1
|
Hindes MT, McElligott AM, Best OG, Ward MP, Selemidis S, Miles MA, Nturubika BD, Gregory PA, Anderson PH, Logan JM, Butler LM, Waugh DJ, O'Leary JJ, Hickey SM, Thurgood LA, Brooks DA. Metabolic reprogramming, malignant transformation and metastasis: Lessons from chronic lymphocytic leukaemia and prostate cancer. Cancer Lett 2025; 611:217441. [PMID: 39755364 DOI: 10.1016/j.canlet.2025.217441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/22/2024] [Accepted: 01/01/2025] [Indexed: 01/06/2025]
Abstract
Metabolic reprogramming is a hallmark of cancer, crucial for malignant transformation and metastasis. Chronic lymphocytic leukaemia (CLL) and prostate cancer exhibit similar metabolic adaptations, particularly in glucose and lipid metabolism. Understanding this metabolic plasticity is crucial for identifying mechanisms contributing to metastasis. This review considers glucose and lipid metabolism in CLL and prostate cancer, exploring their roles in healthy and malignant states and during disease progression. In CLL, lipid metabolism supports cell survival and migration, with aggressive disease characterised by increased fatty acid oxidation and altered sphingolipids. Richter's transformation and aggressive lymphoma, however, exhibit a metabolic shift towards increased glycolysis. Similarly, prostate cell metabolism is unique, relying on citrate production in the healthy state and undergoing metabolic reprogramming during malignant transformation. Early-stage prostate cancer cells increase lipid synthesis and uptake, and decrease glycolysis, whereas metastatic cells re-adopt glucose metabolism, likely driven by interactions with the tumour microenvironment. Genetic drivers including TP53 and ATM mutations connect metabolic alterations to disease severity in these two malignancies. The bone microenvironment supports the metabolic demands of these malignancies, serving as an initiation niche for CLL and a homing site for prostate cancer metastases. By comparing these malignancies, this review underscores the importance of metabolic plasticity in cancer progression and highlights how CLL and prostate cancer may be models of circulating and solid tumours more broadly. The metabolic phenotypes throughout cancer cell transformation and metastasis, and the microenvironment in which these processes occur, present opportunities for interventions that could disrupt metastatic processes and improve patient outcomes.
Collapse
Affiliation(s)
- Madison T Hindes
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia.
| | - Anthony M McElligott
- Discipline of Haematology, School of Medicine, Trinity Translational Medicine Institute, St. James's Hospital and Trinity College, Dublin, Ireland
| | - Oliver G Best
- Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, Australia
| | - Mark P Ward
- Department of Histopathology, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Stavros Selemidis
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Bundoora, Victoria, Australia
| | - Mark A Miles
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Bundoora, Victoria, Australia
| | - Bukuru D Nturubika
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Philip A Gregory
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Paul H Anderson
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Jessica M Logan
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Lisa M Butler
- South Australian ImmunoGENomics Cancer Institute and Freemasons Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, Australia; Solid Tumour Program, Precision Cancer Medicine theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - David J Waugh
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Shane M Hickey
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Lauren A Thurgood
- Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, Australia
| | - Douglas A Brooks
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia; Department of Histopathology, Trinity College Dublin, St. James's Hospital, Dublin, Ireland.
| |
Collapse
|
2
|
Din ZU, Cui B, Wang C, Zhang X, Mehmood A, Peng F, Liu Q. Crosstalk between lipid metabolism and EMT: emerging mechanisms and cancer therapy. Mol Cell Biochem 2025; 480:103-118. [PMID: 38622439 DOI: 10.1007/s11010-024-04995-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/19/2024] [Indexed: 04/17/2024]
Abstract
Lipids are the key component of all membranes composed of a variety of molecules that transduce intracellular signaling and provide energy to the cells in the absence of nutrients. Alteration in lipid metabolism is a major factor for cancer heterogeneity and a newly identified cancer hallmark. Reprogramming of lipid metabolism affects the diverse cancer phenotypes, especially epithelial-mesenchymal transition (EMT). EMT activation is considered to be an essential step for tumor metastasis, which exhibits a crucial role in the biological processes including development, wound healing, and stem cell maintenance, and has been widely reported to contribute pathologically to cancer progression. Altered lipid metabolism triggers EMT and activates multiple EMT-associated oncogenic pathways. Although the role of lipid metabolism-induced EMT in tumorigenesis is an attractive field of research, there are still significant gaps in understanding the underlying mechanisms and the precise contributions of this interplay. Further study is needed to clarify the specific molecular mechanisms driving the crosstalk between lipid metabolism and EMT, as well as to determine the potential therapeutic implications. The increased dependency of tumor cells on lipid metabolism represents a novel therapeutic target, and targeting altered lipid metabolism holds promise as a strategy to suppress EMT and ultimately inhibit metastasis.
Collapse
Affiliation(s)
- Zaheer Ud Din
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research, Guangdong Medical University, Dongguan, China
| | - Bai Cui
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Cenxin Wang
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China
| | - Xiaoyu Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China
| | - Arshad Mehmood
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Fei Peng
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China.
| | - Quentin Liu
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China.
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, 510060, China.
| |
Collapse
|
3
|
Tomecka P, Kunachowicz D, Górczyńska J, Gebuza M, Kuźnicki J, Skinderowicz K, Choromańska A. Factors Determining Epithelial-Mesenchymal Transition in Cancer Progression. Int J Mol Sci 2024; 25:8972. [PMID: 39201656 PMCID: PMC11354349 DOI: 10.3390/ijms25168972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 09/02/2024] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a process in which an epithelial cell undergoes multiple modifications, acquiring both morphological and functional characteristics of a mesenchymal cell. This dynamic process is initiated by various inducing signals that activate numerous signaling pathways, leading to the stimulation of transcription factors. EMT plays a significant role in cancer progression, such as metastasis and tumor heterogeneity, as well as in drug resistance. In this article, we studied molecular mechanisms, epigenetic regulation, and cellular plasticity of EMT, as well as microenvironmental factors influencing this process. We included both in vivo and in vitro models in EMT investigation and clinical implications of EMT, such as the use of EMT in curing oncological patients and targeting its use in therapies. Additionally, this review concludes with future directions and challenges in the wide field of EMT.
Collapse
Affiliation(s)
- Paulina Tomecka
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (P.T.); (J.G.); (M.G.); (J.K.); (K.S.)
| | - Dominika Kunachowicz
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland;
| | - Julia Górczyńska
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (P.T.); (J.G.); (M.G.); (J.K.); (K.S.)
| | - Michał Gebuza
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (P.T.); (J.G.); (M.G.); (J.K.); (K.S.)
| | - Jacek Kuźnicki
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (P.T.); (J.G.); (M.G.); (J.K.); (K.S.)
| | - Katarzyna Skinderowicz
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (P.T.); (J.G.); (M.G.); (J.K.); (K.S.)
| | - Anna Choromańska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland
| |
Collapse
|
4
|
Salloom RJ, Ahmad IM, Abdalla MY. Targeting heme degradation pathway augments prostate cancer cell sensitivity to docetaxel-induced apoptosis and attenuates migration. Front Oncol 2024; 14:1431362. [PMID: 39091910 PMCID: PMC11291216 DOI: 10.3389/fonc.2024.1431362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/02/2024] [Indexed: 08/04/2024] Open
Abstract
Introduction Chemotherapy, notably docetaxel (Doc), stands as the primary treatment for castration-resistant prostate cancer (CRPC). However, its efficacy is hindered by side effects and chemoresistance. Hypoxia in prostate cancer (PC) correlates with chemoresistance to Doc-induced apoptosis via Heme Oxygenase-1 (HO-1) modulation, a key enzyme in heme metabolism. This study investigated targeting heme degradation pathway via HO-1 inhibition to potentiate the therapeutic efficacy of Doc in PC. Methods Utilizing diverse PC cell lines, we evaluated HO-1 inhibition alone and with Doc on viability, apoptosis, migration, and epithelial- to- mesenchymal transition (EMT) markers and elucidated the underlying mechanisms. Results HO-1 inhibition significantly reduced PC cell viability under hypoxic and normoxic conditions, enhancing Doc-induced apoptosis through interconnected mechanisms, including elevated reactive oxygen species (ROS) levels, glutathione cycle disruption, and modulation of Signal Transducer and Activator of Transcription 1 (STAT1) pathway. The interplay between STAT1 and HO-1 suggests its reliance on HO-1 activation. Additionally, a decrease in cell migration and downregulation of EMT markers (vimentin and snail) were observed, indicating attenuation of mesenchymal phenotype. Discussion In conclusion, the combination of HO-1 inhibition with Doc holds promise for improving therapeutic outcomes and advancing clinical management in PC.
Collapse
Affiliation(s)
- Ramia J. Salloom
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Iman M. Ahmad
- Department of Clinical, Diagnostic, and Therapeutic Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Maher Y. Abdalla
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
5
|
Shi J, Lv Q, Miao D, Xiong Z, Wei Z, Wu S, Tan D, Wang K, Zhang X. HIF2α Promotes Cancer Metastasis through TCF7L2-Dependent Fatty Acid Synthesis in ccRCC. RESEARCH (WASHINGTON, D.C.) 2024; 7:0322. [PMID: 38390305 PMCID: PMC10882601 DOI: 10.34133/research.0322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 01/27/2024] [Indexed: 02/24/2024]
Abstract
Recent studies have highlighted the notable involvement of the crosstalk between hypoxia-inducible factor 2 alpha (HIF2α) and Wnt signaling components in tumorigenesis. However, the cellular function and precise regulatory mechanisms of HIF2α and Wnt signaling interactions in clear cell renal cell carcinoma (ccRCC) remain elusive. To analyze the correlation between HIF2α and Wnt signaling, we utilized the Cancer Genome Atlas - Kidney Renal Clear Cell Carcinoma (TCGA-KIRC) public database, HIF2α RNA sequencing data, and conducted luciferase reporter assays. A Wnt-related gene set was employed to identify key regulators of Wnt signaling controlled by HIF2α in ccRCC. Furthermore, we assessed the biological effects of TCF7L2 on ccRCC metastasis and lipid metabolism in both in vivo and in vitro settings. Our outcomes confirm TCF7L2 as a key gene involved in HIF2α-mediated regulation of the canonical Wnt pathway. Functional studies demonstrate that TCF7L2 promotes metastasis in ccRCC. Mechanistic investigations reveal that HIF2α stabilizes TCF7L2 mRNA in a method based on m6A by transcriptionally regulating METTL3. Up-regulation of TCF7L2 enhances cellular fatty acid oxidation, which promotes histone acetylation. This facilitates the transcription of genes connected to epithelial-mesenchymal transition and ultimately enhances metastasis of ccRCC. These outcomes offer a novel understanding into the involvement of lipid metabolism in the signaling pathway regulation, offering valuable implications for targeted treatment in ccRCC.
Collapse
Affiliation(s)
- Jian Shi
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
| | - Qingyang Lv
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
| | - Daojia Miao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
| | - Zhiyong Xiong
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
| | - Zhihao Wei
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
| | - Songming Wu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
| | - Diaoyi Tan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
| | - Keshan Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
| |
Collapse
|
6
|
Mukerjee N, Nag S, Bhattacharya B, Alexiou A, Mirgh D, Mukherjee D, Adhikari MD, Anand K, Muthusamy R, Gorai S, Thorat N. Clinical impact of epithelial–mesenchymal transition for cancer therapy. CLINICAL AND TRANSLATIONAL DISCOVERY 2024; 4. [DOI: 10.1002/ctd2.260] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2025]
Abstract
AbstractThe epithelial–mesenchymal transition (EMT) represents a pivotal frontier in oncology, playing a central role in the metastatic cascade of cancer—a leading global health challenge. This comprehensive review delves into the complexities of EMT, a process where cancer cells gain exceptional mobility, facilitating their invasion into distant organs and the establishment of secondary malignancies. We thoroughly examine the myriad of factors influencing EMT, encompassing transcription factors, signalling pathways, metabolic alterations, microRNAs, long non‐coding RNAs, epigenetic changes, exosomal interactions and the intricate dynamics of the tumour microenvironment. Particularly, the review emphasises the advanced stages of EMT, crucial for the development of highly aggressive cancer phenotypes. During this phase, cancer cells penetrate the vascular barrier and exploit the bloodstream to propagate life‐threatening metastases through the mesenchymal–epithelial transition. We also explore EMT's significant role in fostering tumour dormancy, senescence, the emergence of cancer stem cells and the formidable challenge of therapeutic resistance. Our review transcends a mere inventory of EMT‐inducing elements; it critically assesses the current state of EMT‐focused clinical trials, revealing both the hurdles and significant breakthroughs. Highlighting the potential of EMT research, we project its transformative impact on the future of cancer therapy. This exploration is aimed at paving the way towards an era of effectively managing this relentless disease, positioning EMT at the forefront of innovative cancer research strategies.
Collapse
Affiliation(s)
- Nobendu Mukerjee
- Department of Microbiology West Bengal State University, Barasat Kolkata India
| | - Sagnik Nag
- Department of Bio‐Sciences School of Biosciences & Technology Vellore Institute of Technology Vellore Tamil Nadu India
| | - Bikramjit Bhattacharya
- Department of Applied Microbiology School of Biosciences and Technology Vellore Institute of Technology Vellore Tamil Nadu India
| | - Athanasios Alexiou
- Department of Science and Engineering Novel Global Community Educational Foundation Hebersham New South Wales Australia
| | - Divya Mirgh
- Vaccine and Immunotherapy Canter Massachusetts General Hospital Boston Massachusetts USA
| | | | - Manab Deb Adhikari
- Department of Biotechnology University of North Bengal Darjeeling West Bengal India
| | - Krishnan Anand
- Department of Chemical Pathology School of Pathology Faculty of Health Sciences University of the Free State Bloemfontein South Africa
| | - Raman Muthusamy
- Center for Global Health Research Saveetha Medical College & Hospitals, Saveetha Institute of Medical and Technical Sciences Chennai Tamil Nadu India
| | | | - Nanasaheb Thorat
- Limerick Digital Cancer Research Centre and Department of Physics Bernal Institute University of Limerick, Castletroy Limerick Ireland
| |
Collapse
|
7
|
Saha A, Kolonin MG, DiGiovanni J. Obesity and prostate cancer - microenvironmental roles of adipose tissue. Nat Rev Urol 2023; 20:579-596. [PMID: 37198266 DOI: 10.1038/s41585-023-00764-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2023] [Indexed: 05/19/2023]
Abstract
Obesity is known to have important roles in driving prostate cancer aggressiveness and increased mortality. Multiple mechanisms have been postulated for these clinical observations, including effects of diet and lifestyle, systemic changes in energy balance and hormonal regulation and activation of signalling by growth factors and cytokines and other components of the immune system. Over the past decade, research on obesity has shifted towards investigating the role of peri-prostatic white adipose tissue as an important source of locally produced factors that stimulate prostate cancer progression. Cells that comprise white adipose tissue, the adipocytes and their progenitor adipose stromal cells (ASCs), which proliferate to accommodate white adipose tissue expansion in obesity, have been identified as important drivers of obesity-associated cancer progression. Accumulating evidence suggests that adipocytes are a source of lipids that are used by adjacent prostate cancer cells. However, results of preclinical studies indicate that ASCs promote tumour growth by remodelling extracellular matrix and supporting neovascularization, contributing to the recruitment of immunosuppressive cells, and inducing epithelial-mesenchymal transition through paracrine signalling. Because epithelial-mesenchymal transition is associated with cancer chemotherapy resistance and metastasis, ASCs are considered to be potential targets of therapies that could be developed to suppress cancer aggressiveness in patients with obesity.
Collapse
Affiliation(s)
- Achinto Saha
- Division of Pharmacology and Toxicology and Dell Paediatric Research Institute, The University of Texas at Austin, Austin, TX, USA
- Center for Molecular Carcinogenesis and Toxicology, The University of Texas at Austin, Austin, TX, USA
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Mikhail G Kolonin
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Disease, The University of Texas Health Sciences Center at Houston, Houston, Texas, USA.
| | - John DiGiovanni
- Division of Pharmacology and Toxicology and Dell Paediatric Research Institute, The University of Texas at Austin, Austin, TX, USA.
- Center for Molecular Carcinogenesis and Toxicology, The University of Texas at Austin, Austin, TX, USA.
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
8
|
Haerinck J, Goossens S, Berx G. The epithelial-mesenchymal plasticity landscape: principles of design and mechanisms of regulation. Nat Rev Genet 2023; 24:590-609. [PMID: 37169858 DOI: 10.1038/s41576-023-00601-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2023] [Indexed: 05/13/2023]
Abstract
Epithelial-mesenchymal plasticity (EMP) enables cells to interconvert between several states across the epithelial-mesenchymal landscape, thereby acquiring hybrid epithelial/mesenchymal phenotypic features. This plasticity is crucial for embryonic development and wound healing, but also underlies the acquisition of several malignant traits during cancer progression. Recent research using systems biology and single-cell profiling methods has provided novel insights into the main forces that shape EMP, which include the microenvironment, lineage specification and cell identity, and the genome. Additionally, key roles have emerged for hysteresis (cell memory) and cellular noise, which can drive stochastic transitions between cell states. Here, we review these forces and the distinct but interwoven layers of regulatory control that stabilize EMP states or facilitate epithelial-mesenchymal transitions (EMTs) and discuss the therapeutic potential of manipulating the EMP landscape.
Collapse
Affiliation(s)
- Jef Haerinck
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Steven Goossens
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Unit for Translational Research in Oncology, Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Geert Berx
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| |
Collapse
|
9
|
Wang R, Zhao J, Jin J, Tian Y, Lan L, Wang X, Zhu L, Wang J. WY-14643 attenuates lipid deposition via activation of the PPARα/CPT1A axis by targeting Gly335 to inhibit cell proliferation and migration in ccRCC. Lipids Health Dis 2022; 21:121. [DOI: 10.1186/s12944-022-01726-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/19/2022] [Indexed: 11/17/2022] Open
Abstract
Abstract
Background
Histologically, cytoplasmic deposits of lipids and glycogen are common in clear cell renal cell carcinoma (ccRCC). Owing to the significance of lipid deposition in ccRCC, numerous trials targeting lipid metabolism have shown certain therapeutic potential. The agonism of peroxisome proliferator-activated receptor-α (PPARα) via ligands, including WY-14,643, has been considered a promising intervention for cancers.
Methods
First, the effects of WY-14,643 on malignant behaviors were investigated in ccRCC in vitro. After RNA sequencing, the changes in lipid metabolism, especially neutral lipids and glycerol, were further evaluated. Finally, the underlying mechanisms were revealed.
Results
Phenotypically, the proliferation and migration of ccRCC cells treated with WY-14,643 were significantly inhibited in vitro. A theoretical functional mechanism was proposed in ccRCC: WY-14,643 mediates lipid consumption by recognizing carnitine palmitoyltransferase 1 A (CPT1A). Activation of PPARα using WY-14,643 reduces lipid deposition by increasing the CPT1A level, which also suppresses the NF-κB signaling pathway. Spatially, WY-14,643 binds and activates PPARα by targeting Gly335.
Conclusion
Overall, WY-14,643 suppresses the biological behaviors of ccRCC in terms of cell proliferation, migration, and cell cycle arrest. Furthermore, its anticancer properties are mediated by the inhibition of lipid accumulation, at least in part, through the PPARα/CPT1A axis by targeting Gly335, as part of the process, NF-κB signaling is also suppressed. Pharmacological activation of PPARα might offer a new treatment option for ccRCC.
Collapse
|
10
|
Stearoyl-CoA desaturase 1 as a therapeutic target for cancer: a focus on hepatocellular carcinoma. Mol Biol Rep 2022; 49:8871-8882. [PMID: 35906508 DOI: 10.1007/s11033-021-07094-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 12/16/2021] [Indexed: 01/14/2023]
Abstract
One of the main characteristics of cancer cells is the alteration in lipid composition, which is associated with a significant monounsaturated fatty acids (MUFAs) enrichment. In addition to their structural functions in newly synthesized membranes in proliferating cancer cells, these fatty acids are involved in tumorigenic signaling. Increased expression and activity of stearoyl CoA desaturase (SCD1), i.e., an enzyme converting saturated fatty acids to Δ9-monounsaturated fatty acids, has been observed in various cancer cells. This increase in expression and activity has also been associated with cancer aggressiveness and poor patient outcome. Previous studies have also indicated the SCD1 involvement in increased cancer cells proliferation, growth, migration, epithelial to mesenchymal transition, metastasis, chemoresistance, and maintenance of cancer stem cells properties. Hence, SCD1 seems to be a player in malignancy development and may be considered a novel therapeutic target in cancers, including hepatocellular carcinoma (HCC). This review study aims to discuss the impact of SCD1 as a major component in lipid signaling in HCC.
Collapse
|
11
|
Zhou J, Ji N, Wang G, Zhang Y, Song H, Yuan Y, Yang C, Jin Y, Zhang Z, Zhang L, Yin Y. Metabolic detection of malignant brain gliomas through plasma lipidomic analysis and support vector machine-based machine learning. EBioMedicine 2022; 81:104097. [PMID: 35687958 PMCID: PMC9189781 DOI: 10.1016/j.ebiom.2022.104097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/01/2022] [Accepted: 05/20/2022] [Indexed: 12/25/2022] Open
Abstract
Background Most malignant brain gliomas (MBGs) are associated with dismal outcomes, mainly due to their late diagnosis. Current diagnostic methods for MBGs are based on imaging and histological examination, which limits their early detection. Here, we aimed to identify reliable plasma lipid biomarkers for non-invasive diagnosis for MBGs. Methods Untargeted lipidomic analysis was firstly performed using a discovery cohort (n=107). The data were processed by a support vector machine (SVM)-based discriminating model to retrieve a panel of candidate biomarkers. Then, a targeted quantification method was developed, and the SVM-based diagnostic model was constructed using a training cohort (n=750) and tested using a test cohort (n=225). Finally, the performance of the diagnostic model was further evaluated in an independent validation cohort (n=920) enrolled from multiple medical centers. Findings A panel of 11 plasma lipids was identified as candidate biomarkers with an accuracy of 0.999. The diagnostic model developed achieved a high performance in distinguishing MBGs patients from normal controls with an area under the receiver-operating characteristic curve (AUC) of 0.9877 and 0.9869 in the training and test cohorts, respectively. In the validation cohort, the 11 lipid panel still achieved an accuracy of 0.9641 and an AUC of 0.9866. Interpretation The present study demonstrates the applicability and robustness of utilizing a machine learning algorithm to analyze lipidomic data for efficient and reliable biomarker screening. The 11 lipid biomarkers show great potential for the non-invasive diagnosis of MBGs with high throughput. Funding A full list of funding bodies that contributed to this study can be found in the Acknowledgments section.
Collapse
Affiliation(s)
- Juntuo Zhou
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen 518036, China; Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Nan Ji
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - Guangxi Wang
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yang Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - Huajie Song
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yuyao Yuan
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Chunyuan Yang
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yan Jin
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Zhe Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, National Clinical Research Center for Neurological Diseases, Beijing 100070, China.
| | - Yuxin Yin
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen 518036, China; Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
12
|
Comparison of Multivariate ANOVA-Based Approaches for the Determination of Relevant Variables in Experimentally Designed Metabolomic Studies. Molecules 2022; 27:molecules27103304. [PMID: 35630781 PMCID: PMC9147242 DOI: 10.3390/molecules27103304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 05/08/2022] [Accepted: 05/19/2022] [Indexed: 02/01/2023] Open
Abstract
The use of chemometric methods based on the analysis of variances (ANOVA) allows evaluation of the statistical significance of the experimental factors used in a study. However, classical multivariate ANOVA (MANOVA) has a number of requirements that make it impractical for dealing with metabolomics data. For this reason, in recent years, different options have appeared that overcome these limitations. In this work, we evaluate the performance of three of these multivariate ANOVA-based methods (ANOVA simultaneous component analysis—ASCA, regularized MANOVA–rMANOVA, and Group-wise ANOVA-simultaneous component analysis—GASCA) in the framework of metabolomics studies. Our main goals are to compare these various ANOVA-based approaches and evaluate their performance on experimentally designed metabolomic studies to find the significant factors and identify the most relevant variables (potential markers) from the obtained results. Two experimental data sets were generated employing liquid chromatography coupled to mass spectrometry (LC-MS) with different complexity in the design to evaluate the performance of the statistical approaches. Results show that the three considered ANOVA-based methods have a similar performance in detecting statistically significant factors. However, relevant variables pointed by GASCA seem to be more reliable as there is a strong similarity with those variables detected by the widely used partial least squares discriminant analysis (PLS-DA) method.
Collapse
|
13
|
Morano C, Zulueta A, Caretti A, Roda G, Paroni R, Dei Cas M. An Update on Sphingolipidomics: Is Something Still Missing? Some Considerations on the Analysis of Complex Sphingolipids and Free-Sphingoid Bases in Plasma and Red Blood Cells. Metabolites 2022; 12:metabo12050450. [PMID: 35629954 PMCID: PMC9147510 DOI: 10.3390/metabo12050450] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 02/01/2023] Open
Abstract
The main concerns in targeted “sphingolipidomics” are the extraction and proper handling of biological samples to avoid interferences and achieve a quantitative yield well representing all the sphingolipids in the matrix. Our work aimed to compare different pre-analytical procedures and to evaluate a derivatization step for sphingoid bases quantification, to avoid interferences and improve sensitivity. We tested four protocols for the extraction of sphingolipids from human plasma, at different temperatures and durations, and two derivatization procedures for the conversion of sphingoid bases into phenylthiourea derivatives. Different columns and LC-MS/MS chromatographic conditions were also tested. The protocol that worked better for sphingolipids analysis involved a single-phase extraction in methanol/chloroform mixture (2:1, v/v) for 1 h at 38 °C, followed by a 2 h alkaline methanolysis at 38 °C, for the suppression of phospholipids signals. The derivatization of sphingoid bases promotes the sensibility of non-phosphorylated species but we proved that it is not superior to a careful choice of the appropriate column and a full-length elution gradient. Our procedure was eventually validated by analyzing plasma and erythrocyte samples of 20 volunteers. While both extraction and methanolysis are pivotal steps, our final consideration is to analyze sphingolipids and sphingoid bases under different chromatographic conditions, minding the interferences.
Collapse
Affiliation(s)
- Camillo Morano
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (C.M.); (G.R.)
| | - Aida Zulueta
- Neurorehabilitation Department, IRCCS Istituti Clinici Scientifici Maugeri di Milano, 20138 Milan, Italy;
| | - Anna Caretti
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (A.C.); (R.P.)
| | - Gabriella Roda
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (C.M.); (G.R.)
| | - Rita Paroni
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (A.C.); (R.P.)
| | - Michele Dei Cas
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (A.C.); (R.P.)
- Correspondence:
| |
Collapse
|
14
|
Quantifying the Patterns of Metabolic Plasticity and Heterogeneity along the Epithelial–Hybrid–Mesenchymal Spectrum in Cancer. Biomolecules 2022; 12:biom12020297. [PMID: 35204797 PMCID: PMC8961667 DOI: 10.3390/biom12020297] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/03/2022] [Accepted: 02/07/2022] [Indexed: 12/04/2022] Open
Abstract
Cancer metastasis is the leading cause of cancer-related mortality and the process of the epithelial-to-mesenchymal transition (EMT) is crucial for cancer metastasis. Both partial and complete EMT have been reported to influence the metabolic plasticity of cancer cells in terms of switching among the oxidative phosphorylation, fatty acid oxidation and glycolysis pathways. However, a comprehensive analysis of these major metabolic pathways and their associations with EMT across different cancers is lacking. Here, we analyse more than 180 cancer cell datasets and show the diverse associations of these metabolic pathways with the EMT status of cancer cells. Our bulk data analysis shows that EMT generally positively correlates with glycolysis but negatively with oxidative phosphorylation and fatty acid metabolism. These correlations are also consistent at the level of their molecular master regulators, namely AMPK and HIF1α. Yet, these associations are shown to not be universal. The analysis of single-cell data for EMT induction shows dynamic changes along the different axes of metabolic pathways, consistent with general trends seen in bulk samples. Further, assessing the association of EMT and metabolic activity with patient survival shows that a higher extent of EMT and glycolysis predicts a worse prognosis in many cancers. Together, our results reveal the underlying patterns of metabolic plasticity and heterogeneity as cancer cells traverse through the epithelial–hybrid–mesenchymal spectrum of states.
Collapse
|
15
|
The Epithelial-Mesenchymal Transition at the Crossroads between Metabolism and Tumor Progression. Int J Mol Sci 2022; 23:ijms23020800. [PMID: 35054987 PMCID: PMC8776206 DOI: 10.3390/ijms23020800] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
The transition between epithelial and mesenchymal phenotype is emerging as a key determinant of tumor cell invasion and metastasis. It is a plastic process in which epithelial cells first acquire the ability to invade the extracellular matrix and migrate into the bloodstream via transdifferentiation into mesenchymal cells, a phenomenon known as epithelial–mesenchymal transition (EMT), and then reacquire the epithelial phenotype, the reverse process called mesenchymal–epithelial transition (MET), to colonize a new organ. During all metastatic stages, metabolic changes, which give cancer cells the ability to adapt to increased energy demand and to withstand a hostile new environment, are also important determinants of successful cancer progression. In this review, we describe the complex interaction between EMT and metabolism during tumor progression. First, we outline the main connections between the two processes, with particular emphasis on the role of cancer stem cells and LncRNAs. Then, we focus on some specific cancers, such as breast, lung, and thyroid cancer.
Collapse
|
16
|
Menéndez-Pedriza A, Jaumot J, Bedia C. Lipidomic analysis of single and combined effects of polyethylene microplastics and polychlorinated biphenyls on human hepatoma cells. JOURNAL OF HAZARDOUS MATERIALS 2022; 421:126777. [PMID: 34364209 DOI: 10.1016/j.jhazmat.2021.126777] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/27/2021] [Accepted: 07/27/2021] [Indexed: 06/13/2023]
Abstract
Microplastics are an emerging environmental issue as a result of their ubiquity, persistence, and intrinsic toxic potential. In addition, their ability to sorb and transport a wide variety of environmental pollutants (i.e. "Trojan Horse" effect) exerts significant adverse impacts upon ecosystems. The toxicological evaluation of the single and combined effects produced by polyethylene microplastics and two polychlorinated biphenyl congeners was performed on the human hepatoma cell line HepG2 by cell viability assessment and an untargeted lipidomic study. The cell lethality evaluation evinced that MPs did not induce relevant cell lethality at any of the concentration range tested, while both PCBs presented a hormetic behavior. The lipidomic analysis suggested that both single PCB exposures induced significant lipidomic changes, especially for glycerophospholipids and glycerolipids. In contrast, for MPs single exposure, the most remarkable change was the substantial enhancement of triglyceride content. Regarding combined exposures, results showed that MPs could induce even more harmful effects than those produced intrinsically as a result of desorbing previously sorbed toxic pollutants. To the best of our knowledge, this is the first study assessing the toxicity of microplastics and their possible "Trojan Horse" effect by applying an untargeted lipidomic methodology.
Collapse
Affiliation(s)
- Albert Menéndez-Pedriza
- Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Joaquim Jaumot
- Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Carmen Bedia
- Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, 08034 Barcelona, Spain.
| |
Collapse
|
17
|
Interplay between Metabolism Reprogramming and Epithelial-to-Mesenchymal Transition in Cancer Stem Cells. Cancers (Basel) 2021; 13:cancers13081973. [PMID: 33923958 PMCID: PMC8072988 DOI: 10.3390/cancers13081973] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Tumor cells display important plasticity potential. Notably, tumor cells have the ability to change toward immature cells called cancer stem cells under the influence of the tumor environment. Importantly, cancer stem cells are a small subset of relatively quiescent cells that, unlike rapidly dividing differentiated tumor cells, escape standard chemotherapies, causing relapse or recurrence of cancer. Interestingly, these cells adopt a specific metabolism. Most often, they mainly rely on glucose uptake and metabolism to sustain their energy needs. This metabolic reprogramming is set off by environmental factors such as pro-inflammatory signals or catecholamine hormones (epinephrine, norepinephrine). A better understanding of this process could provide opportunities to kill cancer stem cells. Indeed, it would become possible to develop drugs that act specifically on metabolic pathways used by these cells. These new drugs could be used to strengthen the effects of current chemotherapies and overcome cancers with poor prognoses. Abstract Tumor cells display important plasticity potential, which contributes to intratumoral heterogeneity. Notably, tumor cells have the ability to retrodifferentiate toward immature states under the influence of their microenvironment. Importantly, this phenotypical conversion is paralleled by a metabolic rewiring, and according to the metabostemness theory, metabolic reprogramming represents the first step of epithelial-to-mesenchymal transition (EMT) and acquisition of stemness features. Most cancer stem cells (CSC) adopt a glycolytic phenotype even though cells retain functional mitochondria. Such adaptation is suggested to reduce the production of reactive oxygen species (ROS), protecting CSC from detrimental effects of ROS. CSC may also rely on glutaminolysis or fatty acid metabolism to sustain their energy needs. Besides pro-inflammatory cytokines that are well-known to initiate the retrodifferentiation process, the release of catecholamines in the microenvironment of the tumor can modulate both EMT and metabolic changes in cancer cells through the activation of EMT transcription factors (ZEB1, Snail, or Slug (SNAI2)). Importantly, the acquisition of stem cell properties favors the resistance to standard care chemotherapies. Hence, a better understanding of this process could pave the way for the development of therapies targeting CSC metabolism, providing new strategies to eradicate the whole tumor mass in cancers with unmet needs.
Collapse
|
18
|
Feng X, Zhou CK, Clish CB, Wilson KM, Pernar CH, Dickerman BA, Loda M, Finn SP, Penney KL, Schmidt DR, Heiden MGV, Giovannucci EL, Ebot EM, Mucci LA. Association of Prediagnostic Blood Metabolomics with Prostate Cancer Defined by ERG or PTEN Molecular Subtypes. Cancer Epidemiol Biomarkers Prev 2021; 30:1000-1008. [PMID: 33627383 DOI: 10.1158/1055-9965.epi-20-1363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/24/2020] [Accepted: 02/19/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The TMPRSS2:ERG gene fusion and PTEN loss are two of the most common somatic molecular alterations in prostate cancer. Here, we investigated the association of prediagnostic-circulating metabolomics and prostate cancer defined by ERG or PTEN status to improve understanding of these etiologically distinct molecular prostate cancer subtypes. METHODS The study was performed among 277 prostate cancer cases with ERG status, 211 with PTEN status, and 294 controls nested in the Health Professionals Follow-up Study (HPFS) and the Physicians' Health Study (PHS). We profiled 223 polar and non-polar metabolites using LC-MS in prediagnostic plasma specimens. We applied enrichment analysis and multinomial logistic regression models to identify biological metabolite classes and individual metabolites associated with prostate cancer defined by ERG or PTEN status. RESULTS Compared with noncancer controls, sphingomyelin (P: 0.01), ceramide (P: 0.04), and phosphatidylethanolamine (P: 0.03) circulating levels were enriched among ERG-positive prostate cancer cases. Sphingomyelins (P: 0.02), ceramides (P: 0.005), and amino acids (P: 0.02) were enriched among tumors exhibiting PTEN-loss; unsaturated diacylglycerols (P: 0.003) were enriched among PTEN-intact cases; and unsaturated triacylglycerols were enriched among both PTEN-loss (P: 0.001) and PTEN-intact (P: 0.0001) cases. Although several individual metabolites identified in the above categories were nominally associated with ERG or PTEN-defined prostate cancer, none remained significant after accounting for multiple testing. CONCLUSIONS The molecular process of prostate carcinogenesis may be distinct for men with different metabolomic profiles. IMPACT These novel findings provide insights into the metabolic environment for the development of prostate cancer.
Collapse
Affiliation(s)
- Xiaoshuang Feng
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.
| | - Cindy Ke Zhou
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | | | - Kathryn M Wilson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Claire H Pernar
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Barbra A Dickerman
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Massimo Loda
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Stephen P Finn
- Department of Histopathology Research, Trinity College, Dublin, Ireland
| | - Kathryn L Penney
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Daniel R Schmidt
- David H. Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Matthew G Vander Heiden
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,David H. Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Edward L Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Ericka M Ebot
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
19
|
Liu RZ, Choi WS, Jain S, Dinakaran D, Xu X, Han WH, Yang XH, Glubrecht DD, Moore RB, Lemieux H, Godbout R. The FABP12/PPARγ pathway promotes metastatic transformation by inducing epithelial-to-mesenchymal transition and lipid-derived energy production in prostate cancer cells. Mol Oncol 2020; 14:3100-3120. [PMID: 33031638 PMCID: PMC7718947 DOI: 10.1002/1878-0261.12818] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/11/2020] [Accepted: 10/05/2020] [Indexed: 12/31/2022] Open
Abstract
Early stage localized prostate cancer (PCa) has an excellent prognosis; however, patient survival drops dramatically when PCa metastasizes. The molecular mechanisms underlying PCa metastasis are complex and remain unclear. Here, we examine the role of a new member of the fatty acid‐binding protein (FABP) family, FABP12, in PCa progression. FABP12 is preferentially amplified and/or overexpressed in metastatic compared to primary tumors from both PCa patients and xenograft animal models. We show that FABP12 concurrently triggers metastatic phenotypes (induced epithelial‐to‐mesenchymal transition (EMT) leading to increased cell motility and invasion) and lipid bioenergetics (increased fatty acid uptake and accumulation, increased ATP production from fatty acid β‐oxidation) in PCa cells, supporting increased reliance on fatty acids for energy production. Mechanistically, we show that FABP12 is a driver of PPARγ activation which, in turn, regulates FABP12's role in lipid metabolism and PCa progression. Our results point to a novel role for a FABP‐PPAR pathway in promoting PCa metastasis through induction of EMT and lipid bioenergetics.
Collapse
Affiliation(s)
- Rong-Zong Liu
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| | - Won-Shik Choi
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| | - Saket Jain
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| | - Deepak Dinakaran
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| | - Xia Xu
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| | - Woo Hyun Han
- Faculty Saint-Jean, University of Alberta, Edmonton, AB, Canada
| | - Xiao-Hong Yang
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| | - Darryl D Glubrecht
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| | - Ronald B Moore
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada.,Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Hélène Lemieux
- Faculty Saint-Jean, University of Alberta, Edmonton, AB, Canada
| | - Roseline Godbout
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
20
|
FASN Protein Overexpression Indicates Poor Biochemical Recurrence-Free Survival in Prostate Cancer. DISEASE MARKERS 2020; 2020:3904947. [PMID: 32655718 PMCID: PMC7321525 DOI: 10.1155/2020/3904947] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 05/12/2020] [Accepted: 06/05/2020] [Indexed: 11/18/2022]
Abstract
Backgrounds Fatty acid synthase (FASN) has been regarded as a prognostic marker in prostate cancer (PCa). In this study, we evaluated FASN expression at both mRNA and protein levels and assessed the association between FASN expression and prognosis in male Han Chinese with PCa treated with radical prostatectomy (RP). Methods Expression profile and prognostic value of FASN were analyzed in tissue microarray (TMA) and data retrieved from databases including TCGA public database, GEO database, and our sequencing data with whole clinicopathological characteristics. Results FASN expression was associated with clinical parameters and biochemical recurrence of prostate cancer. The relative expression of FASN mRNA was higher in the tumor tissue in all public databases and our sequencing data (p < 0.001). A similar result was seen in tissue microarray (TMA) (p < 0.001). Analysis of our sequencing data indicated that FASN's relative expression was associated with tumor stage (p = 0.048), and FASN expression was positively associated with the Gleason score (p = 0.004) and seminal vesicle invasion (p = 0.011) in TMA. We found that high FASN expression was an independent predictor of shorter BCR-free survival with univariate and multivariate survival analysis (p < 0.05), rendering FASN an optimal prognostic biomarker in male Han Chinese with prostate cancer. Conclusions Our study demonstrated that FASN was overexpressed at mRNA and protein levels in PCa. We found that patients with high FASN expression had a shorter BCR-free survival, showing its value as a prognostic biomarker in male Han Chinese with PCa.
Collapse
|
21
|
Abstract
Epithelial–mesenchymal plasticity contributes to many biological processes, including tumor progression. Various epithelial–mesenchymal transition (EMT) responses have been reported and no common, EMT-defining gene expression program has been identified. Here, we have performed a comparative analysis of the EMT response, leveraging highly multiplexed single-cell RNA sequencing (scRNA-seq) to measure expression profiles of 103,999 cells from 960 samples, comprising 12 EMT time course experiments and independent kinase inhibitor screens for each. We demonstrate that the EMT is vastly context specific, with an average of only 22% of response genes being shared between any two conditions, and over half of all response genes were restricted to 1–2 time course experiments. Further, kinase inhibitor screens revealed signaling dependencies and modularity of these responses. These findings suggest that the EMT is not simply a single, linear process, but is highly variable and modular, warranting quantitative frameworks for understanding nuances of the transition. It is unclear if a common EMT expression program exists. Here, the authors perform multiplexed single-cell RNA sequencing across 12 EMT time courses and 16 kinase inhibitor screens, and find that EMT transcriptional responses are context specific and EMT is not a single, linear transition.
Collapse
|
22
|
TGFβ2-induced formation of lipid droplets supports acidosis-driven EMT and the metastatic spreading of cancer cells. Nat Commun 2020; 11:454. [PMID: 31974393 PMCID: PMC6978517 DOI: 10.1038/s41467-019-14262-3] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 12/23/2019] [Indexed: 11/10/2022] Open
Abstract
Acidosis, a common characteristic of the tumor microenvironment, is associated with alterations in metabolic preferences of cancer cells and progression of the disease. Here we identify the TGF-β2 isoform at the interface between these observations. We document that acidic pH promotes autocrine TGF-β2 signaling, which in turn favors the formation of lipid droplets (LD) that represent energy stores readily available to support anoikis resistance and cancer cell invasiveness. We find that, in cancer cells of various origins, acidosis-induced TGF-β2 activation promotes both partial epithelial-to-mesenchymal transition (EMT) and fatty acid metabolism, the latter supporting Smad2 acetylation. We show that upon TGF-β2 stimulation, PKC-zeta-mediated translocation of CD36 facilitates the uptake of fatty acids that are either stored as triglycerides in LD through DGAT1 or oxidized to generate ATP to fulfill immediate cellular needs. We also address how, by preventing fatty acid mobilization from LD, distant metastatic spreading may be inhibited. The tumour microenvironment is known to have an acidic pH but how this influences cancer cell phenotype is unclear. Here, the authors show that tumour cells upregulate TGF-β2 under acidosis, which leads to the increased formation of lipid droplets allowing for invasiveness and metastases.
Collapse
|
23
|
Rubino FM, Dei Cas M, Bignotto M, Ghidoni R, Iriti M, Paroni R. Discovery of Unexpected Sphingolipids in Almonds and Pistachios with an Innovative Use of Triple Quadrupole Tandem Mass Spectrometry. Foods 2020; 9:foods9020110. [PMID: 31972966 PMCID: PMC7074565 DOI: 10.3390/foods9020110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/15/2020] [Accepted: 01/19/2020] [Indexed: 01/29/2023] Open
Abstract
The densely packed storage of valuable nutrients (carbohydrates, lipids, proteins, micronutrients) in the endosperm of nuts and seeds makes the study of their complex composition a topic of great importance. Ceramides in the total lipid extract of some ground almonds and pistachios were searched with a systematic innovative discovery precursor ion scan in a triple quadrupole tandem mass spectrometry, where iso-energetic collision activated dissociation was performed. Five descriptors were used to search components with different C18 long chain bases containing different structural motifs (d18:0, d18:1, d18:2, t18:0, t18:1). The presence of hexoside unit was screened with a specific neutral loss experiment under iso-energetic collision activated dissociation conditions. The discovery scans highlighted the presence of two specific hexosyl-ceramides with a modified sphingosine component (d18:2) and C16:0 or C16:0 hydroxy-fatty acids. The hexosyl-ceramide with the non-hydroxylated fatty acid seemed specific of pistachios and was undetected in almonds. The fast and comprehensive mass spectrometric method used here can be useful to screen lipid extracts of several more seeds of nutraceutical interest, searching for unusual and/or specific sphingosides with chemically decorated long chain bases.
Collapse
Affiliation(s)
- Federico Maria Rubino
- Dipartimento di Scienze della Salute, Universita’ degli Studi di Milano, I-20142 Milano, Italy; (F.M.R.); (M.D.C.); (M.B.); (R.G.)
| | - Michele Dei Cas
- Dipartimento di Scienze della Salute, Universita’ degli Studi di Milano, I-20142 Milano, Italy; (F.M.R.); (M.D.C.); (M.B.); (R.G.)
| | - Monica Bignotto
- Dipartimento di Scienze della Salute, Universita’ degli Studi di Milano, I-20142 Milano, Italy; (F.M.R.); (M.D.C.); (M.B.); (R.G.)
| | - Riccardo Ghidoni
- Dipartimento di Scienze della Salute, Universita’ degli Studi di Milano, I-20142 Milano, Italy; (F.M.R.); (M.D.C.); (M.B.); (R.G.)
- Aldo Ravelli Center for Neurotechnology and Experimental Brain Therapeutics, Dipartimento di Scienze della Salute, Universita’ degli Studi di Milano, I-20142 Milano, Italy
| | - Marcello Iriti
- Dipartimento di Scienze Agrarie e Ambientali—Produzione, Territorio, Agroenergia, Universita’ degli Studi di Milano, I-20133 Milano, Italy;
| | - Rita Paroni
- Dipartimento di Scienze della Salute, Universita’ degli Studi di Milano, I-20142 Milano, Italy; (F.M.R.); (M.D.C.); (M.B.); (R.G.)
- Correspondence:
| |
Collapse
|
24
|
Platania CBM, Dei Cas M, Cianciolo S, Fidilio A, Lazzara F, Paroni R, Pignatello R, Strettoi E, Ghidoni R, Drago F, Bucolo C. Novel ophthalmic formulation of myriocin: implications in retinitis pigmentosa. Drug Deliv 2019; 26:237-243. [PMID: 30883241 PMCID: PMC6419690 DOI: 10.1080/10717544.2019.1574936] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/17/2019] [Accepted: 01/21/2019] [Indexed: 11/09/2022] Open
Abstract
Myriocin is an antibiotic derived from Mycelia sterilia, and is a potent inhibitor of serine palmitoyltransferase, the enzyme involved in the first step of sphingosine synthesis. Myriocin, inhibiting ceramide synthesis, has a great potential for treatment of diseases characterized by high ceramide levels in affected tissues, such as retinitis pigmentosa (RP). Drug delivery to the retina is a challenging task, which is generally by-passed through intravitreal injection, that represents a risky invasive procedure. We, therefore, developed and characterized an ophthalmic topical nanotechnological formulation based on a nanostructured lipid carrier (NLC) and containing myriocin. The ocular distribution of myriocin in the back of the eye was assessed both in rabbits and mice using LC-MS/MS. Moreover, rabbit retinal sphingolipid and ceramides levels, after myriocin-NLC (Myr-NLC) eye drops treatment, were assessed. The results demonstrated that Myr-NLC formulation is well tolerated and provided effective levels of myriocin in the back of the eye both in rabbits and mice. We found that Myr-NLC eye drops treatment was able to significantly decrease retinal sphingolipid levels. In conclusion, these data suggest that the Myr-NLC ophthalmic formulation is suitable for pharmaceutical development and warrants further clinical evaluation of this eye drops for the treatment of RP.
Collapse
Affiliation(s)
- Chiara Bianca Maria Platania
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Michele Dei Cas
- Department of Health Sciences, University of Milano, Milano, Italy
| | | | - Annamaria Fidilio
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Francesca Lazzara
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Rita Paroni
- Department of Health Sciences, University of Milano, Milano, Italy
| | - Rosario Pignatello
- Drug Sciences Department, University of Catania, Catania, Italy
- NANO-i – Research Center on Ocular Nanotechnology University of Catania, Catania, Italy
| | | | - Riccardo Ghidoni
- Department of Health Sciences, University of Milano, Milano, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
- Center for Research in Ocular Pharmacology-CERFO, University of Catania, Catania, Italy
| | - Claudio Bucolo
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
- Center for Research in Ocular Pharmacology-CERFO, University of Catania, Catania, Italy
| |
Collapse
|
25
|
Paroni R, Dei Cas M, Rizzo J, Ghidoni R, Montagna MT, Rubino FM, Iriti M. Bioactive phytochemicals of tree nuts. Determination of the melatonin and sphingolipid content in almonds and pistachios. J Food Compost Anal 2019. [DOI: 10.1016/j.jfca.2019.05.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
26
|
Tracz-Gaszewska Z, Dobrzyn P. Stearoyl-CoA Desaturase 1 as a Therapeutic Target for the Treatment of Cancer. Cancers (Basel) 2019; 11:cancers11070948. [PMID: 31284458 PMCID: PMC6678606 DOI: 10.3390/cancers11070948] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 06/29/2019] [Accepted: 07/03/2019] [Indexed: 12/12/2022] Open
Abstract
A distinctive feature of cancer cells of various origins involves alterations of the composition of lipids, with significant enrichment in monounsaturated fatty acids. These molecules, in addition to being structural components of newly formed cell membranes of intensely proliferating cancer cells, support tumorigenic signaling. An increase in the expression of stearoyl-CoA desaturase 1 (SCD1), the enzyme that converts saturated fatty acids to ∆9-monounsaturated fatty acids, has been observed in a wide range of cancer cells, and this increase is correlated with cancer aggressiveness and poor outcomes for patients. Studies have demonstrated the involvement of SCD1 in the promotion of cancer cell proliferation, migration, metastasis, and tumor growth. Many studies have reported a role for this lipogenic factor in maintaining the characteristics of cancer stem cells (i.e., the population of cells that contributes to cancer progression and resistance to chemotherapy). Importantly, both the products of SCD1 activity and its direct impact on tumorigenic pathways have been demonstrated. Based on these findings, SCD1 appears to be a significant player in the development of malignant disease and may be a promising target for anticancer therapy. Numerous chemical compounds that exert inhibitory effects on SCD1 have been developed and preclinically tested. The present review summarizes our current knowledge of the ways in which SCD1 contributes to the progression of cancer and discusses opportunities and challenges of using SCD1 inhibitors for the treatment of cancer.
Collapse
Affiliation(s)
- Zuzanna Tracz-Gaszewska
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Pawel Dobrzyn
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology Polish Academy of Sciences, 02-093 Warsaw, Poland.
| |
Collapse
|
27
|
Kang H, Kim H, Lee S, Youn H, Youn B. Role of Metabolic Reprogramming in Epithelial⁻Mesenchymal Transition (EMT). Int J Mol Sci 2019; 20:ijms20082042. [PMID: 31027222 PMCID: PMC6514888 DOI: 10.3390/ijms20082042] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/08/2019] [Accepted: 04/23/2019] [Indexed: 02/07/2023] Open
Abstract
Activation of epithelial–mesenchymal transition (EMT) is thought to be an essential step for cancer metastasis. Tumor cells undergo EMT in response to a diverse range of extra- and intracellular stimulants. Recently, it was reported that metabolic shifts control EMT progression and induce tumor aggressiveness. In this review, we summarize the involvement of altered glucose, lipid, and amino acid metabolic enzyme expression and the underlying molecular mechanisms in EMT induction in tumor cells. Moreover, we propose that metabolic regulation through gene-specific or pharmacological inhibition may suppress EMT and this treatment strategy may be applied to prevent tumor progression and improve anti-tumor therapeutic efficacy. This review presents evidence for the importance of metabolic changes in tumor progression and emphasizes the need for further studies to better understand tumor metabolism.
Collapse
Affiliation(s)
- Hyunkoo Kang
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
| | - Hyunwoo Kim
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
| | - Sungmin Lee
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
| | - HyeSook Youn
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul 05006, Korea.
| | - BuHyun Youn
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
- Department of Biological Sciences, Pusan National University, Busan 46241, Korea.
| |
Collapse
|
28
|
Carvalho TM, Cardoso HJ, Figueira MI, Vaz CV, Socorro S. The peculiarities of cancer cell metabolism: A route to metastasization and a target for therapy. Eur J Med Chem 2019; 171:343-363. [PMID: 30928707 DOI: 10.1016/j.ejmech.2019.03.053] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/19/2019] [Accepted: 03/21/2019] [Indexed: 02/06/2023]
Abstract
The last decade has witnessed the peculiarities of metabolic reprogramming in tumour onset and progression, and their relevance in cancer therapy. Also, it has been indicated that the metastatic process may depend on the metabolic rewiring and adaptation of cancer cells to the pressure of tumour microenvironment and limiting nutrient availability. The present review gatherers the existent knowledge on the influence of tumour microenvironment and metabolic routes driving metastasis. A focus will be given to glycolysis, fatty acid metabolism, glutaminolysis, and amino acid handling. In addition, the role of metabolic waste driving metastasization will be explored. Finally, we discuss the status of cancer treatment approaches targeting metabolism. This knowledge revision will highlight the critical metabolic targets in metastasis and the chemicals already used in preclinical studies and clinical trials, providing clues that would be further exploited in medicinal chemistry research.
Collapse
Affiliation(s)
- Tiago Ma Carvalho
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Henrique J Cardoso
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Marília I Figueira
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Cátia V Vaz
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Sílvia Socorro
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.
| |
Collapse
|
29
|
Bhattacharya D, Scimè A. Metabolic Regulation of Epithelial to Mesenchymal Transition: Implications for Endocrine Cancer. Front Endocrinol (Lausanne) 2019; 10:773. [PMID: 31849832 PMCID: PMC6901924 DOI: 10.3389/fendo.2019.00773] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/23/2019] [Indexed: 12/13/2022] Open
Abstract
The last few decades have witnessed an outstanding advancement in our understanding of the hallmarks of endocrine cancers. This includes the epithelial to mesenchymal transition (EMT), a process that alters the morphology and functional characteristics of carcinoma cells. The mesenchymal stem cell like phenotype produced by EMT allows the dislocation of cancer cells from the primary tumor site with inheritance of motility, metastatic and invasive properties. A fundamental driver thought to initiate and propagate EMT is metabolic reprogramming that occur during these transitions. Though there remains a paucity of data regarding the alterations that occur during EMT in endocrine cancers, the contribution of deregulated metabolism is a prominent feature. This mini review focuses on metabolic reprogramming events that occur in cancer cells and in particular those of endocrine origin. It highlights the main metabolic reprogramming outcomes of EMT, encompassing glycolysis, mitochondria oxidative phosphorylation and function, glutamine and lipid metabolism. Comprehending the metabolic changes that occur during EMT will help formulate potential bioenergetic targets as therapies for endocrine cancer metastasis.
Collapse
|
30
|
Navarro-Reig M, Tauler R, Iriondo-Frias G, Jaumot J. Untargeted lipidomic evaluation of hydric and heat stresses on rice growth. J Chromatogr B Analyt Technol Biomed Life Sci 2018; 1104:148-156. [PMID: 30471516 DOI: 10.1016/j.jchromb.2018.11.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 11/10/2018] [Accepted: 11/13/2018] [Indexed: 02/02/2023]
Abstract
Environmental stresses are the major factors that limit the geographical distribution of plants. As a consequence, plants have developed different strategies to adapt to these environmental changes among which can be outlined the maintenance of membranes' integrity and fluidity. Lipids are key molecules for this environmental adaptation and a comprehensive understand of the molecular mechanisms underlying is still required. Here, lipidome changes in Japanese rice (Oryza sativa var. Japonica) upon heat and hydric stresses are assessed using an untargeted approach based on liquid chromatography coupled with mass spectrometry (LC-MS). The obtained data were analyzed using different multivariate data analysis tools. A total number of 298 lipids responded to these abiotic stresses, and 128 of them were tentatively identified. Diacylglycerols (DG), triacylglycerols (TG), phosphatidylcholines (PC) and phosphatidylethanolamines (PE) were the most altered lipid families heat and hydric stress. Interpretation of the obtained results showed relevant changes related to the unsaturation degree in the identified lipids. In the case of heat stress, a decrease in the unsaturation degree of lipids can be linked to an increase in the cell membranes' rigidity. In contrast, the hydric stress produced an increase in the lipids unsaturation degree causing an increase in the cell membranes' fluidity, in an attempt to adapt to these non-optimal conditions.
Collapse
Affiliation(s)
- Meritxell Navarro-Reig
- Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, 08034 Barcelona, Spain.
| | - Romà Tauler
- Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Guillermo Iriondo-Frias
- Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Joaquim Jaumot
- Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, 08034 Barcelona, Spain
| |
Collapse
|
31
|
Puig-Castellví F, Pérez Y, Piña B, Tauler R, Alfonso I. Comparative analysis of 1H NMR and 1H- 13C HSQC NMR metabolomics to understand the effects of medium composition in yeast growth. Anal Chem 2018; 90:12422-12430. [PMID: 30350620 DOI: 10.1021/acs.analchem.8b01196] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In nuclear magnetic resonance (NMR) metabolomics, most of the studies have been focused on the analysis of one-dimensional proton (1D 1H) NMR, whereas the analysis of other nuclei, such as 13C, or other NMR experiments are still underrepresented. The preference of 1D 1H NMR metabolomics lies on the fact that it has good sensitivity and a short acquisition time, but it lacks spectral resolution because it presents a high degree of overlap. In this study, the growth metabolism of yeast ( Saccharomyces cerevisiae) was analyzed by 1D 1H NMR and by two-dimensional (2D) 1H-13C heteronuclear single quantum coherence (HSQC) NMR spectroscopy, leading to the detection of more than 50 metabolites with both analytical approaches. These two analyses allow for a better understanding of the strengths and intrinsic limitations of the two types of NMR approaches. The two data sets (1D and 2D NMR) were investigated with PCA, ASCA, and PLS DA chemometric methods, and similar results were obtained regardless of the data type used. However, data-analysis time for the 2D NMR data set was substantially reduced when compared with the data analysis of the corresponding 1H NMR data set because, for the 2D NMR data, signal overlap was not a major problem and deconvolution was not required. The comparative study described in this work can be useful for the future design of metabolomics workflows, to assist in the selection of the most convenient NMR platform and to guide the posterior data analysis of biomarker selection.
Collapse
Affiliation(s)
- Francesc Puig-Castellví
- Department of Environmental Chemistry , Institute of Environmental Assessment and Water Research (IDAEA-CSIC) , Jordi Girona 18-26 , 08034 Barcelona , Spain
| | - Yolanda Pérez
- NMR Facility , Institute of Advanced Chemistry of Catalonia (IQAC-CSIC) , Jordi Girona 18-26 , 08034 Barcelona , Spain
| | - Benjamín Piña
- Department of Environmental Chemistry , Institute of Environmental Assessment and Water Research (IDAEA-CSIC) , Jordi Girona 18-26 , 08034 Barcelona , Spain
| | - Romà Tauler
- Department of Environmental Chemistry , Institute of Environmental Assessment and Water Research (IDAEA-CSIC) , Jordi Girona 18-26 , 08034 Barcelona , Spain
| | - Ignacio Alfonso
- Department of Biological Chemistry , Institute of Advanced Chemistry of Catalonia (IQAC-CSIC) , Jordi Girona 18-26 , 08034 Barcelona , Spain
| |
Collapse
|
32
|
Eiriksson FF, Rolfsson O, Ogmundsdottir HM, Haraldsson GG, Thorsteinsdottir M, Halldorsson S. Altered plasmalogen content and fatty acid saturation following epithelial to mesenchymal transition in breast epithelial cell lines. Int J Biochem Cell Biol 2018; 103:99-104. [DOI: 10.1016/j.biocel.2018.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/27/2018] [Accepted: 08/06/2018] [Indexed: 12/21/2022]
|
33
|
Brzozowski JS, Jankowski H, Bond DR, McCague SB, Munro BR, Predebon MJ, Scarlett CJ, Skelding KA, Weidenhofer J. Lipidomic profiling of extracellular vesicles derived from prostate and prostate cancer cell lines. Lipids Health Dis 2018; 17:211. [PMID: 30193584 PMCID: PMC6128989 DOI: 10.1186/s12944-018-0854-x] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 08/28/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) are produced and secreted from most cells of the body and can be recovered in biological fluids. Although there has been extensive characterisation of the protein and nucleic acid component of EVs, their lipidome has received little attention and may represent a unique and untapped source of biomarkers for prostate cancer diagnosis and prognosis. METHODS EVs were isolated from non-tumourigenic (RWPE1), tumourigenic (NB26) and metastatic (PC-3) prostate cell lines. Lipids were extracted and subsequently used for targeted lipidomics analysis for the quantitation of molecular lipid species. RESULTS A total of 187 molecular lipid species were quantitatively identified in EV samples showing differential abundance between RWPE1, NB26 and PC-3 EV samples. Fatty acids, glycerolipids and prenol lipids were more highly abundant in EVs from non-tumourigenic cells, whereas sterol lipids, sphingolipids and glycerophospholipids were more highly abundant in EVs from tumourigenic or metastatic cells. CONCLUSIONS This study identified differences in the molecular lipid species of prostate cell-derived EVs, increasing our understanding of the changes that occur to the EV lipidome during prostate cancer progression. These differences highlight the importance of characterising the EV lipidome, which may lead to improved diagnostic and prognostic biomarkers for prostate cancer.
Collapse
Affiliation(s)
- Joshua S. Brzozowski
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Ourimbah, NSW Australia
- Cancer Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW Australia
| | - Helen Jankowski
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Ourimbah, NSW Australia
- Cancer Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW Australia
| | - Danielle R. Bond
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Ourimbah, NSW Australia
- Cancer Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW Australia
| | - Siobhan B. McCague
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Ourimbah, NSW Australia
- Cancer Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW Australia
| | - Benjamin R. Munro
- Cancer Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW Australia
- School of Environmental and Life Sciences, University of Newcastle, Ourimbah, NSW Australia
| | - Melanie J. Predebon
- Cancer Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW Australia
- School of Environmental and Life Sciences, University of Newcastle, Ourimbah, NSW Australia
| | - Christopher J. Scarlett
- Cancer Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW Australia
- School of Environmental and Life Sciences, University of Newcastle, Ourimbah, NSW Australia
| | - Kathryn A. Skelding
- Cancer Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW Australia
| | - Judith Weidenhofer
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Ourimbah, NSW Australia
- Cancer Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW Australia
| |
Collapse
|
34
|
Dalmau N, Bedia C, Tauler R. Validation of the Regions of Interest Multivariate Curve Resolution (ROIMCR) procedure for untargeted LC-MS lipidomic analysis. Anal Chim Acta 2018; 1025:80-91. [DOI: 10.1016/j.aca.2018.04.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 02/21/2018] [Accepted: 04/03/2018] [Indexed: 01/04/2023]
|
35
|
Dalmau N, Andrieu-Abadie N, Tauler R, Bedia C. Phenotypic and lipidomic characterization of primary human epidermal keratinocytes exposed to simulated solar UV radiation. J Dermatol Sci 2018; 92:97-105. [PMID: 30017509 DOI: 10.1016/j.jdermsci.2018.07.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/04/2018] [Accepted: 07/05/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Ultraviolet (UV) radiation is known to be one of the most important environmental hazards acting on the skin. The most part of UV radiation is absorbed in the epidermis, where keratinocytes are the most abundant and exposed cell type. Lipids have an important role in skin biology, not only for their important contribution to the maintenance of the permeability barrier but also for the production and storage of energy, membrane organization and cell signalling functions. However, the effects on the lipid composition of keratinocytes under UV radiation are little explored. OBJECTIVE The present work aims to explore the effects on the phenotype and lipid content of primary human keratinocytes exposed to simulated solar UV radiation. METHODS Keratinocytes were exposed to a single (acute exposure) and repeated simulated solar UV irradiations for 4 weeks (chronic exposure). Cell viability and morphology were explored, as well as the production of reactive oxygen species. Then, lipid extracts were analysed through liquid chromatography coupled to mass spectrometry (LC-MS) and the data generated was processed using the ROIMCR chemometric methodology together with partial least squares discriminant analysis (PLS-DA), to finally reveal the most relevant lipid changes that occurred in keratinocytes upon UV irradiation. Also, the potential induction of keratinocyte differentiation was explored by measuring the increase of involucrin. RESULTS Under acute irradiation, cell viability and morphology were not altered. However, a general increase of phosphatidylcholines (PC) phosphatidylethanolamines (PE) and phosphatidylglycerol (PG) together with a slight sphingomyelin (SM) decrease were found in UV irradiated cells, among other changes. In addition, keratinocyte cultures did not present any differentiation hallmark. Contrary to acute-irradiated cells, in chronic exposures, cell viability was reduced and keratinocytes presented an altered morphology. Also, hallmarks of differentiation, such as the increase of involucrin protein and the autophagy induction were detected. Among the main lipid changes that accompanied this phenotype, the increase of long-chain ceramides, lysoPC and glycerolipid species were found. CONCLUSION Important lipid changes were detected under acute and chronic UV irradiation. The lipid profile under chronic exposure may represent a lipid fingerprint of the keratinocyte differentiation phenotype.
Collapse
Affiliation(s)
- Núria Dalmau
- Department of Environmental Chemistry, Institute of Environmental Assessment and Water Research (IDAEA-CSIC), c/ Jordi Girona 18-24, 08034 Barcelona, Spain
| | - Nathalie Andrieu-Abadie
- INSERM UMR 1037, Centre de Recherches en Cancérologie de Toulouse (CRCT), 31037, Toulouse, France
| | - Romà Tauler
- Department of Environmental Chemistry, Institute of Environmental Assessment and Water Research (IDAEA-CSIC), c/ Jordi Girona 18-24, 08034 Barcelona, Spain
| | - Carmen Bedia
- Department of Environmental Chemistry, Institute of Environmental Assessment and Water Research (IDAEA-CSIC), c/ Jordi Girona 18-24, 08034 Barcelona, Spain.
| |
Collapse
|
36
|
Puig-Castellví F, Bedia C, Alfonso I, Piña B, Tauler R. Deciphering the Underlying Metabolomic and Lipidomic Patterns Linked to Thermal Acclimation in Saccharomyces cerevisiae. J Proteome Res 2018; 17:2034-2044. [PMID: 29707950 DOI: 10.1021/acs.jproteome.7b00921] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Temperature is one of the most critical parameters for yeast growth, and it has deep consequences in many industrial processes where yeast is involved. Nevertheless, the metabolic changes required to accommodate yeast cells at high or low temperatures are still poorly understood. In this work, the ultimate responses of these induced transcriptomic effects have been examined using metabolomics-derived strategies. The yeast metabolome and lipidome have been characterized by 1D proton nuclear magnetic resonance spectroscopy and ultra-high-performance liquid chromatography-mass spectrometry at four temperatures, corresponding to low, optimal, high, and extreme thermal conditions. The underlying pathways that drive the acclimation response of yeast to these nonoptimal temperatures were evaluated using multivariate curve resolution-alternating least-squares. The analysis revealed three different thermal profiles (cold, optimal, and high temperature), which include changes in the lipid composition, secondary metabolic pathways, and energy metabolism, and we propose that they reflect the acclimation strategy of yeast cells to low and high temperatures. The data suggest that yeast adjusts membrane fluidity by changing the relative proportions of the different lipid families (acylglycerides, phospholipids, and ceramides, among others) rather than modifying the average length and unsaturation levels of the corresponding fatty acids.
Collapse
Affiliation(s)
- Francesc Puig-Castellví
- Department of Environmental Chemistry , IDAEA-CSIC , Jordi Girona 18-26 , Barcelona 08034 , Catalonia , Spain
| | - Carmen Bedia
- Department of Environmental Chemistry , IDAEA-CSIC , Jordi Girona 18-26 , Barcelona 08034 , Catalonia , Spain
| | - Ignacio Alfonso
- Department of Biological Chemistry and Molecular Modelling , IQAC-CSIC , Jordi Girona 18-26 , Barcelona 08034 , Catalonia , Spain
| | - Benjamin Piña
- Department of Environmental Chemistry , IDAEA-CSIC , Jordi Girona 18-26 , Barcelona 08034 , Catalonia , Spain
| | - Romà Tauler
- Department of Environmental Chemistry , IDAEA-CSIC , Jordi Girona 18-26 , Barcelona 08034 , Catalonia , Spain
| |
Collapse
|
37
|
Galbraith L, Leung HY, Ahmad I. Lipid pathway deregulation in advanced prostate cancer. Pharmacol Res 2018; 131:177-184. [PMID: 29466694 DOI: 10.1016/j.phrs.2018.02.022] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 02/08/2018] [Accepted: 02/14/2018] [Indexed: 01/03/2023]
Abstract
The link between prostate cancer (PC) development and lipid metabolism is well established, with AR intimately involved in a number of lipogenic processes involving SREBP1, PPARG, FASN, ACC, ACLY and SCD1. Recently, there is growing evidence implicating the role of obesity and peri-prostatic adipose tissue (PPAT) in PC aggressiveness and related mortality, suggesting the importance of lipid pathways in both localised and disseminated disease. A number of promising agents are in development to target the lipogenic axis in PC, and the likelihood is that these agents will form part of combination drug strategies, with targeting of multiple metabolic pathways (e.g. FASN and CPT1), or in combination with AR pathway inhibitors (SCD1 and AR).
Collapse
Affiliation(s)
- Laura Galbraith
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Hing Y Leung
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Imran Ahmad
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK.
| |
Collapse
|
38
|
Dalmau N, Andrieu-Abadie N, Tauler R, Bedia C. Untargeted lipidomic analysis of primary human epidermal melanocytes acutely and chronically exposed to UV radiation. Mol Omics 2018; 14:170-180. [DOI: 10.1039/c8mo00060c] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Ultraviolet (UV) radiation present in sunlight has been related to harmful effects on skin such as premature aging and skin cancer.
Collapse
Affiliation(s)
- Núria Dalmau
- Department of Environmental Chemistry
- Institute of Environmental Assessment and Water Research (IDAEA-CSIC)
- 08034 Barcelona
- Spain
| | | | - Romà Tauler
- Department of Environmental Chemistry
- Institute of Environmental Assessment and Water Research (IDAEA-CSIC)
- 08034 Barcelona
- Spain
| | - Carmen Bedia
- Department of Environmental Chemistry
- Institute of Environmental Assessment and Water Research (IDAEA-CSIC)
- 08034 Barcelona
- Spain
| |
Collapse
|
39
|
Zhang L, Min W. Bioorthogonal chemical imaging of metabolic changes during epithelial-mesenchymal transition of cancer cells by stimulated Raman scattering microscopy. JOURNAL OF BIOMEDICAL OPTICS 2017; 22:1-7. [PMID: 29043713 PMCID: PMC5644606 DOI: 10.1117/1.jbo.22.10.106010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/27/2017] [Indexed: 05/30/2023]
Abstract
Study of metabolic changes during epithelial-mesenchymal transition (EMT) of cancer cells is important for basic understanding and therapeutic management of cancer progression. We here used metabolic labeling and stimulated Raman scattering (SRS) microscopy, a strategy of bioorthogonal chemical imaging, to directly visualize changes in anabolic metabolism during cancer EMT at a single-cell level. MCF-7 breast cancer cell is employed as a model system. Four types of metabolites (amino acids, glucose, fatty acids, and choline) are labeled with either deuterium or alkyne (C≡C) tag. Their intracellular incorporations into MCF-7 cells before or after EMT are visualized by SRS imaging targeted at the signature vibration frequency of C-D or C≡C bonds. Overall, after EMT, anabolism of amino acids, glucose, and choline is less active, reflecting slower protein and membrane synthesis in mesenchymal cells. Interestingly, we also observed less incorporation of glucose and palmitate acids into membrane lipids, but more of them into lipid droplets in mesenchymal cells. This result indicates that, although mesenchymal cells synthesize fewer membrane lipids, they are actively storing energy into lipid droplets, either through de novo lipogenesis from glucose or direct scavenging of exogenous free fatty acids. Hence, metabolic labeling coupled with SRS can be a straightforward method in imaging cancer metabolism.
Collapse
Affiliation(s)
- Luyuan Zhang
- Columbia University, Department of Chemistry, New York, New York, United States
| | - Wei Min
- Columbia University, Department of Chemistry, New York, New York, United States
- Columbia University, Kavli Institute for Brain Science, New York, New York, United States
| |
Collapse
|
40
|
Sciacovelli M, Frezza C. Metabolic reprogramming and epithelial-to-mesenchymal transition in cancer. FEBS J 2017; 284:3132-3144. [PMID: 28444969 DOI: 10.1111/febs.14090] [Citation(s) in RCA: 221] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/23/2017] [Accepted: 04/24/2017] [Indexed: 12/16/2022]
Abstract
Several lines of evidence indicate that during transformation epithelial cancer cells can acquire mesenchymal features via a process called epithelial-to-mesenchymal transition (EMT). This process endows cancer cells with increased invasive and migratory capacity, enabling tumour dissemination and metastasis. EMT is associated with a complex metabolic reprogramming, orchestrated by EMT transcription factors, which support the energy requirements of increased motility and growth in harsh environmental conditions. The discovery that mutations in metabolic genes such as FH, SDH and IDH activate EMT provided further evidence that EMT and metabolism are intertwined. In this review, we discuss the role of EMT in cancer and the underpinning metabolic reprogramming. We also put forward the hypothesis that, by altering chromatin structure and function, metabolic pathways engaged by EMT are necessary for its full activation.
Collapse
Affiliation(s)
- Marco Sciacovelli
- Medical Research Council Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, UK
| | - Christian Frezza
- Medical Research Council Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, UK
| |
Collapse
|
41
|
Zhang C, Li J, Lan L, Cheng JX. Quantification of Lipid Metabolism in Living Cells through the Dynamics of Lipid Droplets Measured by Stimulated Raman Scattering Imaging. Anal Chem 2017; 89:4502-4507. [PMID: 28345862 DOI: 10.1021/acs.analchem.6b04699] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Dysregulation of lipid metabolism is associated with many diseases including cancer. Lipid droplet (LD), a ubiquitous organelle in mammalian cells, serves as a hub for lipid metabolism. Conventional assays on the measurement of lipid metabolism rely on the quantification of the lipid composition or amount. Such methods cannot distinguish LDs having different biofunctionalities in living cells, and thus could be inaccurate in measuring the instantaneous lipogenesis of the living cells. We applied label-free stimulated Raman scattering microscopy to quantify the LDs' spatial-temporal dynamics, which showed direct links to cellular lipid metabolisms and can separate LDs involved in different metabolic events. In human cancer cells, we found that changes in the maximum displacement of LDs reflected variations in cellular lipogenic activity, and changes in the average speed of LDs revealed alterations in LD size. The LD dynamics analysis allowed for more accurate measurement in the lipogenesis and LD dimensions, and can break the optical diffraction limit to detect small variation in lipid metabolism that was conventionally undetectable. By this method, we revealed changes in the lipogenic activity and LD sizes during glucose starvation of HeLa cells and transforming growth factor beta-induced epithelial-to-mesenchymal transition of SKOV-3 cells. This method opens a way to quantify lipid metabolism in living cells during cellular development and transition.
Collapse
Affiliation(s)
- Chi Zhang
- Weldon School of Biomedical Engineering, Purdue University . 206 S. Martin Jischke Drive, West Lafayette, Indiana 47906, United States
| | - Junjie Li
- Weldon School of Biomedical Engineering, Purdue University . 206 S. Martin Jischke Drive, West Lafayette, Indiana 47906, United States
| | - Lu Lan
- Weldon School of Biomedical Engineering, Purdue University . 206 S. Martin Jischke Drive, West Lafayette, Indiana 47906, United States
| | - Ji-Xin Cheng
- Weldon School of Biomedical Engineering, Purdue University . 206 S. Martin Jischke Drive, West Lafayette, Indiana 47906, United States
| |
Collapse
|
42
|
Morandi A, Taddei ML, Chiarugi P, Giannoni E. Targeting the Metabolic Reprogramming That Controls Epithelial-to-Mesenchymal Transition in Aggressive Tumors. Front Oncol 2017; 7:40. [PMID: 28352611 PMCID: PMC5348536 DOI: 10.3389/fonc.2017.00040] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 02/28/2017] [Indexed: 01/06/2023] Open
Abstract
The epithelial-to-mesenchymal transition (EMT) process allows the trans-differentiation of a cell with epithelial features into a cell with mesenchymal characteristics. This process has been reported to be a key priming event for tumor development and therefore EMT activation is now considered an established trait of malignancy. The transcriptional and epigenetic reprogramming that governs EMT has been extensively characterized and reviewed in the last decade. However, increasing evidence demonstrates a correlation between metabolic reprogramming and EMT execution. The aim of the current review is to gather the recent findings that illustrate this correlation to help deciphering whether metabolic changes are causative or just a bystander effect of EMT activation. The review is divided accordingly to the catabolic and anabolic pathways that characterize carbohydrate, aminoacid, and lipid metabolism. Moreover, at the end of each part, we have discussed a series of potential metabolic targets involved in EMT promotion and execution for which drugs are either available or that could be further investigated for therapeutic intervention.
Collapse
Affiliation(s)
- Andrea Morandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence , Florence , Italy
| | - Maria Letizia Taddei
- Department of Experimental and Clinical Medicine, University of Florence , Florence , Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy; Excellence Centre for Research, Transfer and High Education DenoTHE, University of Florence, Florence, Italy
| | - Elisa Giannoni
- Department of Experimental and Clinical Biomedical Sciences, University of Florence , Florence , Italy
| |
Collapse
|