Mapping and Profiling Lipid Distribution in a 3D Model of Breast Cancer ProgressionClick to copy article linkArticle link copied!
- Netta VidavskyNetta VidavskyDepartment of Materials Science and Engineering, Cornell University, Ithaca, New York 14850, United StatesMore by Netta Vidavsky
- Jennie A. M. R. KunitakeJennie A. M. R. KunitakeDepartment of Materials Science and Engineering, Cornell University, Ithaca, New York 14850, United StatesMore by Jennie A. M. R. Kunitake
- Maria Elena Diaz-RubioMaria Elena Diaz-RubioMetabolomics Facility, Institute of Biotechnology, Cornell University, Ithaca, New York 14850, United StatesMore by Maria Elena Diaz-Rubio
- Aaron E. ChiouAaron E. ChiouNancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14850, United StatesMore by Aaron E. Chiou
- Hyun-Chae LohHyun-Chae LohDepartment of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United StatesMore by Hyun-Chae Loh
- Sheng ZhangSheng ZhangMetabolomics Facility, Institute of Biotechnology, Cornell University, Ithaca, New York 14850, United StatesMore by Sheng Zhang
- Admir MasicAdmir MasicDepartment of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United StatesMore by Admir Masic
- Claudia FischbachClaudia FischbachNancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14850, United StatesKavli Institute at Cornell for Nanoscale Science, Ithaca, New York 14850, United StatesMore by Claudia Fischbach
- Lara A. Estroff*Lara A. Estroff*E-mail: [email protected] (L.A.E.).Department of Materials Science and Engineering, Cornell University, Ithaca, New York 14850, United StatesKavli Institute at Cornell for Nanoscale Science, Ithaca, New York 14850, United StatesMore by Lara A. Estroff
Abstract
Aberrant lipid accumulation and marked changes in cellular lipid profiles are related to breast cancer metabolism and disease progression. In vitro, these phenomena are primarily studied using cells cultured in monolayers (2D). Here, we employ multicellular spheroids, generated using the MCF10A cell line series of increasing malignancy potential, to better recapitulate the 3D microenvironmental conditions that cells experience in vivo. Breast cancer cell lipid compositions were assessed in 2D and 3D culture models as a function of malignancy using liquid chromatography coupled with mass spectrometry. Further, the spatial distribution of lipids was examined using Raman chemical imaging and lipid staining. We show that with changes in the cellular microenvironment when moving from 2D to 3D cell cultures, total lipid amounts decrease significantly, while the ratio of acylglycerols to membrane lipids increases. This ratio increase could be associated with the formation of large lipid droplets (>10 μm) that are spatially evident throughout the spheroids but absent in 2D cultures. Additionally, we found a significant difference in lipid profiles between the more and less malignant spheroids, including changes that support de novo sphingolipid production and a reduction in ether-linked lipid fractions in the invasive spheroids. These differences in lipid profiles as a function of cell malignancy and microenvironment highlight the importance of coupled spatial and lipidomic studies to better understand the connections between lipid metabolism and cancer.
Synopsis
The profiles and spatial distribution of lipids in breast cancer cells change as a function of culture dimensionality and malignancy potential, yielding insight into cancer-related lipid metabolism.
Introduction
Results
Spatial Distribution of Lipids Depends on the Dimensionality (2D vs 3D) and Malignancy Potential of the Cells
Figure 1
Figure 1. Relation between lipid amounts and the malignancy potential of breast cancer cells in 2D and 3D. Oil-Red-O lipid staining of cell lines from the MCF10A-based breast cancer progression series cultured in 2D (a–f) and as multicellular spheroids (g–l). (a, d, g, j) Nonmalignant MCF10A cells; (b, e, h, k) precancer MCF10DCIS.com cells; (c, f, i, l) invasive MCF10CA1a cells. Lipids are stained red, and cell nuclei are stained purple. Arrows: lipid droplet aggregates close to cell nuclei. For a high resolution version of Figure 1 see Supporting Data.
Lipid Profiling of Precancer and Invasive Cells in 2D and 3D Culture
Lipid Amounts and Profiles Differ Significantly Depending on Dimensionality
Figure 2
Figure 2. Lipid profiles in 2D and 3D culture of precancer and invasive cells detected using LCMS. (a) Heatmap showing the clustering of lipid species in 2D and 3D cultures of MCF10DCIS.com (precancer) and MCF10CA1a cells (invasive). Color bar indicates the scaled distance from the row mean of the normalized transformed data. For assessment of the variation within each group, the biological replicates for each condition are shown. For the 2D samples, each group consists of three biological replicates, and for the 3D samples, each group consists of four biological replicates. Lipid classes are color coded as indicated. Coenzyme Q10 is shown in white. To the right, representative example lipid structures of the color-coded lipid classes in part a are shown. (b) Lipid class distribution in precancer and invasive 2D and 3D cultures as detected with LCMS, calculated from areas in LCMS normalized per microgram of protein in the sample and presented as a fraction of total lipids in the sample. Error bars are the standard error of the mean. 34 Cs = summed acyl chain and sphingoid base chain lengths of 34 carbons. See Supporting Figure S2 for a version of this figure with individual lipid identifiers associated with the heat map.
Neutral Glycerolipids Are Significantly Increased in 3D Compared to 2D
Alkyl and Alkenyl Ether Lipid Fractions Are Significantly Increased in 3D
Sphingomyelin and Lysophosphatidylcholine Show Differences between 2D and 3D Based on Number of Carbons
Lipid Profiles Differ between Precancer and Invasive in 3D Spheroids
Figure 3
Figure 3. Heat map showing clustering of the 25 lipid species with the most significant changes, selected by t-test, across 3D cultures of MCF10DCIS.com (precancer) and MCF10CA1a (invasive) cells as detected with LCMS. Each group consists of four biological replicates. So(d18:0) = sphinganine. Color bar indicates the scaled distance from the row mean of the normalized transformed data.
Mapping Spatial Distribution of Lipids and Other ECM Components in 3D Spheroids Using Raman Microscopy
Figure 4
Figure 4. Characterization of the lipid droplets formed in 3D multicellular spheroids of malignant cell lines. (a, b, e) Precancer spheroid cross sections. (c, d, g) Invasive spheroid cross sections. Asterisks show the necrotic core. (a, c) Oil-Red-O lipid staining showing lipid accumulation in the necrotic core area of the spheroids as well as lipid droplets in the invasive spheroid (arrows). (b, d) Cross-polarized light images of the spheroid cross sections showing birefringence (arrows in part d point at the same areas as in part c). (e, g) Overlaid spatial distribution maps of key basis spectra resultant from NMF multivariate analysis of Raman mapping. (f) Corresponding NMF basis spectra (precancer: black, invasive: gray). The precancer overlay map (e) shows an increase in lipid content toward the spheroid center, while the invasive overlay map confirms the birefringent regions are lipid-rich droplets (cyan). Arrows indicate the same bodies as in parts c and d.
Raman Mapping Localizes Multiple Signatures: Lipids, Proteins, Cells, Glycogen, and Cytochrome c
Raman Mapping Shows That Lipid Droplets Are Consistent with Unsaturated Acylglycerols
Raman Mapping of a Clinical Sample Shows Spatially Distinct Distribution of Lipids within DCIS
Figure 5
Figure 5. Spatial characterization of DCIS (precancer) from human tissue. (a) H&E stained cryo-section showing the duct cross section containing cells (purple), necrosis (dark pink and purple), and surrounding stromal tissue (pink). (b) Confocal Raman mapping and component analysis of a serial section from the same duct in part a, showing the spatial distribution of the tissue components. Lipids, cyan; collagen, green; protein/cytochrome c, magenta; noncollagenous proteins, blue; cells, yellow. A distinct lipid signature consistent with neutral lipids, including cholesterol ester(s), is observed in the necrotic regions and occurs in spatially discrete domains (arrow). (c) The corresponding Raman component spectra for the Raman map in part b.
Discussion
Figure 6
Figure 6. A scheme describing the effects of dimensionality (top left) and the cell malignancy potential (bottom left) on lipid species amounts in the MCF10A breast cancer in vitro model. Lipid-rich areas (red) are observed in the necrotic core with decreasing amounts toward the spheroid periphery alongside large lipid droplets at the periphery. The observed increase in the ratio of neutral to membrane lipid species in 3D versus 2D may be related to large lipid droplet formation through TG synthesis and/or lipid droplet coalescence (top right). The observed increase in sphingolipid species in invasive versus precancer spheroids could be due to enhancement of the de novo sphingolipid synthesis pathway (bottom right, adapted from Ogretmen (46)). Species of lipids increased out of total lipids in 3D invasive spheroids are underlined. CoA = coenzyme A, SPT = serine palmitoyltransferase, KDSR = 3-ketosphinganine reductase, CERS = (dihydro)ceramide synthases, DES = dihydroceramide desaturase, SMS = sphingomyelin synthase, CDase = ceramidases.
Cell Culture Dimensionality Affects Lipid Production and Distribution
Lipid Composition and Distribution in Multicellular Spheroids May Relate to Energy Storage in Nutrient Deprived Conditions
Sphingolipid de Novo Synthesis Is Upregulated in Invasive Cells
Ether Lipid Fractions Depend on Both Malignancy and Dimensionality
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acscentsci.8b00932.
Methods, LCMS and lipid staining data, Raman analysis information, NMF and NMF validations (PDF)
High Resolution Figure 1 and Metaboanalyst Download (ZIP)
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 Drs. Clifford A. Hudis, Neil M. Iyengar, and Monica Morrow of the Memorial Sloan Kettering Cancer Center for providing the human sample and Dr. Daniel Sudilovsky of the Department of Pathology and Laboratory Medicine, Cayuga Medical Center at Ithaca for his pathological expertise. We thank Dr. Lynn M. Johnson from Cornell Statistical Consulting Unit for her help with statistical analysis, Prof. Elizabeth Johnson for lipid pathways discussion, Janille Maragh for assistance with Raman setup, and Dr. Minjee Kang and Dr. Neta Varsano for helpful discussions. We thank the Animal Health Diagnostic Center for help with staining, the Cornell College of Veterinary Medicine for the use of Scanscope, the Bonassar lab for the use of cross-polarized microscope, and the Schaffer-Nishimura lab for use of the cryotome. We thank WITec and Tavis Ezell for kindly allowing access to the WITec Suite FIVE software, Project FIVE Plus. Research reported in this publication was supported by the Center on the Physics of Cancer Metabolism through Award 1-U54-CA210184 and by the Human Science Frontiers Program (RGP0016/2017). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. This work made use of the Cornell Center for Materials Research Shared Facilities, which are supported through the NSF MRSEC program (DMR-1719875). Additional imaging data were acquired through the Cornell University Biotechnology Resource Center with NIH 1-S10-OD012287 funding. N.V. acknowledges funding from The Israeli Council for Higher Education and Ben-Gurion University. A.E.C. acknowledges funding from the National Institutes of Health through Award F31-CA228448.
MAG | monoacylglycerol |
DAG | diacylglycerol |
TG | triradylglcerol (includes triacylglycerol, alkyldiacylglycerol, and alkenyldiacylglycerol) |
So | sphingosine/sphinganine |
Cer | ceramide (also includes dihydroceramide, hexosylceramide, and dihexosylceramide) |
SM | sphingomyelin |
PE | Phosphatidylethanolamine |
PC | Phosphatidylcholine |
(O−) | alkyl ether |
(P-) | alkenyl ether |
PI | Phosphatidylinositol |
PS | Phosphatidylserine |
PG | Phosphatidylglycerol |
LPC | Lysophosphatidylcholine (also includes alkyl ether LPCs, alkenyl ether LPCs) |
CoQ | coenzyme Q10 |
References
This article references 77 other publications.
- 1DeBerardinis, R. J.; Chandel, N. S. Fundamentals of Cancer Metabolism. Sci. Adv. 2016, 2 (5), e1600200– e1600200, DOI: 10.1126/sciadv.1600200Google ScholarThere is no corresponding record for this reference.
- 2Furuta, E.; Pai, S. K.; Zhan, R.; Bandyopadhyay, S.; Watabe, M.; Mo, Y.-Y.; Hirota, S.; Hosobe, S.; Tsukada, T.; Miura, K. Fatty Acid Synthase Gene Is Up-Regulated by Hypoxia via Activation of Akt and Sterol Regulatory Element Binding Protein-1. Cancer Res. 2008, 68 (4), 1003– 1011, DOI: 10.1158/0008-5472.CAN-07-2489Google ScholarThere is no corresponding record for this reference.
- 3Koizume, S.; Miyagi, Y. Lipid Droplets : A Key Cellular Organelle Associated with Cancer Cell Survival under Normoxia and Hypoxia. Int. J. Mol. Sci. 2016, 17, 1– 23, DOI: 10.3390/ijms17091430Google ScholarThere is no corresponding record for this reference.
- 4Kamphorst, J. J.; Cross, J. R.; Fan, J.; de Stanchina, E.; Mathew, R.; White, E. P.; Thompson, C. B.; Rabinowitz, J. D. Hypoxic and Ras-Transformed Cells Support Growth by Scavenging Unsaturated Fatty Acids from Lysophospholipids. Proc. Natl. Acad. Sci. U. S. A. 2013, 110 (22), 8882– 8887, DOI: 10.1073/pnas.1307237110Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFalsbnN&md5=4deaa2c9fa2b5c2b482cfef5cabfb5eaHypoxic and Ras-transformed cells support growth by scavenging unsaturated fatty acids from lysophospholipidsKamphorst, Jurre J.; Cross, Justin R.; Fan, Jing; de Stanchina, Elisa; Mathew, Robin; White, Eileen P.; Thompson, Craig B.; Rabinowitz, Joshua D.Proceedings of the National Academy of Sciences of the United States of America (2013), 110 (22), 8882-8887, S8882/1-S8882/5CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Cancer cell growth requires fatty acids to replicate cellular membranes. The kinase Akt is known to up-regulate fatty acid synthesis and desatn., which is carried out by the oxygen-consuming enzyme stearoyl-CoA desaturase (SCD)1. We used 13C tracers and lipidomics to probe fatty acid metab., including desatn., as a function of oncogene expression and oxygen availability. During hypoxia, flux from glucose to acetyl-CoA decreases, and the fractional contribution of glutamine to fatty acid synthesis increases. In addn., we find that hypoxic cells bypass de novo lipogenesis, and thus, both the need for acetyl-CoA and the oxygen-dependent SCD1-reaction, by scavenging serum fatty acids. The preferred substrates for scavenging are phospholipids with one fatty acid tail (lysophospholipids). Hypoxic reprogramming of de novo lipogenesis can be reproduced in normoxic cells by Ras activation. This renders Ras-driven cells, both in culture and in allografts, resistant to SCD1 inhibition. Thus, a mechanism by which oncogenic Ras confers metabolic robustness is through lipid scavenging.
- 5Counihan, J. L.; Grossman, E. A.; Nomura, D. K. Cancer Metabolism: Current Understanding and Therapies. Chem. Rev. 2018, 118 (14), 6893– 6923, DOI: 10.1021/acs.chemrev.7b00775Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtFyisrvL&md5=4239fa36e326abc1abbe5269522aeeb0Cancer Metabolism: Current Understanding and TherapiesCounihan, Jessica L.; Grossman, Elizabeth A.; Nomura, Daniel K.Chemical Reviews (Washington, DC, United States) (2018), 118 (14), 6893-6923CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review. Dysregulation of cancer cell metab. contributes to abnormal cell growth, the biol. endpoint of cancer. We review here numerous affected oncogenes and metabolic pathways common in cancer, and how they contribute to cancer pathogenesis and malignancy. This review also discusses various pharmacol. manipulations that take advantage of these metabolic abnormalities, and the current targeted therapies that have arisen from this research.
- 6Santos, C. R.; Schulze, A. Lipid Metabolism in Cancer. FEBS J. 2012, 279 (15), 2610– 2623, DOI: 10.1111/j.1742-4658.2012.08644.xGoogle Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtVWhurvI&md5=53fa5afbd3de338e22986d6869a60769Lipid metabolism in cancerSantos, Claudio R.; Schulze, AlmutFEBS Journal (2012), 279 (15), 2610-2623CODEN: FJEOAC; ISSN:1742-464X. (Wiley-Blackwell)A review. Lipids form a diverse group of water-insol. mols. that include triacylglycerides, phosphoglycerides, sterols and sphingolipids. They play several important roles at cellular and organismal levels. Fatty acids are the major building blocks for the synthesis of triacylglycerides, which are mainly used for energy storage. Phosphoglycerides, together with sterols and sphingolipids, represent the major structural components of biol. membranes. Lipids can also have important roles in signaling, functioning as second messengers and as hormones. There is increasing evidence that cancer cells show specific alterations in different aspects of lipid metab. These alterations can affect the availability of structural lipids for the synthesis of membranes, the synthesis and degrdn. of lipids that contribute to energy homeostasis and the abundance of lipids with signaling functions. Changes in lipid metab. can affect numerous cellular processes, including cell growth, proliferation, differentiation and motility. This review will examine some of the alterations in lipid metab. that have been reported in cancer, at both cellular and organismal levels, and discuss how they contribute to different aspects of tumorigenesis.
- 7Schulze, A.; Harris, A. L. How Cancer Metabolism Is Tuned for Proliferation and Vulnerable to Disruption. Nature 2012, 491 (7424), 364– 373, DOI: 10.1038/nature11706Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhs1Ogt77F&md5=73cdad5f0a8652993ff162b9b1ba6fe7How cancer metabolism is tuned for proliferation and vulnerable to disruptionSchulze, Almut; Harris, Adrian L.Nature (London, United Kingdom) (2012), 491 (7424), 364-373CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)A review. Cancer metab. has received a substantial amt. of interest over the past decade. The advances in anal. tools have, along with the rapid progress of cancer genomics, generated an increasingly complex understanding of metabolic reprogramming in cancer. As numerous connections between oncogenic signalling pathways and metabolic activities emerge, the importance of metabolic reprogramming in cancer is being increasingly recognized. The identification of metabolic weaknesses of cancer cells has been used to create strategies for treating cancer, but there are still challenges to be faced in bringing the drugs that target cancer metab. to the clinic.
- 8Thiam, A. R.; Beller, M. The Why, When and How of Lipid Droplet Diversity. J. Cell Sci. 2017, 130 (2), 315– 324, DOI: 10.1242/jcs.192021Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtF2gsL7M&md5=fe4c29e481d35619b6442d58a2375450The why, when and how of lipid droplet diversityThiam, Abdou Rachid; Beller, MathiasJournal of Cell Science (2017), 130 (2), 315-324CODEN: JNCSAI; ISSN:1477-9137. (Company of Biologists Ltd.)Lipid droplets are the universal cellular organelles for the transient or long-term storage of lipids. The no., size and compn. of lipid droplets vary greatly within cells in a homogenous population as well as in different cell types. The variability of intracellular lipid-storage organelles reflects the diversification of lipid droplet compn. and function. Lipid droplet diversification results, for example, in two cellular lipid droplet populations that are prone to diminish and grow, resp. The aberrant accumulation or depletion of lipids are hallmarks or causes of various human pathologies. Thus, a better understanding of the origins of lipid droplet diversification is not only a fascinating cell biol. question but also potentially serves to improve comprehension of pathologies that entail the accumulation of lipids. This Commentary covers the lipid droplet life cycle and highlights the early steps during lipid droplet biogenesis, which we propose to be the potential driving forces of lipid droplet diversification.
- 9Tirinato, L.; Pagliari, F.; Limongi, T.; Marini, M.; Falqui, A.; Seco, J.; Candeloro, P.; Liberale, C.; Di Fabrizio, E. An Overview of Lipid Droplets in Cancer and Cancer Stem Cells. Stem Cells Int. 2017, 2017, 1– 17, DOI: 10.1155/2017/1656053Google ScholarThere is no corresponding record for this reference.
- 10Wu, H.; Volponi, J. V.; Oliver, A. E.; Parikh, A. N.; Simmons, B. A.; Singh, S. In Vivo Lipidomics Using Single-Cell Raman Spectroscopy. Proc. Natl. Acad. Sci. U. S. A. 2011, 108 (9), 3809– 3814, DOI: 10.1073/pnas.1009043108Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXivFOqt78%253D&md5=40bb2127d3598103a795f761dab09c9fIn vivo lipidomics using single-cell raman spectroscopyWu, Huawen; Volponi, Joanne V.; Oliver, Ann E.; Parikh, Atul N.; Simmons, Blake A.; Singh, SeemaProceedings of the National Academy of Sciences of the United States of America (2011), 108 (9), 3809-3814, S3809/1-S3809/11CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)We describe a method for direct, quant., in vivo lipid profiling of oil-producing microalgae using single-cell laser-trapping Raman spectroscopy. This approach is demonstrated in the quant. detn. of the degree of unsatn. and transition temps. of constituent lipids within microalgae. These properties are important markers for detg. engine compatibility and performance metrics of algal biodiesel. We show that these factors can be directly measured from a single living microalgal cell held in place with an optical trap while simultaneously collecting Raman data. Cellular response to different growth conditions is monitored in real time. Our approach circumvents the need for lipid extn. and anal. that is both slow and invasive. Furthermore, this technique yields real-time chem. information in a label-free manner, thus eliminating the limitations of impermeability, toxicity, and specificity of the fluorescent probes common in currently used protocols. Although the single-cell Raman spectroscopy demonstrated here is focused on the study of the microalgal lipids with biofuel applications, the anal. capability and quantitation algorithms demonstrated are applicable to many different organisms and should prove useful for a diverse range of applications in lipidomics.
- 11Balog, J.; Sasi-Szabó, L.; Kinross, J.; Lewis, M. R.; Muirhead, L. J.; Veselkov, K.; Mirnezami, R.; Dezső, B.; Damjanovich, L.; Darzi, A. Intraoperative Tissue Identification Using Rapid Evaporative Ionization Mass Spectrometry. Sci. Transl. Med. 2013, 5 (194), 194ra93, DOI: 10.1126/scitranslmed.3005623Google ScholarThere is no corresponding record for this reference.
- 12Wenk, M. R. Lipidomics: New Tools and Applications. Cell 2010, 143 (6), 888– 895, DOI: 10.1016/j.cell.2010.11.033Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsFGgsrbI&md5=83b0ba44ac3132aa385a24ffa348d7f8Lipidomics: New Tools and ApplicationsWenk, Markus R.Cell (Cambridge, MA, United States) (2010), 143 (6), 888-895CODEN: CELLB5; ISSN:0092-8674. (Cell Press)A review. Once viewed simply as a reservoir for carbon storage, lipids are no longer cast as bystanders in the drama of biol. systems. The emerging field of lipidomics is driven by technol., most notably mass spectrometry, but also by complementary approaches for the detection and characterization of lipids and their biosynthetic enzymes in living cells. The development of these integrated tools promises to greatly advance our understanding of the diverse biol. roles of lipids.
- 13Bergholt, M. S.; Serio, A.; McKenzie, J. S.; Boyd, A.; Soares, R. F.; Tillner, J.; Chiappini, C.; Wu, V.; Dannhorn, A.; Takats, Z. Correlated Heterospectral Lipidomics for Biomolecular Profiling of Remyelination in Multiple Sclerosis. ACS Cent. Sci. 2018, 4 (1), 39– 51, DOI: 10.1021/acscentsci.7b00367Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXitVejurnF&md5=1a0a2def89f5d9768b22886babe0698cCorrelated Heterospectral Lipidomics for Biomolecular Profiling of Remyelination in Multiple SclerosisBergholt, Mads S.; Serio, Andrea; McKenzie, James S.; Boyd, Amanda; Soares, Renata F.; Tillner, Jocelyn; Chiappini, Ciro; Wu, Vincen; Dannhorn, Andreas; Takats, Zoltan; Williams, Anna; Stevens, Molly M.ACS Central Science (2018), 4 (1), 39-51CODEN: ACSCII; ISSN:2374-7951. (American Chemical Society)Analyzing lipid compn. and distribution within the brain is important to study white matter pathologies that present focal demyelination lesions, such as multiple sclerosis. Some lesions can endogenously re-form myelin sheaths. Therapies aim to enhance this repair process in order to reduce neurodegeneration and disability progression in patients. In this context, a lipidomic anal. providing both precise mol. classification and well-defined localization is crucial to detect changes in myelin lipid content. Here we develop a correlated heterospectral lipidomic (HSL) approach based on coregistered Raman spectroscopy, desorption electrospray ionization mass spectrometry (DESI-MS), and immunofluorescence imaging. We employ HSL to study the structural and compositional lipid profile of demyelination and remyelination in an induced focal demyelination mouse model and in multiple sclerosis lesions from patients ex vivo. Pixelwise coregistration of Raman spectroscopy and DESI-MS imaging generated a heterospectral map used to interrelate biomol. structure and compn. of myelin. Multivariate regression anal. enabled Raman-based assessment of highly specific lipid subtypes in complex tissue for the first time. This method revealed the temporal dynamics of remyelination and provided the first indication that newly formed myelin has a different lipid compn. compared to normal myelin. HSL enables detailed mol. myelin characterization that can substantially improve upon the current understanding of remyelination in multiple sclerosis and provides a strategy to assess remyelination treatments in animal models.
- 14Hosokawa, M.; Ando, M.; Mukai, S.; Osada, K.; Yoshino, T.; Hamaguchi, H.; Tanaka, T. In Vivo Live Cell Imaging for the Quantitative Monitoring of Lipids by Using Raman Microspectroscopy. Anal. Chem. 2014, 86 (16), 8224– 8230, DOI: 10.1021/ac501591dGoogle Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXht1aht7%252FE&md5=d89090139173010147f5c4870aab8be6In Vivo Live Cell Imaging for the Quantitative Monitoring of Lipids by Using Raman MicrospectroscopyHosokawa, Masahito; Ando, Masahiro; Mukai, Shoichiro; Osada, Kyoko; Yoshino, Tomoko; Hamaguchi, Hiro-o; Tanaka, TsuyoshiAnalytical Chemistry (Washington, DC, United States) (2014), 86 (16), 8224-8230CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)A straightforward in vivo monitoring technique for biomols. would be an advantageous approach for understanding their spatiotemporal dynamics in living cells. However, the lack of adequate probes has hampered the quant. detn. of the chem. compn. and metabolomics of cellular lipids at single-cell resoln. Here, we describe a method for the rapid, direct, and quant. detn. of lipid mols. from living cells using single-cell Raman imaging. In vivo localization of lipids in the form of triacylglycerol (TAG) within oleaginous microalga and their mol. compns. are monitored with high spatial resoln. in a nondestructive and label-free manner. This method can provide quant. and real-time information on compns., chain lengths, and degree of unsatn. of fatty acids in living cells for improving the cultivating parameters or for detg. the harvest timing during large-scale cultivations for microalgal lipid accumulation toward biodiesel prodn. Therefore, this technique is a potential tool for in vivo lipidomics for understanding the dynamics of lipid metabs. in various organisms.
- 15Abramczyk, H.; Surmacki, J.; Kopeć, M.; Olejnik, A. K.; Lubecka-Pietruszewska, K.; Fabianowska-Majewska, K. The Role of Lipid Droplets and Adipocytes in Cancer. Raman Imaging of Cell Cultures: MCF10A, MCF7, and MDA-MB-231 Compared to Adipocytes in Cancerous Human Breast Tissue. Analyst 2015, 140 (7), 2224– 2235, DOI: 10.1039/C4AN01875CGoogle Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXivFagt74%253D&md5=f91ccebb5206022668da2bf6607594c4The role of lipid droplets and adipocytes in cancer. Raman imaging of cell cultures: MCF10A, MCF7, and MDA-MB-231 compared to adipocytes in cancerous human breast tissueAbramczyk, Halina; Surmacki, Jakub; Kopec, Monika; Olejnik, Alicja Klaudia; Lubecka-Pietruszewska, Katarzyna; Fabianowska-Majewska, KrystynaAnalyst (Cambridge, United Kingdom) (2015), 140 (7), 2224-2235CODEN: ANALAO; ISSN:0003-2654. (Royal Society of Chemistry)We have studied live non-malignant (MCF10A), mildly malignant (MCF7) and malignant (MDA-MB-231) breast cancer cells and human breast cancer tissue. We demonstrate the first application of Raman imaging and spectroscopy in diagnosing the role of lipid droplets in cell line cultures that closely mimic an in vivo environment of various stages in human breast cancer tissue. We have analyzed the compn. of the lipid droplets in non-malignant and malignant human breast epithelial cell lines and discussed the potential of lipid droplets as a prognostic marker in breast cancer. To identify any difference in the lipid droplet-assocd. biochem. and to correlate it with different stages of breast cancer, the PCA method was employed. The chem. compn. of lipids and proteins, both in the cell line models and in human breast tissue has been analyzed. The paper shows the alterations in lipid metab. that have been reported in cancer, at both the cellular and tissue levels, and discusses how they contribute to the different aspects of tumorigenesis.
- 16Dória, M. L.; Cotrim, C. Z.; Simões, C.; Macedo, B.; Domingues, P.; Domingues, M. R.; Helguero, L. A. Lipidomic Analysis of Phospholipids from Human Mammary Epithelial and Breast Cancer Cell Lines. J. Cell. Physiol. 2013, 228 (2), 457– 468, DOI: 10.1002/jcp.24152Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsFGqt7vO&md5=c18a4b2c4ca678908c8f5ce79f3329b3Lipidomic analysis of phospholipids from human mammary epithelial and breast cancer cell linesDoria, M. Luisa; Cotrim, Candida Z.; Simoes, Claudia; Macedo, Barbara; Domingues, Pedro; Domingues, M. Rosario; Helguero, Luisa A.Journal of Cellular Physiology (2013), 228 (2), 457-468CODEN: JCLLAX; ISSN:0021-9541. (Wiley-Blackwell)Alterations of phospholipid (PL) profiles have been assocd. to disease and specific lipids may be involved in the onset and evolution of cancer; yet, anal. of PL profiles using mass spectrometry (MS) in breast cancer cells is a novel approach. Previously, we reported a lipidomic anal. of PLs from mouse mammary epithelial and breast cancer cells using off-line thin layer chromatog. (TLC)-MS, where several changes in PL profile were found to be assocd. with the degree of malignancy of cells. In the present study, lipidomic anal. has been extended to human mammary epithelial cells and breast cancer cell lines (MCF10A, T47-D, and MDA-MB-231), using TLC-MS, validated by hydrophilic interaction liq. chromatog.-MS. Differences in phosphatidylethanolamine (PE) content relative to total amt. of PLs was highest in non-malignant cells while phosphatidic acid was present with highest relative abundance in metastatic cells. In addn., the following differences in PL mol. species assocd. to cancer phenotype, metastatic potential, and cell morphol. were found: higher levels of alkylacyl PCs and phosphatidylinositol (PI; 22:5/18:0) were detected in migratory cells, epithelial cells had less unsatd. fatty acyl chains and shorter aliph. tails in PE and sphingomyelin classes, while PI (18:0/18:1) was lowest in non-malignant cells compared to cancer cells. To date, information about PL changes in cancer progression is scarce, therefore results presented in this work will be useful as a starting point to define possible PLs with prospective as biomarkers and disclose metabolic pathways with potential for cancer therapy.
- 17Baumann, J.; Sevinsky, C.; Conklin, D. S. Lipid Biology of Breast Cancer. Biochim. Biophys. Acta, Mol. Cell Biol. Lipids 2013, 1831 (10), 1509– 1517, DOI: 10.1016/j.bbalip.2013.03.011Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXmvVeru74%253D&md5=d64d37246f4de8e324197252e5b6ab8cLipid biology of breast cancerBaumann, Jan; Sevinsky, Christopher; Conklin, Douglas S.Biochimica et Biophysica Acta, Molecular and Cell Biology of Lipids (2013), 1831 (10), 1509-1517CODEN: BBMLFG; ISSN:1388-1981. (Elsevier B. V.)A review. Alterations in lipid metab. have been reported in many types of cancer. Lipids have been implicated in the regulation of proliferation, differentiation, apoptosis, inflammation, autophagy, motility and membrane homeostasis. It is required that their biosynthesis is tightly regulated to ensure homeostasis and to prevent unnecessary energy expenditure. This review focuses on the emerging understanding of the role of lipids and lipogenic pathway regulation in breast cancer, including parallels drawn from the study of metabolic disease models, and suggestions on how these findings can potentially be exploited to promote gains in HER2/neu-pos. breast cancer research. This article is part of a Special Issue entitled Lipid Metab. in Cancer.
- 18Liu, Q.; Luo, Q.; Halim, A.; Song, G. Targeting Lipid Metabolism of Cancer Cells: A Promising Therapeutic Strategy for Cancer. Cancer Lett. 2017, 401, 39– 45, DOI: 10.1016/j.canlet.2017.05.002Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXotFynsr8%253D&md5=6e71f7f51e06bf38e99411ec9cf887a6Targeting lipid metabolism of cancer cells: A promising therapeutic strategy for cancerLiu, Qiuping; Luo, Qing; Halim, Alexander; Song, GuanbinCancer Letters (New York, NY, United States) (2017), 401 (), 39-45CODEN: CALEDQ; ISSN:0304-3835. (Elsevier)One of the most important metabolic hallmarks of cancer cells is deregulation of lipid metab. In addn., enhancing de novo fatty acid (FA) synthesis, increasing lipid uptake and lipolysis have also been considered as means of FA acquisition in cancer cells. FAs are involved in various aspects of tumorigenesis and tumor progression. Therefore, targeting lipid metab. is a promising therapeutic strategy for human cancer. Recent studies have shown that reprogramming lipid metab. plays important roles in providing energy, macromols. for membrane synthesis, and lipid signals during cancer progression. Moreover, accumulation of lipid droplets in cancer cells acts as a pivotal adaptive response to harmful conditions. Here, we provide a brief review of the crucial roles of FA metab. in cancer development, and place emphasis on FA origin, utilization and storage in cancer cells. Understanding the regulation of lipid metab. in cancer cells has important implications for exploring a new therapeutic strategy for management and treatment of cancer.
- 19Infanger, D. W.; Lynch, M. E.; Fischbach, C. Engineered Culture Models for Studies of Tumor-Microenvironment Interactions. Annu. Rev. Biomed. Eng. 2013, 15 (1), 29– 53, DOI: 10.1146/annurev-bioeng-071811-150028Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVSqtL7M&md5=6a03fb7facd2c94b135cc8a690cf380eEngineered culture models for studies of tumor-microenvironment interactionsInfanger, David W.; Lynch, Maureen E.; Fischbach, ClaudiaAnnual Review of Biomedical Engineering (2013), 15 (), 29-53CODEN: ARBEF7; ISSN:1523-9829. (Annual Reviews)A review. Heterogeneous microenvironmental conditions play crit. roles in cancer pathogenesis and therapy resistance and arise from changes in tissue dimensionality, cell-extracellular matrix (ECM) interactions, sol. factor signaling, oxygen as well as metabolic gradients, and exogenous biomech. cues. Traditional cell culture approaches are restricted in their ability to mimic this complexity with physiol. relevance, offering only partial explanation as to why novel therapeutic compds. are frequently efficacious in vitro but disappoint in preclin. and clin. studies. In an effort to overcome these limitations, phys. sciences-based strategies were employed to model specific aspects of the cancer microenvironment. Although these strategies offer promise to reveal the contributions of microenvironmental parameters on tumor initiation, progression, and therapy resistance, they, too, frequently suffer from limitations. This review highlights physicochem. and biol. key features of the tumor microenvironment, critically discusses advantages and limitations of current engineering strategies, and provides a perspective on future opportunities for engineered tumor models.
- 20Fischbach, C.; Chen, R.; Matsumoto, T.; Schmelzle, T.; Brugge, J. S.; Polverini, P. J.; Mooney, D. J. Engineering Tumors with 3D Scaffolds. Nat. Methods 2007, 4 (10), 855– 860, DOI: 10.1038/nmeth1085Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtV2mtr%252FE&md5=3ea950e03c308dfb0b931c53f28bff5aEngineering tumors with 3D scaffoldsFischbach, Claudia; Chen, Ruth; Matsumoto, Takuya; Schmelzle, Tobias; Brugge, Joan S.; Polverini, Peter J.; Mooney, David J.Nature Methods (2007), 4 (10), 855-860CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Microenvironmental conditions control tumorigenesis and biomimetic culture systems that allow for in vitro and in vivo tumor modeling may greatly aid studies of cancer cells' dependency on these conditions. The authors engineered 3-dimensional (3D) human tumor models using carcinoma cells in polymeric scaffolds that recreated microenvironmental characteristics representative of tumors in vivo. Strikingly, the angiogenic characteristics of tumor cells were dramatically altered upon 3D culture within this system, and corresponded much more closely to tumors formed in vivo. Cells in this model were also less sensitive to chemotherapy and yielded tumors with enhanced malignant potential. The authors assessed the broad relevance of these findings with 3D culture of other tumor cell lines in this same model, comparison with std. 3D Matrigel culture and in vivo expts. This new biomimetic model may provide a broadly applicable 3D culture system to study the effect of microenvironmental conditions on tumor malignancy in vitro and in vivo.
- 21Shekhar, M. P. V.V.; Tait, L.; Pauley, R. J.; Wu, G. S.; Santner, S. J.; Nangia-Makker, P.; Shekhar, V.; Nassar, H.; Visscher, D. W.; Heppner, G. H. Comedo-Ductal Carcinoma in Situ: A Paradoxical Role for Programmed Cell Death. Cancer Biol. Ther. 2008, 7 (11), 1774– 1782, DOI: 10.4161/cbt.7.11.6781Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhvVGgu70%253D&md5=da65d844da20239f0ef0337f47e29a30Comedo-ductal carcinoma in situ: a paradoxical role for programmed cell deathShekhar, Malathy P. V.; Tait, Larry; Pauley, Robert J.; Wu, Gen Sheng; Santner, Steven J.; Nangia-Makker, Pratima; Shekhar, Varun; Nassar, Hind; Visscher, Daniel W.; Heppner, Gloria H.; Miller, Fred R.Cancer Biology & Therapy (2008), 7 (11), 1774-1782CODEN: CBTAAO; ISSN:1538-4047. (Landes Bioscience)Comedo-DCIS is a histol. subtype of preinvasive breast neoplasia that is characterized by prominent apoptotic cell death and has greater malignant potential than other DCIS subtypes. We investigated the mechanisms of apoptosis in comedo-DCIS and its role in conversion of comedo-DCIS to invasive cancer. Clin. comedo-DCIS excisions and the MCF10DCIS.com human breast cancer model which produces lesions resembling comedo-DCIS were analyzed. Apoptotic luminal and myoepithelial cells were identified by TUNEL and reactivity to cleaved PARP antibody and cell death assessed by Western blotting, Mitocapture and immunohistochem. assays. MCF10DCIS.com cells undergo spontaneous apoptosis in vitro, both in monolayers and multicellular spheroids; it is assocd. with increased mitochondrial membrane permeability, increase in Bax/Bcl-2 ratio and occurs via caspase-9-dependent p53-independent pathway. This suggests that apoptosis is stromal-independent and that the cells are programmed to undergo apoptosis. Immunostaining with cleaved PARP antibody showed that myoepithelial apoptosis occurs before lesions progress to comedo-DCIS in both clin. comedo-DCIS and in vivo MCF10DCIS.com lesions. Intense staining for MMP-2, MMP-3, MMP-9 and MMP-11 was obsd. in the stroma and epithelia of solid DCIS lesions prior to conversion to comedo-DCIS in clin. and MCF10DCIS.com lesions. Gelatin zymog. showed higher MMP-2 levels in lysates and conditioned media of MCF10DCIS.com cells undergoing apoptosis. These data suggest that signals arising from the outside (microenvironmental) and inside (internal genetic alterations) of the duct act in concert to trigger apoptosis of myoepithelial and luminal epithelial cells. Our findings implicate spontaneous apoptosis in both the etiol. and progression of comedo-DCIS. It is possible that spontaneous apoptosis facilitates elimination of cells thus permitting expansion and malignant transformation of cancer cells that are resistant to spontaneous apoptosis.
- 22McElwee, J. L.; Mohanan, S.; Griffith, O. L.; Breuer, H. C.; Anguish, L. J.; Cherrington, B. D.; Palmer, A. M.; Howe, L. R.; Subramanian, V.; Causey, C. P.; Identification of PADI2 as a Potential Breast Cancer Biomarker and Therapeutic Target. BMC Cancer 2012. DOI: 10.1186/1471-2407-12-500 .Google ScholarThere is no corresponding record for this reference.
- 23Vidavsky, N.; Kunitake, J. A.; Chiou, A. E.; Northrup, P. A.; Porri, T. J.; Ling, L.; Fischbach, C.; Estroff, L. A. Studying Biomineralization Pathways in a 3D Culture Model of Breast Cancer Microcalcifications. Biomaterials 2018, 179, 71– 82, DOI: 10.1016/j.biomaterials.2018.06.030Google ScholarThere is no corresponding record for this reference.
- 24DelNero, P.; Lane, M.; Verbridge, S. S.; Kwee, B.; Kermani, P.; Hempstead, B.; Stroock, A.; Fischbach, C. 3D Culture Broadly Regulates Tumor Cell Hypoxia Response and Angiogenesis via Pro-Inflammatory Pathways. Biomaterials 2015, 55, 110– 118, DOI: 10.1016/j.biomaterials.2015.03.035Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXlsl2iurw%253D&md5=a471c90cc02529c0e25aabf8a8e9a9b73D culture broadly regulates tumor cell hypoxia response and angiogenesis via pro-inflammatory pathwaysDelNero, Peter; Lane, Maureen; Verbridge, Scott S.; Kwee, Brian; Kermani, Pouneh; Hempstead, Barbara; Stroock, Abraham; Fischbach, ClaudiaBiomaterials (2015), 55 (), 110-118CODEN: BIMADU; ISSN:0142-9612. (Elsevier Ltd.)Oxygen status and tissue dimensionality are crit. determinants of tumor angiogenesis, a hallmark of cancer and an enduring target for therapeutic intervention. However, it is unclear how these microenvironmental conditions interact to promote neovascularization, due in part to a lack of comprehensive, unbiased data sets describing tumor cell gene expression as a function of oxygen levels within three-dimensional (3D) culture. Here, we utilized alginate-based, oxygen-controlled 3D tumor models to study the interdependence of culture context and the hypoxia response. Microarray gene expression anal. of tumor cells cultured in 2D vs. 3D under ambient or hypoxic conditions revealed striking interdependence between culture dimensionality and hypoxia response, which was mediated in part by pro-inflammatory signaling pathways. In particular, interleukin-8 (IL-8) emerged as a major player in the microenvironmental regulation of the hypoxia program. Notably, this interaction between dimensionality and oxygen status via IL-8 increased angiogenic sprouting in a 3D endothelial invasion assay. Taken together, our data suggest that pro-inflammatory pathways are crit. regulators of tumor hypoxia response within 3D environments that ultimately impact tumor angiogenesis, potentially providing important therapeutic targets. Furthermore, these results highlight the importance of pathol. relevant tissue culture models to study the complex phys. and chem. processes by which the cancer microenvironment mediates new vessel formation.
- 25Kang, H. S.; Lee, S. C.; Park, Y. S.; Jeon, Y. E.; Lee, J. H.; Jung, S. Y.; Park, I. H.; Jang, S. H.; Park, H. M.; Yoo, C. W. Protein and Lipid MALDI Profiles Classify Breast Cancers According to the Intrinsic Subtype. BMC Cancer 2011, 11 (1), 465, DOI: 10.1186/1471-2407-11-465Google ScholarThere is no corresponding record for this reference.
- 26Kawashima, M.; Iwamoto, N.; Kawaguchi-Sakita, N.; Sugimoto, M.; Ueno, T.; Mikami, Y.; Terasawa, K.; Sato, T. A.; Tanaka, K.; Shimizu, K. High-Resolution Imaging Mass Spectrometry Reveals Detailed Spatial Distribution of Phosphatidylinositols in Human Breast Cancer. Cancer Sci. 2013, 104 (10), 1372– 1379, DOI: 10.1111/cas.12229Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsFOgs7rI&md5=15e8c8ac9b5687624be3607117e60508High-resolution imaging mass spectrometry reveals detailed spatial distribution of phosphatidylinositols in human breast cancerKawashima, Masahiro; Iwamoto, Noriko; Kawaguchi-Sakita, Nobuko; Sugimoto, Masahiro; Ueno, Takayuki; Mikami, Yoshiki; Terasawa, Kazuya; Sato, Taka-Aki; Tanaka, Koichi; Shimizu, Kazuharu; Toi, MasakazuCancer Science (2013), 104 (10), 1372-1379CODEN: CSACCM; ISSN:1349-7006. (Wiley-Blackwell)High-resoln. matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI IMS) is an emerging application for lipid research that provides a comprehensive and detailed spatial distribution of ionized mols. Recent lipidomic approach has identified several phospholipids and phosphatidylinositols (PIs) are accumulated in breast cancer tissues and are therefore novel biomarker candidates. Because their distribution and significance remain unclear, we investigated the precise spatial distribution of PIs in human breast cancer tissues using high-resoln. MALDI IMS. We evaluated tissues from nine human breast cancers and one normal mammary gland by neg. ion MALDI IMS at a resoln. of 10 μm. We detected 10 PIs with different fatty acid compns., and their proportions were remarkably variable in the malignant epithelial regions. High-resoln. imaging enabled us to discriminate cancer cell clusters from the adjacent stromal tissue within epithelial regions; moreover, this technique revealed that several PIs were specifically localized to cancer cell clusters. These PIs were heterogeneously distributed within cancer cell clusters, allowing us to identify two different populations of cancer cells that predominantly expressed either PI(18:0/18:1) or PI(18:0/20:3). Tracing the expression level of PIs during cancer cell progression suggested that the latter population is assocd. with the invasion. Our study documents a novel model for phospholipid anal. of breast cancer tissues by using high-resoln. MALDI IMS and identifies candidate PIs that can describe a specific phenotype of cancer cells.
- 27Angerer, T. B.; Magnusson, Y.; Landberg, G.; Fletcher, J. S. Lipid Heterogeneity Resulting from Fatty Acid Processing in the Human Breast Cancer Microenvironment Identified by GCIB-ToF-SIMS Imaging. Anal. Chem. 2016, 88 (23), 11946– 11954, DOI: 10.1021/acs.analchem.6b03884Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslCktrvL&md5=aa98a45a841a4038143f72454d1b0e2eLipid Heterogeneity Resulting from Fatty Acid Processing in the Human Breast Cancer Microenvironment Identified by GCIB-ToF-SIMS ImagingAngerer, Tina B.; Magnusson, Ylva; Landberg, Goeran; Fletcher, John S.Analytical Chemistry (Washington, DC, United States) (2016), 88 (23), 11946-11954CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Breast cancer is an umbrella term used to describe a collection of different diseases with broad inter- and intra-tumor heterogeneity. Understanding this variation is crit. in order to develop, and precisely prescribe, new treatments. Changes in the lipid metab. of cancerous cells can provide important indications as to the metabolic state of the cells but are difficult to investigate with conventional histol. methods. Due to the introduction of new higher energy (40 kV) gas cluster ion beams (GCIBs) time-of-flight secondary ion mass spectrometry (ToF-SIMS) imaging is now capable of providing information on the distribution of hundreds of mol. species simultaneously on a cellular to sub-cellular scale. GCIB-ToF-SIMS was used to elucidate changes in lipid compn. in 9 breast cancer biopsy samples. Improved mol. signal generation by the GCIB produced location specific information that revealed elevated levels of essential lipids to be related to inflammatory cells in the stroma while cancerous areas are dominated by non-essential fatty acids and a variety of phosphatidylinositol species with further in-tumor variety arising from decreased desaturase activity. These changes in lipid compn. due to different enzyme activity are seemingly independent of oxygen availability and can be linked to favorable cell membrane properties for either proliferation/invasion or drug resistance/survival.
- 28Cífková, E.; Holčapek, M.; Lísa, M.; Vrána, D.; Melichar, B.; Študent, V. Lipidomic Differentiation between Human Kidney Tumors and Surrounding Normal Tissues Using HILIC-HPLC/ESI-MS and Multivariate Data Analysis. J. Chromatogr. B: Anal. Technol. Biomed. Life Sci. 2015, 1000, 14– 21, DOI: 10.1016/j.jchromb.2015.07.011Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtlajur3J&md5=f3311392bf0df83fec64d7a59d9cc933Lipidomic differentiation between human kidney tumors and surrounding normal tissues using HILIC-HPLC/ESI-MS and multivariate data analysisCifkova, Eva; Holcapek, Michal; Lisa, Miroslav; Vrana, David; Melichar, Bohuslav; Student, VladimirJournal of Chromatography B: Analytical Technologies in the Biomedical and Life Sciences (2015), 1000 (), 14-21CODEN: JCBAAI; ISSN:1570-0232. (Elsevier B.V.)The characterization of differences among polar lipid classes in tumors and surrounding normal tissues of 20 kidney cancer patients is performed by hydrophilic interaction liq. chromatog. (HILIC) coupled to electrospray ionization mass spectrometry (ESI-MS). The detailed anal. of identified lipid classes using relative abundances of characteristic ions in neg.- and pos.-ion modes is used for the detn. of more than 120 individual lipid species contg. attached fatty acyls of different chain length and double bond no. Lipid species are described using relative abundances, providing a better visualization of lipidomic differences between tumor and normal tissues. The multivariate data anal. methods using unsupervised principal component anal. (PCA) and supervised orthogonal partial least square (OPLS) are used for the characterization of statistically significant differences in identified lipid species. Ten most significant up- and down-regulated lipids in OPLS score plots are also displayed by box plots. A notable increase of relative abundances of lipids contg. four and more double bonds is detected in tumor compared to normal tissues.
- 29Hilvo, M.; Denkert, C.; Lehtinen, L.; Muller, B.; Brockmoller, S.; Seppanen-Laakso, T.; Budczies, J.; Bucher, E.; Yetukuri, L.; Castillo, S. Novel Theranostic Opportunities Offered by Characterization of Altered Membrane Lipid Metabolism in Breast Cancer Progression. Cancer Res. 2011, 71 (9), 3236– 3245, DOI: 10.1158/0008-5472.CAN-10-3894Google ScholarThere is no corresponding record for this reference.
- 30Perrotti, F.; Rosa, C.; Cicalini, I.; Sacchetta, P.; Del Boccio, P.; Genovesi, D.; Pieragostino, D. Advances in Lipidomics for Cancer Biomarkers Discovery. Int. J. Mol. Sci. 2016, 17 (12), 1992, DOI: 10.3390/ijms17121992Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXht1eks7c%253D&md5=c767aaf1f2da0027563b3900d38cb35aAdvances in lipidomics for cancer biomarkers discoveryPerrotti, Francesca; Rosa, Consuelo; Cicalini, Ilaria; Sacchetta, Paolo; Del Boccio, Piero; Genovesi, Domenico; Pieragostino, DamianaInternational Journal of Molecular Sciences (2016), 17 (12), 1992/1-1992/26CODEN: IJMCFK; ISSN:1422-0067. (MDPI AG)Lipids play crit. functions in cellular survival, proliferation, interaction and death, since they are involved in chem.-energy storage, cellular signaling, cell membranes, and cell-cell interactions. These cellular processes are strongly related to carcinogenesis pathways, particularly to transformation, progression, and metastasis, suggesting the bioactive lipids are mediators of a no. of oncogenic processes. The current review gives a synopsis of a lipidomic approach in tumor characterization; we provide an overview on potential lipid biomarkers in the oncol. field and on the principal lipidomic methodologies applied. The novel lipidomic biomarkers are reviewed in an effort to underline their role in diagnosis, in prognostic characterization and in prediction of therapeutic outcomes. A lipidomic investigation through mass spectrometry highlights new insights on mol. mechanisms underlying cancer disease. This new understanding will promote clin. applications in drug discovery and personalized therapy.
- 31Chughtai, K.; Jiang, L.; Greenwood, T. R.; Glunde, K.; Heeren, R. M. A. Mass Spectrometry Images Acylcarnitines, Phosphatidylcholines, and Sphingomyelin in MDA-MB-231 Breast Tumor Models. J. Lipid Res. 2013, 54 (2), 333– 344, DOI: 10.1194/jlr.M027961Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXosF2htQ%253D%253D&md5=f43e68b6c9f3a1c996abcd87e61498eaMass spectrometry images acylcarnitines, phosphatidylcholines, and sphingomyelin in MDA-MB-231 breast tumor modelsChughtai, Kamila; Lu, Jiang; Greenwood, Tiffany R.; Glunde, Kristine; Heeren, Ron M. A.Journal of Lipid Research (2013), 54 (2), 333-344CODEN: JLPRAW; ISSN:0022-2275. (American Society for Biochemistry and Molecular Biology, Inc.)The lipid compns. of different breast tumor microenvironments are largely unknown due to limitations in lipid imaging techniques. Imaging lipid distributions would enhance our understanding of processes occurring inside growing tumors, such as cancer cell proliferation, invasion, and metastasis. Recent developments in MALDI mass spectrometry imaging (MSI) enable rapid and specific detection of lipids directly from thin tissue sections. In this study, we performed multimodal imaging of acylcarnitines, phosphatidylcholines (PC), a lysophosphatidylcholine (LPC), and a sphingomyelin (SM) from different microenvironments of breast tumor xenograft models, which carried tdTomato red fluorescent protein as a hypoxia-response element-driven reporter gene. The MSI mol. lipid images revealed spatially heterogeneous lipid distributions within tumor tissue. Four of the most-abundant lipid species, namely PC(16:0/16:0), PC(16:0/18:1), PC(18:1/18:1), and PC(18:0/18:1), were localized in viable tumor regions, whereas LPC(16:0/0:0) was detected in necrotic tumor regions. We identified a heterogeneous distribution of palmitoylcarnitine, stearoylcarnitine, PC(16:0/22:1), and SM(d18:1/16:0) sodium adduct, which colocalized primarily with hypoxic tumor regions. For the first time, we have applied a multimodal imaging approach that has combined optical imaging and MALDI-MSI with ion mobility sepn. to spatially localize and structurally identify acylcarnitines and a variety of lipid species present in breast tumor xenograft models.
- 32Confocal Raman Microscopy; Dieing, T., Hollricher, O., Toporski, J., Eds.; Springer Berlin Heidelberg: Berlin, Heidelberg, 2011.Google ScholarThere is no corresponding record for this reference.
- 33Butler, H. J.; Ashton, L.; Bird, B.; Cinque, G.; Curtis, K.; Dorney, J.; Esmonde-White, K.; Fullwood, N. J.; Gardner, B.; Martin-Hirsch, P. L. Using Raman Spectroscopy to Characterize Biological Materials. Nat. Protoc. 2016, 11 (4), 664– 687, DOI: 10.1038/nprot.2016.036Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xjs1yjs7k%253D&md5=563e9a3c9684196390e2001a791a6e86Using Raman spectroscopy to characterize biological materialsButler, Holly J.; Ashton, Lorna; Bird, Benjamin; Cinque, Gianfelice; Curtis, Kelly; Dorney, Jennifer; Esmonde-White, Karen; Fullwood, Nigel J.; Gardner, Benjamin; Martin-Hirsch, Pierre L.; Walsh, Michael J.; McAinsh, Martin R.; Stone, Nicholas; Martin, Francis L.Nature Protocols (2016), 11 (4), 664-687CODEN: NPARDW; ISSN:1750-2799. (Nature Publishing Group)Raman spectroscopy can be used to measure the chem. compn. of a sample, which can in turn be used to ext. biol. information. Many materials have characteristic Raman spectra, which means that Raman spectroscopy has proven to be an effective anal. approach in geol., semiconductor, materials and polymer science fields. The application of Raman spectroscopy and microscopy within biol. is rapidly increasing because it can provide chem. and compositional information, but it does not typically suffer from interference from water mols. Anal. does not conventionally require extensive sample prepn.; biochem. and structural information can usually be obtained without labeling. In this protocol, we aim to standardize and bring together multiple exptl. approaches from key leaders in the field for obtaining Raman spectra using a microspectrometer. As examples of the range of biol. samples that can be analyzed, we provide instructions for acquiring Raman spectra, maps and images for fresh plant tissue, formalin-fixed and fresh frozen mammalian tissue, fixed cells and biofluids. We explore a robust approach for sample prepn., instrumentation, acquisition parameters and data processing. By using this approach, we expect that a typical Raman expt. can be performed by a nonspecialist user to generate high-quality data for biol. materials anal.
- 34Talari, A. C. S.; Movasaghi, Z.; Rehman, S.; Rehman, I. U. Raman Spectroscopy of Biological Tissues. Appl. Spectrosc. Rev. 2015, 50 (1), 46– 111, DOI: 10.1080/05704928.2014.923902Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtlyrsrbI&md5=fde96a5868714fbb53e87ac1756a2a7bRaman Spectroscopy of Biological TissuesTalari, Abdullah Chandra Sekhar; Movasaghi, Zanyar; Rehman, Shazza; Rehman, Ihtesham urApplied Spectroscopy Reviews (2015), 50 (1), 46-111CODEN: APSRBB; ISSN:0570-4928. (Taylor & Francis, Inc.)A review. We previously published a comprehensive review paper reviewing the Raman spectroscopy of biol. mols. This research area has expanded rapidly, which warranted an update to the existing review paper by adding the recently reported studies in literature. This article reviews some of the recent advances of Raman spectroscopy in relation to biomedical applications starting from natural tissues to cancer biol. Raman spectroscopy, an optical mol. detective, is a vibrational spectroscopic technique that has potential not only in cancer diagnosis but also in understanding progression of the disease. This article summarizes some of the most widely obsd. peak frequencies and their assignments. The aim of this review is to develop a database of mol. fingerprints, which will facilitate researchers in identifying the chem. structure of the biol. tissues including most of the significant peaks reported both in the normal and cancerous tissues. It has covered a variety of Raman approaches and its quant. and qual. biochem. information. In addn., it covers the use of Raman spectroscopy to analyze a variety of different malignancies including breast, brain, cervical, gastrointestinal, lung, oral, and skin cancer. Multivariate anal. approaches used in these studies have also been covered.
- 35Soule, H. D.; Maloney, T. M.; Wolman, S. R.; Peterson, W. D.; Brenz, R.; McGrath, C. M.; Russo, J.; Pauley, R. J.; Jones, R. F.; Brooks, S. C. Isolation and Characterization of a Spontaneously Immortalized Human Breast Epithelial Cell Line, MCF-10. Cancer Res. 1990, 50 (18), 6075– 6086Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXktFSq&md5=bca00d50e0a1ef6366053785c0534acdIsolation and characterization of a spontaneously immortalized human breast epithelial cell line, MCF-10Soule, Herbert D.; Maloney, Terry M.; Wolman, Sandra R.; Peterson, Ward D., Jr.; Brenz, Richard, Jr.; McGrath, Charles M.; Russo, Jose; Pauley, Robert J.; Jones, Richard F.; Brooks, S. C.Cancer Research (1990), 50 (18), 6075-86CODEN: CNREA8; ISSN:0008-5472.Two sublines of a breast epithelial cell culture, MCF-10, derived from human fibrocystic mammary tissue exhibit immortality after extended cultivation in low calcium concns. (0.03-0.06 mM) and floating transfers in low calcium (MCF-10F), or by trypsin-Versene passages in the customary (normal) calcium levels, 1.05 mM (MCF-10A). Both sublines have been maintained as sep. entities after 2.3 yr (849 days) in vitro and at present have been in culture for longer than 4 yr. MCF-10 has the characteristics of normal breast epithelium by the following criteria: (a) lack of tumorigenicity in nude mice; (b) three-dimensional growth in collagen; (c) growth in culture that is controlled by hormones and growth factors; (d) lack of anchorage-independent growth; and (e) dome formation in confluent cultures. Cytogenetic anal. prior to immortalization showed normal diploid cells; although later passages showed minimal rearrangement and near-diploidy, the immortal cells were not karyotypically normal. The emergence of an immortal culture in normal calcium media was not an inherent characteristic of the original tissue from which MCF-10 was derived since reactivated cryo-preserved cells from cultures grown for 0.3 to 1.2 yr in low calcium were incapable of sustained growth in normal calcium.
- 36Miller, F. R.; Santner, S. J.; Tait, L.; Dawson, P. J. MCF10DCIS.Com Xenograft Model of Human Comedo Ductal Carcinoma in Situ. J. Natl. Cancer Inst. 2000, 92 (14), 1185– 1186, DOI: 10.1093/jnci/92.14.1185aGoogle Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3cvlvFentQ%253D%253D&md5=5ed501454dc8b8f787ea9837f03aa72fMCF10DCIS.com xenograft model of human comedo ductal carcinoma in situMiller F R; Santner S J; Tait L; Dawson P JJournal of the National Cancer Institute (2000), 92 (14), 1185-6 ISSN:0027-8874.There is no expanded citation for this reference.
- 37Barnabas, N.; Cohen, D. Phenotypic and Molecular Characterization of MCF10DCIS and SUM Breast Cancer Cell Lines. Int. J. Breast Cancer 2013, 2013, 872743, DOI: 10.1155/2013/872743Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3sznsFyjuw%253D%253D&md5=fc9e492c1ba5859a9e5d7f5f4c95657ePhenotypic and Molecular Characterization of MCF10DCIS and SUM Breast Cancer Cell LinesBarnabas Nandita; Cohen DaliaInternational journal of breast cancer (2013), 2013 (), 872743 ISSN:2090-3170.We reviewed the phenotypic and molecular characteristics of MCF10DCIS.com and the SUM cell lines based on numerous studies performed over the years. The major signaling pathways that give rise to the phenotype of these cells may serve as a good resource of information when researchers in drug discovery and development use these cells to identify novel targets and biomarkers. Major signaling pathways and mutations affecting the coding sequence are also described providing important information when using these cells as a model in a variety of studies.
- 38Santner, S. J.; Dawson, P. J.; Tait, L.; Soule, H. D.; Eliason, J.; Mohamed, A. N.; Wolman, S. R.; Heppner, G. H.; Miller, F. R. Malignant MCF10CA1 Cell Lines Derived from Premalignant Human Breast Epithelial MCF10AT Cells. Breast Cancer Res. Treat. 2001, 65 (2), 101– 110, DOI: 10.1023/A:1006461422273Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3M7mtVWitA%253D%253D&md5=a1e5b7147e918dbdca3e33d6e281c119Malignant MCF10CA1 cell lines derived from premalignant human breast epithelial MCF10AT cellsSantner S J; Dawson P J; Tait L; Soule H D; Eliason J; Mohamed A N; Wolman S R; Heppner G H; Miller F RBreast cancer research and treatment (2001), 65 (2), 101-10 ISSN:0167-6806.The MCF10 series of cell lines was derived from benign breast tissue from a woman with fibrocystic disease. The MCF10 human breast epithelial model system consists of mortal MCF10M and MCF10MS (mortal cells grown in serum-free and serum-containing media, respectively), immortalized but otherwise normal MCF10F and MCF10A lines (free-floating versus growth as attached cells), transformed MCF10AneoT cells transfected with T24 Ha-ras, and premalignant MCF10AT cells with potential for neoplastic progression. The MCF10AT, derived from xenograft-passaged MCF10-AneoT cells, generates carcinomas in approximately 25% of xenografts. We now report the derivation of fully malignant MCF10CA1 lines that complete the spectrum of progression from relatively normal breast epithelial cells to breast cancer cells capable of metastasis. MCF10CA1 lines display histologic variations ranging from undifferentiated carcinomas, sometimes with focal squamous differentiation, to well-differentiated adenocarcinomas. At least two metastasize to the lung following injection of cells into the tail vein; one line grows very rapidly in the lung, with animals moribund within 4 weeks, whereas the other requires 15 weeks to reach the same endpoint. In addition to variations in efficiency of tumor production, the MCF10CA1 lines show differences in morphology in culture, anchorage-independent growth, karyotype, and immunocytochemistry profiles. The MCF10 model provides a unique tool for the investigation of molecular changes during progression of human breast neoplasia and the generation of tumor heterogeneity on a common genetic background.
- 39Alo, P. L.; Visca, P.; Marci, A.; Mangoni, A.; Botti, C.; Di Tondo, U. Expression of Fatty Acid Synthase (FAS) as a Predictor of Recurrence in Stage I Breast Carcinoma Patients. Cancer 1996, 77 (3), 474– 482, DOI: 10.1002/(SICI)1097-0142(19960201)77:3<474::AID-CNCR8>3.0.CO;2-KGoogle Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28Xht12jsL0%253D&md5=9f47ad6c3a97ada40fc288552f638c59Expression of fatty acid synthase (FAS) as a predictor of recurrence in stage I breast carcinoma patientsAlo, Piero L.; Visca, Paolo; Marci, Adele; Mangoni, Antonella; Botti, Claudio; Di Tondo, UgoCancer (New York) (1996), 77 (3), 474-82CODEN: CANCAR; ISSN:0008-543X. (Wiley-Liss)Fatty acid synthase (FAS) is a mol. found in tumor cells from breast carcinomas of patients whose prognosis is very poor. Recently, this mol. has been identified as the key enzyme in fatty acid biosynthesis. This study was done to test the strength of FAS as a prognostic indicator for disease free survival (DFS) and overall survival (OS). Clin. records, histol. features, immunohistochem. expression of cathepsin D and c-erbB-2, and estrogen and progesterone receptor status of 110 Stage I breast carcinoma patients were all assocd. with FAS by a chi-square test. The patterns of DFS and OS were estd. over a ten-year follow-up period using the Kaplan-Meier method. Univariate and multivariate anal. were evaluated using a log logistic regression model. Multivariate regression anal. was based on the Cox proportional hazard model. To detect FAS, cathepsin D and c-erbB-2 expression as well as estrogen and progesterone receptor status, the authors used the unlabeled immunoperoxidase technique on formalin fixed, paraffin embedded tissue. FAS was significantly assocd. with a higher risk of recurrence because it predicted both DFS and OS when evaluated as a continuous variable and DFS when evaluated with other prognostic markers. Peritumoral lymphatic vessel invasion was the other most significant independent predictor for DFS and OS. FAS is a reliable prognostic marker to predict DFS and OS in patients with early breast cancer.
- 40Milgraum, L. Z.; Witters, L. A.; Pasternack, G. R.; Kuhajda, F. P. Enzymes of the Fatty Acid Synthesis Pathway Are Highly Expressed in in Situ Breast Carcinoma. Clin. Cancer Res. 1997, 3 (11), 2115– 2120Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXns1Ogt78%253D&md5=bb85cb2753c93f199c01517e3887f15aEnzymes of the fatty acid synthesis pathway are highly expressed in in situ breast carcinomaMilgraum, Lea Z.; Witters, Lee A.; Pasternack, Gary R.; Kuhajda, Francis P.Clinical Cancer Research (1997), 3 (11), 2115-2120CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Expression of high levels of fatty acid synthase (FAS), an important enzyme in fatty acid synthesis, has been identified in a wide variety of human carcinomas. In breast and prostate carcinoma, FAS expression appears to be assocd. with aggressive disease. Recent biochem. studies have demonstrated that FAS expression in cancer cells connotes activation of the entire fatty acid synthesis pathway leading to the prodn. of palmitic acid. Here, we explore the immunohistochem. expression of FAS and human acetyl-CoA carboxylase (HACC), the rate-limiting enzyme in fatty acid synthesis, in breast cancer progression from histol. normal breast through the development of in situ duct and lobular carcinoma to infiltrating carcinoma. Both FAS and the Mr 275,000 isoform of HACC are expressed in a small subset of cells in normal breast lobules and terminal ducts. Upon development of either in situ duct or lobular carcinoma, FAS and both isoforms of HACC are expressed at higher levels and in a majority of the cells. These findings suggest that expression of the enzymes of fatty acid synthesis are frequently altered early in the progression of human breast carcinoma.
- 41Menendez, J. A.; Lupu, R. Fatty Acid Synthase and the Lipogenic Phenotype in Cancer Pathogenesis. Nat. Rev. Cancer 2007, 7 (10), 763– 777, DOI: 10.1038/nrc2222Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtVOnsr3P&md5=04e65d93150e39aef32d00cb0339fc92Fatty acid synthase and the lipogenic phenotype in cancer pathogenesisMenendez, Javier A.; Lupu, RuthNature Reviews Cancer (2007), 7 (10), 763-777CODEN: NRCAC4; ISSN:1474-175X. (Nature Publishing Group)A review. Fatty acid synthase (FASN) catalyzes the synthesis of fatty acids, and this synthetic pathway is upregulated in many tumors. How might FASN and increased lipogenesis be involved in cancer, and is FASN a valid therapeutic target. There is a renewed interest in the ultimate role of fatty acid synthase (FASN) - a key lipogenic enzyme catalyzing the terminal steps in the de novo biogenesis of fatty acids - in cancer pathogenesis. Tumor-assocd. FASN, by conferring growth and survival advantages rather than functioning as an anabolic energy-storage pathway, appears to necessarily accompany the natural history of most human cancers. A recent identification of cross-talk between FASN and well-established cancer-controlling networks begins to delineate the oncogenic nature of FASN-driven lipogenesis. FASN, a nearly-universal druggable target in many human carcinomas and their precursor lesions, offers new therapeutic opportunities for metabolically treating and preventing cancer.
- 42Ghosh, C.; Nandi, S.; Bhattacharyya, K. Probing Micro-Environment of Lipid Droplets in a Live Breast Cell: MCF7 and MCF10A. Chem. Phys. Lett. 2017, 670, 27– 31, DOI: 10.1016/j.cplett.2016.12.068Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXlsFagtw%253D%253D&md5=6979891b49e8e0e3614c17302804c273Probing micro-environment of lipid droplets in a live breast cell: MCF7 and MCF10AGhosh, Catherine; Nandi, Somen; Bhattacharyya, KankanChemical Physics Letters (2017), 670 (), 27-31CODEN: CHPLBC; ISSN:0009-2614. (Elsevier B.V.)Local environment of the lipid droplets inside the breast cancer cells, MCF7 and in non-malignant breast cells, MCF10A is monitored using time-resolved confocal microscopy. For this study, a coumarin-based dye C153 has been used. The local polarity and the solvation dynamics indicate that a cytoplasmic lipid droplet is less polar and displays slower solvation dynamics compared to the cytosol. Significant differences in terms of no. of lipid droplets, polarity and solvation dynamics are obsd. between the cancer cell (MCF7) and its non-malignant cell (MCF10A).
- 43Wilmanski, T.; Buhman, K.; Donkin, S. S.; Burgess, J. R.; Teegarden, D. 1α,25-Dihydroxyvitamin D Inhibits de Novo Fatty Acid Synthesis and Lipid Accumulation in Metastatic Breast Cancer Cells through down-Regulation of Pyruvate Carboxylase. J. Nutr. Biochem. 2017, 40, 194– 200, DOI: 10.1016/j.jnutbio.2016.11.006Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitVajs7vM&md5=e5284356e8a8274dfdf10c310cf208f61α,25-dihydroxyvitamin D inhibits de novo fatty acid synthesis and lipid accumulation in metastatic breast cancer cells through down-regulation of pyruvate carboxylaseWilmanski, Tomasz; Buhman, Kimberly; Donkin, Shawn S.; Burgess, John R.; Teegarden, DorothyJournal of Nutritional Biochemistry (2017), 40 (), 194-200CODEN: JNBIEL; ISSN:0955-2863. (Elsevier)Both increased de novo fatty acid synthesis and higher neutral lipid accumulation are a common phenotype obsd. in aggressive breast cancer cells, making lipid metab. a promising target for breast cancer prevention. In the present studies, we demonstrate a novel effect of the active metabolite of vitamin D, 1α,25-dihydroxyvitamin D (1,25(OH)2D) on lipid metab. in malignant breast epithelial cells. Treatment of MCF10CA1a breast epithelial cells with 1,25(OH)2D (10 nM) for 5 and 7 days decreased the level of triacylglycerol, the most abundant form of neutral lipids, by 20%(±3.9) and 50%(±5.9), resp. In addn., 1,25(OH)2D treatment for 5 days decreased palmitate synthesis from glucose, the major fatty acid synthesized de novo (48% ± 5.5 relative to vehicle). We have further identified the anaplerotic enzyme pyruvate carboxylase (PC) as a target of 1,25(OH)2D-mediated regulation and hypothesized that 1,25(OH)2D regulates breast cancer cell lipid metab. through inhibition of PC. PC mRNA expression was down-regulated with 1,25(OH)2D treatment at 2 (73% ± 6 relative to vehicle) and 5 (56% ± 8 relative to vehicle) days. Decrease in mRNA abundance corresponded with a decrease in PC protein expression at 5 days of treatment (54% ± 12 relative to vehicle). Constitutive overexpression of PC in MCF10CA1a cells using a pCMV6-PC plasmid inhibited the effect of 1,25(OH)2D on both TAG accumulation and de novo palmitate synthesis from glucose. Together, these studies demonstrate a novel mechanism through which 1,25(OH)2D regulates lipid metab. in malignant breast epithelial cells.
- 44Liu, X.; Kim, C. N.; Yang, J.; Jemmerson, R.; Wang, X. Induction of Apoptotic Program in Cell-Free Extracts: Requirement for DATP and Cytochrome C. Cell 1996, 86 (1), 147– 157, DOI: 10.1016/S0092-8674(00)80085-9Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28XktlGnsbY%253D&md5=5789cef4b6161cbf1faff51ecc0e0096Induction of apoptotic program in cell-free extracts: requirement for dATP and cytochrome cLiu, Xuesong; Kim, Caryn Naekyung; Yang, Jie; Jemmerson, Ronald; Wang, XiaodongCell (Cambridge, Massachusetts) (1996), 86 (1), 147-157CODEN: CELLB5; ISSN:0092-8674. (Cell Press)A cell-free system based on cytosols of normally growing cells was established that reproduces aspects of the apoptotic program in vitro. The apoptotic program was initiated by the addn. of dATP. Fractionation of cytosol yielded a 15-kDa protein that was required for in vitro apoptosis. The absorption spectrum and protein sequence revealed that this protein is cytochrome c. Elimination of cytochrome c from cytosol by immunodepletion, or inclusion of sucrose to stabilize mitochondria during cytosol prepn., diminished the apoptotic activity. Adding cytochrome c back to the cytochrome c-depleted exts. restored their apoptotic activity. Cells undergoing apoptosis in vivo showed increased release of cytochrome c to their cytosol, suggesting that mitochondria may function in apoptosis by releasing cytochrome c.
- 45Rygula, A.; Majzner, K.; Marzec, K. M.; Kaczor, A.; Pilarczyk, M.; Baranska, M. Raman Spectroscopy of Proteins: A Review. J. Raman Spectrosc. 2013, 44 (8), 1061– 1076, DOI: 10.1002/jrs.4335Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFSgsLnI&md5=34557bc0aff6ebe5b4f23288a3ab91a6Raman spectroscopy of proteins: a reviewRygula, A.; Majzner, K.; Marzec, K. M.; Kaczor, A.; Pilarczyk, M.; Baranska, M.Journal of Raman Spectroscopy (2013), 44 (8), 1061-1076CODEN: JRSPAF; ISSN:0377-0486. (John Wiley & Sons Ltd.)A review. In this work, 26 proteins of different structure, function and properties are investigated by Raman spectroscopy with 488, 532 and 1064 nm laser lines. The excitation lines were chosen in NIR and Vis range as the most common and to show the difference due to normal and resonance effect, sometimes accompanied by the fluorescence. The selected proteins were divided, according to the Structural Classification of Proteins, into four classes according to their secondary structure, i.e. α-helical (α), β-sheet (β), mixed structures (α/β, α + β, s) and others. For all compds., FT-Raman and two Vis spectra are presented along with the detailed band assignment. To the best of our knowledge, this is the first review showing the potential of Raman spectroscopy for the measurement and anal. of such a large collection of individual proteins. This work can serve as a comprehensive vibrational spectra library, based on our and previous Raman measurements. Copyright © 2013 John Wiley & Sons, Ltd.
- 46Ogretmen, B. Sphingolipid Metabolism in Cancer Signalling and Therapy. Nat. Rev. Cancer 2017, 18 (1), 33– 50, DOI: 10.1038/nrc.2017.96Google ScholarThere is no corresponding record for this reference.
- 47Kenny, P. A.; Lee, G. Y.; Myers, C. A.; Neve, R. M.; Semeiks, J. R.; Spellman, P. T.; Lorenz, K.; Lee, E. H.; Barcellos-Hoff, M. H.; Petersen, O. W. The Morphologies of Breast Cancer Cell Lines in Three-Dimensional Assays Correlate with Their Profiles of Gene Expression. Mol. Oncol. 2007, 1 (1), 84– 96, DOI: 10.1016/j.molonc.2007.02.004Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtlOitbbO&md5=9370b0e1d910303d9dd3e98cac32e298The morphologies of breast cancer cell lines in three-dimensional assays correlate with their profiles of gene expressionKenny, Paraic A.; Lee, Genee Y.; Myers, Connie A.; Neve, Richard M.; Semeiks, Jeremy R.; Spellman, Paul T.; Lorenz, Katrin; Lee, Eva H.; Barcellos-Hoff, Mary Helen; Petersen, Ole W.; Gray, Joe W.; Bissell, Mina J.Molecular Oncology (2007), 1 (1), 84-96CODEN: MOONC3; ISSN:1574-7891. (Elsevier B.V.)3D cell cultures are rapidly becoming the method of choice for the physiol. relevant modeling of many aspects of non-malignant and malignant cell behavior ex vivo. Nevertheless, only a limited no. of distinct cell types have been evaluated in this assay to date. Here we report the first large scale comparison of the transcriptional profiles and 3D cell culture phenotypes of a substantial panel of human breast cancer cell lines. Each cell line adopts a colony morphol. of one of four main classes in 3D culture. These morphologies reflect, at least in part, the underlying gene expression profile and protein expression patterns of the cell lines, and distinct morphologies were also assocd. with tumor cell invasiveness and with cell lines originating from metastases. We further demonstrate that consistent differences in genes encoding signal transduction proteins emerge when even tumor cells are cultured in 3D microenvironments.
- 48Nath, S.; Devi, G. R. Three-Dimensional Culture Systems in Cancer Research: Focus on Tumor Spheroid Model. Pharmacol. Ther. 2016, 163, 94– 108, DOI: 10.1016/j.pharmthera.2016.03.013Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XmtVSgsLk%253D&md5=5ea2141b4cd53d1959ebf2065e866ea2Three-dimensional culture systems in cancer research: Focus on tumor spheroid modelNath, Sritama; Devi, Gayathri R.Pharmacology & Therapeutics (2016), 163 (), 94-108CODEN: PHTHDT; ISSN:0163-7258. (Elsevier)Cancer cells propagated in three-dimensional (3D) culture systems exhibit physiol. relevant cell-cell and cell-matrix interactions, gene expression and signaling pathway profiles, heterogeneity and structural complexity that reflect in vivo tumors. In recent years, development of various 3D models has improved the study of host-tumor interaction and use of high-throughput screening platforms for anti-cancer drug discovery and development. This review attempts to summarize the various 3D culture systems, with an emphasis on the most well characterized and widely applied model - multicellular tumor spheroids. This review also highlights the various techniques to generate tumor spheroids, methods to characterize them, and its applicability in cancer research.
- 49Edmondson, R.; Broglie, J. J.; Adcock, A. F.; Yang, L. Three-Dimensional Cell Culture Systems and Their Applications in Drug Discovery and Cell-Based Biosensors. Assay Drug Dev. Technol. 2014, 12 (4), 207– 218, DOI: 10.1089/adt.2014.573Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXotVantLY%253D&md5=8bf702b6f1bf095faa1c712fd802ebdbThree-Dimensional Cell Culture Systems and Their Applications in Drug Discovery and Cell-Based BiosensorsEdmondson, Rasheena; Broglie, Jessica Jenkins; Adcock, Audrey F.; Yang, LijuAssay and Drug Development Technologies (2014), 12 (4), 207-218CODEN: ADDTAR; ISSN:1540-658X. (Mary Ann Liebert, Inc.)A review. Three-dimensional (3D) cell culture systems have gained increasing interest in drug discovery and tissue engineering due to their evident advantages in providing more physiol. relevant information and more predictive data for in vivo tests. In this review, we discuss the characteristics of 3D cell culture systems in comparison to the two-dimensional (2D) monolayer culture, focusing on cell growth conditions, cell proliferation, population, and gene and protein expression profiles. The innovations and development in 3D culture systems for drug discovery over the past 5 years are also reviewed in the article, emphasizing the cellular response to different classes of anticancer drugs, focusing particularly on similarities and differences between 3D and 2D models across the field. The progression and advancement in the application of 3D cell cultures in cell-based biosensors is another focal point of this review.
- 50Tauchi-Sato, K.; Ozeki, S.; Houjou, T.; Taguchi, R.; Fujimoto, T. The Surface of Lipid Droplets Is a Phospholipid Monolayer with a Unique Fatty Acid Composition. J. Biol. Chem. 2002, 277 (46), 44507– 44512, DOI: 10.1074/jbc.M207712200Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38Xos1akur8%253D&md5=00ecf3ba28f4744ae8c57834e818cb58The Surface of Lipid Droplets Is a Phospholipid Monolayer with a Unique Fatty Acid CompositionTauchi-Sato, Kumi; Ozeki, Shintaro; Houjou, Toshiaki; Taguchi, Ryo; Fujimoto, ToyoshiJournal of Biological Chemistry (2002), 277 (46), 44507-44512CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)We found that caveolin-2 is targeted to the surface of lipid droplets (Fujimoto, T., Kogo, H., Ishiguro, K., Tauchi, K., and Nomura, R. (2001) J. Cell Biol. 152, 1079-1085) and hypothesized that the lipid droplet surface is a kind of membrane. To elucidate the characteristics of the lipid droplet surface, we isolated lipid droplets from HepG2 cells and analyzed them by cryoelectron microscopy and by mass spectrometry. By use of cryoelectron microscopy at the stage temp. of 4.2 K, the lipid droplet surface was obsd. as a single line without any fixation or staining, indicating the presence of a single layer of phospholipids. This result appeared consistent with the hypothesis that the lipid droplet surface is derived from the cytoplasmic leaflet of the endoplasmic reticulum membrane and may be continuous to it. However, mass spectrometry revealed that the fatty acid compn. of phosphatidylcholine and lysophosphatidylcholine in lipid droplets is different from that of the rough endoplasmic reticulum. The ample presence of free cholesterol in lipid droplets also suggests that their surface is differentiated from the bulk endoplasmic reticulum membrane. On the other hand, although caveolin-2β and adipose differentiation-related protein, both localizing in lipid droplets, were enriched in the low d. floating fraction, the fatty acid compn. of the fraction was distinct from lipid droplets. Collectively, the result indicates that the lipid droplet surface is a hemi-membrane or a phospholipid monolayer contg. cholesterol but is compositionally different from the endoplasmic reticulum membrane or the sphingolipid/cholesterol-rich microdomain.
- 51Penno, A.; Hackenbroich, G.; Thiele, C. Phospholipids and Lipid Droplets. Biochim. Biophys. Acta, Mol. Cell Biol. Lipids 2013, 1831 (3), 589– 594, DOI: 10.1016/j.bbalip.2012.12.001Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXovVOmug%253D%253D&md5=4bfa8e971f0d7401b239e57d66de16ebPhospholipids and lipid dropletsPenno, Anke; Hackenbroich, Gregor; Thiele, ChristophBiochimica et Biophysica Acta, Molecular and Cell Biology of Lipids (2013), 1831 (3), 589-594CODEN: BBMLFG; ISSN:1388-1981. (Elsevier B. V.)A review. Lipid droplets are ubiquitous cellular organelles that allow cells to store large amts. of neutral lipids for membrane synthesis and energy supply in times of starvation. Compared to other cellular organelles, lipid droplets are structurally unique as they are made of a hydrophobic core of neutral lipids and are sepd. to the cytosol only by a surrounding phospholipid monolayer. This phospholipid monolayer consists of over a hundred different phospholipid mol. species of which phosphatidylcholine is the most abundant lipid class. However, lipid droplets lack some indispensable activities of the phosphatidylcholine biogenic pathways suggesting that they partially depend on other organelles for phosphatidylcholine synthesis. Here, the authors discuss very recent data on the compn., origin, transport and function of the phospholipid monolayer with a particular emphasis on the phosphatidylcholine metab. on and for lipid droplets. In addn., we highlight two very important quant. aspects: (i) The amt. of phospholipid required for lipid droplet monolayer expansion is remarkably small and (ii) to maintain the invariably round shape of lipid droplets, a cell must have a highly sensitive but so far unknown mechanism that regulates the ratio of phospholipid to neutral lipid in lipid droplets. This article is part of a Special Issue entitled Phospholipids and Phospholipid Metab.
- 52Wilfling, F.; Wang, H.; Haas, J. T.; Krahmer, N.; Gould, T. J.; Uchida, A.; Cheng, J.-X.; Graham, M.; Christiano, R.; Fröhlich, F. Triacylglycerol Synthesis Enzymes Mediate Lipid Droplet Growth by Relocalizing from the ER to Lipid Droplets. Dev. Cell 2013, 24 (4), 384– 399, DOI: 10.1016/j.devcel.2013.01.013Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXis1Oms7c%253D&md5=b5e30e8f6afd2ffe9e6f2f508263c4e5Triacylglycerol Synthesis Enzymes Mediate Lipid Droplet Growth by Relocalizing from the ER to Lipid DropletsWilfling, Florian; Wang, Huajin; Haas, Joel T.; Krahmer, Natalie; Gould, Travis J.; Uchida, Aki; Cheng, Ji-Xin; Graham, Morven; Christiano, Romain; Frohlich, Florian; Liu, Xinran; Buhman, Kimberly K.; Coleman, Rosalind A.; Bewersdorf, Joerg; Farese, Robert V.; Walther, Tobias C.Developmental Cell (2013), 24 (4), 384-399CODEN: DCEEBE; ISSN:1534-5807. (Cell Press)Lipid droplets (LDs) store metabolic energy and membrane lipid precursors. With excess metabolic energy, cells synthesize triacylglycerol (TG) and form LDs that grow dramatically. It is unclear how TG synthesis relates to LD formation and growth. Here, we identify two LD subpopulations: smaller LDs of relatively const. size, and LDs that grow larger. The latter population contains isoenzymes for each step of TG synthesis. Glycerol-3-phosphate acyltransferase 4 (GPAT4), which catalyzes the first and rate-limiting step, relocalizes from the endoplasmic reticulum (ER) to a subset of forming LDs, where it becomes stably assocd. ER-to-LD targeting of GPAT4 and other LD-localized TG synthesis isoenzymes is required for LD growth. Key features of GPAT4 ER-to-LD targeting and function in LD growth are conserved between Drosophila and mammalian cells. Our results explain how TG synthesis is coupled with LD growth and identify two distinct LD subpopulations based on their capacity for localized TG synthesis.
- 53Mouritsen, O. G.; Bagatolli, L. A. Life-as a Matter of Fat, 2nd ed.; Springer-Verlag: Heidelberg, 2005.Google ScholarThere is no corresponding record for this reference.
- 54Walther, T. C.; Farese, R. V. The Life of Lipid Droplets. Biochim. Biophys. Acta, Mol. Cell Biol. Lipids 2009, 1791 (6), 459– 466, DOI: 10.1016/j.bbalip.2008.10.009Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXntVKls70%253D&md5=7285b4c3d445dcb874c61e56926f0abcThe life of lipid dropletsWalther, Tobias C.; Farese, Robert V.Biochimica et Biophysica Acta, Molecular and Cell Biology of Lipids (2009), 1791 (6), 459-466CODEN: BBMLFG; ISSN:1388-1981. (Elsevier B. V.)A review. Lipid droplets are the least characterized of cellular organelles. Long considered simple lipid storage depots, these dynamic and remarkable organelles have recently been implicated in many biol. processes, and investigators are only now beginning to gain insights into their fascinating lives in cells. Here, the authors examine what is known of the life of lipid droplets. The authors review emerging data concerning their cellular biol. and present their thoughts on some of the most salient questions for investigation.
- 55Nieman, K. M.; Kenny, H. A.; Penicka, C. V.; Ladanyi, A.; Buell-Gutbrod, R.; Zillhardt, M. R.; Romero, I. L.; Carey, M. S.; Mills, G. B.; Hotamisligil, G. S. Adipocytes Promote Ovarian Cancer Metastasis and Provide Energy for Rapid Tumor Growth. Nat. Med. 2011, 17, 1498– 1503, DOI: 10.1038/nm.2492Google Scholar55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtlyis73N&md5=5a208c6ec3ad22910c8a30cc2391bd90Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growthNieman, Kristin M.; Kenny, Hilary A.; Penicka, Carla V.; Ladanyi, Andras; Buell-Gutbrod, Rebecca; Zillhardt, Marion R.; Romero, Iris L.; Carey, Mark S.; Mills, Gordon B.; Hotamisligil, Goekhan S.; Yamada, S. Diane; Peter, Marcus E.; Gwin, Katja; Lengyel, ErnstNature Medicine (New York, NY, United States) (2011), 17 (11), 1498-1503CODEN: NAMEFI; ISSN:1078-8956. (Nature Publishing Group)Intra-abdominal tumors, such as ovarian cancer, have a clear predilection for metastasis to the omentum, an organ primarily composed of adipocytes. Currently, it is unclear why tumor cells preferentially home to and proliferate in the omentum, yet omental metastases typically represent the largest tumor in the abdominal cavities of women with ovarian cancer. We show here that primary human omental adipocytes promote homing, migration and invasion of ovarian cancer cells, and that adipokines including interleukin-8 (IL-8) mediate these activities. Adipocyte-ovarian cancer cell coculture led to the direct transfer of lipids from adipocytes to ovarian cancer cells and promoted in vitro and in vivo tumor growth. Furthermore, coculture induced lipolysis in adipocytes and β-oxidn. in cancer cells, suggesting adipocytes act as an energy source for the cancer cells. A protein array identified upregulation of fatty acid-binding protein 4 (FABP4, also known as aP2) in omental metastases as compared to primary ovarian tumors, and FABP4 expression was detected in ovarian cancer cells at the adipocyte-tumor cell interface. FABP4 deficiency substantially impaired metastatic tumor growth in mice, indicating that FABP4 has a key role in ovarian cancer metastasis. These data indicate adipocytes provide fatty acids for rapid tumor growth, identifying lipid metab. and transport as new targets for the treatment of cancers where adipocytes are a major component of the microenvironment.
- 56Nieman, K. M.; Romero, I. L.; Van Houten, B.; Lengyel, E. Adipose Tissue and Adipocytes Support Tumorigenesis and Metastasis. Biochim. Biophys. Acta, Mol. Cell Biol. Lipids 2013, 1831 (10), 1533– 1541, DOI: 10.1016/j.bbalip.2013.02.010Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXkslamsbs%253D&md5=df984d32b4483f2bfed210751383ce11Adipose tissue and adipocytes support tumorigenesis and metastasisNieman, Kristin M.; Romero, Iris L.; Van Houten, Bennett; Lengyel, ErnstBiochimica et Biophysica Acta, Molecular and Cell Biology of Lipids (2013), 1831 (10), 1533-1541CODEN: BBMLFG; ISSN:1388-1981. (Elsevier B. V.)A review. Adipose tissue influences tumor development in two major ways. First, obese individuals have a higher risk of developing certain cancers (endometrial, esophageal, and renal cell cancer). However, the risk of developing other cancers (melanoma, rectal, and ovarian) is not altered by body mass. In obesity, hypertrophied adipose tissue depots are characterized by a state of low grade inflammation. In this activated state, adipocytes and inflammatory cells secrete adipokines and cytokines which are known to promote tumor development. In addn., the adipocyte mediated conversion of androgens to estrogen specifically contributes to the development of endometrial cancer, which shows the greatest relative risk (6.3-fold) increase between lean and obese individuals. Second, many tumor types (gastric, breast, colon, renal, and ovarian) grow in the anatomical vicinity of adipose tissue. During their interaction with cancer cells, adipocytes dedifferentiate into pre-adipocytes or are reprogrammed into cancer-assocd. adipocytes (CAA). CAA secrete adipokines which stimulate the adhesion, migration, and invasion of tumor cells. Cancer cells and CAA also engage in a dynamic exchange of metabolites. Specifically, CAA release fatty acids through lipolysis which are then transferred to cancer cells and used for energy prodn. through β-oxidn. The abundant availability of lipids from adipocytes in the tumor microenvironment, supports tumor progression and uncontrolled growth. Given that adipocytes are a major source of adipokines and energy for the cancer cell, understanding the mechanisms of metabolic symbiosis between cancer cells and adipocytes, should reveal new therapeutic possibilities. This article is part of a Special Issue entitled Lipid Metab. in Cancer.
- 57Thiam, A. R.; Farese, R. V.; Walther, T. C. The Biophysics and Cell Biology of Lipid Droplets. Nat. Rev. Mol. Cell Biol. 2013, 14 (12), 775– 786, DOI: 10.1038/nrm3699Google Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslGntLrK&md5=f38fca0797834390268b8180715fa3e6The biophysics and cell biology of lipid dropletsThiam, Abdou Rachid; Farese, Robert V., Jr.; Walther, Tobias C.Nature Reviews Molecular Cell Biology (2013), 14 (12), 775-786CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. Lipid droplets are intracellular organelles that are found in most cells, where they have fundamental roles in metab. They function prominently in storing oil-based reserves of metabolic energy and components of membrane lipids. Lipid droplets are the dispersed phase of an oil-in-water emulsion in the aq. cytosol of cells, and the importance of basic biophys. principles of emulsions for lipid droplet biol. is now being appreciated. Because of their unique architecture, with an interface between the dispersed oil phase and the aq. cytosol, specific mechanisms underlie their formation, growth and shrinkage. Such mechanisms enable cells to use emulsified oil when the demands for metabolic energy or membrane synthesis change. The regulation of the compn. of the phospholipid surfactants at the surface of lipid droplets is crucial for lipid droplet homeostasis and protein targeting to their surfaces.
- 58Pelletier, J.; Bellot, G.; Gounon, P.; Lacas-Gervais, S.; Pouysségur, J.; Mazure, N. M. Glycogen Synthesis Is Induced in Hypoxia by the Hypoxia-Inducible Factor and Promotes Cancer Cell Survival. Front. Oncol. 2012, 2, 18, DOI: 10.3389/fonc.2012.00018Google Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC38nnslShsA%253D%253D&md5=bee5fdd5bf92a923514e66dae65113e3Glycogen Synthesis is Induced in Hypoxia by the Hypoxia-Inducible Factor and Promotes Cancer Cell SurvivalPelletier Joffrey; Bellot Gregory; Gounon Pierre; Lacas-Gervais Sandra; Pouyssegur Jacques; Mazure Nathalie MFrontiers in oncology (2012), 2 (), 18 ISSN:.The hypoxia-inducible factor 1 (HIF-1), in addition to genetic and epigenetic changes, is largely responsible for alterations in cell metabolism in hypoxic tumor cells. This transcription factor not only favors cell proliferation through the metabolic shift from oxidative phosphorylation to glycolysis and lactic acid production but also stimulates nutrient supply by mediating adaptive survival mechanisms. In this study we showed that glycogen synthesis is enhanced in non-cancer and cancer cells when exposed to hypoxia, resulting in a large increase in glycogen stores. Furthermore, we demonstrated that the mRNA and protein levels of the first enzyme of glycogenesis, phosphoglucomutase1 (PGM1), were increased in hypoxia. We showed that induction of glycogen storage as well as PGM1 expression were dependent on HIF-1 and HIF-2. We established that hypoxia-induced glycogen stores are rapidly mobilized in cells that are starved of glucose. Glycogenolysis allows these "hypoxia-preconditioned" cells to confront and survive glucose deprivation. In contrast normoxic control cells exhibit a high rate of cell death following glucose removal. These findings point to the important role of hypoxia and HIF in inducing mechanisms of rapid adaptation and survival in response to a decrease in oxygen tension. We propose that a decrease in pO(2) acts as an "alarm" that prepares the cells to face subsequent nutrient depletion and to survive.
- 59Pescador, N.; Villar, D.; Cifuentes, D.; Garcia-Rocha, M.; Ortiz-Barahona, A.; Vazquez, S.; Ordoñez, A.; Cuevas, Y.; Saez-Morales, D.; Garcia-Bermejo, M. L. Hypoxia Promotes Glycogen Accumulation through Hypoxia Inducible Factor (HIF)-Mediated Induction of Glycogen Synthase 1. PLoS One 2010, 5 (3), e9644 DOI: 10.1371/journal.pone.0009644Google ScholarThere is no corresponding record for this reference.
- 60Favaro, E.; Bensaad, K.; Chong, M. G.; Tennant, D. A.; Ferguson, D. J. P.; Snell, C.; Steers, G.; Turley, H.; Li, J.-L.; Günther, U. L. Glucose Utilization via Glycogen Phosphorylase Sustains Proliferation and Prevents Premature Senescence in Cancer Cells. Cell Metab. 2012, 16 (6), 751– 764, DOI: 10.1016/j.cmet.2012.10.017Google Scholar60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhslWqur7I&md5=b74e88cc2c3cea7b355b22439d14874aGlucose Utilization via Glycogen Phosphorylase Sustains Proliferation and Prevents Premature Senescence in Cancer CellsFavaro, Elena; Bensaad, Karim; Chong, Mei G.; Tennant, Daniel A.; Ferguson, David J. P.; Snell, Cameron; Steers, Graham; Turley, Helen; Li, Ji-Liang; Guenther, Ulrich L.; Buffa, Francesca M.; McIntyre, Alan; Harris, Adrian L.Cell Metabolism (2012), 16 (6), 751-764CODEN: CMEEB5; ISSN:1550-4131. (Elsevier Inc.)Metabolic reprogramming of cancer cells provides energy and multiple intermediates crit. for cell growth. Hypoxia in tumors represents a hostile environment that can encourage these transformations. We report that glycogen metab. is upregulated in tumors in vivo and in cancer cells in vitro in response to hypoxia. In vitro, hypoxia induced an early accumulation of glycogen, followed by a gradual decline. Concordantly, glycogen synthase (GYS1) showed a rapid induction, followed by a later increase of glycogen phosphorylase (PYGL). PYGL depletion and the consequent glycogen accumulation led to increased reactive oxygen species (ROS) levels that contributed to a p53-dependent induction of senescence and markedly impaired tumorigenesis in vivo. Metabolic analyses indicated that glycogen degrdn. by PYGL is important for the optimal function of the pentose phosphate pathway. Thus, glycogen metab. is a key pathway induced by hypoxia, necessary for optimal glucose utilization, which represents a targetable mechanism of metabolic adaptation.
- 61Okada, M.; Smith, N. I.; Palonpon, A. F.; Endo, H.; Kawata, S.; Sodeoka, M.; Fujita, K. Label-Free Raman Observation of Cytochrome c Dynamics during Apoptosis. Proc. Natl. Acad. Sci. U. S. A. 2012, 109 (1), 28– 32, DOI: 10.1073/pnas.1107524108Google Scholar61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsVehsrg%253D&md5=05f3ec458114cd4d6d2091bf7e201114Label-free Raman observation of cytochrome c dynamics during apoptosisOkada, Masaya; Smith, Nicholas Isaac; Palonpon, Almar Flotildes; Endo, Hiromi; Kawata, Satoshi; Sodeoka, Mikiko; Fujita, KatsumasaProceedings of the National Academy of Sciences of the United States of America (2012), 109 (1), 28-32, S28/1-S28/2CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)We performed label-free observation of mol. dynamics in apoptotic cells by Raman microscopy. Dynamic changes in cytochrome c distribution at the Raman band of 750 cm-1 were obsd. after adding an apoptosis inducer to the cells. The comparison of mitochondria fluorescence images and Raman images of cytochrome c confirmed that changes in cytochrome c distribution can be distinguished as release of cytochrome c from mitochondria. Our observation also revealed that the redox state of cytochrome c was maintained during the release from the mitochondria. Monitoring mitochondrial membrane potential with JC-1 dye confirmed that the obsd. cytochrome c release was assocd. with apoptosis.
- 62Vazquez, A.; Kamphorst, J. J.; Markert, E. K.; Schug, Z. T.; Tardito, S.; Gottlieb, E. Cancer Metabolism at a Glance. J. Cell Sci. 2016, 129 (18), 3367– 3373, DOI: 10.1242/jcs.181016Google Scholar62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitVyht73F&md5=04c4e6b18ac946a397558d1ec98e5e4dCancer metabolism at a glanceVazquez, Alexei; Kamphorst, Jurre J.; Markert, Elke K.; Schug, Zachary T.; Tardito, Saverio; Gottlieb, EyalJournal of Cell Science (2016), 129 (18), 3367-3373CODEN: JNCSAI; ISSN:0021-9533. (Company of Biologists Ltd.)A defining hallmark of cancer is uncontrolled cell proliferation. This is initiated once cells have accumulated alterations in signaling pathways that control metab. and proliferation, wherein the metabolic alterations provide the energetic and anabolic demands of enhanced cell proliferation. How these metabolic requirements are satisfied depends, in part, on the tumor microenvironment, which dets. the availability of nutrients and oxygen. In this Cell Science at a Glance paper and the accompanying poster, we summarize our current understanding of cancer metab., emphasizing pathways of nutrient utilization and metab. that either appear or have been proven essential for cancer cells. We also review how this knowledge has contributed to the development of anticancer therapies that target cancer metab.
- 63Worgall, T. S. Sphingolipids: Major Regulators of Lipid Metabolism. Curr. Opin. Clin. Nutr. Metab. Care 2007, 10 (2), 149– 155, DOI: 10.1097/MCO.0b013e328028fda3Google Scholar63https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXjs1ylsro%253D&md5=052c779add3bd4133a3371ccd2ec5f26Sphingolipids: major regulators of lipid metabolismWorgall, Tilla S.Current Opinion in Clinical Nutrition and Metabolic Care (2007), 10 (2), 149-155CODEN: COCMF3; ISSN:1363-1950. (Lippincott Williams & Wilkins)A review. Emerging data strongly suggest a role of sphingolipid synthesis in the regulation of transcription factors and regulatory proteins that control cellular lipid homeostasis.
- 64Eichmann, T. O.; Lass, A. DAG Tales: The Multiple Faces of Diacylglycerol—stereochemistry, Metabolism, and Signaling. Cell. Mol. Life Sci. 2015, 72 (20), 3931– 3952, DOI: 10.1007/s00018-015-1982-3Google Scholar64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFCjtLnL&md5=31e9b2808d906fd24aa4ef4ab4d9aba8DAG tales: the multiple faces of diacylglycerol-stereochemistry, metabolism, and signalingEichmann, Thomas Oliver; Lass, AchimCellular and Molecular Life Sciences (2015), 72 (20), 3931-3952CODEN: CMLSFI; ISSN:1420-682X. (Birkhaeuser Basel)The neutral lipids diacylglycerols (DAGs) are involved in a plethora of metabolic pathways. They function as components of cellular membranes, as building blocks for glycero(phospho)lipids, and as lipid second messengers. Considering their central role in multiple metabolic processes and signaling pathways, cellular DAG levels require a tight regulation to ensure a const. and controlled availability. Interestingly, DAG species are versatile in their chem. structure. Besides the different fatty acid species esterified to the glycerol backbone, DAGs can occur in three different stereo/regioisoforms, each with unique biol. properties. Recent scientific advances have revealed that DAG metabolizing enzymes generate and distinguish different DAG isoforms, and that only one DAG isoform holds signaling properties. Herein, we review the current knowledge of DAG stereochem. and their impact on cellular metab. and signaling. Further, we describe intracellular DAG turnover and its stereochem. in a 3-pool model to illustrate the spatial and stereochem. sepn. and hereby the diversity of cellular DAG metab.
- 65Kadota, M.; Yang, H. H.; Gomez, B.; Sato, M.; Clifford, R. J.; Meerzaman, D.; Dunn, B. K.; Wakefield, L. M.; Lee, M. P. Delineating Genetic Alterations for Tumor Progression in the MCF10A Series of Breast Cancer Cell Lines. PLoS One 2010, 5 (2), e9201 DOI: 10.1371/journal.pone.0009201Google ScholarThere is no corresponding record for this reference.
- 66Han, G.; Gupta, S. D.; Gable, K.; Niranjanakumari, S.; Moitra, P.; Eichler, F.; Brown, R. H.; Harmon, J. M.; Dunn, T. M. Identification of Small Subunits of Mammalian Serine Palmitoyltransferase That Confer Distinct Acyl-CoA Substrate Specificities. Proc. Natl. Acad. Sci. U. S. A. 2009, 106 (20), 8186– 8191, DOI: 10.1073/pnas.0811269106Google ScholarThere is no corresponding record for this reference.
- 67Knapp, P.; Baranowski, M.; Knapp, M.; Zabielski, P.; Błachnio-Zabielska, A. U.; Górski, J. Altered Sphingolipid Metabolism in Human Endometrial Cancer. Prostaglandins Other Lipid Mediators 2010, 92 (1–4), 62– 66, DOI: 10.1016/j.prostaglandins.2010.03.002Google Scholar67https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXmtFeqsbY%253D&md5=489c4c99690000987f5d1f964d0c35e0Altered sphingolipid metabolism in human endometrial cancerKnapp, Pawel; Baranowski, Marcin; Knapp, Malgorzata; Zabielski, Piotr; Blachnio-Zabielska, Agnieszka U.; Gorski, JanProstaglandins & Other Lipid Mediators (2010), 92 (1-4), 62-66CODEN: POLMFL; ISSN:1098-8823. (Elsevier)There is a growing body of evidence indicating that bioactive sphingolipids play a key role in cancer development, progression and metastasis. However, sphingolipid metab. in malignant tumors is poorly investigated. Therefore, the aim of the present study was to examine the content of selected intermediates of ceramide metab. and the activity of key enzymes of ceramide de novo synthesis and sphingosine-1-phosphate (S1P) prodn. in the endometrial cancer. The specimens of cancer tissue and healthy endometrium were obtained from women undergoing surgery because of the cancer (n = 23) and because of myomas (n = 18), resp. The content of sphinganine, dihydroceramide, ceramide, sphingosine and S1P was measured using high pressure liq. chromatog. The activity of the enzymes was detd. using radioactive substrates. It has been found that the content of each examd. sphingolipid was markedly elevated in the cancer tissue compared with the healthy endometrium. Namely, sphinganine, sphingosine and dihydroceramide by 3-4.6-fold, ceramide and S1P by 1.9- and 1.6-fold, resp. Interestingly, the ratio of S1P to ceramide remained stable. The activity of serine palmitoyltransferase and sphingosine kinase 1 was increased by 2.3- and 2.6-fold, resp. We conclude that endometrial carcinoma is characterized by profound changes in sphingolipid metab. that likely contribute to its progression and chemoresistance.
- 68Patra, S. K. Dissecting Lipid Raft Facilitated Cell Signaling Pathways in Cancer. Biochim. Biophys. Acta, Rev. Cancer 2008, 1785 (2), 182– 206, DOI: 10.1016/j.bbcan.2007.11.002Google Scholar68https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXkvFWlur8%253D&md5=c3334d247415b40d2693a93d0e7fe4fbDissecting lipid raft facilitated cell signaling pathways in cancerPatra, Samir KumarBiochimica et Biophysica Acta, Reviews on Cancer (2008), 1785 (2), 182-206CODEN: BBACEU; ISSN:0304-419X. (Elsevier Ltd.)A review. Cancer is one of the most devastating disorders in our lives. Higher rate of proliferation than death of cells is one of the essential factors for development of cancer. The dynamicity of cell membrane plays some vital roles in cell survival and cell death, including protection, endocytosis, signaling, and increases in mech. stability during cell division, as well as decrease of shear forces during sepn. of two cells after division, and cell sepn. from tissues for cancer metastasis. Within the membrane, there are specialized domains, known as lipid rafts. A raft can coordinate various signaling pathways. Recent data on the proteomics of lipid rafts/caveolae have highlighted the enigmatic role of various signaling proteins in cancer development. Anal. of these data of raft proteome from various tumors, cancer tissues, and cell lines cultured without and with therapeutic agents, as well as from model rafts revealed that there may be two subsets of raft assemblage in cell membrane. One subset of raft is enriched with cholesterol-sphingomyelins-ganglioside-cav-1/Src/EGFR (hereafter, "chol-raft") that is involved in normal cell signaling, and when dysregulated promotes cell transformation and tumor progression; another subset of raft is enriched with ceramide-sphingomyeline-ganglioside-FAS/Ezrin (hereafter, "cer-raft") that generally promotes apoptosis. In view of this, and to focus insight into the cancer cell physiol. caused by the lipid rafts mediated signals and their receptors, and the downstream transmitters, either proliferative (for example, EGF and EGFR) or death-inducing (for example, FASL and FAS), and the precise roles of some therapeutic drugs and endogenous acid sphingomylenase in this scenario in in situ transformation of "chol-raft" into "cer-raft" are summarized and discussed in this contribution.
- 69Hannun, Y. A.; Obeid, L. M. Principles of Bioactive Lipid Signalling: Lessons from Sphingolipids. Nat. Rev. Mol. Cell Biol. 2008, 9 (2), 139– 150, DOI: 10.1038/nrm2329Google Scholar69https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXovFOntg%253D%253D&md5=0bee4e560d083ef4c43f7d0b591a0997Principles of bioactive lipid signalling: lessons from sphingolipidsHannun, Yusuf A.; Obeid, Lina M.Nature Reviews Molecular Cell Biology (2008), 9 (2), 139-150CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. It has become increasingly difficult to find an area of cell biol. in which lipids do not have important, if not key, roles as signaling and regulatory mols. The rapidly expanding field of bioactive lipids is exemplified by many sphingolipids, such as ceramide, sphingosine, sphingosine-1-phosphate (S1P), ceramide-1-phosphate, and lyso-sphingomyelin, which have roles in the regulation of cell growth, death, senescence, adhesion, migration, inflammation, angiogenesis, and intracellular trafficking. Deciphering the mechanisms of these varied cell functions necessitates an understanding of the complex pathways of sphingolipid metab. and the mechanisms that regulate lipid generation and lipid action.
- 70Wallner, S.; Schmitz, G. Plasmalogens the Neglected Regulatory and Scavenging Lipid Species. Chem. Phys. Lipids 2011, 164 (6), 573– 589, DOI: 10.1016/j.chemphyslip.2011.06.008Google Scholar70https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXpvFCnurc%253D&md5=762062e2e7f960550f689b5fd05952c9Plasmalogens the neglected regulatory and scavenging lipid speciesWallner, Stefan; Schmitz, GerdChemistry and Physics of Lipids (2011), 164 (6), 573-589CODEN: CPLIA4; ISSN:0009-3084. (Elsevier Ltd.)A review. Plasmalogens are a class of phospholipids carrying a vinyl ether bond in sn-1 and an ester bond in sn-2 position of the glycerol backbone. Although they are widespread in all tissues and represent up to 18% of the total phospholipid mass in humans, their physiol. function is still poorly understood. The aim of this review is to give an overview over the current knowledge in plasmalogen biol. and pathol. with an emphasis on neglected aspects of their involvement in neurol. and metabolic diseases. Furthermore a better understanding of plasmalogen biol. in health and disease could also lead to the development of better diagnostic and prognostic biomarkers for vascular and metabolic diseases such as obesity and diabetes mellitus, inflammation, neuro-degeneration and cancer.
- 71Benjamin, D. I.; Cozzo, A.; Ji, X.; Roberts, L. S.; Louie, S. M.; Mulvihill, M. M.; Luo, K.; Nomura, D. K. Ether Lipid Generating Enzyme AGPS Alters the Balance of Structural and Signaling Lipids to Fuel Cancer Pathogenicity. Proc. Natl. Acad. Sci. U. S. A. 2013, 110 (37), 14912– 14917, DOI: 10.1073/pnas.1310894110Google Scholar71https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsFWrsrrP&md5=06519e3d17983acbc3d159d5f19c21afEther lipid generating enzyme AGPS alters the balance of structural and signaling lipids to fuel cancer pathogenicityBenjamin, Daniel I.; Cozzo, Alyssa; Ji, Xiaodan; Roberts, Lindsay S.; Louie, Sharon M.; Mulvihill, Melinda M.; Luo, Kunxin; Nomura, Daniel K.Proceedings of the National Academy of Sciences of the United States of America (2013), 110 (37), 14912-14917,S14912/1-S14912/9CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Aberrant lipid metab. is an established hallmark of cancer cells. In particular, ether lipid levels have been shown to be elevated in tumors, but their specific function in cancer remains elusive. We show here that the metabolic enzyme alkylglyceronephosphate synthase (AGPS), a crit. step in the synthesis of ether lipids, is up-regulated across multiple types of aggressive human cancer cells and primary tumors. We demonstrate that ablation of AGPS in cancer cells results in reduced cell survival, cancer aggressiveness, and tumor growth through altering the balance of ether lipid, fatty acid, eicosanoid, and fatty acid-derived glycerophospholipid metab., resulting in an overall redn. in the levels of several oncogenic signaling lipids. Taken together, our results reveal that AGPS, in addn. to maintaining ether lipids, also controls cellular utilization of fatty acids, favoring the generation of signaling lipids necessary for promoting the aggressive features of cancer.
- 72Piano, V.; Benjamin, D. I.; Valente, S.; Nenci, S.; Marrocco, B.; Mai, A.; Aliverti, A.; Nomura, D. K.; Mattevi, A. Discovery of Inhibitors for the Ether Lipid-Generating Enzyme AGPS as Anti-Cancer Agents. ACS Chem. Biol. 2015, 10 (11), 2589– 2597, DOI: 10.1021/acschembio.5b00466Google ScholarThere is no corresponding record for this reference.
- 73Dean, J. M.; Lodhi, I. J. Structural and Functional Roles of Ether Lipids. Protein Cell 2018, 9 (2), 196– 206, DOI: 10.1007/s13238-017-0423-5Google Scholar73https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXotVKmtrs%253D&md5=f0e2a74a3f4b51f9397531847d92d79dStructural and functional roles of ether lipidsDean, John M.; Lodhi, Irfan J.Protein & Cell (2018), 9 (2), 196-206CODEN: PCREFB; ISSN:1674-800X. (Higher Education Press)Ether lipids, such as plasmalogens, are peroxisome-derived glycerophospholipids in which the hydrocarbon chain at the sn-1 position of the glycerol backbone is attached by an ether bond, as opposed to an ester bond in the more common diacyl phospholipids. This seemingly simple biochem. change has profound structural and functional implications. Notably, the tendency of ether lipids to form non-lamellar inverted hexagonal structures in model membranes suggests that they have a role in facilitating membrane fusion processes. Ether lipids are also important for the organization and stability of lipid raft microdomains, cholesterol-rich membrane regions involved in cellular signaling. In addn. to their structural roles, a subset of ether lipids are thought to function as endogenous antioxidants, and emerging studies suggest that they are involved in cell differentiation and signaling pathways. Here, we review the biol. of ether lipids and their potential significance in human disorders, including neurol. diseases, cancer, and metabolic disorders.
- 74Koivuniemi, A. The Biophysical Properties of Plasmalogens Originating from Their Unique Molecular Architecture. FEBS Lett. 2017, 591 (18), 2700– 2713, DOI: 10.1002/1873-3468.12754Google Scholar74https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXht1GgtbnF&md5=cb7c33b4ba34883680edb584e32205f4The biophysical properties of plasmalogens originating from their unique molecular architectureKoivuniemi, ArtturiFEBS Letters (2017), 591 (18), 2700-2713CODEN: FEBLAL; ISSN:0014-5793. (Wiley-Blackwell)A review. Plasmalogens are a unique class of phospholipids that are present in many organisms. Their presence in cell membranes has intrigued researchers for decades due to their unique mol. structure, namely the vinyl-ether bond at the sn-1 position, and their assocn. with brain related disorders. Apparently, based on their amt. in the cell membranes, their function is to provide exclusive structural and dynamical properties to these complex mol. assemblies. Yet, many of their physiol. roles manifested through their biophys. properties have been challenging to identify. In this review, the biophys. properties of plasmalogens are discussed and compared to other lipid species. The role of plasmalogens is examd. in the context of cell membrane function, and some future directions are given.
- 75Zeno, W. F.; Baul, U.; Snead, W. T.; DeGroot, A. C. M.; Wang, L.; Lafer, E. M.; Thirumalai, D.; Stachowiak, J. C. Synergy between Intrinsically Disordered Domains and Structured Proteins Amplifies Membrane Curvature Sensing. Nat. Commun. 2018, 9, 4152, DOI: 10.1038/s41467-018-06532-3Google Scholar75https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3czntVSrsQ%253D%253D&md5=d04d08deb4eb772d317e0da978f09d14Synergy between intrinsically disordered domains and structured proteins amplifies membrane curvature sensingZeno Wade F; Snead Wilton T; DeGroot Andre C M; Stachowiak Jeanne C; Baul Upayan; Thirumalai D; Wang Liping; Lafer Eileen M; Stachowiak Jeanne CNature communications (2018), 9 (1), 4152 ISSN:.The ability of proteins to sense membrane curvature is essential to cellular function. All known sensing mechanisms rely on protein domains with specific structural features such as wedge-like amphipathic helices and crescent-shaped BAR domains. Yet many proteins that contain these domains also contain large intrinsically disordered regions. Here we report that disordered domains are themselves potent sensors of membrane curvature. Comparison of Monte Carlo simulations with in vitro and live-cell measurements demonstrates that the polymer-like behavior of disordered domains found in endocytic proteins drives them to partition preferentially to convex membrane surfaces, which place fewer geometric constraints on their conformational entropy. Further, proteins containing both structured curvature sensors and disordered regions are more than twice as curvature sensitive as their respective structured domains alone. These findings demonstrate an entropic mechanism of curvature sensing that is independent of protein structure and illustrate how structured and disordered domains can synergistically enhance curvature sensitivity.
- 76Huang, C.; Cao, Z.; Ma, J.; Shen, Y.; Bu, Y.; Khoshaba, R.; Shi, G.; Huang, D.; Liao, D.-F.; Ji, H. AKR1B10 Activates Diacylglycerol (DAG) Second Messenger in Breast Cancer Cells. Mol. Carcinog. 2018, 57 (10), 1300– 1310, DOI: 10.1002/mc.22844Google Scholar76https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXht1ans7rF&md5=c1427c667bdb536e64be522e34c93fe8AKR1B10 activates diacylglycerol (DAG) second messenger in breast cancer cellsHuang, Chenfei; Cao, Zhe; Ma, Jun; Shen, Yi; Bu, Yiwen; Khoshaba, Ramina; Shi, Guiyuan; Huang, Dan; Liao, Duan-Fang; Ji, Haitao; Jin, Junfei; Cao, DeliangMolecular Carcinogenesis (2018), 57 (10), 1300-1310CODEN: MOCAE8; ISSN:0899-1987. (Wiley-Blackwell)Aldo-keto reductase 1B10 (AKR1B10) is upregulated in breast cancer and promotes tumor growth and metastasis. However, little is known of the mol. mechanisms of action. Herein we report that AKR1B10 activates lipid second messengers to stimulate cell proliferation. Our data showed that ectopic expression of AKR1B10 in breast cancer cells MCF-7 promoted lipogenesis and enhanced levels of lipid second messengers, including phosphatidylinositol bisphosphate (PIP2), diacylglycerol (DAG), and inositol triphosphate (IP3). In contrast, silencing of AKR1B10 in breast cancer cells BT-20 and colon cancer cells HCT-8 led to decrease of these lipid messengers. Qual. analyses by liq. chromatog.-mass spectrum (LC-MS) revealed that AKR1B10 regulated the cellular levels of total DAG and majority of subspecies. This in turn modulated the phosphorylation of protein kinase C (PKC) isoforms PKCδ (Thr505), PKCμ (Ser744/748), and PKCα/βII (Thr638/641) and activity of the PKC-mediated c-Raf/MEK/ERK signaling cascade. A pan inhibitor of PKC (Go6983) blocked ERK1/2 activation by AKR1B10. In these cells, phospho-p90RSK, phospho-MSK, and Cyclin D1 expression was increased by AKR1B10, and pharmacol. inhibition of the ERK signaling cascade with MEK1/2 inhibitors U0126 and PD98059 eradicated induction of phospho-p90RSK, phospho-MSK, and Cyclin D1. In breast cancer cells, AKR1B10 promoted the clonogenic growth and proliferation of breast cancer cells in two-dimension (2D) and three-dimension (3D) cultures and tumor growth in immunodeficient female nude mice through activation of the PKC/ERK pathway. These data suggest that AKR1B10 stimulates breast cancer cell growth and proliferation through activation of DAG-mediated PKC/ERK signaling pathway.
- 77Kunitake, J. A. M. R.; Choi, S.; Nguyen, K. X.; Lee, M. M.; He, F.; Sudilovsky, D.; Morris, P. G.; Jochelson, M. S.; Hudis, C. A.; Muller, D. A. Correlative Imaging Reveals Physiochemical Heterogeneity of Microcalcifications in Human Breast Carcinomas. J. Struct. Biol. 2018, 202 (1), 25– 34, DOI: 10.1016/j.jsb.2017.12.002Google ScholarThere is no corresponding record for this reference.
Cited By
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by ACS Publications if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
This article is cited by 45 publications.
- Peng Zhou, Yu Xiao, Xin Zhou, Jinghui Fang, Jingwen Zhang, Jianjun Liu, Ling Guo, Jiuhong Zhang, Ning Zhang, Ke Chen, Chao Zhao. Mapping Spatiotemporal Heterogeneity in Multifocal Breast Tumor Progression by Noninvasive Ultrasound Elastography-Guided Mass Spectrometry Imaging Strategy. JACS Au 2024, 4
(2)
, 465-475. https://doi.org/10.1021/jacsau.3c00589
- Hao Liu, Hui Jiang, Xiaohui Liu, Xuemei Wang. Tunable Nanomaterials of Intracellular Crystallization for In Situ Biolabeling and Biomedical Imaging. Chemical & Biomedical Imaging 2023, 1
(9)
, 767-784. https://doi.org/10.1021/cbmi.3c00021
- Jing Chen, Peisi Xie, Pengfei Wu, Zian Lin, Yu He, Zongwei Cai. Spatial Metabolomics and Lipidomics Reveal the Mechanisms of the Enhanced Growth of Breast Cancer Cell Spheroids Exposed to Triclosan. Environmental Science & Technology 2023, 57
(29)
, 10542-10553. https://doi.org/10.1021/acs.est.3c01746
- Hao Jia, Shuhua Yue. Stimulated Raman Scattering Imaging Sheds New Light on Lipid Droplet Biology. The Journal of Physical Chemistry B 2023, 127
(11)
, 2381-2394. https://doi.org/10.1021/acs.jpcb.3c00038
- Javier Plou, Beatriz Molina-Martínez, Clara García-Astrain, Judith Langer, Isabel García, Amaia Ercilla, Govindaraj Perumal, Arkaitz Carracedo, Luis M. Liz-Marzán. Nanocomposite Scaffolds for Monitoring of Drug Diffusion in Three-Dimensional Cell Environments by Surface-Enhanced Raman Spectroscopy. Nano Letters 2021, 21
(20)
, 8785-8793. https://doi.org/10.1021/acs.nanolett.1c03070
- Fernando Tobias, Julie C. McIntosh, Gabriel J. LaBonia, Matthew W. Boyce, Matthew R. Lockett, Amanda B. Hummon. Developing a Drug Screening Platform: MALDI-Mass Spectrometry Imaging of Paper-Based Cultures. Analytical Chemistry 2019, 91
(24)
, 15370-15376. https://doi.org/10.1021/acs.analchem.9b03536
- Cha Yang, Gwang Bin Lee, Ling Hao, Fenghua Hu. TMEM106B deficiency leads to alterations in lipid metabolism and obesity in the TDP-43Q331K knock-in mouse model. Communications Biology 2025, 8
(1)
https://doi.org/10.1038/s42003-025-07752-2
- Jiaojiao Xu, Wanxia Fang, Huanhuan Zhou, Ruiyuan Jiang, Zhanhong Chen, Xiaojia Wang. Application and progress of 3D tumor models in breast cancer. Biotechnology and Bioengineering 2025, 122
(1)
, 30-43. https://doi.org/10.1002/bit.28860
- Takahisa Matsuzaki, Mai Fujii, Hayata Noro, Shodai Togo, Mami Watanabe, Masami Suganuma, Shivani Sharma, Naritaka Kobayashi, Ryuzo Kawamura, Seiichiro Nakabayashi, Hiroshi Y. Yoshikawa. Simultaneous visualization of membrane fluidity and morphology defines adhesion signatures of cancer cells. Proceedings of the National Academy of Sciences 2024, 121
(50)
https://doi.org/10.1073/pnas.2412914121
- Anmi Jose, Pallavi Kulkarni, Jaya Thilakan, Murali Munisamy, Anvita Gupta Malhotra, Jitendra Singh, Ashok Kumar, Vivek M. Rangnekar, Neha Arya, Mahadev Rao. Integration of pan-omics technologies and three-dimensional in vitro tumor models: an approach toward drug discovery and precision medicine. Molecular Cancer 2024, 23
(1)
https://doi.org/10.1186/s12943-023-01916-6
- Xiao-Yu Zheng, Yao Lv, Ling-Yan Xu, Dong-Ming Zhou, Lan Yu, Zheng-Yan Zhao. A novel approach for breast cancer treatment: the multifaceted antitumor effects of rMeV-Hu191. Hereditas 2024, 161
(1)
https://doi.org/10.1186/s41065-024-00337-9
- Daan J. Kloosterman, Martina Farber, Menno Boon, Johanna Erbani, Leila Akkari. Protocol for studying macrophage lipid crosstalk with murine tumor cells. STAR Protocols 2024, 5
(4)
, 103421. https://doi.org/10.1016/j.xpro.2024.103421
- Patrik Sioris, Meri Mäkelä, Anton Kontunen, Markus Karjalainen, Antti Vehkaoja, Niku Oksala, Antti Roine. Identification of Phospholipids Relevant to Cancer Tissue Using Differential Ion Mobility Spectrometry. International Journal of Molecular Sciences 2024, 25
(20)
, 11002. https://doi.org/10.3390/ijms252011002
- Marjolein M. G. Kes, Celia R. Berkers, Jarno Drost. Bridging the gap: advancing cancer cell culture to reveal key metabolic targets. Frontiers in Oncology 2024, 14 https://doi.org/10.3389/fonc.2024.1480613
- Senol Dogan, Jenny Leopold, Daniel T. Hoffmann, Hans Kubitschke, Eliane Blauth, Carlotta Ficorella, Amelie Zschau, Jürgen Schiller, Josef A. Käs. Identification of Lipid Droplet-Associated Genes in Breast Cancer Patients. Lipidology 2024, 1
(1)
, 52-74. https://doi.org/10.3390/lipidology1010005
- Fahad Khan, Deena Elsori, Meenakshi Verma, Shivam Pandey, Safia Obaidur Rab, Samra Siddiqui, Nadiyah M. Alabdallah, Mohd Saeed, Pratibha Pandey. Unraveling the intricate relationship between lipid metabolism and oncogenic signaling pathways. Frontiers in Cell and Developmental Biology 2024, 12 https://doi.org/10.3389/fcell.2024.1399065
- Jiun-Han Lin, Tien-Wei Hsu, Wei-Chung Cheng, Chen-Chi Liu, Anna Fen-Yau Li, Mien-Chie Hung, Han-Shui Hsu, Shih-Chieh Hung. TAG Synthesis and Oxidation Activated by YAP/TEAD Confers ROS Resistance in Cancer Stem Cells. 2024https://doi.org/10.21203/rs.3.rs-3936782/v1
- Shanjing Wen, Xinyi Tu, Qingce Zang, Ying Zhu, Limei Li, Ruiping Zhang, Zeper Abliz. Liquid chromatography–mass spectrometry‐based metabolomics and fluxomics reveals the metabolic alterations in glioma U87MG multicellular tumor spheroids versus two‐dimensional cell cultures. Rapid Communications in Mass Spectrometry 2024, 38
(2)
https://doi.org/10.1002/rcm.9670
- Lara Pierantoni, Rui L. Reis, Joana Silva-Correia, Joaquim M. Oliveira, Susan Heavey. Spatial -omics technologies: the new enterprise in 3D breast cancer models. Trends in Biotechnology 2023, 41
(12)
, 1488-1500. https://doi.org/10.1016/j.tibtech.2023.07.003
- Rachel C. Avard, Megan L. Broad, Fereshteh Zandkarimi, Alexander J. Devanny, Joseph L. Hammer, Karen Yu, Asja Guzman, Laura J. Kaufman. DISC-3D: dual-hydrogel system enhances optical imaging and enables correlative mass spectrometry imaging of invading multicellular tumor spheroids. Scientific Reports 2023, 13
(1)
https://doi.org/10.1038/s41598-023-38699-1
- Amit Cohen, Lotem Gotnayer, Sahar Gal, Dina Aranovich, Netta Vidavsky. Multicellular spheroids containing synthetic mineral particles: an advanced 3D tumor model system to investigate breast precancer malignancy potential according to the mineral type. Journal of Materials Chemistry B 2023, 11
(33)
, 8033-8045. https://doi.org/10.1039/D3TB00439B
- Tiantian Wang, Judith Desmet, Elisabet Pérez-Albaladejo, Cinta Porte. Development of fish liver PLHC-1 spheroids and its applicability to investigate the toxicity of plastic additives. Ecotoxicology and Environmental Safety 2023, 259 , 115016. https://doi.org/10.1016/j.ecoenv.2023.115016
- Saman Khalil, Uroosa Nawaz, Zubariah, Zohaib Mushtaq, Saad Arif, Muhammad Zia ur Rehman, Muhammad Farrukh Qureshi, Abdul Malik, Adham Aleid, Khalid Alhussaini. Enhancing Ductal Carcinoma Classification Using Transfer Learning with 3D U-Net Models in Breast Cancer Imaging. Applied Sciences 2023, 13
(7)
, 4255. https://doi.org/10.3390/app13074255
- Xiaotian Hao, Sheng Miao, Zhiquan Li, Taoran Wang, Baobao Xue, Jingdi Chen, Chunxing Xian, Long Bi. 3D printed structured porous hydrogel promotes osteogenic differentiation of BMSCs. Materials & Design 2023, 227 , 111729. https://doi.org/10.1016/j.matdes.2023.111729
- Amit Cohen, Lotem Gotnayer, Dina Aranovich, Netta Vidavsky. Microcalcifications can either trigger or suppress breast precancer malignancy potential according to the mineral type in a 3D tumor model. 2023https://doi.org/10.1101/2023.02.20.529220
- Peisi Xie, Jinghui Zhang, Pengfei Wu, Yongjiang Wu, Yanjun Hong, Jianing Wang, Zongwei Cai. Multicellular tumor spheroids bridge the gap between two-dimensional cancer cells and solid tumors: The role of lipid metabolism and distribution. Chinese Chemical Letters 2023, 34
(2)
, 107349. https://doi.org/10.1016/j.cclet.2022.03.072
- N. Leitner, J. Hlavatý, R. Ertl, S. Gabner, A. Fuchs-Baumgartinger, Ingrid Walter. Lipid droplets and perilipins in canine osteosarcoma. Investigations on tumor tissue, 2D and 3D cell culture models. Veterinary Research Communications 2022, 46
(4)
, 1175-1193. https://doi.org/10.1007/s11259-022-09975-8
- Yichen Zhu, Elliot Kang, Matthew Wilson, Taylor Basso, Evelynn Chen, Yanqi Yu, Yan-Ruide Li. 3D Tumor Spheroid and Organoid to Model Tumor Microenvironment for Cancer Immunotherapy. Organoids 2022, 1
(2)
, 149-167. https://doi.org/10.3390/organoids1020012
- Senol Dogan, Jenny Leopold, Daniel T. Hoffmann, Hans Kubitschke, Eliane Blauth, Carlotta Ficorella, Amelie Zschau, Jürgen Schiller, Josef A. Käs. Identification of lipid droplet-associated genes in breast cancer patients. 2022https://doi.org/10.21203/rs.3.rs-2157661/v1
- Muhammad Azaz Farooq, Zhaoxuan Gong, Yu Liu, Muhammad Zubair, Arslan Manzoor, Guodong Zhang, , , , . Breast cancer detection from ultrasound images using attention U-nets model. 2022, 49. https://doi.org/10.1117/12.2643599
- Alana Smith, Xueyuan Cao, Qingqing Gu, Ernestine Kubi Amos-Abanyie, Elizabeth A. Tolley, Gregory Vidal, Beverly Lyn-Cook, Athena Starlard-Davenport. Characterization of the Metabolome of Breast Tissues from Non-Hispanic Black and Non-Hispanic White Women Reveals Correlations between Microbial Dysbiosis and Enhanced Lipid Metabolism Pathways in Triple-Negative Breast Tumors. Cancers 2022, 14
(17)
, 4075. https://doi.org/10.3390/cancers14174075
- Fernando Tobias, Amanda B. Hummon. Lipidomic comparison of 2D and 3D colon cancer cell culture models. Journal of Mass Spectrometry 2022, 57
(8)
https://doi.org/10.1002/jms.4880
- Mauriane Maja, Danahe Mohammed, Andra C. Dumitru, Sandrine Verstraeten, Maxime Lingurski, Marie-Paule Mingeot-Leclercq, David Alsteens, Donatienne Tyteca. Surface cholesterol-enriched domains specifically promote invasion of breast cancer cell lines by controlling invadopodia and extracellular matrix degradation. Cellular and Molecular Life Sciences 2022, 79
(8)
https://doi.org/10.1007/s00018-022-04426-8
- Xiaobo Zhou, Ke Zhang, Chaojie Yang, Yuetian Pei, Lingfeng Zhao, Xiaoxia Kang, Zhenhua Li, Fuyou Li, Yuling Qin, Li Wu. Ultrabright and Highly Polarity‐Sensitive NIR‐I/NIR‐II Fluorophores for the Tracking of Lipid Droplets and Staging of Fatty Liver Disease. Advanced Functional Materials 2022, 32
(12)
https://doi.org/10.1002/adfm.202109929
- Ashley V. Ward, Steven M. Anderson, Carol A. Sartorius. Advances in Analyzing the Breast Cancer Lipidome and Its Relevance to Disease Progression and Treatment. Journal of Mammary Gland Biology and Neoplasia 2021, 26
(4)
, 399-417. https://doi.org/10.1007/s10911-021-09505-3
- Isidora Tošić, David A. Frank. STAT3 as a mediator of oncogenic cellular metabolism: Pathogenic and therapeutic implications. Neoplasia 2021, 23
(12)
, 1167-1178. https://doi.org/10.1016/j.neo.2021.10.003
- Yanyan Chen, Tao Wang, Peisi Xie, Yuanyuan Song, Jianing Wang, Zongwei Cai. Mass spectrometry imaging revealed alterations of lipid metabolites in multicellular tumor spheroids in response to hydroxychloroquine. Analytica Chimica Acta 2021, 1184 , 339011. https://doi.org/10.1016/j.aca.2021.339011
- Mengting ZHANG, Yulu ZHANG, Haojiang WANG, Ning LI, Bo LI, Hong XIAO, Wei BIAN, Zongwei CAI. 质谱成像技术及其在乳腺癌研究中的应用. Chinese Journal of Chromatography 2021, 39
(6)
, 578-587. https://doi.org/10.3724/SP.J.1123.2020.10005
- Lorenzo Scipioni, Alessandro Rossetta, Giulia Tedeschi, Enrico Gratton. Phasor S-FLIM: a new paradigm for fast and robust spectral fluorescence lifetime imaging. Nature Methods 2021, 18
(5)
, 542-550. https://doi.org/10.1038/s41592-021-01108-4
- Vera M. Pieters, Ileana L. Co, Nila C. Wu, Alison P. McGuigan. Applications of Omics Technologies for Three-Dimensional
In Vitro
Disease Models. Tissue Engineering Part C: Methods 2021, 27
(3)
, 183-199. https://doi.org/10.1089/ten.tec.2020.0300
- Vaishali Aggarwal, Oshin Miranda, Paul A. Johnston, Shilpa Sant. Three dimensional engineered models to study hypoxia biology in breast cancer. Cancer Letters 2020, 490 , 124-142. https://doi.org/10.1016/j.canlet.2020.05.030
- Sandip Kumar Patel, Bhawana George, Vineeta Rai. Artificial Intelligence to Decode Cancer Mechanism: Beyond Patient Stratification for Precision Oncology. Frontiers in Pharmacology 2020, 11 https://doi.org/10.3389/fphar.2020.01177
- Tarapong Srisongkram, Natthida Weerapreeyakul, Kanjana Thumanu. Evaluation of Melanoma (SK-MEL-2) Cell Growth between Three-Dimensional (3D) and Two-Dimensional (2D) Cell Cultures with Fourier Transform Infrared (FTIR) Microspectroscopy. International Journal of Molecular Sciences 2020, 21
(11)
, 4141. https://doi.org/10.3390/ijms21114141
- Jeremy P. Koelmel, Michael P. Napolitano, Candice Z. Ulmer, Vasilis Vasiliou, Timothy J. Garrett, Richard A. Yost, M. N. V. Prasad, Krystal J. Godri Pollitt, John A. Bowden. Environmental lipidomics: understanding the response of organisms and ecosystems to a changing world. Metabolomics 2020, 16
(5)
https://doi.org/10.1007/s11306-020-01665-3
- Finnur Freyr Eiriksson, Martha Kampp Nøhr, Margarida Costa, Sigridur Klara Bödvarsdottir, Helga Margret Ögmundsdottir, Margret Thorsteinsdottir, . Lipidomic study of cell lines reveals differences between breast cancer subtypes. PLOS ONE 2020, 15
(4)
, e0231289. https://doi.org/10.1371/journal.pone.0231289
- Ieva Palubeckaitė, Lucy Crooks, David P. Smith, Laura M. Cole, Heijs Bram, Christine Le Maitre, Malcolm R. Clench, Neil A. Cross. Mass spectrometry imaging of endogenous metabolites in response to doxorubicin in a novel 3D osteosarcoma cell culture model. Journal of Mass Spectrometry 2020, 55
(4)
https://doi.org/10.1002/jms.4461
- Bhagyashree D. Rao, Hirak Chakraborty, Arunima Chaudhuri, Amitabha Chattopadhyay. Differential sensitivity of pHLIP to ester and ether lipids. Chemistry and Physics of Lipids 2020, 226 , 104849. https://doi.org/10.1016/j.chemphyslip.2019.104849
- Nguyen Hoang Anh, Nguyen Phuoc Long, Sun Jo Kim, Jung Eun Min, Sang Jun Yoon, Hyung Min Kim, Eugine Yang, Eun Sook Hwang, Jeong Hill Park, Soon-Sun Hong, Sung Won Kwon. Steroidomics for the Prevention, Assessment, and Management of Cancers: A Systematic Review and Functional Analysis. Metabolites 2019, 9
(10)
, 199. https://doi.org/10.3390/metabo9100199
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
Abstract
Figure 1
Figure 1. Relation between lipid amounts and the malignancy potential of breast cancer cells in 2D and 3D. Oil-Red-O lipid staining of cell lines from the MCF10A-based breast cancer progression series cultured in 2D (a–f) and as multicellular spheroids (g–l). (a, d, g, j) Nonmalignant MCF10A cells; (b, e, h, k) precancer MCF10DCIS.com cells; (c, f, i, l) invasive MCF10CA1a cells. Lipids are stained red, and cell nuclei are stained purple. Arrows: lipid droplet aggregates close to cell nuclei. For a high resolution version of Figure 1 see Supporting Data.
Figure 2
Figure 2. Lipid profiles in 2D and 3D culture of precancer and invasive cells detected using LCMS. (a) Heatmap showing the clustering of lipid species in 2D and 3D cultures of MCF10DCIS.com (precancer) and MCF10CA1a cells (invasive). Color bar indicates the scaled distance from the row mean of the normalized transformed data. For assessment of the variation within each group, the biological replicates for each condition are shown. For the 2D samples, each group consists of three biological replicates, and for the 3D samples, each group consists of four biological replicates. Lipid classes are color coded as indicated. Coenzyme Q10 is shown in white. To the right, representative example lipid structures of the color-coded lipid classes in part a are shown. (b) Lipid class distribution in precancer and invasive 2D and 3D cultures as detected with LCMS, calculated from areas in LCMS normalized per microgram of protein in the sample and presented as a fraction of total lipids in the sample. Error bars are the standard error of the mean. 34 Cs = summed acyl chain and sphingoid base chain lengths of 34 carbons. See Supporting Figure S2 for a version of this figure with individual lipid identifiers associated with the heat map.
Figure 3
Figure 3. Heat map showing clustering of the 25 lipid species with the most significant changes, selected by t-test, across 3D cultures of MCF10DCIS.com (precancer) and MCF10CA1a (invasive) cells as detected with LCMS. Each group consists of four biological replicates. So(d18:0) = sphinganine. Color bar indicates the scaled distance from the row mean of the normalized transformed data.
Figure 4
Figure 4. Characterization of the lipid droplets formed in 3D multicellular spheroids of malignant cell lines. (a, b, e) Precancer spheroid cross sections. (c, d, g) Invasive spheroid cross sections. Asterisks show the necrotic core. (a, c) Oil-Red-O lipid staining showing lipid accumulation in the necrotic core area of the spheroids as well as lipid droplets in the invasive spheroid (arrows). (b, d) Cross-polarized light images of the spheroid cross sections showing birefringence (arrows in part d point at the same areas as in part c). (e, g) Overlaid spatial distribution maps of key basis spectra resultant from NMF multivariate analysis of Raman mapping. (f) Corresponding NMF basis spectra (precancer: black, invasive: gray). The precancer overlay map (e) shows an increase in lipid content toward the spheroid center, while the invasive overlay map confirms the birefringent regions are lipid-rich droplets (cyan). Arrows indicate the same bodies as in parts c and d.
Figure 5
Figure 5. Spatial characterization of DCIS (precancer) from human tissue. (a) H&E stained cryo-section showing the duct cross section containing cells (purple), necrosis (dark pink and purple), and surrounding stromal tissue (pink). (b) Confocal Raman mapping and component analysis of a serial section from the same duct in part a, showing the spatial distribution of the tissue components. Lipids, cyan; collagen, green; protein/cytochrome c, magenta; noncollagenous proteins, blue; cells, yellow. A distinct lipid signature consistent with neutral lipids, including cholesterol ester(s), is observed in the necrotic regions and occurs in spatially discrete domains (arrow). (c) The corresponding Raman component spectra for the Raman map in part b.
Figure 6
Figure 6. A scheme describing the effects of dimensionality (top left) and the cell malignancy potential (bottom left) on lipid species amounts in the MCF10A breast cancer in vitro model. Lipid-rich areas (red) are observed in the necrotic core with decreasing amounts toward the spheroid periphery alongside large lipid droplets at the periphery. The observed increase in the ratio of neutral to membrane lipid species in 3D versus 2D may be related to large lipid droplet formation through TG synthesis and/or lipid droplet coalescence (top right). The observed increase in sphingolipid species in invasive versus precancer spheroids could be due to enhancement of the de novo sphingolipid synthesis pathway (bottom right, adapted from Ogretmen (46)). Species of lipids increased out of total lipids in 3D invasive spheroids are underlined. CoA = coenzyme A, SPT = serine palmitoyltransferase, KDSR = 3-ketosphinganine reductase, CERS = (dihydro)ceramide synthases, DES = dihydroceramide desaturase, SMS = sphingomyelin synthase, CDase = ceramidases.
References
This article references 77 other publications.
- 1DeBerardinis, R. J.; Chandel, N. S. Fundamentals of Cancer Metabolism. Sci. Adv. 2016, 2 (5), e1600200– e1600200, DOI: 10.1126/sciadv.1600200There is no corresponding record for this reference.
- 2Furuta, E.; Pai, S. K.; Zhan, R.; Bandyopadhyay, S.; Watabe, M.; Mo, Y.-Y.; Hirota, S.; Hosobe, S.; Tsukada, T.; Miura, K. Fatty Acid Synthase Gene Is Up-Regulated by Hypoxia via Activation of Akt and Sterol Regulatory Element Binding Protein-1. Cancer Res. 2008, 68 (4), 1003– 1011, DOI: 10.1158/0008-5472.CAN-07-2489There is no corresponding record for this reference.
- 3Koizume, S.; Miyagi, Y. Lipid Droplets : A Key Cellular Organelle Associated with Cancer Cell Survival under Normoxia and Hypoxia. Int. J. Mol. Sci. 2016, 17, 1– 23, DOI: 10.3390/ijms17091430There is no corresponding record for this reference.
- 4Kamphorst, J. J.; Cross, J. R.; Fan, J.; de Stanchina, E.; Mathew, R.; White, E. P.; Thompson, C. B.; Rabinowitz, J. D. Hypoxic and Ras-Transformed Cells Support Growth by Scavenging Unsaturated Fatty Acids from Lysophospholipids. Proc. Natl. Acad. Sci. U. S. A. 2013, 110 (22), 8882– 8887, DOI: 10.1073/pnas.13072371104https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFalsbnN&md5=4deaa2c9fa2b5c2b482cfef5cabfb5eaHypoxic and Ras-transformed cells support growth by scavenging unsaturated fatty acids from lysophospholipidsKamphorst, Jurre J.; Cross, Justin R.; Fan, Jing; de Stanchina, Elisa; Mathew, Robin; White, Eileen P.; Thompson, Craig B.; Rabinowitz, Joshua D.Proceedings of the National Academy of Sciences of the United States of America (2013), 110 (22), 8882-8887, S8882/1-S8882/5CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Cancer cell growth requires fatty acids to replicate cellular membranes. The kinase Akt is known to up-regulate fatty acid synthesis and desatn., which is carried out by the oxygen-consuming enzyme stearoyl-CoA desaturase (SCD)1. We used 13C tracers and lipidomics to probe fatty acid metab., including desatn., as a function of oncogene expression and oxygen availability. During hypoxia, flux from glucose to acetyl-CoA decreases, and the fractional contribution of glutamine to fatty acid synthesis increases. In addn., we find that hypoxic cells bypass de novo lipogenesis, and thus, both the need for acetyl-CoA and the oxygen-dependent SCD1-reaction, by scavenging serum fatty acids. The preferred substrates for scavenging are phospholipids with one fatty acid tail (lysophospholipids). Hypoxic reprogramming of de novo lipogenesis can be reproduced in normoxic cells by Ras activation. This renders Ras-driven cells, both in culture and in allografts, resistant to SCD1 inhibition. Thus, a mechanism by which oncogenic Ras confers metabolic robustness is through lipid scavenging.
- 5Counihan, J. L.; Grossman, E. A.; Nomura, D. K. Cancer Metabolism: Current Understanding and Therapies. Chem. Rev. 2018, 118 (14), 6893– 6923, DOI: 10.1021/acs.chemrev.7b007755https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtFyisrvL&md5=4239fa36e326abc1abbe5269522aeeb0Cancer Metabolism: Current Understanding and TherapiesCounihan, Jessica L.; Grossman, Elizabeth A.; Nomura, Daniel K.Chemical Reviews (Washington, DC, United States) (2018), 118 (14), 6893-6923CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review. Dysregulation of cancer cell metab. contributes to abnormal cell growth, the biol. endpoint of cancer. We review here numerous affected oncogenes and metabolic pathways common in cancer, and how they contribute to cancer pathogenesis and malignancy. This review also discusses various pharmacol. manipulations that take advantage of these metabolic abnormalities, and the current targeted therapies that have arisen from this research.
- 6Santos, C. R.; Schulze, A. Lipid Metabolism in Cancer. FEBS J. 2012, 279 (15), 2610– 2623, DOI: 10.1111/j.1742-4658.2012.08644.x6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtVWhurvI&md5=53fa5afbd3de338e22986d6869a60769Lipid metabolism in cancerSantos, Claudio R.; Schulze, AlmutFEBS Journal (2012), 279 (15), 2610-2623CODEN: FJEOAC; ISSN:1742-464X. (Wiley-Blackwell)A review. Lipids form a diverse group of water-insol. mols. that include triacylglycerides, phosphoglycerides, sterols and sphingolipids. They play several important roles at cellular and organismal levels. Fatty acids are the major building blocks for the synthesis of triacylglycerides, which are mainly used for energy storage. Phosphoglycerides, together with sterols and sphingolipids, represent the major structural components of biol. membranes. Lipids can also have important roles in signaling, functioning as second messengers and as hormones. There is increasing evidence that cancer cells show specific alterations in different aspects of lipid metab. These alterations can affect the availability of structural lipids for the synthesis of membranes, the synthesis and degrdn. of lipids that contribute to energy homeostasis and the abundance of lipids with signaling functions. Changes in lipid metab. can affect numerous cellular processes, including cell growth, proliferation, differentiation and motility. This review will examine some of the alterations in lipid metab. that have been reported in cancer, at both cellular and organismal levels, and discuss how they contribute to different aspects of tumorigenesis.
- 7Schulze, A.; Harris, A. L. How Cancer Metabolism Is Tuned for Proliferation and Vulnerable to Disruption. Nature 2012, 491 (7424), 364– 373, DOI: 10.1038/nature117067https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhs1Ogt77F&md5=73cdad5f0a8652993ff162b9b1ba6fe7How cancer metabolism is tuned for proliferation and vulnerable to disruptionSchulze, Almut; Harris, Adrian L.Nature (London, United Kingdom) (2012), 491 (7424), 364-373CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)A review. Cancer metab. has received a substantial amt. of interest over the past decade. The advances in anal. tools have, along with the rapid progress of cancer genomics, generated an increasingly complex understanding of metabolic reprogramming in cancer. As numerous connections between oncogenic signalling pathways and metabolic activities emerge, the importance of metabolic reprogramming in cancer is being increasingly recognized. The identification of metabolic weaknesses of cancer cells has been used to create strategies for treating cancer, but there are still challenges to be faced in bringing the drugs that target cancer metab. to the clinic.
- 8Thiam, A. R.; Beller, M. The Why, When and How of Lipid Droplet Diversity. J. Cell Sci. 2017, 130 (2), 315– 324, DOI: 10.1242/jcs.1920218https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtF2gsL7M&md5=fe4c29e481d35619b6442d58a2375450The why, when and how of lipid droplet diversityThiam, Abdou Rachid; Beller, MathiasJournal of Cell Science (2017), 130 (2), 315-324CODEN: JNCSAI; ISSN:1477-9137. (Company of Biologists Ltd.)Lipid droplets are the universal cellular organelles for the transient or long-term storage of lipids. The no., size and compn. of lipid droplets vary greatly within cells in a homogenous population as well as in different cell types. The variability of intracellular lipid-storage organelles reflects the diversification of lipid droplet compn. and function. Lipid droplet diversification results, for example, in two cellular lipid droplet populations that are prone to diminish and grow, resp. The aberrant accumulation or depletion of lipids are hallmarks or causes of various human pathologies. Thus, a better understanding of the origins of lipid droplet diversification is not only a fascinating cell biol. question but also potentially serves to improve comprehension of pathologies that entail the accumulation of lipids. This Commentary covers the lipid droplet life cycle and highlights the early steps during lipid droplet biogenesis, which we propose to be the potential driving forces of lipid droplet diversification.
- 9Tirinato, L.; Pagliari, F.; Limongi, T.; Marini, M.; Falqui, A.; Seco, J.; Candeloro, P.; Liberale, C.; Di Fabrizio, E. An Overview of Lipid Droplets in Cancer and Cancer Stem Cells. Stem Cells Int. 2017, 2017, 1– 17, DOI: 10.1155/2017/1656053There is no corresponding record for this reference.
- 10Wu, H.; Volponi, J. V.; Oliver, A. E.; Parikh, A. N.; Simmons, B. A.; Singh, S. In Vivo Lipidomics Using Single-Cell Raman Spectroscopy. Proc. Natl. Acad. Sci. U. S. A. 2011, 108 (9), 3809– 3814, DOI: 10.1073/pnas.100904310810https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXivFOqt78%253D&md5=40bb2127d3598103a795f761dab09c9fIn vivo lipidomics using single-cell raman spectroscopyWu, Huawen; Volponi, Joanne V.; Oliver, Ann E.; Parikh, Atul N.; Simmons, Blake A.; Singh, SeemaProceedings of the National Academy of Sciences of the United States of America (2011), 108 (9), 3809-3814, S3809/1-S3809/11CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)We describe a method for direct, quant., in vivo lipid profiling of oil-producing microalgae using single-cell laser-trapping Raman spectroscopy. This approach is demonstrated in the quant. detn. of the degree of unsatn. and transition temps. of constituent lipids within microalgae. These properties are important markers for detg. engine compatibility and performance metrics of algal biodiesel. We show that these factors can be directly measured from a single living microalgal cell held in place with an optical trap while simultaneously collecting Raman data. Cellular response to different growth conditions is monitored in real time. Our approach circumvents the need for lipid extn. and anal. that is both slow and invasive. Furthermore, this technique yields real-time chem. information in a label-free manner, thus eliminating the limitations of impermeability, toxicity, and specificity of the fluorescent probes common in currently used protocols. Although the single-cell Raman spectroscopy demonstrated here is focused on the study of the microalgal lipids with biofuel applications, the anal. capability and quantitation algorithms demonstrated are applicable to many different organisms and should prove useful for a diverse range of applications in lipidomics.
- 11Balog, J.; Sasi-Szabó, L.; Kinross, J.; Lewis, M. R.; Muirhead, L. J.; Veselkov, K.; Mirnezami, R.; Dezső, B.; Damjanovich, L.; Darzi, A. Intraoperative Tissue Identification Using Rapid Evaporative Ionization Mass Spectrometry. Sci. Transl. Med. 2013, 5 (194), 194ra93, DOI: 10.1126/scitranslmed.3005623There is no corresponding record for this reference.
- 12Wenk, M. R. Lipidomics: New Tools and Applications. Cell 2010, 143 (6), 888– 895, DOI: 10.1016/j.cell.2010.11.03312https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsFGgsrbI&md5=83b0ba44ac3132aa385a24ffa348d7f8Lipidomics: New Tools and ApplicationsWenk, Markus R.Cell (Cambridge, MA, United States) (2010), 143 (6), 888-895CODEN: CELLB5; ISSN:0092-8674. (Cell Press)A review. Once viewed simply as a reservoir for carbon storage, lipids are no longer cast as bystanders in the drama of biol. systems. The emerging field of lipidomics is driven by technol., most notably mass spectrometry, but also by complementary approaches for the detection and characterization of lipids and their biosynthetic enzymes in living cells. The development of these integrated tools promises to greatly advance our understanding of the diverse biol. roles of lipids.
- 13Bergholt, M. S.; Serio, A.; McKenzie, J. S.; Boyd, A.; Soares, R. F.; Tillner, J.; Chiappini, C.; Wu, V.; Dannhorn, A.; Takats, Z. Correlated Heterospectral Lipidomics for Biomolecular Profiling of Remyelination in Multiple Sclerosis. ACS Cent. Sci. 2018, 4 (1), 39– 51, DOI: 10.1021/acscentsci.7b0036713https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXitVejurnF&md5=1a0a2def89f5d9768b22886babe0698cCorrelated Heterospectral Lipidomics for Biomolecular Profiling of Remyelination in Multiple SclerosisBergholt, Mads S.; Serio, Andrea; McKenzie, James S.; Boyd, Amanda; Soares, Renata F.; Tillner, Jocelyn; Chiappini, Ciro; Wu, Vincen; Dannhorn, Andreas; Takats, Zoltan; Williams, Anna; Stevens, Molly M.ACS Central Science (2018), 4 (1), 39-51CODEN: ACSCII; ISSN:2374-7951. (American Chemical Society)Analyzing lipid compn. and distribution within the brain is important to study white matter pathologies that present focal demyelination lesions, such as multiple sclerosis. Some lesions can endogenously re-form myelin sheaths. Therapies aim to enhance this repair process in order to reduce neurodegeneration and disability progression in patients. In this context, a lipidomic anal. providing both precise mol. classification and well-defined localization is crucial to detect changes in myelin lipid content. Here we develop a correlated heterospectral lipidomic (HSL) approach based on coregistered Raman spectroscopy, desorption electrospray ionization mass spectrometry (DESI-MS), and immunofluorescence imaging. We employ HSL to study the structural and compositional lipid profile of demyelination and remyelination in an induced focal demyelination mouse model and in multiple sclerosis lesions from patients ex vivo. Pixelwise coregistration of Raman spectroscopy and DESI-MS imaging generated a heterospectral map used to interrelate biomol. structure and compn. of myelin. Multivariate regression anal. enabled Raman-based assessment of highly specific lipid subtypes in complex tissue for the first time. This method revealed the temporal dynamics of remyelination and provided the first indication that newly formed myelin has a different lipid compn. compared to normal myelin. HSL enables detailed mol. myelin characterization that can substantially improve upon the current understanding of remyelination in multiple sclerosis and provides a strategy to assess remyelination treatments in animal models.
- 14Hosokawa, M.; Ando, M.; Mukai, S.; Osada, K.; Yoshino, T.; Hamaguchi, H.; Tanaka, T. In Vivo Live Cell Imaging for the Quantitative Monitoring of Lipids by Using Raman Microspectroscopy. Anal. Chem. 2014, 86 (16), 8224– 8230, DOI: 10.1021/ac501591d14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXht1aht7%252FE&md5=d89090139173010147f5c4870aab8be6In Vivo Live Cell Imaging for the Quantitative Monitoring of Lipids by Using Raman MicrospectroscopyHosokawa, Masahito; Ando, Masahiro; Mukai, Shoichiro; Osada, Kyoko; Yoshino, Tomoko; Hamaguchi, Hiro-o; Tanaka, TsuyoshiAnalytical Chemistry (Washington, DC, United States) (2014), 86 (16), 8224-8230CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)A straightforward in vivo monitoring technique for biomols. would be an advantageous approach for understanding their spatiotemporal dynamics in living cells. However, the lack of adequate probes has hampered the quant. detn. of the chem. compn. and metabolomics of cellular lipids at single-cell resoln. Here, we describe a method for the rapid, direct, and quant. detn. of lipid mols. from living cells using single-cell Raman imaging. In vivo localization of lipids in the form of triacylglycerol (TAG) within oleaginous microalga and their mol. compns. are monitored with high spatial resoln. in a nondestructive and label-free manner. This method can provide quant. and real-time information on compns., chain lengths, and degree of unsatn. of fatty acids in living cells for improving the cultivating parameters or for detg. the harvest timing during large-scale cultivations for microalgal lipid accumulation toward biodiesel prodn. Therefore, this technique is a potential tool for in vivo lipidomics for understanding the dynamics of lipid metabs. in various organisms.
- 15Abramczyk, H.; Surmacki, J.; Kopeć, M.; Olejnik, A. K.; Lubecka-Pietruszewska, K.; Fabianowska-Majewska, K. The Role of Lipid Droplets and Adipocytes in Cancer. Raman Imaging of Cell Cultures: MCF10A, MCF7, and MDA-MB-231 Compared to Adipocytes in Cancerous Human Breast Tissue. Analyst 2015, 140 (7), 2224– 2235, DOI: 10.1039/C4AN01875C15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXivFagt74%253D&md5=f91ccebb5206022668da2bf6607594c4The role of lipid droplets and adipocytes in cancer. Raman imaging of cell cultures: MCF10A, MCF7, and MDA-MB-231 compared to adipocytes in cancerous human breast tissueAbramczyk, Halina; Surmacki, Jakub; Kopec, Monika; Olejnik, Alicja Klaudia; Lubecka-Pietruszewska, Katarzyna; Fabianowska-Majewska, KrystynaAnalyst (Cambridge, United Kingdom) (2015), 140 (7), 2224-2235CODEN: ANALAO; ISSN:0003-2654. (Royal Society of Chemistry)We have studied live non-malignant (MCF10A), mildly malignant (MCF7) and malignant (MDA-MB-231) breast cancer cells and human breast cancer tissue. We demonstrate the first application of Raman imaging and spectroscopy in diagnosing the role of lipid droplets in cell line cultures that closely mimic an in vivo environment of various stages in human breast cancer tissue. We have analyzed the compn. of the lipid droplets in non-malignant and malignant human breast epithelial cell lines and discussed the potential of lipid droplets as a prognostic marker in breast cancer. To identify any difference in the lipid droplet-assocd. biochem. and to correlate it with different stages of breast cancer, the PCA method was employed. The chem. compn. of lipids and proteins, both in the cell line models and in human breast tissue has been analyzed. The paper shows the alterations in lipid metab. that have been reported in cancer, at both the cellular and tissue levels, and discusses how they contribute to the different aspects of tumorigenesis.
- 16Dória, M. L.; Cotrim, C. Z.; Simões, C.; Macedo, B.; Domingues, P.; Domingues, M. R.; Helguero, L. A. Lipidomic Analysis of Phospholipids from Human Mammary Epithelial and Breast Cancer Cell Lines. J. Cell. Physiol. 2013, 228 (2), 457– 468, DOI: 10.1002/jcp.2415216https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsFGqt7vO&md5=c18a4b2c4ca678908c8f5ce79f3329b3Lipidomic analysis of phospholipids from human mammary epithelial and breast cancer cell linesDoria, M. Luisa; Cotrim, Candida Z.; Simoes, Claudia; Macedo, Barbara; Domingues, Pedro; Domingues, M. Rosario; Helguero, Luisa A.Journal of Cellular Physiology (2013), 228 (2), 457-468CODEN: JCLLAX; ISSN:0021-9541. (Wiley-Blackwell)Alterations of phospholipid (PL) profiles have been assocd. to disease and specific lipids may be involved in the onset and evolution of cancer; yet, anal. of PL profiles using mass spectrometry (MS) in breast cancer cells is a novel approach. Previously, we reported a lipidomic anal. of PLs from mouse mammary epithelial and breast cancer cells using off-line thin layer chromatog. (TLC)-MS, where several changes in PL profile were found to be assocd. with the degree of malignancy of cells. In the present study, lipidomic anal. has been extended to human mammary epithelial cells and breast cancer cell lines (MCF10A, T47-D, and MDA-MB-231), using TLC-MS, validated by hydrophilic interaction liq. chromatog.-MS. Differences in phosphatidylethanolamine (PE) content relative to total amt. of PLs was highest in non-malignant cells while phosphatidic acid was present with highest relative abundance in metastatic cells. In addn., the following differences in PL mol. species assocd. to cancer phenotype, metastatic potential, and cell morphol. were found: higher levels of alkylacyl PCs and phosphatidylinositol (PI; 22:5/18:0) were detected in migratory cells, epithelial cells had less unsatd. fatty acyl chains and shorter aliph. tails in PE and sphingomyelin classes, while PI (18:0/18:1) was lowest in non-malignant cells compared to cancer cells. To date, information about PL changes in cancer progression is scarce, therefore results presented in this work will be useful as a starting point to define possible PLs with prospective as biomarkers and disclose metabolic pathways with potential for cancer therapy.
- 17Baumann, J.; Sevinsky, C.; Conklin, D. S. Lipid Biology of Breast Cancer. Biochim. Biophys. Acta, Mol. Cell Biol. Lipids 2013, 1831 (10), 1509– 1517, DOI: 10.1016/j.bbalip.2013.03.01117https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXmvVeru74%253D&md5=d64d37246f4de8e324197252e5b6ab8cLipid biology of breast cancerBaumann, Jan; Sevinsky, Christopher; Conklin, Douglas S.Biochimica et Biophysica Acta, Molecular and Cell Biology of Lipids (2013), 1831 (10), 1509-1517CODEN: BBMLFG; ISSN:1388-1981. (Elsevier B. V.)A review. Alterations in lipid metab. have been reported in many types of cancer. Lipids have been implicated in the regulation of proliferation, differentiation, apoptosis, inflammation, autophagy, motility and membrane homeostasis. It is required that their biosynthesis is tightly regulated to ensure homeostasis and to prevent unnecessary energy expenditure. This review focuses on the emerging understanding of the role of lipids and lipogenic pathway regulation in breast cancer, including parallels drawn from the study of metabolic disease models, and suggestions on how these findings can potentially be exploited to promote gains in HER2/neu-pos. breast cancer research. This article is part of a Special Issue entitled Lipid Metab. in Cancer.
- 18Liu, Q.; Luo, Q.; Halim, A.; Song, G. Targeting Lipid Metabolism of Cancer Cells: A Promising Therapeutic Strategy for Cancer. Cancer Lett. 2017, 401, 39– 45, DOI: 10.1016/j.canlet.2017.05.00218https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXotFynsr8%253D&md5=6e71f7f51e06bf38e99411ec9cf887a6Targeting lipid metabolism of cancer cells: A promising therapeutic strategy for cancerLiu, Qiuping; Luo, Qing; Halim, Alexander; Song, GuanbinCancer Letters (New York, NY, United States) (2017), 401 (), 39-45CODEN: CALEDQ; ISSN:0304-3835. (Elsevier)One of the most important metabolic hallmarks of cancer cells is deregulation of lipid metab. In addn., enhancing de novo fatty acid (FA) synthesis, increasing lipid uptake and lipolysis have also been considered as means of FA acquisition in cancer cells. FAs are involved in various aspects of tumorigenesis and tumor progression. Therefore, targeting lipid metab. is a promising therapeutic strategy for human cancer. Recent studies have shown that reprogramming lipid metab. plays important roles in providing energy, macromols. for membrane synthesis, and lipid signals during cancer progression. Moreover, accumulation of lipid droplets in cancer cells acts as a pivotal adaptive response to harmful conditions. Here, we provide a brief review of the crucial roles of FA metab. in cancer development, and place emphasis on FA origin, utilization and storage in cancer cells. Understanding the regulation of lipid metab. in cancer cells has important implications for exploring a new therapeutic strategy for management and treatment of cancer.
- 19Infanger, D. W.; Lynch, M. E.; Fischbach, C. Engineered Culture Models for Studies of Tumor-Microenvironment Interactions. Annu. Rev. Biomed. Eng. 2013, 15 (1), 29– 53, DOI: 10.1146/annurev-bioeng-071811-15002819https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVSqtL7M&md5=6a03fb7facd2c94b135cc8a690cf380eEngineered culture models for studies of tumor-microenvironment interactionsInfanger, David W.; Lynch, Maureen E.; Fischbach, ClaudiaAnnual Review of Biomedical Engineering (2013), 15 (), 29-53CODEN: ARBEF7; ISSN:1523-9829. (Annual Reviews)A review. Heterogeneous microenvironmental conditions play crit. roles in cancer pathogenesis and therapy resistance and arise from changes in tissue dimensionality, cell-extracellular matrix (ECM) interactions, sol. factor signaling, oxygen as well as metabolic gradients, and exogenous biomech. cues. Traditional cell culture approaches are restricted in their ability to mimic this complexity with physiol. relevance, offering only partial explanation as to why novel therapeutic compds. are frequently efficacious in vitro but disappoint in preclin. and clin. studies. In an effort to overcome these limitations, phys. sciences-based strategies were employed to model specific aspects of the cancer microenvironment. Although these strategies offer promise to reveal the contributions of microenvironmental parameters on tumor initiation, progression, and therapy resistance, they, too, frequently suffer from limitations. This review highlights physicochem. and biol. key features of the tumor microenvironment, critically discusses advantages and limitations of current engineering strategies, and provides a perspective on future opportunities for engineered tumor models.
- 20Fischbach, C.; Chen, R.; Matsumoto, T.; Schmelzle, T.; Brugge, J. S.; Polverini, P. J.; Mooney, D. J. Engineering Tumors with 3D Scaffolds. Nat. Methods 2007, 4 (10), 855– 860, DOI: 10.1038/nmeth108520https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtV2mtr%252FE&md5=3ea950e03c308dfb0b931c53f28bff5aEngineering tumors with 3D scaffoldsFischbach, Claudia; Chen, Ruth; Matsumoto, Takuya; Schmelzle, Tobias; Brugge, Joan S.; Polverini, Peter J.; Mooney, David J.Nature Methods (2007), 4 (10), 855-860CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Microenvironmental conditions control tumorigenesis and biomimetic culture systems that allow for in vitro and in vivo tumor modeling may greatly aid studies of cancer cells' dependency on these conditions. The authors engineered 3-dimensional (3D) human tumor models using carcinoma cells in polymeric scaffolds that recreated microenvironmental characteristics representative of tumors in vivo. Strikingly, the angiogenic characteristics of tumor cells were dramatically altered upon 3D culture within this system, and corresponded much more closely to tumors formed in vivo. Cells in this model were also less sensitive to chemotherapy and yielded tumors with enhanced malignant potential. The authors assessed the broad relevance of these findings with 3D culture of other tumor cell lines in this same model, comparison with std. 3D Matrigel culture and in vivo expts. This new biomimetic model may provide a broadly applicable 3D culture system to study the effect of microenvironmental conditions on tumor malignancy in vitro and in vivo.
- 21Shekhar, M. P. V.V.; Tait, L.; Pauley, R. J.; Wu, G. S.; Santner, S. J.; Nangia-Makker, P.; Shekhar, V.; Nassar, H.; Visscher, D. W.; Heppner, G. H. Comedo-Ductal Carcinoma in Situ: A Paradoxical Role for Programmed Cell Death. Cancer Biol. Ther. 2008, 7 (11), 1774– 1782, DOI: 10.4161/cbt.7.11.678121https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhvVGgu70%253D&md5=da65d844da20239f0ef0337f47e29a30Comedo-ductal carcinoma in situ: a paradoxical role for programmed cell deathShekhar, Malathy P. V.; Tait, Larry; Pauley, Robert J.; Wu, Gen Sheng; Santner, Steven J.; Nangia-Makker, Pratima; Shekhar, Varun; Nassar, Hind; Visscher, Daniel W.; Heppner, Gloria H.; Miller, Fred R.Cancer Biology & Therapy (2008), 7 (11), 1774-1782CODEN: CBTAAO; ISSN:1538-4047. (Landes Bioscience)Comedo-DCIS is a histol. subtype of preinvasive breast neoplasia that is characterized by prominent apoptotic cell death and has greater malignant potential than other DCIS subtypes. We investigated the mechanisms of apoptosis in comedo-DCIS and its role in conversion of comedo-DCIS to invasive cancer. Clin. comedo-DCIS excisions and the MCF10DCIS.com human breast cancer model which produces lesions resembling comedo-DCIS were analyzed. Apoptotic luminal and myoepithelial cells were identified by TUNEL and reactivity to cleaved PARP antibody and cell death assessed by Western blotting, Mitocapture and immunohistochem. assays. MCF10DCIS.com cells undergo spontaneous apoptosis in vitro, both in monolayers and multicellular spheroids; it is assocd. with increased mitochondrial membrane permeability, increase in Bax/Bcl-2 ratio and occurs via caspase-9-dependent p53-independent pathway. This suggests that apoptosis is stromal-independent and that the cells are programmed to undergo apoptosis. Immunostaining with cleaved PARP antibody showed that myoepithelial apoptosis occurs before lesions progress to comedo-DCIS in both clin. comedo-DCIS and in vivo MCF10DCIS.com lesions. Intense staining for MMP-2, MMP-3, MMP-9 and MMP-11 was obsd. in the stroma and epithelia of solid DCIS lesions prior to conversion to comedo-DCIS in clin. and MCF10DCIS.com lesions. Gelatin zymog. showed higher MMP-2 levels in lysates and conditioned media of MCF10DCIS.com cells undergoing apoptosis. These data suggest that signals arising from the outside (microenvironmental) and inside (internal genetic alterations) of the duct act in concert to trigger apoptosis of myoepithelial and luminal epithelial cells. Our findings implicate spontaneous apoptosis in both the etiol. and progression of comedo-DCIS. It is possible that spontaneous apoptosis facilitates elimination of cells thus permitting expansion and malignant transformation of cancer cells that are resistant to spontaneous apoptosis.
- 22McElwee, J. L.; Mohanan, S.; Griffith, O. L.; Breuer, H. C.; Anguish, L. J.; Cherrington, B. D.; Palmer, A. M.; Howe, L. R.; Subramanian, V.; Causey, C. P.; Identification of PADI2 as a Potential Breast Cancer Biomarker and Therapeutic Target. BMC Cancer 2012. DOI: 10.1186/1471-2407-12-500 .There is no corresponding record for this reference.
- 23Vidavsky, N.; Kunitake, J. A.; Chiou, A. E.; Northrup, P. A.; Porri, T. J.; Ling, L.; Fischbach, C.; Estroff, L. A. Studying Biomineralization Pathways in a 3D Culture Model of Breast Cancer Microcalcifications. Biomaterials 2018, 179, 71– 82, DOI: 10.1016/j.biomaterials.2018.06.030There is no corresponding record for this reference.
- 24DelNero, P.; Lane, M.; Verbridge, S. S.; Kwee, B.; Kermani, P.; Hempstead, B.; Stroock, A.; Fischbach, C. 3D Culture Broadly Regulates Tumor Cell Hypoxia Response and Angiogenesis via Pro-Inflammatory Pathways. Biomaterials 2015, 55, 110– 118, DOI: 10.1016/j.biomaterials.2015.03.03524https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXlsl2iurw%253D&md5=a471c90cc02529c0e25aabf8a8e9a9b73D culture broadly regulates tumor cell hypoxia response and angiogenesis via pro-inflammatory pathwaysDelNero, Peter; Lane, Maureen; Verbridge, Scott S.; Kwee, Brian; Kermani, Pouneh; Hempstead, Barbara; Stroock, Abraham; Fischbach, ClaudiaBiomaterials (2015), 55 (), 110-118CODEN: BIMADU; ISSN:0142-9612. (Elsevier Ltd.)Oxygen status and tissue dimensionality are crit. determinants of tumor angiogenesis, a hallmark of cancer and an enduring target for therapeutic intervention. However, it is unclear how these microenvironmental conditions interact to promote neovascularization, due in part to a lack of comprehensive, unbiased data sets describing tumor cell gene expression as a function of oxygen levels within three-dimensional (3D) culture. Here, we utilized alginate-based, oxygen-controlled 3D tumor models to study the interdependence of culture context and the hypoxia response. Microarray gene expression anal. of tumor cells cultured in 2D vs. 3D under ambient or hypoxic conditions revealed striking interdependence between culture dimensionality and hypoxia response, which was mediated in part by pro-inflammatory signaling pathways. In particular, interleukin-8 (IL-8) emerged as a major player in the microenvironmental regulation of the hypoxia program. Notably, this interaction between dimensionality and oxygen status via IL-8 increased angiogenic sprouting in a 3D endothelial invasion assay. Taken together, our data suggest that pro-inflammatory pathways are crit. regulators of tumor hypoxia response within 3D environments that ultimately impact tumor angiogenesis, potentially providing important therapeutic targets. Furthermore, these results highlight the importance of pathol. relevant tissue culture models to study the complex phys. and chem. processes by which the cancer microenvironment mediates new vessel formation.
- 25Kang, H. S.; Lee, S. C.; Park, Y. S.; Jeon, Y. E.; Lee, J. H.; Jung, S. Y.; Park, I. H.; Jang, S. H.; Park, H. M.; Yoo, C. W. Protein and Lipid MALDI Profiles Classify Breast Cancers According to the Intrinsic Subtype. BMC Cancer 2011, 11 (1), 465, DOI: 10.1186/1471-2407-11-465There is no corresponding record for this reference.
- 26Kawashima, M.; Iwamoto, N.; Kawaguchi-Sakita, N.; Sugimoto, M.; Ueno, T.; Mikami, Y.; Terasawa, K.; Sato, T. A.; Tanaka, K.; Shimizu, K. High-Resolution Imaging Mass Spectrometry Reveals Detailed Spatial Distribution of Phosphatidylinositols in Human Breast Cancer. Cancer Sci. 2013, 104 (10), 1372– 1379, DOI: 10.1111/cas.1222926https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsFOgs7rI&md5=15e8c8ac9b5687624be3607117e60508High-resolution imaging mass spectrometry reveals detailed spatial distribution of phosphatidylinositols in human breast cancerKawashima, Masahiro; Iwamoto, Noriko; Kawaguchi-Sakita, Nobuko; Sugimoto, Masahiro; Ueno, Takayuki; Mikami, Yoshiki; Terasawa, Kazuya; Sato, Taka-Aki; Tanaka, Koichi; Shimizu, Kazuharu; Toi, MasakazuCancer Science (2013), 104 (10), 1372-1379CODEN: CSACCM; ISSN:1349-7006. (Wiley-Blackwell)High-resoln. matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI IMS) is an emerging application for lipid research that provides a comprehensive and detailed spatial distribution of ionized mols. Recent lipidomic approach has identified several phospholipids and phosphatidylinositols (PIs) are accumulated in breast cancer tissues and are therefore novel biomarker candidates. Because their distribution and significance remain unclear, we investigated the precise spatial distribution of PIs in human breast cancer tissues using high-resoln. MALDI IMS. We evaluated tissues from nine human breast cancers and one normal mammary gland by neg. ion MALDI IMS at a resoln. of 10 μm. We detected 10 PIs with different fatty acid compns., and their proportions were remarkably variable in the malignant epithelial regions. High-resoln. imaging enabled us to discriminate cancer cell clusters from the adjacent stromal tissue within epithelial regions; moreover, this technique revealed that several PIs were specifically localized to cancer cell clusters. These PIs were heterogeneously distributed within cancer cell clusters, allowing us to identify two different populations of cancer cells that predominantly expressed either PI(18:0/18:1) or PI(18:0/20:3). Tracing the expression level of PIs during cancer cell progression suggested that the latter population is assocd. with the invasion. Our study documents a novel model for phospholipid anal. of breast cancer tissues by using high-resoln. MALDI IMS and identifies candidate PIs that can describe a specific phenotype of cancer cells.
- 27Angerer, T. B.; Magnusson, Y.; Landberg, G.; Fletcher, J. S. Lipid Heterogeneity Resulting from Fatty Acid Processing in the Human Breast Cancer Microenvironment Identified by GCIB-ToF-SIMS Imaging. Anal. Chem. 2016, 88 (23), 11946– 11954, DOI: 10.1021/acs.analchem.6b0388427https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslCktrvL&md5=aa98a45a841a4038143f72454d1b0e2eLipid Heterogeneity Resulting from Fatty Acid Processing in the Human Breast Cancer Microenvironment Identified by GCIB-ToF-SIMS ImagingAngerer, Tina B.; Magnusson, Ylva; Landberg, Goeran; Fletcher, John S.Analytical Chemistry (Washington, DC, United States) (2016), 88 (23), 11946-11954CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Breast cancer is an umbrella term used to describe a collection of different diseases with broad inter- and intra-tumor heterogeneity. Understanding this variation is crit. in order to develop, and precisely prescribe, new treatments. Changes in the lipid metab. of cancerous cells can provide important indications as to the metabolic state of the cells but are difficult to investigate with conventional histol. methods. Due to the introduction of new higher energy (40 kV) gas cluster ion beams (GCIBs) time-of-flight secondary ion mass spectrometry (ToF-SIMS) imaging is now capable of providing information on the distribution of hundreds of mol. species simultaneously on a cellular to sub-cellular scale. GCIB-ToF-SIMS was used to elucidate changes in lipid compn. in 9 breast cancer biopsy samples. Improved mol. signal generation by the GCIB produced location specific information that revealed elevated levels of essential lipids to be related to inflammatory cells in the stroma while cancerous areas are dominated by non-essential fatty acids and a variety of phosphatidylinositol species with further in-tumor variety arising from decreased desaturase activity. These changes in lipid compn. due to different enzyme activity are seemingly independent of oxygen availability and can be linked to favorable cell membrane properties for either proliferation/invasion or drug resistance/survival.
- 28Cífková, E.; Holčapek, M.; Lísa, M.; Vrána, D.; Melichar, B.; Študent, V. Lipidomic Differentiation between Human Kidney Tumors and Surrounding Normal Tissues Using HILIC-HPLC/ESI-MS and Multivariate Data Analysis. J. Chromatogr. B: Anal. Technol. Biomed. Life Sci. 2015, 1000, 14– 21, DOI: 10.1016/j.jchromb.2015.07.01128https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtlajur3J&md5=f3311392bf0df83fec64d7a59d9cc933Lipidomic differentiation between human kidney tumors and surrounding normal tissues using HILIC-HPLC/ESI-MS and multivariate data analysisCifkova, Eva; Holcapek, Michal; Lisa, Miroslav; Vrana, David; Melichar, Bohuslav; Student, VladimirJournal of Chromatography B: Analytical Technologies in the Biomedical and Life Sciences (2015), 1000 (), 14-21CODEN: JCBAAI; ISSN:1570-0232. (Elsevier B.V.)The characterization of differences among polar lipid classes in tumors and surrounding normal tissues of 20 kidney cancer patients is performed by hydrophilic interaction liq. chromatog. (HILIC) coupled to electrospray ionization mass spectrometry (ESI-MS). The detailed anal. of identified lipid classes using relative abundances of characteristic ions in neg.- and pos.-ion modes is used for the detn. of more than 120 individual lipid species contg. attached fatty acyls of different chain length and double bond no. Lipid species are described using relative abundances, providing a better visualization of lipidomic differences between tumor and normal tissues. The multivariate data anal. methods using unsupervised principal component anal. (PCA) and supervised orthogonal partial least square (OPLS) are used for the characterization of statistically significant differences in identified lipid species. Ten most significant up- and down-regulated lipids in OPLS score plots are also displayed by box plots. A notable increase of relative abundances of lipids contg. four and more double bonds is detected in tumor compared to normal tissues.
- 29Hilvo, M.; Denkert, C.; Lehtinen, L.; Muller, B.; Brockmoller, S.; Seppanen-Laakso, T.; Budczies, J.; Bucher, E.; Yetukuri, L.; Castillo, S. Novel Theranostic Opportunities Offered by Characterization of Altered Membrane Lipid Metabolism in Breast Cancer Progression. Cancer Res. 2011, 71 (9), 3236– 3245, DOI: 10.1158/0008-5472.CAN-10-3894There is no corresponding record for this reference.
- 30Perrotti, F.; Rosa, C.; Cicalini, I.; Sacchetta, P.; Del Boccio, P.; Genovesi, D.; Pieragostino, D. Advances in Lipidomics for Cancer Biomarkers Discovery. Int. J. Mol. Sci. 2016, 17 (12), 1992, DOI: 10.3390/ijms1712199230https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXht1eks7c%253D&md5=c767aaf1f2da0027563b3900d38cb35aAdvances in lipidomics for cancer biomarkers discoveryPerrotti, Francesca; Rosa, Consuelo; Cicalini, Ilaria; Sacchetta, Paolo; Del Boccio, Piero; Genovesi, Domenico; Pieragostino, DamianaInternational Journal of Molecular Sciences (2016), 17 (12), 1992/1-1992/26CODEN: IJMCFK; ISSN:1422-0067. (MDPI AG)Lipids play crit. functions in cellular survival, proliferation, interaction and death, since they are involved in chem.-energy storage, cellular signaling, cell membranes, and cell-cell interactions. These cellular processes are strongly related to carcinogenesis pathways, particularly to transformation, progression, and metastasis, suggesting the bioactive lipids are mediators of a no. of oncogenic processes. The current review gives a synopsis of a lipidomic approach in tumor characterization; we provide an overview on potential lipid biomarkers in the oncol. field and on the principal lipidomic methodologies applied. The novel lipidomic biomarkers are reviewed in an effort to underline their role in diagnosis, in prognostic characterization and in prediction of therapeutic outcomes. A lipidomic investigation through mass spectrometry highlights new insights on mol. mechanisms underlying cancer disease. This new understanding will promote clin. applications in drug discovery and personalized therapy.
- 31Chughtai, K.; Jiang, L.; Greenwood, T. R.; Glunde, K.; Heeren, R. M. A. Mass Spectrometry Images Acylcarnitines, Phosphatidylcholines, and Sphingomyelin in MDA-MB-231 Breast Tumor Models. J. Lipid Res. 2013, 54 (2), 333– 344, DOI: 10.1194/jlr.M02796131https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXosF2htQ%253D%253D&md5=f43e68b6c9f3a1c996abcd87e61498eaMass spectrometry images acylcarnitines, phosphatidylcholines, and sphingomyelin in MDA-MB-231 breast tumor modelsChughtai, Kamila; Lu, Jiang; Greenwood, Tiffany R.; Glunde, Kristine; Heeren, Ron M. A.Journal of Lipid Research (2013), 54 (2), 333-344CODEN: JLPRAW; ISSN:0022-2275. (American Society for Biochemistry and Molecular Biology, Inc.)The lipid compns. of different breast tumor microenvironments are largely unknown due to limitations in lipid imaging techniques. Imaging lipid distributions would enhance our understanding of processes occurring inside growing tumors, such as cancer cell proliferation, invasion, and metastasis. Recent developments in MALDI mass spectrometry imaging (MSI) enable rapid and specific detection of lipids directly from thin tissue sections. In this study, we performed multimodal imaging of acylcarnitines, phosphatidylcholines (PC), a lysophosphatidylcholine (LPC), and a sphingomyelin (SM) from different microenvironments of breast tumor xenograft models, which carried tdTomato red fluorescent protein as a hypoxia-response element-driven reporter gene. The MSI mol. lipid images revealed spatially heterogeneous lipid distributions within tumor tissue. Four of the most-abundant lipid species, namely PC(16:0/16:0), PC(16:0/18:1), PC(18:1/18:1), and PC(18:0/18:1), were localized in viable tumor regions, whereas LPC(16:0/0:0) was detected in necrotic tumor regions. We identified a heterogeneous distribution of palmitoylcarnitine, stearoylcarnitine, PC(16:0/22:1), and SM(d18:1/16:0) sodium adduct, which colocalized primarily with hypoxic tumor regions. For the first time, we have applied a multimodal imaging approach that has combined optical imaging and MALDI-MSI with ion mobility sepn. to spatially localize and structurally identify acylcarnitines and a variety of lipid species present in breast tumor xenograft models.
- 32Confocal Raman Microscopy; Dieing, T., Hollricher, O., Toporski, J., Eds.; Springer Berlin Heidelberg: Berlin, Heidelberg, 2011.There is no corresponding record for this reference.
- 33Butler, H. J.; Ashton, L.; Bird, B.; Cinque, G.; Curtis, K.; Dorney, J.; Esmonde-White, K.; Fullwood, N. J.; Gardner, B.; Martin-Hirsch, P. L. Using Raman Spectroscopy to Characterize Biological Materials. Nat. Protoc. 2016, 11 (4), 664– 687, DOI: 10.1038/nprot.2016.03633https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xjs1yjs7k%253D&md5=563e9a3c9684196390e2001a791a6e86Using Raman spectroscopy to characterize biological materialsButler, Holly J.; Ashton, Lorna; Bird, Benjamin; Cinque, Gianfelice; Curtis, Kelly; Dorney, Jennifer; Esmonde-White, Karen; Fullwood, Nigel J.; Gardner, Benjamin; Martin-Hirsch, Pierre L.; Walsh, Michael J.; McAinsh, Martin R.; Stone, Nicholas; Martin, Francis L.Nature Protocols (2016), 11 (4), 664-687CODEN: NPARDW; ISSN:1750-2799. (Nature Publishing Group)Raman spectroscopy can be used to measure the chem. compn. of a sample, which can in turn be used to ext. biol. information. Many materials have characteristic Raman spectra, which means that Raman spectroscopy has proven to be an effective anal. approach in geol., semiconductor, materials and polymer science fields. The application of Raman spectroscopy and microscopy within biol. is rapidly increasing because it can provide chem. and compositional information, but it does not typically suffer from interference from water mols. Anal. does not conventionally require extensive sample prepn.; biochem. and structural information can usually be obtained without labeling. In this protocol, we aim to standardize and bring together multiple exptl. approaches from key leaders in the field for obtaining Raman spectra using a microspectrometer. As examples of the range of biol. samples that can be analyzed, we provide instructions for acquiring Raman spectra, maps and images for fresh plant tissue, formalin-fixed and fresh frozen mammalian tissue, fixed cells and biofluids. We explore a robust approach for sample prepn., instrumentation, acquisition parameters and data processing. By using this approach, we expect that a typical Raman expt. can be performed by a nonspecialist user to generate high-quality data for biol. materials anal.
- 34Talari, A. C. S.; Movasaghi, Z.; Rehman, S.; Rehman, I. U. Raman Spectroscopy of Biological Tissues. Appl. Spectrosc. Rev. 2015, 50 (1), 46– 111, DOI: 10.1080/05704928.2014.92390234https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtlyrsrbI&md5=fde96a5868714fbb53e87ac1756a2a7bRaman Spectroscopy of Biological TissuesTalari, Abdullah Chandra Sekhar; Movasaghi, Zanyar; Rehman, Shazza; Rehman, Ihtesham urApplied Spectroscopy Reviews (2015), 50 (1), 46-111CODEN: APSRBB; ISSN:0570-4928. (Taylor & Francis, Inc.)A review. We previously published a comprehensive review paper reviewing the Raman spectroscopy of biol. mols. This research area has expanded rapidly, which warranted an update to the existing review paper by adding the recently reported studies in literature. This article reviews some of the recent advances of Raman spectroscopy in relation to biomedical applications starting from natural tissues to cancer biol. Raman spectroscopy, an optical mol. detective, is a vibrational spectroscopic technique that has potential not only in cancer diagnosis but also in understanding progression of the disease. This article summarizes some of the most widely obsd. peak frequencies and their assignments. The aim of this review is to develop a database of mol. fingerprints, which will facilitate researchers in identifying the chem. structure of the biol. tissues including most of the significant peaks reported both in the normal and cancerous tissues. It has covered a variety of Raman approaches and its quant. and qual. biochem. information. In addn., it covers the use of Raman spectroscopy to analyze a variety of different malignancies including breast, brain, cervical, gastrointestinal, lung, oral, and skin cancer. Multivariate anal. approaches used in these studies have also been covered.
- 35Soule, H. D.; Maloney, T. M.; Wolman, S. R.; Peterson, W. D.; Brenz, R.; McGrath, C. M.; Russo, J.; Pauley, R. J.; Jones, R. F.; Brooks, S. C. Isolation and Characterization of a Spontaneously Immortalized Human Breast Epithelial Cell Line, MCF-10. Cancer Res. 1990, 50 (18), 6075– 608635https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXktFSq&md5=bca00d50e0a1ef6366053785c0534acdIsolation and characterization of a spontaneously immortalized human breast epithelial cell line, MCF-10Soule, Herbert D.; Maloney, Terry M.; Wolman, Sandra R.; Peterson, Ward D., Jr.; Brenz, Richard, Jr.; McGrath, Charles M.; Russo, Jose; Pauley, Robert J.; Jones, Richard F.; Brooks, S. C.Cancer Research (1990), 50 (18), 6075-86CODEN: CNREA8; ISSN:0008-5472.Two sublines of a breast epithelial cell culture, MCF-10, derived from human fibrocystic mammary tissue exhibit immortality after extended cultivation in low calcium concns. (0.03-0.06 mM) and floating transfers in low calcium (MCF-10F), or by trypsin-Versene passages in the customary (normal) calcium levels, 1.05 mM (MCF-10A). Both sublines have been maintained as sep. entities after 2.3 yr (849 days) in vitro and at present have been in culture for longer than 4 yr. MCF-10 has the characteristics of normal breast epithelium by the following criteria: (a) lack of tumorigenicity in nude mice; (b) three-dimensional growth in collagen; (c) growth in culture that is controlled by hormones and growth factors; (d) lack of anchorage-independent growth; and (e) dome formation in confluent cultures. Cytogenetic anal. prior to immortalization showed normal diploid cells; although later passages showed minimal rearrangement and near-diploidy, the immortal cells were not karyotypically normal. The emergence of an immortal culture in normal calcium media was not an inherent characteristic of the original tissue from which MCF-10 was derived since reactivated cryo-preserved cells from cultures grown for 0.3 to 1.2 yr in low calcium were incapable of sustained growth in normal calcium.
- 36Miller, F. R.; Santner, S. J.; Tait, L.; Dawson, P. J. MCF10DCIS.Com Xenograft Model of Human Comedo Ductal Carcinoma in Situ. J. Natl. Cancer Inst. 2000, 92 (14), 1185– 1186, DOI: 10.1093/jnci/92.14.1185a36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3cvlvFentQ%253D%253D&md5=5ed501454dc8b8f787ea9837f03aa72fMCF10DCIS.com xenograft model of human comedo ductal carcinoma in situMiller F R; Santner S J; Tait L; Dawson P JJournal of the National Cancer Institute (2000), 92 (14), 1185-6 ISSN:0027-8874.There is no expanded citation for this reference.
- 37Barnabas, N.; Cohen, D. Phenotypic and Molecular Characterization of MCF10DCIS and SUM Breast Cancer Cell Lines. Int. J. Breast Cancer 2013, 2013, 872743, DOI: 10.1155/2013/87274337https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3sznsFyjuw%253D%253D&md5=fc9e492c1ba5859a9e5d7f5f4c95657ePhenotypic and Molecular Characterization of MCF10DCIS and SUM Breast Cancer Cell LinesBarnabas Nandita; Cohen DaliaInternational journal of breast cancer (2013), 2013 (), 872743 ISSN:2090-3170.We reviewed the phenotypic and molecular characteristics of MCF10DCIS.com and the SUM cell lines based on numerous studies performed over the years. The major signaling pathways that give rise to the phenotype of these cells may serve as a good resource of information when researchers in drug discovery and development use these cells to identify novel targets and biomarkers. Major signaling pathways and mutations affecting the coding sequence are also described providing important information when using these cells as a model in a variety of studies.
- 38Santner, S. J.; Dawson, P. J.; Tait, L.; Soule, H. D.; Eliason, J.; Mohamed, A. N.; Wolman, S. R.; Heppner, G. H.; Miller, F. R. Malignant MCF10CA1 Cell Lines Derived from Premalignant Human Breast Epithelial MCF10AT Cells. Breast Cancer Res. Treat. 2001, 65 (2), 101– 110, DOI: 10.1023/A:100646142227338https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3M7mtVWitA%253D%253D&md5=a1e5b7147e918dbdca3e33d6e281c119Malignant MCF10CA1 cell lines derived from premalignant human breast epithelial MCF10AT cellsSantner S J; Dawson P J; Tait L; Soule H D; Eliason J; Mohamed A N; Wolman S R; Heppner G H; Miller F RBreast cancer research and treatment (2001), 65 (2), 101-10 ISSN:0167-6806.The MCF10 series of cell lines was derived from benign breast tissue from a woman with fibrocystic disease. The MCF10 human breast epithelial model system consists of mortal MCF10M and MCF10MS (mortal cells grown in serum-free and serum-containing media, respectively), immortalized but otherwise normal MCF10F and MCF10A lines (free-floating versus growth as attached cells), transformed MCF10AneoT cells transfected with T24 Ha-ras, and premalignant MCF10AT cells with potential for neoplastic progression. The MCF10AT, derived from xenograft-passaged MCF10-AneoT cells, generates carcinomas in approximately 25% of xenografts. We now report the derivation of fully malignant MCF10CA1 lines that complete the spectrum of progression from relatively normal breast epithelial cells to breast cancer cells capable of metastasis. MCF10CA1 lines display histologic variations ranging from undifferentiated carcinomas, sometimes with focal squamous differentiation, to well-differentiated adenocarcinomas. At least two metastasize to the lung following injection of cells into the tail vein; one line grows very rapidly in the lung, with animals moribund within 4 weeks, whereas the other requires 15 weeks to reach the same endpoint. In addition to variations in efficiency of tumor production, the MCF10CA1 lines show differences in morphology in culture, anchorage-independent growth, karyotype, and immunocytochemistry profiles. The MCF10 model provides a unique tool for the investigation of molecular changes during progression of human breast neoplasia and the generation of tumor heterogeneity on a common genetic background.
- 39Alo, P. L.; Visca, P.; Marci, A.; Mangoni, A.; Botti, C.; Di Tondo, U. Expression of Fatty Acid Synthase (FAS) as a Predictor of Recurrence in Stage I Breast Carcinoma Patients. Cancer 1996, 77 (3), 474– 482, DOI: 10.1002/(SICI)1097-0142(19960201)77:3<474::AID-CNCR8>3.0.CO;2-K39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28Xht12jsL0%253D&md5=9f47ad6c3a97ada40fc288552f638c59Expression of fatty acid synthase (FAS) as a predictor of recurrence in stage I breast carcinoma patientsAlo, Piero L.; Visca, Paolo; Marci, Adele; Mangoni, Antonella; Botti, Claudio; Di Tondo, UgoCancer (New York) (1996), 77 (3), 474-82CODEN: CANCAR; ISSN:0008-543X. (Wiley-Liss)Fatty acid synthase (FAS) is a mol. found in tumor cells from breast carcinomas of patients whose prognosis is very poor. Recently, this mol. has been identified as the key enzyme in fatty acid biosynthesis. This study was done to test the strength of FAS as a prognostic indicator for disease free survival (DFS) and overall survival (OS). Clin. records, histol. features, immunohistochem. expression of cathepsin D and c-erbB-2, and estrogen and progesterone receptor status of 110 Stage I breast carcinoma patients were all assocd. with FAS by a chi-square test. The patterns of DFS and OS were estd. over a ten-year follow-up period using the Kaplan-Meier method. Univariate and multivariate anal. were evaluated using a log logistic regression model. Multivariate regression anal. was based on the Cox proportional hazard model. To detect FAS, cathepsin D and c-erbB-2 expression as well as estrogen and progesterone receptor status, the authors used the unlabeled immunoperoxidase technique on formalin fixed, paraffin embedded tissue. FAS was significantly assocd. with a higher risk of recurrence because it predicted both DFS and OS when evaluated as a continuous variable and DFS when evaluated with other prognostic markers. Peritumoral lymphatic vessel invasion was the other most significant independent predictor for DFS and OS. FAS is a reliable prognostic marker to predict DFS and OS in patients with early breast cancer.
- 40Milgraum, L. Z.; Witters, L. A.; Pasternack, G. R.; Kuhajda, F. P. Enzymes of the Fatty Acid Synthesis Pathway Are Highly Expressed in in Situ Breast Carcinoma. Clin. Cancer Res. 1997, 3 (11), 2115– 212040https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXns1Ogt78%253D&md5=bb85cb2753c93f199c01517e3887f15aEnzymes of the fatty acid synthesis pathway are highly expressed in in situ breast carcinomaMilgraum, Lea Z.; Witters, Lee A.; Pasternack, Gary R.; Kuhajda, Francis P.Clinical Cancer Research (1997), 3 (11), 2115-2120CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Expression of high levels of fatty acid synthase (FAS), an important enzyme in fatty acid synthesis, has been identified in a wide variety of human carcinomas. In breast and prostate carcinoma, FAS expression appears to be assocd. with aggressive disease. Recent biochem. studies have demonstrated that FAS expression in cancer cells connotes activation of the entire fatty acid synthesis pathway leading to the prodn. of palmitic acid. Here, we explore the immunohistochem. expression of FAS and human acetyl-CoA carboxylase (HACC), the rate-limiting enzyme in fatty acid synthesis, in breast cancer progression from histol. normal breast through the development of in situ duct and lobular carcinoma to infiltrating carcinoma. Both FAS and the Mr 275,000 isoform of HACC are expressed in a small subset of cells in normal breast lobules and terminal ducts. Upon development of either in situ duct or lobular carcinoma, FAS and both isoforms of HACC are expressed at higher levels and in a majority of the cells. These findings suggest that expression of the enzymes of fatty acid synthesis are frequently altered early in the progression of human breast carcinoma.
- 41Menendez, J. A.; Lupu, R. Fatty Acid Synthase and the Lipogenic Phenotype in Cancer Pathogenesis. Nat. Rev. Cancer 2007, 7 (10), 763– 777, DOI: 10.1038/nrc222241https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtVOnsr3P&md5=04e65d93150e39aef32d00cb0339fc92Fatty acid synthase and the lipogenic phenotype in cancer pathogenesisMenendez, Javier A.; Lupu, RuthNature Reviews Cancer (2007), 7 (10), 763-777CODEN: NRCAC4; ISSN:1474-175X. (Nature Publishing Group)A review. Fatty acid synthase (FASN) catalyzes the synthesis of fatty acids, and this synthetic pathway is upregulated in many tumors. How might FASN and increased lipogenesis be involved in cancer, and is FASN a valid therapeutic target. There is a renewed interest in the ultimate role of fatty acid synthase (FASN) - a key lipogenic enzyme catalyzing the terminal steps in the de novo biogenesis of fatty acids - in cancer pathogenesis. Tumor-assocd. FASN, by conferring growth and survival advantages rather than functioning as an anabolic energy-storage pathway, appears to necessarily accompany the natural history of most human cancers. A recent identification of cross-talk between FASN and well-established cancer-controlling networks begins to delineate the oncogenic nature of FASN-driven lipogenesis. FASN, a nearly-universal druggable target in many human carcinomas and their precursor lesions, offers new therapeutic opportunities for metabolically treating and preventing cancer.
- 42Ghosh, C.; Nandi, S.; Bhattacharyya, K. Probing Micro-Environment of Lipid Droplets in a Live Breast Cell: MCF7 and MCF10A. Chem. Phys. Lett. 2017, 670, 27– 31, DOI: 10.1016/j.cplett.2016.12.06842https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXlsFagtw%253D%253D&md5=6979891b49e8e0e3614c17302804c273Probing micro-environment of lipid droplets in a live breast cell: MCF7 and MCF10AGhosh, Catherine; Nandi, Somen; Bhattacharyya, KankanChemical Physics Letters (2017), 670 (), 27-31CODEN: CHPLBC; ISSN:0009-2614. (Elsevier B.V.)Local environment of the lipid droplets inside the breast cancer cells, MCF7 and in non-malignant breast cells, MCF10A is monitored using time-resolved confocal microscopy. For this study, a coumarin-based dye C153 has been used. The local polarity and the solvation dynamics indicate that a cytoplasmic lipid droplet is less polar and displays slower solvation dynamics compared to the cytosol. Significant differences in terms of no. of lipid droplets, polarity and solvation dynamics are obsd. between the cancer cell (MCF7) and its non-malignant cell (MCF10A).
- 43Wilmanski, T.; Buhman, K.; Donkin, S. S.; Burgess, J. R.; Teegarden, D. 1α,25-Dihydroxyvitamin D Inhibits de Novo Fatty Acid Synthesis and Lipid Accumulation in Metastatic Breast Cancer Cells through down-Regulation of Pyruvate Carboxylase. J. Nutr. Biochem. 2017, 40, 194– 200, DOI: 10.1016/j.jnutbio.2016.11.00643https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitVajs7vM&md5=e5284356e8a8274dfdf10c310cf208f61α,25-dihydroxyvitamin D inhibits de novo fatty acid synthesis and lipid accumulation in metastatic breast cancer cells through down-regulation of pyruvate carboxylaseWilmanski, Tomasz; Buhman, Kimberly; Donkin, Shawn S.; Burgess, John R.; Teegarden, DorothyJournal of Nutritional Biochemistry (2017), 40 (), 194-200CODEN: JNBIEL; ISSN:0955-2863. (Elsevier)Both increased de novo fatty acid synthesis and higher neutral lipid accumulation are a common phenotype obsd. in aggressive breast cancer cells, making lipid metab. a promising target for breast cancer prevention. In the present studies, we demonstrate a novel effect of the active metabolite of vitamin D, 1α,25-dihydroxyvitamin D (1,25(OH)2D) on lipid metab. in malignant breast epithelial cells. Treatment of MCF10CA1a breast epithelial cells with 1,25(OH)2D (10 nM) for 5 and 7 days decreased the level of triacylglycerol, the most abundant form of neutral lipids, by 20%(±3.9) and 50%(±5.9), resp. In addn., 1,25(OH)2D treatment for 5 days decreased palmitate synthesis from glucose, the major fatty acid synthesized de novo (48% ± 5.5 relative to vehicle). We have further identified the anaplerotic enzyme pyruvate carboxylase (PC) as a target of 1,25(OH)2D-mediated regulation and hypothesized that 1,25(OH)2D regulates breast cancer cell lipid metab. through inhibition of PC. PC mRNA expression was down-regulated with 1,25(OH)2D treatment at 2 (73% ± 6 relative to vehicle) and 5 (56% ± 8 relative to vehicle) days. Decrease in mRNA abundance corresponded with a decrease in PC protein expression at 5 days of treatment (54% ± 12 relative to vehicle). Constitutive overexpression of PC in MCF10CA1a cells using a pCMV6-PC plasmid inhibited the effect of 1,25(OH)2D on both TAG accumulation and de novo palmitate synthesis from glucose. Together, these studies demonstrate a novel mechanism through which 1,25(OH)2D regulates lipid metab. in malignant breast epithelial cells.
- 44Liu, X.; Kim, C. N.; Yang, J.; Jemmerson, R.; Wang, X. Induction of Apoptotic Program in Cell-Free Extracts: Requirement for DATP and Cytochrome C. Cell 1996, 86 (1), 147– 157, DOI: 10.1016/S0092-8674(00)80085-944https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28XktlGnsbY%253D&md5=5789cef4b6161cbf1faff51ecc0e0096Induction of apoptotic program in cell-free extracts: requirement for dATP and cytochrome cLiu, Xuesong; Kim, Caryn Naekyung; Yang, Jie; Jemmerson, Ronald; Wang, XiaodongCell (Cambridge, Massachusetts) (1996), 86 (1), 147-157CODEN: CELLB5; ISSN:0092-8674. (Cell Press)A cell-free system based on cytosols of normally growing cells was established that reproduces aspects of the apoptotic program in vitro. The apoptotic program was initiated by the addn. of dATP. Fractionation of cytosol yielded a 15-kDa protein that was required for in vitro apoptosis. The absorption spectrum and protein sequence revealed that this protein is cytochrome c. Elimination of cytochrome c from cytosol by immunodepletion, or inclusion of sucrose to stabilize mitochondria during cytosol prepn., diminished the apoptotic activity. Adding cytochrome c back to the cytochrome c-depleted exts. restored their apoptotic activity. Cells undergoing apoptosis in vivo showed increased release of cytochrome c to their cytosol, suggesting that mitochondria may function in apoptosis by releasing cytochrome c.
- 45Rygula, A.; Majzner, K.; Marzec, K. M.; Kaczor, A.; Pilarczyk, M.; Baranska, M. Raman Spectroscopy of Proteins: A Review. J. Raman Spectrosc. 2013, 44 (8), 1061– 1076, DOI: 10.1002/jrs.433545https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFSgsLnI&md5=34557bc0aff6ebe5b4f23288a3ab91a6Raman spectroscopy of proteins: a reviewRygula, A.; Majzner, K.; Marzec, K. M.; Kaczor, A.; Pilarczyk, M.; Baranska, M.Journal of Raman Spectroscopy (2013), 44 (8), 1061-1076CODEN: JRSPAF; ISSN:0377-0486. (John Wiley & Sons Ltd.)A review. In this work, 26 proteins of different structure, function and properties are investigated by Raman spectroscopy with 488, 532 and 1064 nm laser lines. The excitation lines were chosen in NIR and Vis range as the most common and to show the difference due to normal and resonance effect, sometimes accompanied by the fluorescence. The selected proteins were divided, according to the Structural Classification of Proteins, into four classes according to their secondary structure, i.e. α-helical (α), β-sheet (β), mixed structures (α/β, α + β, s) and others. For all compds., FT-Raman and two Vis spectra are presented along with the detailed band assignment. To the best of our knowledge, this is the first review showing the potential of Raman spectroscopy for the measurement and anal. of such a large collection of individual proteins. This work can serve as a comprehensive vibrational spectra library, based on our and previous Raman measurements. Copyright © 2013 John Wiley & Sons, Ltd.
- 46Ogretmen, B. Sphingolipid Metabolism in Cancer Signalling and Therapy. Nat. Rev. Cancer 2017, 18 (1), 33– 50, DOI: 10.1038/nrc.2017.96There is no corresponding record for this reference.
- 47Kenny, P. A.; Lee, G. Y.; Myers, C. A.; Neve, R. M.; Semeiks, J. R.; Spellman, P. T.; Lorenz, K.; Lee, E. H.; Barcellos-Hoff, M. H.; Petersen, O. W. The Morphologies of Breast Cancer Cell Lines in Three-Dimensional Assays Correlate with Their Profiles of Gene Expression. Mol. Oncol. 2007, 1 (1), 84– 96, DOI: 10.1016/j.molonc.2007.02.00447https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtlOitbbO&md5=9370b0e1d910303d9dd3e98cac32e298The morphologies of breast cancer cell lines in three-dimensional assays correlate with their profiles of gene expressionKenny, Paraic A.; Lee, Genee Y.; Myers, Connie A.; Neve, Richard M.; Semeiks, Jeremy R.; Spellman, Paul T.; Lorenz, Katrin; Lee, Eva H.; Barcellos-Hoff, Mary Helen; Petersen, Ole W.; Gray, Joe W.; Bissell, Mina J.Molecular Oncology (2007), 1 (1), 84-96CODEN: MOONC3; ISSN:1574-7891. (Elsevier B.V.)3D cell cultures are rapidly becoming the method of choice for the physiol. relevant modeling of many aspects of non-malignant and malignant cell behavior ex vivo. Nevertheless, only a limited no. of distinct cell types have been evaluated in this assay to date. Here we report the first large scale comparison of the transcriptional profiles and 3D cell culture phenotypes of a substantial panel of human breast cancer cell lines. Each cell line adopts a colony morphol. of one of four main classes in 3D culture. These morphologies reflect, at least in part, the underlying gene expression profile and protein expression patterns of the cell lines, and distinct morphologies were also assocd. with tumor cell invasiveness and with cell lines originating from metastases. We further demonstrate that consistent differences in genes encoding signal transduction proteins emerge when even tumor cells are cultured in 3D microenvironments.
- 48Nath, S.; Devi, G. R. Three-Dimensional Culture Systems in Cancer Research: Focus on Tumor Spheroid Model. Pharmacol. Ther. 2016, 163, 94– 108, DOI: 10.1016/j.pharmthera.2016.03.01348https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XmtVSgsLk%253D&md5=5ea2141b4cd53d1959ebf2065e866ea2Three-dimensional culture systems in cancer research: Focus on tumor spheroid modelNath, Sritama; Devi, Gayathri R.Pharmacology & Therapeutics (2016), 163 (), 94-108CODEN: PHTHDT; ISSN:0163-7258. (Elsevier)Cancer cells propagated in three-dimensional (3D) culture systems exhibit physiol. relevant cell-cell and cell-matrix interactions, gene expression and signaling pathway profiles, heterogeneity and structural complexity that reflect in vivo tumors. In recent years, development of various 3D models has improved the study of host-tumor interaction and use of high-throughput screening platforms for anti-cancer drug discovery and development. This review attempts to summarize the various 3D culture systems, with an emphasis on the most well characterized and widely applied model - multicellular tumor spheroids. This review also highlights the various techniques to generate tumor spheroids, methods to characterize them, and its applicability in cancer research.
- 49Edmondson, R.; Broglie, J. J.; Adcock, A. F.; Yang, L. Three-Dimensional Cell Culture Systems and Their Applications in Drug Discovery and Cell-Based Biosensors. Assay Drug Dev. Technol. 2014, 12 (4), 207– 218, DOI: 10.1089/adt.2014.57349https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXotVantLY%253D&md5=8bf702b6f1bf095faa1c712fd802ebdbThree-Dimensional Cell Culture Systems and Their Applications in Drug Discovery and Cell-Based BiosensorsEdmondson, Rasheena; Broglie, Jessica Jenkins; Adcock, Audrey F.; Yang, LijuAssay and Drug Development Technologies (2014), 12 (4), 207-218CODEN: ADDTAR; ISSN:1540-658X. (Mary Ann Liebert, Inc.)A review. Three-dimensional (3D) cell culture systems have gained increasing interest in drug discovery and tissue engineering due to their evident advantages in providing more physiol. relevant information and more predictive data for in vivo tests. In this review, we discuss the characteristics of 3D cell culture systems in comparison to the two-dimensional (2D) monolayer culture, focusing on cell growth conditions, cell proliferation, population, and gene and protein expression profiles. The innovations and development in 3D culture systems for drug discovery over the past 5 years are also reviewed in the article, emphasizing the cellular response to different classes of anticancer drugs, focusing particularly on similarities and differences between 3D and 2D models across the field. The progression and advancement in the application of 3D cell cultures in cell-based biosensors is another focal point of this review.
- 50Tauchi-Sato, K.; Ozeki, S.; Houjou, T.; Taguchi, R.; Fujimoto, T. The Surface of Lipid Droplets Is a Phospholipid Monolayer with a Unique Fatty Acid Composition. J. Biol. Chem. 2002, 277 (46), 44507– 44512, DOI: 10.1074/jbc.M20771220050https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38Xos1akur8%253D&md5=00ecf3ba28f4744ae8c57834e818cb58The Surface of Lipid Droplets Is a Phospholipid Monolayer with a Unique Fatty Acid CompositionTauchi-Sato, Kumi; Ozeki, Shintaro; Houjou, Toshiaki; Taguchi, Ryo; Fujimoto, ToyoshiJournal of Biological Chemistry (2002), 277 (46), 44507-44512CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)We found that caveolin-2 is targeted to the surface of lipid droplets (Fujimoto, T., Kogo, H., Ishiguro, K., Tauchi, K., and Nomura, R. (2001) J. Cell Biol. 152, 1079-1085) and hypothesized that the lipid droplet surface is a kind of membrane. To elucidate the characteristics of the lipid droplet surface, we isolated lipid droplets from HepG2 cells and analyzed them by cryoelectron microscopy and by mass spectrometry. By use of cryoelectron microscopy at the stage temp. of 4.2 K, the lipid droplet surface was obsd. as a single line without any fixation or staining, indicating the presence of a single layer of phospholipids. This result appeared consistent with the hypothesis that the lipid droplet surface is derived from the cytoplasmic leaflet of the endoplasmic reticulum membrane and may be continuous to it. However, mass spectrometry revealed that the fatty acid compn. of phosphatidylcholine and lysophosphatidylcholine in lipid droplets is different from that of the rough endoplasmic reticulum. The ample presence of free cholesterol in lipid droplets also suggests that their surface is differentiated from the bulk endoplasmic reticulum membrane. On the other hand, although caveolin-2β and adipose differentiation-related protein, both localizing in lipid droplets, were enriched in the low d. floating fraction, the fatty acid compn. of the fraction was distinct from lipid droplets. Collectively, the result indicates that the lipid droplet surface is a hemi-membrane or a phospholipid monolayer contg. cholesterol but is compositionally different from the endoplasmic reticulum membrane or the sphingolipid/cholesterol-rich microdomain.
- 51Penno, A.; Hackenbroich, G.; Thiele, C. Phospholipids and Lipid Droplets. Biochim. Biophys. Acta, Mol. Cell Biol. Lipids 2013, 1831 (3), 589– 594, DOI: 10.1016/j.bbalip.2012.12.00151https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXovVOmug%253D%253D&md5=4bfa8e971f0d7401b239e57d66de16ebPhospholipids and lipid dropletsPenno, Anke; Hackenbroich, Gregor; Thiele, ChristophBiochimica et Biophysica Acta, Molecular and Cell Biology of Lipids (2013), 1831 (3), 589-594CODEN: BBMLFG; ISSN:1388-1981. (Elsevier B. V.)A review. Lipid droplets are ubiquitous cellular organelles that allow cells to store large amts. of neutral lipids for membrane synthesis and energy supply in times of starvation. Compared to other cellular organelles, lipid droplets are structurally unique as they are made of a hydrophobic core of neutral lipids and are sepd. to the cytosol only by a surrounding phospholipid monolayer. This phospholipid monolayer consists of over a hundred different phospholipid mol. species of which phosphatidylcholine is the most abundant lipid class. However, lipid droplets lack some indispensable activities of the phosphatidylcholine biogenic pathways suggesting that they partially depend on other organelles for phosphatidylcholine synthesis. Here, the authors discuss very recent data on the compn., origin, transport and function of the phospholipid monolayer with a particular emphasis on the phosphatidylcholine metab. on and for lipid droplets. In addn., we highlight two very important quant. aspects: (i) The amt. of phospholipid required for lipid droplet monolayer expansion is remarkably small and (ii) to maintain the invariably round shape of lipid droplets, a cell must have a highly sensitive but so far unknown mechanism that regulates the ratio of phospholipid to neutral lipid in lipid droplets. This article is part of a Special Issue entitled Phospholipids and Phospholipid Metab.
- 52Wilfling, F.; Wang, H.; Haas, J. T.; Krahmer, N.; Gould, T. J.; Uchida, A.; Cheng, J.-X.; Graham, M.; Christiano, R.; Fröhlich, F. Triacylglycerol Synthesis Enzymes Mediate Lipid Droplet Growth by Relocalizing from the ER to Lipid Droplets. Dev. Cell 2013, 24 (4), 384– 399, DOI: 10.1016/j.devcel.2013.01.01352https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXis1Oms7c%253D&md5=b5e30e8f6afd2ffe9e6f2f508263c4e5Triacylglycerol Synthesis Enzymes Mediate Lipid Droplet Growth by Relocalizing from the ER to Lipid DropletsWilfling, Florian; Wang, Huajin; Haas, Joel T.; Krahmer, Natalie; Gould, Travis J.; Uchida, Aki; Cheng, Ji-Xin; Graham, Morven; Christiano, Romain; Frohlich, Florian; Liu, Xinran; Buhman, Kimberly K.; Coleman, Rosalind A.; Bewersdorf, Joerg; Farese, Robert V.; Walther, Tobias C.Developmental Cell (2013), 24 (4), 384-399CODEN: DCEEBE; ISSN:1534-5807. (Cell Press)Lipid droplets (LDs) store metabolic energy and membrane lipid precursors. With excess metabolic energy, cells synthesize triacylglycerol (TG) and form LDs that grow dramatically. It is unclear how TG synthesis relates to LD formation and growth. Here, we identify two LD subpopulations: smaller LDs of relatively const. size, and LDs that grow larger. The latter population contains isoenzymes for each step of TG synthesis. Glycerol-3-phosphate acyltransferase 4 (GPAT4), which catalyzes the first and rate-limiting step, relocalizes from the endoplasmic reticulum (ER) to a subset of forming LDs, where it becomes stably assocd. ER-to-LD targeting of GPAT4 and other LD-localized TG synthesis isoenzymes is required for LD growth. Key features of GPAT4 ER-to-LD targeting and function in LD growth are conserved between Drosophila and mammalian cells. Our results explain how TG synthesis is coupled with LD growth and identify two distinct LD subpopulations based on their capacity for localized TG synthesis.
- 53Mouritsen, O. G.; Bagatolli, L. A. Life-as a Matter of Fat, 2nd ed.; Springer-Verlag: Heidelberg, 2005.There is no corresponding record for this reference.
- 54Walther, T. C.; Farese, R. V. The Life of Lipid Droplets. Biochim. Biophys. Acta, Mol. Cell Biol. Lipids 2009, 1791 (6), 459– 466, DOI: 10.1016/j.bbalip.2008.10.00954https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXntVKls70%253D&md5=7285b4c3d445dcb874c61e56926f0abcThe life of lipid dropletsWalther, Tobias C.; Farese, Robert V.Biochimica et Biophysica Acta, Molecular and Cell Biology of Lipids (2009), 1791 (6), 459-466CODEN: BBMLFG; ISSN:1388-1981. (Elsevier B. V.)A review. Lipid droplets are the least characterized of cellular organelles. Long considered simple lipid storage depots, these dynamic and remarkable organelles have recently been implicated in many biol. processes, and investigators are only now beginning to gain insights into their fascinating lives in cells. Here, the authors examine what is known of the life of lipid droplets. The authors review emerging data concerning their cellular biol. and present their thoughts on some of the most salient questions for investigation.
- 55Nieman, K. M.; Kenny, H. A.; Penicka, C. V.; Ladanyi, A.; Buell-Gutbrod, R.; Zillhardt, M. R.; Romero, I. L.; Carey, M. S.; Mills, G. B.; Hotamisligil, G. S. Adipocytes Promote Ovarian Cancer Metastasis and Provide Energy for Rapid Tumor Growth. Nat. Med. 2011, 17, 1498– 1503, DOI: 10.1038/nm.249255https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtlyis73N&md5=5a208c6ec3ad22910c8a30cc2391bd90Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growthNieman, Kristin M.; Kenny, Hilary A.; Penicka, Carla V.; Ladanyi, Andras; Buell-Gutbrod, Rebecca; Zillhardt, Marion R.; Romero, Iris L.; Carey, Mark S.; Mills, Gordon B.; Hotamisligil, Goekhan S.; Yamada, S. Diane; Peter, Marcus E.; Gwin, Katja; Lengyel, ErnstNature Medicine (New York, NY, United States) (2011), 17 (11), 1498-1503CODEN: NAMEFI; ISSN:1078-8956. (Nature Publishing Group)Intra-abdominal tumors, such as ovarian cancer, have a clear predilection for metastasis to the omentum, an organ primarily composed of adipocytes. Currently, it is unclear why tumor cells preferentially home to and proliferate in the omentum, yet omental metastases typically represent the largest tumor in the abdominal cavities of women with ovarian cancer. We show here that primary human omental adipocytes promote homing, migration and invasion of ovarian cancer cells, and that adipokines including interleukin-8 (IL-8) mediate these activities. Adipocyte-ovarian cancer cell coculture led to the direct transfer of lipids from adipocytes to ovarian cancer cells and promoted in vitro and in vivo tumor growth. Furthermore, coculture induced lipolysis in adipocytes and β-oxidn. in cancer cells, suggesting adipocytes act as an energy source for the cancer cells. A protein array identified upregulation of fatty acid-binding protein 4 (FABP4, also known as aP2) in omental metastases as compared to primary ovarian tumors, and FABP4 expression was detected in ovarian cancer cells at the adipocyte-tumor cell interface. FABP4 deficiency substantially impaired metastatic tumor growth in mice, indicating that FABP4 has a key role in ovarian cancer metastasis. These data indicate adipocytes provide fatty acids for rapid tumor growth, identifying lipid metab. and transport as new targets for the treatment of cancers where adipocytes are a major component of the microenvironment.
- 56Nieman, K. M.; Romero, I. L.; Van Houten, B.; Lengyel, E. Adipose Tissue and Adipocytes Support Tumorigenesis and Metastasis. Biochim. Biophys. Acta, Mol. Cell Biol. Lipids 2013, 1831 (10), 1533– 1541, DOI: 10.1016/j.bbalip.2013.02.01056https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXkslamsbs%253D&md5=df984d32b4483f2bfed210751383ce11Adipose tissue and adipocytes support tumorigenesis and metastasisNieman, Kristin M.; Romero, Iris L.; Van Houten, Bennett; Lengyel, ErnstBiochimica et Biophysica Acta, Molecular and Cell Biology of Lipids (2013), 1831 (10), 1533-1541CODEN: BBMLFG; ISSN:1388-1981. (Elsevier B. V.)A review. Adipose tissue influences tumor development in two major ways. First, obese individuals have a higher risk of developing certain cancers (endometrial, esophageal, and renal cell cancer). However, the risk of developing other cancers (melanoma, rectal, and ovarian) is not altered by body mass. In obesity, hypertrophied adipose tissue depots are characterized by a state of low grade inflammation. In this activated state, adipocytes and inflammatory cells secrete adipokines and cytokines which are known to promote tumor development. In addn., the adipocyte mediated conversion of androgens to estrogen specifically contributes to the development of endometrial cancer, which shows the greatest relative risk (6.3-fold) increase between lean and obese individuals. Second, many tumor types (gastric, breast, colon, renal, and ovarian) grow in the anatomical vicinity of adipose tissue. During their interaction with cancer cells, adipocytes dedifferentiate into pre-adipocytes or are reprogrammed into cancer-assocd. adipocytes (CAA). CAA secrete adipokines which stimulate the adhesion, migration, and invasion of tumor cells. Cancer cells and CAA also engage in a dynamic exchange of metabolites. Specifically, CAA release fatty acids through lipolysis which are then transferred to cancer cells and used for energy prodn. through β-oxidn. The abundant availability of lipids from adipocytes in the tumor microenvironment, supports tumor progression and uncontrolled growth. Given that adipocytes are a major source of adipokines and energy for the cancer cell, understanding the mechanisms of metabolic symbiosis between cancer cells and adipocytes, should reveal new therapeutic possibilities. This article is part of a Special Issue entitled Lipid Metab. in Cancer.
- 57Thiam, A. R.; Farese, R. V.; Walther, T. C. The Biophysics and Cell Biology of Lipid Droplets. Nat. Rev. Mol. Cell Biol. 2013, 14 (12), 775– 786, DOI: 10.1038/nrm369957https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslGntLrK&md5=f38fca0797834390268b8180715fa3e6The biophysics and cell biology of lipid dropletsThiam, Abdou Rachid; Farese, Robert V., Jr.; Walther, Tobias C.Nature Reviews Molecular Cell Biology (2013), 14 (12), 775-786CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. Lipid droplets are intracellular organelles that are found in most cells, where they have fundamental roles in metab. They function prominently in storing oil-based reserves of metabolic energy and components of membrane lipids. Lipid droplets are the dispersed phase of an oil-in-water emulsion in the aq. cytosol of cells, and the importance of basic biophys. principles of emulsions for lipid droplet biol. is now being appreciated. Because of their unique architecture, with an interface between the dispersed oil phase and the aq. cytosol, specific mechanisms underlie their formation, growth and shrinkage. Such mechanisms enable cells to use emulsified oil when the demands for metabolic energy or membrane synthesis change. The regulation of the compn. of the phospholipid surfactants at the surface of lipid droplets is crucial for lipid droplet homeostasis and protein targeting to their surfaces.
- 58Pelletier, J.; Bellot, G.; Gounon, P.; Lacas-Gervais, S.; Pouysségur, J.; Mazure, N. M. Glycogen Synthesis Is Induced in Hypoxia by the Hypoxia-Inducible Factor and Promotes Cancer Cell Survival. Front. Oncol. 2012, 2, 18, DOI: 10.3389/fonc.2012.0001858https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC38nnslShsA%253D%253D&md5=bee5fdd5bf92a923514e66dae65113e3Glycogen Synthesis is Induced in Hypoxia by the Hypoxia-Inducible Factor and Promotes Cancer Cell SurvivalPelletier Joffrey; Bellot Gregory; Gounon Pierre; Lacas-Gervais Sandra; Pouyssegur Jacques; Mazure Nathalie MFrontiers in oncology (2012), 2 (), 18 ISSN:.The hypoxia-inducible factor 1 (HIF-1), in addition to genetic and epigenetic changes, is largely responsible for alterations in cell metabolism in hypoxic tumor cells. This transcription factor not only favors cell proliferation through the metabolic shift from oxidative phosphorylation to glycolysis and lactic acid production but also stimulates nutrient supply by mediating adaptive survival mechanisms. In this study we showed that glycogen synthesis is enhanced in non-cancer and cancer cells when exposed to hypoxia, resulting in a large increase in glycogen stores. Furthermore, we demonstrated that the mRNA and protein levels of the first enzyme of glycogenesis, phosphoglucomutase1 (PGM1), were increased in hypoxia. We showed that induction of glycogen storage as well as PGM1 expression were dependent on HIF-1 and HIF-2. We established that hypoxia-induced glycogen stores are rapidly mobilized in cells that are starved of glucose. Glycogenolysis allows these "hypoxia-preconditioned" cells to confront and survive glucose deprivation. In contrast normoxic control cells exhibit a high rate of cell death following glucose removal. These findings point to the important role of hypoxia and HIF in inducing mechanisms of rapid adaptation and survival in response to a decrease in oxygen tension. We propose that a decrease in pO(2) acts as an "alarm" that prepares the cells to face subsequent nutrient depletion and to survive.
- 59Pescador, N.; Villar, D.; Cifuentes, D.; Garcia-Rocha, M.; Ortiz-Barahona, A.; Vazquez, S.; Ordoñez, A.; Cuevas, Y.; Saez-Morales, D.; Garcia-Bermejo, M. L. Hypoxia Promotes Glycogen Accumulation through Hypoxia Inducible Factor (HIF)-Mediated Induction of Glycogen Synthase 1. PLoS One 2010, 5 (3), e9644 DOI: 10.1371/journal.pone.0009644There is no corresponding record for this reference.
- 60Favaro, E.; Bensaad, K.; Chong, M. G.; Tennant, D. A.; Ferguson, D. J. P.; Snell, C.; Steers, G.; Turley, H.; Li, J.-L.; Günther, U. L. Glucose Utilization via Glycogen Phosphorylase Sustains Proliferation and Prevents Premature Senescence in Cancer Cells. Cell Metab. 2012, 16 (6), 751– 764, DOI: 10.1016/j.cmet.2012.10.01760https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhslWqur7I&md5=b74e88cc2c3cea7b355b22439d14874aGlucose Utilization via Glycogen Phosphorylase Sustains Proliferation and Prevents Premature Senescence in Cancer CellsFavaro, Elena; Bensaad, Karim; Chong, Mei G.; Tennant, Daniel A.; Ferguson, David J. P.; Snell, Cameron; Steers, Graham; Turley, Helen; Li, Ji-Liang; Guenther, Ulrich L.; Buffa, Francesca M.; McIntyre, Alan; Harris, Adrian L.Cell Metabolism (2012), 16 (6), 751-764CODEN: CMEEB5; ISSN:1550-4131. (Elsevier Inc.)Metabolic reprogramming of cancer cells provides energy and multiple intermediates crit. for cell growth. Hypoxia in tumors represents a hostile environment that can encourage these transformations. We report that glycogen metab. is upregulated in tumors in vivo and in cancer cells in vitro in response to hypoxia. In vitro, hypoxia induced an early accumulation of glycogen, followed by a gradual decline. Concordantly, glycogen synthase (GYS1) showed a rapid induction, followed by a later increase of glycogen phosphorylase (PYGL). PYGL depletion and the consequent glycogen accumulation led to increased reactive oxygen species (ROS) levels that contributed to a p53-dependent induction of senescence and markedly impaired tumorigenesis in vivo. Metabolic analyses indicated that glycogen degrdn. by PYGL is important for the optimal function of the pentose phosphate pathway. Thus, glycogen metab. is a key pathway induced by hypoxia, necessary for optimal glucose utilization, which represents a targetable mechanism of metabolic adaptation.
- 61Okada, M.; Smith, N. I.; Palonpon, A. F.; Endo, H.; Kawata, S.; Sodeoka, M.; Fujita, K. Label-Free Raman Observation of Cytochrome c Dynamics during Apoptosis. Proc. Natl. Acad. Sci. U. S. A. 2012, 109 (1), 28– 32, DOI: 10.1073/pnas.110752410861https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsVehsrg%253D&md5=05f3ec458114cd4d6d2091bf7e201114Label-free Raman observation of cytochrome c dynamics during apoptosisOkada, Masaya; Smith, Nicholas Isaac; Palonpon, Almar Flotildes; Endo, Hiromi; Kawata, Satoshi; Sodeoka, Mikiko; Fujita, KatsumasaProceedings of the National Academy of Sciences of the United States of America (2012), 109 (1), 28-32, S28/1-S28/2CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)We performed label-free observation of mol. dynamics in apoptotic cells by Raman microscopy. Dynamic changes in cytochrome c distribution at the Raman band of 750 cm-1 were obsd. after adding an apoptosis inducer to the cells. The comparison of mitochondria fluorescence images and Raman images of cytochrome c confirmed that changes in cytochrome c distribution can be distinguished as release of cytochrome c from mitochondria. Our observation also revealed that the redox state of cytochrome c was maintained during the release from the mitochondria. Monitoring mitochondrial membrane potential with JC-1 dye confirmed that the obsd. cytochrome c release was assocd. with apoptosis.
- 62Vazquez, A.; Kamphorst, J. J.; Markert, E. K.; Schug, Z. T.; Tardito, S.; Gottlieb, E. Cancer Metabolism at a Glance. J. Cell Sci. 2016, 129 (18), 3367– 3373, DOI: 10.1242/jcs.18101662https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitVyht73F&md5=04c4e6b18ac946a397558d1ec98e5e4dCancer metabolism at a glanceVazquez, Alexei; Kamphorst, Jurre J.; Markert, Elke K.; Schug, Zachary T.; Tardito, Saverio; Gottlieb, EyalJournal of Cell Science (2016), 129 (18), 3367-3373CODEN: JNCSAI; ISSN:0021-9533. (Company of Biologists Ltd.)A defining hallmark of cancer is uncontrolled cell proliferation. This is initiated once cells have accumulated alterations in signaling pathways that control metab. and proliferation, wherein the metabolic alterations provide the energetic and anabolic demands of enhanced cell proliferation. How these metabolic requirements are satisfied depends, in part, on the tumor microenvironment, which dets. the availability of nutrients and oxygen. In this Cell Science at a Glance paper and the accompanying poster, we summarize our current understanding of cancer metab., emphasizing pathways of nutrient utilization and metab. that either appear or have been proven essential for cancer cells. We also review how this knowledge has contributed to the development of anticancer therapies that target cancer metab.
- 63Worgall, T. S. Sphingolipids: Major Regulators of Lipid Metabolism. Curr. Opin. Clin. Nutr. Metab. Care 2007, 10 (2), 149– 155, DOI: 10.1097/MCO.0b013e328028fda363https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXjs1ylsro%253D&md5=052c779add3bd4133a3371ccd2ec5f26Sphingolipids: major regulators of lipid metabolismWorgall, Tilla S.Current Opinion in Clinical Nutrition and Metabolic Care (2007), 10 (2), 149-155CODEN: COCMF3; ISSN:1363-1950. (Lippincott Williams & Wilkins)A review. Emerging data strongly suggest a role of sphingolipid synthesis in the regulation of transcription factors and regulatory proteins that control cellular lipid homeostasis.
- 64Eichmann, T. O.; Lass, A. DAG Tales: The Multiple Faces of Diacylglycerol—stereochemistry, Metabolism, and Signaling. Cell. Mol. Life Sci. 2015, 72 (20), 3931– 3952, DOI: 10.1007/s00018-015-1982-364https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFCjtLnL&md5=31e9b2808d906fd24aa4ef4ab4d9aba8DAG tales: the multiple faces of diacylglycerol-stereochemistry, metabolism, and signalingEichmann, Thomas Oliver; Lass, AchimCellular and Molecular Life Sciences (2015), 72 (20), 3931-3952CODEN: CMLSFI; ISSN:1420-682X. (Birkhaeuser Basel)The neutral lipids diacylglycerols (DAGs) are involved in a plethora of metabolic pathways. They function as components of cellular membranes, as building blocks for glycero(phospho)lipids, and as lipid second messengers. Considering their central role in multiple metabolic processes and signaling pathways, cellular DAG levels require a tight regulation to ensure a const. and controlled availability. Interestingly, DAG species are versatile in their chem. structure. Besides the different fatty acid species esterified to the glycerol backbone, DAGs can occur in three different stereo/regioisoforms, each with unique biol. properties. Recent scientific advances have revealed that DAG metabolizing enzymes generate and distinguish different DAG isoforms, and that only one DAG isoform holds signaling properties. Herein, we review the current knowledge of DAG stereochem. and their impact on cellular metab. and signaling. Further, we describe intracellular DAG turnover and its stereochem. in a 3-pool model to illustrate the spatial and stereochem. sepn. and hereby the diversity of cellular DAG metab.
- 65Kadota, M.; Yang, H. H.; Gomez, B.; Sato, M.; Clifford, R. J.; Meerzaman, D.; Dunn, B. K.; Wakefield, L. M.; Lee, M. P. Delineating Genetic Alterations for Tumor Progression in the MCF10A Series of Breast Cancer Cell Lines. PLoS One 2010, 5 (2), e9201 DOI: 10.1371/journal.pone.0009201There is no corresponding record for this reference.
- 66Han, G.; Gupta, S. D.; Gable, K.; Niranjanakumari, S.; Moitra, P.; Eichler, F.; Brown, R. H.; Harmon, J. M.; Dunn, T. M. Identification of Small Subunits of Mammalian Serine Palmitoyltransferase That Confer Distinct Acyl-CoA Substrate Specificities. Proc. Natl. Acad. Sci. U. S. A. 2009, 106 (20), 8186– 8191, DOI: 10.1073/pnas.0811269106There is no corresponding record for this reference.
- 67Knapp, P.; Baranowski, M.; Knapp, M.; Zabielski, P.; Błachnio-Zabielska, A. U.; Górski, J. Altered Sphingolipid Metabolism in Human Endometrial Cancer. Prostaglandins Other Lipid Mediators 2010, 92 (1–4), 62– 66, DOI: 10.1016/j.prostaglandins.2010.03.00267https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXmtFeqsbY%253D&md5=489c4c99690000987f5d1f964d0c35e0Altered sphingolipid metabolism in human endometrial cancerKnapp, Pawel; Baranowski, Marcin; Knapp, Malgorzata; Zabielski, Piotr; Blachnio-Zabielska, Agnieszka U.; Gorski, JanProstaglandins & Other Lipid Mediators (2010), 92 (1-4), 62-66CODEN: POLMFL; ISSN:1098-8823. (Elsevier)There is a growing body of evidence indicating that bioactive sphingolipids play a key role in cancer development, progression and metastasis. However, sphingolipid metab. in malignant tumors is poorly investigated. Therefore, the aim of the present study was to examine the content of selected intermediates of ceramide metab. and the activity of key enzymes of ceramide de novo synthesis and sphingosine-1-phosphate (S1P) prodn. in the endometrial cancer. The specimens of cancer tissue and healthy endometrium were obtained from women undergoing surgery because of the cancer (n = 23) and because of myomas (n = 18), resp. The content of sphinganine, dihydroceramide, ceramide, sphingosine and S1P was measured using high pressure liq. chromatog. The activity of the enzymes was detd. using radioactive substrates. It has been found that the content of each examd. sphingolipid was markedly elevated in the cancer tissue compared with the healthy endometrium. Namely, sphinganine, sphingosine and dihydroceramide by 3-4.6-fold, ceramide and S1P by 1.9- and 1.6-fold, resp. Interestingly, the ratio of S1P to ceramide remained stable. The activity of serine palmitoyltransferase and sphingosine kinase 1 was increased by 2.3- and 2.6-fold, resp. We conclude that endometrial carcinoma is characterized by profound changes in sphingolipid metab. that likely contribute to its progression and chemoresistance.
- 68Patra, S. K. Dissecting Lipid Raft Facilitated Cell Signaling Pathways in Cancer. Biochim. Biophys. Acta, Rev. Cancer 2008, 1785 (2), 182– 206, DOI: 10.1016/j.bbcan.2007.11.00268https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXkvFWlur8%253D&md5=c3334d247415b40d2693a93d0e7fe4fbDissecting lipid raft facilitated cell signaling pathways in cancerPatra, Samir KumarBiochimica et Biophysica Acta, Reviews on Cancer (2008), 1785 (2), 182-206CODEN: BBACEU; ISSN:0304-419X. (Elsevier Ltd.)A review. Cancer is one of the most devastating disorders in our lives. Higher rate of proliferation than death of cells is one of the essential factors for development of cancer. The dynamicity of cell membrane plays some vital roles in cell survival and cell death, including protection, endocytosis, signaling, and increases in mech. stability during cell division, as well as decrease of shear forces during sepn. of two cells after division, and cell sepn. from tissues for cancer metastasis. Within the membrane, there are specialized domains, known as lipid rafts. A raft can coordinate various signaling pathways. Recent data on the proteomics of lipid rafts/caveolae have highlighted the enigmatic role of various signaling proteins in cancer development. Anal. of these data of raft proteome from various tumors, cancer tissues, and cell lines cultured without and with therapeutic agents, as well as from model rafts revealed that there may be two subsets of raft assemblage in cell membrane. One subset of raft is enriched with cholesterol-sphingomyelins-ganglioside-cav-1/Src/EGFR (hereafter, "chol-raft") that is involved in normal cell signaling, and when dysregulated promotes cell transformation and tumor progression; another subset of raft is enriched with ceramide-sphingomyeline-ganglioside-FAS/Ezrin (hereafter, "cer-raft") that generally promotes apoptosis. In view of this, and to focus insight into the cancer cell physiol. caused by the lipid rafts mediated signals and their receptors, and the downstream transmitters, either proliferative (for example, EGF and EGFR) or death-inducing (for example, FASL and FAS), and the precise roles of some therapeutic drugs and endogenous acid sphingomylenase in this scenario in in situ transformation of "chol-raft" into "cer-raft" are summarized and discussed in this contribution.
- 69Hannun, Y. A.; Obeid, L. M. Principles of Bioactive Lipid Signalling: Lessons from Sphingolipids. Nat. Rev. Mol. Cell Biol. 2008, 9 (2), 139– 150, DOI: 10.1038/nrm232969https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXovFOntg%253D%253D&md5=0bee4e560d083ef4c43f7d0b591a0997Principles of bioactive lipid signalling: lessons from sphingolipidsHannun, Yusuf A.; Obeid, Lina M.Nature Reviews Molecular Cell Biology (2008), 9 (2), 139-150CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. It has become increasingly difficult to find an area of cell biol. in which lipids do not have important, if not key, roles as signaling and regulatory mols. The rapidly expanding field of bioactive lipids is exemplified by many sphingolipids, such as ceramide, sphingosine, sphingosine-1-phosphate (S1P), ceramide-1-phosphate, and lyso-sphingomyelin, which have roles in the regulation of cell growth, death, senescence, adhesion, migration, inflammation, angiogenesis, and intracellular trafficking. Deciphering the mechanisms of these varied cell functions necessitates an understanding of the complex pathways of sphingolipid metab. and the mechanisms that regulate lipid generation and lipid action.
- 70Wallner, S.; Schmitz, G. Plasmalogens the Neglected Regulatory and Scavenging Lipid Species. Chem. Phys. Lipids 2011, 164 (6), 573– 589, DOI: 10.1016/j.chemphyslip.2011.06.00870https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXpvFCnurc%253D&md5=762062e2e7f960550f689b5fd05952c9Plasmalogens the neglected regulatory and scavenging lipid speciesWallner, Stefan; Schmitz, GerdChemistry and Physics of Lipids (2011), 164 (6), 573-589CODEN: CPLIA4; ISSN:0009-3084. (Elsevier Ltd.)A review. Plasmalogens are a class of phospholipids carrying a vinyl ether bond in sn-1 and an ester bond in sn-2 position of the glycerol backbone. Although they are widespread in all tissues and represent up to 18% of the total phospholipid mass in humans, their physiol. function is still poorly understood. The aim of this review is to give an overview over the current knowledge in plasmalogen biol. and pathol. with an emphasis on neglected aspects of their involvement in neurol. and metabolic diseases. Furthermore a better understanding of plasmalogen biol. in health and disease could also lead to the development of better diagnostic and prognostic biomarkers for vascular and metabolic diseases such as obesity and diabetes mellitus, inflammation, neuro-degeneration and cancer.
- 71Benjamin, D. I.; Cozzo, A.; Ji, X.; Roberts, L. S.; Louie, S. M.; Mulvihill, M. M.; Luo, K.; Nomura, D. K. Ether Lipid Generating Enzyme AGPS Alters the Balance of Structural and Signaling Lipids to Fuel Cancer Pathogenicity. Proc. Natl. Acad. Sci. U. S. A. 2013, 110 (37), 14912– 14917, DOI: 10.1073/pnas.131089411071https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsFWrsrrP&md5=06519e3d17983acbc3d159d5f19c21afEther lipid generating enzyme AGPS alters the balance of structural and signaling lipids to fuel cancer pathogenicityBenjamin, Daniel I.; Cozzo, Alyssa; Ji, Xiaodan; Roberts, Lindsay S.; Louie, Sharon M.; Mulvihill, Melinda M.; Luo, Kunxin; Nomura, Daniel K.Proceedings of the National Academy of Sciences of the United States of America (2013), 110 (37), 14912-14917,S14912/1-S14912/9CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Aberrant lipid metab. is an established hallmark of cancer cells. In particular, ether lipid levels have been shown to be elevated in tumors, but their specific function in cancer remains elusive. We show here that the metabolic enzyme alkylglyceronephosphate synthase (AGPS), a crit. step in the synthesis of ether lipids, is up-regulated across multiple types of aggressive human cancer cells and primary tumors. We demonstrate that ablation of AGPS in cancer cells results in reduced cell survival, cancer aggressiveness, and tumor growth through altering the balance of ether lipid, fatty acid, eicosanoid, and fatty acid-derived glycerophospholipid metab., resulting in an overall redn. in the levels of several oncogenic signaling lipids. Taken together, our results reveal that AGPS, in addn. to maintaining ether lipids, also controls cellular utilization of fatty acids, favoring the generation of signaling lipids necessary for promoting the aggressive features of cancer.
- 72Piano, V.; Benjamin, D. I.; Valente, S.; Nenci, S.; Marrocco, B.; Mai, A.; Aliverti, A.; Nomura, D. K.; Mattevi, A. Discovery of Inhibitors for the Ether Lipid-Generating Enzyme AGPS as Anti-Cancer Agents. ACS Chem. Biol. 2015, 10 (11), 2589– 2597, DOI: 10.1021/acschembio.5b00466There is no corresponding record for this reference.
- 73Dean, J. M.; Lodhi, I. J. Structural and Functional Roles of Ether Lipids. Protein Cell 2018, 9 (2), 196– 206, DOI: 10.1007/s13238-017-0423-573https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXotVKmtrs%253D&md5=f0e2a74a3f4b51f9397531847d92d79dStructural and functional roles of ether lipidsDean, John M.; Lodhi, Irfan J.Protein & Cell (2018), 9 (2), 196-206CODEN: PCREFB; ISSN:1674-800X. (Higher Education Press)Ether lipids, such as plasmalogens, are peroxisome-derived glycerophospholipids in which the hydrocarbon chain at the sn-1 position of the glycerol backbone is attached by an ether bond, as opposed to an ester bond in the more common diacyl phospholipids. This seemingly simple biochem. change has profound structural and functional implications. Notably, the tendency of ether lipids to form non-lamellar inverted hexagonal structures in model membranes suggests that they have a role in facilitating membrane fusion processes. Ether lipids are also important for the organization and stability of lipid raft microdomains, cholesterol-rich membrane regions involved in cellular signaling. In addn. to their structural roles, a subset of ether lipids are thought to function as endogenous antioxidants, and emerging studies suggest that they are involved in cell differentiation and signaling pathways. Here, we review the biol. of ether lipids and their potential significance in human disorders, including neurol. diseases, cancer, and metabolic disorders.
- 74Koivuniemi, A. The Biophysical Properties of Plasmalogens Originating from Their Unique Molecular Architecture. FEBS Lett. 2017, 591 (18), 2700– 2713, DOI: 10.1002/1873-3468.1275474https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXht1GgtbnF&md5=cb7c33b4ba34883680edb584e32205f4The biophysical properties of plasmalogens originating from their unique molecular architectureKoivuniemi, ArtturiFEBS Letters (2017), 591 (18), 2700-2713CODEN: FEBLAL; ISSN:0014-5793. (Wiley-Blackwell)A review. Plasmalogens are a unique class of phospholipids that are present in many organisms. Their presence in cell membranes has intrigued researchers for decades due to their unique mol. structure, namely the vinyl-ether bond at the sn-1 position, and their assocn. with brain related disorders. Apparently, based on their amt. in the cell membranes, their function is to provide exclusive structural and dynamical properties to these complex mol. assemblies. Yet, many of their physiol. roles manifested through their biophys. properties have been challenging to identify. In this review, the biophys. properties of plasmalogens are discussed and compared to other lipid species. The role of plasmalogens is examd. in the context of cell membrane function, and some future directions are given.
- 75Zeno, W. F.; Baul, U.; Snead, W. T.; DeGroot, A. C. M.; Wang, L.; Lafer, E. M.; Thirumalai, D.; Stachowiak, J. C. Synergy between Intrinsically Disordered Domains and Structured Proteins Amplifies Membrane Curvature Sensing. Nat. Commun. 2018, 9, 4152, DOI: 10.1038/s41467-018-06532-375https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3czntVSrsQ%253D%253D&md5=d04d08deb4eb772d317e0da978f09d14Synergy between intrinsically disordered domains and structured proteins amplifies membrane curvature sensingZeno Wade F; Snead Wilton T; DeGroot Andre C M; Stachowiak Jeanne C; Baul Upayan; Thirumalai D; Wang Liping; Lafer Eileen M; Stachowiak Jeanne CNature communications (2018), 9 (1), 4152 ISSN:.The ability of proteins to sense membrane curvature is essential to cellular function. All known sensing mechanisms rely on protein domains with specific structural features such as wedge-like amphipathic helices and crescent-shaped BAR domains. Yet many proteins that contain these domains also contain large intrinsically disordered regions. Here we report that disordered domains are themselves potent sensors of membrane curvature. Comparison of Monte Carlo simulations with in vitro and live-cell measurements demonstrates that the polymer-like behavior of disordered domains found in endocytic proteins drives them to partition preferentially to convex membrane surfaces, which place fewer geometric constraints on their conformational entropy. Further, proteins containing both structured curvature sensors and disordered regions are more than twice as curvature sensitive as their respective structured domains alone. These findings demonstrate an entropic mechanism of curvature sensing that is independent of protein structure and illustrate how structured and disordered domains can synergistically enhance curvature sensitivity.
- 76Huang, C.; Cao, Z.; Ma, J.; Shen, Y.; Bu, Y.; Khoshaba, R.; Shi, G.; Huang, D.; Liao, D.-F.; Ji, H. AKR1B10 Activates Diacylglycerol (DAG) Second Messenger in Breast Cancer Cells. Mol. Carcinog. 2018, 57 (10), 1300– 1310, DOI: 10.1002/mc.2284476https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXht1ans7rF&md5=c1427c667bdb536e64be522e34c93fe8AKR1B10 activates diacylglycerol (DAG) second messenger in breast cancer cellsHuang, Chenfei; Cao, Zhe; Ma, Jun; Shen, Yi; Bu, Yiwen; Khoshaba, Ramina; Shi, Guiyuan; Huang, Dan; Liao, Duan-Fang; Ji, Haitao; Jin, Junfei; Cao, DeliangMolecular Carcinogenesis (2018), 57 (10), 1300-1310CODEN: MOCAE8; ISSN:0899-1987. (Wiley-Blackwell)Aldo-keto reductase 1B10 (AKR1B10) is upregulated in breast cancer and promotes tumor growth and metastasis. However, little is known of the mol. mechanisms of action. Herein we report that AKR1B10 activates lipid second messengers to stimulate cell proliferation. Our data showed that ectopic expression of AKR1B10 in breast cancer cells MCF-7 promoted lipogenesis and enhanced levels of lipid second messengers, including phosphatidylinositol bisphosphate (PIP2), diacylglycerol (DAG), and inositol triphosphate (IP3). In contrast, silencing of AKR1B10 in breast cancer cells BT-20 and colon cancer cells HCT-8 led to decrease of these lipid messengers. Qual. analyses by liq. chromatog.-mass spectrum (LC-MS) revealed that AKR1B10 regulated the cellular levels of total DAG and majority of subspecies. This in turn modulated the phosphorylation of protein kinase C (PKC) isoforms PKCδ (Thr505), PKCμ (Ser744/748), and PKCα/βII (Thr638/641) and activity of the PKC-mediated c-Raf/MEK/ERK signaling cascade. A pan inhibitor of PKC (Go6983) blocked ERK1/2 activation by AKR1B10. In these cells, phospho-p90RSK, phospho-MSK, and Cyclin D1 expression was increased by AKR1B10, and pharmacol. inhibition of the ERK signaling cascade with MEK1/2 inhibitors U0126 and PD98059 eradicated induction of phospho-p90RSK, phospho-MSK, and Cyclin D1. In breast cancer cells, AKR1B10 promoted the clonogenic growth and proliferation of breast cancer cells in two-dimension (2D) and three-dimension (3D) cultures and tumor growth in immunodeficient female nude mice through activation of the PKC/ERK pathway. These data suggest that AKR1B10 stimulates breast cancer cell growth and proliferation through activation of DAG-mediated PKC/ERK signaling pathway.
- 77Kunitake, J. A. M. R.; Choi, S.; Nguyen, K. X.; Lee, M. M.; He, F.; Sudilovsky, D.; Morris, P. G.; Jochelson, M. S.; Hudis, C. A.; Muller, D. A. Correlative Imaging Reveals Physiochemical Heterogeneity of Microcalcifications in Human Breast Carcinomas. J. Struct. Biol. 2018, 202 (1), 25– 34, DOI: 10.1016/j.jsb.2017.12.002There is no corresponding record for this reference.
Supporting Information
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acscentsci.8b00932.
Methods, LCMS and lipid staining data, Raman analysis information, NMF and NMF validations (PDF)
High Resolution Figure 1 and Metaboanalyst Download (ZIP)
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.