl. constraints and information as a bias to guide docking. This article provides an overview of the program structure and features and compares rDock to two ref. programs, AutoDock Vina (open source) and Schrodinger's Glide (com.). In terms of computational speed for VS, rDock is faster than Vina and comparable to Glide. For binding mode prediction, rDock and Vina are superior to Glide. The VS performance of rDock is significantly better than Vina, but inferior to Glide for most systems unless pharmacophore constraints are used; in that case rDock and Glide are of equal performance. The program is released under the Lesser General Public License and is freely available for download, together with the manuals, example files and the complete test sets, at online.
- 64Lee, J., Patel, D. S., Ståhle, J., Park, S. J., Kern, N. R., Kim, S., Lee, J., Cheng, X., Valvano, M. A., Holst, O. (2019) CHARMM-GUI Membrane Builder for Complex Biological Membrane Simulations with Glycolipids and Lipoglycans. J. Chem. Theory Comput. 15, 775– 786, DOI: 10.1021/acs.jctc.8b01066Google Scholar64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisVOitrbO&md5=a212a0bfce1818bc74f2b317eff3af93CHARMM-GUI Membrane Builder for Complex Biological Membrane Simulations with Glycolipids and LipoglycansLee, Jumin; Patel, Dhilon S.; Stahle, Jonas; Park, Sang-Jun; Kern, Nathan R.; Kim, Seonghoon; Lee, Joonseong; Cheng, Xi; Valvano, Miguel A.; Holst, Otto; Knirel, Yuriy A.; Qi, Yifei; Jo, Sunhwan; Klauda, Jeffery B.; Widmalm, Goran; Im, WonpilJournal of Chemical Theory and Computation (2019), 15 (1), 775-786CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Glycolipids (such as glycoglycerolipids, glycosphingolipids, and glycosylphosphatidylinositol) and lipoglycans (such as lipopolysaccharides (LPS), lipooligosaccharides (LOS), mycobacterial lipoarabinomannan, and mycoplasma lipoglycans) are typically found on the surface of cell membranes and play crucial roles in various cellular functions. Characterizing their structure and dynamics at the mol. level is essential to understand their biol. roles, but systematic generation of glycolipid and lipoglycan structures is challenging because of great variations in lipid structures and glycan sequences (i.e., carbohydrate types and their linkages). To facilitate the generation of all-atom glycolipid/LPS/LOS structures, we have developed Glycolipid Modeler and LPS Modeler in CHARMM-GUI (http://www.charmm-gui.org), a web-based interface that simplifies building of complex biol. simulation systems. In addn., we have incorporated these modules into Membrane Builder so that users can readily build a complex sym. or asym. biol. membrane system with various glycolipids and LPS/LOS. These tools are expected to be useful in innovative and novel glycolipid/LPS/LOS modeling and simulation research by easing tedious and intricate steps in modeling complex biol. systems and shall provide insight into structures, dynamics, and underlying mechanisms of complex glycolipid-/LPS-/LOS-contg. biol. membrane systems.
- 65Sousa da Silva, A. W and Vranken, W. F (2012) ACPYPE-Antechamber Python Parser Interface. BMC Res. Notes 5, 367, DOI: 10.1186/1756-0500-5-367Google Scholar65https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC38fitlWjtg%253D%253D&md5=60f750ba98c975374b351a39a1fa0ec4ACPYPE - AnteChamber PYthon Parser interfacESousa da Silva Alan W; Vranken Wim FBMC research notes (2012), 5 (), 367 ISSN:.BACKGROUND: ACPYPE (or AnteChamber PYthon Parser interfacE) is a wrapper script around the ANTECHAMBER software that simplifies the generation of small molecule topologies and parameters for a variety of molecular dynamics programmes like GROMACS, CHARMM and CNS. It is written in the Python programming language and was developed as a tool for interfacing with other Python based applications such as the CCPN software suite (for NMR data analysis) and ARIA (for structure calculations from NMR data). ACPYPE is open source code, under GNU GPL v3, and is available as a stand-alone application at http://www.ccpn.ac.uk/acpype and as a web portal application at http://webapps.ccpn.ac.uk/acpype. FINDINGS: We verified the topologies generated by ACPYPE in three ways: by comparing with default AMBER topologies for standard amino acids; by generating and verifying topologies for a large set of ligands from the PDB; and by recalculating the structures for 5 protein-ligand complexes from the PDB. CONCLUSIONS: ACPYPE is a tool that simplifies the automatic generation of topology and parameters in different formats for different molecular mechanics programmes, including calculation of partial charges, while being object oriented for integration with other applications.
- 66Kim, S., Lee, J., Jo, S., Brooks, C. L., 3rd, Lee, H. S., and Im, W. (2017) CHARMM-GUI Ligand Reader and Modeler for CHARMM Force Field Generation of Small Molecules. J. Comput. Chem. 38, 1879– 1886, DOI: 10.1002/jcc.24829Google Scholar66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXnslKhtLk%253D&md5=3e872e011783f98bc14594d91bc142efCHARMM-GUI ligand reader and modeler for CHARMM force field generation of small moleculesKim, Seonghoon; Lee, Jumin; Jo, Sunhwan; Brooks, Charles L., III; Lee, Hui Sun; Im, WonpilJournal of Computational Chemistry (2017), 38 (21), 1879-1886CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)Reading ligand structures into any simulation program is often nontrivial and time consuming, esp. when the force field parameters and/or structure files of the corresponding mols. are not available. To address this problem, we have developed Ligand Reader & Modeler in CHARMM-GUI. Users can upload ligand structure information in various forms (using PDB ID, ligand ID, SMILES, MOL/MOL2/SDF file, or PDB/mmCIF file), and the uploaded structure is displayed on a sketchpad for verification and further modification. Based on the displayed structure, Ligand Reader & Modeler generates the ligand force field parameters and necessary structure files by searching for the ligand in the CHARMM force field library or using the CHARMM general force field (CGenFF). In addn., users can define chem. substitution sites and draw substituents in each site on the sketchpad to generate a set of combinatorial structure files and corresponding force field parameters for throughput or alchem. free energy simulations. Finally, the output from Ligand Reader & Modeler can be used in other CHARMM-GUI modules to build a protein-ligand simulation system for all supported simulation programs, such as CHARMM, NAMD, GROMACS, AMBER, GENESIS, LAMMPS, Desmond, OpenMM, and CHARMM/OpenMM. Ligand Reader & Modeler is available as a functional module of CHARMM-GUI at http://www.charmm-gui.org/input/ligandrm.
- 67Van Der Spoel, D., Lindahl, E., Hess, B., Groenhof, G., Mark, A. E., and Berendsen, H. J. (2005) GROMACS: Fast, Flexible, and Free. J. Comput. Chem. 26, 1701– 1718, DOI: 10.1002/jcc.20291Google Scholar67https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXht1SlsbbO&md5=4aa71b1dc0b5b4978a88b0568245a265GROMACS: Fast, flexible, and freeVan Der Spoel, David; Lindahl, Erik; Hess, Berk; Groenhof, Gerrit; Mark, Alan E.; Berendsen, Herman J. C.Journal of Computational Chemistry (2005), 26 (16), 1701-1718CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)This article describes the software suite GROMACS (Groningen MAchine for Chem. Simulation) that was developed at the University of Groningen, The Netherlands, in the early 1990s. The software, written in ANSI C, originates from a parallel hardware project, and is well suited for parallelization on processor clusters. By careful optimization of neighbor searching and of inner loop performance, GROMACS is a very fast program for mol. dynamics simulation. It does not have a force field of its own, but is compatible with GROMOS, OPLS, AMBER, and ENCAD force fields. In addn., it can handle polarizable shell models and flexible constraints. The program is versatile, as force routines can be added by the user, tabulated functions can be specified, and analyses can be easily customized. Nonequil. dynamics and free energy detns. are incorporated. Interfaces with popular quantum-chem. packages (MOPAC, GAMES-UK, GAUSSIAN) are provided to perform mixed MM/QM simulations. The package includes about 100 utility and anal. programs. GROMACS is in the public domain and distributed (with source code and documentation) under the GNU General Public License. It is maintained by a group of developers from the Universities of Groningen, Uppsala, and Stockholm, and the Max Planck Institute for Polymer Research in Mainz. Its Web site is http://www.gromacs.org.
- 68Cheung, Y. T., Lau, W. K., Yu, M. S., Lai, C. S., Yeung, S. C., So, K. F., and Chang, R. C. (2009) Effects of All-Trans-Retinoic Acid on Human SH-SY5Y Neuroblastoma as In Vitro Model in Neurotoxicity Research. NeuroToxicology 30 (1), 127– 135, DOI: 10.1016/j.neuro.2008.11.001Google Scholar68https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtFeqsbw%253D&md5=c0fd9e12f367cc8258ce304212571828Effects of all-trans-retinoic acid on human SH-SY5Y neuroblastoma as in vitro model in neurotoxicity researchCheung, Yuen-Ting; Lau, Way Kwok-Wai; Yu, Man-Shan; Lai, Cora Sau-Wan; Yeung, Sze-Chun; So, Kwok-Fai; Chang, Raymond Chuen-ChungNeuroToxicology (2009), 30 (1), 127-135CODEN: NRTXDN; ISSN:0161-813X. (Elsevier B.V.)Human neuroblastoma SH-SY5Y is a dopaminergic neuronal cell line which has been used as an in vitro model for neurotoxicity expts. Although the neuroblastoma is usually differentiated by all-trans-retinoic acid (RA), both RA-differentiated and undifferentiated SH-SY5Y cells have been used in neuroscience research. However, the changes in neuronal properties triggered by RA as well as the subsequent responsiveness to neurotoxins have not been comprehensively studied. Therefore, we aim to re-evaluate the differentiation property of RA on this cell line. We hypothesize that modulation of signaling pathways and neuronal properties during RA-mediated differentiation in SH-SY5Y cells can affect their susceptibility to neurotoxins. The differentiation property of RA was confirmed by showing an extensive outgrowth of neurites, increased expressions of neuronal nuclei, neuron specific enolase, synaptophysin and synaptic assocd. protein-97, and decreased expression of inhibitor of differentiation-1. While undifferentiated SH-SY5Y cells were susceptible to 6-OHDA and MPP+, RA-differentiation conferred SH-SY5Y cells higher tolerance, potentially by up-regulating survival signaling, including Akt pathway as inhibition of Akt removed RA-induced neuroprotection against 6-OHDA. As a result, the real toxicity cannot be revealed in RA-differentiated cells. Therefore, undifferentiated SH-SY5Y is more appropriate for studying neurotoxicity or neuroprotection in exptl. Parkinson's disease research.
- 69Nosi, D., Mercatelli, R., Chellini, F., Soria, S., Pini, A., Formigli, L., and Quercioli, F. (2013) A Molecular Imaging Analysis of Cx43 Association with Cdo During Skeletal Myoblast Differentiation. J. Biophotonics 6 (8), 612– 621, DOI: 10.1002/jbio.201200063Google Scholar69https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXht1amu77P&md5=cd23bd3959355f60143523005ad14929A molecular imaging analysis of Cx43 association with Cdo during skeletal myoblast differentiationNosi, Daniele; Mercatelli, Raffaella; Chellini, Flaminia; Soria, Silvia; Pini, Alessandro; Formigli, Lucia; Quercioli, FrancoJournal of Biophotonics (2013), 6 (8), 612-621CODEN: JBOIBX; ISSN:1864-063X. (Wiley-VCH Verlag GmbH & Co. KGaA)Cell-to-cell contacts are crucial for cell differentiation. The promyogenic cell surface protein, Cdo, functions as a component of multiprotein clusters to mediate cell adhesion signaling. Connexin 43, the main connexin forming gap junctions, also plays a key role in myogenesis. At least part of its effects is independent of the intercellular channel function, but the mechanisms underlying are unknown. Here, using multiple optical approaches, we provided the first evidence that Cx43 phys. interacts with Cdo to form dynamic complexes during myoblast differentiation, offering clues for considering this interaction a structural basis of the channel-independent function of Cx43.
Cited By
This article is cited by 22 publications.
- Bakri Alaziqi, Liam Beckitt, David J. Townsend, Jasmine Morgan, Rebecca Price, Alana Maerivoet, Jillian Madine, David Rochester, Geoffrey Akien, David A. Middleton. Characterization of Olive Oil Phenolic Extracts and Their Effects on the Aggregation of the Alzheimer’s Amyloid-β Peptide and Tau. ACS Omega 2024, 9
(30)
, 32557-32578. https://doi.org/10.1021/acsomega.4c01281
- Efrat Naaman, Amanda Qarawani, Rony Ben-Zvi Elimelech, Michal Harel, Shahaf Sigal-Dror, Shadi Safuri, Vytautas Smirnovas, Ieva Baronaite, Nina V. Romanova, Ludmilla A. Morozova-Roche, Shiri Zayit-Soudry. The Surprising Nonlinear Effects of S100A9 Proteins in the Retina. ACS Chemical Neuroscience 2024, 15
(4)
, 735-744. https://doi.org/10.1021/acschemneuro.3c00650
- Natalia-Maria Christopoulou, Vasiliki Mamoulaki, Aglaia Mitsiakou, Eleni Samolada, Despina P. Kalogianni, Theodore K. Christopoulos. Screening Method for the Visual Discrimination of Olive Oil from Other Vegetable Oils by a Multispecies DNA Sensor. Analytical Chemistry 2024, 96
(4)
, 1803-1811. https://doi.org/10.1021/acs.analchem.3c05507
- Goro Nishide, Keesiang Lim, Maiki Tamura, Akiko Kobayashi, Qingci Zhao, Masaharu Hazawa, Toshio Ando, Noritaka Nishida, Richard W. Wong. Nanoscopic Elucidation of Spontaneous Self-Assembly of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Open Reading Frame 6 (ORF6) Protein. The Journal of Physical Chemistry Letters 2023, 14
(38)
, 8385-8396. https://doi.org/10.1021/acs.jpclett.3c01440
- Vinod Kumar Meena, Vijay Kumar, Shivani Karalia, Garima, Monica Sundd. Ellagic Acid Modulates Uninduced as well as Mutation and Metal-Induced Aggregation of α-Synuclein: Implications for Parkinson’s Disease. ACS Chemical Neuroscience 2021, 12
(19)
, 3598-3614. https://doi.org/10.1021/acschemneuro.1c00317
- Jiarui Zhao, Yuan Peng, Zhenfang Lin, Yulai Gong. Association between Mediterranean diet adherence and Parkinson's disease: a systematic review and meta-analysis. The Journal of nutrition, health and aging 2025, 29
(2)
, 100451. https://doi.org/10.1016/j.jnha.2024.100451
- Qi Tian, Zhijie Li, Ziang Yan, Shengming Jiang, Xincan Zhao, Lei Wang, Mingchang Li. Inflammatory role of S100A8/A9 in the central nervous system non-neoplastic diseases. Brain Research Bulletin 2024, 218 , 111100. https://doi.org/10.1016/j.brainresbull.2024.111100
- Junyi Chen, Wen-Zhu Yang, Huilan Chen, Xiuxia Ding, Hongxu Chen, Cai-Hong Zhan, Zhigang Jin. Targeting protein aggregation: the promising application of polyoxometalates in neurodegenerative diseases. Inorganic Chemistry Frontiers 2024, 11
(21)
, 7238-7255. https://doi.org/10.1039/D4QI01945H
- Manuela Leri, Dan Sun, Željko M. Svedružic, Darius Šulskis, Vytautas Smirnovas, Massimo Stefani, Ludmilla Morozova-Roche, Monica Bucciantini. Pro-inflammatory protein S100A9 targeted by a natural molecule to prevent neurodegeneration onset. International Journal of Biological Macromolecules 2024, 276 , 133838. https://doi.org/10.1016/j.ijbiomac.2024.133838
- Debojyoti Halder, Subham Das, Alex Joseph. An insight into structure-activity relationship of naturally derived biological macromolecules for the treatment of Alzheimer’s disease: a review. Journal of Biomolecular Structure and Dynamics 2024, 42
(12)
, 6455-6471. https://doi.org/10.1080/07391102.2023.2230279
- Natalia-Maria Christopoulou, Eleftheria Figgou, Panagiotis Kalaitzis, Despina P. Kalogianni, Theodore K. Christopoulos. Multiallelic DNA sensors for molecular traceability of olive oil varietal origin. Sensors and Actuators B: Chemical 2024, 406 , 135423. https://doi.org/10.1016/j.snb.2024.135423
- Di Zhang, Juanli Zhang, Zhongying Ma, Qianwen Wu, Meiyou Liu, Tingting Fan, Likun Ding, Danjun Ren, Aidong Wen, Jingwen Wang. Luteoloside inhibits Aβ1–42 fibrillogenesis, disintegrates preformed fibrils, and alleviates amyloid-induced cytotoxicity. Biophysical Chemistry 2024, 306 , 107171. https://doi.org/10.1016/j.bpc.2023.107171
- Simone Filardo, Mattioli Roberto, Daniel Di Risola, Luciana Mosca, Marisa Di Pietro, Rosa Sessa. Olea europaea L-derived secoiridoids: Beneficial health effects and potential therapeutic approaches. Pharmacology & Therapeutics 2024, 254 , 108595. https://doi.org/10.1016/j.pharmthera.2024.108595
- Ines Lopez del Castillo, Laura Vegas-Gomez, Joana Loureiro, Ines Moreno-Gonzalez. Natural molecules in neuroprotection and neurotoxicity in neurodegenerative diseases. 2024, 3-28. https://doi.org/10.1016/B978-0-443-23763-8.00001-4
- Rimgailė Tamulytė, Evelina Jankaitytė, Zigmantas Toleikis, Vytautas Smirnovas, Marija Jankunec. Pro-inflammatory protein S100A9 alters membrane organization by dispersing ordered domains. Biochimica et Biophysica Acta (BBA) - Biomembranes 2023, 1865
(3)
, 184113. https://doi.org/10.1016/j.bbamem.2022.184113
- Wei Wei, Shixu Wang, Chongchong Xu, Xuemei Zhou, Xinqing Lian, Lin He, Kuan Li. Gut microbiota, pathogenic proteins and neurodegenerative diseases. Frontiers in Microbiology 2022, 13 https://doi.org/10.3389/fmicb.2022.959856
- Vinod Kumar Meena, Vijay Kumar, Shivani Karalia, Rohit Singh Dangi, Monica Sundd. Structural and mechanistic insights into modulation of α-Synuclein fibril formation by aloin and emodin. Biochimica et Biophysica Acta (BBA) - General Subjects 2022, 1866
(7)
, 130151. https://doi.org/10.1016/j.bbagen.2022.130151
- Yijing Tang, Dong Zhang, Xiong Gong, Jie Zheng. Repurposing of intestinal defensins as multi-target, dual-function amyloid inhibitors
via
cross-seeding. Chemical Science 2022, 13
(24)
, 7143-7156. https://doi.org/10.1039/D2SC01447E
- Xing Ge, Tingting Yao, Chaoran Zhang, Qingqing Wang, Xuxu Wang, Li-Chun Xu. Human microRNA-4433 (hsa-miR-4443) Targets 18 Genes to be a Risk
Factor of Neurodegenerative Diseases. Current Alzheimer Research 2022, 19
(7)
, 511-522. https://doi.org/10.2174/1567205019666220805120303
- Basavaraj Vastrad, Chanabasayya Vastrad. Bioinformatics and next generation sequencing data analysis to identify key genes and pathways influencing in Parkinson’s disease. 2022https://doi.org/10.1101/2022.02.27.482208
- Masood Sadiq Butt, Urwa Tariq, Iahtisham‐Ul‐Haq, Ambreen Naz, Muhammad Rizwan. Neuroprotective effects of oleuropein: Recent developments and contemporary research. Journal of Food Biochemistry 2021, 45
(12)
https://doi.org/10.1111/jfbc.13967
- Alma Jahić Mujkić, Magda Tušek Žnidarič, Selma Berbić, Eva Žerovnik. Synergy of the Inhibitory Action of Polyphenols Plus Vitamin C on Amyloid Fibril Formation: Case Study of Human Stefin B. Antioxidants 2021, 10
(9)
, 1471. https://doi.org/10.3390/antiox10091471
- Lili Arabuli, Igor A. Iashchishyn, Nina V. Romanova, Greta Musteikyte, Vytautas Smirnovas, Himanshu Chaudhary, Željko M. Svedružić, Ludmilla A. Morozova-Roche. Co-Aggregation of S100A9 with DOPA and Cyclen-Based Compounds Manifested in Amyloid Fibril Thickening without Altering Rates of Self-Assembly. International Journal of Molecular Sciences 2021, 22
(16)
, 8556. https://doi.org/10.3390/ijms22168556
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 69 other publications.
- 1Akiyama, H., Barger, S., Barnum, S., Bradt, B., Bauer, J., Cole, G. M., Cooper, N. R., Eikelenboom, P., Emmerling, M., Fiebich, B. L. (2000) Inflammation and Alzheimer’s disease. Neurobiol. Aging 21 (3), 383– 421, DOI: 10.1016/S0197-4580(00)00124-X1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXktVSmt74%253D&md5=409a0ca2920ef2cba12474cb7d05f4aaInflammation and Alzheimer's diseaseAkiyama, H.; Barger, S.; Barnum, S.; Bradt, B.; Bauer, J.; Cole, G. M.; Cooper, N. R.; Eikelenboom, P.; Emmerling, M.; Fiebich, B. L.; Finch, C. E.; Frautschy, S.; Griffin, W. S. T.; Hampel, H.; Hull, M.; Landreth, G.; Lue, L.-F.; Mrak, R.; Mackenzie, I. R.; McGeer, P. L.; O'Banion, M. K.; Pachter, J.; Pasinetti, G.; Plata-Salaman, C.; Rogers, J.; Rydel, R.; Shen, Y.; Streit, W.; Strohmeyer, R.; Tooyoma, I.; Van Muiswinkel, F. L.; Veerhuis, R.; Walker, D.; Webster, S.; Wegrzyniak, B.; Wenk, G.; Wyss-Coray, T.Neurobiology of Aging (2000), 21 (3), 383-421CODEN: NEAGDO; ISSN:0197-4580. (Elsevier Science Inc.)A review, with 636 refs. Inflammation clearly occurs in pathol. vulnerable regions of the Alzheimer's disease (AD) brain, and it does so with the full complexity of local peripheral inflammatory responses. In the periphery, degenerating tissue and the deposition of highly insol. abnormal materials are classical stimulants of inflammation. Likewise, in the AD brain damaged neurons and neurites and highly insol. amyloid β peptide deposits and neurofibrillary tangles provide obvious stimuli for inflammation. Because these stimuli are discrete, microlocalized, and present from early preclin. to terminal stages of AD, local upregulation of complement, cytokines, acute-phase reactants, and other inflammatory mediators is also discrete, microlocalized, and chronic. Cumulated over many years, direct and bystander damage from AD inflammatory mechanisms is likely to exacerbate the very pathogenic processes that gave rise to it. Thus, animal models and clin. studies, although still in their infancy, strongly suggest that AD inflammation contributes to AD pathogenesis. By better understanding AD inflammatory and immunoregulatory processes, it should be possible to develop anti-inflammatory approaches that may not cure AD but will likely help slow the progression or delay the onset of this devastating disorder.
- 2Hardy, J. and Selkoe, D. J. (2002) The Amyloid Hypothesis of Alzheimer’ s Disease: Progress and Problems on the Road to Therapeutics. Science 297 (5580), 353, DOI: 10.1126/science.10729942https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38Xls1Cju7s%253D&md5=f7b8db1bc6f13e85f887b73f3042e86eThe amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeuticsHardy, John; Selkoe, Dennis J.Science (Washington, DC, United States) (2002), 297 (5580), 353-356CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)A review. It has been more than 10 yr since it was first proposed that the neurodegeneration in Alzheimer's disease (AD) may be caused by deposition of amyloid β-peptide (Aβ) in plaques in brain tissue. According to the amyloid hypothesis, accumulation of Aβ in the brain is the primary influence driving AD pathogenesis. The rest of the disease process, including formation of neurofibrillary tangles contg. tau protein, is proposed to result from an imbalance between Aβ prodn. and Aβ clearance.
- 3Zhu, S., Wang, J., Zhang, Y., He, J., Kong, J., Wang, J.-F., and Li, X.-M. (2017) The Role of Neuroinflammation and Amyloid in Cognitive Impairment in an APP/PS1 Transgenic Mouse Model of Alzheimer’s Disease. CNS Neurosci. Ther. 23 (4), 310– 320, DOI: 10.1111/cns.126773https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXksFaktr8%253D&md5=34feee5613ff847c8e2615f444238bdcThe role of neuroinflammation and amyloid in cognitive impairment in an APP/PS1 transgenic mouse model of Alzheimer's diseaseZhu, Shenghua; Wang, Junhui; Zhang, Yanbo; He, Jue; Kong, Jiming; Wang, Jun-Feng; Li, Xin-MinCNS Neuroscience & Therapeutics (2017), 23 (4), 310-320CODEN: CNTNAB; ISSN:1755-5930. (Wiley-Blackwell)Aims : Both amyloid deposition and neuroinflammation appear in the early course of Alzheimer's disease (AD). However, the progression of neuroinflammation and its relationship with amyloid deposition and behavioral changes have not been fully elucidated. A better understanding the role of neuroinflammation in AD might extend our current knowledge to therapeutic intervention possibilities. Methods : This study systematically characterized changes in behavioral abnormalities in APP/PS1 transgenic mice. Brain pathol. measures were performed in post-mortem brain tissues of mice from 2 to 22 mo. Results : APP/PS1 mice exhibited significant memory deficits from 5 mo old, which were aggravated at the later stage of life. However, the degree of memory impairments reached a plateau at 12 mo. An early appearance of amyloid plaques was at 3 mo with a linear increase throughout the disease course. CD11b-pos. microglia and glial fibrillary acidic protein-(GFAP) pos. astrocytes were first detected at 3 mo with a close assocn. with amyloid plaques. Yet, the rate of changes in glial activation slowed down from 12 mo despite the steady increase in Aβ. Conclusion : These findings provided evidence that neuroinflammation might be involved in the development and progression of cognitive deficits in APP/PS1 mice, suggesting novel intervention and prevention strategies for AD.
- 4Demuro, A., Mina, E., Kayed, R., Milton, S. C., Parker, I., and Glabe, C. G. (2005) Calcium Dysregulation and Membrane Disruption as a Ubiquitous Neurotoxic Mechanism of Soluble Amyloid Oligomers. J. Biol. Chem. 280 (17), 17294– 17300, DOI: 10.1074/jbc.M5009972004https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXjsFOguro%253D&md5=bbecbaadfc5f74d42bd09f2cddcf5763Calcium Dysregulation and Membrane Disruption as a Ubiquitous Neurotoxic Mechanism of Soluble Amyloid OligomersDemuro, Angelo; Mina, Erene; Kayed, Rakez; Milton, Saskia C.; Parker, Ian; Glabe, Charles G.Journal of Biological Chemistry (2005), 280 (17), 17294-17300CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Increasing evidence suggests that amyloid peptides assocd. with a variety of degenerative diseases induce neurotoxicity in their intermediate oligomeric state, rather than as monomers or fibrils. To test this hypothesis and investigate the possible involvement of Ca2+ signaling disruptions in amyloid-induced cytotoxicity, the authors made homogeneous prepns. of disease-related amyloids (Aβ, prion, islet amyloid polypeptide, polyglutamine, and lysozyme) in various aggregation states and tested their actions on fluo-3-loaded SH-SY5Y cells. Application of oligomeric forms of all amyloids tested (0.6-6 μg ml-1) rapidly (∼5 s) elevated intracellular Ca2+, whereas equiv. amts. of monomers and fibrils did not. Ca2+ signals evoked by Aβ42 oligomers persisted after depletion of intracellular Ca2+ stores, and small signals remained in Ca2+-free medium, indicating contributions from both extracellular and intracellular Ca2+ sources. The increased membrane permeability to Ca2+ cannot be attributed to activation of endogenous Ca2+ channels, because responses were unaffected by the potent Ca2+-channel blocker cobalt (20 μm). Instead, observations that Aβ42 and other oligomers caused rapid cellular leakage of anionic fluorescent dyes point to a generalized increase in membrane permeability. The resulting unregulated flux of ions and mols. may provide a common mechanism for oligomer-mediated toxicity in many amyloidogenic diseases, with dysregulation of Ca2+ ions playing a crucial role because of their strong trans-membrane concn. gradient and involvement in cell dysfunction and death.
- 5Perry, V. H., Nicoll, J. A. R., and Holmes, C. (2010) Microglia in Neurodegenerative Disease. Nat. Rev. Neurol. 6 (4), 193– 201, DOI: 10.1038/nrneurol.2010.175https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3c3kslyntw%253D%253D&md5=efb215a8a737c924905e7aa04a5b6706Microglia in neurodegenerative diseasePerry V Hugh; Nicoll James A R; Holmes CliveNature reviews. Neurology (2010), 6 (4), 193-201 ISSN:.Microglia, the resident macrophages of the CNS, are exquisitely sensitive to brain injury and disease, altering their morphology and phenotype to adopt a so-called activated state in response to pathophysiological brain insults. Morphologically activated microglia, like other tissue macrophages, exist as many different phenotypes, depending on the nature of the tissue injury. Microglial responsiveness to injury suggests that these cells have the potential to act as diagnostic markers of disease onset or progression, and could contribute to the outcome of neurodegenerative diseases. The persistence of activated microglia long after acute injury and in chronic disease suggests that these cells have an innate immune memory of tissue injury and degeneration. Microglial phenotype is also modified by systemic infection or inflammation. Evidence from some preclinical models shows that systemic manipulations can ameliorate disease progression, although data from other models indicates that systemic inflammation exacerbates disease progression. Systemic inflammation is associated with a decline in function in patients with chronic neurodegenerative disease, both acutely and in the long term. The fact that diseases with a chronic systemic inflammatory component are risk factors for Alzheimer disease implies that crosstalk occurs between systemic inflammation and microglia in the CNS.
- 6Paradisi, S., Sacchetti, B., Balduzzi, M., Gaudi, S., and Malchiodi-Albedi, F. (2004) Astrocyte Modulation of In Vitro β-Amyloid Neurotoxicity. Glia 46 (3), 252– 260, DOI: 10.1002/glia.200056https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2c7lsVerug%253D%253D&md5=d8fce7b7e08c280b10ed9f8b444a8936Astrocyte modulation of in vitro beta-amyloid neurotoxicityParadisi Silvia; Sacchetti Benedetto; Balduzzi Maria; Gaudi Simona; Malchiodi-Albedi FiorellaGlia (2004), 46 (3), 252-60 ISSN:0894-1491.In Alzheimer's disease brain, beta-amyloid (Abeta) deposition is accompanied by astrocyte activation, whose role in the pathogenesis of the disease is still unclear. To explore the subject, we compared Abeta neurotoxicity in pure hippocampal cultures and neuronal-astrocytic cocultures, where astrocytes conditioned neurons but were not in contact with them or Abeta. In the presence of astrocytes, neurons were protected from Abeta neurotoxicity. Neuritic dystrophy was reduced, synapses were partially preserved, and apoptosis was contrasted. The protection disappeared when astrocytes were also treated with Abeta, suggesting that Abeta-astrocyte interaction is deleterious for neurons. This was supported by comparing Abeta neurotoxicity in pure neurons and neurons grown on astrocytes. In this case, where astrocytes were also in contact with Abeta, neuritic damage was enhanced and expression of synaptic vesicle proteins decreased. Our results suggest that astrocytes can protect neurons from Abeta neurotoxicity, but when they interact with Abeta, the protection is undermined and neurotoxicity enhanced.
- 7Eikelenboom, P., Hoozemans, J. J., Veerhuis, R., van Exel, E., Rozemuller, A. J., and van Gool, W. A. (2012) Whether, When and How Chronic Inflammation Increases the Risk of Developing Late-onset Alzheimer’s Disease. Alzheimer's Res. Ther. 4 (3), 15– 15, DOI: 10.1186/alzrt1187https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xpt1ajurw%253D&md5=a2a92116ef2fb0e70170b35ee945713cWhether, when and how chronic inflammation increases the risk of developing late-onset Alzheimer's diseaseEikelenboom, Piet; Hoozemans, Jeroen J. M.; Veerhuis, Rob; van Exel, Eric; Rozemuller, Annemieke J. M.; van Gool, Willem A.Alzheimer's Research & Therapy (2012), 4 (3), 15CODEN: ARTLCD; ISSN:1758-9193. (BioMed Central Ltd.)A review. Neuropathol. studies have revealed the presence of a broad variety of inflammation-related proteins (complement factors, acute-phase proteins, pro-inflammatory cytokines) in Alzheimer's disease (AD) brains. These constituents of innate immunity are involved in several crucial pathogenic events of the underlying pathol. cascade in AD and recent studies have shown that innate immunity is involved in the etiol. of late-onset AD. Genome-wide assocn. studies have demonstrated gene loci that are linked to the complement system. Neuropathol. and exptl. studies indicate that fibrillar amyloid-β (Aβ) can activate the innate immunity-related CD14 and Toll-like receptor signaling pathways of glial cells for pro-inflammatory cytokine prodn. The prodn. capacity of this pathway is under genetic control and offspring with a parental history of late-onset AD have a higher prodn. capacity for pro-inflammatory cytokines. The activation of microglia by fibrillar Aβ deposits in the early preclin. stages of AD can make the brain susceptible later on for a second immune challenge leading to enhanced prodn. of pro-inflammatory cytokines. An example of a second immune challenge could be systemic inflammation in patients with preclin. AD. Prospective epidemiol. studies show that elevated serum levels of acute phase reactants can be considered as a risk factor for AD. Clin. studies suggest that peripheral inflammation increases the risk of dementia, esp. in patients with preexistent cognitive impairment and accelerates further deterioration in demented patients. The view that peripheral inflammation can increase the risk of dementia in older people provides scope for prevention.
- 8Lim, S., Chun, Y., Lee, J. S., and Lee, S.-J. (2016) Neuroinflammation in Synucleinopathies. Brain Pathol. 26 (3), 404– 409, DOI: 10.1111/bpa.123718https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC28jmslyjsw%253D%253D&md5=0db4eb9a45b73151df154d7ce22d55a9Neuroinflammation in SynucleinopathiesLim Somin; Chun Yewon; Lee Jun Sung; Lee Seung-JaeBrain pathology (Zurich, Switzerland) (2016), 26 (3), 404-9 ISSN:.The causes of most neurodegenerative diseases are attributed to multiple genetic and environmental factors interacting with one another. Above all, inflammation in the nervous system has been implicated in many neurodegenerative diseases. Still, the roles of neuroinflammation in disease mechanisms and the triggers of inflammatory responses in disease-inflicted brain tissues seem to remain unclear. This review will examine previous studies that had been done from genetic, pathological and epidemiological perspectives. These studies assess the involvement of neuroinflammation in synucleinopathies, a group of neurodegenerative diseases that are characterized by deposition of α-synuclein aggregates such as Parkinson's disease, dementia with Lewy bodies and multiple system atrophy. The review will also discuss the role of α-synuclein aggregates in triggering inflammatory responses from glial cells. It is expected that a precise assessment of the roles and mechanisms of neuroinflammation in neurodegenerative diseases will pave the way for the development of disease-modifying drugs.
- 9Wang, C., Iashchishyn, I. A., Pansieri, J., Nystrom, S., Klementieva, O., Kara, J., Horvath, I., Moskalenko, R., Rofougaran, R., Gouras, G. (2018) S100A9-Driven Amyloid-Neuroinflammatory Cascade in Traumatic Brain Injury as a Precursor State for Alzheimer’s Disease. Sci. Rep. 8, 12836, DOI: 10.1038/s41598-018-31141-x9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3c3itlOitw%253D%253D&md5=f4623ac4ea93ef3ab98aa20a3c70f5cfS100A9-Driven Amyloid-Neuroinflammatory Cascade in Traumatic Brain Injury as a Precursor State for Alzheimer's DiseaseWang Chao; Iashchishyn Igor A; Pansieri Jonathan; Kara John; Horvath Istvan; Moskalenko Roman; Rofougaran Reza; Morozova-Roche Ludmilla A; Iashchishyn Igor A; Nystrom Sofie; Klementieva Oxana; Gouras Gunnar; Moskalenko Roman; Kovacs Gabor G; Shankar S KScientific reports (2018), 8 (1), 12836 ISSN:.Pro-inflammatory and amyloidogenic S100A9 protein is an important contributor to Alzheimer's disease (AD) pathology. Traumatic brain injury (TBI) is viewed as a precursor state for AD. Here we have shown that S100A9-driven amyloid-neuroinflammatory cascade was initiated in TBI and may serve as a mechanistic link between TBI and AD. By analyzing the TBI and AD human brain tissues, we demonstrated that in post-TBI tissues S100A9, produced by neurons and microglia, becomes drastically abundant compared to Aβ and contributes to both precursor-plaque formation and intracellular amyloid oligomerization. Conditions implicated in TBI, such as elevated S100A9 concentration, acidification and fever, provide strong positive feedback for S100A9 nucleation-dependent amyloid formation and delay in its proteinase clearance. Consequently, both intracellular and extracellular S100A9 oligomerization correlated with TBI secondary neuronal loss. Common morphology of TBI and AD plaques indicated their similar initiation around multiple aggregation centers. Importantly, in AD and TBI we found S100A9 plaques without Aβ. S100A9 and Aβ plaque pathology was significantly advanced in AD cases with TBI history at earlier age, signifying TBI as a risk factor. These new findings highlight the detrimental consequences of prolonged post-TBI neuroinflammation, which can sustain S100A9-driven amyloid-neurodegenerative cascade as a specific mechanism leading to AD development.
- 10Horvath, I., Iashchishyn, I. A., Moskalenko, R. A., Wang, C., Warmlander, S. K. T. S., Wallin, C., Graslund, A., Kovacs, G. G., and Morozova-Roche, L. A. (2018) Co-aggregation of Pro-inflammatory S100A9 with Alpha-Synuclein in Parkinson’s Disease: Ex Vivo and In Vitro Studies. J. Neuroinflammation 15, 172, DOI: 10.1186/s12974-018-1210-910https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisFWgurrK&md5=713ff8fce99cbbd976b097ec98515dbcCo-aggregation of pro-inflammatory S100A9 with α-synuclein in Parkinson's disease: ex vivo and in vitro studiesHorvath, Istvan; Iashchishyn, Igor A.; Moskalenko, Roman A.; Wang, Chao; Waermlaender, Sebastian K. T. S.; Wallin, Cecilia; Graeslund, Astrid; Kovacs, Gabor G.; Morozova-Roche, Ludmilla A.Journal of Neuroinflammation (2018), 15 (), 172/1-172/16CODEN: JNOEB3; ISSN:1742-2094. (BioMed Central Ltd.)Background: Chronic neuroinflammation is a hallmark of Parkinson's disease (PD) pathophysiol., assocd. with increased levels of pro-inflammatory factors in PD brain tissues. The pro-inflammatory mediator and highly amyloidogenic protein S100A9 is involved in the amyloid-neuroinflammatory cascade in Alzheimer's disease. This is the first report on the co-aggregation of α-synuclein (α-syn) and S100A9 both in vitro and ex vivo in PD brain. Methods: Single and sequential immunohistochem., immunofluorescence, scanning electron and at. force (AFM) microscopies were used to analyze the ex vivo PD brain tissues for S100A9 and α-syn location and aggregation. In vitro studies revealing S100A9 and α-syn interaction and co-aggregation were conducted by NMR, CD, Thioflavin-T fluorescence, AFM, and surface plasmon resonance methods. Results: Co-localized and co-aggregated S100A9 and α-syn were found in 20% Lewy bodies and 77% neuronal cells in the substantia nigra; both proteins were also obsd. in Lewy bodies in PD frontal lobe (Braak stages 4-6). Lewy bodies were characterized by ca. 10-23 μm outer diam., with S100A9 and α-syn being co-localized in the same lamellar structures. S100A9 was also detected in neurons and blood vessels of the aged patients without PD, but in much lesser extent. In vitro S100A9 and α-syn were shown to interact with each other via the α-syn C-terminus with an apparent dissocn. const. of ca. 5 μM. Their co-aggregation occurred significantly faster and led to formation of larger amyloid aggregates than the self-assembly of individual proteins. S100A9 amyloid oligomers were more toxic than those of α-syn, while co-aggregation of both proteins mitigated the cytotoxicity of S100A9 oligomers. Conclusions: We suggest that sustained neuroinflammation promoting the spread of amyloidogenic S100A9 in the brain tissues may trigger the amyloid cascade involving α-syn and S100A9 and leading to PD, similar to the effect of S100A9 and Aβ co-aggregation in Alzheimer's disease. The finding of S100A9 involvement in PD may open a new avenue for therapeutic interventions targeting S100A9 and preventing its amyloid self-assembly in affected brain tissues.
- 11Wang, C., Klechikov, A. G., Gharibyan, A. L., Wärmländer, S. K. T. S., Jarvet, J., Zhao, L., Jia, X., Narayana, V. K., Shankar, S. K., Olofsson, A. (2014) The Role of Pro-inflammatory S100A9 in Alzheimer’s Disease Amyloid-neuroinflammatory Cascade. Acta Neuropathol. 127 (4), 507– 522, DOI: 10.1007/s00401-013-1208-411https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslymu7bF&md5=af7f01a5c3f6f24fc24aa907b72e9e43The role of pro-inflammatory S100A9 in Alzheimer's disease amyloid-neuroinflammatory cascadeWang, Chao; Klechikov, Alexey G.; Gharibyan, Anna L.; Waermlaender, Sebastian K. T. S.; Jarvet, Juri; Zhao, Lina; Jia, Xueen; Shankar, S. K.; Olofsson, Anders; Braennstroem, Thomas; Mu, Yuguang; Graeslund, Astrid; Morozova-Roche, Ludmilla A.Acta Neuropathologica (2014), 127 (4), 507-522CODEN: ANPTAL; ISSN:0001-6322. (Springer)Pro-inflammatory S100A9 protein is increasingly recognized as an important contributor to inflammation-related neurodegeneration. Here, we provide insights into S100A9 specific mechanisms of action in Alzheimer's disease (AD). Due to its inherent amyloidogenicity S100A9 contributes to amyloid plaque formation together with Aβ. In traumatic brain injury (TBI) S100A9 itself rapidly forms amyloid plaques, which were reactive with oligomer-specific antibodies, but not with Aβ and amyloid fibrillar antibodies. They may serve as precursor-plaques for AD, implicating TBI as an AD risk factor. S100A9 was obsd. in some hippocampal and cortical neurons in TBI, AD and non-demented aging. In vitro S100A9 forms neurotoxic linear and annular amyloids resembling Aβ protofilaments. S100A9 amyloid cytotoxicity and native S100A9 pro-inflammatory signaling can be mitigated by its co-aggregation with Aβ, which results in a variety of micron-scale amyloid complexes. NMR and mol. docking demonstrated transient interactions between native S100A9 and Aβ. Thus, abundantly present in AD brain pro-inflammatory S100A9, possessing also intrinsic amyloidogenic properties and ability to modulate Aβ aggregation, can serve as a link between the AD amyloid and neuroinflammatory cascades and as a prospective therapeutic target.
- 12Pruenster, M., Vogl, T., Roth, J., and Sperandio, M. (2016) S100A8/A9: From Basic Science to Clinical Application. Pharmacol. Ther. 167, 120– 131, DOI: 10.1016/j.pharmthera.2016.07.01512https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtlWjurzJ&md5=99ce16a70a32600c9f393ae6bfef9b90S100A8/A9: From basic science to clinical applicationPruenster, Monika; Vogl, Thomas; Roth, Johannes; Sperandio, MarkusPharmacology & Therapeutics (2016), 167 (), 120-131CODEN: PHTHDT; ISSN:0163-7258. (Elsevier)Neutrophils and monocytes belong to the first line of immune defense cells and are recruited to sites of inflammation during infection or sterile injury. Both cells contain huge amts. of the heterodimeric protein S100A8/A9 in their cytoplasm. S100A8/A9 belongs to the Ca2 + binding S100 protein family and has recently gained a lot of interest as a crit. alarmin modulating the inflammatory response after its release (extracellular S100A8/A9) from neutrophils and monocytes. Extracellular S100A8/A9 interacts with the pattern recognition receptors Toll-like receptor 4 (TLR4) and Receptor for Advanced Glycation Endproducts (RAGE) promoting cell activation and recruitment. Besides its biol. function, S100A8/A9 (also known as myeloid related protein 8/14, MRP8/14) was identified as interesting biomarker to monitor disease activity in chronic inflammatory disorders including inflammatory bowel disease and rheumatoid arthritis. Furthermore, S100A8/A9 has been tested successfully in pre-clin. imaging studies to localize sites of infection or sterile injury. Finally, recent evidence using small mol. inhibitors for S100A8/A9 also suggests that blocking S100A8/A9 activity exerts beneficial effects on disease activity in animal models of autoimmune diseases including multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis and inflammatory bowel disease. This review will provide a comprehensive and detailed overview into the structure and biol. function of S100A8/A9 and also will give an outlook in terms of diagnostic and therapeutic applications targeting S100A8/A9.
- 13Markowitz, J. and Carson, W. E., 3rd. (2013) Review of S100A9 Biology and Its Role in Cancer. Biochim. Biophys. Acta, Rev. Cancer 1835 (1), 100– 109, DOI: 10.1016/j.bbcan.2012.10.00313https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhvVKqurzJ&md5=b417186ba1010bf09ea0e350e2acd708Review of S100A9 biology and its role in cancerMarkowitz, Joseph; Carson, William E.Biochimica et Biophysica Acta, Reviews on Cancer (2013), 1835 (1), 100-109CODEN: BBACEU; ISSN:0304-419X. (Elsevier B.V.)A review. S100A9 is a calcium binding protein with multiple ligands and post-translation modifications that is involved in inflammatory events and the initial development of the cancer cell through to the development of metastatic disease. This review has a threefold purpose: 1) describe the S100A9 structural elements important for its biol. activity, 2) describe the S100A9 biol. in the context of the immune system, and 3) illustrate the role of S100A9 in the development of malignancy via interactions with the immune system and other cellular processes.
- 14Schluesener, H. J., Kremsner, P. G., and Meyermann, R. (1998) Widespread Expression of MRP8 and MRP14 in Human Cerebral Malaria by Microglial Cells. Acta Neuropathol. 96 (6), 575– 580, DOI: 10.1007/s00401005093814https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXmvFyitLo%253D&md5=4db5ed68a417b831c80894978e554c30Widespread expression of MRP8 and MRP14 in human cerebral malaria by microglial cellsSchluesener, H. J.; Kremsner, Peter G.; Meyermann, RichardActa Neuropathologica (1998), 96 (6), 575-580CODEN: ANPTAL; ISSN:0001-6322. (Springer-Verlag)Human cerebral malaria (CM) is an often fatal infection. The cascades of signaling events resulting in tissue trauma and coma are only slowly becoming unraveled. Here the authors report that microglial cells, sensitive cellular sensors of threats to the central nervous system, in CM express the myeloid-related proteins MRP8 (S100A8) and MRP14 (S100A9), Ca2+-binding sensor proteins of activated monocytes. Surprisingly, microglial activation was widespread throughout the brain in white and gray matter and not limited to areas of petechial bleedings or sequestration of infected erythrocytes. Further, apoptosis/necrosis is prominent in CM; not only leukocytes appeared apoptotic, neurons also appeared damaged and DNA fragmentation was revealed by in situ nick translation. Thus, a prominent feature of human CM is activation of microglia, and anal. of these reactive microglia might further promote the authors' understanding of CM pathol. and guide development of future therapeutic intervention of the local reactive processes.
- 15Postler, E., Lehr, A., Schluesener, H., and Meyermann, R. (1997) Expression of the S-100 proteins MRP-8 and −14 in Ischemic Brain Lesions. Glia 19 (1), 27– 34, DOI: 10.1002/(SICI)1098-1136(199701)19:1<27::AID-GLIA3>3.0.CO;2-715https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK2s7ktlGmtg%253D%253D&md5=b269af3ced945688d66f310da4d3e5dcExpression of the S-100 proteins MRP-8 and -14 in ischemic brain lesionsPostler E; Lehr A; Schluesener H; Meyermann RGlia (1997), 19 (1), 27-34 ISSN:0894-1491.So far, microglial activation in cerebral ischemia has only been studied in different animal models. We have investigated the activation of microglial cells in human cerebral ischemia. As a marker for the activation of these "brain macrophages," we have used the macrophage inhibitor factor-related-proteins MRP-8 and MRP-14, which belong to the calcium binding S-100 protein family. The proteins can be detected on microglial cells in bacterial encephalitis and Alzheimer's disease but have so far not been studied in non-inflammatory diseases, in which microglial activation also occurs. Antibodies against MRP-8 and -14 detected ramified microglial cells within the first 3 days after cerebral infarction. Labeled cells were found selectively in the periinfarctional area. To support the notion that these cells belong to the locally activated resident microglial population, we studied their proliferation rate by staining the Ki-67 antigen with the antibody MIB-1. Double-labeling clearly showed that in the early phase of cerebral infarction microglial cells in the periinfarctional area express MRP-8 and -14 and also proliferate. Surprisingly, MRPs are expressed no longer than 3 days post infarction. This indicates that the activation of the resident microglia is an early step of tissue reaction after cerebral infarction. Additionally, we found evidence that microglial cells contribute to the population of phagocytes only during the first 3 days post infarction. The majority of lipid phagocytes found in the later stages are obviously recruited from the blood-borne macrophage pool.
- 16Nagareddy, P. R., Murphy, A. J., Stirzaker, R. A., Hu, Y., Yu, S., Miller, R. G., Ramkhelawon, B., Distel, E., Westerterp, M., Huang, L.-S. (2013) Hyperglycemia Promotes Myelopoiesis and Impairs the Resolution of Atherosclerosis. Cell Metab. 17 (5), 695– 708, DOI: 10.1016/j.cmet.2013.04.00116https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXnsVyqs7Y%253D&md5=ff11d96d88b7b22191ca54f90233b5d4Hyperglycemia Promotes Myelopoiesis and Impairs the Resolution of AtherosclerosisNagareddy, Prabhakara R.; Murphy, Andrew J.; Stirzaker, Roslynn A.; Hu, Yunying; Yu, Shiquing; Miller, Rachel G.; Ramkhelawon, Bhama; Distel, Emilie; Westerterp, Marit; Huang, Li-Shin; Schmidt, Ann Marie; Orchard, Trevor J.; Fisher, Edward A.; Tall, Alan R.; Goldberg, Ira J.Cell Metabolism (2013), 17 (5), 695-708CODEN: CMEEB5; ISSN:1550-4131. (Elsevier Inc.)Diabetes is a major risk factor for atherosclerosis. Although atherosclerosis is initiated by deposition of cholesterol-rich lipoproteins in the artery wall, the entry of inflammatory leukocytes into lesions fuels disease progression and impairs resoln. We show that diabetic mice have increased nos. of circulating neutrophils and Ly6-Chi monocytes, reflecting hyperglycemia-induced proliferation and expansion of bone marrow myeloid progenitors and release of monocytes into the circulation. Increased neutrophil prodn. of S100A8/S100A9, and its subsequent interaction with the receptor for advanced glycation end products on common myeloid progenitor cells, leads to enhanced myelopoiesis. Treatment of hyperglycemia reduces monocytosis, entry of monocytes into atherosclerotic lesions, and promotes regression. In patients with type 1 diabetes, plasma S100A8/S100A9 levels correlate with leukocyte counts and coronary artery disease. Thus, hyperglycemia drives myelopoiesis and promotes atherogenesis in diabetes.
- 17Ma, L.-P., Haugen, E., Ikemoto, M., Fujita, M., Terasaki, F., and Fu, M. (2012) S100A8/A9 Complex as a New Biomarker in Prediction of Mortality in Elderly Patients with Severe Heart Failure. Int. J. Cardiol. 155 (1), 26– 32, DOI: 10.1016/j.ijcard.2011.01.08217https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC383gsFyiuw%253D%253D&md5=9e31f29e08d75738e968ef7fbe218022S100A8/A9 complex as a new biomarker in prediction of mortality in elderly patients with severe heart failureMa Li-Ping; Haugen Espen; Ikemoto Masaki; Fujita Masatoshi; Terasaki Fumio; Fu MichaelInternational journal of cardiology (2012), 155 (1), 26-32 ISSN:.BACKGROUND: S100A8/A9 complex is a new inflammation-related protein and has a positive correlation with C-reaction protein level. However its role in chronic heart failure (CHF) remains unclear. METHODS AND RESULTS: Circulating levels of S100A8/A9 complex and other biomarkers (IL-6, IL-8, TNF-α, and BNP) were measured in CHF (n = 54) and hypertensive without CHF (n = 31) as well as healthy subjects (n = 23), with follow up to 1480 days. During follow-up, cumulative mortality rate for CHF patients was 63%. Plasma levels of S100A8/A9 complex, IL-6, IL-8 and TNF-α were significantly higher in CHF than the hypertensive patients and healthy subjects. A significant positive correlation was found between S100A8/A9 complex and IL-6 and IL-8 respectively. Cox regression analysis showed that IL-6 and IL-8 were predictors for mortality for 6 months, and S100A8/A9 complex, IL-6, IL-8 and age were predictors for mortality for one year whereas BNP, TNF-α, IL-6 and IL-8 remained predictors for mortality for two years. A combination of S100A8/A9 complex and IL-6 provided powerful predictive value in mortality for both 6 and 12 months. CONCLUSIONS: S100A8/A9 complex is a useful biomarker as a predictor for one year mortality and its combination with IL-6 is able to provide additive prognostic information in this vulnerable heart failure population in the elderly.
- 18Swindell, W. R., Johnston, A., Xing, X., Little, A., Robichaud, P., Voorhees, J. J., Fisher, G., and Gudjonsson, J. E. (2013) Robust Shifts in S100A9 Expression with Aging: A Novel Mechanism for Chronic Inflammation. Sci. Rep. 3 (1), 1215, DOI: 10.1038/srep0121518https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXpvFWjt74%253D&md5=8ff1b1f10a81db6c7d8496626fc2c72bRobust shifts in S100a9 expression with aging: a novel mechanism for chronic inflammationSwindell, William R.; Johnston, Andrew; Xing, Xianying; Little, Andrew; Robichaud, Patrick; Voorhees, John J.; Fisher, Gary; Gudjonsson, Johann E.Scientific Reports (2013), 3 (), 1215, 13 pp.CODEN: SRCEC3; ISSN:2045-2322. (Nature Publishing Group)The S100a8 and S100a9 genes encode a pro-inflammatory protein (calgranulin) that has been implicated in multiple diseases. However, involvement of S100a8/a9 in the basic mechanisms of intrinsic aging has not been established. In this study, we show that shifts in the abundance of S100a8 and S100a9 mRNA are a robust feature of aging in mammalian tissues, involving a range of cell types including the central nervous system. To identify transcription factors that control S100a9 expression, we performed a large-scale transcriptome anal. of 62 mouse and human cell types. We identified cell type-specific trends, as well as robust assocns. linking S100a9 coexpression to elevated frequency of ETS family motifs and in particular, to motifs recognized by the transcription factor SPI/PU.1. Sparse occurrence of SATB1 motifs was also a strong predictor of S100a9 coexpression. These findings offer support for a novel mechanism by which a SPI1/PU.1-S100a9 axis sustains chronic inflammation during aging.
- 19Iashchishyn, I. A., Sulskis, D., Nguyen Ngoc, M., Smirnovas, V., and Morozova-Roche, L. A. (2017) Finke–Watzky Two-Step Nucleation–Autocatalysis Model of S100A9 Amyloid Formation: Protein Misfolding as “Nucleation” Event. ACS Chem. Neurosci. 8 (10), 2152– 2158, DOI: 10.1021/acschemneuro.7b0025119https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXht1Cltb3P&md5=9e6270edc1a9e7e2d0277ab483d7f14fFinke-Watzky Two-Step Nucleation-Autocatalysis Model of S100A9 Amyloid Formation: Protein Misfolding as "Nucleation" EventIashchishyn, Igor A.; Sulskis, Darius; Nguyen Ngoc, Mai; Smirnovas, Vytautas; Morozova-Roche, Ludmilla A.ACS Chemical Neuroscience (2017), 8 (10), 2152-2158CODEN: ACNCDM; ISSN:1948-7193. (American Chemical Society)Quant. kinetic anal. is crit. for understanding amyloid mechanisms. Here, we demonstrate the application of the generic Finke-Watzky (F-W; 1997, 2008) 2-step nucleation-autocatalytic growth model to concn.-dependent amyloid kinetics of proinflammatory α-helical S100A9 protein/calgranulin B at pH 7.4 and temps. of 37 and 42°. The model is based on 2 pseudo-elementary reaction steps applied without further anal. constraints and its treatment of S100A9 amyloid self-assembly demonstrates that initial misfolding and β-sheet formation, defined as a "nucleation" step, spontaneously takes place within individual S100A9 mols. at a higher rate than the subsequent fibrillar growth. The latter, described as an autocatalytic process, will proceed if misfolded amyloid-prone S100A9 is populated on a macroscopic time scale. Short lengths of S100A9 fibrils were consistent with the F-W model. The anal. of fibrillar length distribution by the Bekker-Doering model demonstrated independently that such distribution was solely detd. by slow fibril growth and that there was no fragmentation or secondary pathways decreasing fibrillar length.
- 20Yanamandra, K., Alexeyev, O., Zamotin, V., Srivastava, V., Shchukarev, A., Brorsson, A.-C., Tartaglia, G. G., Vogl, T., Kayed, R., Wingsle, G. (2009) Amyloid Formation by the Pro-inflammatory S100A8/A9 Proteins in the Ageing Prostate. PLoS One 4 (5), e5562– e5562, DOI: 10.1371/journal.pone.0005562There is no corresponding record for this reference.
- 21Eremenko, E., Ben-Zvi, A., Morozova-Roche, L. A., and Raveh, D. (2013) Aggregation of Human S100A8 and S100A9 Amyloidogenic Proteins Perturbs Proteostasis in a Yeast Model. PLoS One 8 (3), e58218 DOI: 10.1371/journal.pone.005821821https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXktlWjsrg%253D&md5=adb676af9433824aad66ff794da89062Aggregation of human S100A8 and S100A9 amyloidogenic proteins perturbs proteostasis in a yeast modelEremenko, Ekaterina; Ben-Zvi, Anat; Morozova-Roche, Ludmilla A.; Raveh, DinaPLoS One (2013), 8 (3), e58218CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)Amyloid aggregates of the calcium-binding EF-hand proteins S100A8 and S100A9 have been found in the corpora amylacea of patients with prostate cancer and may play a role in carcinogenesis. Here, the authors present a novel model system using the yeast Saccharomyces cerevisiae to study human S100A8 and S100A9 aggregation and toxicity. They found that S100A8, S100A9 and S100A8/9 cotransfomants form SDS-resistant non-toxic aggregates in yeast cells. Using fluorescently tagged proteins, the authors showed that S100A8 and S100A9 accumulate in foci. After prolonged induction, S100A8 foci localized to the cell vacuole, whereas the S100A9 foci remained in the cytoplasm when present alone, but entered the vacuole in cotransformants. Biochem. anal. of the proteins indicated that S100A8 and S100A9 alone or coexpressed together form amyloid-like aggregates in yeast. Expression of S100A8 and S100A9 in wild-type yeast did not affect cell viability, but these proteins were toxic when expressed on a background of unrelated metastable temp.-sensitive mutant proteins, Cdc53-1p, Cdc34-2p, Srp1-31p and Sec27-1p. This finding suggests that the expression and aggregation of S100A8 and S100A9 may limit the capacity of the cellular proteostasis machinery. To test this hypothesis, the authors screened a set of chaperone deletion mutants and found that reducing the levels of the heat-shock proteins Hsp104p and Hsp70p was sufficient to induce S100A8 and S100A9 toxicity. This result indicates that the chaperone activity of the Hsp104/Hsp70 bichaperone system in wild type cells is sufficient to reduce S100A8 and S100A9 amyloid toxicity and preserve cellular proteostasis. Expression of human S100A8 and S100A9 in yeast thus provides a novel model system for the study of the interaction of amyloid deposits with the proteostasis machinery.
- 22Horvath, I., Jia, X., Johansson, P., Wang, C., Moskalenko, R., Steinau, A., Forsgren, L., Wågberg, T., Svensson, J., Zetterberg, H., and Morozova-Roche, L. A. (2016) Pro-inflammatory S100A9 Protein as a Robust Biomarker Differentiating Early Stages of Cognitive Impairment in Alzheimer’s Disease. ACS Chem. Neurosci. 7 (1), 34– 39, DOI: 10.1021/acschemneuro.5b0026522https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhslyktbjM&md5=9a2362b63b01d23b69f4dd2b4e6c5c88Pro-inflammatory S100A9 Protein as a Robust Biomarker Differentiating Early Stages of Cognitive Impairment in Alzheimer's DiseaseHorvath, Istvan; Jia, Xueen; Johansson, Per; Wang, Chao; Moskalenko, Roman; Steinau, Andreas; Forsgren, Lars; Waagberg, Thomas; Svensson, Johan; Zetterberg, Henrik; Morozova-Roche, Ludmilla A.ACS Chemical Neuroscience (2016), 7 (1), 34-39CODEN: ACNCDM; ISSN:1948-7193. (American Chemical Society)Pro-inflammatory protein S100A9 was established as a biomarker of dementia progression and compared with others such as Aβ1-42 and tau-proteins. CSF samples from 104 stringently diagnosed individuals divided into five subgroups were analyzed, including nondemented controls, stable mild cognitive impairment (SMCI), mild cognitive impairment due to Alzheimer's disease (MCI-AD), Alzheimer's disease (AD), and vascular dementia (VaD) patients. ELISA, dot-blotting, and electrochem. impedance spectroscopy were used as research methods. The S100A9 and Aβ1-42 levels correlated with each other: their CSF content decreased already at the SMCI stage and declined further under MCI-AD, AD, and VaD conditions. Immunohistochem. anal. also revealed involvement of both Aβ1-42 and S100A9 in the amyloid-neuroinflammatory cascade already during SMCI. Tau proteins were not yet altered in SMCI; however their contents increased during MCI-AD and AD, diagnosing later dementia stages. Thus, four biomarkers together, reflecting different underlying pathol. causes, can accurately differentiate dementia progression and also distinguish AD from VaD.
- 23Stefani, M. and Rigacci, S. (2014) Beneficial Properties of Natural Phenols: Highlight on Protection Against Pathological Conditions Associated with Amyloid Aggregation. BioFactors 40 (5), 482– 493, DOI: 10.1002/biof.117123https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvVWnsLfM&md5=6fe575d1bd7e30a74c54978a209a92f9Beneficial properties of natural phenols: Highlight on protection against pathological conditions associated with amyloid aggregationStefani, Massimo; Rigacci, StefaniaBioFactors (2014), 40 (5), 482-493CODEN: BIFAEU; ISSN:1872-8081. (Wiley-Blackwell)A review. Mediterranean and Asian diets are currently considered as the most healthy traditional feeding habits effective against risk of age-assocd., particularly cardiovascular and neurodegenerative, diseases. A common feature of these two regimens is the abundance of foods and beverages of plant origin (green tea, extra virgin olive oil, red wine, spices, berries, and arom. herbs) that are considered responsible for the obsd. beneficial effects. Epidemiol. data suggest that the phenolic component remarkably enriched in these foods plays an important role in reducing the incidence of amyloid diseases, pathol. conditions assocd. to tissue deposition of toxic protein aggregates responsible for progressive functional deterioration. Great effort is being spent to provide knowledge on the effects of several natural phenols in this context, moving from the test tube to animal models and, more slowly, to the patient's bed. An emerging feature that makes these mols. increasingly attractive for amyloid disease prevention and therapy is their wide spectrum of activity: recent pieces of evidence suggest that they can inhibit the prodn. of amyloidogenic peptides from precursors, increase antioxidant enzyme activity, activate autophagy and reduce inflammation. Our concept should than shift from considering natural phenols simply as antioxidants or, at the best, as amyloid aggregation inhibitors, to describing them as potentially multitargeting drugs. A main concern is the low bioavailability of such compds. and efforts aimed at improving it are underway, with encapsulation strategies being the most promising ones. © 2014 BioFactors, 40(5):482-493, 2014.
- 24Leri, M., Scuto, M., Ontario, M. L., Calabrese, V., Calabrese, E. J., Bucciantini, M., and Stefani, M. (2020) Healthy Effects of Plant Polyphenols: Molecular Mechanisms. Int. J. Mol. Sci. 21 (4), 1250, DOI: 10.3390/ijms2104125024https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhslGmt7%252FN&md5=8bfe06c440f984df718a7db0c12e74e0Healthy effects of plant polyphenols molecular mechanismsLeri, Manuela; Scuto, Maria; Ontario, Maria Laura; Calabrese, Vittorio; Calabrese, Edward J.; Bucciantini, Monica; Stefani, MassimoInternational Journal of Molecular Sciences (2020), 21 (4), 1250CODEN: IJMCFK; ISSN:1422-0067. (MDPI AG)The increasing extension in life expectancy of human beings in developed countries is accompanied by a progressively greater rate of degenerative diseases assocd. with lifestyle and aging, most of which are still waiting for effective, not merely symptomatic, therapies. Accordingly, at present, the recommendations aimed at reducing the prevalence of these conditions in the population are limited to a safer lifestyle including phys./mental exercise, a reduced caloric intake, and a proper diet in a convivial environment. The claimed health benefits of the Mediterranean and Asian diets have been confirmed in many clin. trials and epidemiol. surveys. These diets are characterized by several features, including low meat consumption, the intake of oils instead of fats as lipid sources, moderate amts. of red wine, and significant amts. of fresh fruit and vegetables. In particular, the latter have attracted popular and scientific attention for their content, though in reduced amts., of a no. of mols. increasingly investigated for their healthy properties. Among the latter, plant polyphenols have raised remarkable interest in the scientific community; in fact, several clin. trials have confirmed that many health benefits of the Mediterranean/Asian diets can be traced back to the presence of significant amts. of these mols., even though, in some cases, contradictory results have been reported, which highlights the need for further investigation. In light of the results of these trials, recent research has sought to provide information on the biochem., mol., epigenetic, and cell biol. modifications by plant polyphenols in cell, organismal, animal, and human models of cancer, metabolic, and neurodegenerative pathologies, notably Alzheimer's and Parkinson disease. The findings reported in the last decade are starting to help to decipher the complex relations between plant polyphenols and cell homeostatic systems including metabolic and redox equil., proteostasis, and the inflammatory response, establishing an increasingly solid mol. basis for the healthy effects of these mols. Taken together, the data currently available, though still incomplete, are providing a rationale for the possible use of natural polyphenols, or their mol. scaffolds, as nutraceuticals to contrast aging and to combat many assocd. pathologies.
- 25Rigacci, S., Guidotti, V., Bucciantini, M., Parri, M., Nediani, C., Cerbai, E., Stefani, M., and Berti, A. (2010) Oleuropein Aglycon Prevents Cytotoxic Amyloid Aggregation of Human Amylin. J. Nutr. Biochem. 21 (8), 726– 735, DOI: 10.1016/j.jnutbio.2009.04.01025https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXptVamt78%253D&md5=8571339f9624289049a490f0e25d965aOleuropein aglycon prevents cytotoxic amyloid aggregation of human amylinRigacci, Stefania; Guidotti, Valentina; Bucciantini, Monica; Parri, Matteo; Nediani, Chiara; Cerbai, Elisabetta; Stefani, Massimo; Berti, AndreaJournal of Nutritional Biochemistry (2010), 21 (8), 726-735CODEN: JNBIEL; ISSN:0955-2863. (Elsevier)Pancreatic amyloid deposits of amylin are a hallmark of Type II diabetes and considerable evidence indicates that amylin oligomers are cytotoxic to β-cells. Many efforts are presently spent to find out naturally occurring mols., or to design synthetic ones, able to hinder amylin aggregation or to protect cells against aggregate cytotoxicity. In this context, a protective effect of some polyphenols against amyloid cytotoxicity was reported. Actually dietary polyphenols are endowed with multiple health benefits, and extra virgin olive oil is attracting increasing interest as a source of these substances. Here, we investigated the effects on amylin aggregation and cytotoxicity of the secoiridoid oleuropein aglycon, the main phenolic component of extra virgin olive oil. We found that oleuropein, when present during the aggregation of amylin, consistently prevented its cytotoxicity to RIN-5F pancreatic β-cells, as detd. by the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide test and caspase-3 activity assay. A lack of interaction with the cell membrane of amylin aggregates grown in the presence of oleuropein was shown by fluorescence microscopy and synthetic lipid vesicle permeabilization. Moreover, our ThT assay, CD anal. and electron microscopy images suggested that oleuropein interferes with amylin aggregation, resulting in a different path skipping the formation of toxic pre-fibrillar aggregates. These results provide a mol. basis for some of the benefits potentially coming from extra virgin olive oil consumption and pave the way to further studies on the possible pharmacol. use of oleuropein to prevent or to slow down the progression of type II diabetes.
- 26Leri, M., Nosi, D., Natalello, A., Porcari, R., Ramazzotti, M., Chiti, F., Bellotti, V., Doglia, S. M., Stefani, M., and Bucciantini, M. (2016) The Polyphenol Oleuropein Aglycone Hinders the Growth of Toxic Transthyretin Amyloid Assemblies. J. Nutr. Biochem. 30, 153– 166, DOI: 10.1016/j.jnutbio.2015.12.00926https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xis1Gis7g%253D&md5=df648a9675770f47caa514e982d62264The polyphenol Oleuropein aglycone hinders the growth of toxic transthyretin amyloid assembliesLeri, Manuela; Nosi, Daniele; Natalello, Antonino; Porcari, Riccardo; Ramazzotti, Matteo; Chiti, Fabrizio; Bellotti, Vittorio; Doglia, Silvia Maria; Stefani, Massimo; Bucciantini, MonicaJournal of Nutritional Biochemistry (2016), 30 (), 153-166CODEN: JNBIEL; ISSN:0955-2863. (Elsevier)Transthyretin (TTR) is involved in a subset of familial or sporadic amyloid diseases including senile systemic amyloidosis (SSA), familial amyloid polyneuropathy and cardiomyopathy (FAP/FAC) for which no effective therapy has been found yet. These conditions are characterized by extracellular deposits primarily found in the heart parenchyma and in peripheral nerves whose main component are amyloid fibrils, presently considered the main culprits of cell sufferance. The latter are polymeric assemblies grown from misfolded TTR, either wt or carrying one out of many identified mutations. The recent introduction in the clin. practice of synthetic TTR-stabilizing mols. that reduce protein aggregation provides the rationale to search natural effective mols. able to interfere with TTR amyloid aggregation by hindering the appearance of toxic species or by favoring the growth of harmless aggregates. Here we carried out an in depth biophys. and morphol. study on the mol. features of the aggregation of wt- and L55P-TTR involved in SSA or FAP/FAC, resp., and on the interference with fibril aggregation, stability and toxicity to cardiac HL-1 cells to demonstrate the ability of Oleuropein aglycon (OleA), the main phenolic component of the extra virgin olive oil. We describe the mol. basis of such interference and the resulting redn. of TTR amyloid aggregate cytotoxicity. Our data offer the possibility to validate and optimize the use of OleA or its mol. scaffold to rationally design promising drugs against TTR-related pathologies that could enter a clin. exptl. phase.
- 27Rigacci, S., Guidotti, V., Bucciantini, M., Nichino, D., Relini, A., Berti, A., and Stefani, M. (2011) Aβ(1–42) Aggregates into Non-Toxic Amyloid Assemblies in the Presence of the Natural Polyphenol Oleuropein Aglycon. Curr. Alzheimer Res. 8 (8), 841– 852, DOI: 10.2174/15672051179819268227https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs1ShtrvN&md5=4280c13b299c367bc10a8e52145a8ffcAβ(1-42) aggregates into non-toxic amyloid assemblies in the presence of the natural polyphenol oleuropein aglyconRigacci, Stefania; Guidotti, Valentina; Bucciantini, Monica; Nichino, Daniela; Relini, Annalisa; Berti, Andrea; Stefani, MassimoCurrent Alzheimer Research (2011), 8 (8), 841-852CODEN: CARUBY; ISSN:1567-2050. (Bentham Science Publishers Ltd.)Amyloid aggregation starts with the initial misfolding of peptide/protein precursors, with subsequent structural rearrangement into oligomers and protofibrils; the latter eventually organize into fibrils with shared basic structural features, found deposited in amyloid diseases. Mounting evidence indicates early oligomers as the most toxic amyloid species; accordingly, the search of inhibitors of their growth is considered a promising target to prevent amyloid toxicity. We recently showed that oleuropein aglycon, a polyphenol abundant in the extra virgin olive oil, interferes with the aggregation of amylin (involved in type-2 diabetes), eliminating its cytotoxicity. Here we report that oleuropein aglycon also hinders amyloid aggregation of Aβ(1-42) and its cytotoxicity, suggesting a general effect of such polyphenol. In particular, by a wide panel of different spectroscopic, immunol., cell viability and imaging techniques we provide a more detailed description of Aβ(1-42) structural modifications arising in the presence of the inhibitor and the resulting cytotoxicity. We here report that the polyphenol eliminates the appearance of early toxic oligomers favoring the formation of stable harmless protofibrils, structurally different from the typical Aβ(1-42) fibrils. We also show that oleuropein aglycon is maximally effective when is present at the beginning of the aggregation process; furthermore, when added to pre-formed fibrils, it does not induce the release of toxic oligomers but, rather, neutralizes any residual toxicity possibly arising from the residual presence of traces of sol. oligomers and other toxic aggregates. The possible use of this polyphenol as anti-aggregation mol. is discussed in the light of these data.
- 28Palazzi, L., Bruzzone, E., Bisello, G., Leri, M., Stefani, M., Bucciantini, M., and Polverino de Laureto, P. (2018) Oleuropein Aglycone Stabilizes the Monomeric α-Synuclein and Favours the Growth of Non-Toxic Aggregates. Sci. Rep. 8 (1), 8337, DOI: 10.1038/s41598-018-26645-528https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MbhsFemtg%253D%253D&md5=e9bed4333ac2bf282a12dc0f4994ee47Oleuropein aglycone stabilizes the monomeric α-synuclein and favours the growth of non-toxic aggregatesPalazzi Luana; Bisello Giovanni; Polverino de Laureto Patrizia; Bruzzone Elena; Leri Manuela; Stefani Massimo; Bucciantini Monica; Leri ManuelaScientific reports (2018), 8 (1), 8337 ISSN:.α-synuclein plays a key role in the pathogenesis of Parkinson's disease (PD); its deposits are found as amyloid fibrils in Lewy bodies and Lewy neurites, the histopathological hallmarks of PD. Amyloid fibrillation is a progressive polymerization path starting from peptide/protein misfolding and proceeding through the transient growth of oligomeric intermediates widely considered as the most toxic species. Consequently, a promising approach of intervention against PD might be preventing α-synuclein build-up, misfolding and aggregation. A possible strategy involves the use of small molecules able to slow down the aggregation process or to alter oligomer conformation favouring the growth of non-pathogenic species. Here, we show that oleuropein aglycone (OleA), the main olive oil polyphenol, exhibits anti-amyloidogenic power in vitro by interacting with, and stabilizing, α-synuclein monomers thus hampering the growth of on-pathway oligomers and favouring the growth of stable and harmless aggregates with no tendency to evolve into other cytotoxic amyloids. We investigated the molecular basis of such interference by both biophysical techniques and limited proteolysis; aggregate morphology was monitored by electron microscopy. We also found that OleA reduces the cytotoxicity of α-synuclein aggregates by hindering their binding to cell membrane components and preventing the resulting oxidative damage to cells.
- 29Leri, M., Natalello, A., Bruzzone, E., Stefani, M., and Bucciantini, M. (2019) Oleuropein Aglycone and Hydroxytyrosol Interfere Differently with Toxic Aβ1–42 Aggregation. Food Chem. Toxicol. 129, 1– 12, DOI: 10.1016/j.fct.2019.04.01529https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXnvF2ht7s%253D&md5=3863c374b1298e87ce4fb3add2a6419fOleuropein aglycone and hydroxytyrosol interfere differently with toxic Aβ1-42 aggregationLeri, Manuela; Natalello, Antonino; Bruzzone, Elena; Stefani, Massimo; Bucciantini, MonicaFood and Chemical Toxicology (2019), 129 (), 1-12CODEN: FCTOD7; ISSN:0278-6915. (Elsevier Ltd.)Oleuropein aglycon (OleA), the most abundant polyphenol in extra virgin olive oil (EVOO), and Hydroxythyrosol (HT), the OleA main metabolite, have attracted our interest due to their multitarget effects, including the interference with amyloid aggregation path. However, the mechanistic details of their anti-amyloid effect are not known yet. We report here a broad biophys. approach and cell biol. techniques that enabled us to characterize the different mol. mechanisms by which OleA and HT modulate the Aβ1-42 fibrillation, a main histopathol. feature of Alzheimer's disease (AD). In particular, OleA prevents the growth of toxic Aβ1-42 oligomers and blocks their successive growth into mature fibrils following its interaction with the peptide N-terminus, while HT speeds up harmless fibril formation. Our data demonstrate that, by stabilizing oligomers and fibrils, both polyphenols reduce their seeding activity and aggregate/membrane interaction on human neuroblastoma SH-SY5Y cells. These findings highlight the great potential of EVOO polyphenols and offer the possibility to validate and to optimize their use for possible AD prevention and therapy.
- 30Palazzi, L., Leri, M., Cesaro, S., Stefani, M., Bucciantini, M., and Polverino de Laureto, P. (2020) Insight into the Molecular Mechanism Underlying the Inhibition of α-Synuclein Aggregation by Hydroxytyrosol. Biochem. Pharmacol. 173, 113722, DOI: 10.1016/j.bcp.2019.11372230https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXit12ktL7F&md5=a4445287929f44b1f225758d549bf706Insight into the molecular mechanism underlying the inhibition of α-synuclein aggregation by hydroxytyrosolPalazzi, Luana; Leri, Manuela; Cesaro, Samuele; Stefani, Massimo; Bucciantini, Monica; Polverino de Laureto, PatriziaBiochemical Pharmacology (Amsterdam, Netherlands) (2020), 173 (), 113722CODEN: BCPCA6; ISSN:0006-2952. (Elsevier B.V.)Parkinson's disease (PD) is the second most prevalent neurodegenerative disease in the elderly people. To date, drugs able to reverse the disease are not available; the gold std. is levodopa that only relieves clin. symptoms, yet with severe side effects after prolonged administration. Many efforts are underway to find alternative targets for PD prevention or treatment, the most promising being α-synuclein (Syn). Recently, we reported that oleuropein aglycon (OleA) interferes with amyloid aggregation of Syn both stabilizing its monomeric state and inducing the formation of harmless, off-pathway oligomers. This study is focused at describing the interaction between Syn and hydroxytyrosol (HT), the phenolic moiety and main metabolite of OleA, and the interferences with Syn aggregation by using biophys. and biol. techniques. Our results show that HT dose-dependently inhibits Syn aggregation and that covalent and non-covalent binding mediate HT-Syn interaction. HT does not modify the natively unfolded structure of Syn, rather, it stabilizes specific regions of the mol. leading to inhibition of protein fibrillation. Cellular assays showed that HT reduces the toxicity of Syn aggregates. Moreover, Syn aggregates interaction with the cell membrane, an important factor for prion-like properties of Syn on-pathway oligomers, was reduced in cells exposed to Syn aggregates grown in the presence of HT.
- 31Daccache, A., Lion, C., Sibille, N., Gerard, M., Slomianny, C., Lippens, G., and Cotelle, P. (2011) Oleuropein and Derivatives From Olives as Tau Aggregation Inhibitors. Neurochem. Int. 58 (6), 700– 707, DOI: 10.1016/j.neuint.2011.02.01031https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXksFGmtLw%253D&md5=94c9cf92c95514c8dc8b024f96d53ce2Oleuropein and derivatives from olives as Tau aggregation inhibitorsDaccache, Anthony; Lion, Cedric; Sibille, Nathalie; Gerard, Melanie; Slomianny, Christian; Lippens, Guy; Cotelle, PhilippeNeurochemistry International (2011), 58 (6), 700-707CODEN: NEUIDS; ISSN:0197-0186. (Elsevier Ltd.)Tau isoforms constitute a family of microtubule-assocd. proteins that are mainly expressed in neurons of the central nervous system. They promote the assembly of tubulin monomers into microtubules and modulate their stability, thus playing a key structural role in the distal portion of axons. In Alzheimer's disease and related tauopathies, Tau aggregation into fibrillary tangles contributes to intraneuronal and glial lesions. We report herein the ability of three natural phenolic derivs. obtained from olives and derived food products to prevent such Tau fibrillization in vitro, namely hydroxytyrosol, oleuropein, and oleuropein aglycon. The latter was found to be more active than the ref. Tau aggregation inhibitor methylene blue on both wild-type and P301L Tau proteins, inhibiting fibrillization at low micromolar concns. These findings might provide further exptl. support for the beneficial nutritional properties of olives and olive oil as well as a chem. scaffold for the development of new drugs aiming at neurodegenerative tauopathies.
- 32Leri, M., Oropesa-Nuñez, R., Canale, C., Raimondi, S., Giorgetti, S., Bruzzone, E., Bellotti, V., Stefani, M., and Bucciantini, M. (2018) Oleuropein Aglycone: A Polyphenol with Different Targets Against Amyloid Toxicity. Biochim. Biophys. Acta, Gen. Subj. 1862 (6), 1432– 1442, DOI: 10.1016/j.bbagen.2018.03.02332https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXmsVKitL4%253D&md5=e3762313d1252ea3bf5f2cd7a8361f18Oleuropein aglycone: A polyphenol with different targets against amyloid toxicityLeri, Manuela; Oropesa-Nunez, Reiner; Canale, Claudio; Raimondi, Sara; Giorgetti, Sofia; Bruzzone, Elena; Bellotti, Vittorio; Stefani, Massimo; Bucciantini, MonicaBiochimica et Biophysica Acta, General Subjects (2018), 1862 (6), 1432-1442CODEN: BBGSB3; ISSN:0304-4165. (Elsevier B.V.)Many data highlight the benefits of the Mediterranean diet and its main lipid component, extra-virgin olive oil (EVOO). EVOO contains many phenolic compds. that have been found effective against several aging- and lifestyle-related diseases, including neurodegeneration. Oleuropein, a phenolic secoiroid glycoside, is the main polyphenol in the olive oil. It has been reported that the aglycon form of Oleuropein (OleA) interferes in vitro and in vivo with amyloid aggregation of a no. of proteins/peptides involved in amyloid, particularly neurodegenerative, diseases avoiding the growth of toxic oligomers and displaying protection against cognitive deterioration. In this study, we carried out a cellular and biophys. study on the relationships between the effects of OleA on the aggregation and cell interactions of the D76N β2-microglobulin (D76N b2m) variant assocd. with a familial form of systemic amyloidosis with progressive bowel dysfunction and extensive visceral amyloid deposits. Our results indicate that OleA protection against D76N b2m cytotoxicity results from (i) a modification of the conformational and biophys. properties of its amyloid fibrils; (ii) a modification of the cell bilayer surface properties of exposed cells. This study reveals that OleA remodels not only D76N b2m aggregates but also the cell membrane interfering with the misfolded proteins-cell membrane assocn., in most cases an early event triggering amyloid-mediated cytotoxicity. The data provided in the present article focus on OleA protection, featuring this polyphenol as a promising plant mol. useful against amyloid diseases.
- 33Rigacci, S., Miceli, C., Nediani, C., Berti, A., Cascella, R., Pantano, D., Nardiello, P., Luccarini, I., Casamenti, F., and Stefani, M. (2015) Oleuropein Aglycone Induces Autophagy via the AMPK/mTOR Signalling Pathway: A Mechanistic Insight. Oncotarget 6 (34), 35344– 35357, DOI: 10.18632/oncotarget.611933https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC28zitlGmuw%253D%253D&md5=4ed55c7e2bd9c10917d22d132d6a7752Oleuropein aglycone induces autophagy via the AMPK/mTOR signalling pathway: a mechanistic insightRigacci Stefania; Miceli Caterina; Nediani Chiara; Berti Andrea; Cascella Roberta; Stefani Massimo; Pantano Daniela; Nardiello Pamela; Luccarini Ilaria; Casamenti FiorellaOncotarget (2015), 6 (34), 35344-57 ISSN:.The healthy effects of plant polyphenols, some of which characterize the so-called Mediterranean diet, have been shown to arise from epigenetic and biological modifications resulting, among others, in autophagy stimulation. Our previous work highlighted the beneficial effects of oleuropein aglycone (OLE), the main polyphenol found in the extra virgin olive oil, against neurodegeneration both in cultured cells and in model organisms, focusing, in particular, autophagy activation. In this study we investigated more in depth the molecular and cellular mechanisms of autophagy induction by OLE using cultured neuroblastoma cells and an OLE-fed mouse model of amylod beta (Aβ) deposition. We found that OLE triggers autophagy in cultured cells through the Ca2+-CAMKKβ-AMPK axis. In particular, in these cells OLE induces a rapid release of Ca2+ from the SR stores which, in turn, activates CAMKKβ, with subsequent phosphorylation and activation of AMPK. The link between AMPK activation and mTOR inhibition was shown in the OLE-fed animal model in which we found that decreased phospho-mTOR immunoreactivity and phosphorylated mTOR substrate p70 S6K levels match enhanced phospho-AMPK levels, supporting the idea that autophagy activation by OLE proceeds through mTOR inhibition. Our results agree with those reported for other plant polyphenols, suggesting a shared molecular mechanism underlying the healthy effects of these substances against ageing, neurodegeneration, cancer, diabetes and other diseases implying autophagy dysfunction.
- 34Grossi, C., Rigacci, S., Ambrosini, S., Ed Dami, T., Luccarini, I., Traini, C., Failli, P., Berti, A., Casamenti, F., and Stefani, M. (2013) The Polyphenol Oleuropein Aglycone Protects TgCRND8Mice against Aß Plaque Pathology. PLoS One 8 (8), e71702 DOI: 10.1371/journal.pone.007170234https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtlamsrfI&md5=0cabef823648e2452389ed35a0320d17The polyphenol oleuropein aglycone protects TgCRND8 mice against Aβ plaque pathologyGrossi, Cristina; Rigacci, Stefania; Ambrosini, Stefano; Ed Dami, Teresa; Luccarini, Ilaria; Traini, Chiara; Failli, Paola; Berti, Andrea; Casamenti, Fiorella; Stefani, MassimoPLoS One (2013), 8 (8), e71702CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)The claimed beneficial effects of the Mediterranean diet include prevention of several age-related dysfunctions including neurodegenerative diseases and Alzheimer-like pathol. These effects have been related to the protection against cognitive decline assocd. with aging and disease by a no. of polyphenols found in red wine and extra virgin olive oil. The double transgenic TgCRND8 mice (overexpressing the Swedish and Indiana mutations in the human amyloid precursor protein), aged 1.5 and 4, and age-matched wild type control mice were used to examine in vivo the effects of 8 wk dietary supplementation of oleuropein aglycon (50 mg/kg of diet), the main polyphenol found in extra virgin olive oil. We report here that dietary supplementation of oleuropein aglycon strongly improves the cognitive performance of young/middle-aged TgCRND8 mice, a model of amyloid-β deposition, respect to age-matched littermates with un-supplemented diet. Immunofluorescence anal. of cerebral tissue in oleuropein aglycon-fed transgenic mice showed remarkably reduced β-amyloid levels and plaque deposits, which appeared less compact and "fluffy"; moreover, microglia migration to the plaques for phagocytosis and a remarkable redn. of the astrocyte reaction were evident. Finally, oleuropein aglycon-fed mice brain displayed an astonishingly intense autophagic reaction, as shown by the increase of autophagic markers expression and of lysosomal activity. Data obtained with cultured cells confirmed the latter evidence, suggesting mTOR regulation by oleuropein aglycon. Our results support, and provide mechanistic insights into, the beneficial effects against Alzheimer-assocd. neurodegeneration of a polyphenol enriched in the extra virgin olive oil, a major component of the Mediterranean diet.
- 35Luccarini, I., Pantano, D., Nardiello, P., Cavone, L., Lapucci, A., Miceli, C., Nediani, C., Berti, A., Stefani, M., and Casamenti, F. (2016) The Polyphenol Oleuropein Aglycone Modulates the PARP1-SIRT1 Interplay: An In Vitro and In Vivo Study. J. Alzheimer's Dis. 54 (2), 737– 750, DOI: 10.3233/JAD-16047135https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsVyisrrN&md5=41ee6b88a89ab8643d3fa9cf6f5a723fThe Polyphenol Oleuropein Aglycone Modulates the PARP1-SIRT1 Interplay: An In Vitro and In Vivo StudyLuccarini, Ilaria; Pantano, Daniela; Nardiello, Pamela; Cavone, Leonardo; Lapucci, Andrea; Miceli, Caterina; Nediani, Chiara; Berti, Andrea; Stefani, Massimo; Casamenti, Fiorella; Cassano, TommasoJournal of Alzheimer's Disease (2016), 54 (2), 737-750CODEN: JADIF9; ISSN:1387-2877. (IOS Press)Poly(ADP-ribose) polymerase-1 (PARP1) activation contributes to the cascade of events initiated by amyloid-β (Aβ) peptide eventually leading to cell death in Alzheimer's disease brain. A significant accumulation of PAR polymers and increase of PARP1 expression were detected in the cortex at the early (3.5 mo) and intermediate (6 mo) stage of Aβ deposition in the TgCRND8 mouse model. Our previous data highlighted the beneficial effects of oleuropein aglycon (OLE), the main polyphenol found in the olive oil, against neurodegeneration both in cultured cells and in model organisms. Here we found that 8-wk OLE treatment (50mg/kg of diet) to 6-mo-old TgCRND8 mice rescued to control values PARP1 activation and the levels of its product, PAR. In N2a neuroblastoma cells, PARP1 activation and PAR formation upon exposure to N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) were abolished by pretreatment for 24h with either OLE (100μM) or PARP inhibitors. A significant redn. of the NAD+ content, compared to controls, was found in N2a cells exposed to MNNG (100μM) for 90min; the latter was slightly attenuated by cell treatment for 24h with PJ-34 or with OLE. In vitro and in vivo, the OLE-induced redn. of PARP1 activation was paralleled by the overexpression of Sirtuin1 (SIRT1), and, in vivo, by a decrease of NF-κB and the pro-apoptotic marker p53. In N2a cells, we also found that OLE potentiates the MNNG-induced increase of Beclin1 levels. In conclusion, our data show that OLE treatment counteracts neuronal damage through modulation of the PARP1-SIRT1 interplay.
- 36LeVine, H. (1995) Thioflavine T Interaction with Amyloid β-sheet Structures. Amyloid 2 (1), 1– 6, DOI: 10.3109/1350612950903188136https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXlsFyhs7g%253D&md5=4e7b589b31c163fd215a025a16664d49Thioflavine T interaction with amyloid β-sheet structuresLeVine, Harry IIIAmyloid (1995), 2 (1), 1-6CODEN: AIJIET; ISSN:1350-6129.Thioflavine T (ThT) interacts in tissue sections with amyloid deposits comprised of a variety of protein species giving a characteristic fluorescent complex. Binding of the dye to amyloid fibrils in suspension generates an amyloid-specific fluorescent signal. This interaction with amyloid fibrils formed from different polypeptides and proteins contg. antiparallel β-pleated sheet secondary structure is selective, occurring strongly with amyloid fibrils formed from insulin, transthyretin, polyglycine(I), Aβ(1-40), and weakly with β2-microglobulin. No fluorescence changes were seen with β-sheet fibrillar poly-L-lysine, islet amyloid peptide (20-29), or poly-L-serine. Native forms of transthyretin, insulin, β2-microglobulin, poly-L-lysine, Aβ(1-40), or several proteins contg. high percentages of β-sheet were also unreactive. The affinity of the amyloid fibrils for ThT varied: (apparent Kd's 0.033 - 10 μM; Amyloid A protein < insulin < ApoAII < polyglycine < transthyretin < Aβ(1-40)). The spectral changes induced by the different amyloid fibrils are qual. the same regardless of the pKd of the interaction with ThT or the identity of the amyloid fibrils, suggesting that the quaternary and secondary, but not the primary structure of the amyloid fibrils are important in forming the amyloid fibril-specific ThT species. Thus, Thioflavine T provides a useful tool for the investigation of amyloidogenesis in a variety of systems.
- 37Pansieri, J., Ostojić, L., Iashchishyn, I. A., Magzoub, M., Wallin, C., Wärmländer, S. K. T. S., Gräslund, A., Nguyen Ngoc, M., Smirnovas, V., Svedružić, Ž. (2019) Pro-Inflammatory S100A9 Protein Aggregation Promoted by NCAM1 Peptide Constructs. ACS Chem. Biol. 14 (7), 1410– 1417, DOI: 10.1021/acschembio.9b0039437https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtV2lsLjM&md5=994b31d58e7e135544a71406e5148697Pro-inflammatory S100A9 protein aggregation promoted by NCAM1 peptide constructsPansieri, Jonathan; Ostojic, Lucija; Iashchishyn, Igor A.; Magzoub, Mazin; Wallin, Cecilia; Waermlaender, Sebastian K. T. S.; Graeslund, Astrid; Nguyen Ngoc, Mai; Smirnovas, Vytautas; Svedruzic, Zeljko; Morozova-Roche, Ludmilla A.ACS Chemical Biology (2019), 14 (7), 1410-1417CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Amyloid cascade and neuroinflammation are hallmarks of neurodegenerative diseases, and pro-inflammatory S100A9 protein is central to both of them. Here, we have shown that NCAM1 peptide constructs carrying polycationic sequences derived from Aβ peptide (KKLVFF) and PrP protein (KKRPKP) significantly promote the S100A9 amyloid self-assembly in a concn.-dependent manner by making transient interactions with individual S100A9 mols., perturbing its native structure and acting as catalysts. Since the individual mol. misfolding is a rate-limiting step in S100A9 amyloid aggregation, the effects of the NCAM1 construct on the native S100A9 are so crit. for its amyloid self-assembly. S100A9 rapid self-assembly into large aggregated clumps may prevent its amyloid tissue propagation, and by modulating S100A9 aggregation as a part of the amyloid cascade, the whole process may be effectively tuned.
- 38Ferrone, F. (1999) Analysis of Protein Aggregation Kinetics. In Amyloid, Prions, and Other Protein Aggregates, Methods in Enzymology, Vol. 309, pp 256– 274, Academic Press, New York. DOI: 10.1016/S0076-6879(99)09019-9 .There is no corresponding record for this reference.
- 39Pansieri, J., Iashchishyn, I. A., Fakhouri, H., Ostojić, L., Malisauskas, M., Musteikyte, G., Smirnovas, V., Schneider, M. M., Scheidt, T., Xu, C. K. (2020) Templating S100A9 Amyloids on Aβ Fibrillar Surfaces Revealed by Charge Detection Mass Spectrometry, Microscopy, Kinetic and Microfluidic Analyses. Chem. Sci. 11 (27), 7031– 7039, DOI: 10.1039/C9SC05905A39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtFOqtrjM&md5=870909e223e6273b417714be73311647Templating S100A9 amyloids on Aβ fibrillar surfaces revealed by charge detection mass spectrometry, microscopy, kinetic and microfluidic analysesPansieri, Jonathan; Iashchishyn, Igor A.; Fakhouri, Hussein; Ostojic, Lucija; Malisauskas, Mantas; Musteikyte, Greta; Smirnovas, Vytautas; Schneider, Matthias M.; Scheidt, Tom; Xu, Catherine K.; Meisl, Georg; Knowles, Tuomas P. J.; Gazit, Ehud; Antoine, Rodolphe; Morozova-Roche, Ludmilla A.Chemical Science (2020), 11 (27), 7031-7039CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)The mechanism of amyloid co-aggregation and its nucleation process are not fully understood in spite of extensive studies. Deciphering the interactions between proinflammatory S100A9 protein and Aβ42 peptide in Alzheimer's disease is fundamental since inflammation plays a central role in the disease onset. Here we use innovative charge detection mass spectrometry (CDMS) together with biophys. techniques to provide mechanistic insight into the co-aggregation process and differentiate amyloid complexes at a single particle level. Combination of mass and charge distributions of amyloids together with reconstruction of the differences between them and detailed microscopy reveals that co-aggregation involves templating of S100A9 fibrils on the surface of Aβ42 amyloids. Kinetic anal. further corroborates that the surfaces available for the Aβ42 secondary nucleation are diminished due to the coating by S100A9 amyloids, while the binding of S100A9 to Aβ42 fibrils is validated by a microfluidic assay. We demonstrate that synergy between CDMS, microscopy, kinetic and microfluidic analyses opens new directions in interdisciplinary research.
- 40Lambert de Malezieu, M., Ferron, S., Sauvager, A., Courtel, P., Ramassamy, C., Tomasi, S., and Abasq, M.-L. (2019) UV-Vis Spectroelectrochemistry of Oleuropein, Tyrosol, and p-Coumaric Acid Individually and in an Equimolar Combination. Differences in LC-ESI-MS2 Profiles of Oxidation Products and Their Neuroprotective Properties. Biomolecules 9 (12), 802, DOI: 10.3390/biom912080240https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXisVOisbzF&md5=bce247f0495156061750c7fcec67e4e5UV-vis spectroelectrochemistry of oleuropein, tyrosol, and p-coumaric acid individually and in an equimolar combination. Differences in LC-ESI-MS2 profiles of oxidation products and their neuroprotective propertiesLambert de Malezieu, Morgane; Ferron, Solenn; Sauvager, Aurelie; Courtel, Patricia; Ramassamy, Charles; Tomasi, Sophie; Abasq, Marie-LaurenceBiomolecules (2019), 9 (12), 802CODEN: BIOMHC; ISSN:2218-273X. (MDPI AG)Major phenolic compds. from olive oil (ArOH-EVOO), oleuropein (Ole), tyrosol (Tyr), and p-coumaric acid (p-Cou), are known for their antioxidant and neuroprotective properties. We previously demonstrated that their combination could potentiate their antioxidant activity in vitro and in cellulo. To further our knowledge of their electron-transfer properties, Ole, Tyr, and p-Cou underwent a spectroelectrochem. study, performed either individually or in equimolar mixts. Two mixts. (Mix and Mix-seq) were prepd. in order to det. whether distinct mols. could arise from their simultaneous or sequential oxidn. The comparison of Liq. Chromatog.-Electrospray Ionization-Tandem Mass Spectrometry (LC-ESI-MS2) profiles highlighted the presence of specific oxidized products found in the mixes. We hypothesized that they derived from the dimerization between Tyr and Ole or p-Cou, which have reacted either in their native or oxidized forms. Moreover, Ole regenerates when the Mix undergoes oxidn. Our study also showed significant neuroprotection by oxidized Ole and oxidized Mix against H2O2 toxicity on SK-N-SH cells, after 24 h of treatment with very low concns. (1 and 5 nM). This suggests the putative relevant role of oxidized Ole products to protect or delay neuronal death.
- 41Chang, C. C., Khan, I., Tsai, K. L., Li, H., Yang, L. W., Chou, R. H., and Yu, C. (2016) Blocking the Interaction between S100A9 and RAGE V Domain using CHAPS Molecule: A novel route to Drug Development Against Cell Proliferation. Biochim. Biophys. Acta, Proteins Proteomics 1864 (11), 1558– 69, DOI: 10.1016/j.bbapap.2016.08.00841https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtleitrrJ&md5=edf7f6363b5d98376e5294ef68a8fe16Blocking the interaction between S100A9 and RAGE V domain using CHAPS molecule: A novel route to drug development against cell proliferationChang, Chin-Chi; Khan, Imran; Tsai, Kun-Lin; Li, Hongchun; Yang, Lee-Wei; Chou, Ruey-Hwang; Yu, ChinBiochimica et Biophysica Acta, Proteins and Proteomics (2016), 1864 (11), 1558-1569CODEN: BBAPBW; ISSN:1570-9639. (Elsevier B.V.)Human S100A9 (Calgranulin B) is a Ca2+-binding protein, from the S100 family, that often presents as a homodimer in myeloid cells. It becomes an important mediator during inflammation once calcium binds to its EF-hand motifs. Human RAGE protein (receptor for advanced glycation end products) is one of the target-proteins. RAGE binds to a hydrophobic surface on S100A9. Interactions between these proteins trigger signal transduction cascades, promoting cell growth, proliferation, and tumorigenesis. Here, we present the soln. structure of mutant S100A9 (C3S) homodimer, detd. by multi-dimensional NMR expts. We further characterize the soln. interactions between mS100A9 and the RAGE V domain via NMR spectroscopy. CHAPS is a zwitterionic and non-denaturing mol. widely used for protein solubilizing and stabilization. We found out that CHAPS and RAGE V domain would interact with mS100A9 by using 1H-15N HSQC NMR titrns. Therefore, using the HADDOCK program, we superimpose two binary complex models mS100A9-RAGE V domain and mS100A9-CHAPS and demonstrate that CHAPS mols. could play a crucial role in blocking the interaction between mS100A9 and the RAGE V domain. WST-1 assay results also support the conclusion that CHAPS inhibits the bioactivity of mS100A9. This report will help to inform new drug development against cell proliferation.
- 42Ferrer, I., Blanco, R., Carmona, M., Puig, B., Ribera, R., Rey, M. J., and Ribalta, T. (2001) Prion Protein Expression in Senile Plaques in Alzheimer’s Disease. Acta Neuropathol. 101 (1), 49– 56, DOI: 10.1007/s00401000027142https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXhtValurs%253D&md5=ad313fb97a3424f0f9f87f3af05b2e20Prion protein expression in senile plaques in Alzheimer's diseaseFerrer, I.; Blanco, R.; Carmona, M.; Puig, B.; Ribera, R.; Rey, M. J.; Ribalta, T.Acta Neuropathologica (2001), 101 (1), 49-56CODEN: ANPTAL; ISSN:0001-6322. (Springer-Verlag)Prion protein (PrPC) is a glycolipid-anchored cell membrane sialoglycoprotein that localizes in presynaptic membranes. Since synapses are vulnerable to Alzheimer's disease (AD), the present study examines PrPC expression in senile plaques, one of the major structural abnormalities in AD, by single- and double-labeling immunohistochem. Punctate PrPC immunoreactivity is found in diffuse plaques, whereas isolated large coarse PrPC-pos. granules reminiscent of dystrophic neurites are obsd. in neuritic plaques. Finally, PrPC deposition also occurs as dense filamentous and amorphous ppts. in amyloid cores of senile plaques, but not in the walls of blood vessels with amyloid angiopathy. In contrast to PrPC, βA4-amyloid immunoreactivity is preserved and even enhanced following incubation of the tissue sections with proteinase K prior to immunohistochem., thus indicating no PrPC and βA4-amyloid cross-reactivity in dense amyloid cores of senile plaques. Punctate PrPC deposition in diffuse plaques is similar to that of synaptophysin, a synaptic vesicle-assocd. protein, as already reported in other studies. Immunopptn., electrophoresis and Western blot studies have shown that synaptophysin, amyloid precursor protein (APP) and βA4 do not co-ppt. with PrP. These results suggest that synaptophysin, APP and βA4 are likely not bound to PrP. PrPC accumulation in βA4-amyloid dense cores may be the consequence of the release of PrP into the extracellular space. Whether PrPC accumulation in the extracellular space is the result of impaired endocytosis and subsequent hydrolysis in the endosomal compartment, in contrast to normal degrdn. of PrPC, resulting from or occurring in parallel to abnormal APP degrdn., deserves further study.
- 43Vogl, T., Eisenblätter, M., Völler, T., Zenker, S., Hermann, S., van Lent, P., Faust, A., Geyer, C., Petersen, B., Roebrock, K. (2014) Alarmin S100A8/S100A9 as a Biomarker for Molecular Imaging of Local Inflammatory Activity. Nat. Commun. 5 (1), 4593, DOI: 10.1038/ncomms559343https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitVaksLfN&md5=12a921b769ee55ecc080d53474332119Alarmin S100A8/S100A9 as a biomarker for molecular imaging of local inflammatory activityVogl, Thomas; Eisenblaetter, Michel; Voeller, Tom; Zenker, Stefanie; Hermann, Sven; van Lent, Peter; Faust, Andreas; Geyer, Christiane; Petersen, Beatrix; Roebrock, Kirsten; Schaefers, Michael; Bremer, Christoph; Roth, JohannesNature Communications (2014), 5 (), 4593CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)Inflammation has a key role in the pathogenesis of various human diseases. The early detection, localization and monitoring of inflammation are crucial for tailoring individual therapies. However, reliable biomarkers to detect local inflammatory activities and to predict disease outcome are still missing. Alarmins, which are locally released during cellular stress, are early amplifiers of inflammation. Here, using optical mol. imaging, we demonstrate that the alarmin S100A8/S100A9 serves as a sensitive local and systemic marker for the detection of even sub-clin. disease activity in inflammatory and immunol. processes like irritative and allergic contact dermatitis. In a model of collagen-induced arthritis, we use S100A8/S100A9 imaging to predict the development of disease activity. Furthermore, S100A8/S100A9 can act as a very early and sensitive biomarker in exptl. leishmaniasis for phagocyte activation linked to an effective Th1-response. In conclusion, the alarmin S100A8/S100A9 is a valuable and sensitive mol. target for novel imaging approaches to monitor clin. relevant inflammatory disorders on a mol. level.
- 44Bucciantini, M., Calloni, G., Chiti, F., Formigli, L., Nosi, D., Dobson, C. M., and Stefani, M. (2004) Prefibrillar Amyloid Protein Aggregates Share Common Features of Cytotoxicity. J. Biol. Chem. 279 (30), 31374– 31382, DOI: 10.1074/jbc.M40034820044https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXlslOrs7g%253D&md5=f0ab3f11b00cc2b8b23acb99069545e6Prefibrillar Amyloid Protein Aggregates Share Common Features of CytotoxicityBucciantini, Monica; Calloni, Giulia; Chiti, Fabrizio; Formigli, Lucia; Nosi, Daniele; Dobson, Christopher M.; Stefani, MassimoJournal of Biological Chemistry (2004), 279 (30), 31374-31382CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)The intracellular free Ca2+ concn. and redox status of murine fibroblasts exposed to prefibrillar aggregates of the HypF N-terminal domain have been investigated in vitro and in vivo using a range of fluorescent probes. Aggregate entrance into the cytoplasm is followed by an early rise of reactive oxygen species and free Ca2+ levels and eventually by cell death. Such changes correlate directly with the viability of the cells and are not obsd. when cell are cultured in the presence of reducing agents or in Ca2+-free media. In addn., moderate cell stress following exposure to the aggregates was found to be fully reversible. The results show that the cytotoxicity of prefibrillar aggregates of HypF-N, a protein not assocd. with clin. disease, has the same fundamental origin as that produced by similar types of aggregates of proteins linked with specific amyloidoses. These findings suggest that misfolded proteinaceous aggregates stimulate generic cellular responses as a result of the exposure of regions of the structure (such as hydrophobic residues and the polypeptide main chain) that are buried in the normally folded proteins. They also support the idea that a higher no. of degenerative pathologies than previously known might be considered as protein deposition diseases.
- 45Novitskaya, V., Bocharova, O. V., Bronstein, I., and Baskakov, I. V. (2006) Amyloid Fibrils of Mammalian Prion Protein Are Highly Toxic to Cultured Cells and Primary Neurons. J. Biol. Chem. 281 (19), 13828– 13836, DOI: 10.1074/jbc.M51117420045https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xkt1ymurY%253D&md5=7dd9e8722bc1997f4d260444248ddd58Amyloid Fibrils of Mammalian Prion Protein Are Highly Toxic to Cultured Cells and Primary NeuronsNovitskaya, Vera; Bocharova, Olga V.; Bronstein, Igor; Baskakov, Ilia V.Journal of Biological Chemistry (2006), 281 (19), 13828-13836CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)A growing body of evidence indicates that small, sol. oligomeric species generated from a variety of proteins and peptides rather than mature amyloid fibrils are inherently highly cytotoxic. Here, we show for the first time that mature amyloid fibrils produced from full-length recombinant mammalian prion protein (rPrP) were highly toxic to cultured cells and primary hippocampal and cerebellar neurons. Fibrils induced apoptotic cell death in a time- and dose-dependent manner. The toxic effect of fibrils was comparable with that exhibited by sol. small β-oligomers generated from the same protein. Fibrils prepd. from insulin were not toxic, suggesting that the toxic effect was not solely due to the highly polymeric nature of the fibrillar form. The cell death caused by rPrP fibrils or β-oligomers was substantially reduced when expression of endogenous PrPC was down-regulated by small interfering RNAs. In opposition to the β-oligomer and amyloid fibrils of rPrP, the monomeric α-helical form of rPrP stimulated neurite out-growth and survival of neurons. These studies illustrated that both sol. β-oligomer and amyloid fibrils of the prion protein are intrinsically toxic and confirmed that endogenously expressed PrPC is required for mediating the toxicity of abnormally folded external PrP aggregates.
- 46Canale, C., Oropesa-Nuñez, R., Diaspro, A., and Dante, S. (2018) Amyloid and Membrane Complexity: The Toxic Interplay Revealed by AFM. Semin. Cell Dev. Biol. 73, 82– 94, DOI: 10.1016/j.semcdb.2017.08.04646https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsFSmsL3J&md5=912b964f5577766e290127a998df02e5Amyloid and membrane complexity: The toxic interplay revealed by AFMCanale, Claudio; Oropesa-Nunez, Reinier; Diaspro, Alberto; Dante, SilviaSeminars in Cell & Developmental Biology (2018), 73 (), 82-94CODEN: SCDBFX; ISSN:1084-9521. (Elsevier Ltd.)A review. Lipid membranes play a fundamental role in the pathol. development of protein misfolding diseases. Several pieces of evidence suggest that the lipid membrane could act as a catalytic surface for protein aggregation. Furthermore, a leading theory indicates the interaction between the cell membrane and misfolded oligomer species as the responsible for cytotoxicity, hence, for neurodegeneration in disorders such as Alzheimer's and Parkinson's disease. The definition of the mechanisms that drive the interaction between pathol. protein aggregates and plasma membrane is fundamental for the development of effective therapies for a large class of diseases. Atomic force microscopy (AFM) has been employed to study how amyloid aggregates affect the cell physiol. properties. Considerable efforts were spent to characterize the interaction with model systems, i.e., planar supported lipid bilayers, but some works also addressed the problem directly on living cells. Here, an overview of the main works involving the use of the AFM on both model system and living cells will be provided. Different kind of approaches will be presented, as well as the main results derived from the AFM anal.
- 47Walsh, P., Vanderlee, G., Yau, J., Campeau, J., Sim, V. L., Yip, C. M., and Sharpe, S. (2014) The Mechanism of Membrane Disruption by Cytotoxic Amyloid Oligomers Formed by Prion Protein (106–126) is Dependent on Bilayer Composition. J. Biol. Chem. 289 (15), 10419– 10430, DOI: 10.1074/jbc.M113.51586647https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXmtlCnsbo%253D&md5=6e3cdf9ca126d6309eff637155f6ada0The Mechanism of Membrane Disruption by Cytotoxic Amyloid Oligomers Formed by Prion Protein(106-126) Is Dependent on Bilayer CompositionWalsh, Patrick; Vanderlee, Gillian; Yau, Jason; Campeau, Jody; Sim, Valerie L.; Yip, Christopher M.; Sharpe, SimonJournal of Biological Chemistry (2014), 289 (15), 10419-10430CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)The formation of fibrillar aggregates has long been assocd. with neurodegenerative disorders such as Alzheimer and Parkinson diseases. Although fibrils are still considered important to the pathol. of these disorders, it is now widely understood that smaller amyloid oligomers are the toxic entities along the misfolding pathway. One characteristic shared by the majority of amyloid oligomers is the ability to disrupt membranes, a commonality proposed to be responsible for their toxicity, although the mechanisms linking this to cell death are poorly understood. Here, we describe the phys. basis for the cytotoxicity of oligomers formed by the prion protein (PrP)-derived amyloid peptide PrP(106-126). We show that oligomers of this peptide kill several mammalian cells lines, as well as mouse cerebellar organotypic cultures, and we also show that they exhibit antimicrobial activity. Phys. perturbation of model membranes mimicking bacterial or mammalian cells was investigated using at. force microscopy, polarized total internal reflection fluorescence microscopy, and NMR spectroscopy. Disruption of anionic membranes proceeds through a carpet or detergent model as proposed for other antimicrobial peptides. By contrast, when added to zwitterionic membranes contg. cholesterol-rich ordered domains, PrP(106-126) oligomers induce a loss of domain sepn. and decreased membrane disorder. Loss of raft-like domains may lead to activation of apoptotic pathways, resulting in cell death. This work sheds new light on the phys. mechanisms of amyloid cytotoxicity and is the first to clearly show membrane type-specific modes of action for a cytotoxic peptide.
- 48Leri, M., Bemporad, F., Oropesa-Nuñez, R., Canale, C., Calamai, M., Nosi, D., Ramazzotti, M., Giorgetti, S., Pavone, F. S., Bellotti, V. (2016) Molecular Insights into Cell Toxicity of a Novel Familial Amyloidogenic Variant of β2-microglobulin. J. Cell. Mol. Med. 20 (8), 1443– 1456, DOI: 10.1111/jcmm.1283348https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xht1SmtL%252FI&md5=569e255a9a3564a7f0a53904b708a902Molecular insights into cell toxicity of a novel familial amyloidogenic variant of β2-microglobulinLeri, Manuela; Bemporad, Francesco; Oropesa-Nunez, Reinier; Canale, Claudio; Calamai, Martino; Nosi, Daniele; Ramazzotti, Matteo; Giorgetti, Sofia; Pavone, Francesco S.; Bellotti, Vittorio; Stefani, Massimo; Bucciantini, MonicaJournal of Cellular and Molecular Medicine (2016), 20 (8), 1443-1456CODEN: JCMMC9; ISSN:1582-4934. (Wiley-Blackwell)The first genetic variant of β2-microglobulin (b2M) assocd. with a familial form of systemic amyloidosis has been recently described. The mutated protein, carrying a substitution of Asp at position 76 with an Asn (D76N b2M), exhibits a strongly enhanced amyloidogenic tendency to aggregate with respect to the wild-type protein. In this study, we characterized the D76N b2M aggregation path and performed an unprecedented anal. of the biochem. mechanisms underlying aggregate cytotoxicity. We showed that, contrarily to what expected from other amyloid studies, early aggregates of the mutant are not the most toxic species, despite their higher surface hydrophobicity. By modulating ganglioside GM1 content in cell membrane or synthetic lipid bilayers, we confirmed the pivotal role of this lipid as aggregate recruiter favoring their cytotoxicity. We finally obsd. that the aggregates bind to the cell membrane inducing an alteration of its elasticity (with possible functional unbalance and cytotoxicity) in GM1-enriched domains only, thus establishing a link between aggregate-membrane contact and cell damage.
- 49Cicerale, S., Conlan, X. A., Sinclair, A. J., and Keast, R. S. J. (2008) Chemistry and Health of Olive Oil Phenolics. Crit. Rev. Food Sci. Nutr. 49 (3), 218– 236, DOI: 10.1080/10408390701856223There is no corresponding record for this reference.
- 50Bucciantini, M., Nosi, D., Forzan, M., Russo, E., Calamai, M., Pieri, L., Formigli, L., Quercioli, F., Soria, S., Pavone, F. (2012) Toxic Effects of Amyloid Fibrils on Cell Membranes: The Importance of Ganglioside GM1. FASEB J. 26 (2), 818– 831, DOI: 10.1096/fj.11-18938150https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XitFOnt7o%253D&md5=3e235091b37469527df80656e9941281Toxic effects of amyloid fibrils on cell membranes: the importance of ganglioside GM1Bucciantini, Monica; Nosi, Daniele; Forzan, Mario; Russo, Edda; Calamai, Martino; Pieri, Laura; Formigli, Lucia; Quercioli, Franco; Soria, Silvia; Pavone, Francesco; Savistchenko, Jimmy; Melki, Ronald; Stefani, MassimoFASEB Journal (2012), 26 (2), 818-831, 10.1096/fj.11-189381CODEN: FAJOEC; ISSN:0892-6638. (Federation of American Societies for Experimental Biology)The interaction of amyloid aggregates with the cell plasma membrane is currently considered among the basic mechanisms of neuronal dysfunction in amyloid neurodegeneration. We used amyloid oligomers and fibrils grown from the yeast prion Sup35p, responsible for the specific prion trait [PSI+], to investigate how membrane lipids modulate fibril interaction with the membranes of cultured H-END cells and cytotoxicity. Sup35p shares no homol. with endogenous mammalian polypeptide chains. Thus, the generic toxicity of amyloids and the mol. events underlying cell degeneration can be investigated without interference with analogous polypeptides encoded by the cell genome. Sup35 fibrils bound to the cell membrane without increasing its permeability to Ca2+. Fibril binding resulted in structural reorganization and aggregation of membrane rafts, with GM1 clustering and alteration of its mobility. Sup35 fibril binding was affected by GM1 or its sialic acid moiety, but not by cholesterol membrane content, with complete inhibition after treatment with fumonisin B1 or neuraminidase. Finally, cell impairment resulted from caspase-8 activation after Fas receptor translocation on fibril binding to the plasma membrane. Our observations suggest that amyloid fibrils induce abnormal accumulation and overstabilization of raft domains in the cell membrane and provide a reasonable, although not unique, mechanistic and mol. explanation for fibril toxicity.
- 51Calamai, M. and Pavone, F. S. (2013) Partitioning and Confinement of GM1 Ganglioside induced by Amyloid Aggregates. FEBS Lett. 587 (9), 1385– 1391, DOI: 10.1016/j.febslet.2013.03.01451https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXltFCktL0%253D&md5=8a6120f6badbd0c18f63fd5ec3a1a42ePartitioning and confinement of GM1 ganglioside induced by amyloid aggregatesCalamai, Martino; Pavone, Francesco S.FEBS Letters (2013), 587 (9), 1385-1391CODEN: FEBLAL; ISSN:0014-5793. (Elsevier B.V.)Growing evidence shows that GM1 ganglioside is involved in amyloid deposition and toxicity. By means of real-time single particle tracking, we show that amyloid oligomers and aggregates formed by Aβ1-42 and amylin, two peptides assocd., resp., with the development of Alzheimer's disease and type II diabetes, interact with GM1 and decrease dramatically its lateral diffusion on the plasma membrane of living neuroblastoma cells. The confinement of GM1, a constituent of membrane rafts involved in neuroprotection, at the level of both types of amyloid aggregates can interfere with cell signaling pathways and contribute to the loss of neuroprotection.
- 52Pellistri, F., Bucciantini, M., Relini, A., Nosi, D., Gliozzi, A., Robello, M., and Stefani, M. (2008) Nonspecific Interaction of Prefibrillar Amyloid Aggregates with Glutamatergic Receptors Results in Ca2+ Increase in Primary Neuronal Cells. J. Biol. Chem. 283 (44), 29950– 29960, DOI: 10.1074/jbc.M80399220052https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXht1ynt7bM&md5=3450bdbfbcc79be0a3bd6efcfa9cb12cNonspecific Interaction of Prefibrillar Amyloid Aggregates with Glutamatergic Receptors Results in Ca2+ Increase in Primary Neuronal CellsPellistri, Francesca; Bucciantini, Monica; Relini, Annalisa; Nosi, Daniele; Gliozzi, Alessandra; Robello, Mauro; Stefani, MassimoJournal of Biological Chemistry (2008), 283 (44), 29950-29960CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)It is widely reported that the Ca2+ increase following nonspecific cell membrane permeabilization is among the earliest biochem. modifications in cells exposed to toxic amyloid aggregates. However, more recently receptors with Ca2+ channel activity such as α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA), N-Me D-aspartate (NMDA), ryanodine, and inositol 1,4,5-trisphosphate receptors have been proposed as mediators of the Ca2+ increase in neuronal cells challenged with β-amyloid peptides. We previously showed that prefibrillar aggregates of proteins not assocd. with amyloid diseases are toxic to exposed cells similarly to comparable aggregates of disease-assocd. proteins. In particular, prefibrillar aggregates of the prokaryotic HypF-N were shown to be toxic to different cultured cell lines by eliciting Ca2+ and reactive oxygen species increases. This study was aimed at assessing whether NMDA and AMPA receptor activations could be considered a generic feature of cell interaction with amyloid aggregates rather than a specific effect of some aggregated protein. Therefore, we investigated whether NMDA and AMPA receptors were involved in the Ca2+ increase following exposure of rat cerebellar granule cells to HypF-N prefibrillar aggregates. We found that the intracellular Ca2+ increase was assocd. with the early activation of NMDA and AMPA receptors, although some nonspecific membrane permeabilization was also obsd. at longer times of exposure. This result matched a significant co-localization of the aggregates with both receptors on the plasma membrane. Our data support the possibility that glutamatergic channels are generic sites of interaction with the cell membrane of prefibrillar aggregates of different peptides and proteins as well as the key structures responsible for the resulting early membrane permeabilization to Ca2+.
- 53Salazar, S. V. and Strittmatter, S. M. (2017) Cellular Prion Protein as a Receptor for Amyloid-β Oligomers in Alzheimer’s Disease. Biochem. Biophys. Res. Commun. 483 (4), 1143– 1147, DOI: 10.1016/j.bbrc.2016.09.06253https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFCrsbfO&md5=ca66bd363a8321200220fab42dd8a539Cellular prion protein as a receptor for amyloid-β oligomers in Alzheimer's diseaseSalazar, Santiago V.; Strittmatter, Stephen M.Biochemical and Biophysical Research Communications (2017), 483 (4), 1143-1147CODEN: BBRCA9; ISSN:0006-291X. (Elsevier B.V.)Sol. oligomers of amyloid-beta (Aβo) are implicated by biochem. and genetic evidence as a trigger for Alzheimer's disease (AD) pathophysiol. A key step is Aβo interaction with the neuronal surface to initiate a cascade of altered signal transduction leading to synaptic dysfunction and damage. This review discusses neuronal cell surface mols. with high affinity selectively for oligomeric disease-assocd. states of Aβ, with a particular focus on the role of cellular prion protein (PrPC) in this process. Addnl. receptors may contribute to mediation of Aβo action, but PrPC appears to play a primary role in a no. of systems. The specificity of binding, the genetic necessity in mouse models of disease and downstream signaling pathways are considered. Signal transduction downstream of Aβo complexes with PrPC involves metabotropic glutamate receptor 5 (mGluR5), Fyn kinase and Pyk2 kinase, with deleterious effects on synaptic transmission and maintenance. Current data support the hypothesis that a substantial portion of Aβo toxicity in AD is mediated after initial interaction with PrPC on the neuronal surface. As such, the interaction of Aβo with PrPC is a potential therapeutic intervention site for AD.
- 54Haas, L. T. and Strittmatter, S. M. (2016) Oligomers of Amyloid β Prevent Physiological Activation of the Cellular Prion Protein-Metabotropic Glutamate Receptor 5 Complex by Glutamate in Alzheimer Disease. J. Biol. Chem. 291 (33), 17112– 17121, DOI: 10.1074/jbc.M116.72066454https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslaktL3P&md5=b338b288603f6edc0d4b6ed1f615df82Oligomers of amyloid β prevent physiological activation of the cellular prion protein-metabotropic glutamate receptor 5 complex by glutamate in Alzheimer diseaseHaas, Laura T.; Strittmatter, Stephen M.Journal of Biological Chemistry (2016), 291 (33), 17112-17121CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)The dysfunction and loss of synapses in Alzheimer disease are central to dementia symptoms. We have recently demonstrated that pathol. Amyloid β oligomer (Aβo) regulates the assocn. between intracellular protein mediators and the synaptic receptor complex composed of cellular prion protein (PrPC) and metabotropic glutamate receptor 5 (mGluR5). Here we sought to det. whether Aβo alters the physiol. signaling of the PrPC-mGluR5 complex upon glutamate activation. We provide evidence that acute exposure to Aβo as well as chronic expression of familial Alzheimer disease mutant transgenes in model mice prevents protein-protein interaction changes of the complex induced by the glutamate analog 3,5-dihydroxyphenylglycine. We further show that 3,5-dihydroxyphenylglycine triggers the phosphorylation and activation of protein tyrosine kinase 2-β (PTK2B, also referred to as Pyk2) and of calcium/calmodulin-dependent protein kinase II in wild-type brain slices but not in Alzheimer disease transgenic brain slices or wild-type slices incubated with Aβo. This study further distinguishes two sep. Aβo-dependent signaling cascades, one dependent on extracellular Ca2+ and Fyn kinase activation and the other dependent on the release of Ca2+ from intracellular stores. Thus, Aβo triggers multiple distinct PrPC-mGluR5-dependent events implicated in neurodegeneration and dementia. We propose that targeting the PrPC-mGluR5 complex will reverse aberrant Aβo-triggered states of the complex to allow physiol. fluctuations of glutamate signaling.
- 55Abedini, A., Cao, P., Plesner, A., Zhang, J., He, M., Derk, J., Patil, S. A., Rosario, R., Lonier, J., Song, F. (2018) RAGE Binds Preamyloid IAPP Intermediates and Mediates Pancreatic β Cell Proteotoxicity. J. Clin. Invest. 128 (2), 682– 698, DOI: 10.1172/JCI8521055https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MvhtVKntA%253D%253D&md5=e8bdc41817d48f49d22e4f4b3695b129RAGE binds preamyloid IAPP intermediates and mediates pancreatic β cell proteotoxicityAbedini Andisheh; Zhang Jinghua; He Meilun; Derk Julia; Patil Sachi A; Rosario Rosa; Lonier Jacqueline; Song Fei; Schmidt Ann Marie; Cao Ping; Raleigh Daniel P; Plesner Annette; Koh Hyunwook; Li HuilinThe Journal of clinical investigation (2018), 128 (2), 682-698 ISSN:.Islet amyloidosis is characterized by the aberrant accumulation of islet amyloid polypeptide (IAPP) in pancreatic islets, resulting in β cell toxicity, which exacerbates type 2 diabetes and islet transplant failure. It is not fully clear how IAPP induces cellular stress or how IAPP-induced toxicity can be prevented or treated. We recently defined the properties of toxic IAPP species. Here, we have identified a receptor-mediated mechanism of islet amyloidosis-induced proteotoxicity. In human diabetic pancreas and in cellular and mouse models of islet amyloidosis, increased expression of the receptor for advanced glycation endproducts (RAGE) correlated with human IAPP-induced (h-IAPP-induced) β cell and islet inflammation, toxicity, and apoptosis. RAGE selectively bound toxic intermediates, but not nontoxic forms of h-IAPP, including amyloid fibrils. The isolated extracellular ligand-binding domains of soluble RAGE (sRAGE) blocked both h-IAPP toxicity and amyloid formation. Inhibition of the interaction between h-IAPP and RAGE by sRAGE, RAGE-blocking antibodies, or genetic RAGE deletion protected pancreatic islets, β cells, and smooth muscle cells from h-IAPP-induced inflammation and metabolic dysfunction. sRAGE-treated h-IAPP Tg mice were protected from amyloid deposition, loss of β cell area, β cell inflammation, stress, apoptosis, and glucose intolerance. These findings establish RAGE as a mediator of IAPP-induced toxicity and suggest that targeting the IAPP/RAGE axis is a potential strategy to mitigate this source of β cell dysfunction in metabolic disease.
- 56Vogl, T., Leukert, N., Barczyk, K., Strupat, K., and Roth, J. (2006) Biophysical Characterization of S100A8 and S100A9 in the Absence and Presence of Bivalent Cations. Biochim. Biophys. Acta, Mol. Cell Res. 1763 (11), 1298– 1306, DOI: 10.1016/j.bbamcr.2006.08.02856https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xht1CnsLbK&md5=f4bc53c5e2577488e442d1034458f067Biophysical characterization of S100A8 and S100A9 in the absence and presence of bivalent cationsVogl, Thomas; Leukert, Nadja; Barczyk, Katarzyna; Strupat, Kerstin; Roth, JohannesBiochimica et Biophysica Acta, Molecular Cell Research (2006), 1763 (11), 1298-1306CODEN: BBAMCO; ISSN:0167-4889. (Elsevier Ltd.)S100A8 and S100A9 are two proinflammatory mols. belonging to the S100 family of calcium-binding proteins. Common to all S100 proteins, S100A8 and S100A9 form non-covalently assocd. complexes which have been shown to exhibit different functional properties. Besides dimerization, recent research is focused on the importance of higher oligomeric structures of S100 proteins induced by bivalent cations. While S100A8/S100A9-heterodimers are formed in the absence of calcium, tetramerization is strictly calcium-dependent. Heterodimer formation is not a simple process and our biophys. analyses (FRET, ESI-MS) demonstrate that simply mixing both subunits is not sufficient to induce complex formation. Steps of denaturation/renaturation are necessary for the recombinant complex to show identical biophys. properties as S100A8/S100A9 obtained from granulocytes. In addn. to calcium, both proteins are able to bind zinc with high affinity. Here, we demonstrate for the first time by different biophys. methods (MALDI-MS, ESI-MS, fluorescence spectroscopy) that zinc-binding, like calcium, induces (S100A8/S100A9)2-tetramers. Using mass spectrometric investigations, we demonstrate that zinc triggers the formation of (S100A8/S100A9)2-tetramers by zinc-specific binding sites rather than by interactions with calcium-specific EF-hands. The zinc-induced tetramer is structurally very similar to the calcium-induced tetramer. Thus, like calcium, zinc acts as a regulatory factor in S100A8/S100A9-dependent signaling pathways.
- 57Wang, C., Iashchishyn, I. A., Kara, J., Foderà, V., Vetri, V., Sancataldo, G., Marklund, M., and Morozova-Roche, L. A. (2019) Proinflammatory and Amyloidogenic S100A9 Induced by Traumatic Brain Injury in Mouse Model. Neurosci. Lett. 699, 199– 205, DOI: 10.1016/j.neulet.2019.02.01257https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXjtFOkurY%253D&md5=00ce57ce6592952a11a2f6a928d0cd73Proinflammatory and amyloidogenic S100A9 induced by traumatic brain injury in mouse modelWang, Chao; Iashchishyn, Igor A.; Kara, John; Fodera, Vito; Vetri, Valeria; Sancataldo, Giuseppe; Marklund, Niklas; Morozova-Roche, Ludmilla A.Neuroscience Letters (2019), 699 (), 199-205CODEN: NELED5; ISSN:0304-3940. (Elsevier Ireland Ltd.)Traumatic brain injury (TBI) represents a significant risk factor for development of neurodegenerative diseases such as Alzheimer's and Parkinson's. The S100A9-driven amyloid-neuroinflammatory cascade occurring during primary and secondary TBI events can serve as a mechanistic link between TBI and Alzheimer's as demonstrated recently in the human brain tissues. Here by using immunohistochem. in the controlled cortical impact TBI mouse model we have found pro-inflammatory S100A9 in the brain tissues of all mice on the first and third post-TBI days, while 70% of mice did not show any S100A9 presence on seventh post-TBI day similar to controls. This indicates that defensive mechanisms effectively cleared S100A9 in these mouse brain tissues during post-TBI recovery. By using sequential immunohistochem. we have shown that S100A9 was produced by both neuronal and microglial cells. However, Aβ peptide deposits characteristic for Alzheimer's disease were not detected in any post-TBI animals. On the first and third post-TBI days S100A9 was found to aggregate intracellularly into amyloid oligomers, similar to what was previously obsd. in human TBI tissues. Complementary, by using Rayleigh scatting, intrinsic fluorescence and at. force microscopy we demonstrated that in vitro S100A9 self-assembles into amyloid oligomers within minutes. Its amyloid aggregation is highly dependent on changes of environmental conditions such as variation of calcium levels, pH, temp. and redn./oxidn., which might be relevant to perturbation of cellular and tissues homeostasis under TBI. Present results demonstrate that S100A9 induction mechanisms in TBI are similar in mice and humans, emphasizing that S100A9 is an important marker of brain injury and therefore can be a potential therapeutic target.
- 58Kayed, R., Canto, I., Breydo, L., Rasool, S., Lukacsovich, T., Wu, J., Albay, R., Pensalfini, A., Yeung, S., Head, E. (2010) Conformation Dependent Monoclonal Antibodies Distinguish Different Replicating Strains or Conformers of Prefibrillar Aβ Oligomers. Mol. Neurodegener. 5 (1), 57, DOI: 10.1186/1750-1326-5-5758https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhs1ams7fE&md5=54b7c4aca24f41e66a682f2a78efc8e0Conformation dependent monoclonal antibodies distinguish different replicating strains or conformers of prefibrillar Aβ oligomersKayed, Rakez; Canto, Isabel; Breydo, Leonid; Rasool, Suhail; Lukacsovich, Tamas; Wu, Jessica; Albay, Ricardo, III; Pensalfini, Anna; Yeung, Stephen; Head, Elizabeth; Marsh, J. Lawrence; Glabe, CharlesMolecular Neurodegeneration (2010), 5 (), 57CODEN: MNOEAZ; ISSN:1750-1326. (BioMed Central Ltd.)Background: Age-related neurodegenerative diseases share a no. of important pathol. features, such as accumulation of misfolded proteins as amyloid oligomers and fibrils. Recent evidence suggests that sol. amyloid oligomers and not the insol. amyloid fibrils may represent the primary pathol. species of protein aggregates. Results: We have produced several monoclonal antibodies that specifically recognize prefibrillar oligomers and do not recognize amyloid fibrils, monomer or natively folded proteins. Like the polyclonal antisera, the individual monoclonals recognize generic epitopes that do not depend on a specific linear amino acid sequence, but they display distinct preferences for different subsets of prefibrillar oligomers. Immunol. anal. of a no. of different prefibrillar Aβ oligomer prepns. show that structural polymorphisms exist in Aβ prefibrillar oligomers that can be distinguished on the basis of their reactivity with monoclonal antibodies. Western blot anal. demonstrates that the conformers defined by the monoclonal antibodies have distinct size distributions, indicating that oligomer structure varies with size. The different conformational types of Aβ prefibrillar oligomers can serve as they serve as templates for monomer addn., indicating that they seed the conversion of Aβ monomer into more prefibrillar oligomers of the same type. Conclusions: These results indicate that distinct structural variants or conformers of prefibrillar Aβ oligomers exist that are capable of seeding their own replication. These conformers may be analogous to different strains of prions.
- 59Eilers, P. H. C. (2003) A Perfect Smoother. Anal. Chem. 75 (14), 3631– 3636, DOI: 10.1021/ac034173t59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXktFGqsLw%253D&md5=eba4aed2ebbf3e48bd80049d8e0ce165A Perfect SmootherEilers, Paul H. C.Analytical Chemistry (2003), 75 (14), 3631-3636CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)The A. Savitzky and M. Golay (1964) filter has several disadvantages. A very attractive alternative is a smoother based on penalized least squares, extending ideas presented by E. T. Whittaker (1923). This smoother is extremely fast, gives continuous control over smoothness, interpolates automatically, and allows fast leave-one-out cross-validation. It can be programmed in a few lines of Matlab code. Theory, implementation, and applications are presented.
- 60Wasylewski, Z., Koloczek, H., and Wasniowska, A. (1988) Fluorescence-Quenching-Resolved Spectroscopy of Proteins. Eur. J. Biochem. 172 (3), 719– 724, DOI: 10.1111/j.1432-1033.1988.tb13948.x60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1cXhsVKmt70%253D&md5=bb0cf515871a8a98d9b958cd066dde9eFluorescence-quenching-resolved spectroscopy of proteinsWasylewski, Zygmunt; Koloczek, Henryk; Wasniowska, AlicjaEuropean Journal of Biochemistry (1988), 172 (3), 719-24CODEN: EJBCAI; ISSN:0014-2956.A new procedure is described for using fluorescence-quenching data of tryptophan residues in proteins to resolve their fluorescence emission spectra. In this concept, the Stern-Volmer quenching plot is detd. at each particular emission wavelength and iterative nonlinear least-squares fitting procedure allowed to resolve the steady-state emission spectra into components. The resolved components, attributed to each of tryptophan residue, can be characterized by different accessibility to the quencher. The ability to resolve fluorescence emission spectra can be improved by using different kinds of efficient quenchers, which can selectively quench the emission of exposed or both exposed and buried fluorophores. The method was used to decomp. emission fluorescence spectra in 2-tryptophan-contg. proteins: horse liver dehydrogenase, sperm whale apomyoglobin, and metalloprotease from Staphylococcus aureus. The resolved spectra of alc. dehydrogenase and metalloprotease are in excellent agreement with those previously obtained by single-photon counting or phase methods. The method presented here is tech. simple and does not require expensive instrumentation.
- 61Pettersen, E. F., Goddard, T. D., Huang, C. C., Couch, G. S., Greenblatt, D. M., Meng, E. C., and Ferrin, T. E. (2004) UCSF Chimera-AVisualization Sytem for Exploratory Research and Analysis Vesrion. J. Comput. Chem. 25, 1605– 1612, DOI: 10.1002/jcc.2008461https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXmvVOhsbs%253D&md5=944b175f440c1ff323705987cf937ee7UCSF Chimera-A visualization system for exploratory research and analysisPettersen, Eric F.; Goddard, Thomas D.; Huang, Conrad C.; Couch, Gregory S.; Greenblatt, Daniel M.; Meng, Elaine C.; Ferrin, Thomas E.Journal of Computational Chemistry (2004), 25 (13), 1605-1612CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)The design, implementation, and capabilities of an extensible visualization system, UCSF Chimera, are discussed. Chimera is segmented into a core that provides basic services and visualization, and extensions that provide most higher level functionality. This architecture ensures that the extension mechanism satisfies the demands of outside developers who wish to incorporate new features. Two unusual extensions are presented: Multiscale, which adds the ability to visualize large-scale mol. assemblies such as viral coats, and Collab., which allows researchers to share a Chimera session interactively despite being at sep. locales. Other extensions include Multalign Viewer, for showing multiple sequence alignments and assocd. structures; ViewDock, for screening docked ligand orientations; Movie, for replaying mol. dynamics trajectories; and Vol. Viewer, for display and anal. of volumetric data. A discussion of the usage of Chimera in real-world situations is given, along with anticipated future directions. Chimera includes full user documentation, is free to academic and nonprofit users, and is available for Microsoft Windows, Linux, Apple Mac OS X, SGI IRIX, and HP Tru64 Unix from http://www.cgl.ucsf.edu/chimera/.
- 62Miletić, V., Odorčić, I., Nikolić, P., and Svedružić, Ž. M. (2017) In Silico Design of the First DNA-Independent Mechanism-Based Inhibitor of Mammalian DNA Methyltransferase Dnmt1. PLoS One 12, e0174410 DOI: 10.1371/journal.pone.017441062https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsValsrbL&md5=e52a25b05645fb00f338b89d0193afa6In silico design of the first DNA-independent mechanism-based inhibitor of mammalian DNA methyltransferase Dnmt1Miletic, Vedran; Odorcic, Ivica; Nikolic, Patrik; Svedruzic, Zeljko M.PLoS One (2017), 12 (4), e0174410/1-e0174410/21CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)We use our earlier exptl. studies of the catalytic mechanism of DNA methyltransferases to prep. in silico a family of novel mechanism-based inhibitors of human Dnmt1. Highly specific inhibitors of DNA methylation can be used for anal. of human epigenome and for the creation of iPS cells. We describe a set of adenosyl-1-methyl-pyrimidin-2-one derivs. as novel mechanism based inhibitors of mammalian DNA methyltransferase Dnmt1. The inhibitors have been designed to bind simultaneously in the active site and the cofactor site and thus act as transition- state analogs. Mol. dynamics studies showed that the lead compd. can form between 6 to 9 binding interactions with Dnmt1. QM/MM anal. showed that the upon binding to Dnmt1 the inhibitor can form a covalent adduct with active site Cys1226 and thus act as a mechanism-based suicide-inhibitor. The inhibitor can target DNA-bond and DNAfree form of Dnmt1, however the suicide-inhibition step is more likely to happen when DNA is bound to Dnmt1. The validity of presented anal. is described in detail using 69 modifications in the lead compd. structure. In total 18 of the presented 69 modifications can be used to prep. a family of highly specific inhibitors that can differentiate even between closely related enzymes such as Dnmt1 and Dnmt3a DNA methyltransferases. Presented results can be used for prepn. of some highly specific and potent inhibitors of mammalian DNA methylation with specific pharmacol. properties.
- 63Ruiz-Carmona, S., Alvarez-Garcia, D., Foloppe, N., Garmendia-Doval, A. B., Juhos, S., Schmidtke, P., Barril, X., Hubbard, R. E., and Morley, S. D. (2014) rDock: a Fast, Versatile and Open Source Program for Docking Ligands to Proteins and Nucleic Acids. PLoS Comput. Biol. 10, e1003571 DOI: 10.1371/journal.pcbi.100357163https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhsVGlsL%252FO&md5=e4cb786d6567fdc7f2a46f64955a9992rDock: a fast, versatile and open source program for docking ligands to proteins and nucleic acidsRuiz-Carmona, Sergio; Alvarez-Garcia, Daniel; Foloppe, Nicolas; Garmendia-Doval, A. Beatriz; Juhos, Szilveszter; Schmidtke, Peter; Barril, Xavier; Hubbard, Roderick E.; Morley, S. DavidPLoS Computational Biology (2014), 10 (4), e1003571/1-e1003571/7, 7 pp.CODEN: PCBLBG; ISSN:1553-7358. (Public Library of Science)Identification of chem. compds. with specific biol. activities is an important step in both chem. biol. and drug discovery. When the structure of the intended target is available, one approach is to use mol. docking programs to assess the chem. complementarity of small mols. with the target; such calcns. provide a qual. measure of affinity that can be used in virtual screening (VS) to rank order a list of compds. according to their potential to be active. rDock is a mol. docking program developed at Vernalis for high-throughput VS (HTVS) applications. Evolved from RiboDock, the program can be used against proteins and nucleic acids, is designed to be computationally very efficient and allows the user to incorporate addnl. constraints and information as a bias to guide docking. This article provides an overview of the program structure and features and compares rDock to two ref. programs, AutoDock Vina (open source) and Schrodinger's Glide (com.). In terms of computational speed for VS, rDock is faster than Vina and comparable to Glide. For binding mode prediction, rDock and Vina are superior to Glide. The VS performance of rDock is significantly better than Vina, but inferior to Glide for most systems unless pharmacophore constraints are used; in that case rDock and Glide are of equal performance. The program is released under the Lesser General Public License and is freely available for download, together with the manuals, example files and the complete test sets, at online.
- 64Lee, J., Patel, D. S., Ståhle, J., Park, S. J., Kern, N. R., Kim, S., Lee, J., Cheng, X., Valvano, M. A., Holst, O. (2019) CHARMM-GUI Membrane Builder for Complex Biological Membrane Simulations with Glycolipids and Lipoglycans. J. Chem. Theory Comput. 15, 775– 786, DOI: 10.1021/acs.jctc.8b0106664https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisVOitrbO&md5=a212a0bfce1818bc74f2b317eff3af93CHARMM-GUI Membrane Builder for Complex Biological Membrane Simulations with Glycolipids and LipoglycansLee, Jumin; Patel, Dhilon S.; Stahle, Jonas; Park, Sang-Jun; Kern, Nathan R.; Kim, Seonghoon; Lee, Joonseong; Cheng, Xi; Valvano, Miguel A.; Holst, Otto; Knirel, Yuriy A.; Qi, Yifei; Jo, Sunhwan; Klauda, Jeffery B.; Widmalm, Goran; Im, WonpilJournal of Chemical Theory and Computation (2019), 15 (1), 775-786CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Glycolipids (such as glycoglycerolipids, glycosphingolipids, and glycosylphosphatidylinositol) and lipoglycans (such as lipopolysaccharides (LPS), lipooligosaccharides (LOS), mycobacterial lipoarabinomannan, and mycoplasma lipoglycans) are typically found on the surface of cell membranes and play crucial roles in various cellular functions. Characterizing their structure and dynamics at the mol. level is essential to understand their biol. roles, but systematic generation of glycolipid and lipoglycan structures is challenging because of great variations in lipid structures and glycan sequences (i.e., carbohydrate types and their linkages). To facilitate the generation of all-atom glycolipid/LPS/LOS structures, we have developed Glycolipid Modeler and LPS Modeler in CHARMM-GUI (http://www.charmm-gui.org), a web-based interface that simplifies building of complex biol. simulation systems. In addn., we have incorporated these modules into Membrane Builder so that users can readily build a complex sym. or asym. biol. membrane system with various glycolipids and LPS/LOS. These tools are expected to be useful in innovative and novel glycolipid/LPS/LOS modeling and simulation research by easing tedious and intricate steps in modeling complex biol. systems and shall provide insight into structures, dynamics, and underlying mechanisms of complex glycolipid-/LPS-/LOS-contg. biol. membrane systems.
- 65Sousa da Silva, A. W and Vranken, W. F (2012) ACPYPE-Antechamber Python Parser Interface. BMC Res. Notes 5, 367, DOI: 10.1186/1756-0500-5-36765https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC38fitlWjtg%253D%253D&md5=60f750ba98c975374b351a39a1fa0ec4ACPYPE - AnteChamber PYthon Parser interfacESousa da Silva Alan W; Vranken Wim FBMC research notes (2012), 5 (), 367 ISSN:.BACKGROUND: ACPYPE (or AnteChamber PYthon Parser interfacE) is a wrapper script around the ANTECHAMBER software that simplifies the generation of small molecule topologies and parameters for a variety of molecular dynamics programmes like GROMACS, CHARMM and CNS. It is written in the Python programming language and was developed as a tool for interfacing with other Python based applications such as the CCPN software suite (for NMR data analysis) and ARIA (for structure calculations from NMR data). ACPYPE is open source code, under GNU GPL v3, and is available as a stand-alone application at http://www.ccpn.ac.uk/acpype and as a web portal application at http://webapps.ccpn.ac.uk/acpype. FINDINGS: We verified the topologies generated by ACPYPE in three ways: by comparing with default AMBER topologies for standard amino acids; by generating and verifying topologies for a large set of ligands from the PDB; and by recalculating the structures for 5 protein-ligand complexes from the PDB. CONCLUSIONS: ACPYPE is a tool that simplifies the automatic generation of topology and parameters in different formats for different molecular mechanics programmes, including calculation of partial charges, while being object oriented for integration with other applications.
- 66Kim, S., Lee, J., Jo, S., Brooks, C. L., 3rd, Lee, H. S., and Im, W. (2017) CHARMM-GUI Ligand Reader and Modeler for CHARMM Force Field Generation of Small Molecules. J. Comput. Chem. 38, 1879– 1886, DOI: 10.1002/jcc.2482966https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXnslKhtLk%253D&md5=3e872e011783f98bc14594d91bc142efCHARMM-GUI ligand reader and modeler for CHARMM force field generation of small moleculesKim, Seonghoon; Lee, Jumin; Jo, Sunhwan; Brooks, Charles L., III; Lee, Hui Sun; Im, WonpilJournal of Computational Chemistry (2017), 38 (21), 1879-1886CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)Reading ligand structures into any simulation program is often nontrivial and time consuming, esp. when the force field parameters and/or structure files of the corresponding mols. are not available. To address this problem, we have developed Ligand Reader & Modeler in CHARMM-GUI. Users can upload ligand structure information in various forms (using PDB ID, ligand ID, SMILES, MOL/MOL2/SDF file, or PDB/mmCIF file), and the uploaded structure is displayed on a sketchpad for verification and further modification. Based on the displayed structure, Ligand Reader & Modeler generates the ligand force field parameters and necessary structure files by searching for the ligand in the CHARMM force field library or using the CHARMM general force field (CGenFF). In addn., users can define chem. substitution sites and draw substituents in each site on the sketchpad to generate a set of combinatorial structure files and corresponding force field parameters for throughput or alchem. free energy simulations. Finally, the output from Ligand Reader & Modeler can be used in other CHARMM-GUI modules to build a protein-ligand simulation system for all supported simulation programs, such as CHARMM, NAMD, GROMACS, AMBER, GENESIS, LAMMPS, Desmond, OpenMM, and CHARMM/OpenMM. Ligand Reader & Modeler is available as a functional module of CHARMM-GUI at http://www.charmm-gui.org/input/ligandrm.
- 67Van Der Spoel, D., Lindahl, E., Hess, B., Groenhof, G., Mark, A. E., and Berendsen, H. J. (2005) GROMACS: Fast, Flexible, and Free. J. Comput. Chem. 26, 1701– 1718, DOI: 10.1002/jcc.2029167https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXht1SlsbbO&md5=4aa71b1dc0b5b4978a88b0568245a265GROMACS: Fast, flexible, and freeVan Der Spoel, David; Lindahl, Erik; Hess, Berk; Groenhof, Gerrit; Mark, Alan E.; Berendsen, Herman J. C.Journal of Computational Chemistry (2005), 26 (16), 1701-1718CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)This article describes the software suite GROMACS (Groningen MAchine for Chem. Simulation) that was developed at the University of Groningen, The Netherlands, in the early 1990s. The software, written in ANSI C, originates from a parallel hardware project, and is well suited for parallelization on processor clusters. By careful optimization of neighbor searching and of inner loop performance, GROMACS is a very fast program for mol. dynamics simulation. It does not have a force field of its own, but is compatible with GROMOS, OPLS, AMBER, and ENCAD force fields. In addn., it can handle polarizable shell models and flexible constraints. The program is versatile, as force routines can be added by the user, tabulated functions can be specified, and analyses can be easily customized. Nonequil. dynamics and free energy detns. are incorporated. Interfaces with popular quantum-chem. packages (MOPAC, GAMES-UK, GAUSSIAN) are provided to perform mixed MM/QM simulations. The package includes about 100 utility and anal. programs. GROMACS is in the public domain and distributed (with source code and documentation) under the GNU General Public License. It is maintained by a group of developers from the Universities of Groningen, Uppsala, and Stockholm, and the Max Planck Institute for Polymer Research in Mainz. Its Web site is http://www.gromacs.org.
- 68Cheung, Y. T., Lau, W. K., Yu, M. S., Lai, C. S., Yeung, S. C., So, K. F., and Chang, R. C. (2009) Effects of All-Trans-Retinoic Acid on Human SH-SY5Y Neuroblastoma as In Vitro Model in Neurotoxicity Research. NeuroToxicology 30 (1), 127– 135, DOI: 10.1016/j.neuro.2008.11.00168https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtFeqsbw%253D&md5=c0fd9e12f367cc8258ce304212571828Effects of all-trans-retinoic acid on human SH-SY5Y neuroblastoma as in vitro model in neurotoxicity researchCheung, Yuen-Ting; Lau, Way Kwok-Wai; Yu, Man-Shan; Lai, Cora Sau-Wan; Yeung, Sze-Chun; So, Kwok-Fai; Chang, Raymond Chuen-ChungNeuroToxicology (2009), 30 (1), 127-135CODEN: NRTXDN; ISSN:0161-813X. (Elsevier B.V.)Human neuroblastoma SH-SY5Y is a dopaminergic neuronal cell line which has been used as an in vitro model for neurotoxicity expts. Although the neuroblastoma is usually differentiated by all-trans-retinoic acid (RA), both RA-differentiated and undifferentiated SH-SY5Y cells have been used in neuroscience research. However, the changes in neuronal properties triggered by RA as well as the subsequent responsiveness to neurotoxins have not been comprehensively studied. Therefore, we aim to re-evaluate the differentiation property of RA on this cell line. We hypothesize that modulation of signaling pathways and neuronal properties during RA-mediated differentiation in SH-SY5Y cells can affect their susceptibility to neurotoxins. The differentiation property of RA was confirmed by showing an extensive outgrowth of neurites, increased expressions of neuronal nuclei, neuron specific enolase, synaptophysin and synaptic assocd. protein-97, and decreased expression of inhibitor of differentiation-1. While undifferentiated SH-SY5Y cells were susceptible to 6-OHDA and MPP+, RA-differentiation conferred SH-SY5Y cells higher tolerance, potentially by up-regulating survival signaling, including Akt pathway as inhibition of Akt removed RA-induced neuroprotection against 6-OHDA. As a result, the real toxicity cannot be revealed in RA-differentiated cells. Therefore, undifferentiated SH-SY5Y is more appropriate for studying neurotoxicity or neuroprotection in exptl. Parkinson's disease research.
- 69Nosi, D., Mercatelli, R., Chellini, F., Soria, S., Pini, A., Formigli, L., and Quercioli, F. (2013) A Molecular Imaging Analysis of Cx43 Association with Cdo During Skeletal Myoblast Differentiation. J. Biophotonics 6 (8), 612– 621, DOI: 10.1002/jbio.20120006369https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXht1amu77P&md5=cd23bd3959355f60143523005ad14929A molecular imaging analysis of Cx43 association with Cdo during skeletal myoblast differentiationNosi, Daniele; Mercatelli, Raffaella; Chellini, Flaminia; Soria, Silvia; Pini, Alessandro; Formigli, Lucia; Quercioli, FrancoJournal of Biophotonics (2013), 6 (8), 612-621CODEN: JBOIBX; ISSN:1864-063X. (Wiley-VCH Verlag GmbH & Co. KGaA)Cell-to-cell contacts are crucial for cell differentiation. The promyogenic cell surface protein, Cdo, functions as a component of multiprotein clusters to mediate cell adhesion signaling. Connexin 43, the main connexin forming gap junctions, also plays a key role in myogenesis. At least part of its effects is independent of the intercellular channel function, but the mechanisms underlying are unknown. Here, using multiple optical approaches, we provided the first evidence that Cx43 phys. interacts with Cdo to form dynamic complexes during myoblast differentiation, offering clues for considering this interaction a structural basis of the channel-independent function of Cx43.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acschemneuro.0c00828.
Time dependences of ThT fluorescence during OleA incubation; interaction of S100A9 amyloids with A11 amyloid oligomer specific antibodies upon increasing OleA concentration; cell viability and intracellular free Ca2+ level in SH-SY5Y cells affected by S100A9 amyloids; preformed S100A9 fibrils treated with OleA during various times; cytotoxicity of S100A9 fibrils to undifferentiated and differentiated SH-SY5Y cells (PDF)
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.