1
|
Lingampelly SS, Naviaux JC, Heuer LS, Monk JM, Li K, Wang L, Haapanen L, Kelland CA, Van de Water J, Naviaux RK. Metabolic network analysis of pre-ASD newborns and 5-year-old children with autism spectrum disorder. Commun Biol 2024; 7:536. [PMID: 38729981 PMCID: PMC11549098 DOI: 10.1038/s42003-024-06102-y] [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: 10/19/2023] [Accepted: 03/22/2024] [Indexed: 05/12/2024] Open
Abstract
Classical metabolomic and new metabolic network methods were used to study the developmental features of autism spectrum disorder (ASD) in newborns (n = 205) and 5-year-old children (n = 53). Eighty percent of the metabolic impact in ASD was caused by 14 shared biochemical pathways that led to decreased anti-inflammatory and antioxidant defenses, and to increased physiologic stress molecules like lactate, glycerol, cholesterol, and ceramides. CIRCOS plots and a new metabolic network parameter,V ° net, revealed differences in both the kind and degree of network connectivity. Of 50 biochemical pathways and 450 polar and lipid metabolites examined, the developmental regulation of the purine network was most changed. Purine network hub analysis revealed a 17-fold reversal in typically developing children. This purine network reversal did not occur in ASD. These results revealed previously unknown metabolic phenotypes, identified new developmental states of the metabolic correlation network, and underscored the role of mitochondrial functional changes, purine metabolism, and purinergic signaling in autism spectrum disorder.
Collapse
Grants
- UL1 TR001442 NCATS NIH HHS
- 7274 Autism Speaks (Autism Speaks Inc.)
- This work was funded in part by philanthropic gifts to the Naviaux Lab from the UCSD Christini Fund, the Lennox Foundation, the William Wright Family Foundation, Malone Family Foundation, the Brain Foundation, the Westreich Foundation, the Aloe family, the Harb family, Marc Spilo and all the others who contributed to the Aloe family autism research fund, the N of One Autism Research Foundation, the UCSD Mitochondrial Disease Research Fund, the JMS Fund, Linda Clark, Jeanne Conrad, David Cannistraro, the Kirby and Katie Mano Family, Simon and Evelyn Foo, Wing-kun Tam, Gita and Anurag Gupta, the Brent Kaufman Family, and the Daniel and Kelly White Family, and grassroots support from over 2000 individuals from around the world who have each provided gifts in the past year to support Naviaux Lab research. The REDCap software system used in this study was provided by the UCSD Clinical and Translational Research Center and supported by Award Number UL1TR001442 from the National Center for Research Resources. Financial supporters for this study had no role in study design, data collection, analysis, interpretation, writing, or publication of this work.
Collapse
Affiliation(s)
- Sai Sachin Lingampelly
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA
| | - Jane C Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA
- Department of Neuroscience, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA
| | - Luke S Heuer
- The UC Davis MIND Institute, University of California, Davis, Davis, CA, 95616, USA
| | - Jonathan M Monk
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA
| | - Kefeng Li
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA
- Macao Polytechnic University, Macau, China
| | - Lin Wang
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA
| | - Lori Haapanen
- The UC Davis MIND Institute, University of California, Davis, Davis, CA, 95616, USA
| | - Chelsea A Kelland
- The UC Davis MIND Institute, University of California, Davis, Davis, CA, 95616, USA
| | - Judy Van de Water
- The UC Davis MIND Institute, University of California, Davis, Davis, CA, 95616, USA
- Department of Rheumatology and Allergy, School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Robert K Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA.
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA.
- Department of Pediatrics, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA.
- Department of Pathology, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA.
| |
Collapse
|
2
|
Tomar MS, Kumar A, Shrivastava A. Mitochondrial metabolism as a dynamic regulatory hub to malignant transformation and anti-cancer drug resistance. Biochem Biophys Res Commun 2024; 694:149382. [PMID: 38128382 DOI: 10.1016/j.bbrc.2023.149382] [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: 08/17/2023] [Revised: 12/02/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023]
Abstract
Glycolysis is the fundamental cellular process that permits cancer cells to convert energy and grow anaerobically. Recent developments in molecular biology have made it evident that mitochondrial respiration is critical to tumor growth and treatment response. As the principal organelle of cellular energy conversion, mitochondria can rapidly alter cellular metabolic processes, thereby fueling malignancies and contributing to treatment resistance. This review emphasizes the significance of mitochondrial biogenesis, turnover, DNA copy number, and mutations in bioenergetic system regulation. Tumorigenesis requires an intricate cascade of metabolic pathways that includes rewiring of the tricarboxylic acid (TCA) cycle, electron transport chain and oxidative phosphorylation, supply of intermediate metabolites of the TCA cycle through amino acids, and the interaction between mitochondria and lipid metabolism. Cancer recurrence or resistance to therapy often results from the cooperation of several cellular defense mechanisms, most of which are connected to mitochondria. Many clinical trials are underway to assess the effectiveness of inhibiting mitochondrial respiration as a potential cancer therapeutic. We aim to summarize innovative strategies and therapeutic targets by conducting a comprehensive review of recent studies on the relationship between mitochondrial metabolism, tumor development and therapeutic resistance.
Collapse
Affiliation(s)
- Manendra Singh Tomar
- Center for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow, 226003, Uttar Pradesh, India
| | - Ashok Kumar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal, 462020, Madhya Pradesh, India
| | - Ashutosh Shrivastava
- Center for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow, 226003, Uttar Pradesh, India.
| |
Collapse
|
3
|
Razi S, Haghparast A, Chodari Khameneh S, Ebrahimi Sadrabadi A, Aziziyan F, Bakhtiyari M, Nabi-Afjadi M, Tarhriz V, Jalili A, Zalpoor H. The role of tumor microenvironment on cancer stem cell fate in solid tumors. Cell Commun Signal 2023; 21:143. [PMID: 37328876 PMCID: PMC10273768 DOI: 10.1186/s12964-023-01129-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/15/2023] [Indexed: 06/18/2023] Open
Abstract
In the last few decades, the role of cancer stem cells in initiating tumors, metastasis, invasion, and resistance to therapies has been recognized as a potential target for tumor therapy. Understanding the mechanisms by which CSCs contribute to cancer progression can help to provide novel therapeutic approaches against solid tumors. In this line, the effects of mechanical forces on CSCs such as epithelial-mesenchymal transition, cellular plasticity, etc., the metabolism pathways of CSCs, players of the tumor microenvironment, and their influence on the regulating of CSCs can lead to cancer progression. This review focused on some of these mechanisms of CSCs, paving the way for a better understanding of their regulatory mechanisms and developing platforms for targeted therapies. While progress has been made in research, more studies will be required in the future to explore more aspects of how CSCs contribute to cancer progression. Video Abstract.
Collapse
Affiliation(s)
- Sara Razi
- Vira Pioneers of Modern Science (VIPOMS), Tehran, Iran
| | | | | | - Amin Ebrahimi Sadrabadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACER, Tehran, Iran
- Cytotech and Bioinformatics Research Group, Tehran, Iran
| | - Fatemeh Aziziyan
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Maryam Bakhtiyari
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Vahideh Tarhriz
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, P.O. Box 5163639888, Tabriz, Iran.
| | - Arsalan Jalili
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACER, Tehran, Iran.
- Parvaz Research Ideas Supporter Institute, Tehran, Iran.
| | - Hamidreza Zalpoor
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
4
|
Shi H, Pan M, Jia E, Lu W, Zhou Y, Sheng Y, Zhao X, Cai L, Ge Q. A comprehensive characterization of cell-free RNA in spent blastocyst medium and quality prediction for blastocyst. Clin Sci (Lond) 2023; 137:129-0. [PMID: 36597876 DOI: 10.1042/cs20220495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 12/16/2022] [Accepted: 01/03/2023] [Indexed: 01/05/2023]
Abstract
The rate of pregnancy can be affected by many factors in assisted reproductive technology (ART), and one of which is the quality of embryos. Therefore, selecting the embryos with high potential is crucial for the outcome. Fifteen spent blastocyst medium (SBM) samples were collected from 14 patients who received in vitro fertilization (IVF) or intracytoplasmic sperm injection (ICSI), seven from high-grade embryos and eight from low-grade embryos. Cell-free RNA (cf-RNA) profile of SBM samples were analyzed by RNA sequencing in the present study. It was found that a large amount of cf-RNA were released into SBM, including protein-coding genes (68.9%) and long noncoding RNAs (lncRNAs) (17.26%). Furthermore, a high correlation was observed between blastocyst genes and SBM genes. And the cf-mRNAs of SBM were highly fragmented, and coding sequence (CDS) and untranslated (UTR) regions were released equally. Two hundred and thirty-two differentially expressed genes were identified in high-grade SBM (hSBM) and low-grade SBM (lSBM), which could be potential biomarker in distinguishing the embryos with different quality as an alternative or supplementary approach for subjective morphology criteria. Hence, cf-RNAs sequencing revealed the characterization of circulating transcriptomes of embryos with different quality. Based on the results, the genes related to blastocyst quality were screened, including the genes closely related to translation, immune-signaling pathway, and amino acid metabolism. Overall, the present study showed the types of SBM cf-RNAs, and the integrated analysis of cf-RNAs profiling with morphology grading displayed its potential in predicting blastocyst quality. The present study provided valuable scientific basis for noninvasive embryo selection in ART by RNA-profiling analysis.
Collapse
Affiliation(s)
- Huajuan Shi
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, People's Republic of China
| | - Min Pan
- School of Medicine, Southeast University, Nanjing 210097, China
| | - Erteng Jia
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, People's Republic of China
| | - Wenxiang Lu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, People's Republic of China
| | - Ying Zhou
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, People's Republic of China
| | - Yuqi Sheng
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, People's Republic of China
| | - Xiangwei Zhao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, People's Republic of China
| | - Lingbo Cai
- Clinical Center of Reproductive Medicine, State Key Laboratory of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Qinyu Ge
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, People's Republic of China
| |
Collapse
|
5
|
Limper CB, Bondah N, Zhu D, Villanueva AN, Chukwukere UK, Huang W, August A. Effective differentiation of double negative thymocytes requires high fidelity replication of mitochondrial DNA in an age dependent manner. Front Immunol 2023; 14:1128626. [PMID: 37020546 PMCID: PMC10067910 DOI: 10.3389/fimmu.2023.1128626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/03/2023] [Indexed: 04/07/2023] Open
Abstract
One of the most proliferative periods for T cells occurs during their development in the thymus. Increased DNA replication can result in increased DNA mutations in the nuclear genome, but also in mitochondrial genomes. A high frequency of mitochondrial DNA mutations can lead to abnormal mitochondrial function and have negative implications on human health. Furthermore, aging is accompanied by an increase in such mutations through oxidative damage and replication errors. Increased mitochondrial DNA mutations cause loss of mitochondrial protein function, and decrease energy production, substrates, and metabolites. Here we have evaluated the effect of increased mitochondrial DNA mutations on T cell development in the thymus. Using mice carrying a mutant mitochondrial DNA polymerase γ (PolG) that causes increased mitochondrial DNA mutations, we show that high fidelity replication of mitochondrial DNA is pivotal for proper T cell development. Reducing the fidelity of mitochondrial DNA replication results in a premature age-dependent reduction in the total number of CD4/CD8 double negative and double positive thymocytes. Analysis of mitochondrial density in thymocyte subpopulations suggests that this may be due to reduced proliferation in specific double negative stages. Taken together, this work suggests that T cell development is regulated by the ability of mitochondria to faithfully replicate their DNA.
Collapse
Affiliation(s)
- Candice B. Limper
- Department of Microbiology and Immunology, Cornell Institute of Host-Microbe Interaction and Disease, Cornell Center for Immunology, Cornell University, Ithaca, NY, United States
| | - Narda Bondah
- Department of Microbiology and Immunology, Cornell Institute of Host-Microbe Interaction and Disease, Cornell Center for Immunology, Cornell University, Ithaca, NY, United States
| | - Daphne Zhu
- Department of Microbiology and Immunology, Cornell Institute of Host-Microbe Interaction and Disease, Cornell Center for Immunology, Cornell University, Ithaca, NY, United States
| | - Alanis N. Villanueva
- Department of Microbiology and Immunology, Cornell Institute of Host-Microbe Interaction and Disease, Cornell Center for Immunology, Cornell University, Ithaca, NY, United States
| | - Uchenna K. Chukwukere
- Department of Microbiology and Immunology, Cornell Institute of Host-Microbe Interaction and Disease, Cornell Center for Immunology, Cornell University, Ithaca, NY, United States
| | - Weishan Huang
- Department of Microbiology and Immunology, Cornell Institute of Host-Microbe Interaction and Disease, Cornell Center for Immunology, Cornell University, Ithaca, NY, United States
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Avery August
- Department of Microbiology and Immunology, Cornell Institute of Host-Microbe Interaction and Disease, Cornell Center for Immunology, Cornell University, Ithaca, NY, United States
- Cornell Center for Health Equity, Cornell University, Ithaca, NY, United States
- *Correspondence: Avery August,
| |
Collapse
|
6
|
Luo M, Liu YQ, Zhang H, Luo CH, Liu Q, Wang WY, He ZC, Chen C, Zhang XN, Mao M, Yang KD, Wang C, Chen XQ, Fu WJ, Niu Q, Bian XW, Shi Y, Ping YF. Overexpression of carnitine palmitoyltransferase 1A promotes mitochondrial fusion and differentiation of glioblastoma stem cells. J Transl Med 2022; 102:722-730. [PMID: 34963686 DOI: 10.1038/s41374-021-00724-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 11/09/2022] Open
Abstract
Glioma stem cells (GSCs) are self-renewing tumor cells with multi-lineage differentiation potential and the capacity of construct glioblastoma (GBM) heterogenicity. Mitochondrial morphology is associated with the metabolic plasticity of GBM cells. Previous studies have revealed distinct mitochondrial morphologies and metabolic phenotypes between GSCs and non-stem tumor cells (NSTCs), whereas the molecules regulating mitochondrial dynamics in GBM cells are largely unknown. Herein, we report that carnitine palmitoyltransferase 1A (CPT1A) is preferentially expressed in NSTCs, and governs mitochondrial dynamics and GSC differentiation. Expressions of CPT1A and GSC marker CD133 were mutually exclusive in human GBMs. Overexpression of CPT1A inhibited GSC self-renewal but promoted mitochondrial fusion. In contrast, disruption of CPT1A in NSTCs promoted mitochondrial fission and reprogrammed NSTCs toward GSC feature. Mechanistically, CPT1A overexpression increased the phosphorylation of dynamin-related protein 1 at Ser-637 to promote mitochondrial fusion. In vivo, CPT1A overexpression decreased the percentage of GSCs, impaired GSC-derived xenograft growth and prolonged tumor-bearing mice survival. Our work identified CPT1A as a critical regulator of mitochondrial dynamics and GSC differentiation, indicating that CPT1A could be developed as a molecular target for GBM cell-differentiation strategy.
Collapse
Affiliation(s)
- Min Luo
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Yu-Qi Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Hua Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Chun-Hua Luo
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Qing Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Wen-Ying Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Zhi-Cheng He
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Cong Chen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Xiao-Ning Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Min Mao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Kai-Di Yang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Chao Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Xiao-Qing Chen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Wen-Juan Fu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Qin Niu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Yu Shi
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China.
| | - Yi-Fang Ping
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China.
| |
Collapse
|
7
|
Kang J, li N, Wang F, Wei Y, Zeng Y, Luo Q, Sun X, Xu H, Peng J, Zhou F. Exploration of Reduced Mitochondrial Content–Associated Gene Signature and Immunocyte Infiltration in Colon Adenocarcinoma by an Integrated Bioinformatic Analysis. Front Genet 2022; 13:832331. [PMID: 35464857 PMCID: PMC9024084 DOI: 10.3389/fgene.2022.832331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 03/14/2022] [Indexed: 12/11/2022] Open
Abstract
Purpose: Mitochondrial dysfunction refers to cancer immune evasion. A novel 7-gene prognostic signature related to the mitochondrial DNA copy number was utilized to evaluate the immunocyte infiltration in colon cancer according to the risk scores and to predict the survival for colon cancer.Experimental design: We performed an integrated bioinformatic analysis to analyze transcriptome profiling of the EB-treated mitochondrial DNA–defected NCM460 cell line with differentially expressed genes between tumor and normal tissues of COAD in TCGA. The LASSO analysis was utilized to establish a prognostic signature. ESTIMATE and CIBERSORT validated the differences of immunocyte infiltration between colon cancer patients with high- and low-risk scores.Results: Our study identified a 7-gene prognostic signature (LRRN2, ANKLE1, GPRASP1, PRAME, TCF7L1, RAB6B, and CALB2). Patients with colon cancer were split into the high- and low-risk group by the risk scores in TCGA (training cohort: HR = 2.50 p < 0.0001) and GSE39582 (validation cohort: HR = 1.43 p < 0.05). ESTIMATE and CIBERSORT revealed diverseness of immune infiltration in the two groups, especially downregulated T-cell infiltration in the patients with high-risk scores. Finally, we validated the colon patients with a low expression of the mitochondrial number biomarker TFAM had less CD3+ and CD8+ T-cell infiltration in clinical specimens.Conclusion: An mtDNA copy number-related 7-gene prognostic signature was investigated and evaluated, which may help to predict the prognosis of colon cancer patients and to guide clinical immunotherapy via immunocyte infiltration evaluation.
Collapse
Affiliation(s)
- Jinlin Kang
- Department of Radiation and Medical Oncology, Zhongnan Hospital Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Na li
- Department of Radiation and Medical Oncology, Zhongnan Hospital Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
- Renmin Hospital of Wuhan University, Wuhan, China
| | - Fen Wang
- Department of Radiation and Medical Oncology, Zhongnan Hospital Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Yan Wei
- Department of Radiation and Medical Oncology, Zhongnan Hospital Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Yangyang Zeng
- Department of Radiation and Medical Oncology, Zhongnan Hospital Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Qifan Luo
- Department of Radiation and Medical Oncology, Zhongnan Hospital Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Xuehua Sun
- Department of Radiation and Medical Oncology, Zhongnan Hospital Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Hui Xu
- Department of Radiation and Medical Oncology, Zhongnan Hospital Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Jin Peng
- Department of Radiation and Medical Oncology, Zhongnan Hospital Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
- *Correspondence: Jin Peng, ; Fuxiang Zhou,
| | - Fuxiang Zhou
- Department of Radiation and Medical Oncology, Zhongnan Hospital Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
- *Correspondence: Jin Peng, ; Fuxiang Zhou,
| |
Collapse
|
8
|
Xu J, Liu Y, Zhang Q, Su Z, Yan T, Zhou S, Wang T, Wei X, Chen Z, Hu G, Chen T, Jia G. DNA damage, serum metabolomic alteration and carcinogenic risk associated with low-level air pollution. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 297:118763. [PMID: 34998894 DOI: 10.1016/j.envpol.2021.118763] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 12/23/2021] [Accepted: 12/26/2021] [Indexed: 06/14/2023]
Abstract
Outdoor air pollution has been classified as carcinogenic to humans (Group 1) for lung cancer, but the underlying mechanism and key toxic components remain incompletely understood. Since DNA damage and metabolite alterations are associated with cancer progression, exploring potential mechanisms linking air pollution and cancer might be meaningful. In this study, a real-time ambient air exposure system was established to simulate the real-world environment of adult male SD rats in Beijing from June 13th, 2018, to October 8th, 2018. 8-OHdG in the urine, γ-H2AX in the lungs and mtDNA copy number in the peripheral blood were analyzed to explore DNA damage at different levels. Serum non-targeted metabolomics analysis was performed. Pair-wise spearman was used to explore the correlation between DNA damage biomarkers and serum differential metabolites. Carcinogenic risks of heavy metals and PAHs via inhalation were assessed according to US EPA guidelines. Results showed that PM2.5 and O3 were the major air pollutants in the exposure group and not detected in the control group. Compared with control group, higher levels of 8-OHdG, mtDNA copy number, γ-H2AX and PCNA-positive nuclei cells were observed in the exposure group. Histopathological evaluation suggested ambient air induced alveolar wall thickening and inflammatory cell infiltration in lungs. Perturbed metabolic pathways identified included glycolysis/gluconeogenesis metabolism, purine and pyrimidine metabolism, etc. γ-H2AX was positively correlated with serum ADP, 3-phospho-D-glyceroyl phosphate and N-acetyl-D-glucosamine. The BaPeq was 0.120 ng/m3. Risks of Cr(VI), As, V, BaP, BaA and BbF were above 1 × 10-6. We concluded that low-level air pollution was associated with DNA damage and serum metabolomic alterations in rats. Cr(VI) and BaP were identified as key carcinogenic components in PM2.5. Our results provided experimental evidence for hazard identification and risk assessment of low-level air pollution.
Collapse
Affiliation(s)
- Jiayu Xu
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China
| | - Yu Liu
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China
| | - Qiaojian Zhang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China
| | - Zekang Su
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China
| | - Tenglong Yan
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China
| | - Shupei Zhou
- Department of Laboratory Animal Science, Health Science Center, Peking University, Beijing, 100083, China
| | - Tiancheng Wang
- Department of Clinical Laboratory, Third Hospital of Peking University, Beijing, 100083, China
| | - Xuetao Wei
- Department of Toxicology, School of Public Health, Peking University, Beijing, 100083, China
| | - Zhangjian Chen
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China
| | - Guiping Hu
- School of Medical Science and Engineering, Beihang University, Beijing, 100191, China
| | - Tian Chen
- School of Public Health and Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Guang Jia
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China.
| |
Collapse
|
9
|
Sourty B, Dardaud LM, Bris C, Desquiret-Dumas V, Boisselier B, Basset L, Figarella-Branger D, Morel A, Sanson M, Procaccio V, Rousseau A. Mitochondrial DNA copy number as a prognostic marker is age-dependent in adult glioblastoma. Neurooncol Adv 2022; 4:vdab191. [PMID: 35118384 PMCID: PMC8807107 DOI: 10.1093/noajnl/vdab191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common and aggressive form of glioma. GBM frequently displays chromosome (chr) 7 gain, chr 10 loss and/or EGFR amplification (chr7+/chr10-/EGFRamp). Overall survival (OS) is 15 months after treatment. In young adults, IDH1/2 mutations are associated with longer survival. In children, histone H3 mutations portend a dismal prognosis. Novel reliable prognostic markers are needed in GBM. We assessed the prognostic value of mitochondrial DNA (mtDNA) copy number in adult GBM. METHODS mtDNA copy number was assessed using real-time quantitative PCR in 232 primary GBM. Methylation of POLG and TFAM genes, involved in mtDNA replication, was assessed by bisulfite-pyrosequencing in 44 and 51 cases, respectively. RESULTS Median age at diagnosis was 56.6 years-old and median OS, 13.3 months. 153/232 GBM (66 %) displayed chr7+/chr10-/EGFRamp, 23 (9.9 %) IDH1/2 mutation, 3 (1.3 %) H3 mutation and 53 (22.8 %) no key genetic alterations. GBM were divided into two groups, "Low" (n = 116) and "High" (n = 116), according to the median mtDNA/nuclear DNA ratio (237.7). There was no significant difference in OS between the two groups. By dividing the whole cohort according to the median age at diagnosis, OS was longer in the "High" vs "Low" subgroup (27.3 vs 15 months, P = .0203) in young adult GBM (n = 117) and longer in the "Low" vs "High" subgroup (14.5 vs 10.2 months, P = .0116) in older adult GBM (n = 115). POLG was highly methylated, whereas TFAM remained unmethylated. CONCLUSION mtDNA copy number may be a novel prognostic biomarker in GBM, its impact depending on age.
Collapse
Affiliation(s)
- Baptiste Sourty
- Department of Pathology, University Hospital of Angers, Angers, France
| | | | - Céline Bris
- Department of Genetics, University Hospital of Angers and Angers University, INSERM1083, CNRS6015, MITOVASC, Angers, France
| | - Valérie Desquiret-Dumas
- Department of Genetics, University Hospital of Angers and Angers University, INSERM1083, CNRS6015, MITOVASC, Angers, France
| | - Blandine Boisselier
- Department of Pathology, University Hospital of Angers, Angers, France
- Center for Research in Cancerology and Immunology Nantes/Angers, INSERM, University of Nantes, University of Angers, Angers, France
| | - Laëtitia Basset
- Department of Pathology, University Hospital of Angers, Angers, France
- Center for Research in Cancerology and Immunology Nantes/Angers, INSERM, University of Nantes, University of Angers, Angers, France
| | - Dominique Figarella-Branger
- Aix-Marseille Univ, APHM, CNRS, INP, Inst Neurophysiopathol, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France
| | - Alain Morel
- Institut de Cancérologie de l'Ouest - Paul Papin, Angers, France
| | - Marc Sanson
- Sorbonne University UPMC Univ Paris 06, INSERM CNRS, U1127, UMR 7225, ICM, F-75013, Groupe Hospitalier Pitié-Salpêtrière, Neurology Department 2, Paris, France
| | - Vincent Procaccio
- Department of Genetics, University Hospital of Angers and Angers University, INSERM1083, CNRS6015, MITOVASC, Angers, France
| | - Audrey Rousseau
- Department of Pathology, University Hospital of Angers, Angers, France
- Center for Research in Cancerology and Immunology Nantes/Angers, INSERM, University of Nantes, University of Angers, Angers, France
| |
Collapse
|
10
|
Ji B, Xiao LY, Ren JC, Zhang GH, Wang Y, Dong T, Li J, Zhang F, Xia ZL. Gene-Environment Interactions Between Environmental Response Genes Polymorphisms and Mitochondrial DNA Copy Numbers Among Benzene Workers. J Occup Environ Med 2021; 63:e408-e415. [PMID: 34184658 DOI: 10.1097/jom.0000000000002225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVE To determine the effect of mitochondrial DNA copy number (mtDNAcn) as a biomarker of benzene exposure. METHODS A total of 294 benzene-exposed workers and 102 controls were recruited. Biomarkers of mtDNAcn, cytokinesis-block micronucleus (MN) frequency, and peripheral blood white blood cells (WBC) were detected. Eighteen polymorphism sites in DNA damage repair and metabolic genes were analyzed. RESULTS Benzene exposure increased mtDNAcn and indicated a dose-response relationship (P < 0.001). mtDNAcn was negatively correlated with WBC count and DNA methylation and positively correlated with MN frequency. The AG type in rs1695 interacted with benzene exposure to aggravate mtDNAcn (β = 0.006, 95% CI: 0, 0.012, P = 0.050). rs13181, rs1695, rs1800975, and GSTM1 null were associated with benzene-induced mtDNAcn. Rs1695 interacted with benzene to increase mitochondrial damage. CONCLUSIONS Benzene exposure increases mtDNAcn levels in benzene-exposed workers.
Collapse
Affiliation(s)
- Buqiang Ji
- Department of Hematology, Linyi People's Hospital, 27 Jifang Road, Linyi, China (Ji, Xiao), School of Public Health, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, China (Ren, Zhang, Wang, Dong, Li, Zhang), Department of Occupational Health and Toxicology, School of Public Health, Fudan University, 138 Yixueyuan Road, Shanghai, China (Xia)
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Abstract
Many tumors are now understood to be heterogenous cell populations arising from a minority of epithelial-like cancer stem cells (CSCs). CSCs demonstrate distinctive metabolic signatures from the more differentiated surrounding tumor bulk that confer resistance to traditional chemotherapeutic regimens and potential for tumor relapse. Many CSC phenotypes including metabolism, epithelial-to-mesenchymal transition, cellular signaling pathway activity, and others, arise from altered mitochondrial function and turnover, which are regulated by constant cycles of mitochondrial fusion and fission. Further, recycling of mitochondria through mitophagy in CSCs is associated with maintenance of reactive oxygen species levels that dictate gene expression. The protein machinery that drives mitochondrial dynamics is surprisingly simple and may represent attractive new therapeutic avenues to target CSC metabolism and selectively eradicate tumor-generating cells to reduce the risks of metastasis and relapse for a variety of tumor types.
Collapse
Affiliation(s)
- Dane T Sessions
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - David F Kashatus
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA, 22908, USA.
| |
Collapse
|
12
|
Sravya P, Nimbalkar VP, Kanuri NN, Sugur H, Verma BK, Kundu P, Rao S, Uday Krishna AS, Somanna S, Kondaiah P, Arivazhagan A, Santosh V. Low mitochondrial DNA copy number is associated with poor prognosis and treatment resistance in glioblastoma. Mitochondrion 2020; 55:154-163. [PMID: 33045388 DOI: 10.1016/j.mito.2020.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/27/2020] [Accepted: 10/05/2020] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Mitochondrial DNA (mtDNA) content in several solid tumors was found to be lower than in their normal counterparts. However, there is paucity of literature on the clinical significance of mtDNA content in glioblastoma and its effect on treatment response. Hence, we studied the prognostic significance of mtDNA content in glioblastoma tumor tissue and the effect of mtDNA depletion in glioblastoma cells on response to treatment. MATERIALS AND METHODS 130 newly diagnosed glioblastomas, 32 paired newly diagnosed and recurrent glioblastomas and 35 non-neoplastic brain tissues were utilized for the study. mtDNA content in the patient tumor tissue was assessed and compared with known biomarkers and patient survival. mtDNA was chemically depleted in malignant glioma cell lines, U87, LN229. The biology and treatment response of parent and depleted cells were compared. RESULTS Lower range of mtDNA copy number in glioblastoma was associated with poor overall survival (p = 0.01), progression free survival (p = 0.04) and also with wild type IDH (p = 0.02). In recurrent glioblastoma, mtDNA copy number was higher than newly diagnosed glioblastoma in the patients who received RT (p = 0.01). mtDNA depleted U87 and LN229 cells showed higher survival fraction post radiation exposure when compared to parent lines. The IC50 of TMZ was also higher for mtDNA depleted U87 and LN229 cells. The depleted cells formed more neurospheres than their parent counterparts, thus showing increased stemness of mtDNA depleted cells. CONCLUSION Low mtDNA copy number in glioblastoma is associated with poor patient survival and treatment resistance in cell lines possibly by impacting stemness of the glioblastoma cells.
Collapse
Affiliation(s)
- Palavalasa Sravya
- Department of Clinical Neurosciences, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Vidya Prasad Nimbalkar
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Nandaki Nag Kanuri
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Harsha Sugur
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Brijesh Kumar Verma
- Department of Molecular Reproduction Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Paramita Kundu
- Department of Molecular Reproduction Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Shilpa Rao
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - A S Uday Krishna
- Department of Radiation Oncology, KIDWAI Memorial Institute of Oncology, Bengaluru, India
| | - Sampath Somanna
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Paturu Kondaiah
- Department of Molecular Reproduction Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Arimappamagan Arivazhagan
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, Bengaluru, India.
| | - Vani Santosh
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bengaluru, India.
| |
Collapse
|
13
|
Kobayashi M, Kobayashi J, Shirasuna K, Iwata H. Abundance of cell-free mitochondrial DNA in spent culture medium associated with morphokinetics and blastocyst collapse of expanded blastocysts. Reprod Med Biol 2020; 19:404-414. [PMID: 33071643 PMCID: PMC7542021 DOI: 10.1002/rmb2.12344] [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: 06/10/2020] [Revised: 07/22/2020] [Accepted: 07/25/2020] [Indexed: 01/28/2023] Open
Abstract
Purpose This retrospective observational study investigated relationships between the abundance of cell‐free mitochondrial DNA (cf‐mtDNA) in spent culture medium (SCM) of human‐expanded blastocysts and their morphokinetics to address the question of whether the abundance of cf‐mtDNA in SCM could predict the quality of blastocysts. Methods Embryos (n = 53) were individually cultured in a time‐lapse incubator until they reached the expanded blastocyst stage (5 or 6 days), following which copy numbers of cf‐mtDNA in SCM (20 μL) of expanded blastocysts were determined using real‐time PCR. Results The duration between start of blastulation to expanded blastocyst (tEB–tSB) and between that of the blastocyst stage to expanded blastocyst (tEB–tB) significantly and positively correlated with the abundance of cf‐mtDNA in the SCM (tEB–tSB: r = .46; P < .01; tEB–tB: r = .47; P < .01). The abundance of cf‐mtDNA in the SCM was significantly greater in blastocysts with blastocyst collapse (BC), than without BC, and significantly and positively correlated with the number of BC. Conclusions The abundance of cf‐mtDNA in the SCM was associated with expansion duration and BC. Thus, cf‐mtDNA abundance in the SCM serves as a marker to predict the quality of expanded blastocysts.
Collapse
Affiliation(s)
- Mitsuru Kobayashi
- Department of Animal Science Tokyo University of Agriculture Atsugi Japan.,Kanagawa Ladies Clinic Yokohama Japan
| | | | - Koumei Shirasuna
- Department of Animal Science Tokyo University of Agriculture Atsugi Japan
| | - Hisataka Iwata
- Department of Animal Science Tokyo University of Agriculture Atsugi Japan
| |
Collapse
|
14
|
Dong Z, Pu L, Cui H. Mitoepigenetics and Its Emerging Roles in Cancer. Front Cell Dev Biol 2020; 8:4. [PMID: 32039210 PMCID: PMC6989428 DOI: 10.3389/fcell.2020.00004] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 01/08/2020] [Indexed: 12/11/2022] Open
Abstract
In human beings, there is a ∼16,569 bp circular mitochondrial DNA (mtDNA) encoding 22 tRNAs, 12S and 16S rRNAs, 13 polypeptides that constitute the central core of ETC/OxPhos complexes, and some non-coding RNAs. Recently, mtDNA has been shown to have some covalent modifications such as methylation or hydroxylmethylation, which play pivotal epigenetic roles in mtDNA replication and transcription. Post-translational modifications of proteins in mitochondrial nucleoids such as mitochondrial transcription factor A (TFAM) also emerge as essential epigenetic modulations in mtDNA replication and transcription. Post-transcriptional modifications of mitochondrial RNAs (mtRNAs) including mt-rRNAs, mt-tRNAs and mt-mRNAs are important epigenetic modulations. Besides, mtDNA or nuclear DNA (n-DNA)-derived non-coding RNAs also play important roles in the regulation of translation and function of mitochondrial genes. These evidences introduce a novel concept of mitoepigenetics that refers to the study of modulations in the mitochondria that alter heritable phenotype in mitochondria itself without changing the mtDNA sequence. Since mitochondrial dysfunction contributes to carcinogenesis and tumor development, mitoepigenetics is also essential for cancer. Understanding the mode of actions of mitoepigenetics in cancers may shade light on the clinical diagnosis and prevention of these diseases. In this review, we summarize the present study about modifications in mtDNA, mtRNA and nucleoids and modulations of mtDNA/nDNA-derived non-coding RNAs that affect mtDNA translation/function, and overview recent studies of mitoepigenetic alterations in cancer.
Collapse
Affiliation(s)
- Zhen Dong
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Chongqing, China.,Cancer Center, Medical Research Institute, Southwest University, Chongqing, China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing, China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Chongqing, China
| | - Longjun Pu
- Umeå Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Chongqing, China.,Cancer Center, Medical Research Institute, Southwest University, Chongqing, China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing, China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Chongqing, China
| |
Collapse
|
15
|
Li Z, Fu J, Li Z, Tang Y, Hua Q, Liu L, Zhao J. Air pollution and placental mitochondrial DNA copy number: Mechanistic insights and epidemiological challenges. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 255:113266. [PMID: 31557557 DOI: 10.1016/j.envpol.2019.113266] [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: 05/01/2019] [Revised: 09/12/2019] [Accepted: 09/15/2019] [Indexed: 06/10/2023]
Abstract
During embryogenesis and embryo implantation, the copy number of mtDNA is elaborately regulated to meet the cellular demand for division, growth and differentiation. With large numbers of mitochondria for energy production, placental cells possess strong endocrine functionalities and capacities for efficient signaling communication. Recently, several environmental epidemiological studies have shown an association between mitochondrial DNA copy number, adverse birth outcomes and maternal exposure to air pollution, which has shed light on the possible effect of pollutants on placental molecular events. Because the mtDNA replication is thought to be a direct drive of mtDNA change, we tried to highlight the essential factors involved in the process of mtDNA replication. Then we traced the mtDNA change in the formation of placenta during embryogenesis, and evaluated the importance of mitochondrial genome maintenance during gestation. The possible mechanism from the epidemiological and experimental studies were reviewed and summarized, and recommendations were proposed for future studies to improve the precision of the estimated difference. The issue will be well-understood if the integrated profiles, such as familial genetic tendency, maternal genetic information, identification of mitochondrial DNA copy number in each placental cell type, and total personal exposure assessment, are considered in the future study.
Collapse
Affiliation(s)
- Zhen Li
- Department of Preventative Medicine, Zhejiang Key Laboratory of Pathophysiology, Medicine School of Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang Province 315211, People's Republic of China.
| | - Jianfei Fu
- Department of Medical Records and Statistics, Ningbo First Hospital, Ningbo, Zhejiang Province 315010, People's Republic of China
| | - Zhou Li
- Department of Preventative Medicine, Zhejiang Key Laboratory of Pathophysiology, Medicine School of Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang Province 315211, People's Republic of China
| | - Yuqing Tang
- Department of Preventative Medicine, Zhejiang Key Laboratory of Pathophysiology, Medicine School of Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang Province 315211, People's Republic of China
| | - Qihang Hua
- Department of Preventative Medicine, Zhejiang Key Laboratory of Pathophysiology, Medicine School of Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang Province 315211, People's Republic of China
| | - Liya Liu
- Department of Preventative Medicine, Zhejiang Key Laboratory of Pathophysiology, Medicine School of Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang Province 315211, People's Republic of China
| | - Jinshun Zhao
- Department of Preventative Medicine, Zhejiang Key Laboratory of Pathophysiology, Medicine School of Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang Province 315211, People's Republic of China
| |
Collapse
|
16
|
Genomic Balance: Two Genomes Establishing Synchrony to Modulate Cellular Fate and Function. Cells 2019; 8:cells8111306. [PMID: 31652817 PMCID: PMC6912345 DOI: 10.3390/cells8111306] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/17/2019] [Accepted: 10/22/2019] [Indexed: 01/21/2023] Open
Abstract
It is becoming increasingly apparent that cells require cooperation between the nuclear and mitochondrial genomes to promote effective function. However, it was long thought that the mitochondrial genome was under the strict control of the nuclear genome and the mitochondrial genome had little influence on cell fate unless it was extensively mutated, as in the case of the mitochondrial DNA diseases. However, as our understanding of the roles that epigenetic regulators, including DNA methylation, and metabolism play in cell fate and function, the role of the mitochondrial genome appears to have a greater influence than previously thought. In this review, I draw on examples from tumorigenesis, stem cells, and oocyte pre- and post-fertilisation events to discuss how modulating one genome affects the other and that this results in a compromise to produce functional mature cells. I propose that, during development, both of the genomes interact with each other through intermediaries to establish genomic balance and that establishing genomic balance is a key facet in determining cell fate and viability.
Collapse
|
17
|
Mitochondria and Female Germline Stem Cells-A Mitochondrial DNA Perspective. Cells 2019; 8:cells8080852. [PMID: 31398797 PMCID: PMC6721711 DOI: 10.3390/cells8080852] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/06/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023] Open
Abstract
Mitochondria and mitochondrial DNA have important roles to play in development. In primordial germ cells, they progress from small numbers to populate the maturing oocyte with high numbers to support post-fertilization events. These processes take place under the control of significant changes in DNA methylation and other epigenetic modifiers, as well as changes to the DNA methylation status of the nuclear-encoded mitochondrial DNA replication factors. Consequently, the differentiating germ cell requires significant synchrony between the two genomes in order to ensure that they are fit for purpose. In this review, I examine these processes in the context of female germline stem cells that are isolated from the ovary and those derived from embryonic stem cells and reprogrammed somatic cells. Although our knowledge is limited in this respect, I provide predictions based on other cellular systems of what is expected and provide insight into how these cells could be used in clinical medicine.
Collapse
|
18
|
Sun X, Johnson J, St John JC. Global DNA methylation synergistically regulates the nuclear and mitochondrial genomes in glioblastoma cells. Nucleic Acids Res 2019; 46:5977-5995. [PMID: 29722878 PMCID: PMC6158714 DOI: 10.1093/nar/gky339] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 04/19/2018] [Indexed: 12/12/2022] Open
Abstract
Replication of mitochondrial DNA is strictly regulated during differentiation and development allowing each cell type to acquire its required mtDNA copy number to meet its specific needs for energy. Undifferentiated cells establish the mtDNA set point, which provides low numbers of mtDNA copy but sufficient template for replication once cells commit to specific lineages. However, cancer cells, such as those from the human glioblastoma multiforme cell line, HSR-GBM1, cannot complete differentiation as they fail to enforce the mtDNA set point and are trapped in a ‘pseudo-differentiated’ state. Global DNA methylation is likely to be a major contributing factor, as DNA demethylation treatments promote differentiation of HSR-GBM1 cells. To determine the relationship between DNA methylation and mtDNA copy number in cancer cells, we applied whole genome MeDIP-Seq and RNA-Seq to HSR-GBM1 cells and following their treatment with the DNA demethylation agents 5-azacytidine and vitamin C. We identified key methylated regions modulated by the DNA demethylation agents that also induced synchronous changes to mtDNA copy number and nuclear gene expression. Our findings highlight the control exerted by DNA methylation on the expression of key genes, the regulation of mtDNA copy number and establishment of the mtDNA set point, which collectively contribute to tumorigenesis.
Collapse
Affiliation(s)
- Xin Sun
- Centre for Genetic Diseases, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168, Australia.,Department of Molecular and Translational Sciences, Monash University, 27-31 Wright Street, Clayton, VIC 3168, Australia
| | - Jacqueline Johnson
- Centre for Genetic Diseases, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168, Australia
| | - Justin C St John
- Centre for Genetic Diseases, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168, Australia.,Department of Molecular and Translational Sciences, Monash University, 27-31 Wright Street, Clayton, VIC 3168, Australia
| |
Collapse
|
19
|
Wang T, Xie X, Liu H, Chen F, Du J, Wang X, Jiang X, Yu F, Fan H. Pyridine nucleotide-disulphide oxidoreductase domain 2 (PYROXD2): Role in mitochondrial function. Mitochondrion 2019; 47:114-124. [PMID: 31170524 DOI: 10.1016/j.mito.2019.05.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 05/23/2019] [Accepted: 05/30/2019] [Indexed: 02/05/2023]
Abstract
Pyridine Nucleotide-Disulphide Oxidoreductase Domain 2 (PYROXD2), a Hepatitis B virus X protein (HBx)-interacting protein, is significantly down-regulated in hepatocellular carcinoma (HCC), however its exact biological function remains unclear. The aim of this study is to investigate the subcellular localization and biological function of PYROXD2 in hepatic cells. The results showed that PYROXD2 was imported to the mitochondrial inner membrane/matrix by Tom40 and Tim23, but not Mia40. PYROXD2 151-230aa might be the mitochondrial targeting sequence. PYROXD2 interacted with complex IV subunit COX5B. Knockout of PYROXD2 decreased MMP, intracellular ROS, complex IV activity, cell proliferation, ATP content and mtDNA copy number, but increased mtROS levels and the number of immature mitochondria. In summary, our data illustrated that PYROXD2 localizes to the mitochondrial inner membrane/matrix, and it plays important roles in regulating mitochondrial function.
Collapse
Affiliation(s)
- Tao Wang
- Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou 310036, China
| | - Xiaoyuan Xie
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - HuiLin Liu
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Feng Chen
- Blood center of Zhejiang province, Hangzhou, Zhejiang 310052, China
| | - Jianhua Du
- Nanchang Institute of Science and Technology, Nanchang 330108, China
| | - XingZhi Wang
- Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou 310036, China
| | - XingYan Jiang
- Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou 310036, China
| | - Fang Yu
- Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou 310036, China
| | - Handong Fan
- Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou 310036, China.
| |
Collapse
|
20
|
Srirattana K, St John JC. Transmission of Dysfunctional Mitochondrial DNA and Its Implications for Mammalian Reproduction. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2019; 231:75-103. [PMID: 30617719 DOI: 10.1007/102_2018_3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mitochondrial DNA (mtDNA) encodes proteins for the electron transport chain which produces the vast majority of cellular energy. MtDNA has its own replication and transcription machinery that relies on nuclear-encoded transcription and replication factors. MtDNA is inherited in a non-Mendelian fashion as maternal-only mtDNA is passed onto the next generation. Mutation to mtDNA can cause mitochondrial dysfunction, which affects energy production and tissue and organ function. In somatic cell nuclear transfer (SCNT), there is an issue with the mixing of two populations of mtDNA, namely from the donor cell and recipient oocyte. This review focuses on the transmission of mtDNA in SCNT embryos and offspring. The transmission of donor cell mtDNA can be prevented by depleting the donor cell of its mtDNA using mtDNA depletion agents prior to SCNT. As a result, SCNT embryos harbour oocyte-only mtDNA. Moreover, culturing SCNT embryos derived from mtDNA depleted cells in media supplemented with a nuclear reprograming agent can increase the levels of expression of genes related to embryo development when compared with non-depleted cell-derived embryos. Furthermore, we have reviewed how mitochondrial supplementation in oocytes can have beneficial effects for SCNT embryos by increasing mtDNA copy number and the levels of expression of genes involved in energy production and decreasing the levels of expression of genes involved in embryonic cell death. Notably, there are beneficial effects of mtDNA supplementation over the use of nuclear reprograming agents in terms of regulating gene expression in embryos. Taken together, manipulating mtDNA in donor cells and/or oocytes prior to SCNT could enhance embryo production efficiency.
Collapse
Affiliation(s)
- Kanokwan Srirattana
- Mitochondrial Genetics Group, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Justin C St John
- Mitochondrial Genetics Group, Hudson Institute of Medical Research, Clayton, VIC, Australia. .,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
21
|
Sun X, Vaghjiani V, Jayasekara WSN, Cain JE, St John JC. The degree of mitochondrial DNA methylation in tumor models of glioblastoma and osteosarcoma. Clin Epigenetics 2018; 10:157. [PMID: 30558637 PMCID: PMC6296150 DOI: 10.1186/s13148-018-0590-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 11/21/2018] [Indexed: 12/16/2022] Open
Abstract
Background Different cell types possess different copies of mtDNA to support their specific requirements for cellular metabolism. Cell-specific mtDNA copy numbers are established through cell-specific mtDNA replication during cell differentiation. However, cancer cells are trapped in a “pseudo-differentiated” state as they fail to expand mtDNA copy number. Global DNA methylation can regulate this process, as induced DNA demethylation promotes differentiation of cancer cells and expansion of mtDNA copy number. Results To determine the role that mtDNA methylation plays in regulating mtDNA replication during tumorigenesis, we have characterized the patterns of mtDNA methylation using glioblastoma and osteosarcoma tumor models that have different combinations of mtDNA genotypes and copy number against common nuclear genome backgrounds at different stages of tumor progression. To ensure the reliability of the findings, we have applied a robust experimental pipeline including three approaches, namely whole-mtDNA bisulfite-sequencing with mtDNA-genotype-specific analysis, pyrosequencing, and methylated immunoprecipitation against 5mC and 5hmC. We have determined genotype-specific methylation profiles, which were modulated through tumor progression. Moreover, a strong influence from the nuclear genome was also observed on mtDNA methylation patterns using the same mtDNA genotype under different nuclear genomes. Furthermore, the numbers of mtDNA copy in tumor-initiating cells affected mtDNA methylation levels in late-stage tumors. Conclusions Our findings highlight the influences that the nuclear and mitochondrial genomes have in setting mtDNA methylation patterns to regulate mtDNA copy number in tumorigenesis. They have important implications for assessing global DNA methylation patterns in tumorigenesis and the availability of mtDNA template for mtDNA replication. Electronic supplementary material The online version of this article (10.1186/s13148-018-0590-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xin Sun
- Mitochondrial Genetics Group, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC, 3168, Australia.,Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, 27-31 Wright Street, Clayton, VIC, 3168, Australia
| | - Vijesh Vaghjiani
- Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, 27-31 Wright Street, Clayton, VIC, 3168, Australia.,Centre for Cancer Research, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC, 3168, Australia
| | - W Samantha N Jayasekara
- Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, 27-31 Wright Street, Clayton, VIC, 3168, Australia.,Centre for Cancer Research, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC, 3168, Australia
| | - Jason E Cain
- Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, 27-31 Wright Street, Clayton, VIC, 3168, Australia.,Centre for Cancer Research, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC, 3168, Australia
| | - Justin C St John
- Mitochondrial Genetics Group, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC, 3168, Australia. .,Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, 27-31 Wright Street, Clayton, VIC, 3168, Australia.
| |
Collapse
|
22
|
Ryzhkova AI, Sazonova MA, Sinyov VV, Galitsyna EV, Chicheva MM, Melnichenko AA, Grechko AV, Postnov AY, Orekhov AN, Shkurat TP. Mitochondrial diseases caused by mtDNA mutations: a mini-review. Ther Clin Risk Manag 2018; 14:1933-1942. [PMID: 30349272 PMCID: PMC6186303 DOI: 10.2147/tcrm.s154863] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
There are several types of mitochondrial cytopathies, which cause a set of disorders, arise as a result of mitochondria’s failure. Mitochondria’s functional disruption leads to development of physical, growing and cognitive disabilities and includes multiple organ pathologies, essentially disturbing the nervous and muscular systems. The origins of mitochondrial cytopathies are mutations in genes of nuclear DNA encoding mitochondrial proteins or in mitochondrial DNA. Nowadays, numerous mtDNA mutations significant to the appearance and progress of pathologies in humans are detected. In this mini-review, we accent on the mitochondrial cytopathies related to mutations of mtDNA. As well known, there are definite set of symptoms of mitochondrial cytopathies distinguishing or similar for different syndromes. The present article contains data about mutations linked with cytopathies that facilitate diagnosis of different syndromes by using genetic analysis methods. In addition, for every individual, more effective therapeutic approach could be developed after wide-range mutant background analysis of mitochondrial genome.
Collapse
Affiliation(s)
- Anastasia I Ryzhkova
- Laboratory of Medical Genetics, National Medical Research Center of Cardiology, Moscow, Russian Federation, .,Department of Virology, K.I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology-MVA, Moscow, Russian Federation,
| | - Margarita A Sazonova
- Laboratory of Medical Genetics, National Medical Research Center of Cardiology, Moscow, Russian Federation, .,Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russian Federation
| | - Vasily V Sinyov
- Laboratory of Medical Genetics, National Medical Research Center of Cardiology, Moscow, Russian Federation,
| | - Elena V Galitsyna
- Department of Genetics, Southern Federal University, Rostov-on-Don, Russian Federation
| | - Mariya M Chicheva
- Department of Genetics, Southern Federal University, Rostov-on-Don, Russian Federation
| | | | - Andrey V Grechko
- Federal Research and Clinical Center of Reanimatology and Rehabilitology, Moscow, Russian Federation
| | - Anton Yu Postnov
- Laboratory of Medical Genetics, National Medical Research Center of Cardiology, Moscow, Russian Federation,
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russian Federation.,Institute for Atherosclerosis Research, Skolkovo Innovative Centre, Moscow Region, Russian Federation
| | - Tatiana P Shkurat
- Department of Genetics, Southern Federal University, Rostov-on-Don, Russian Federation
| |
Collapse
|
23
|
Sun X, St John JC. Modulation of mitochondrial DNA copy number in a model of glioblastoma induces changes to DNA methylation and gene expression of the nuclear genome in tumours. Epigenetics Chromatin 2018; 11:53. [PMID: 30208958 PMCID: PMC6136172 DOI: 10.1186/s13072-018-0223-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/06/2018] [Indexed: 01/23/2023] Open
Abstract
Background There are multiple copies of mitochondrial DNA (mtDNA) present in each cell type, and they are strictly regulated in a cell-specific manner by a group of nuclear-encoded mtDNA-specific replication factors. This strict regulation of mtDNA copy number is mediated by cell-specific DNA methylation of these replication factors. Glioblastoma multiforme, HSR-GBM1, cells are hyper-methylated and maintain low mtDNA copy number to support their tumorigenic status. We have previously shown that when HSR-GBM1 cells with 50% of their original mtDNA content were inoculated into mice, tumours grew more aggressively than non-depleted cells. However, when the cells possessed only 3% and 0.2% of their original mtDNA content, tumour formation was less frequent and the initiation of tumorigenesis was significantly delayed. Importantly, the process of tumorigenesis was dependent on mtDNA copy number being restored to pre-depletion levels. Results By performing whole genome MeDIP-Seq and RNA-Seq on tumours generated from cells possessing 100%, 50%, 0.3% and 0.2% of their original mtDNA content, we determined that restoration of mtDNA copy number caused significant changes to both the nuclear methylome and its transcriptome for each tumour type. The affected genes were specifically associated with gene networks and pathways involving behaviour, nervous system development, cell differentiation and regulation of transcription and cellular processes. The mtDNA-specific replication factors were also modulated. Conclusions Our results highlight the bidirectional control of the nuclear and mitochondrial genomes through modulation of DNA methylation to control mtDNA copy number, which, in turn, modulates nuclear gene expression during tumorigenesis. Electronic supplementary material The online version of this article (10.1186/s13072-018-0223-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xin Sun
- Mitochondrial Genetics Group, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC, 3168, Australia.,Department of Molecular and Translational Sciences, Monash University, 27-31 Wright Street, Clayton, VIC, 3168, Australia
| | - Justin C St John
- Mitochondrial Genetics Group, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC, 3168, Australia. .,Department of Molecular and Translational Sciences, Monash University, 27-31 Wright Street, Clayton, VIC, 3168, Australia.
| |
Collapse
|
24
|
Wang Z, Long H, Chang C, Zhao M, Lu Q. Crosstalk between metabolism and epigenetic modifications in autoimmune diseases: a comprehensive overview. Cell Mol Life Sci 2018; 75:3353-3369. [PMID: 29974127 PMCID: PMC11105184 DOI: 10.1007/s00018-018-2864-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 06/20/2018] [Accepted: 06/25/2018] [Indexed: 12/11/2022]
Abstract
Little information is available regarding mechanistic links between epigenetic modifications and autoimmune diseases. It seems plausible to surmise that aberrant gene expression and energy metabolism would disrupt immune tolerance, which could ultimately result in autoimmune responses. Metaboloepigenetics is an emerging paradigm that defines the interrelationships between metabolism and epigenetics. Epigenetic modifications, such as the methylation/demethylation of DNA and histone proteins and histone acetylation/deacetylation can be dynamically produced and eliminated by a group of enzymes that consume several metabolites derived from various physiological pathways. Recent insights into cellular metabolism have demonstrated that environmental stimuli such as dietary exposure and nutritional status act through the variation in concentration of metabolites to affect epigenetic regulation and breakdown biochemical homeostasis. Metabolites, including S-adenosylmethionine, acetyl-CoA, nicotinamide adenine dinucleotide, α-ketoglutarate, and ATP serve as cofactors for chromatin-modifying enzymes, such as methyltransferases, deacetylases and kinases, which are responsible for chromatin remodelling. The concentration of crucial nutrients, such as glucose, glutamine, and oxygen, spatially and temporally modulate epigenetic modifications to regulate gene expression and the reaction to stressful microenvironments in disease pathology. In this review, we focus on the interaction between metabolic intermediates and epigenetic modifications, integrating environmental signals with programmes through modification of the epigenome-metabolome to speculate as to how this may influence autoimmune diseases.
Collapse
Affiliation(s)
- Zijun Wang
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, No. 139 Renmin Middle Rd, Changsha, 410011, Hunan, China
| | - Hai Long
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, No. 139 Renmin Middle Rd, Changsha, 410011, Hunan, China
| | - Christopher Chang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Suite 6510, 451 Health Sciences Drive, Davis, CA, 95616, USA
| | - Ming Zhao
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, No. 139 Renmin Middle Rd, Changsha, 410011, Hunan, China.
| | - Qianjin Lu
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, No. 139 Renmin Middle Rd, Changsha, 410011, Hunan, China.
| |
Collapse
|
25
|
Loureiro R, Mesquita KA, Magalhães-Novais S, Oliveira PJ, Vega-Naredo I. Mitochondrial biology in cancer stem cells. Semin Cancer Biol 2017; 47:18-28. [DOI: 10.1016/j.semcancer.2017.06.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 06/24/2017] [Accepted: 06/27/2017] [Indexed: 02/06/2023]
|
26
|
Thomas TM, Yu JS. Metabolic regulation of glioma stem-like cells in the tumor micro-environment. Cancer Lett 2017; 408:174-181. [PMID: 28743531 PMCID: PMC5790120 DOI: 10.1016/j.canlet.2017.07.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 07/11/2017] [Accepted: 07/15/2017] [Indexed: 12/18/2022]
Abstract
Cancer metabolism has emerged as one of the most interesting old ideas being revisited from a new perspective. In the early 20th century Otto Warburg declared metabolism the prime cause in a disease of many secondary causes, and this statement seems more prescient in view of modern expositions into the true nature of tumor evolution. As the complexity of tumor heterogeneity becomes more clear from a genetic perspective, it is important to consider the inevitably heterogeneous metabolic components of the tumor and the tumor microenvironment. High grade gliomas remain one of the most difficult to treat solid tumors, due in part to the highly vascularized nature of the tumor and the maintenance of more resistant stem-like subpopulations within the tumor. Maintenance of glioma stem cells (GSCs) requires specific alterations within the cells and the greater tumor microenvironment with regards to signaling and metabolism. Specific niches within gliomas help foster the survival of stem-like sub-populations of cells with high tumorigenicity and high metabolic plasticity. Understanding these maintenance pathways and the metabolic dependencies within the niche may highlight potential avenues of addressing tumor resistance and recurrence in glioma patients.
Collapse
Affiliation(s)
- Tom M Thomas
- Maxine-Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - John S Yu
- Maxine-Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
27
|
Wang D, Li GD, Fan Y, Zhang DF, Bi R, Yu XF, Long H, Li YY, Yao YG. The mtDNA replication-related genes TFAM and POLG are associated with leprosy in Han Chinese from Southwest China. J Dermatol Sci 2017; 88:349-356. [PMID: 28958595 DOI: 10.1016/j.jdermsci.2017.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/07/2017] [Accepted: 09/13/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND The pathogen Mycobacterium leprae of leprosy is heavily dependent on the host energy metabolites and nutritional products for survival. Previously we and others have identified associations of several mitochondrion-related genes and mitochondrial DNA (mtDNA) copy number alterations with leprosy and/or its subtype. We hypothesized that genetic variants of mtDNA replication-related genes would affect leprosy. OBJECTIVE We aimed to identify genetic associations between the mtDNA replication-related genes TFAM, POLG and leprosy. METHODS Genetic association study was performed in 2898 individuals from two independent sample sets in Yunnan Province, China. We first screened 7 tag SNPs of TFAM and POLG in 527 leprosy cases and 583 controls (Sample I). Expression quantitative trait loci (eQTL) analysis and differential mRNA expression were analyzed to discern potential effect of risk variants. The entire exon region of TFAM and POLG were further analyzed in 798 leprosy cases and 990 controls (Sample II; 4327 East Asians from the ExAC dataset was included as a reference control) by using targeted gene sequencing for fine mapping potentially causal variants. RESULTS Two tag SNPs of TFAM (rs1049432, P=0.007) and POLG (rs3176238, P=0.006) were associated with multibacillary leprosy (MB) in Sample I and the significance survived correction for multiple comparisons. SNPs rs1937 of TFAM (which was linked with rs1049432) and rs61756401 of POLG were associated with leprosy, whereas no potentially causative coding variants were identified in Sample II. The eQTL analysis showed that rs1049432 was a significant cis eQTL for TFAM in nerve tissue (P=1.20×10-12), and rs3176238 was a significant cis eQTL for POLG in nerve (P=3.90×10-13) and skin tissues (P=2.50×10-11). Consistently, mRNA level of POLG was differentially expressed in leprotic skin lesions. CONCLUSIONS Genetic variants of TFAM and POLG were associated with leprosy in Han Chinese, presumably by affecting gene expression.
Collapse
Affiliation(s)
- Dong Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650223, China
| | - Guo-Dong Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Yu Fan
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650223, China
| | - Deng-Feng Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650223, China
| | - Rui Bi
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650223, China
| | - Xiu-Feng Yu
- Wenshan Institute of Dermatology, Wenshan, Yunnan, 663000, China
| | - Heng Long
- Wenshan Institute of Dermatology, Wenshan, Yunnan, 663000, China
| | - Yu-Ye Li
- Department of Dermatology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650201, China.
| |
Collapse
|
28
|
Vaghjiani V, Cain JE, Lee W, Vaithilingam V, Tuch BE, St John JC. Modulation of Mitochondrial DNA Copy Number to Induce Hepatocytic Differentiation of Human Amniotic Epithelial Cells. Stem Cells Dev 2017; 26:1505-1519. [PMID: 28756741 DOI: 10.1089/scd.2017.0041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mitochondrial deoxyribonucleic acid (mtDNA) copy number is tightly regulated during pluripotency and differentiation. There is increased demand of cellular adenosine triphosphate (ATP) during differentiation for energy-intensive cell types such as hepatocytes and neurons to meet the cell's functional requirements. During hepatocyte differentiation, mtDNA copy number should be synchronously increased to generate sufficient ATP through oxidative phosphorylation. Unlike bone marrow mesenchymal cells, mtDNA copy number failed to increase by 28 days of differentiation of human amniotic epithelial cells (hAEC) into hepatocyte-like cells (HLC) despite their expression of some end-stage hepatic markers. This was due to higher levels of DNA methylation at exon 2 of POLGA, the mtDNA-specific replication factor. Treatment with a DNA demethylation agent, 5-azacytidine, resulted in increased mtDNA copy number, reduced DNA methylation at exon 2 of POLGA, and reduced hepatic gene expression. Depletion of mtDNA followed by subsequent differentiation did not increase mtDNA copy number, but reduced DNA methylation at exon 2 of POLGA and increased expression of hepatic and pluripotency genes. We encapsulated hAEC in barium alginate microcapsules and subsequently differentiated them into HLC. Encapsulation resulted in no net increase of mtDNA copy number but a significant reduction in DNA methylation of POLGA. RNAseq analysis showed that differentiated HLC express hepatocyte-specific genes but also increased expression of inflammatory interferon genes. Differentiation in encapsulated cells showed suppression of inflammatory genes as well as increased expression of genes associated with hepatocyte function pathways and networks. This study demonstrates that an increase in classical hepatic gene expression can be achieved in HLC through encapsulation, although they fail to effectively regulate mtDNA copy number.
Collapse
Affiliation(s)
- Vijesh Vaghjiani
- 1 Centre for Genetic Diseases, Hudson Institute of Medical Research , Clayton, Australia .,2 Department of Molecular and Translational Science, Monash University , Clayton, Australia
| | - Jason E Cain
- 2 Department of Molecular and Translational Science, Monash University , Clayton, Australia .,3 Centre for Cancer Research, Hudson Institute of Medical Research , Clayton, Australia
| | - William Lee
- 1 Centre for Genetic Diseases, Hudson Institute of Medical Research , Clayton, Australia .,2 Department of Molecular and Translational Science, Monash University , Clayton, Australia
| | - Vijayaganapathy Vaithilingam
- 4 Future Manufacturing Flagship, Commonwealth Scientific and Industrial Research Organisation , North Ryde, Australia
| | - Bernard E Tuch
- 4 Future Manufacturing Flagship, Commonwealth Scientific and Industrial Research Organisation , North Ryde, Australia .,5 School of Biomedical Science, Discipline Physiology, University of Sydney , Sydney, Australia
| | - Justin C St John
- 1 Centre for Genetic Diseases, Hudson Institute of Medical Research , Clayton, Australia .,2 Department of Molecular and Translational Science, Monash University , Clayton, Australia
| |
Collapse
|
29
|
Application of pharmacometrics and quantitative systems pharmacology to cancer therapy: The example of luminal a breast cancer. Pharmacol Res 2017; 124:20-33. [PMID: 28735000 DOI: 10.1016/j.phrs.2017.07.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 06/09/2017] [Accepted: 07/14/2017] [Indexed: 12/12/2022]
Abstract
Breast cancer (BC) is the most common cancer in women, and the second most frequent cause of cancer-related deaths in women worldwide. It is a heterogeneous disease composed of multiple subtypes with distinct morphologies and clinical implications. Quantitative systems pharmacology (QSP) is an emerging discipline bridging systems biology with pharmacokinetics (PK) and pharmacodynamics (PD) leveraging the systematic understanding of drugs' efficacy and toxicity. Despite numerous challenges in applying computational methodologies for QSP and mechanism-based PK/PD models to biological, physiological, and pharmacological data, bridging these disciplines has the potential to enhance our understanding of complex disease systems such as BC. In QSP/PK/PD models, various sources of data are combined including large, multi-scale experimental data such as -omics (i.e. genomics, transcriptomics, proteomics, and metabolomics), biomarkers (circulating and bound), PK, and PD endpoints. This offers a means for a translational application from pre-clinical mathematical models to patients, bridging the bench to bedside paradigm. Not only can these models be applied to inform and advance BC drug development, but they also could aid in optimizing combination therapies and rational dosing regimens for BC patients. Here, we review the current literature pertaining to the application of QSP and pharmacometrics-based pharmacotherapy in BC including bottom-up and top-down modeling approaches. Bottom-up modeling approaches employ mechanistic signal transduction pathways to predict the behavior of a biological system. The ones that are addressed in this review include signal transduction and homeostatic feedback modeling approaches. Alternatively, top-down modeling techniques are bioinformatics reconstruction techniques that infer static connections between molecules that make up a biological network and include (1) Bayesian networks, (2) co-expression networks, and (3) module-based approaches. This review also addresses novel techniques which utilize the principles of systems biology, synthetic lethality and tumor priming, both of which are discussed in relationship to novel drug targets and existing BC therapies. By utilizing QSP approaches, clinicians may develop a platform for improved dose individualization for subpopulation of BC patients, strengthen rationale in treatment designs, and explore mechanism elucidation for improving future treatments in BC medicine.
Collapse
|
30
|
Novielli C, Mandò C, Tabano S, Anelli GM, Fontana L, Antonazzo P, Miozzo M, Cetin I. Mitochondrial DNA content and methylation in fetal cord blood of pregnancies with placental insufficiency. Placenta 2017. [PMID: 28623975 DOI: 10.1016/j.placenta.2017.05.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Intrauterine growth restriction (IUGR) and preeclampsia (PE) are pregnancy disorders characterized by placental insufficiency with oxygen/nutrient restriction and oxidative stress, all influencing mitochondria functionality and number. Moreover, IUGR and PE fetuses are predisposed to diseases later in life, and this might occur through epigenetic alterations. Here we analyze content and methylation of mitochondrial DNA (mtDNA), for the first time in IUGR and PE singleton fetuses, to identify possible alterations in mtDNA levels and/or epigenetic control of mitochondrial loci relevant to replication (D-loop) and functionality (mt-TF/RNR1: protein synthesis, mt-CO1: respiratory chain complex). METHODS We studied 35 term and 8 preterm control, 31 IUGR, 17 PE/IUGR and 17 PE human singleton pregnancies with elective cesarean delivery. Fetal cord blood was collected and evaluated for biochemical parameters. Extracted DNA was subjected to Real-time PCR to assess mtDNA content and analyzed for D-loop, mt-TF/RNR1 and mt-CO1 methylation by bisulfite conversion and pyrosequencing. RESULTS mtDNA levels were increased in all pathologic groups compared to controls. Mitochondrial loci showed very low methylation levels in all samples; D-loop methylation was further decreased in the most severe cases and associated to umbilical vein pO2. mt-CO1 methylation levels inversely correlated to mtDNA content. DISCUSSION Increased mtDNA levels in IUGR, PE/IUGR and PE cord blood may denote a fetal response to placental insufficiency. Hypomethylation of D-loop, mt-TF/RNR1 and mt-CO1 loci confirms their relevance in pregnancy.
Collapse
Affiliation(s)
- Chiara Novielli
- "L. Sacco" Department of Biomedical and Clinical Sciences, Laboratory of Maternal-Fetal Translational Research "Giorgio Pardi", Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy
| | - Chiara Mandò
- "L. Sacco" Department of Biomedical and Clinical Sciences, Laboratory of Maternal-Fetal Translational Research "Giorgio Pardi", Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy
| | - Silvia Tabano
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Via F. Sforza 35, 20122 Milano, Italy
| | - Gaia M Anelli
- "L. Sacco" Department of Biomedical and Clinical Sciences, Laboratory of Maternal-Fetal Translational Research "Giorgio Pardi", Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy
| | - Laura Fontana
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Via F. Sforza 35, 20122 Milano, Italy
| | - Patrizio Antonazzo
- Department of Mother and Child, Luigi Sacco Hospital, via G.B. Grassi 74, 20157 Milano, Italy
| | - Monica Miozzo
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Via F. Sforza 35, 20122 Milano, Italy
| | - Irene Cetin
- "L. Sacco" Department of Biomedical and Clinical Sciences, Laboratory of Maternal-Fetal Translational Research "Giorgio Pardi", Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy; Department of Mother and Child, Luigi Sacco Hospital, via G.B. Grassi 74, 20157 Milano, Italy.
| |
Collapse
|
31
|
Williams MD, Xian L, Huso T, Park JJ, Huso D, Cope LM, Gang DR, Siems WF, Resar L, Reeves R, Hill HH. Fecal Metabolome in Hmga1 Transgenic Mice with Polyposis: Evidence for Potential Screen for Early Detection of Precursor Lesions in Colorectal Cancer. J Proteome Res 2016; 15:4176-4187. [PMID: 27696867 DOI: 10.1021/acs.jproteome.6b00035] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Because colorectal cancer (CRC) remains a leading cause of cancer mortality worldwide, more accessible screening tests are urgently needed to identify early stage lesions. We hypothesized that highly sensitive, metabolic profile analysis of stool samples will identify metabolites associated with early stage lesions and could serve as a noninvasive screening test. We therefore applied traveling wave ion mobility mass spectrometry (TWIMMS) coupled with ultraperformance liquid chromatography (UPLC) to investigate metabolic aberrations in stool samples in a transgenic model of premalignant polyposis aberrantly expressing the gene encoding the high mobility group A (Hmga1) chromatin remodeling protein. Here, we report for the first time that the fecal metabolome of Hmga1 mice is distinct from that of control mice and includes metabolites previously identified in human CRC. Significant alterations were observed in fatty acid metabolites and metabolites associated with bile acids (hypoxanthine xanthine, taurine) in Hmga1 mice compared to controls. Surprisingly, a marked increase in the levels of distinctive short, arginine-enriched, tetra-peptide fragments was observed in the transgenic mice. Together these findings suggest that specific metabolites are associated with Hmga1-induced polyposis and abnormal proliferation in intestinal epithelium. Although further studies are needed, these data provide a compelling rationale to develop fecal metabolomic analysis as a noninvasive screening tool to detect early precursor lesions to CRC in humans.
Collapse
Affiliation(s)
- Michael D Williams
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Lingling Xian
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Tait Huso
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Jeong-Jin Park
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - David Huso
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Leslie M Cope
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - David R Gang
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - William F Siems
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Linda Resar
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Raymond Reeves
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Herbert H Hill
- Department of Chemistry, ‡School of Molecular Biosciences, and §Institute of Biological Chemistry, Washington State University , Pullman, Washington 99164, United States.,Department of Medicine, ¶Department of Oncology, and ∥Institute for Cellular Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| |
Collapse
|
32
|
Requirement for the Mitochondrial Pyruvate Carrier in Mammalian Development Revealed by a Hypomorphic Allelic Series. Mol Cell Biol 2016; 36:2089-104. [PMID: 27215380 DOI: 10.1128/mcb.00166-16] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/13/2016] [Indexed: 11/20/2022] Open
Abstract
Glucose and oxygen are two of the most important molecules transferred from mother to fetus during eutherian pregnancy, and the metabolic fates of these nutrients converge at the transport and metabolism of pyruvate in mitochondria. Pyruvate enters the mitochondrial matrix through the mitochondrial pyruvate carrier (MPC), a complex in the inner mitochondrial membrane that consists of two essential components, MPC1 and MPC2. Here, we define the requirement for mitochondrial pyruvate metabolism during development with a progressive allelic series of Mpc1 deficiency in mouse. Mpc1 deletion was homozygous lethal in midgestation, but Mpc1 hypomorphs and tissue-specific deletion of Mpc1 presented as early perinatal lethality. The allelic series demonstrated that graded suppression of MPC resulted in dose-dependent metabolic and transcriptional changes. Steady-state metabolomics analysis of brain and liver from Mpc1 hypomorphic embryos identified compensatory changes in amino acid and lipid metabolism. Flux assays in Mpc1-deficient embryonic fibroblasts also reflected these changes, including a dramatic increase in mitochondrial alanine utilization. The mitochondrial alanine transaminase GPT2 was found to be necessary and sufficient for increased alanine flux upon MPC inhibition. These data show that impaired mitochondrial pyruvate transport results in biosynthetic deficiencies that can be mitigated in part by alternative anaplerotic substrates in utero.
Collapse
|
33
|
Hypoxic regulation of the expression of cell proliferation related genes in U87 glioma cells upon inhibition of IRE1 signaling enzyme. UKRAINIAN BIOCHEMICAL JOURNAL 2016; 88:11-21. [DOI: 10.15407/ubj88.01.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
34
|
Riabovol OO. IRE1 KNOCKDOWN MODIFIES THE GLUTAMINE AND GLUCOSE DEPRIVATION EFFECT ON THE EXPRESSION OF NUCLEAR GENES ENCODING MITOCHONDRIAL PROTEINS IN U87 GLIOMA CELLS. BIOTECHNOLOGIA ACTA 2016. [DOI: 10.15407/biotech9.02.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
35
|
IRE1 KNOCKDOWN MODIFIES GLUCOSE AND GLUTAMINE DEPRIVATION EFFECTS ON THE EXPRESSION OF PROLIFERATION RELATED GENES IN U87 GLIOMA CELLS. BIOTECHNOLOGIA ACTA 2016. [DOI: 10.15407/biotech9.01.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
36
|
Copley AL, Martinez-Outschoorn UE, Pestell RG, Sotgia F, Lisanti MP. [On the physiological roles of fibrinogen and fibrin]. Postepy Biochem 1968; 18:55. [PMID: 27220421 PMCID: PMC4879746 DOI: 10.1186/s13058-016-0712-6] [Citation(s) in RCA: 352] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 04/25/2016] [Indexed: 12/14/2022]
Abstract
Cancer is now viewed as a stem cell disease. There is still no consensus on the metabolic characteristics of cancer stem cells, with several studies indicating that they are mainly glycolytic and others pointing instead to mitochondrial metabolism as their principal source of energy. Cancer stem cells also seem to adapt their metabolism to microenvironmental changes by conveniently shifting energy production from one pathway to another, or by acquiring intermediate metabolic phenotypes. Determining the role of cancer stem cell metabolism in carcinogenesis has become a major focus in cancer research, and substantial efforts are conducted towards discovering clinical targets.
Collapse
Affiliation(s)
| | | | | | - Federica Sotgia
- The Breast Cancer Now Research Unit, Institute of Cancer Sciences, CRUK Manchester Institute, Paterson Building, University of Manchester, Manchester, UK. .,The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, CRUK Manchester Institute, Paterson Building, University of Manchester, Manchester, UK.
| | - Michael P Lisanti
- The Breast Cancer Now Research Unit, Institute of Cancer Sciences, CRUK Manchester Institute, Paterson Building, University of Manchester, Manchester, UK. .,The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, CRUK Manchester Institute, Paterson Building, University of Manchester, Manchester, UK.
| |
Collapse
|