1
|
Pragati, Sarkar S. Targeted downregulation of insulin signaling restricts human tau pathogenesis by reinstating the aberrant heterochromatin loss and mTOR/4EBP/S6K pathway in Drosophila. Brain Res 2025; 1849:149347. [PMID: 39579954 DOI: 10.1016/j.brainres.2024.149347] [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: 09/10/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 11/25/2024]
Abstract
Tauopathies are a group of neurodegenerative diseases characterized by the accumulation of paired helical filaments (PHFs)/or neurofibrillary tangles (NFTs) in neuronal/glial cells. Besides hyperphosphorylation of tau protein, aberrant heterochromatin loss and translation dysfunction have emerged as other important aspects contributing to the disease pathogenesis. We have recently reported that tissue-specific downregulation of insulin signaling or its growth-promoting downstream sub-branch effectively reinstates the tau-mediated overactivated insulin pathway, and restricts pathogenic tau hyperphosphorylation and aggregate formation. We next investigated if the downregulation of the insulin pathway or its growth-promoting downstream sub-branch makes any impact on tau-mediated aberrant heterochromatin loss and translation dysfunction. For the first time, we demonstrate that tissue-specific downregulation of insulin signaling or its growth-promoting branch effectively restricts the pathogenic tau-induced heterochromatin loss. We further report that expression of human tau in Drosophila causes induction of the mTOR/4EBP/S6K pathway and energy disbalance which gets effectively balanced upon downregulation of insulin signaling. Our findings establish an imperative role of insulin signaling in effectively mitigating various aspects of tau etiology in Drosophila ranging from hyperphosphorylation, chromatin relaxation, and translational upsurge. Our findings could be beneficial in establishing novel therapeutic options against tauopathies.
Collapse
Affiliation(s)
- Pragati
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi 110 021, India
| | - Surajit Sarkar
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi 110 021, India.
| |
Collapse
|
2
|
Dikeocha IJ, Wardill HR, Coller JK, Bowen JM. Dietary interventions and tumor response to chemotherapy in breast cancer: A comprehensive review of preclinical and clinical data. Clin Nutr ESPEN 2024; 63:462-475. [PMID: 39018241 DOI: 10.1016/j.clnesp.2024.06.048] [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: 05/21/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND & AIMS Optimizing treatment efficacy is still a critical part in advancing the treatment of breast cancer. Dietary interventions have drawn significant attention for their potential to increase tumor sensitivity, with a plethora of strategies evaluated both preclinically and clinically. The aim of this paper is to explore these strategies, ranging from entire dietary programs to specific supplements, for their potential to directly enhance tumor sensitivity and chemotherapy adherence. METHODS PubMed, Scopus, Embase and Web of Science databases were searched up to September 2023. In this comprehensive review, preclinical and clinical research on dietary interventions used in conjunction with chemotherapy for breast cancer was examined and synthesized, to identify potential causal mechanisms. RESULTS 42 studies in total were identified and synthesized, 32 pre-clinical and 8 clinical studies. CONCLUSION Although a topic of intense interest, the heterogeneity in approaches has resulted in a large but minimally impactful evidence base, further complicated by a limited understanding of the mechanisms at play. This review highlights the areas for further research to increase opportunities for nutritional-based interventions as adjuvant to chemotherapy for breast cancer.
Collapse
Affiliation(s)
- Ifeoma J Dikeocha
- Discipline of Physiology, School of Biomedicine, The University of Adelaide, Level 2 Helen Mayo South, North Terrace, Adelaide, SA 5000, Australia.
| | - Hannah R Wardill
- Supportive Oncology Research Group, Precision Cancer Medicine, The South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Janet K Coller
- Discipline of Pharmacology, School of Biomedicine, The University of Adelaide, Level 2 Helen Mayo South, North Terrace, Adelaide, SA 5000, Australia
| | - Joanne M Bowen
- Discipline of Physiology, School of Biomedicine, The University of Adelaide, Level 2 Helen Mayo South, North Terrace, Adelaide, SA 5000, Australia
| |
Collapse
|
3
|
Vidal CM, Alva-Ornelas JA, Chen NZ, Senapati P, Tomsic J, Robles VM, Resto C, Sanchez N, Sanchez A, Hyslop T, Emwas N, Aljaber D, Bachelder N, Martinez E, Ann D, Jones V, Winn RA, Miele L, Ochoa AC, Dietze EC, Natarajan R, Schones D, Seewaldt VL. Insulin Resistance in Women Correlates with Chromatin Histone Lysine Acetylation, Inflammatory Signaling, and Accelerated Aging. Cancers (Basel) 2024; 16:2735. [PMID: 39123463 PMCID: PMC11311683 DOI: 10.3390/cancers16152735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Epigenetic changes link medical, social, and environmental factors with cardiovascular and kidney disease and, more recently, with cancer. The mechanistic link between metabolic health and epigenetic changes is only starting to be investigated. In our in vitro and in vivo studies, we performed a broad analysis of the link between hyperinsulinemia and chromatin acetylation; our top "hit" was chromatin opening at H3K9ac. METHODS Building on our published preclinical studies, here, we performed a detailed analysis of the link between insulin resistance, chromatin acetylation, and inflammation using an initial test set of 28 women and validation sets of 245, 22, and 53 women. RESULTS ChIP-seq identified chromatin acetylation and opening at the genes coding for TNFα and IL6 in insulin-resistant women. Pathway analysis identified inflammatory response genes, NFκB/TNFα-signaling, reactome cytokine signaling, innate immunity, and senescence. Consistent with this finding, flow cytometry identified increased senescent circulating peripheral T-cells. DNA methylation analysis identified evidence of accelerated aging in insulin-resistant vs. metabolically healthy women. CONCLUSIONS This study shows that insulin-resistant women have increased chromatin acetylation/opening, inflammation, and, perhaps, accelerated aging. Given the role that inflammation plays in cancer initiation and progression, these studies provide a potential mechanistic link between insulin resistance and cancer.
Collapse
Affiliation(s)
- Christina M. Vidal
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Jackelyn A. Alva-Ornelas
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Nancy Zhuo Chen
- Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Duarte, Duarte, CA 91010, USA; (N.Z.C.); (N.B.); (R.N.)
| | - Parijat Senapati
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Jerneja Tomsic
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Vanessa Myriam Robles
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Cristal Resto
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Nancy Sanchez
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Angelica Sanchez
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Terry Hyslop
- Sidney Kimmel Cancer Center, Philadelphia, PA 19107, USA;
| | - Nour Emwas
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Dana Aljaber
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Nick Bachelder
- Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Duarte, Duarte, CA 91010, USA; (N.Z.C.); (N.B.); (R.N.)
| | - Ernest Martinez
- Department of Biochemistry, University of California at Riverside, Riverside, CA 92521, USA;
| | - David Ann
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Veronica Jones
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Robert A. Winn
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Lucio Miele
- School of Medicine, Louisiana State University, New Orleans, LA 70112, USA; (L.M.); (A.C.O.)
| | - Augusto C. Ochoa
- School of Medicine, Louisiana State University, New Orleans, LA 70112, USA; (L.M.); (A.C.O.)
| | - Eric C. Dietze
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| | - Rama Natarajan
- Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Duarte, Duarte, CA 91010, USA; (N.Z.C.); (N.B.); (R.N.)
| | - Dustin Schones
- Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Duarte, Duarte, CA 91010, USA; (N.Z.C.); (N.B.); (R.N.)
| | - Victoria L. Seewaldt
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (C.M.V.); (J.A.A.-O.); (P.S.); (J.T.); (V.M.R.); (C.R.); (N.S.); (A.S.); (N.E.); (D.A.); (D.A.); (V.J.); (E.C.D.)
| |
Collapse
|
4
|
Zhao T, Fan J, Abu-Zaid A, Burley SK, Zheng XS. Nuclear mTOR Signaling Orchestrates Transcriptional Programs Underlying Cellular Growth and Metabolism. Cells 2024; 13:781. [PMID: 38727317 PMCID: PMC11083943 DOI: 10.3390/cells13090781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/26/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024] Open
Abstract
mTOR is a central regulator of cell growth and metabolism in response to mitogenic and nutrient signals. Notably, mTOR is not only found in the cytoplasm but also in the nucleus. This review highlights direct involvement of nuclear mTOR in regulating transcription factors, orchestrating epigenetic modifications, and facilitating chromatin remodeling. These effects intricately modulate gene expression programs associated with growth and metabolic processes. Furthermore, the review underscores the importance of nuclear mTOR in mediating the interplay between metabolism and epigenetic modifications. By integrating its functions in nutrient signaling and gene expression related to growth and metabolism, nuclear mTOR emerges as a central hub governing cellular homeostasis, malignant transformation, and cancer progression. Better understanding of nuclear mTOR signaling has the potential to lead to novel therapies against cancer and other growth-related diseases.
Collapse
Affiliation(s)
- Tinghan Zhao
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Jialin Fan
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Ahmed Abu-Zaid
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Stephen K. Burley
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- RCSB Protein Data Bank and Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, 174 Frelinghuysen Road, Piscataway, NJ 08854, USA
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 174 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - X.F. Steven Zheng
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
5
|
Colak DK, Coskun Yazici ZM, Bolkent S. Chronic administration of delta9-tetrahydrocannabinol protects hyperinsulinemic gastric tissue in rats. Cell Biochem Funct 2023; 41:1543-1551. [PMID: 38032085 DOI: 10.1002/cbf.3894] [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: 07/19/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023]
Abstract
Hyperinsulinemia (HI) can result from some reasons such as an increase in basal/fasting circulating insulin and/or potentiation of postprandial insulin production. Diabetes mellitus (DM) is indirectly related to HI since it both causes and results from insulin resistance. Understanding the causes of HI and treating this is crucial for preventing DM. Previous research has shown that delta9-tetrahydrocannabinol (THC) has medicinal benefits. In light of this, the relationship between THC and oxidative stress, DNA repair mechanism, apoptosis, and its regulatory impact on appetite hormones in the gastric tissue of hyperinsulinemic rats has been investigated for the first time. Male rats (Spraque-Dawley, total = 32) were used, and they were randomly divided into the following groups (n = 8 in each group): control (CTRL), HI, THC administered control (THC, 1.5 mg/kg/day, during 4 weeks), and THC administered HI (HI + THC) groups. The number of poly (ADP-ribose) polymerase-1 and proliferating cell nuclear antigen (PCNA) and caspase-3 immunopositive cells in the HI group was significantly reduced compared to the CTRL group. The number of PCNA and caspase-9 immunopositive cells was significantly increased in the HI + THC group compared to the HI group. Obestatin immunopositive cell numbers in the HI + THC group were higher than in the HI and CTRL groups. The results show that THC administration may affect the regulation of appetite hormones and regeneration in the fundus of rats with HI. Glutathione (GSH) levels were higher in the HI + THC group than in the HI group. Both immunohistochemical and biochemical analyses revealed that THC promotes regeneration and regulates appetite hormones in hyperinsulinemic gastric tissues.
Collapse
Affiliation(s)
- Dilara Kamer Colak
- Department of Medical Biology, Faculty of Cerrahpaşa Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Zeynep Mine Coskun Yazici
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Demiroglu Bilim University, Istanbul, Turkey
| | - Sema Bolkent
- Department of Medical Biology, Faculty of Cerrahpaşa Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| |
Collapse
|
6
|
Ahmed SBM, Radwan N, Amer S, Saheb Sharif-Askari N, Mahdami A, Samara KA, Halwani R, Jelinek HF. Assessing the Link between Diabetic Metabolic Dysregulation and Breast Cancer Progression. Int J Mol Sci 2023; 24:11816. [PMID: 37511575 PMCID: PMC10380477 DOI: 10.3390/ijms241411816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Diabetes mellitus is a burdensome disease that affects various cellular functions through altered glucose metabolism. Several reports have linked diabetes to cancer development; however, the exact molecular mechanism of how diabetes-related traits contribute to cancer progression is not fully understood. The current study aimed to explore the molecular mechanism underlying the potential effect of hyperglycemia combined with hyperinsulinemia on the progression of breast cancer cells. To this end, gene dysregulation induced by the exposure of MCF7 breast cancer cells to hyperglycemia (HG), or a combination of hyperglycemia and hyperinsulinemia (HGI), was analyzed using a microarray gene expression assay. Hyperglycemia combined with hyperinsulinemia induced differential expression of 45 genes (greater than or equal to two-fold), which were not shared by other treatments. On the other hand, in silico analysis performed using a publicly available dataset (GEO: GSE150586) revealed differential upregulation of 15 genes in the breast tumor tissues of diabetic patients with breast cancer when compared with breast cancer patients with no diabetes. SLC26A11, ALDH1A3, MED20, PABPC4 and SCP2 were among the top upregulated genes in both microarray data and the in silico analysis. In conclusion, hyperglycemia combined with hyperinsulinemia caused a likely unique signature that contributes to acquiring more carcinogenic traits. Indeed, these findings might potentially add emphasis on how monitoring diabetes-related metabolic alteration as an adjunct to diabetes therapy is important in improving breast cancer outcomes. However, further detailed studies are required to decipher the role of the highlighted genes, in this study, in the pathogenesis of breast cancer in patients with a different glycemic index.
Collapse
Affiliation(s)
- Samrein B M Ahmed
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Health, Wellbeing and Life Sciences, Department of Biosciences and Chemistry, Sheffield Hallam University, Sheffield S1 1WB, UK
| | - Nada Radwan
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Sara Amer
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Narjes Saheb Sharif-Askari
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Amena Mahdami
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Kamel A Samara
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Rabih Halwani
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Herbert F Jelinek
- Department of Biomedical Engineering and Health Engineering Innovation Center, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| |
Collapse
|
7
|
Felipe Souza E Silva L, Siena Dos Santos A, Mayumi Yuzawa J, Luiz de Barros Torresi J, Ziroldo A, Rosado Rosenstock T. SIRTUINS MODULATORS COUNTERACT MITOCHONDRIAL DYSFUNCTION IN CELLULAR MODELS OF HYPOXIA: RELEVANCE TO SCHIZOPHRENIA. Neuroscience 2023:S0306-4522(23)00200-2. [PMID: 37169164 DOI: 10.1016/j.neuroscience.2023.04.027] [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: 10/24/2022] [Revised: 04/16/2023] [Accepted: 04/27/2023] [Indexed: 05/13/2023]
Abstract
Schizophrenia (SZ) is a neurodevelopmental-associated disorder strongly related to environmental factors, such as hypoxia. Because there is no cure for SZ or any pharmacological approach that could revert hypoxia-induced cellular damages, we evaluated whether modulators of sirtuins could abrogate hypoxia-induced mitochondrial deregulation as a neuroprotective strategy. Firstly, astrocytes from control (Wistar) and Spontaneously Hypertensive Rats (SHR), a model of both SZ and neonatal hypoxia, were submitted to chemical hypoxia. Then, cells were exposed to different concentrations of Nicotinamide (NAM), Resveratrol (Resv), and Sirtinol (Sir) for 48hrs. Our data indicate that sirtuins modulation reduces cell death increasing the acetylation of histone 3. This outcome is related to the rescue of loss of mitochondrial membrane potential, changes in mitochondrial calcium buffering capacity, decreased O2-• levels and increased expression of metabolic regulators (Nrf-1 and Nfe2l2) and mitochondrial content. Such findings are relevant not only for hypoxia-associated conditions, named pre-eclampsia but also for SZ since prenatal hypoxia is a relevant environmental factor related to this burdensome neuropsychiatric disorder.
Collapse
Affiliation(s)
- Luiz Felipe Souza E Silva
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Amanda Siena Dos Santos
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Jessica Mayumi Yuzawa
- Department of Physiological Science, Santa Casa de São Paulo School of Medical Science, São Paulo, Brazil
| | | | - Alan Ziroldo
- Department of Physiological Science, Santa Casa de São Paulo School of Medical Science, São Paulo, Brazil
| | - Tatiana Rosado Rosenstock
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil; Dept. of Bioscience, In-vitro Neuroscience, Sygnature Discovery, Nottingham, United Kingdom.
| |
Collapse
|
8
|
Lin X, Yang Q, Zheng D, Tian H, Chen L, Wu J, Ji Z, Chen Y, Li Z. Scientometric analysis of lipid metabolism in breast neoplasm: 2012-2021. Front Physiol 2023; 14:1042603. [PMID: 37179822 PMCID: PMC10168182 DOI: 10.3389/fphys.2023.1042603] [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: 10/04/2022] [Accepted: 04/10/2023] [Indexed: 05/15/2023] Open
Abstract
Introduction: In recent years, more and more studies have proved that lipid metabolism plays an essential role in breast cancer's proliferation and metastasisand also has a specific significance in predicting survival. Methods: This paper collected data from 725 publications related to lipid metabolism in breast neoplasm from 2012 to 2021 through the Web of Science Core Collection database. Bibliometrix, VOSviewer, and CiteSpace were used for the scientometrics analysis of countries, institutions, journals, authors, keywords, etc. Results: The number of documents published showed an increasing trend, with an average annual growth rate of 14.49%. The United States was the most productive country (n = 223, 30.76%). The journals with the largest number of publications are mostly from developed countries. Except for the retrieved topics, "lipid metabolism" (n = 272) and "breast cancer" (n = 175), the keywords that appeared most frequently were "expression" (n = 151), "fatty-acid synthase" (n = 78), "growth" (n = 72), "metabolism" (n = 67) and "cells" (n = 66). Discussion: These findings and summaries help reveal the current research status and clarify the hot spots in this field.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yexi Chen
- Department of Thyroid, Breast and Hernia Surgery, General Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zhiyang Li
- Department of Thyroid, Breast and Hernia Surgery, General Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
9
|
ACAT1-mediated METTL3 acetylation inhibits cell migration and invasion in triple negative breast cancer. Genes Immun 2023; 24:99-107. [PMID: 36890220 DOI: 10.1038/s41435-023-00202-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 03/10/2023]
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous and aggressive disease with poor prognosis. Acetylation modifications affect a great number of biological processes of malignant tumors. The current study aims at revealing the role of acetylation-related mechanism in TNBC progression. Methyltransferase like-3 (METTL3) was found to be downregulated in TNBC cells via quantitative polymerase chain reaction (qPCR) and western blot analyses. Co-Immunoprecipitation (Co-IP) and GST pulldown assays revealed the interaction between acetyl-CoA acetyltransferase 1 (ACAT1) and METTL3. Through further immunoprecipitation (IP) assay, we determined that ACAT1 stabilizes METTL3 protein via inhibiting the degradation of ubiquitin-proteasome. Functionally, ACAT1 inhibits TNBC cell migration and invasion. Moreover, nuclear receptor subfamily 2 group F member 6 (NR2F6) regulates ACAT1 expression at transcriptional level. Finally, we demonstrated that NR2F6/ACAT/METTL3 axis suppresses the migration and invasion of TNBC cells via METTL3. In conclusion, NR2F6 transcriptionally activates ACAT1 and promotes the suppressive effects of ACAT1-mediated METTL3 acetylation on TNBC cell migration and invasion.
Collapse
|
10
|
Zhu X, Luo X, Long X, Jiang S, Xie X, Zhang Q, Wang H. CircAGO2 promotes colorectal cancer progression by inhibiting heat shock protein family B (small) member 8 via miR-1-3p/retinoblastoma binding protein 4 axis. Funct Integr Genomics 2023; 23:78. [PMID: 36881338 DOI: 10.1007/s10142-023-00990-9] [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: 11/16/2022] [Revised: 02/08/2023] [Accepted: 02/16/2023] [Indexed: 03/08/2023]
Abstract
This paper was to uncover the mechanism of circular RNA Argonaute 2 (circAGO2) in colorectal cancer (CRC) progression. The expression of circAGO2 was detected in CRC cells and tissues, and the relationship between clinicopathological features of CRC and circAGO2 level was evaluated. The growth and invasion of CRC cells and subcutaneous xenograft of nude mice were measured to evaluate the effect of circAGO2 on CRC development. Bioinformatics databases were applied to analyze levels of retinoblastoma binding protein 4 (RBBP4) and heat shock protein family B 8 (HSPB8) in cancer tissues. The relevance of circAGO2 and RBBP4 expression and the relationship between RBBP4 and HSPB8 during histone acetylation were assessed. The targeting relationship between miR-1-3p and circAGO2 or RBBP4 was predicted and confirmed. The effects of miR-1-3p and RBBP4 on biological functions of CRC cells were also verified. CircAGO2 was upregulated in CRC. CircAGO2 promoted the growth and invasion of CRC cells. CircAGO2 competitively bound to miR-1-3p and regulated RBBP4 expression, thus inhibiting HSPB8 transcription by promoting histone deacetylation. Silencing circAGO2 enhanced miR-1-3p expression and reduced RBBP4 expression, while suppression of miR-1-3p downgraded levels of miR-1-3p, up-regulated RBBP4, and facilitated cell proliferation and invasion in the presence of silencing circAGO2. RBBP4 silencing decreased RBBP4 expression and reduced proliferation and invasion of cells where circAGO2 and miR-1-3p were silenced. CircAGO2 overexpression decoyed miR-1-3p to increase RBBP4 expression, which inhibited HSPB8 transcription via histone deacetylation in HSPB8 promoter region, promoting proliferation and invasion of CRC cells.
Collapse
Affiliation(s)
- Xijia Zhu
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Guilin Medical University, No. 212 Renmin Road, Lingui District, Guilin, Guangxi, 541100, People's Republic of China
| | - Xishun Luo
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Guilin Medical University, No. 212 Renmin Road, Lingui District, Guilin, Guangxi, 541100, People's Republic of China
| | - Xiangkai Long
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Guilin Medical University, No. 212 Renmin Road, Lingui District, Guilin, Guangxi, 541100, People's Republic of China
| | - Shiyu Jiang
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Guilin Medical University, No. 212 Renmin Road, Lingui District, Guilin, Guangxi, 541100, People's Republic of China
| | - Xinyang Xie
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Guilin Medical University, No. 212 Renmin Road, Lingui District, Guilin, Guangxi, 541100, People's Republic of China
| | - Qiqi Zhang
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Guilin Medical University, No. 212 Renmin Road, Lingui District, Guilin, Guangxi, 541100, People's Republic of China
| | - Haipeng Wang
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Guilin Medical University, No. 212 Renmin Road, Lingui District, Guilin, Guangxi, 541100, People's Republic of China.
| |
Collapse
|
11
|
Abstract
Few metabolites can claim a more central and versatile role in cell metabolism than acetyl coenzyme A (acetyl-CoA). Acetyl-CoA is produced during nutrient catabolism to fuel the tricarboxylic acid cycle and is the essential building block for fatty acid and isoprenoid biosynthesis. It also functions as a signalling metabolite as the substrate for lysine acetylation reactions, enabling the modulation of protein functions in response to acetyl-CoA availability. Recent years have seen exciting advances in our understanding of acetyl-CoA metabolism in normal physiology and in cancer, buoyed by new mouse models, in vivo stable-isotope tracing approaches and improved methods for measuring acetyl-CoA, including in specific subcellular compartments. Efforts to target acetyl-CoA metabolic enzymes are also advancing, with one therapeutic agent targeting acetyl-CoA synthesis receiving approval from the US Food and Drug Administration. In this Review, we give an overview of the regulation and cancer relevance of major metabolic pathways in which acetyl-CoA participates. We further discuss recent advances in understanding acetyl-CoA metabolism in normal tissues and tumours and the potential for targeting these pathways therapeutically. We conclude with a commentary on emerging nodes of acetyl-CoA metabolism that may impact cancer biology.
Collapse
Affiliation(s)
- David A Guertin
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA, USA.
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Su YH, Wu YZ, Ann DK, Chen JLY, Kuo CY. Obesity promotes radioresistance through SERPINE1-mediated aggressiveness and DNA repair of triple-negative breast cancer. Cell Death Dis 2023; 14:53. [PMID: 36681663 PMCID: PMC9867751 DOI: 10.1038/s41419-023-05576-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/08/2023] [Accepted: 01/09/2023] [Indexed: 01/22/2023]
Abstract
Obesity is a risk factor in various types of cancer, including breast cancer. The disturbance of adipose tissue in obesity highly correlates with cancer progression and resistance to standard treatments such as chemo- and radio-therapies. In this study, in a syngeneic mouse model of triple-negative breast cancer (TNBC), diet-induced obesity (DIO) not only promoted tumor growth, but also reduced tumor response to radiotherapy. Serpine1 (Pai-1) was elevated in the circulation of obese mice and was enriched within tumor microenvironment. In vitro co-culture of human white adipocytes-conditioned medium (hAd-CM) with TNBC cells potentiated the aggressive phenotypes and radioresistance of TNBC cells. Moreover, inhibition of both cancer cell autonomous and non-autonomous SERPINE1 by either genetic or pharmacological strategy markedly dampened the aggressive phenotypes and radioresistance of TNBC cells. Mechanistically, we uncovered a previously unrecognized role of SERPINE1 in DNA damage response. Ionizing radiation-induced DNA double-strand breaks (DSBs) increased the expression of SERPINE1 in cancer cells in an ATM/ATR-dependent manner, and promoted nuclear localization of SERPINE1 to facilitate DSB repair. By analyzing public clinical datasets, higher SERPINE1 expression in TNBC correlated with patients' BMI as well as poor outcomes. Elevated SERPINE1 expression and nuclear localization were also observed in radioresistant breast cancer cells. Collectively, we reveal a link between obesity and radioresistance in TNBC and identify SERPINE1 to be a crucial factor mediating obesity-associated tumor radioresistance.
Collapse
Affiliation(s)
- Yong-Han Su
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Zhen Wu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - David K Ann
- Department of Diabetes Complications & Metabolism, City of Hope, Duarte, CA, USA
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA, USA
| | - Jenny Ling-Yu Chen
- Department of Radiology, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Radiation Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Ching-Ying Kuo
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
13
|
Romanos-Nanclares A, Tabung FK, Willett WC, Rosner B, Holmes MD, Chen WY, Tamimi RM, Eliassen AH. Insulinemic potential of diet and risk of total and subtypes of breast cancer among US females. Am J Clin Nutr 2022; 116:1530-1539. [PMID: 36178066 PMCID: PMC9761760 DOI: 10.1093/ajcn/nqac284] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/15/2022] [Accepted: 09/28/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Insulin resistance and hyperinsulinemia play important roles in the progression of multiple chronic disease and conditions. Diet modulates insulin response; however, evidence is limited regarding whether diets with higher insulinemic potential increase the risk of invasive breast cancer. OBJECTIVES We aimed to prospectively evaluate the association between a food-based empirical dietary index for hyperinsulinemia (EDIH) and the incidence of invasive breast cancer. METHODS We prospectively followed 76,686 women from the Nurses' Health Study (NHS; 1984-2016) and 93,287 women from the Nurses' Health Study II (NHSII; 1991-2017). Diet was assessed by food-frequency questionnaires every 4 y. The insulinemic potential of diet was evaluated using the previously established EDIH based on circulating C-peptide concentrations. Higher scores indicate higher insulinemic potential of the diet. Covariates included reproductive, hormonal, and anthropometric factors (height and BMI at age 18 y); race; socioeconomic status; total alcohol intake; total caloric intake; and physical activity. RESULTS During 4,216,106 person-years of follow-up, we documented 10,602 breast cancer cases (6689 NHS, 3913 NHSII). In the pooled multivariable-adjusted analyses, women in the highest, compared with the lowest, EDIH quintile (Q) were at higher breast cancer risk (HRQ5 vs. Q1 = 1.15; 95% CI: 1.07, 1.24; P-trend < 0.01). Although heterogeneity by estrogen receptor (ER) status was nonsignificant, the strongest association between EDIH and breast cancer was observed for ER-negative tumors (HRQ5 vs. Q1 = 1.21; 95% CI: 1.00, 1.46; P-trend = 0.02). Among tumor molecular subtypes, the strongest associations were observed for human epidermal growth factor receptor 2 (HER2)-enriched tumors (HRQ5 vs. Q1 = 1.62; 95% CI: 1.01, 2.61; P-trend = 0.02). CONCLUSIONS A dietary pattern contributing to hyperinsulinemia and insulin resistance was associated with greater breast cancer risk, especially ER-negative and HER2-enriched tumors. Our findings suggest that dietary modifications to reduce insulinemic potential may reduce the risk of breast cancer.
Collapse
Affiliation(s)
- Andrea Romanos-Nanclares
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Fred K Tabung
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center—Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
| | - Walter C Willett
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Bernard Rosner
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Michelle D Holmes
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Wendy Y Chen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rulla M Tamimi
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - A Heather Eliassen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
14
|
Xing J, Chen C. Hyperinsulinemia: beneficial or harmful or both on glucose homeostasis. Am J Physiol Endocrinol Metab 2022; 323:E2-E7. [PMID: 35635329 DOI: 10.1152/ajpendo.00441.2021] [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] [Indexed: 11/22/2022]
Abstract
Insulin, a principal anabolic hormone produced by pancreatic β-cells, has a primary function of storage of nutrients following excessive energy intake. Pre- or early type 2 diabetes stages present hyperinsulinemia (β-cell dysfunction) and insulin resistance. Initiation of hyperinsulinemia is triggered by a loss of first-phase glucose-stimulated insulin secretion with altered membrane ion channel distribution. More factors, including insulin resistance and excessive proliferation of β-cells, deteriorate the hyperinsulinemia, whereas the hyperinsulinemia contributes to further development of insulin resistance and type 2 diabetes; to develop eventually late-stage diabetes with absolute insulin deficiency. In this mini-review, the major focus was put on the causes and pathophysiology of hyperinsulinemia, and the metabolic consequences and current treatment of hyperinsulinemia were discussed. The data used in this narrative review were collected mainly from relevant discoveries in the past 3 years.
Collapse
Affiliation(s)
- JingJing Xing
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
15
|
Scordamaglia D, Cirillo F, Talia M, Santolla MF, Rigiracciolo DC, Muglia L, Zicarelli A, De Rosis S, Giordano F, Miglietta AM, De Francesco EM, Vella V, Belfiore A, Lappano R, Maggiolini M. Metformin counteracts stimulatory effects induced by insulin in primary breast cancer cells. J Transl Med 2022; 20:263. [PMID: 35672854 PMCID: PMC9172136 DOI: 10.1186/s12967-022-03463-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background Metabolic disorders are associated with increased incidence, aggressive phenotype and poor outcome of breast cancer (BC) patients. For instance, hyperinsulinemia is an independent risk factor for BC and the insulin/insulin receptor (IR) axis is involved in BC growth and metastasis. Of note, the anti-diabetic metformin may be considered in comprehensive therapeutic approaches in BC on the basis of its antiproliferative effects obtained in diverse pre-clinical and clinical studies. Methods Bioinformatics analysis were performed using the information provided by The Invasive Breast Cancer Cohort of The Cancer Genome Atlas (TCGA) project. The naturally immortalized BC cell line, named BCAHC-1, as well as cancer-associated fibroblasts (CAFs) derived from BC patients were used as model systems. In order to identify further mechanisms that characterize the anticancer action of metformin in BC, we performed gene expression and promoter studies as well as western blotting experiments. Moreover, cell cycle analysis, colony and spheroid formation, actin cytoskeleton reorganization, cell migration and matrigel drops evasion assays were carried out to provide novel insights on the anticancer properties of metformin. Results We first assessed that elevated expression and activation of IR correlate with a worse prognostic outcome in estrogen receptor (ER)-positive BC. Thereafter, we established that metformin inhibits the insulin/IR-mediated activation of transduction pathways, gene changes and proliferative responses in BCAHC-1 cells. Then, we found that metformin interferes with the insulin-induced expression of the metastatic gene CXC chemokine receptor 4 (CXCR4), which we found to be associated with poor disease-free survival in BC patients exhibiting high levels of IR. Next, we ascertained that metformin prevents a motile phenotype of BCAHC-1 cells triggered by the paracrine liaison between tumor cells and CAFs upon insulin activated CXCL12/CXCR4 axis. Conclusions Our findings provide novel mechanistic insights regarding the anti-proliferative and anti-migratory effects of metformin in both BC cells and important components of the tumor microenvironment like CAFs. Further investigations are warranted to corroborate the anticancer action of metformin on the tumor mass toward the assessment of more comprehensive strategies halting BC progression, in particular in patients exhibiting metabolic disorders and altered insulin/IR functions. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03463-y.
Collapse
|
16
|
Samanta S, Mahata R, Santra MK. The Cross-Talk between Epigenetic Gene Regulation and Signaling Pathways Regulates Cancer Pathogenesis. Subcell Biochem 2022; 100:427-472. [PMID: 36301502 DOI: 10.1007/978-3-031-07634-3_13] [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] [Indexed: 06/16/2023]
Abstract
Cancer begins due to uncontrolled cell division. Cancer cells are insensitive to the signals that control normal cell proliferation. This uncontrolled cell division is due to the accumulation of abnormalities in different factors associated with the cell division, including different cyclins, cell cycle checkpoint inhibitors, and cellular signaling. Cellular signaling pathways are aberrantly activated in cancer mainly due to epigenetic regulation and post-translational regulation. In this chapter, the role of epigenetic regulation in aberrant activation of PI3K/AKT, Ras, Wnt, Hedgehog, Notch, JAK/STAT, and mTOR signaling pathways in cancer progression is discussed. The role of epigenetic regulators in controlling the upstream regulatory proteins and downstream effector proteins responsible for abnormal cellular signaling-mediated cancer progression is covered in this chapter. Similarly, the role of signaling pathways in controlling epigenetic gene regulation-mediated cancer progression is also discussed. We have tried to ascertain the current status of potential epigenetic drugs targeting several epigenetic regulators to prevent different cancers.
Collapse
Affiliation(s)
- Snigdha Samanta
- Molecular Oncology Laboratory, National Centre for Cell Science, NCCS Complex, S. P. Pune University Campus, Ganeshkhind Road, Pune, Maharashtra, India
- Department of Biotechnology, Savitribai Phule Pune University, Pune, Maharashtra, India
| | - Rumpa Mahata
- Molecular Oncology Laboratory, National Centre for Cell Science, NCCS Complex, S. P. Pune University Campus, Ganeshkhind Road, Pune, Maharashtra, India
- Department of Biotechnology, Savitribai Phule Pune University, Pune, Maharashtra, India
| | - Manas Kumar Santra
- Molecular Oncology Laboratory, National Centre for Cell Science, NCCS Complex, S. P. Pune University Campus, Ganeshkhind Road, Pune, Maharashtra, India.
| |
Collapse
|
17
|
Abstract
Lysine acetylation is the second most well-studied post-translational modification after phosphorylation. While phosphorylation regulates signaling cascades, one of the most significant roles of acetylation is regulation of chromatin structure. Acetyl-coenzyme A (acetyl-CoA) serves as the acetyl group donor for acetylation reactions mediated by lysine acetyltransferases (KATs). On the other hand, NAD+ serves as the cofactor for lysine deacetylases (KDACs). Both acetyl-CoA and NAD+ are metabolites integral to energy metabolism, and therefore, their metabolic flux can regulate the activity of KATs and KDACs impacting the epigenome. In this chapter, we review our current understanding of how metabolic pathways regulate lysine acetylation in normal and cancer cells.
Collapse
Affiliation(s)
- Siddharth Singh
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, Karnataka, India
| | - Parijat Senapati
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, Karnataka, India
- Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Tapas K Kundu
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, Karnataka, India.
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India.
| |
Collapse
|
18
|
Benedetti R, Benincasa G, Glass K, Chianese U, Vietri MT, Congi R, Altucci L, Napoli C. Effects of novel SGLT2 inhibitors on cancer incidence in hyperglycemic patients: a meta-analysis of randomized clinical trials. Pharmacol Res 2021; 175:106039. [PMID: 34929299 DOI: 10.1016/j.phrs.2021.106039] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 12/10/2021] [Accepted: 12/15/2021] [Indexed: 02/06/2023]
Abstract
Epidemiological evidence shows that diabetic patients have an increased cancer risk and a higher mortality rate. Glucose could play a central role in metabolism and growth of many tumor types, and this possible mechanism is supported by the high rate of glucose demand and uptake in cancer. Thus, growing evidence suggests that hyperglycemia contributes to cancer progression but also to its onset. Many mechanisms underlying this association have been hypothesized, such as insulin resistance, hyperinsulinemia, and increased inflammatory processes. Inflammation is a common pathophysiological feature in both diabetic and oncological patients, and inflammation linked to high glucose levels sensitizes microenvironment to tumorigenesis, promoting the development of malignant lesions by altering and sustaining a pathological condition in tissues. Glycemic control is the first goal of antidiabetic therapy, and glucose level reduction has also been associated with favorable outcomes in cancer. Here, we describe key events in carcinogenesis focusing on hyperglycemia as supporter in tumor progression and in particular, related to the role of a specific hypoglycemic drug class, sodium-glucose linked transporters (SGLTs). We also discuss the use of SGLT2 inhibitors as a novel potential cancer therapy. Our meta-analysis showed that SGLT-2 inhibitors were significantly associated with an overall reduced risk of cancer as compared to placebo (RR = 0.35, CI 0.33-0.37, P = 0. 00) with a particular effectiveness for dapaglifozin and ertuglifozin (RR = 0. 06, CI 0. 06-0. 07 and RR = 0. 22, CI 0. 18-0. 26, respectively). Network Medicine approaches may advance the possible repurposing of these drugs in patients with concomitant diabetes and cancer.
Collapse
Affiliation(s)
- Rosaria Benedetti
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Naples, Italy
| | - Giuditta Benincasa
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Pz. Miraglia 2, 80138 Naples, Italy.
| | - Kimberly Glass
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Ugo Chianese
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Naples, Italy
| | - Maria Teresa Vietri
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Naples, Italy
| | - Raffaella Congi
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Naples, Italy
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Naples, Italy; Biogem Institute of Molecular and Genetic Biology, 83031 Ariano Irpino, Italy.
| | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Pz. Miraglia 2, 80138 Naples, Italy; Clinical Department of Internal Medicine and Specialistics, Division of Clinical Immunology, Transfusion Medicine and Transplant Immunology, AOU University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Naples, Italy.
| |
Collapse
|
19
|
Jemal M, Molla TS, Asmamaw Dejenie T. Ketogenic Diets and their Therapeutic Potential on Breast Cancer: A Systemic Review. Cancer Manag Res 2021; 13:9147-9155. [PMID: 34934359 PMCID: PMC8684375 DOI: 10.2147/cmar.s339970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 11/27/2021] [Indexed: 12/05/2022] Open
Abstract
Breast cancer remains a major cause of morbidity and mortality in women, and there is still a lack of complementary approaches to significantly improve the efficacy of standard therapies. For many kinds of cancers, the usual standard care is the combination of surgery, radiation, and chemotherapy. However, this standard therapy is not effective alone. Therefore, new approaches that increase therapeutic effectiveness are urgently needed. The ketogenic diet is a novel therapeutic approach for certain types of cancers, as indicated by several preclinical and clinical evidences. The ketogenic diet, which consists of a high-fat, low-carbohydrate diet with adequate protein, appears to sensitize most cancers to standard therapy by utilizing the reprogrammed metabolism of cancer cells, making it a promising candidate for adjuvant cancer treatment. The majority of preclinical and clinical studies argue that the use of a ketogenic diet in combination with standard therapies is based on its potential to improve the antitumor effects of conventional chemotherapy, its overall good safety and tolerability, and quality of life improvement. According to new evidence, a ketogenic diet lowers the level of glucose and insulin in the blood, which are necessary for tumor growth. Thus, the ketogenic diet has emerged as a potential treatment option for a variety of cancers, including breast cancer. Besides, implementation of a Ketogenic diet in the clinic could improve progression-free and overall survival for patients with breast cancer. This review summarizes the composition and metabolism of ketogenic diets and their potential mechanisms in breast carcinogenesis in addition to their therapeutic potential on breast cancer.
Collapse
Affiliation(s)
- Mohammed Jemal
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Amhara, Ethiopia
| | - Tewodros Shibabaw Molla
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Amhara, Ethiopia
| | - Tadesse Asmamaw Dejenie
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Amhara, Ethiopia
| |
Collapse
|
20
|
Bompada P, Goncalves I, Wu C, Gao R, Sun J, Mir BA, Luan C, Renström E, Groop L, Weng J, Hansson O, Edsfeldt A, De Marinis Y. Epigenome-Wide Histone Acetylation Changes in Peripheral Blood Mononuclear Cells in Patients with Type 2 Diabetes and Atherosclerotic Disease. Biomedicines 2021; 9:biomedicines9121908. [PMID: 34944721 PMCID: PMC8698994 DOI: 10.3390/biomedicines9121908] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/30/2021] [Accepted: 12/06/2021] [Indexed: 11/16/2022] Open
Abstract
There is emerging evidence of an association between epigenetic modifications, glycemic control and atherosclerosis risk. In this study, we mapped genome-wide epigenetic changes in patients with type 2 diabetes (T2D) and advanced atherosclerotic disease. We performed chromatin immunoprecipitation sequencing (ChIP-seq) using a histone 3 lysine 9 acetylation (H3K9ac) mark in peripheral blood mononuclear cells from patients with atherosclerosis with T2D (n = 8) or without T2D (ND, n = 10). We mapped epigenome changes and identified 23,394 and 13,133 peaks in ND and T2D individuals, respectively. Out of all the peaks, 753 domains near the transcription start site (TSS) were unique to T2D. We found that T2D in atherosclerosis leads to an H3K9ac increase in 118, and loss in 63 genomic regions. Furthermore, we discovered an association between the genomic locations of significant H3K9ac changes with genetic variants identified in previous T2D GWAS. The transcription factor 7-like 2 (TCF7L2) rs7903146, together with several human leukocyte antigen (HLA) variants, were among the domains with the most dramatic changes of H3K9ac enrichments. Pathway analysis revealed multiple activated pathways involved in immunity, including type 1 diabetes. Our results present novel evidence on the interaction between genetics and epigenetics, as well as epigenetic changes related to immunity in patients with T2D and advanced atherosclerotic disease.
Collapse
Affiliation(s)
- Pradeep Bompada
- Department of Clinical Sciences, Lund University, 20502 Malmö, Sweden; (P.B.); (C.W.); (B.A.M.); (C.L.); (E.R.); (L.G.); (O.H.)
| | - Isabel Goncalves
- Cardiovascular Research-Translational Studies, Institution of Clinical Science Malmö, Lund University, 20502 Malmö, Sweden; (I.G.); (J.S.); (A.E.)
- Department of Cardiology, Skåne University Hospital, 20502 Malmö, Sweden
| | - Chuanyan Wu
- Department of Clinical Sciences, Lund University, 20502 Malmö, Sweden; (P.B.); (C.W.); (B.A.M.); (C.L.); (E.R.); (L.G.); (O.H.)
- School of Control Science and Engineering, Shandong University, Jinan 250061, China
- School of Intelligent Engineering, Shandong Management University, Jinan 250100, China
| | - Rui Gao
- School of Control Science and Engineering, Shandong University, Jinan 250061, China
- Correspondence: (R.G.); (Y.D.M.); Tel.: +86-135-0531-8418 (R.G.); +46-760-384-868 (Y.D.M.)
| | - Jiangming Sun
- Cardiovascular Research-Translational Studies, Institution of Clinical Science Malmö, Lund University, 20502 Malmö, Sweden; (I.G.); (J.S.); (A.E.)
| | - Bilal Ahmad Mir
- Department of Clinical Sciences, Lund University, 20502 Malmö, Sweden; (P.B.); (C.W.); (B.A.M.); (C.L.); (E.R.); (L.G.); (O.H.)
| | - Cheng Luan
- Department of Clinical Sciences, Lund University, 20502 Malmö, Sweden; (P.B.); (C.W.); (B.A.M.); (C.L.); (E.R.); (L.G.); (O.H.)
| | - Erik Renström
- Department of Clinical Sciences, Lund University, 20502 Malmö, Sweden; (P.B.); (C.W.); (B.A.M.); (C.L.); (E.R.); (L.G.); (O.H.)
| | - Leif Groop
- Department of Clinical Sciences, Lund University, 20502 Malmö, Sweden; (P.B.); (C.W.); (B.A.M.); (C.L.); (E.R.); (L.G.); (O.H.)
- Finnish Institute for Molecular Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Jianping Weng
- Clinical Research Hospital, Chinese Academy of Sciences, Hefei 230001, China;
- Department of Endocrinology and Metabolism, Division of Life Sciences of Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Ola Hansson
- Department of Clinical Sciences, Lund University, 20502 Malmö, Sweden; (P.B.); (C.W.); (B.A.M.); (C.L.); (E.R.); (L.G.); (O.H.)
- Institute for Molecular Medicine Finland (FIMM), Helsinki University, 00290 Helsinki, Finland
| | - Andreas Edsfeldt
- Cardiovascular Research-Translational Studies, Institution of Clinical Science Malmö, Lund University, 20502 Malmö, Sweden; (I.G.); (J.S.); (A.E.)
- Department of Cardiology, Skåne University Hospital, 20502 Malmö, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, 20502 Malmö, Sweden
| | - Yang De Marinis
- Department of Clinical Sciences, Lund University, 20502 Malmö, Sweden; (P.B.); (C.W.); (B.A.M.); (C.L.); (E.R.); (L.G.); (O.H.)
- School of Control Science and Engineering, Shandong University, Jinan 250061, China
- Clinical Research Hospital, Chinese Academy of Sciences, Hefei 230001, China;
- Department of Endocrinology and Metabolism, Division of Life Sciences of Medicine, University of Science and Technology of China, Hefei 230001, China
- Correspondence: (R.G.); (Y.D.M.); Tel.: +86-135-0531-8418 (R.G.); +46-760-384-868 (Y.D.M.)
| |
Collapse
|
21
|
Simeon J, Thrush J, Bailey TA. Angiopoietin-like protein 4 is a chromatin-bound protein that enhances mammosphere formation in vitro and experimental triple-negative breast cancer brain and liver metastases in vivo. J Carcinog 2021; 20:8. [PMID: 34447288 PMCID: PMC8356708 DOI: 10.4103/jcar.jcar_20_20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/30/2020] [Accepted: 01/06/2021] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION: Metastatic progression in triple-negative breast cancer (TNBC) patients occurs primarily because of nuclear reprogramming that includes chromatin remodeling and epigenetic modifications. The existing and most successful chemotherapies available for metastatic TNBC target nuclear proteins or damage DNA. The objectives here are to investigate an undescribed role for the molecular biology of nuclear angiopoietin-like protein 4 (ANGPTL4) and to characterize the effect of ectopic overexpression of ANGPTL4 in the metastatic biology of TNBC. MATERIALS AND METHODS: Lentiviral-mediated transduction was used to overexpress ANGPTL4 in the TNBC cell line MD Anderson–metastatic breast cancer 231. The overexpression of ANGPTL4 was confirmed by western blot and ELISA. Subcellular fractionation, western blot, and immunofluorescence microscopy were used to characterize the intracellular localization of ANGPTL4. Mammosphere culture and the anchorage-independent growth assay analyzed the metastatic potential of the cell line. Xenograft assays assessed the effect of ANGPTL4 overexpression on TNBC metastases in vivo. RESULTS: The ANGPTL4 overexpressing cell line formed larger mammospheres and anchorage-independent colonies in vitro and developed larger primary tumors, more liver metastases, and brain metastatic outgrowth in vivo in comparison to a cell line that expressed endogenous levels of ANGPTL4. ANGPTL4, aurora kinase A (AURKA), a mitotic kinase, and Tat-interacting protein p60 kDa (Tip60), a lysine acetyltransferase, associated with chromatin in the ANGPTL4 overexpressing cells but not in cells that expressed endogenous levels of ANGPTL4. CONCLUSIONS: The ANGPTL4 overexpressing cell line showed in vitro and in vivo activities that suggest that nuclear ANGPTL4, AURKA, and Tip60 may cooperatively modulate TNBC metastases within chromatin-remodeling complexes or DNA-associated machinery.
Collapse
Affiliation(s)
- Jodi Simeon
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas, USA.,Department of Cell and Molecular Biology Program, University of Arkansas, Fayetteville, Arkansas, USA
| | - Jessica Thrush
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas, USA.,Department of Honors College, University of Arkansas, Fayetteville, Arkansas, USA
| | - Tameka A Bailey
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas, USA.,Department of Cell and Molecular Biology Program, University of Arkansas, Fayetteville, Arkansas, USA
| |
Collapse
|
22
|
Zou Y, Fineberg S, Pearlman A, Feinman RD, Fine EJ. The effect of a ketogenic diet and synergy with rapamycin in a mouse model of breast cancer. PLoS One 2020; 15:e0233662. [PMID: 33270630 PMCID: PMC7714189 DOI: 10.1371/journal.pone.0233662] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 11/06/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The effects of diet in cancer, in general, and breast cancer in particular, are not well understood. Insulin inhibition in ketogenic, high fat diets, modulate downstream signaling molecules and are postulated to have therapeutic benefits. Obesity and diabetes have been associated with higher incidence of breast cancer. Addition of anti-cancer drugs together with diet is also not well studied. METHODS Two diets, one ketogenic, the other standard mouse chow, were tested in a spontaneous breast cancer model in 34 mice. Subgroups of 3-9 mice were assigned, in which the diet were implemented either with or without added rapamycin, an mTOR inhibitor and potential anti-cancer drug. RESULTS Blood glucose and insulin concentrations in mice ingesting the ketogenic diet (KD) were significantly lower, whereas beta hydroxybutyrate (BHB) levels were significantly higher, respectively, than in mice on the standard diet (SD). Growth of primary breast tumors and lung metastases were inhibited, and lifespans were longer in the KD mice compared to mice on the SD (p<0.005). Rapamycin improved survival in both mouse diet groups, but when combined with the KD was more effective than when combined with the SD. CONCLUSIONS The study provides proof of principle that a ketogenic diet a) results in serum insulin reduction and ketosis in a spontaneous breast cancer mouse model; b) can serve as a therapeutic anti-cancer agent; and c) can enhance the effects of rapamycin, an anti-cancer drug, permitting dose reduction for comparable effect. Further, the ketogenic diet in this model produces superior cancer control than standard mouse chow whether with or without added rapamycin.
Collapse
Affiliation(s)
- Yiyu Zou
- Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Susan Fineberg
- Montefiore Medical Center, Bronx, NY, United States of America
| | - Alexander Pearlman
- Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Richard D. Feinman
- SUNY Downstate Health Sciences Center, Brooklyn, NY, United States of America
| | - Eugene J. Fine
- Albert Einstein College of Medicine, Bronx, NY, United States of America
- Montefiore Medical Center, Bronx, NY, United States of America
- * E-mail:
| |
Collapse
|
23
|
Izzo LT, Affronti HC, Wellen KE. The Bidirectional Relationship Between Cancer Epigenetics and Metabolism. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2020; 5:235-257. [PMID: 34109280 PMCID: PMC8186467 DOI: 10.1146/annurev-cancerbio-070820-035832] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Metabolic and epigenetic reprogramming are characteristics of cancer cells that, in many cases, are linked. Oncogenic signaling, diet, and tumor microenvironment each influence the availability of metabolites that are substrates or inhibitors of epigenetic enzymes. Reciprocally, altered expression or activity of chromatin-modifying enzymes can exert direct and indirect effects on cellular metabolism. In this article, we discuss the bidirectional relationship between epigenetics and metabolism in cancer. First, we focus on epigenetic control of metabolism, highlighting evidence that alterations in histone modifications, chromatin remodeling, or the enhancer landscape can drive metabolic features that support growth and proliferation. We then discuss metabolic regulation of chromatin-modifying enzymes and roles in tumor growth and progression. Throughout, we highlight proposed therapeutic and dietary interventions that leverage metabolic-epigenetic cross talk and have the potential to improve cancer therapy.
Collapse
Affiliation(s)
- Luke T Izzo
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Hayley C Affronti
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Kathryn E Wellen
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
24
|
Abstract
Elevated circulating insulin levels are frequently observed in the setting of obesity and early type 2 diabetes, as a result of insensitivity of metabolic tissues to the effects of insulin. Higher levels of circulating insulin have been associated with increased cancer risk and progression in epidemiology studies. Elevated circulating insulin is believed to be a major factor linking obesity, diabetes and cancer. With the development of targeted cancer therapies, insulin signalling has emerged as a mechanism of therapeutic resistance. Although metabolic tissues become insensitive to insulin in the setting of obesity, a number of mechanisms allow cancer cells to maintain their ability to respond to insulin. Significant progress has been made in the past decade in understanding the insulin receptor and its signalling pathways in cancer, and a number of lessons have been learnt from therapeutic failures. These discoveries have led to numerous clinical trials that have aimed to reduce the levels of circulating insulin and to abrogate insulin signalling in cancer cells. With the rising prevalence of obesity and diabetes worldwide, and the realization that hyperinsulinaemia may contribute to therapeutic failures, it is essential to understand how insulin and insulin receptor signalling promote cancer progression.
Collapse
Affiliation(s)
- Emily J Gallagher
- Division of Endocrinology, Diabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Tisch Cancer Institute at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
25
|
Liu JY, Wellen KE. Advances into understanding metabolites as signaling molecules in cancer progression. Curr Opin Cell Biol 2020; 63:144-153. [PMID: 32097832 DOI: 10.1016/j.ceb.2020.01.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/22/2020] [Accepted: 01/22/2020] [Indexed: 12/13/2022]
Abstract
Recent years have seen a great expansion in our knowledge of the roles that metabolites play in cellular signaling. Structural data have provided crucial insights into mechanisms through which amino acids are sensed. New nutrient-coupled protein and RNA modifications have been identified and characterized. A growing list of functions has been ascribed to metabolic regulation of modifications such as acetylation, methylation, and glycosylation. A current challenge lies in developing an integrated understanding of the roles that metabolic signaling mechanisms play in physiology and disease, which will inform the design of strategies to target such mechanisms. In this brief article, we review recent advances in metabolic signaling through post-translational modification during cancer progression, to provide a framework for understanding signaling roles of metabolites in the context of cancer biology and illuminate areas for future investigation.
Collapse
Affiliation(s)
- Joyce Y Liu
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, 19104 PA, USA; Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, 19104 PA, USA; Biochemistry & Molecular Biophysics Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, 19104 PA, USA
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, 19104 PA, USA; Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, 19104 PA, USA.
| |
Collapse
|