1
|
Hao J, Huang Z, Zhang S, Song K, Wang J, Gao C, Fang Z, Zhang N. Deciphering the multifaceted roles and clinical implications of 2-hydroxyglutarate in cancer. Pharmacol Res 2024; 209:107437. [PMID: 39349213 DOI: 10.1016/j.phrs.2024.107437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/13/2024] [Accepted: 09/24/2024] [Indexed: 10/02/2024]
Abstract
Increasing evidence indicates that 2-hydroxyglutarate (2HG) is an oncometabolite that drives tumour formation and progression. Due to mutations in isocitrate dehydrogenase (IDH) and the dysregulation of other enzymes, 2HG accumulates significantly in tumour cells. Due to its structural similarity to α-ketoglutarate (αKG), accumulated 2HG leads to the competitive inhibition of αKG-dependent dioxygenases (αKGDs), such as KDMs, TETs, and EGLNs. This inhibition results in epigenetic alterations in both tumour cells and the tumour microenvironment. This review comprehensively discusses the metabolic pathways of 2HG and the subsequent pathways influenced by elevated 2HG levels. We will delve into the molecular mechanisms by which 2HG exerts its oncogenic effects, particularly focusing on epigenetic modifications. This review will also explore the various methods available for the detection of 2HG, emphasising both current techniques and emerging technologies. Furthermore, 2HG shows promise as a biomarker for clinical diagnosis and treatment. By integrating these perspectives, this review aims to provide a comprehensive overview of the current understanding of 2HG in cancer biology, highlight the importance of ongoing research, and discuss future directions for translating these findings into clinical applications.
Collapse
Affiliation(s)
- Jie Hao
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Ziyi Huang
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Siyue Zhang
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Kefan Song
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Juncheng Wang
- Advanced Medical Research Institute, Shandong University, Jinan, China
| | - Chao Gao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Zhiqing Fang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Ning Zhang
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
2
|
Menendez JA, Cuyàs E, Encinar JA, Vander Steen T, Verdura S, Llop‐Hernández À, López J, Serrano‐Hervás E, Osuna S, Martin‐Castillo B, Lupu R. Fatty acid synthase (FASN) signalome: A molecular guide for precision oncology. Mol Oncol 2024; 18:479-516. [PMID: 38158755 PMCID: PMC10920094 DOI: 10.1002/1878-0261.13582] [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: 10/02/2023] [Revised: 10/27/2023] [Accepted: 12/28/2023] [Indexed: 01/03/2024] Open
Abstract
The initial excitement generated more than two decades ago by the discovery of drugs targeting fatty acid synthase (FASN)-catalyzed de novo lipogenesis for cancer therapy was short-lived. However, the advent of the first clinical-grade FASN inhibitor (TVB-2640; denifanstat), which is currently being studied in various phase II trials, and the exciting advances in understanding the FASN signalome are fueling a renewed interest in FASN-targeted strategies for the treatment and prevention of cancer. Here, we provide a detailed overview of how FASN can drive phenotypic plasticity and cell fate decisions, mitochondrial regulation of cell death, immune escape and organ-specific metastatic potential. We then present a variety of FASN-targeted therapeutic approaches that address the major challenges facing FASN therapy. These include limitations of current FASN inhibitors and the lack of precision tools to maximize the therapeutic potential of FASN inhibitors in the clinic. Rethinking the role of FASN as a signal transducer in cancer pathogenesis may provide molecularly driven strategies to optimize FASN as a long-awaited target for cancer therapeutics.
Collapse
Affiliation(s)
- Javier A. Menendez
- Metabolism & Cancer Group, Program Against Cancer Therapeutic Resistance (ProCURE)Catalan Institute of OncologyGironaSpain
- Girona Biomedical Research InstituteGironaSpain
| | - Elisabet Cuyàs
- Metabolism & Cancer Group, Program Against Cancer Therapeutic Resistance (ProCURE)Catalan Institute of OncologyGironaSpain
- Girona Biomedical Research InstituteGironaSpain
| | - Jose Antonio Encinar
- Institute of Research, Development and Innovation in Biotechnology of Elche (IDiBE) and Molecular and Cell Biology Institute (IBMC)Miguel Hernández University (UMH)ElcheSpain
| | - Travis Vander Steen
- Division of Experimental Pathology, Department of Laboratory Medicine and PathologyMayo ClinicRochesterMNUSA
- Mayo Clinic Cancer CenterRochesterMNUSA
- Department of Biochemistry and Molecular Biology LaboratoryMayo Clinic LaboratoryRochesterMNUSA
| | - Sara Verdura
- Metabolism & Cancer Group, Program Against Cancer Therapeutic Resistance (ProCURE)Catalan Institute of OncologyGironaSpain
- Girona Biomedical Research InstituteGironaSpain
| | - Àngela Llop‐Hernández
- Metabolism & Cancer Group, Program Against Cancer Therapeutic Resistance (ProCURE)Catalan Institute of OncologyGironaSpain
- Girona Biomedical Research InstituteGironaSpain
| | - Júlia López
- Metabolism & Cancer Group, Program Against Cancer Therapeutic Resistance (ProCURE)Catalan Institute of OncologyGironaSpain
- Girona Biomedical Research InstituteGironaSpain
| | - Eila Serrano‐Hervás
- Metabolism & Cancer Group, Program Against Cancer Therapeutic Resistance (ProCURE)Catalan Institute of OncologyGironaSpain
- Girona Biomedical Research InstituteGironaSpain
- CompBioLab Group, Institut de Química Computacional i Catàlisi (IQCC) and Departament de QuímicaUniversitat de GironaGironaSpain
| | - Sílvia Osuna
- CompBioLab Group, Institut de Química Computacional i Catàlisi (IQCC) and Departament de QuímicaUniversitat de GironaGironaSpain
- ICREABarcelonaSpain
| | - Begoña Martin‐Castillo
- Metabolism & Cancer Group, Program Against Cancer Therapeutic Resistance (ProCURE)Catalan Institute of OncologyGironaSpain
- Girona Biomedical Research InstituteGironaSpain
- Unit of Clinical ResearchCatalan Institute of OncologyGironaSpain
| | - Ruth Lupu
- Division of Experimental Pathology, Department of Laboratory Medicine and PathologyMayo ClinicRochesterMNUSA
- Mayo Clinic Cancer CenterRochesterMNUSA
- Department of Biochemistry and Molecular Biology LaboratoryMayo Clinic LaboratoryRochesterMNUSA
| |
Collapse
|
3
|
IDH mutation and cancer stem cell. Essays Biochem 2022; 66:413-422. [PMID: 35611837 DOI: 10.1042/ebc20220008] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/04/2022] [Accepted: 05/12/2022] [Indexed: 12/11/2022]
Abstract
Cancer stem cells (CSCs) are a small population of cells in human malignancies that resemble the biology of human pluripotent stem cells. CSCs are closely related to the critical hallmarks in human cancers, ranging from oncogenesis to disease progression, therapeutic resistance, and overall outcome. Mutations in isocitrate dehydrogenase (IDH) were recently identified as founder mutations for human cancers. An increasing amount of evidence indicates that IDH mutations are closely related to the establishment and maintenance of CSCs. Biosynthesis of oncometabolite, metabolic reprogramming, and epigenetic shifts establish distinctive molecular signatures in IDH-mutated CSCs. Additionally, IDH mutation and IDH-related pathways could be valuable molecular targets to impact the CSC components in human cancers and to improve the disease outcome.
Collapse
|
4
|
Metabostemness in cancer: Linking metaboloepigenetics and mitophagy in remodeling cancer stem cells. Stem Cell Rev Rep 2021; 18:198-213. [PMID: 34355273 DOI: 10.1007/s12015-021-10216-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 01/01/2023]
Abstract
Cancer stem cells (CSCs) are rare populations of malignant cells with stem cell-like features of self-renewal, uninterrupted differentiation, tumorigenicity, and resistance to conventional therapeutic agents, and these cells have a decisive role in treatment failure and tumor relapse. The self-renewal potential of CSCs with atypical activation of developmental signaling pathways involves the maintenance of stemness to support cancer progression. The acquisition of stemness in CSCs has been accomplished through genetic and epigenetic rewiring following the metabolic switch. In this context, "metabostemness" denotes the metabolic parameters that essentially govern the epitranscriptional gene reprogramming mechanism to dedifferentiate tumor cells into CSCs. Several metabolites often referred to as oncometabolites can directly remodel chromatin structure and thereby influence the operation of epitranscriptional circuits. This integrated metaboloepigenetic dimension of CSCs favors the differentiated cells to move in dedifferentiated macrostates. Some metabolic events might perform as early drivers of epitranscriptional reprogramming; however, subsequent metabolic hits may govern the retention of stemness properties in the tumor mass. Interestingly, selective removal of mitochondria through autophagy can promote metabolic plasticity and alter metabolic states during differentiation and dedifferentiation. In this connection, novel metabostemness-specific drugs can be generated as potential cancer therapeutics to target the metaboloepigenetic circuitry to eliminate CSCs.
Collapse
|
5
|
Gupta VK, Sharma NS, Durden B, Garrido VT, Kesh K, Edwards D, Wang D, Myer C, Mateo-Victoriano B, Kollala SS, Ban Y, Gao Z, Bhattacharya SK, Saluja A, Singh PK, Banerjee S. Hypoxia-Driven Oncometabolite L-2HG Maintains Stemness-Differentiation Balance and Facilitates Immune Evasion in Pancreatic Cancer. Cancer Res 2021; 81:4001-4013. [PMID: 33990397 PMCID: PMC8338764 DOI: 10.1158/0008-5472.can-20-2562] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/09/2020] [Accepted: 05/12/2021] [Indexed: 12/16/2022]
Abstract
In pancreatic cancer, the robust fibroinflammatory stroma contributes to immune suppression and renders tumors hypoxic, altering intratumoral metabolic pathways and leading to poor survival. One metabolic enzyme activated during hypoxia is lactate dehydrogenase A (LDHA). As a result of its promiscuous activity under hypoxia, LDHA produces L-2 hydroxyglutarate (L-2HG), an epigenetic modifier, that regulates the tumor transcriptome. However, the role of L-2HG in remodeling the pancreatic tumor microenvironment is not known. Here we used mass spectrometry to detect L-2HG in serum samples from patients with pancreatic cancer, comprising tumor cells as well as stromal cells. Both hypoxic pancreatic tumors as well as serum from patients with pancreatic cancer accumulated L-2HG as a result of promiscuous activity of LDHA. This abnormally accumulated L-2HG led to H3 hypermethylation and altered gene expression, which regulated a critical balance between stemness and differentiation in pancreatic tumors. Secreted L-2HG inhibited T-cell proliferation and migration, suppressing antitumor immunity. In a syngeneic orthotopic model of pancreatic cancer, inhibition of LDH with GSK2837808A decreased L-2HG, induced tumor regression, and sensitized tumors to anti-PD1 therapy. In conclusion, hypoxia-mediated promiscuous activity of LDH produces L-2HG in pancreatic tumor cells, regulating the stemness-differentiation balance and contributing to immune evasion. Targeting LDH can be developed as a potential therapy to sensitize pancreatic tumors to checkpoint inhibitor therapy. SIGNIFICANCE: This study shows that promiscuous LDH activity produces L-2HG in pancreatic tumor and stromal cells, modulating tumor stemness and immune cell function and infiltration in the tumor microenvironment.
Collapse
Affiliation(s)
- Vineet K Gupta
- Department of Surgery, University of Miami, Miami, Florida
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, Florida
| | - Nikita S Sharma
- Department of Surgery, University of Miami, Miami, Florida
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, Florida
| | - Brittany Durden
- Department of Surgery, University of Miami, Miami, Florida
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, Florida
| | - Vanessa T Garrido
- Department of Surgery, University of Miami, Miami, Florida
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, Florida
| | - Kousik Kesh
- Department of Surgery, University of Miami, Miami, Florida
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, Florida
| | - Dujon Edwards
- Department of Surgery, University of Miami, Miami, Florida
| | - Dezhen Wang
- University of Nebraska Medical School, Omaha, Nebraska
| | - Ciara Myer
- Department of Ophthalmology, University of Miami, Miami, Florida
- Miami Integrative Metabolomic Research Center, University of Miami, Miami, Florida
| | | | | | - Yuguang Ban
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, Florida
| | - Zhen Gao
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, Florida
| | - Sanjoy K Bhattacharya
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, Florida
- Department of Ophthalmology, University of Miami, Miami, Florida
- Miami Integrative Metabolomic Research Center, University of Miami, Miami, Florida
| | - Ashok Saluja
- Department of Surgery, University of Miami, Miami, Florida
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, Florida
| | | | - Sulagna Banerjee
- Department of Surgery, University of Miami, Miami, Florida.
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, Florida
- Miami Integrative Metabolomic Research Center, University of Miami, Miami, Florida
| |
Collapse
|
6
|
Alarcón T, Sardanyés J, Guillamon A, Menendez JA. Bivalent chromatin as a therapeutic target in cancer: An in silico predictive approach for combining epigenetic drugs. PLoS Comput Biol 2021; 17:e1008408. [PMID: 34153035 PMCID: PMC8248646 DOI: 10.1371/journal.pcbi.1008408] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 07/01/2021] [Accepted: 04/26/2021] [Indexed: 11/28/2022] Open
Abstract
Tumour cell heterogeneity is a major barrier for efficient design of targeted anti-cancer therapies. A diverse distribution of phenotypically distinct tumour-cell subpopulations prior to drug treatment predisposes to non-uniform responses, leading to the elimination of sensitive cancer cells whilst leaving resistant subpopulations unharmed. Few strategies have been proposed for quantifying the variability associated to individual cancer-cell heterogeneity and minimizing its undesirable impact on clinical outcomes. Here, we report a computational approach that allows the rational design of combinatorial therapies involving epigenetic drugs against chromatin modifiers. We have formulated a stochastic model of a bivalent transcription factor that allows us to characterise three different qualitative behaviours, namely: bistable, high- and low-gene expression. Comparison between analytical results and experimental data determined that the so-called bistable and high-gene expression behaviours can be identified with undifferentiated and differentiated cell types, respectively. Since undifferentiated cells with an aberrant self-renewing potential might exhibit a cancer/metastasis-initiating phenotype, we analysed the efficiency of combining epigenetic drugs against the background of heterogeneity within the bistable sub-ensemble. Whereas single-targeted approaches mostly failed to circumvent the therapeutic problems represented by tumour heterogeneity, combinatorial strategies fared much better. Specifically, the more successful combinations were predicted to involve modulators of the histone H3K4 and H3K27 demethylases KDM5 and KDM6A/UTX. Those strategies involving the H3K4 and H3K27 methyltransferases MLL2 and EZH2, however, were predicted to be less effective. Our theoretical framework provides a coherent basis for the development of an in silico platform capable of identifying the epigenetic drugs combinations best-suited to therapeutically manage non-uniform responses of heterogenous cancer cell populations.
Collapse
Affiliation(s)
- Tomás Alarcón
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Centre de Recerca Matemàtica, Cerdanyola del Vallès, Spain
- Departament de Matemàtiques, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | | | - Antoni Guillamon
- Centre de Recerca Matemàtica, Cerdanyola del Vallès, Spain
- Departament de Matemàtiques, EPSEB, Universitat Politècnica de Catalunya, Barcelona, Spain
- Institut de Matemàtiques de la UPC-BarcelonaTech (IMTech), Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Javier A. Menendez
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain
- Girona Biomedical Research Institute, Salt, Girona, Spain
| |
Collapse
|
7
|
Cuyàs E, Gumuzio J, Lozano-Sánchez J, Segura-Carretero A, Verdura S, Bosch-Barrera J, Martin-Castillo B, Nonell-Canals A, Llebaria A, Cabello S, Serra C, Sanchez-Martinez M, Martin ÁG, Menendez JA. Mimetics of extra virgin olive oil phenols with anti-cancer stem cell activity. Aging (Albany NY) 2020; 12:21057-21075. [PMID: 33168787 PMCID: PMC7695371 DOI: 10.18632/aging.202154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 09/24/2020] [Indexed: 12/30/2022]
Abstract
The extra virgin olive oil (EVOO) dihydroxy-phenol oleacein is a natural inhibitor of multiple metabolic and epigenetic enzymes capable of suppressing the functional traits of cancer stem cells (CSC). Here, we used a natural product-inspired drug discovery approach to identify new compounds that phenotypically mimic the anti-CSC activity of oleacein. We coupled 3D quantitative structure-activity relationship-based virtual profiling with phenotypic analysis using 3D tumorsphere formation as a gold standard for assessing the presence of CSC. Among the top 20 computationally-predicted oleacein mimetics, four fulfilled the phenotypic endpoint of specifically suppressing the tumorsphere-initiating capacity of CSC, in the absence of significant cytotoxicity against differentiated cancer cells growing in 2D cultures in the same low micromolar concentration range. Of these, 3,4-dihydrophenetyl butyrate -a lipophilic ester conjugate of the hydroxytyrosol moiety of oleacein- and (E)-N-allyl-2-((5-nitrofuran-2-yl)methylene)hydrazinecarbothioamide) -an inhibitor of Trypanosoma cruzi triosephosphate isomerase- were also highly effective at significantly reducing the proportion of aldehyde dehydrogenase (ALDH)-positive CSC-like proliferating cells. Preservation of the mTOR/DNMT binding mode of oleacein was dispensable for suppression of the ALDH+-CSC functional phenotype in hydroxytyrosol-unrelated mimetics. The anti-CSC chemistry of complex EVOO phenols such as oleacein can be phenocopied through the use of mimetics capturing its physico-chemical properties.
Collapse
Affiliation(s)
- Elisabet Cuyàs
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | | | - Jesús Lozano-Sánchez
- Research and Development of Functional Food Centre (CIDAF), Granada, Spain
- Department of Food Science and Nutrition, University of Granada, Granada, Spain
| | - Antonio Segura-Carretero
- Research and Development of Functional Food Centre (CIDAF), Granada, Spain
- Department of Analytical Chemistry, University of Granada, Granada, Spain
| | - Sara Verdura
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Joaquim Bosch-Barrera
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain
- Medical Oncology, Catalan Institute of Oncology, Girona, Spain
- Department of Medical Sciences, Medical School University of Girona, Girona, Spain
| | - Begoña Martin-Castillo
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain
- Unit of Clinical Research, Catalan Institute of Oncology, Girona, Spain
| | - Alfons Nonell-Canals
- Mind the Byte, Barcelona, Spain
- Current address: The Patients Resource, Barcelona, Spain
| | - Amadeu Llebaria
- MCS, Laboratory of Medicinal Chemistry and Synthesis, Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Silvia Cabello
- SIMChem, Synthesis of High Added Value Molecules, Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Carme Serra
- MCS, Laboratory of Medicinal Chemistry and Synthesis, Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
- SIMChem, Synthesis of High Added Value Molecules, Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | | | | | - Javier A. Menendez
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| |
Collapse
|
8
|
Xiang K, Jendrossek V, Matschke J. Oncometabolites and the response to radiotherapy. Radiat Oncol 2020; 15:197. [PMID: 32799884 PMCID: PMC7429799 DOI: 10.1186/s13014-020-01638-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/06/2020] [Indexed: 12/18/2022] Open
Abstract
Radiotherapy (RT) is applied in 45-60% of all cancer patients either alone or in multimodal therapy concepts comprising surgery, RT and chemotherapy. However, despite technical innovations approximately only 50% are cured, highlight a high medical need for innovation in RT practice. RT is a multidisciplinary treatment involving medicine and physics, but has always been successful in integrating emerging novel concepts from cancer and radiation biology for improving therapy outcome. Currently, substantial improvements are expected from integration of precision medicine approaches into RT concepts.Altered metabolism is an important feature of cancer cells and a driving force for malignant progression. Proper metabolic processes are essential to maintain and drive all energy-demanding cellular processes, e.g. repair of DNA double-strand breaks (DSBs). Consequently, metabolic bottlenecks might allow therapeutic intervention in cancer patients.Increasing evidence now indicates that oncogenic activation of metabolic enzymes, oncogenic activities of mutated metabolic enzymes, or adverse conditions in the tumor microenvironment can result in abnormal production of metabolites promoting cancer progression, e.g. 2-hyroxyglutarate (2-HG), succinate and fumarate, respectively. Interestingly, these so-called "oncometabolites" not only modulate cell signaling but also impact the response of cancer cells to chemotherapy and RT, presumably by epigenetic modulation of DNA repair.Here we aimed to introduce the biological basis of oncometabolite production and of their actions on epigenetic regulation of DNA repair. Furthermore, the review will highlight innovative therapeutic opportunities arising from the interaction of oncometabolites with DNA repair regulation for specifically enhancing the therapeutic effects of genotoxic treatments including RT in cancer patients.
Collapse
Affiliation(s)
- Kexu Xiang
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Virchowstrasse 173, 45147, Essen, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Virchowstrasse 173, 45147, Essen, Germany
| | - Johann Matschke
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Virchowstrasse 173, 45147, Essen, Germany.
| |
Collapse
|
9
|
Tumor Cell-Intrinsic Immunometabolism and Precision Nutrition in Cancer Immunotherapy. Cancers (Basel) 2020; 12:cancers12071757. [PMID: 32630618 PMCID: PMC7409312 DOI: 10.3390/cancers12071757] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 06/26/2020] [Accepted: 06/30/2020] [Indexed: 12/19/2022] Open
Abstract
One of the greatest challenges in the cancer immunotherapy field is the need to biologically rationalize and broaden the clinical utility of immune checkpoint inhibitors (ICIs). The balance between metabolism and immune response has critical implications for overcoming the major weaknesses of ICIs, including their lack of universality and durability. The last decade has seen tremendous advances in understanding how the immune system's ability to kill tumor cells requires the conspicuous metabolic specialization of T-cells. We have learned that cancer cell-associated metabolic activities trigger shifts in the abundance of some metabolites with immunosuppressory roles in the tumor microenvironment. Yet very little is known about the tumor cell-intrinsic metabolic traits that control the immune checkpoint contexture in cancer cells. Likewise, we lack a comprehensive understanding of how systemic metabolic perturbations in response to dietary interventions can reprogram the immune checkpoint landscape of tumor cells. We here review state-of-the-art molecular- and functional-level interrogation approaches to uncover how cell-autonomous metabolic traits and diet-mediated changes in nutrient availability and utilization might delineate new cancer cell-intrinsic metabolic dependencies of tumor immunogenicity. We propose that clinical monitoring and in-depth molecular evaluation of the cancer cell-intrinsic metabolic traits involved in primary, adaptive, and acquired resistance to cancer immunotherapy can provide the basis for improvements in therapeutic responses to ICIs. Overall, these approaches might guide the use of metabolic therapeutics and dietary approaches as novel strategies to broaden the spectrum of cancer patients and indications that can be effectively treated with ICI-based cancer immunotherapy.
Collapse
|
10
|
Metformin: Sentinel of the Epigenetic Landscapes That Underlie Cell Fate and Identity. Biomolecules 2020; 10:biom10050780. [PMID: 32443566 PMCID: PMC7277648 DOI: 10.3390/biom10050780] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/08/2020] [Accepted: 05/15/2020] [Indexed: 12/14/2022] Open
Abstract
The biguanide metformin is the first drug to be tested as a gerotherapeutic in the clinical trial TAME (Targeting Aging with Metformin). The current consensus is that metformin exerts indirect pleiotropy on core metabolic hallmarks of aging, such as the insulin/insulin-like growth factor 1 and AMP-activated protein kinase/mammalian Target Of Rapamycin signaling pathways, downstream of its primary inhibitory effect on mitochondrial respiratory complex I. Alternatively, but not mutually exclusive, metformin can exert regulatory effects on components of the biologic machinery of aging itself such as chromatin-modifying enzymes. An integrative metabolo-epigenetic outlook supports a new model whereby metformin operates as a guardian of cell identity, capable of retarding cellular aging by preventing the loss of the information-theoretic nature of the epigenome. The ultimate anti-aging mechanism of metformin might involve the global preservation of the epigenome architecture, thereby ensuring cell fate commitment and phenotypic outcomes despite the challenging effects of aging noise. Metformin might therefore inspire the development of new gerotherapeutics capable of preserving the epigenome architecture for cell identity. Such gerotherapeutics should replicate the ability of metformin to halt the erosion of the epigenetic landscape, mitigate the loss of cell fate commitment, delay stochastic/environmental DNA methylation drifts, and alleviate cellular senescence. Yet, it remains a challenge to confirm if regulatory changes in higher-order genomic organizers can connect the capacity of metformin to dynamically regulate the three-dimensional nature of epigenetic landscapes with the 4th dimension, the aging time.
Collapse
|
11
|
Impact of the Monocarboxylate Transporter-1 (MCT1)-Mediated Cellular Import of Lactate on Stemness Properties of Human Pancreatic Adenocarcinoma Cells †. Cancers (Basel) 2020; 12:cancers12030581. [PMID: 32138176 PMCID: PMC7139999 DOI: 10.3390/cancers12030581] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 02/28/2020] [Accepted: 03/01/2020] [Indexed: 02/07/2023] Open
Abstract
Metabolite exchange between stromal and tumor cells or among tumor cells themselves accompanies metabolic reprogramming in cancer including pancreatic adenocarcinoma (PDAC). Some tumor cells import and utilize lactate for oxidative energy production (reverse Warburg-metabolism) and the presence of these “reverse Warburg“ cells associates with a more aggressive phenotype and worse prognosis, though the underlying mechanisms are poorly understood. We now show that PDAC cells (BxPc3, A818-6, T3M4) expressing the lactate-importer monocarboxylate transporter-1 (MCT1) are protected by lactate against gemcitabine-induced apoptosis in a MCT1-dependent fashion, contrary to MCT1-negative PDAC cells (Panc1, Capan2). Moreover, lactate administration under glucose starvation, resembling reverse Warburg co a phenotype of BxPc3 and T3M4 cells that confers greater potential of clonal growth upon re-exposure to glucose, along with drug resistance and elevated expression of the stemness marker Nestin and reprogramming factors (Oct4, KLF4, Nanog). These lactate dependent effects on stemness properties are abrogated by the MCT1/lactate-uptake inhibitor 7ACC2 or MCT1 knock-down. Furthermore, the clinical relevance of these observations was supported by detecting co-expression of MCT1 and reprogramming factors in human PDAC tissues. In conclusion, the MCT1-dependent import of lactate supplies “reverse Warburg “PDAC cells with an efficient driver of metabostemness. This condition may essentially contribute to malignant traits including therapy resistance.
Collapse
|
12
|
Julià-Sapé M, Candiota AP, Arús C. Cancer metabolism in a snapshot: MRS(I). NMR IN BIOMEDICINE 2019; 32:e4054. [PMID: 30633389 DOI: 10.1002/nbm.4054] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 11/02/2018] [Accepted: 11/05/2018] [Indexed: 06/09/2023]
Abstract
The contribution of MRS(I) to the in vivo evaluation of cancer-metabolism-derived metrics, mostly since 2016, is reviewed here. Increased carbon consumption by tumour cells, which are highly glycolytic, is now being sampled by 13 C magnetic resonance spectroscopic imaging (MRSI) following the injection of hyperpolarized [1-13 C] pyruvate (Pyr). Hot-spots of, mostly, increased lactate dehydrogenase activity or flow between Pyr and lactate (Lac) have been seen with cancer progression in prostate (preclinical and in humans), brain and pancreas (both preclinical) tumours. Therapy response is usually signalled by decreased Lac/Pyr 13 C-labelled ratio with respect to untreated or non-responding tumour. For therapeutic agents inducing tumour hypoxia, the 13 C-labelled Lac/bicarbonate ratio may be a better metric than the Lac/Pyr ratio. 31 P MRSI may sample intracellular pH changes from brain tumours (acidification upon antiangiogenic treatment, basification at fast proliferation and relapse). The steady state tumour metabolome pattern is still in use for cancer evaluation. Metrics used for this range from quantification of single oncometabolites (such as 2-hydroxyglutarate in mutant IDH1 glial brain tumours) to selected metabolite ratios (such as total choline to N-acetylaspartate (plain ratio or CNI index)) or the whole 1 H MRSI(I) pattern through pattern recognition analysis. These approaches have been applied to address different questions such as tumour subtype definition, following/predicting the response to therapy or defining better resection or radiosurgery limits.
Collapse
Affiliation(s)
- Margarida Julià-Sapé
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Cerdanyola del Vallès, Spain
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
| | - Ana Paula Candiota
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Cerdanyola del Vallès, Spain
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
| | - Carles Arús
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Cerdanyola del Vallès, Spain
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
| |
Collapse
|
13
|
Crispo F, Condelli V, Lepore S, Notarangelo T, Sgambato A, Esposito F, Maddalena F, Landriscina M. Metabolic Dysregulations and Epigenetics: A Bidirectional Interplay that Drives Tumor Progression. Cells 2019; 8:E798. [PMID: 31366176 PMCID: PMC6721562 DOI: 10.3390/cells8080798] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/24/2019] [Accepted: 07/29/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer has been considered, for a long time, a genetic disease where mutations in keyregulatory genes drive tumor initiation, growth, metastasis, and drug resistance. Instead, theadvent of high-throughput technologies has revolutionized cancer research, allowing to investigatemolecular alterations at multiple levels, including genome, epigenome, transcriptome, proteome,and metabolome and showing the multifaceted aspects of this disease. The multi-omics approachesrevealed an intricate molecular landscape where different cellular functions are interconnected andcooperatively contribute to shaping the malignant phenotype. Recent evidence has brought to lighthow metabolism and epigenetics are highly intertwined, and their aberrant crosstalk can contributeto tumorigenesis. The oncogene-driven metabolic plasticity of tumor cells supports the energeticand anabolic demands of proliferative tumor programs and secondary can alter the epigeneticlandscape via modulating the production and/or the activity of epigenetic metabolites. Conversely,epigenetic mechanisms can regulate the expression of metabolic genes, thereby altering themetabolome, eliciting adaptive responses to rapidly changing environmental conditions, andsustaining malignant cell survival and progression in hostile niches. Thus, cancer cells takeadvantage of the epigenetics-metabolism crosstalk to acquire aggressive traits, promote cellproliferation, metastasis, and pluripotency, and shape tumor microenvironment. Understandingthis bidirectional relationship is crucial to identify potential novel molecular targets for theimplementation of robust anti-cancer therapeutic strategies.
Collapse
Affiliation(s)
- Fabiana Crispo
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata,85028 Rionero in Vulture, PZ, Italy.
| | - Valentina Condelli
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata,85028 Rionero in Vulture, PZ, Italy.
| | - Silvia Lepore
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata,85028 Rionero in Vulture, PZ, Italy.
| | - Tiziana Notarangelo
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata,85028 Rionero in Vulture, PZ, Italy.
| | - Alessandro Sgambato
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata,85028 Rionero in Vulture, PZ, Italy.
| | - Franca Esposito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II,80131 Naples, Italy.
| | - Francesca Maddalena
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata,85028 Rionero in Vulture, PZ, Italy.
| | - Matteo Landriscina
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata,85028 Rionero in Vulture, PZ, Italy.
- Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia,71100 Foggia, Italy.
| |
Collapse
|
14
|
Folguera-Blasco N, Pérez-Carrasco R, Cuyàs E, Menendez JA, Alarcón T. A multiscale model of epigenetic heterogeneity-driven cell fate decision-making. PLoS Comput Biol 2019; 15:e1006592. [PMID: 31039148 PMCID: PMC6510448 DOI: 10.1371/journal.pcbi.1006592] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 05/10/2019] [Accepted: 03/19/2019] [Indexed: 02/06/2023] Open
Abstract
The inherent capacity of somatic cells to switch their phenotypic status in response to damage stimuli in vivo might have a pivotal role in ageing and cancer. However, how the entry-exit mechanisms of phenotype reprogramming are established remains poorly understood. In an attempt to elucidate such mechanisms, we herein introduce a stochastic model of combined epigenetic regulation (ER)-gene regulatory network (GRN) to study the plastic phenotypic behaviours driven by ER heterogeneity. To deal with such complex system, we additionally formulate a multiscale asymptotic method for stochastic model reduction, from which we derive an efficient hybrid simulation scheme. Our analysis of the coupled system reveals a regime of tristability in which pluripotent stem-like and differentiated steady-states coexist with a third indecisive state, with ER driving transitions between these states. Crucially, ER heterogeneity of differentiation genes is for the most part responsible for conferring abnormal robustness to pluripotent stem-like states. We formulate epigenetic heterogeneity-based strategies capable of unlocking and facilitating the transit from differentiation-refractory (stem-like) to differentiation-primed epistates. The application of the hybrid numerical method validates the likelihood of such switching involving solely kinetic changes in epigenetic factors. Our results suggest that epigenetic heterogeneity regulates the mechanisms and kinetics of phenotypic robustness of cell fate reprogramming. The occurrence of tunable switches capable of modifying the nature of cell fate reprogramming might pave the way for new therapeutic strategies to regulate reparative reprogramming in ageing and cancer. Certain modifications of the structure and functioning of the protein/DNA complex called chromatin can allow adult, fully differentiated, cells to adopt a stem cell-like pluripotent state in a purely epigenetic manner, not involving changes in the underlying DNA sequence. Such reprogramming-like phenomena may constitute an innate reparative route through which human tissues respond to injury and could also serve as a novel regenerative strategy in human pathological situations in which tissue or organ repair is impaired. However, it should be noted that in vivo reprogramming would be capable of maintaining tissue homeostasis provided the acquisition of pluripotency features is strictly transient and accompanied by an accurate replenishment of the specific cell types being lost. Crucially, an excessive reprogramming in the absence of controlled re-differentiation would impair the repair or the replacement of damaged cells, thereby promoting pathological alterations of cell fate. A mechanistic understanding of how the degree of chromatin plasticity dictates the reparative versus pathological behaviour of in vivo reprogramming to rejuvenate aged tissues while preventing tumorigenesis is urgently needed, including especially the intrinsic epigenetic heterogeneity of the tissue resident cells being reprogrammed. We here introduce a novel method that mathematically captures how epigenetic heterogeneity is actually the driving force that governs the routes and kinetics to entry into and exit from a pathological stem-like state. Moreover, our approach computationally validates the likelihood of unlocking chronic, unrestrained plastic states and drive their differentiation down the correct path by solely manipulating the intensity and direction of few epigenetic control switches. Our approach could inspire new therapeutic approaches based on in vivo cell reprogramming for efficient tissue regeneration and rejuvenation and cancer treatment.
Collapse
Affiliation(s)
- Núria Folguera-Blasco
- Centre de Recerca Matemàtica, Edifici C, Campus de Bellaterra, 08193 Bellaterra, Barcelona, Spain
- Departament de Matemàtiques, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
- * E-mail:
| | - Rubén Pérez-Carrasco
- Department of Mathematics, University College London, Gower Street, London WC1E 6BT, UK
| | - Elisabet Cuyàs
- ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Javier A. Menendez
- ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Tomás Alarcón
- Centre de Recerca Matemàtica, Edifici C, Campus de Bellaterra, 08193 Bellaterra, Barcelona, Spain
- Departament de Matemàtiques, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
- ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
- Barcelona Graduate School of Mathematics (BGSMath), Barcelona, Spain
| |
Collapse
|
15
|
da Silva I, da Costa Vieira R, Stella C, Loturco E, Carvalho AL, Veo C, Neto C, Silva SM, D'Amora P, Salzgeber M, Matos D, Silva CR, Oliveira JR, Rabelo I, Yamakawa P, Maciel R, Biscolla R, Chiamolera M, Fraietta R, Reis F, Mori M, Marchioni D, Carioca A, Maciel G, Tomioka R, Baracat E, Silva C, Granato C, Diaz R, Scarpellini B, Egle D, Fiegl H, Himmel I, Troi C, Nagourney R. Inborn-like errors of metabolism are determinants of breast cancer risk, clinical response and survival: a study of human biochemical individuality. Oncotarget 2018; 9:31664-31681. [PMID: 30167086 PMCID: PMC6114970 DOI: 10.18632/oncotarget.25839] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/12/2018] [Indexed: 01/16/2023] Open
Abstract
Breast cancer remains a leading cause of morbidity and mortality worldwide yet methods for early detection remain elusive. We describe the discovery and validation of biochemical signatures measured by mass spectrometry, performed upon blood samples from patients and controls that accurately identify (>95%) the presence of clinical breast cancer. Targeted quantitative MS/MS conducted upon 1225 individuals, including patients with breast and other cancers, normal controls as well as individuals with a variety of metabolic disorders provide a biochemical phenotype that accurately identifies the presence of breast cancer and predicts response and survival following the administration of neoadjuvant chemotherapy. The metabolic changes identified are consistent with inborn-like errors of metabolism and define a continuum from normal controls to elevated risk to invasive breast cancer. Similar results were observed in other adenocarcinomas but were not found in squamous cell cancers or hematologic neoplasms. The findings describe a new early detection platform for breast cancer and support a role for pre-existing, inborn-like errors of metabolism in the process of breast carcinogenesis that may also extend to other glandular malignancies. Statement of Significance: Findings provide a powerful tool for early detection and the assessment of prognosis in breast cancer and define a novel concept of breast carcinogenesis that characterizes malignant transformation as the clinical manifestation of underlying metabolic insufficiencies.
Collapse
Affiliation(s)
- Ismael da Silva
- Gynecology Department, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil.,Fleury Laboratories, São Paulo, Brazil.,Barretos Cancer Hospital (HCB), Barretos, Brazil
| | | | - Carolina Stella
- Gynecology Department, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Edson Loturco
- Department of Surgery, Urology Unit, Human Reproduction Division, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | | | - Carlos Veo
- Barretos Cancer Hospital (HCB), Barretos, Brazil
| | | | | | - Paulo D'Amora
- Gynecology Department, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Marcia Salzgeber
- Gynecology Department, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Delcio Matos
- Department of Surgery, Surgical Gastroenterology Division, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Celso R Silva
- Clinical and Experimental Oncology Department, Hematology and Hemotherapy Division, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Jose R Oliveira
- Clinical and Experimental Oncology Department, Hematology and Hemotherapy Division, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Iara Rabelo
- Clinical and Experimental Oncology Department, Hematology and Hemotherapy Division, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Patricia Yamakawa
- Clinical and Experimental Oncology Department, Hematology and Hemotherapy Division, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Rui Maciel
- Fleury Laboratories, São Paulo, Brazil.,Department of Medicine, Endocrinology Division, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Rosa Biscolla
- Department of Medicine, Endocrinology Division, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Maria Chiamolera
- Department of Medicine, Endocrinology Division, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Renato Fraietta
- Department of Surgery, Urology Unit, Human Reproduction Division, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Felipe Reis
- Biophysics Department, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Marcelo Mori
- Department of Biochemistry and Tissue Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Dirce Marchioni
- Nutrition Department, School of Public Health, University of São Paulo School of Medicine (FMUSP), São Paulo, Brazil
| | - Antonio Carioca
- Nutrition Department, School of Public Health, University of São Paulo School of Medicine (FMUSP), São Paulo, Brazil
| | - Gustavo Maciel
- Fleury Laboratories, São Paulo, Brazil.,Department of Obstetrics and Gynecology, University of São Paulo School of Medicine (HCFMUSP), São Paulo, Brazil
| | - Renato Tomioka
- Department of Obstetrics and Gynecology, University of São Paulo School of Medicine (HCFMUSP), São Paulo, Brazil
| | - Edmund Baracat
- Department of Obstetrics and Gynecology, University of São Paulo School of Medicine (HCFMUSP), São Paulo, Brazil
| | - Clovis Silva
- Department of Pediatrics, Children's Hospital, University of São Paulo School of Medicine (HCFMUSP), São Paulo, Brazil
| | - Celso Granato
- Fleury Laboratories, São Paulo, Brazil.,Retrovirology Laboratory, Infectious Diseases Unit, Medicine Department, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Ricardo Diaz
- Retrovirology Laboratory, Infectious Diseases Unit, Medicine Department, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Bruno Scarpellini
- Fleury Laboratories, São Paulo, Brazil.,Retrovirology Laboratory, Infectious Diseases Unit, Medicine Department, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Daniel Egle
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Heidi Fiegl
- Department of Gynecology, Meran Hospital, Meran, Italy
| | | | - Christina Troi
- Department of Gynecology, Brixen Hospital, Brixen, Italy
| | - Robert Nagourney
- Department of Obstetrics and Gynecology, Gynecological Oncology Unit, University of California Irvine (UCI), California, USA
| |
Collapse
|
16
|
Cuyàs E, Verdura S, Folguera-Blasco N, Bastidas-Velez C, Martin ÁG, Alarcón T, Menendez JA. Mitostemness. Cell Cycle 2018; 17:918-926. [PMID: 29886796 DOI: 10.1080/15384101.2018.1467679] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Unraveling the key mechanisms governing the retention versus loss of the cancer stem cell (CSC) state would open new therapeutic avenues to eradicate cancer. Mitochondria are increasingly recognized key drivers in the origin and development of CSC functional traits. We here propose the new term "mitostemness" to designate the mitochondria-dependent signaling functions that, evolutionary rooted in the bacterial origin of mitochondria, regulate the maintenance of CSC self-renewal and resistance to differentiation. Mitostemness traits, namely mitonuclear communication, mitoproteome components, and mitochondrial fission/fusion dynamics, can be therapeutically exploited to target the CSC state. We briefly review the pre-clinical evidence of action of investigational compounds on mitostemness traits and discuss ongoing strategies to accelerate the clinical translation of new mitostemness drugs. The recognition that the bacterial origin of present-day mitochondria can drive decision-making signaling phenomena may open up a new therapeutic dimension against life-threatening CSCs. New therapeutics aimed to target mitochondria not only as biochemical but also as biophysical and morpho-physiological hallmarks of CSC might certainly guide improvements to cancer treatment.
Collapse
Affiliation(s)
- Elisabet Cuyàs
- a Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group , Catalan Institute of Oncology , Girona , Spain.,b Girona Biomedical Research Institute (IDIBGI) , Girona , Spain
| | - Sara Verdura
- a Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group , Catalan Institute of Oncology , Girona , Spain.,b Girona Biomedical Research Institute (IDIBGI) , Girona , Spain
| | | | | | | | - Tomás Alarcón
- c Centre de Recerca Matemàtica , Barcelona , Spain.,e Barcelona Graduate School of Mathematics (BGSMath) , Barcelona , Spain.,f ICREA , Barcelona , Spain.,g Departament de Matemàtiques , Universitat Autònoma de Barcelona , Barcelona , Spain
| | - Javier A Menendez
- a Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group , Catalan Institute of Oncology , Girona , Spain.,b Girona Biomedical Research Institute (IDIBGI) , Girona , Spain
| |
Collapse
|
17
|
Ong AL, Ramasamy TS. Role of Sirtuin1-p53 regulatory axis in aging, cancer and cellular reprogramming. Ageing Res Rev 2018; 43:64-80. [PMID: 29476819 DOI: 10.1016/j.arr.2018.02.004] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 01/23/2018] [Accepted: 02/16/2018] [Indexed: 12/12/2022]
Abstract
Regulatory role of Sirtuin 1 (SIRT1), one of the most extensively studied members of its kind in histone deacetylase family in governing multiple cellular fates, is predominantly linked to p53 activity. SIRT1 deacetylates p53 in a NAD+-dependent manner to inhibit transcription activity of p53, in turn modulate pathways that are implicated in regulation of tissue homoeostasis and many disease states. In this review, we discuss the role of SIRT1-p53 pathway and its regulatory axis in the cellular events which are implicated in cellular aging, cancer and reprogramming. It is noteworthy that these cellular events share few common regulatory pathways, including SIRT1-p53-LDHA-Myc, miR-34a,-Let7 regulatory network, which forms a positive feedback loop that controls cell cycle, metabolism, proliferation, differentiation, epigenetics and many others. In the context of aging, SIRT1 expression is reduced as a protective mechanism against oncogenesis and for maintenance of tissue homeostasis. Interestingly, its activation in aged cells is evidenced in response to DNA damage to protect the cells from p53-dependent apoptosis or senescence, predispose these cells to neoplastic transformation. Importantly, the dual roles of SIRT1-p53 axis in aging and tumourigenesis, either as tumour suppressor or tumour promoter are determined by SIRT1 localisation and type of cells. Conceptualising the distinct similarity between tumorigenesis and cellular reprogramming, this review provides a perspective discussion on involvement of SIRT1 in improving efficiency in the induction and maintenance of pluripotent state. Further research in understanding the role of SIRT1-p53 pathway and their associated regulators and strategies to manipulate this regulatory axis very likely foster the development of therapeutics and strategies for treating cancer and aging-associated degenerative diseases.
Collapse
|
18
|
Folguera-Blasco N, Cuyàs E, Menéndez JA, Alarcón T. Epigenetic regulation of cell fate reprogramming in aging and disease: A predictive computational model. PLoS Comput Biol 2018; 14:e1006052. [PMID: 29543808 PMCID: PMC5871006 DOI: 10.1371/journal.pcbi.1006052] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 03/27/2018] [Accepted: 02/21/2018] [Indexed: 01/12/2023] Open
Abstract
Understanding the control of epigenetic regulation is key to explain and modify the aging process. Because histone-modifying enzymes are sensitive to shifts in availability of cofactors (e.g. metabolites), cellular epigenetic states may be tied to changing conditions associated with cofactor variability. The aim of this study is to analyse the relationships between cofactor fluctuations, epigenetic landscapes, and cell state transitions. Using Approximate Bayesian Computation, we generate an ensemble of epigenetic regulation (ER) systems whose heterogeneity reflects variability in cofactor pools used by histone modifiers. The heterogeneity of epigenetic metabolites, which operates as regulator of the kinetic parameters promoting/preventing histone modifications, stochastically drives phenotypic variability. The ensemble of ER configurations reveals the occurrence of distinct epi-states within the ensemble. Whereas resilient states maintain large epigenetic barriers refractory to reprogramming cellular identity, plastic states lower these barriers, and increase the sensitivity to reprogramming. Moreover, fine-tuning of cofactor levels redirects plastic epigenetic states to re-enter epigenetic resilience, and vice versa. Our ensemble model agrees with a model of metabolism-responsive loss of epigenetic resilience as a cellular aging mechanism. Our findings support the notion that cellular aging, and its reversal, might result from stochastic translation of metabolic inputs into resilient/plastic cell states via ER systems.
Collapse
Affiliation(s)
- Núria Folguera-Blasco
- Centre de Recerca Matemàtica, Edifici C, Campus de Bellaterra, Bellaterra (Barcelona), Spain
- Departament de Matemàtiques, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain
| | - Elisabet Cuyàs
- Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain
- MetaboStem, Barcelona, Spain
| | - Javier A. Menéndez
- Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain
- MetaboStem, Barcelona, Spain
- ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain
| | - Tomás Alarcón
- Centre de Recerca Matemàtica, Edifici C, Campus de Bellaterra, Bellaterra (Barcelona), Spain
- Departament de Matemàtiques, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain
- ICREA, Pg. Lluís Companys 23, Barcelona, Spain
- Barcelona Graduate School of Mathematics (BGSMath), Barcelona, Spain
| |
Collapse
|
19
|
El-Badawy A, Ghoneim MA, Gabr MM, Salah RA, Mohamed IK, Amer M, El-Badri N. Cancer cell-soluble factors reprogram mesenchymal stromal cells to slow cycling, chemoresistant cells with a more stem-like state. Stem Cell Res Ther 2017; 8:254. [PMID: 29115987 PMCID: PMC5688803 DOI: 10.1186/s13287-017-0709-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/24/2017] [Accepted: 10/23/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) play different roles in modulating tumor progression, growth, and metastasis. MSCs are recruited to the tumor site in large numbers and subsequently have an important microenvironmental role in modulating tumor progression and drug sensitivity. However, the effect of the tumor microenvironment on MSC plasticity remains poorly understood. Herein, we report a paracrine effect of cancer cells, in which they secrete soluble factors that promote a more stem-like state in bone marrow mesenchymal stem cells (BM-MSCs). METHODS The effect of soluble factors secreted from MCF7, Hela, and HepG2 cancer cell lines on BM-MSCs was assessed using a Transwell indirect coculture system. After 5 days of coculture, BM-MSCs were characterized by flow cytometry for surface marker expression, by qPCR for gene expression profile, and by confocal immunofluorescence for marker expression. We then measured the sensitivity of cocultured BM-MSCs to chemotherapeutic agents, their cell cycle profile, and their response to DNA damage. The sphere formation, invasive properties, and in-vivo performance of BM-MSCs after coculture with cancer cells were also measured. RESULTS Indirect coculture of cancer cells and BM-MSCs, without direct cell contact, generated slow cycling, chemoresistant spheroid stem cells that highly expressed markers of pluripotency, cancer cells, and cancer stem cells (CSCs). They also displayed properties of a side population and enhanced sphere formation in culture. Accordingly, these cells were termed cancer-induced stem cells (CiSCs). CiSCs showed a more mesenchymal phenotype that was further augmented upon TGF-β stimulation and demonstrated a high expression of the β-catenin pathway and ALDH1A1. CONCLUSIONS These findings demonstrate that MSCs, recruited to the tumor microenvironment in large numbers, may display cellular plasticity, acquire a more stem-like state, and acquire some properties of CSCs upon exposure to cancer cell-secreted factors. These acquired characteristics may contribute to tumor progression, survival, and metastasis. Our findings provide new insights into the interactions between MSCs and cancer cells, with the potential to identify novel molecular targets for cancer therapy.
Collapse
Affiliation(s)
- Ahmed El-Badawy
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Sheikh Zayed District, 12588, 6th of October City, Giza, Egypt
| | | | - Mahmoud M. Gabr
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Radwa Ayman Salah
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Sheikh Zayed District, 12588, 6th of October City, Giza, Egypt
| | - Ihab K. Mohamed
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Marwa Amer
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Sheikh Zayed District, 12588, 6th of October City, Giza, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Sheikh Zayed District, 12588, 6th of October City, Giza, Egypt
| |
Collapse
|
20
|
Metformin regulates global DNA methylation via mitochondrial one-carbon metabolism. Oncogene 2017; 37:963-970. [PMID: 29059169 DOI: 10.1038/onc.2017.367] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 08/15/2017] [Accepted: 08/16/2017] [Indexed: 12/11/2022]
Abstract
The anti-diabetic biguanide metformin may exert health-promoting effects via metabolic regulation of the epigenome. Here we show that metformin promotes global DNA methylation in non-cancerous, cancer-prone and metastatic cancer cells by decreasing S-adenosylhomocysteine (SAH), a strong feedback inhibitor of S-adenosylmethionine (SAM)-dependent DNA methyltransferases, while promoting the accumulation of SAM, the universal methyl donor for cellular methylation. Using metformin and a mitochondria/complex I (mCI)-targeted analog of metformin (norMitoMet) in experimental pairs of wild-type and AMP-activated protein kinase (AMPK)-, serine hydroxymethyltransferase 2 (SHMT2)- and mCI-null cells, we provide evidence that metformin increases the SAM:SAH ratio-related methylation capacity by targeting the coupling between serine mitochondrial one-carbon flux and CI activity. By increasing the contribution of one-carbon units to the SAM from folate stores while decreasing SAH in response to AMPK-sensed energetic crisis, metformin can operate as a metabolo-epigenetic regulator capable of reprogramming one of the key conduits linking cellular metabolism to the DNA methylation machinery.
Collapse
|
21
|
Cuyàs E, Verdura S, Fernández-Arroyo S, Bosch-Barrera J, Martin-Castillo B, Joven J, Menendez JA. Metabolomic mapping of cancer stem cells for reducing and exploiting tumor heterogeneity. Oncotarget 2017; 8:99223-99236. [PMID: 29245896 PMCID: PMC5725087 DOI: 10.18632/oncotarget.21834] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 09/29/2017] [Indexed: 02/06/2023] Open
Abstract
Personalized cancer medicine based on the analysis of tumors en masse is limited by tumor heterogeneity, which has become a major obstacle to effective cancer treatment. Cancer stem cells (CSC) are emerging as key drivers of inter- and intratumoral heterogeneity. CSC have unique metabolic dependencies that are required not only for specific bioenergetic/biosynthetic demands but also for sustaining their operational epigenetic traits, i.e. self-renewal, tumor-initiation, and plasticity. Given that the metabolome is the final downstream product of all the –omic layers and, therefore, most representative of the biological phenotype, we here propose that a novel approach to better understand the complexity of tumor heterogeneity is by mapping and cataloging small numbers of CSC metabolomic phenotypes. The narrower metabolomic diversity of CSC states could be employed to reduce multidimensional tumor heterogeneity into dynamic models of fewer actionable sub-phenotypes. The identification of the driver nodes that are used differentially by CSC states to metabolically regulate self-renewal and tumor initation and escape chemotherapy might open new preventive and therapeutic avenues. The mapping of CSC metabolomic states could become a pioneering strategy to reduce the dimensionality of tumor heterogeneity and improve our ability to examine changes in tumor cell populations for cancer detection, prognosis, prediction/monitoring of therapy response, and detection of therapy resistance and recurrent disease. The identification of driver metabolites and metabolic nodes accounting for a large amount of variance within the CSC metabolomic sub-phenotypes might offer new unforeseen opportunities for reducing and exploiting tumor heterogeneity via metabolic targeting of CSC.
Collapse
Affiliation(s)
- Elisabet Cuyàs
- Metabolism and Cancer Group, Program Against Cancer Therapeutic Resistance, Catalan Institute of Oncology, Girona, Spain.,Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Sara Verdura
- Metabolism and Cancer Group, Program Against Cancer Therapeutic Resistance, Catalan Institute of Oncology, Girona, Spain.,Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Salvador Fernández-Arroyo
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Catalonia, Spain.,Campus of International Excellence Southern Catalonia, Tarragona, Catalonia, Spain
| | | | | | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Catalonia, Spain.,Campus of International Excellence Southern Catalonia, Tarragona, Catalonia, Spain
| | - Javier A Menendez
- Metabolism and Cancer Group, Program Against Cancer Therapeutic Resistance, Catalan Institute of Oncology, Girona, Spain.,Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| |
Collapse
|
22
|
Diehl K, Dinges LA, Helm O, Ammar N, Plundrich D, Arlt A, Röcken C, Sebens S, Schäfer H. Nuclear factor E2-related factor-2 has a differential impact on MCT1 and MCT4 lactate carrier expression in colonic epithelial cells: a condition favoring metabolic symbiosis between colorectal cancer and stromal cells. Oncogene 2017; 37:39-51. [DOI: 10.1038/onc.2017.299] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 07/03/2017] [Accepted: 07/21/2017] [Indexed: 12/28/2022]
|
23
|
Menendez JA, Alarcón T. Senescence-Inflammatory Regulation of Reparative Cellular Reprogramming in Aging and Cancer. Front Cell Dev Biol 2017; 5:49. [PMID: 28529938 PMCID: PMC5418360 DOI: 10.3389/fcell.2017.00049] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 04/18/2017] [Indexed: 12/16/2022] Open
Abstract
The inability of adult tissues to transitorily generate cells with functional stem cell-like properties is a major obstacle to tissue self-repair. Nuclear reprogramming-like phenomena that induce a transient acquisition of epigenetic plasticity and phenotype malleability may constitute a reparative route through which human tissues respond to injury, stress, and disease. However, tissue rejuvenation should involve not only the transient epigenetic reprogramming of differentiated cells, but also the committed re-acquisition of the original or alternative committed cell fate. Chronic or unrestrained epigenetic plasticity would drive aging phenotypes by impairing the repair or the replacement of damaged cells; such uncontrolled phenomena of in vivo reprogramming might also generate cancer-like cellular states. We herein propose that the ability of senescence-associated inflammatory signaling to regulate in vivo reprogramming cycles of tissue repair outlines a threshold model of aging and cancer. The degree of senescence/inflammation-associated deviation from the homeostatic state may delineate a type of thresholding algorithm distinguishing beneficial from deleterious effects of in vivo reprogramming. First, transient activation of NF-κB-related innate immunity and senescence-associated inflammatory components (e.g., IL-6) might facilitate reparative cellular reprogramming in response to acute inflammatory events. Second, para-inflammation switches might promote long-lasting but reversible refractoriness to reparative cellular reprogramming. Third, chronic senescence-associated inflammatory signaling might lock cells in highly plastic epigenetic states disabled for reparative differentiation. The consideration of a cellular reprogramming-centered view of epigenetic plasticity as a fundamental element of a tissue's capacity to undergo successful repair, aging degeneration or malignant transformation should provide challenging stochastic insights into the current deterministic genetic paradigm for most chronic diseases, thereby increasing the spectrum of therapeutic approaches for physiological aging and cancer.
Collapse
Affiliation(s)
- Javier A Menendez
- Metabolism and Cancer Group, Program Against Cancer Therapeutic Resistance, Catalan Institute of OncologyGirona, Spain.,Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI)Girona, Spain.,METABOSTEMBarcelona, Spain
| | - Tomás Alarcón
- Institució Catalana de Recerca i Estudis Avançats (ICREA)Barcelona, Spain.,Computational and Mathematical Biology Research Group, Centre de Recerca MatemàticaBarcelona, Spain.,Departament de Matemàtiques, Universitat Autònoma de BarcelonaBarcelona, Spain.,Barcelona Graduate School of MathematicsBarcelona, Spain
| |
Collapse
|
24
|
Menendez JA, Alarcón T. Nuclear reprogramming of cancer stem cells: Corrupting the epigenetic code of cell identity with oncometabolites. Mol Cell Oncol 2017; 3:e1160854. [PMID: 28090573 DOI: 10.1080/23723556.2016.1160854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 02/27/2016] [Accepted: 02/28/2016] [Indexed: 01/24/2023]
Abstract
Generation of cancer stem cell (CSC)-like cells might occur through metabolic corruption of the epigenetic codes that govern cell identity. We recently identified how archetypal oncometabolites, without altering the baseline expression of endogenous stem cell maintenance genes but endowing cells with epigenetic states refractory to differentiation, considerably enhance the global kinetic efficiency of nuclear reprogramming processes that generate CSC-like states de novo. This study highlights that metabolo-epigenetic axes of communication can direct the development and maintenance of CSCs during the natural history of cancer diseases.
Collapse
Affiliation(s)
- Javier A Menendez
- ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Catalonia, Spain; Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI), Salt, Catalonia, Spain
| | - Tomás Alarcón
- Computational & Mathematical Biology Research Group, Center de Recerca Matemàtica, Barcelona, Catalonia, Spain; ICREA (Institució Catalana d'Estudis i Recerca Avançats), Barcelona, Spain; Departament de Matemàtiques, Universitat Autónoma de Barcelona, Barcelona, Spain; Barcelona Graduate School of Mathematics (BGSMath), Barcelona, Spain
| |
Collapse
|
25
|
Cuyàs E, Fernández-Arroyo S, Joven J, Menendez JA. Metformin targets histone acetylation in cancer-prone epithelial cells. Cell Cycle 2016; 15:3355-3361. [PMID: 27792453 DOI: 10.1080/15384101.2016.1249547] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The usage of metabolic intermediates as substrates for chromatin-modifying enzymes provides a direct link between the metabolic state of the cell and epigenetics. Because this metabolism-epigenetics axis can regulate not only normal but also diseased states, it is reasonable to suggest that manipulating the epigenome via metabolic interventions may improve the clinical manifestation of age-related diseases including cancer. Using a model of BRCA1 haploinsufficiency-driven accelerated geroncogenesis, we recently tested the hypothesis that: 1.) metabolic rewiring of the mitochondrial biosynthetic nodes that overproduce epigenetic metabolites such as acetyl-CoA should promote cancer-related acetylation of histone H3 marks; 2.) metformin-induced restriction of mitochondrial biosynthetic capacity should manifest in the epigenetic regulation of histone acetylation. We now provide one of the first examples of how metformin-driven metabolic shifts such as reduction of the 2-carbon epigenetic substrate acetyl-CoA is sufficient to correct specific histone H3 acetylation marks in cancer-prone human epithelial cells. The ability of metformin to regulate mitonuclear communication and modulate the epigenetic landscape in genomically unstable pre-cancerous cells might guide the development of new metabolo-epigenetic strategies for cancer prevention and therapy.
Collapse
Affiliation(s)
- Elisabet Cuyàs
- a ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism & Cancer Group, Catalan Institute of Oncology , Girona , Catalonia , Spain.,b Girona Biomedical Research Institute (IDIBGI) , Girona , Catalonia , Spain
| | - Salvador Fernández-Arroyo
- c Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain, The Campus of International Excellence Southern Catalonia , Tarragona , Spain
| | - Jorge Joven
- c Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain, The Campus of International Excellence Southern Catalonia , Tarragona , Spain
| | - Javier A Menendez
- a ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism & Cancer Group, Catalan Institute of Oncology , Girona , Catalonia , Spain.,b Girona Biomedical Research Institute (IDIBGI) , Girona , Catalonia , Spain
| |
Collapse
|
26
|
Corominas-Faja B, Vellon L, Cuyàs E, Buxó M, Martin-Castillo B, Serra D, García J, Lupu R, Menendez JA. Clinical and therapeutic relevance of the metabolic oncogene fatty acid synthase in HER2+ breast cancer. Histol Histopathol 2016; 32:687-698. [PMID: 27714708 DOI: 10.14670/hh-11-830] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Fatty acid synthase (FASN) is a key lipogenic enzyme for de novo fatty acid biosynthesis and a druggable metabolic oncoprotein that is activated in most human cancers. We evaluated whether the HER2-driven lipogenic phenotype might represent a biomarker for sensitivity to pharmacological FASN blockade. A majority of clinically HER2-positive tumors were scored as FASN overexpressors in a series of almost 200 patients with invasive breast carcinoma. Re-classification of HER2-positive breast tumors based on FASN gene expression predicted a significantly inferior relapse-free and distant metastasis-free survival in HER2+/FASN+ patients. Notably, non-tumorigenic MCF10A breast epithelial cells engineered to overexpress HER2 upregulated FASN gene expression, and the FASN inhibitor C75 abolished HER2-induced anchorage-independent growth and survival. Furthermore, in the presence of high concentrations of C75, HER2-negative MCF-7 breast cancer cells overexpressing HER2 (MCF-7/HER2) had significantly higher levels of apoptosis than HER2-negative cells. Finally, C75 at non-cytotoxic concentrations significantly reduced the capacity of MCF-7/HER2 cells to form mammospheres, an in vitro indicator of cancer stem-like cells. Collectively, our findings strongly suggest that the HER2-FASN lipogenic axis delineates a group of breast cancer patients that might benefit from treatment with therapeutic regimens containing FASN inhibitors.
Collapse
Affiliation(s)
- Bruna Corominas-Faja
- ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain.,Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Luciano Vellon
- IBYME, CONICET-Laboratorio de Immunohematología, Buenos Aires, Argentina
| | - Elisabet Cuyàs
- ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain.,Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Maria Buxó
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Begoña Martin-Castillo
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain.,Unit of Clinical Research, Catalan Institute of Oncology, Girona, Spain
| | - Dolors Serra
- Department of Biochemistry and Molecular Biology, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jordi García
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.,Departament de Química Orgànica, Facultat de Química, Institut de Biomedicina de la UB (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Ruth Lupu
- Mayo Clinic, Department of Laboratory Medicine and Pathology, Division of Experimental Pathology, Rochester, MN, USA.,Mayo Clinic Cancer Center, Rochester, MN, USA.
| | - Javier A Menendez
- ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain.,Girona Biomedical Research Institute (IDIBGI), Girona, Spain.
| |
Collapse
|
27
|
Hernández-Aguilera A, Fernández-Arroyo S, Cuyàs E, Luciano-Mateo F, Cabre N, Camps J, Lopez-Miranda J, Menendez JA, Joven J. Epigenetics and nutrition-related epidemics of metabolic diseases: Current perspectives and challenges. Food Chem Toxicol 2016; 96:191-204. [PMID: 27503834 DOI: 10.1016/j.fct.2016.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/03/2016] [Accepted: 08/04/2016] [Indexed: 02/07/2023]
Abstract
We live in a world fascinated by the relationship between disease and nutritional disequilibrium. The subtle and slow effects of chronic nutrient toxicity are a major public health concern. Since food is potentially important for the development of "metabolic memory", there is a need for more information on the type of nutrients causing adverse or toxic effects. We now know that metabolic alterations produced by excessive intake of some nutrients, drugs and chemicals directly impact epigenetic regulation. We envision that understanding how metabolic pathways are coordinated by environmental and genetic factors will provide novel insights for the treatment of metabolic diseases. New methods will enable the assembly and analysis of large sets of complex molecular and clinical data for understanding how inflammation and mitochondria affect bioenergetics, epigenetics and health. Collectively, the observations we highlight indicate that energy utilization and disease are intimately connected by epigenetics. The challenge is to incorporate metabolo-epigenetic data in better interpretations of disease, to expedite therapeutic targeting of key pathways linking nutritional toxicity and metabolism. An additional concern is that changes in the parental phenotype are detectable in the methylome of subsequent offspring. The effect might create a menace to future generations and preconceptional considerations.
Collapse
Affiliation(s)
- Anna Hernández-Aguilera
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Salvador Fernández-Arroyo
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Elisabet Cuyàs
- Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain; ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain
| | - Fedra Luciano-Mateo
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Noemi Cabre
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Jordi Camps
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Jose Lopez-Miranda
- Lipid and Atherosclerosis Unit, IMIBIC, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain; CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier A Menendez
- Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain; ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain.
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain; The Campus of International Excellence Southern Catalonia, Tarragona, Spain.
| |
Collapse
|