301
|
Volobueva AS, Melnichenko AA, Grechko AV, Orekhov AN. Mitochondrial genome variability: the effect on cellular functional activity. Ther Clin Risk Manag 2018; 14:237-245. [PMID: 29467576 PMCID: PMC5811183 DOI: 10.2147/tcrm.s153895] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mitochondria are the key players in cell metabolism, calcium homeostasis, and reactive oxygen species (ROS) production. Mitochondrial genome alterations are reported to be associated with numerous human disorders affecting nearly all tissues. In this review, we discuss the available information on the involvement of mitochondrial DNA (mtDNA) mutations in cell dysfunction.
Collapse
Affiliation(s)
| | - Alexandra A Melnichenko
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow, Russia
| | - Andrey V Grechko
- Federal Scientific Clinical Center for Resuscitation and Rehabilitation, Moscow, Russia
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow, Russia.,Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| |
Collapse
|
302
|
Mitochondrial regulation of hematopoietic stem cells. Curr Opin Cell Biol 2018; 49:91-98. [PMID: 29309987 DOI: 10.1016/j.ceb.2017.12.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/12/2017] [Accepted: 12/16/2017] [Indexed: 12/16/2022]
Abstract
Hematopoietic stem cells (HSCs) preferentially use glycolysis rather than mitochondrial oxidative phosphorylation for energy production. While glycolysis in HSC is typically viewed as response to a hypoxic bone marrow environment that protects HSC from damaging reactive oxygen species, other interpretations are possible. Furthermore, recent evidence directly supports a critical role for mitochondria in the maintenance and function of HSCs that goes beyond ATP production. Here, we review recent advances in our understanding of metabolism and the role of mitochondria in the biology of HSCs.
Collapse
|
303
|
Mitohormesis, an Antiaging Paradigm. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 340:35-77. [DOI: 10.1016/bs.ircmb.2018.05.002] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
304
|
Jeong JS, Kim SR, Lee YC. Can Controlling Endoplasmic Reticulum Dysfunction Treat Allergic Inflammation in Severe Asthma With Fungal Sensitization? ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2018; 10:106-120. [PMID: 29411551 PMCID: PMC5809759 DOI: 10.4168/aair.2018.10.2.106] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 09/05/2017] [Accepted: 09/21/2017] [Indexed: 12/14/2022]
Abstract
Severe asthma is a heterogeneous disease entity to which diverse cellular components and pathogenetic mechanisms contribute. Current asthma therapies, including new biologic agents, are mainly targeting T helper type 2 cell-dominant inflammation, so that they are often unsatisfactory in the treatment of severe asthma. Respiratory fungal exposure has long been regarded as a precipitating factor for severe asthma phenotype. Moreover, as seen in clinical definitions of allergic bronchopulmonary aspergillosis (ABPA) and severe asthma with fungal sensitization (SAFS), fungal allergy-associated severe asthma phenotype is increasingly thought to have distinct pathobiologic mechanisms requiring different therapeutic approaches other than conventional treatment. However, there are still many unanswered questions on the direct causality of fungal sensitization in inducing severe allergic inflammation in SAFS. Recently, growing evidence suggests that stress response from the largest organelle, endoplasmic reticulum (ER), is closely interconnected to diverse cellular immune/inflammatory platforms, thereby being implicated in severe allergic lung inflammation. Interestingly, a recent study on this issue has suggested that ER stress responses and several associated molecular platforms, including phosphoinositide 3-kinase-δ and mitochondria, may be crucial players in the development of severe allergic inflammation in the SAFS. Defining emerging roles of ER and associated cellular platforms in SAFS may offer promising therapeutic options in the near future.
Collapse
Affiliation(s)
- Jae Seok Jeong
- Department of Internal Medicine, Research Center for Pulmonary Disorders, Chonbuk National University Medical School, Jeonju, Korea
| | - So Ri Kim
- Department of Internal Medicine, Research Center for Pulmonary Disorders, Chonbuk National University Medical School, Jeonju, Korea.,Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Medical School, Jeonju, Korea
| | - Yong Chul Lee
- Department of Internal Medicine, Research Center for Pulmonary Disorders, Chonbuk National University Medical School, Jeonju, Korea.,Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Medical School, Jeonju, Korea.
| |
Collapse
|
305
|
Lei Y, Liu L, Tang X, Yang D, Yang X, He F. Sanguinarine and chelerythrine: two natural products for mitochondria-imaging with aggregation-induced emission enhancement and pH-sensitive characteristics. RSC Adv 2018. [DOI: 10.1039/c7ra12920c] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
In this paper, two natural products: chelerythrine (Che) and sanguinarine (San) were systematically explored for aggregation-induced emission enhancement (AIEE) characteristics by a series of spectroscopic and theoretical experiments.
Collapse
Affiliation(s)
- Yaohui Lei
- Department of Medicinal Chemistry
- School of Pharmaceutical Science
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Liyan Liu
- Department of Medicinal Chemistry
- School of Pharmaceutical Science
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Xuemei Tang
- Department of Medicinal Chemistry
- School of Pharmaceutical Science
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Depo Yang
- Department of Medicinal Chemistry
- School of Pharmaceutical Science
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Xuhui Yang
- Assisted Reproductive Center
- Guangdong Women and Children's Hospital
- Guangzhou
- P. R. China
| | - Feng He
- Department of Medicinal Chemistry
- School of Pharmaceutical Science
- Sun Yat-sen University
- Guangzhou
- P. R. China
| |
Collapse
|
306
|
Abstract
Reactive oxygen species (ROS) mediate redox signaling necessary for numerous cellular functions. Yet, high levels of ROS in cells and tissues can cause damage and cell death. Therefore, regulation of redox homeostasis is essential for ROS-dependent signaling that does not incur cellular damage. Cells achieve this optimal balance by coordinating ROS production and elimination. In this Minireview, we discuss the mechanisms by which proliferating cancer and T cells maintain a carefully controlled redox balance. Greater insight into such redox biology may enable precisely targeted manipulation of ROS for effective medical therapies against cancer or immunological disorders.
Collapse
Affiliation(s)
- Hyewon Kong
- From the Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | - Navdeep S Chandel
- From the Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| |
Collapse
|
307
|
More than a powerplant: the influence of mitochondrial transfer on the epigenome. CURRENT OPINION IN PHYSIOLOGY 2017; 3:16-24. [PMID: 29750205 DOI: 10.1016/j.cophys.2017.11.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Each cell in the human body, with the exception of red blood cells, contains multiple copies of mitochondria that house their own genetic material, the maternally inherited mitochondrial DNA. Mitochondria are the cell's powerplant due to their massive ATP generation. However, the mitochondrion is also a hub for metabolite production from the TCA cycle, fatty acid beta-oxidation, and ketogenesis. In addition to producing macromolecules for biosynthetic reactions and cell replication, several mitochondrial intermediate metabolites serve as cofactors or substrates for epigenome modifying enzymes that regulate chromatin structure and impact gene expression. Here, we discuss connections between mitochondrial metabolites and enzymatic writers and erasers of chromatin modifications. We do this from the unique perspective of cell-to-cell mitochondrial transfer and its potential impact on mitochondrial replacement therapies.
Collapse
|
308
|
Frazier AE, Thorburn DR, Compton AG. Mitochondrial energy generation disorders: genes, mechanisms, and clues to pathology. J Biol Chem 2017; 294:5386-5395. [PMID: 29233888 DOI: 10.1074/jbc.r117.809194] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Inherited disorders of oxidative phosphorylation cause the clinically and genetically heterogeneous diseases known as mitochondrial energy generation disorders, or mitochondrial diseases. Over the last three decades, mutations causing these disorders have been identified in almost 290 genes, but many patients still remain without a molecular diagnosis. Moreover, while our knowledge of the genetic causes is continually expanding, our understanding into how these defects lead to cellular dysfunction and organ pathology is still incomplete. Here, we review recent developments in disease gene discovery, functional characterization, and shared pathogenic parameters influencing disease pathology that offer promising avenues toward the development of effective therapies.
Collapse
Affiliation(s)
- Ann E Frazier
- From the Murdoch Children's Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, and
| | - David R Thorburn
- From the Murdoch Children's Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, and.,Victorian Clinical Genetic Services, Royal Children's Hospital, Melbourne, Victoria 3052, Australia
| | - Alison G Compton
- From the Murdoch Children's Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, and
| |
Collapse
|
309
|
Weyandt JD, Thompson CB, Giaccia AJ, Rathmell WK. Metabolic Alterations in Cancer and Their Potential as Therapeutic Targets. AMERICAN SOCIETY OF CLINICAL ONCOLOGY EDUCATIONAL BOOK. AMERICAN SOCIETY OF CLINICAL ONCOLOGY. ANNUAL MEETING 2017. [PMID: 28561705 DOI: 10.14694/edbk_175561] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Otto Warburg's discovery in the 1920s that tumor cells took up more glucose and produced more lactate than normal cells provided the first clues that cancer cells reprogrammed their metabolism. For many years, however, it was unclear as to whether these metabolic alterations were a consequence of tumor growth or an adaptation that provided a survival advantage to these cells. In more recent years, interest in the metabolic differences in cancer cells has surged, as tumor proliferation and survival have been shown to be dependent upon these metabolic changes. In this educational review, we discuss some of the mechanisms that tumor cells use for reprogramming their metabolism to provide the energy and nutrients that they need for quick or sustained proliferation and discuss the potential for therapeutic targeting of these pathways to improve patient outcomes.
Collapse
Affiliation(s)
- Jamie D Weyandt
- From the Department of Medicine, Division of Hematology and Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN; Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Craig B Thompson
- From the Department of Medicine, Division of Hematology and Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN; Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Amato J Giaccia
- From the Department of Medicine, Division of Hematology and Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN; Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - W Kimryn Rathmell
- From the Department of Medicine, Division of Hematology and Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN; Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
310
|
Schito L, Rey S. Cell-Autonomous Metabolic Reprogramming in Hypoxia. Trends Cell Biol 2017; 28:128-142. [PMID: 29191366 DOI: 10.1016/j.tcb.2017.10.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 10/25/2017] [Accepted: 10/25/2017] [Indexed: 12/31/2022]
Abstract
Molecular oxygen (O2) is a universal electron acceptor that enables ATP synthesis through mitochondrial respiration in all metazoans. Consequently, hypoxia (low O2) has arisen as an organizing principle for cellular evolution, metabolism, and (patho)biology, eliciting a remarkable panoply of metabolic adaptations that trigger transcriptional, translational, post-translational, and epigenetic responses to determine cellular fitness. In this review we summarize current and emerging cell-autonomous molecular mechanisms that induce hypoxic metabolic reprogramming in health and disease.
Collapse
Affiliation(s)
- Luana Schito
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada.
| | - Sergio Rey
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada.
| |
Collapse
|
311
|
Zhao S, Torres A, Henry RA, Trefely S, Wallace M, Lee JV, Carrer A, Sengupta A, Campbell SL, Kuo YM, Frey AJ, Meurs N, Viola JM, Blair IA, Weljie AM, Metallo CM, Snyder NW, Andrews AJ, Wellen KE. ATP-Citrate Lyase Controls a Glucose-to-Acetate Metabolic Switch. Cell Rep 2017; 17:1037-1052. [PMID: 27760311 DOI: 10.1016/j.celrep.2016.09.069] [Citation(s) in RCA: 267] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 08/09/2016] [Accepted: 09/21/2016] [Indexed: 12/22/2022] Open
Abstract
Mechanisms of metabolic flexibility enable cells to survive under stressful conditions and can thwart therapeutic responses. Acetyl-coenzyme A (CoA) plays central roles in energy production, lipid metabolism, and epigenomic modifications. Here, we show that, upon genetic deletion of Acly, the gene coding for ATP-citrate lyase (ACLY), cells remain viable and proliferate, although at an impaired rate. In the absence of ACLY, cells upregulate ACSS2 and utilize exogenous acetate to provide acetyl-CoA for de novo lipogenesis (DNL) and histone acetylation. A physiological level of acetate is sufficient for cell viability and abundant acetyl-CoA production, although histone acetylation levels remain low in ACLY-deficient cells unless supplemented with high levels of acetate. ACLY-deficient adipocytes accumulate lipid in vivo, exhibit increased acetyl-CoA and malonyl-CoA production from acetate, and display some differences in fatty acid content and synthesis. Together, these data indicate that engagement of acetate metabolism is a crucial, although partial, mechanism of compensation for ACLY deficiency.
Collapse
Affiliation(s)
- Steven Zhao
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - AnnMarie Torres
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ryan A Henry
- Department of Cancer Biology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Sophie Trefely
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; A.J. Drexel Autism Institute, Drexel University, Philadelphia, PA 19104, USA
| | - Martina Wallace
- Department of Bioengineering and Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Joyce V Lee
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alessandro Carrer
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Arjun Sengupta
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sydney L Campbell
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yin-Ming Kuo
- Department of Cancer Biology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Alexander J Frey
- A.J. Drexel Autism Institute, Drexel University, Philadelphia, PA 19104, USA
| | - Noah Meurs
- Department of Bioengineering and Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - John M Viola
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ian A Blair
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aalim M Weljie
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christian M Metallo
- Department of Bioengineering and Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nathaniel W Snyder
- A.J. Drexel Autism Institute, Drexel University, Philadelphia, PA 19104, USA
| | - Andrew J Andrews
- Department of Cancer Biology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Kathryn E Wellen
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
312
|
Mitochondrial DNA density homeostasis accounts for a threshold effect in a cybrid model of a human mitochondrial disease. Biochem J 2017; 474:4019-4034. [PMID: 29079678 PMCID: PMC5705840 DOI: 10.1042/bcj20170651] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/24/2017] [Accepted: 10/27/2017] [Indexed: 12/16/2022]
Abstract
Mitochondrial dysfunction is involved in a wide array of devastating diseases, but the heterogeneity and complexity of the symptoms of these diseases challenges theoretical understanding of their causation. With the explosion of omics data, we have the unprecedented opportunity to gain deep understanding of the biochemical mechanisms of mitochondrial dysfunction. This goal raises the outstanding need to make these complex datasets interpretable. Quantitative modelling allows us to translate such datasets into intuition and suggest rational biomedical treatments. Taking an interdisciplinary approach, we use a recently published large-scale dataset and develop a descriptive and predictive mathematical model of progressive increase in mutant load of the MELAS 3243A>G mtDNA mutation. The experimentally observed behaviour is surprisingly rich, but we find that our simple, biophysically motivated model intuitively accounts for this heterogeneity and yields a wealth of biological predictions. Our findings suggest that cells attempt to maintain wild-type mtDNA density through cell volume reduction, and thus power demand reduction, until a minimum cell volume is reached. Thereafter, cells toggle from demand reduction to supply increase, up-regulating energy production pathways. Our analysis provides further evidence for the physiological significance of mtDNA density and emphasizes the need for performing single-cell volume measurements jointly with mtDNA quantification. We propose novel experiments to verify the hypotheses made here to further develop our understanding of the threshold effect and connect with rational choices for mtDNA disease therapies.
Collapse
|
313
|
Vazquez-Martin A, Van den Haute C, Cufí S, Corominas-Faja B, Cuyàs E, Lopez-Bonet E, Rodriguez-Gallego E, Fernández-Arroyo S, Joven J, Baekelandt V, Menendez JA. Mitophagy-driven mitochondrial rejuvenation regulates stem cell fate. Aging (Albany NY) 2017; 8:1330-52. [PMID: 27295498 PMCID: PMC4993334 DOI: 10.18632/aging.100976] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 05/30/2016] [Indexed: 12/12/2022]
Abstract
Our understanding on how selective mitochondrial autophagy, or mitophagy, can sustain the archetypal properties of stem cells is incomplete. PTEN-induced putative kinase 1 (PINK1) plays a key role in the maintenance of mitochondrial morphology and function and in the selective degradation of damaged mitochondria by mitophagy. Here, using embryonic fibroblasts from PINK1 gene-knockout (KO) mice, we evaluated whether mitophagy is a causal mechanism for the control of cell-fate plasticity and maintenance of pluripotency. Loss of PINK1-dependent mitophagy was sufficient to dramatically decrease the speed and efficiency of induced pluripotent stem cell (iPSC) reprogramming. Mitophagy-deficient iPSC colonies, which were characterized by a mixture of mature and immature mitochondria, seemed unstable, with a strong tendency to spontaneously differentiate and form heterogeneous populations of cells. Although mitophagy-deficient iPSC colonies normally expressed pluripotent markers, functional monitoring of cellular bioenergetics revealed an attenuated glycolysis in mitophagy-deficient iPSC cells. Targeted metabolomics showed a notable alteration in numerous glycolysis- and TCA-related metabolites in mitophagy-deficient iPSC cells, including a significant decrease in the intracellular levels of α-ketoglutarate -a key suppressor of the differentiation path in stem cells. Mitophagy-deficient iPSC colonies exhibited a notably reduced teratoma-initiating capacity, but fully retained their pluripotency and multi-germ layer differentiation capacity in vivo. PINK1-dependent mitophagy pathway is an important mitochondrial switch that determines the efficiency and quality of somatic reprogramming. Mitophagy-driven mitochondrial rejuvenation might contribute to the ability of iPSCs to suppress differentiation by directing bioenergetic transition and metabolome remodeling traits. These findings provide new insights into how mitophagy might influence the stem cell decisions to retain pluripotency or differentiate in tissue regeneration and aging, tumor growth, and regenerative medicine.
Collapse
Affiliation(s)
| | - Chris Van den Haute
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Katholieke Universiteit Leuven, Leuven, Flanders, Belgium
| | - Sílvia Cufí
- Josep Carreras Leukemia Research Institute, Stem Cell Lab, Barcelona, Spain
| | - Bruna Corominas-Faja
- Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Elisabet Cuyàs
- Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Eugeni Lopez-Bonet
- Department of Anatomical Pathology, Dr. Josep Trueta Hospital of Girona, Girona, Catalonia, Spain
| | - Esther Rodriguez-Gallego
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain
| | - Salvador Fernández-Arroyo
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Katholieke Universiteit Leuven, Leuven, Flanders, Belgium
| | - Javier A Menendez
- Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain.,ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain
| |
Collapse
|
314
|
Su G, Wang H, Gao Y, Chen G, Pei Y, Bai J. ¹H-NMR-Based Metabonomics of the Protective Effect of Coptis chinensis and Berberine on Cinnabar-Induced Hepatotoxicity and Nephrotoxicity in Rats. Molecules 2017; 22:molecules22111855. [PMID: 29099071 PMCID: PMC6150353 DOI: 10.3390/molecules22111855] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/15/2017] [Accepted: 10/26/2017] [Indexed: 02/06/2023] Open
Abstract
Coptis chinensis Franch has been used in Traditional Chinese Medicine (TCM) for treating infectious and inflammatory diseases for over two thousand years. Berberine (BN), an isoquinoline alkaloid, is the main component of Coptis chinensis. The pharmacological basis for its therapeutic effects, which include hepatoprotective effects on liver injuries, has been studied intensively, yet the therapy of liver injuries and underlying mechanism remain unclear. We investigated the detoxification mechanism of Coptis chinensis and berberine using metabolomics of urine and serum in the present study. After the treatment with Coptis chinensis and berberine, compared with the cinnabar group, Coptis chinensis and berberine can regulate the concentration of the endogenous metabolites. PLS-DA score plots demonstrated that the urine and serum metabolic profiles in rats of the Coptis chinensis and berberine groups were similar those of the control group, yet remarkably apart from the cinnabar group. The mechanism may be related to the endogenous metabolites including energy metabolism, amino acid metabolism and metabolism of intestinal flora in rats. Meanwhile, liver and kidney histopathology examinations and serum clinical chemistry analysis verified the experimental results of metabonomics.
Collapse
Affiliation(s)
- Guangyue Su
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Haifeng Wang
- Department of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China.
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Yuxian Gao
- Department of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Gang Chen
- Department of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China.
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Yuehu Pei
- Department of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China.
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Jiao Bai
- Department of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China.
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
315
|
Franchina DG, He F, Brenner D. Survival of the fittest: Cancer challenges T cell metabolism. Cancer Lett 2017; 412:216-223. [PMID: 29074426 DOI: 10.1016/j.canlet.2017.10.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 10/10/2017] [Accepted: 10/12/2017] [Indexed: 02/07/2023]
Abstract
T cells represent the major contributors to antitumor-specific immunity among the tumor-infiltrating lymphocytes. However, tumors acquire ways to evade immunosurveillance and anti-tumor responses are too weak to eradicate the disease. T cells are often functionally impaired as a result of interaction with, or signals from, transformed cells and the tumor microenvironment, including stromal cells. Among these, nutrients use and consumption is critically important for the control of differentiation and effector mechanisms of T cells. Moreover, Treg cells-skewing conditions often coexist within the cancer milieu, which sustains the notion of immune privileged tumors. Additionally, cancer cells contend with tumor infiltrating lymphocytes for nutrients and can outcompete the immune response. PD1- and CTLA-based immunotherapies partially remodel cell metabolism leading the way to clinical approaches of metabolic reprogramming for therapeutic purposes. Here we shortly discuss T cell fates during anti-tumor immune responses and how signals within tumor microenvironment influence T cell metabolism, altering functions and longevity of the cell.
Collapse
Affiliation(s)
- Davide G Franchina
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg
| | - Feng He
- Department of Infection and Immunity, Immune Systems Biology, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg
| | - Dirk Brenner
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis (ORCA), Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
316
|
Platelet mitochondrial dysfunction and the correlation with human diseases. Biochem Soc Trans 2017; 45:1213-1223. [PMID: 29054925 DOI: 10.1042/bst20170291] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/10/2017] [Accepted: 09/14/2017] [Indexed: 12/20/2022]
Abstract
The platelet is considered as an accessible and valuable tool to study mitochondrial function, owing to its greater content of fully functional mitochondria compared with other metabolically active organelles. Different lines of studies have demonstrated that mitochondria in platelets have function far more than thrombogenesis regulation, and beyond hemostasis, platelet mitochondrial dysfunction has also been used for studying mitochondrial-related diseases. In this review, the interplay between platelet mitochondrial dysfunction and oxidative stress, mitochondrial DNA lesions, electron transfer chain impairments, mitochondrial apoptosis and mitophagy has been outlined. Meanwhile, considerable efforts have been made towards understanding the role of platelet mitochondrial dysfunction in human diseases, such as diabetes mellitus, sepsis and neurodegenerative disorders. Alongside this, we have also articulated our perspectives on the development of potential biomarkers of platelet mitochondrial dysfunction in mitochondrial-related diseases.
Collapse
|
317
|
Packaging and transfer of mitochondrial DNA via exosomes regulate escape from dormancy in hormonal therapy-resistant breast cancer. Proc Natl Acad Sci U S A 2017; 114:E9066-E9075. [PMID: 29073103 PMCID: PMC5664494 DOI: 10.1073/pnas.1704862114] [Citation(s) in RCA: 462] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Increasing evidence suggests that extracellular vesicles (EVs) can transfer genetic material to recipient cells. However, the mechanism and role of this phenomenon are largely unknown. Here we have made a remarkable discovery: EVs can harbor the full mitochondrial genome. These extracellular vesicles can in turn transfer their mtDNA to cells with impaired metabolism, leading to restoration of metabolic activity. We determined that hormonal therapy induces oxidative phosphorylation-deficient breast cancer cells, which can be rescued via the transfer of mtDNA-laden extracellular vesicles. Horizontal transfer of mtDNA occurred in cancer stem-like cells and was associated with increased self-renewal potential of these cells, leading to resistance to hormonal therapy. We propose that mtDNA transfer occurs in human cancer via EVs. The horizontal transfer of mtDNA and its role in mediating resistance to therapy and an exit from dormancy have never been investigated. Here we identified the full mitochondrial genome in circulating extracellular vesicles (EVs) from patients with hormonal therapy-resistant (HTR) metastatic breast cancer. We generated xenograft models of HTR metastatic disease characterized by EVs in the peripheral circulation containing mtDNA. Moreover, these human HTR cells had acquired host-derived (murine) mtDNA promoting estrogen receptor-independent oxidative phosphorylation (OXPHOS). Functional studies identified cancer-associated fibroblast (CAF)-derived EVs (from patients and xenograft models) laden with whole genomic mtDNA as a mediator of this phenotype. Specifically, the treatment of hormone therapy (HT)-naive cells or HT-treated metabolically dormant populations with CAF-derived mtDNAhi EVs promoted an escape from metabolic quiescence and HTR disease both in vitro and in vivo. Moreover, this phenotype was associated with the acquisition of EV mtDNA, especially in cancer stem-like cells, expression of EV mtRNA, and restoration of OXPHOS. In summary, we have demonstrated that the horizontal transfer of mtDNA from EVs acts as an oncogenic signal promoting an exit from dormancy of therapy-induced cancer stem-like cells and leading to endocrine therapy resistance in OXPHOS-dependent breast cancer.
Collapse
|
318
|
Rabinovitch RC, Samborska B, Faubert B, Ma EH, Gravel SP, Andrzejewski S, Raissi TC, Pause A, St.-Pierre J, Jones RG. AMPK Maintains Cellular Metabolic Homeostasis through Regulation of Mitochondrial Reactive Oxygen Species. Cell Rep 2017; 21:1-9. [DOI: 10.1016/j.celrep.2017.09.026] [Citation(s) in RCA: 258] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 08/01/2017] [Accepted: 09/06/2017] [Indexed: 12/13/2022] Open
|
319
|
Montemurro C, Vadrevu S, Gurlo T, Butler AE, Vongbunyong KE, Petcherski A, Shirihai OS, Satin LS, Braas D, Butler PC, Tudzarova S. Cell cycle-related metabolism and mitochondrial dynamics in a replication-competent pancreatic beta-cell line. Cell Cycle 2017; 16:2086-2099. [PMID: 28820316 DOI: 10.1080/15384101.2017.1361069] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cell replication is a fundamental attribute of growth and repair in multicellular organisms. Pancreatic beta-cells in adults rarely enter cell cycle, hindering the capacity for regeneration in diabetes. Efforts to drive beta-cells into cell cycle have so far largely focused on regulatory molecules such as cyclins and cyclin-dependent kinases (CDKs). Investigations in cancer biology have uncovered that adaptive changes in metabolism, the mitochondrial network, and cellular Ca2+ are critical for permitting cells to progress through the cell cycle. Here, we investigated these parameters in the replication-competent beta-cell line INS 832/13. Cell cycle synchronization of this line permitted evaluation of cell metabolism, mitochondrial network, and cellular Ca2+ compartmentalization at key cell cycle stages. The mitochondrial network is interconnected and filamentous at G1/S but fragments during the S and G2/M phases, presumably to permit sorting to daughter cells. Pyruvate anaplerosis peaks at G1/S, consistent with generation of biomass for daughter cells, whereas mitochondrial Ca2+ and respiration increase during S and G2/M, consistent with increased energy requirements for DNA and lipid synthesis. This synchronization approach may be of value to investigators performing live cell imaging of Ca2+ or mitochondrial dynamics commonly undertaken in INS cell lines because without synchrony widely disparate data from cell to cell would be expected depending on position within cell cycle. Our findings also offer insight into why replicating beta-cells are relatively nonfunctional secreting insulin in response to glucose. They also provide guidance on metabolic requirements of beta-cells for the transition through the cell cycle that may complement the efforts currently restricted to manipulating cell cycle to drive beta-cells through cell cycle.
Collapse
Affiliation(s)
- Chiara Montemurro
- a Larry L. Hillblom Islet Research Center , University of California, Los Angeles, David Geffen School of Medicine , Los Angeles , CA , USA
| | - Suryakiran Vadrevu
- b Department of Pharmacology and Brehm Diabetes Research Center , University of Michigan , Ann Arbor , MI , USA
| | - Tatyana Gurlo
- a Larry L. Hillblom Islet Research Center , University of California, Los Angeles, David Geffen School of Medicine , Los Angeles , CA , USA
| | - Alexandra E Butler
- a Larry L. Hillblom Islet Research Center , University of California, Los Angeles, David Geffen School of Medicine , Los Angeles , CA , USA
| | - Kenny E Vongbunyong
- a Larry L. Hillblom Islet Research Center , University of California, Los Angeles, David Geffen School of Medicine , Los Angeles , CA , USA
| | - Anton Petcherski
- c Division of Endocrinology, Department of Medicine, David Geffen School of Medicine , University of California, Los Angeles , Los Angeles , CA , USA
| | - Orian S Shirihai
- c Division of Endocrinology, Department of Medicine, David Geffen School of Medicine , University of California, Los Angeles , Los Angeles , CA , USA
| | - Leslie S Satin
- b Department of Pharmacology and Brehm Diabetes Research Center , University of Michigan , Ann Arbor , MI , USA
| | - Daniel Braas
- d Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging , University of California, Los Angeles, David Geffen School of Medicine , Los Angeles , CA , USA ; UCLA Metabolomics Center , University of California, Los Angeles , Los Angeles , CA , USA
| | - Peter C Butler
- a Larry L. Hillblom Islet Research Center , University of California, Los Angeles, David Geffen School of Medicine , Los Angeles , CA , USA
| | - Slavica Tudzarova
- a Larry L. Hillblom Islet Research Center , University of California, Los Angeles, David Geffen School of Medicine , Los Angeles , CA , USA.,e Jonsson Comprehensive Cancer Center , University of California, Los Angeles, David Geffen School of Medicine , Los Angeles , CA , USA
| |
Collapse
|
320
|
Mathieu J, Ruohola-Baker H. Metabolic remodeling during the loss and acquisition of pluripotency. Development 2017; 144:541-551. [PMID: 28196802 DOI: 10.1242/dev.128389] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pluripotent cells from the early stages of embryonic development have the unlimited capacity to self-renew and undergo differentiation into all of the cell types of the adult organism. These properties are regulated by tightly controlled networks of gene expression, which in turn are governed by the availability of transcription factors and their interaction with the underlying epigenetic landscape. Recent data suggest that, perhaps unexpectedly, some key epigenetic marks, and thereby gene expression, are regulated by the levels of specific metabolites. Hence, cellular metabolism plays a vital role beyond simply the production of energy, and may be involved in the regulation of cell fate. In this Review, we discuss the metabolic changes that occur during the transitions between different pluripotent states both in vitro and in vivo, including during reprogramming to pluripotency and the onset of differentiation, and we discuss the extent to which distinct metabolites might regulate these transitions.
Collapse
Affiliation(s)
- Julie Mathieu
- Department of Biochemistry, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Hannele Ruohola-Baker
- Department of Biochemistry, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
321
|
Abstract
Due to their varied metabolic and signalling roles, mitochondria are important in mediating cell behaviour. By altering mitochondrial function, two studies now identify metabolite-induced epigenetic changes that have profound effects on haematopoietic stem cell fate and function.
Collapse
|
322
|
Lee WT, Sun X, Tsai TS, Johnson JL, Gould JA, Garama DJ, Gough DJ, McKenzie M, Trounce IA, St. John JC. Mitochondrial DNA haplotypes induce differential patterns of DNA methylation that result in differential chromosomal gene expression patterns. Cell Death Discov 2017; 3:17062. [PMID: 28900542 PMCID: PMC5592988 DOI: 10.1038/cddiscovery.2017.62] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 08/02/2017] [Indexed: 01/05/2023] Open
Abstract
Mitochondrial DNA copy number is strictly regulated during development as naive cells differentiate into mature cells to ensure that specific cell types have sufficient copies of mitochondrial DNA to perform their specialised functions. Mitochondrial DNA haplotypes are defined as specific regions of mitochondrial DNA that cluster with other mitochondrial sequences to show the phylogenetic origins of maternal lineages. Mitochondrial DNA haplotypes are associated with a range of phenotypes and disease. To understand how mitochondrial DNA haplotypes induce these characteristics, we used four embryonic stem cell lines that have the same set of chromosomes but possess different mitochondrial DNA haplotypes. We show that mitochondrial DNA haplotypes influence changes in chromosomal gene expression and affinity for nuclear-encoded mitochondrial DNA replication factors to modulate mitochondrial DNA copy number, two events that act synchronously during differentiation. Global DNA methylation analysis showed that each haplotype induces distinct DNA methylation patterns, which, when modulated by DNA demethylation agents, resulted in skewed gene expression patterns that highlight the effectiveness of the new DNA methylation patterns established by each haplotype. The haplotypes differentially regulate α-ketoglutarate, a metabolite from the TCA cycle that modulates the TET family of proteins, which catalyse the transition from 5-methylcytosine, indicative of DNA methylation, to 5-hydroxymethylcytosine, indicative of DNA demethylation. Our outcomes show that mitochondrial DNA haplotypes differentially modulate chromosomal gene expression patterns of naive and differentiating cells by establishing mitochondrial DNA haplotype-specific DNA methylation patterns.
Collapse
Affiliation(s)
- William T Lee
- Centre for Genetic Diseases, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria 3168, Australia
- Department of Molecular and Translational Science, Monash University, 27-31 Wright Street, Clayton, Victoria 3168, Australia
| | - Xin Sun
- Centre for Genetic Diseases, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria 3168, Australia
- Department of Molecular and Translational Science, Monash University, 27-31 Wright Street, Clayton, Victoria 3168, Australia
| | - Te-Sha Tsai
- Centre for Genetic Diseases, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria 3168, Australia
- Department of Molecular and Translational Science, Monash University, 27-31 Wright Street, Clayton, Victoria 3168, Australia
| | - Jacqueline L Johnson
- Centre for Genetic Diseases, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria 3168, Australia
- Department of Molecular and Translational Science, Monash University, 27-31 Wright Street, Clayton, Victoria 3168, Australia
| | - Jodee A Gould
- Medical Genomic Facility, Monash Health Translational Precinct, 27-31 Wright Street, Clayton, Victoria 3168, Australia
| | - Daniel J Garama
- Department of Molecular and Translational Science, Monash University, 27-31 Wright Street, Clayton, Victoria 3168, Australia
- Centre for Cancer Research, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria 3168, Australia
| | - Daniel J Gough
- Department of Molecular and Translational Science, Monash University, 27-31 Wright Street, Clayton, Victoria 3168, Australia
- Centre for Cancer Research, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria 3168, Australia
| | - Matthew McKenzie
- Centre for Genetic Diseases, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria 3168, Australia
- Department of Molecular and Translational Science, Monash University, 27-31 Wright Street, Clayton, Victoria 3168, Australia
| | - Ian A Trounce
- Centre for Eye Research Australia, Ophthalmology, University of Melbourne Department of Surgery, 32 Gisborne Street, East Melbourne, Victoria 3002, Australia
| | - Justin C St. John
- Centre for Genetic Diseases, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria 3168, Australia
- Department of Molecular and Translational Science, Monash University, 27-31 Wright Street, Clayton, Victoria 3168, Australia
| |
Collapse
|
323
|
Olenchock BA, Rathmell JC, Vander Heiden MG. Biochemical Underpinnings of Immune Cell Metabolic Phenotypes. Immunity 2017; 46:703-713. [PMID: 28514672 DOI: 10.1016/j.immuni.2017.04.013] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/23/2017] [Accepted: 04/25/2017] [Indexed: 02/06/2023]
Abstract
The metabolism of immune cells affects their function and influences host immunity. This review explores how immune cell metabolic phenotypes reflect biochemical dependencies and highlights evidence that both the metabolic state of immune cells and nutrient availability can alter immune responses. The central importance of oxygen, energetics, and redox homeostasis in immune cell metabolism, and how these factors are reflected in different metabolic phenotypes, is also discussed. Linking immune cell metabolic phenotype to effector functions is important to understand how altering metabolism can impact the way in which immune cells meet their metabolic demands and affect the immune response in various disease contexts.
Collapse
Affiliation(s)
- Benjamin A Olenchock
- Division of Cardiovascular Medicine, Department of Medicine, The Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA.
| | - Jeffrey C Rathmell
- Department of Pathology, Microbiology and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232-2363, USA
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Dana-Farber Cancer Institute, Boston, MA 02115, USA.
| |
Collapse
|
324
|
Abstract
Stem cell aging and exhaustion are considered important drivers of organismal aging. Age-associated declines in stem cell function are characterized by metabolic and epigenetic changes. Understanding the mechanisms underlying these changes will likely reveal novel therapeutic targets for ameliorating age-associated phenotypes and for prolonging human healthspan. Recent studies have shown that metabolism plays an important role in regulating epigenetic modifications and that this regulation dramatically affects the aging process. This review focuses on current knowledge regarding the mechanisms of stem cell aging, and the links between cellular metabolism and epigenetic regulation. In addition, we discuss how these interactions sense and respond to environmental stress in order to maintain stem cell homeostasis, and how environmental stimuli regulate stem cell function. Additionally, we highlight recent advances in the development of therapeutic strategies to rejuvenate dysfunctional aged stem cells.
Collapse
Affiliation(s)
- Ruotong Ren
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Alejandro Ocampo
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Guang-Hui Liu
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Brain Disorders, Beijing 100069, China.
| | - Juan Carlos Izpisua Belmonte
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
325
|
Schell JC, Wisidagama DR, Bensard C, Zhao H, Wei P, Tanner J, Flores A, Mohlman J, Sorensen LK, Earl CS, Olson KA, Miao R, Waller TC, Delker D, Kanth P, Jiang L, DeBerardinis RJ, Bronner MP, Li DY, Cox JE, Christofk HR, Lowry WE, Thummel CS, Rutter J. Control of intestinal stem cell function and proliferation by mitochondrial pyruvate metabolism. Nat Cell Biol 2017; 19:1027-1036. [PMID: 28812582 PMCID: PMC6137334 DOI: 10.1038/ncb3593] [Citation(s) in RCA: 220] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 07/18/2017] [Indexed: 02/06/2023]
Abstract
Most differentiated cells convert glucose to pyruvate in the cytosol through glycolysis, followed by pyruvate oxidation in the mitochondria. These processes are linked by the mitochondrial pyruvate carrier (MPC), which is required for efficient mitochondrial pyruvate uptake. In contrast, proliferative cells, including many cancer and stem cells, perform glycolysis robustly but limit fractional mitochondrial pyruvate oxidation. We sought to understand the role this transition from glycolysis to pyruvate oxidation plays in stem cell maintenance and differentiation. Loss of the MPC in Lgr5-EGFP-positive stem cells, or treatment of intestinal organoids with an MPC inhibitor, increases proliferation and expands the stem cell compartment. Similarly, genetic deletion of the MPC in Drosophila intestinal stem cells also increases proliferation, whereas MPC overexpression suppresses stem cell proliferation. These data demonstrate that limiting mitochondrial pyruvate metabolism is necessary and sufficient to maintain the proliferation of intestinal stem cells.
Collapse
Affiliation(s)
- John C. Schell
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Dona R. Wisidagama
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Claire Bensard
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Helong Zhao
- Department of Molecular Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Peng Wei
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Jason Tanner
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Aimee Flores
- Eli and Edythe Broad Center for Regenerative Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Jeffrey Mohlman
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Lise K. Sorensen
- Department of Molecular Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Christian S. Earl
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Kristofor A. Olson
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Ren Miao
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - T. Cameron Waller
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Don Delker
- Department of Molecular Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Priyanka Kanth
- Department of Molecular Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Lei Jiang
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
- Department of Molecular and Cellular Endocrinology, Beckman Research Institute at City of Hope, Duarte, CA 91010, USA
| | - Ralph J. DeBerardinis
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Mary P. Bronner
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Dean Y. Li
- Department of Molecular Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - James E. Cox
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Heather R. Christofk
- Department of Biological Chemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- Eli and Edythe Broad Center for Regenerative Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - William E. Lowry
- Eli and Edythe Broad Center for Regenerative Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Carl S. Thummel
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Jared Rutter
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- Department of Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| |
Collapse
|
326
|
Young A, Gardiner D, Brosnan ME, Brosnan JT, Mailloux RJ. Physiological levels of formate activate mitochondrial superoxide/hydrogen peroxide release from mouse liver mitochondria. FEBS Lett 2017; 591:2426-2438. [PMID: 28771687 DOI: 10.1002/1873-3468.12777] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 07/30/2017] [Accepted: 07/31/2017] [Indexed: 11/11/2022]
Abstract
Here, we found that formate, an essential one-carbon metabolite, activates superoxide (O2·-)/hydrogen peroxide (H2 O2 ) release from mitochondria. Sodium formate (30 μm) induces a significant increase in O2·-/H2 O2 production in liver mitochondria metabolizing pyruvate (50 μm). At concentrations deemed to be toxic, formate does not increase O2·-/H2 O2 production further. It was observed that the formate-mediated increase in O2·-/H2 O2 production is not associated with cytochrome c oxidase (COX) inhibition or changes in membrane potential and NAD(P)H levels. Sodium formate supplementation increases phosphorylating respiration without altering proton leaks. Finally, it was observed that the 2-oxoglutarate dehydrogenase (OGDH) inhibitors 3-methyl-2-oxovaleric acid (KMV) and CPI-613 inhibit the formate-induced increase in pyruvate-driven ROS production. The importance of these findings in one-carbon metabolism and physiology are discussed herein.
Collapse
Affiliation(s)
- Adrian Young
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Danielle Gardiner
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Margaret E Brosnan
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - John T Brosnan
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Ryan J Mailloux
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| |
Collapse
|
327
|
Ceco E, Weinberg SE, Chandel NS, Sznajder JI. Metabolism and Skeletal Muscle Homeostasis in Lung Disease. Am J Respir Cell Mol Biol 2017; 57:28-34. [PMID: 28085493 DOI: 10.1165/rcmb.2016-0355tr] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
There is increased awareness that patients with lung diseases develop muscle dysfunction. Muscle dysfunction is a major contributor to a decreased quality of life in patients with chronic pulmonary diseases. Furthermore, muscle dysfunction exacerbates lung disease outcome, as a decrease in muscle mass and function are associated with increased morbidity, often long after critical illness or lung disease has been resolved. As we are learning more about the role of metabolism in health and disease, we are appreciating more the direct role of metabolism in skeletal muscle homeostasis. Altered metabolism is associated with numerous skeletal muscle pathologies and, conversely, skeletal muscle diseases are associated with significant changes in metabolic pathways. In this review, we highlight the role of metabolism in the regulation of skeletal muscle homeostasis. Understanding the metabolic pathways that underlie skeletal muscle wasting is of significant clinical interest for critically ill patients as well as patients with chronic lung disease, in which proper skeletal muscle function is essential to disease outcome.
Collapse
Affiliation(s)
- Ermelinda Ceco
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, Illinois
| | - Samuel E Weinberg
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, Illinois
| | - Navdeep S Chandel
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, Illinois
| | - Jacob I Sznajder
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
328
|
Blajszczak C, Bonini MG. Mitochondria targeting by environmental stressors: Implications for redox cellular signaling. Toxicology 2017; 391:84-89. [PMID: 28750850 DOI: 10.1016/j.tox.2017.07.013] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 06/22/2017] [Accepted: 07/21/2017] [Indexed: 01/07/2023]
Abstract
Mitochondria are cellular powerhouses as well as metabolic and signaling hubs regulating diverse cellular functions, from basic physiology to phenotypic fate determination. It is widely accepted that reactive oxygen species (ROS) generated in mitochondria participate in the regulation of cellular signaling, and that some mitochondria chronically operate at a high ROS baseline. However, it is not completely understood how mitochondria adapt to persistently high ROS states and to environmental stressors that disturb the redox balance. Here we will review some of the current concepts regarding how mitochondria resist oxidative damage, how they are replaced when excessive oxidative damage compromises function, and the effect of environmental toxicants (i.e. heavy metals) on the regulation of mitochondrial ROS (mtROS) production and subsequent impact.
Collapse
Affiliation(s)
- Chuck Blajszczak
- Departments of Medicine and Pathology, University of Illinois College of Medicine at Chicago, IL, USA
| | - Marcelo G Bonini
- Departments of Medicine and Pathology, University of Illinois College of Medicine at Chicago, IL, USA.
| |
Collapse
|
329
|
Xue Z, Zhao H, Liu J, Han J, Han S. Defining Cancer Cell Bioenergetic Profiles Using a Dual Organelle-Oriented Chemosensor Responsive to pH Values and Electropotential Changes. Anal Chem 2017. [DOI: 10.1021/acs.analchem.7b01934] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Zhongwei Xue
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory for Physical Chemistry of Solid Surfaces, the Key Laboratory for Chemical Biology of Fujian Province, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Innovation Center for Cell Signaling Network, Xiamen University, Xiamen, 361005, China
| | - Hu Zhao
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory for Physical Chemistry of Solid Surfaces, the Key Laboratory for Chemical Biology of Fujian Province, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Innovation Center for Cell Signaling Network, Xiamen University, Xiamen, 361005, China
| | - Jian Liu
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory for Physical Chemistry of Solid Surfaces, the Key Laboratory for Chemical Biology of Fujian Province, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Innovation Center for Cell Signaling Network, Xiamen University, Xiamen, 361005, China
| | - Jiahuai Han
- State
key Laboratory of Cellular Stress Biology, Innovation Center for Cell
Signaling Network, School of Life Sciences, Xiamen University, Xiamen, 361005, China
| | - Shoufa Han
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory for Physical Chemistry of Solid Surfaces, the Key Laboratory for Chemical Biology of Fujian Province, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Innovation Center for Cell Signaling Network, Xiamen University, Xiamen, 361005, China
| |
Collapse
|
330
|
Quirós PM, Prado MA, Zamboni N, D'Amico D, Williams RW, Finley D, Gygi SP, Auwerx J. Multi-omics analysis identifies ATF4 as a key regulator of the mitochondrial stress response in mammals. J Cell Biol 2017; 216:2027-2045. [PMID: 28566324 PMCID: PMC5496626 DOI: 10.1083/jcb.201702058] [Citation(s) in RCA: 537] [Impact Index Per Article: 76.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/04/2017] [Accepted: 04/18/2017] [Indexed: 01/25/2023] Open
Abstract
Mitochondrial stress activates a mitonuclear response to safeguard and repair mitochondrial function and to adapt cellular metabolism to stress. Using a multiomics approach in mammalian cells treated with four types of mitochondrial stressors, we identify activating transcription factor 4 (ATF4) as the main regulator of the stress response. Surprisingly, canonical mitochondrial unfolded protein response genes mediated by ATF5 are not activated. Instead, ATF4 activates the expression of cytoprotective genes, which reprogram cellular metabolism through activation of the integrated stress response (ISR). Mitochondrial stress promotes a local proteostatic response by reducing mitochondrial ribosomal proteins, inhibiting mitochondrial translation, and coupling the activation of the ISR with the attenuation of mitochondrial function. Through a trans-expression quantitative trait locus analysis, we provide genetic evidence supporting a role for Fh1 in the control of Atf4 expression in mammals. Using gene expression data from mice and humans with mitochondrial diseases, we show that the ATF4 pathway is activated in vivo upon mitochondrial stress. Our data illustrate the value of a multiomics approach to characterize complex cellular networks and provide a versatile resource to identify new regulators of mitochondrial-related diseases.
Collapse
Affiliation(s)
- Pedro M Quirós
- Laboratory for Integrative and Systems Physiology, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Miguel A Prado
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Nicola Zamboni
- Department of Biology, Institute of Molecular Systems Biology, Eidgenössische Technische Hochschule Zürich, Zurich, Switzerland
| | - Davide D'Amico
- Laboratory for Integrative and Systems Physiology, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN
| | - Daniel Finley
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Johan Auwerx
- Laboratory for Integrative and Systems Physiology, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
331
|
Interplay between mitochondrial metabolism and oxidative stress in ischemic stroke: An epigenetic connection. Mol Cell Neurosci 2017; 82:176-194. [DOI: 10.1016/j.mcn.2017.05.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/26/2017] [Accepted: 05/24/2017] [Indexed: 12/18/2022] Open
|
332
|
Yuan Y, Zhang A, Qi J, Wang H, Liu X, Zhao M, Duan S, Huang Z, Zhang C, Wu L, Zhang B, Zhang A, Xing C. p53/Drp1-dependent mitochondrial fission mediates aldosterone-induced podocyte injury and mitochondrial dysfunction. Am J Physiol Renal Physiol 2017; 314:F798-F808. [PMID: 28659272 DOI: 10.1152/ajprenal.00055.2017] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Mitochondrial dysfunction is increasingly recognized as an important factor in glomerular diseases. Previous study has shown that mitochondrial fission contributed to mitochondrial dysfunction. However, the mechanism of mitochondrial fission on mitochondrial dysfunction in aldosterone-induced podocyte injury remains ambiguous. This study aimed to investigate the pathogenic effect of mitochondrial fission both in vivo and in vitro. In an animal model of aldosterone-induced nephropathy, inhibition of the mitochondrial fission protein dynamin-related protein 1 (Drp1) suppressed aldosterone-induced podocyte injury. In cultured podocytes, aldosterone dose dependently induced Drp1 expression. Knockdown of Drp1 inhibited aldosterone-induced mitochondrial fission, mitochondrial dysfunction, and podocyte apoptosis. Furthermore, aldosterone dose dependently induced p53 expression. Knockdown of p53 inhibited aldosterone-induced Drp1 expression, mitochondrial dysfunction, and podocyte apoptosis. These findings implicated that aldosterone induced mitochondrial dysfunction and podocyte injury mediated by p53/Drp1-dependent mitochondrial fission, which may provide opportunities for therapeutic intervention for podocyte injury.
Collapse
Affiliation(s)
- Yanggang Yuan
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University , Nanjing , China
| | - Aiqing Zhang
- Department of Pediatric Nephrology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing Medical University , Nanjing , China
| | - Jia Qi
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Hui Wang
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University , Nanjing , China
| | - Xi Liu
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University , Nanjing , China
| | - Min Zhao
- Department of Nephrology, Nanjing Children's Hospital, Nanjing Medical University , Nanjing , China
| | - Suyan Duan
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University , Nanjing , China
| | - Zhimin Huang
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University , Nanjing , China
| | - Chengning Zhang
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University , Nanjing , China
| | - Lin Wu
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University , Nanjing , China
| | - Bo Zhang
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University , Nanjing , China
| | - Aihua Zhang
- Department of Nephrology, Nanjing Children's Hospital, Nanjing Medical University , Nanjing , China
| | - Changying Xing
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University , Nanjing , China
| |
Collapse
|
333
|
Abstract
Background Cattle are bred for, amongst other factors, specific traits, including parasite resistance and adaptation to climate. However, the influence and inheritance of mitochondrial DNA (mtDNA) are not usually considered in breeding programmes. In this study, we analysed the mtDNA profiles of cattle from Victoria (VIC), southern Australia, which is a temperate climate, and the Northern Territory (NT), the northern part of Australia, which has a tropical climate, to determine if the mtDNA profiles of these cattle are indicative of breed and phenotype, and whether these profiles are appropriate for their environments. Results A phylogenetic tree of the full mtDNA sequences of different breeds of cattle, which were obtained from the NCBI database, showed that the mtDNA profiles of cattle do not always reflect their phenotype as some cattle with Bos taurus phenotypes had Bos indicus mtDNA, whilst some cattle with Bos indicus phenotypes had Bos taurus mtDNA. Using D-loop sequencing, we were able to contrast the phenotypes and mtDNA profiles from different species of cattle from the 2 distinct cattle breeding regions of Australia. We found that 67 of the 121 cattle with Bos indicus phenotypes from NT (55.4%) had Bos taurus mtDNA. In VIC, 92 of the 225 cattle with Bos taurus phenotypes (40.9%) possessed Bos indicus mtDNA. When focusing on oocytes from cattle with the Bos taurus phenotype in VIC, their respective oocytes with Bos indicus mtDNA had significantly lower levels of mtDNA copy number compared with oocytes possessing Bos taurus mtDNA (P < 0.01). However, embryos derived from oocytes with Bos indicus mtDNA had the same ability to develop to the blastocyst stage and the levels of mtDNA copy number in their blastocysts were similar to blastocysts derived from oocytes harbouring Bos taurus mtDNA. Nevertheless, oocytes originating from the Bos indicus phenotype exhibited lower developmental potential due to low mtDNA copy number when compared with oocytes from cattle with a Bos taurus phenotype. Conclusions The phenotype of cattle is not always related to their mtDNA profiles. MtDNA profiles should be considered for breeding programmes as they also influence phenotypic traits and reproductive capacity in terms of oocyte quality. Electronic supplementary material The online version of this article (doi:10.1186/s12863-017-0523-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kanokwan Srirattana
- Centre for Genetic Diseases, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, 3168, Australia
| | - Kieren McCosker
- Department of Primary Industry and Resources, Darwin, NT, 0800, Australia
| | - Tim Schatz
- Department of Primary Industry and Resources, Darwin, NT, 0800, Australia
| | - Justin C St John
- Centre for Genetic Diseases, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia. .,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, 3168, Australia.
| |
Collapse
|
334
|
Ward NP, Poff AM, Koutnik AP, D’Agostino DP. Complex I inhibition augments dichloroacetate cytotoxicity through enhancing oxidative stress in VM-M3 glioblastoma cells. PLoS One 2017. [PMID: 28644886 PMCID: PMC5482478 DOI: 10.1371/journal.pone.0180061] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The robust glycolytic metabolism of glioblastoma multiforme (GBM) has proven them susceptible to increases in oxidative metabolism induced by the pyruvate mimetic dichloroacetate (DCA). Recent reports demonstrate that the anti-diabetic drug metformin enhances the damaging oxidative stress associated with DCA treatment in cancer cells. We sought to elucidate the role of metformin's reported activity as a mitochondrial complex I inhibitor in the enhancement of DCA cytotoxicity in VM-M3 GBM cells. Metformin potentiated DCA-induced superoxide production, which was required for enhanced cytotoxicity towards VM-M3 cells observed with the combination. Similarly, rotenone enhanced oxidative stress resultant from DCA treatment and this too was required for the noted augmentation of cytotoxicity. Adenosine monophosphate kinase (AMPK) activation was not observed with the concentration of metformin required to enhance DCA activity. Moreover, addition of an activator of AMPK did not enhance DCA cytotoxicity, whereas an inhibitor of AMPK heightened the cytotoxicity of the combination. Our data indicate that metformin enhancement of DCA cytotoxicity is dependent on complex I inhibition. Particularly, that complex I inhibition cooperates with DCA-induction of glucose oxidation to enhance cytotoxic oxidative stress in VM-M3 GBM cells.
Collapse
Affiliation(s)
- Nathan P. Ward
- Department of Molecular Pharmacology & Physiology, University of South Florida, Tampa, FL, United States of America
| | - Angela M. Poff
- Department of Molecular Pharmacology & Physiology, University of South Florida, Tampa, FL, United States of America
| | - Andrew P. Koutnik
- Department of Molecular Pharmacology & Physiology, University of South Florida, Tampa, FL, United States of America
| | - Dominic P. D’Agostino
- Department of Molecular Pharmacology & Physiology, University of South Florida, Tampa, FL, United States of America
- * E-mail:
| |
Collapse
|
335
|
Sommer N, Hüttemann M, Pak O, Scheibe S, Knoepp F, Sinkler C, Malczyk M, Gierhardt M, Esfandiary A, Kraut S, Jonas F, Veith C, Aras S, Sydykov A, Alebrahimdehkordi N, Giehl K, Hecker M, Brandes RP, Seeger W, Grimminger F, Ghofrani HA, Schermuly RT, Grossman LI, Weissmann N. Mitochondrial Complex IV Subunit 4 Isoform 2 Is Essential for Acute Pulmonary Oxygen Sensing. Circ Res 2017; 121:424-438. [PMID: 28620066 DOI: 10.1161/circresaha.116.310482] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 12/14/2016] [Accepted: 06/14/2017] [Indexed: 12/17/2022]
Abstract
RATIONALE Acute pulmonary oxygen sensing is essential to avoid life-threatening hypoxemia via hypoxic pulmonary vasoconstriction (HPV) which matches perfusion to ventilation. Hypoxia-induced mitochondrial superoxide release has been suggested as a critical step in the signaling pathway underlying HPV. However, the identity of the primary oxygen sensor and the mechanism of superoxide release in acute hypoxia, as well as its relevance for chronic pulmonary oxygen sensing, remain unresolved. OBJECTIVES To investigate the role of the pulmonary-specific isoform 2 of subunit 4 of the mitochondrial complex IV (Cox4i2) and the subsequent mediators superoxide and hydrogen peroxide for pulmonary oxygen sensing and signaling. METHODS AND RESULTS Isolated ventilated and perfused lungs from Cox4i2-/- mice lacked acute HPV. In parallel, pulmonary arterial smooth muscle cells (PASMCs) from Cox4i2-/- mice showed no hypoxia-induced increase of intracellular calcium. Hypoxia-induced superoxide release which was detected by electron spin resonance spectroscopy in wild-type PASMCs was absent in Cox4i2-/- PASMCs and was dependent on cysteine residues of Cox4i2. HPV could be inhibited by mitochondrial superoxide inhibitors proving the functional relevance of superoxide release for HPV. Mitochondrial hyperpolarization, which can promote mitochondrial superoxide release, was detected during acute hypoxia in wild-type but not Cox4i2-/- PASMCs. Downstream signaling determined by patch-clamp measurements showed decreased hypoxia-induced cellular membrane depolarization in Cox4i2-/- PASMCs compared with wild-type PASMCs, which could be normalized by the application of hydrogen peroxide. In contrast, chronic hypoxia-induced pulmonary hypertension and pulmonary vascular remodeling were not or only slightly affected by Cox4i2 deficiency, respectively. CONCLUSIONS Cox4i2 is essential for acute but not chronic pulmonary oxygen sensing by triggering mitochondrial hyperpolarization and release of mitochondrial superoxide which, after conversion to hydrogen peroxide, contributes to cellular membrane depolarization and HPV. These findings provide a new model for oxygen-sensing processes in the lung and possibly also in other organs.
Collapse
Affiliation(s)
- Natascha Sommer
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Maik Hüttemann
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Oleg Pak
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Susan Scheibe
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Fenja Knoepp
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Christopher Sinkler
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Monika Malczyk
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Mareike Gierhardt
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Azadeh Esfandiary
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Simone Kraut
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Felix Jonas
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Christine Veith
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Siddhesh Aras
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Akylbek Sydykov
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Nasim Alebrahimdehkordi
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Klaudia Giehl
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Matthias Hecker
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Ralf P Brandes
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Werner Seeger
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Friedrich Grimminger
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Hossein A Ghofrani
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Ralph T Schermuly
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| | - Lawrence I Grossman
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.).
| | - Norbert Weissmann
- From the Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany (N.S., O.P., S.S., F.K., M.M., M.G., A.E., S.K., F.J., C.V., A.S., N.A., K.G., M.H., W.S., F.G., H.A.G., R.T.S., N.W.); Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., C.S., S.A., L.I.G.); Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany (R.P.B.); and Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (W.S.)
| |
Collapse
|
336
|
He H, Lai Y, Hao Y, Liu Y, Zhang Z, Liu X, Guo C, Zhang M, Zhou H, Wang N, Luo XG, Huo L, Ma W, Zhang TC. Selective p300 inhibitor C646 inhibited HPV E6-E7 genes, altered glucose metabolism and induced apoptosis in cervical cancer cells. Eur J Pharmacol 2017; 812:206-215. [PMID: 28619596 DOI: 10.1016/j.ejphar.2017.06.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 05/29/2017] [Accepted: 06/07/2017] [Indexed: 11/27/2022]
Abstract
High risk HPV infection is a causative factor of cervical cancer. The constitutive expression of HPV E6-E7 genes is important for the maintenance of cancer phenotypes. The cellular transcription co-activator p300 plays a crucial role in the regulation of HPV genes thus it was targeted for the inhibition of HPV-associated cervical cancer. In the present study, HPV positive cervical cells were treated with C646, a selective inhibitor of p300, to investigate its influence on HPV E6-E7 expression and cancer cell growth. Results of RT-qPCR, Western-blot and promoter activity assays showed that C646 inhibited the transcription of HPV E6-E7, which was accompanied with the accumulation of p53 protein. Meanwhile, cell proliferation was suppressed, glucose metabolism was disrupted and apoptosis was induced via the intrinsic pathway. Generally, the anti-cervical cancer potential of C646 was demonstrated and a novel mechanism was proposed in this study.
Collapse
Affiliation(s)
- Hongpeng He
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Yongwei Lai
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Yunpeng Hao
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Yupeng Liu
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Zijiang Zhang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Xiang Liu
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Chenhong Guo
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Mengmeng Zhang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Hao Zhou
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Nan Wang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Xue-Gang Luo
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Lihong Huo
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Wenjian Ma
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Tong-Cun Zhang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China; College of Life Sciences, Wuhan University of Science and Technology, Wuhan 430081, PR China.
| |
Collapse
|
337
|
Sodium butyrate protects against oxidative stress in HepG2 cells through modulating Nrf2 pathway and mitochondrial function. J Physiol Biochem 2017; 73:405-414. [DOI: 10.1007/s13105-017-0568-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 05/31/2017] [Indexed: 01/04/2023]
|
338
|
Abstract
Reactive oxygen species (ROS) are important signaling molecules that act through the oxidation of nucleic acids, proteins, and lipids. Several hallmarks of cancer, including uncontrolled proliferation, angiogenesis, and genomic instability, are promoted by the increased ROS levels commonly found in tumor cells. To counteract excessive ROS accumulation, oxidative stress, and death, cancer cells tightly regulate ROS levels by enhancing scavenging enzymes, which are dependent on the reducing cofactor nicotinamide adenine dinucleotide phosphate (NADPH). This review focuses on mitochondrial ROS homeostasis with a description of six pathways of NADPH production in mitochondria and a discussion of the possible strategies of pharmacological intervention to selectively eliminate cancer cells by increasing their ROS levels.
Collapse
Affiliation(s)
- Francesco Ciccarese
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Vincenzo Ciminale
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy.,Veneto Institute of Oncology - IRCCS, Padua, Italy
| |
Collapse
|
339
|
Rodrigues-Silva E, Siqueira-Santos ES, Ruas JS, Ignarro RS, Figueira TR, Rogério F, Castilho RF. Evaluation of mitochondrial respiratory function in highly glycolytic glioma cells reveals low ADP phosphorylation in relation to oxidative capacity. J Neurooncol 2017; 133:519-529. [PMID: 28540666 DOI: 10.1007/s11060-017-2482-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 05/14/2017] [Indexed: 01/25/2023]
Abstract
High-grade gliomas are aggressive and intensely glycolytic tumors. In the present study, we evaluated the mitochondrial respiratory function of glioma cells (T98G and U-87MG) and fresh human glioblastoma (GBM) tissue. To this end, measurements of oxygen consumption rate (OCR) were performed under various experimental conditions. The OCR of T98G and U-87MG cells was well coupled to ADP phosphorylation based on the ratio of ATP produced per oxygen consumed of ~2.5. In agreement, the basal OCR of GBM tissue was also partially associated with ADP phosphorylation. The basal respiration of intact T98G and U-87MG cells was not limited by the supply of endogenous substrates, as indicated by the increased OCR in response to a protonophore. These cells also displayed a high affinity for oxygen, as evidenced by the values of the partial pressure of oxygen when respiration is half maximal (p 50). In permeabilized glioma cells, ADP-stimulated OCR was only approximately 50% of that obtained in the presence of protonophore, revealing a significant limitation in oxidative phosphorylation (OXPHOS) relative to the activity of the electron transport system (ETS). This characteristic was maintained when the cells were grown under low glucose conditions. Flux control coefficient analyses demonstrated that the impaired OXPHOS was associated with the function of both mitochondrial ATP synthase and the adenine nucleotide translocator, but not the phosphate carrier. Altogether, these data indicate that the availability and metabolism of respiratory substrates and mitochondrial ETS are preserved in T98G and U-87MG glioma cells even though these cells possess a relatively restrained OXPHOS capability.
Collapse
Affiliation(s)
- Erika Rodrigues-Silva
- Departamento de Patologia Clínica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, 13083-887, Brazil
| | - Edilene S Siqueira-Santos
- Departamento de Patologia Clínica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, 13083-887, Brazil
| | - Juliana S Ruas
- Departamento de Patologia Clínica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, 13083-887, Brazil
| | - Raffaela S Ignarro
- Departamento de Anatomia Patológica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Tiago R Figueira
- Departamento de Patologia Clínica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, 13083-887, Brazil
| | - Fábio Rogério
- Departamento de Anatomia Patológica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Roger F Castilho
- Departamento de Patologia Clínica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, 13083-887, Brazil.
| |
Collapse
|
340
|
Su G, Chen G, An X, Wang H, Pei YH. Metabolic Profiling Analysis of the Alleviation Effect of Treatment with Baicalin on Cinnabar Induced Toxicity in Rats Urine and Serum. Front Pharmacol 2017; 8:271. [PMID: 28567014 PMCID: PMC5434134 DOI: 10.3389/fphar.2017.00271] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 05/01/2017] [Indexed: 01/23/2023] Open
Abstract
Objectives: Baicalin is the main bioactive flavonoid constituent isolated from Scutellaria baicalensis Georgi. The mechanisms of protection of liver remain unclear. In this study, 1H NMR-based metabonomics approach has been used to investigate the alleviation effect of Baicalin. Method:1H NMR metabolomics analyses of urine and serum from rats, was performed to illuminate the alleviation effect of Baicalin on mineral medicine (cinnabar)-induced liver and kidney toxicity. Results: The metabolic profiles of groups receiving Baicalin at a dose of 80 mg/kg were remarkably different from cinnabar, and meanwhile, the level of endogenous metabolites returned to normal compared to group cinnabar. PLS-DA scores plots demonstrated that the variation tendency of control and Baicalein are apart from Cinnabar. The metabolic profiles of group Baicalein were similar to those of group control. Statistics results were confirmed by the histopathological examination and biochemical assay. Conclusion: Baicalin have the alleviation effect to the liver and kidney damage induced by cinnabar. The Baicalin could regulate endogenous metabolites associated with the energy metabolism, choline metabolism, amino acid metabolism, and gut flora.
Collapse
Affiliation(s)
- Guangyue Su
- Department of Traditional Chinese Materia Medica, Shenyang Pharmaceutical UniversityShenyang, China.,Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical UniversityShenyang, China
| | - Gang Chen
- Department of Traditional Chinese Materia Medica, Shenyang Pharmaceutical UniversityShenyang, China.,Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical UniversityShenyang, China
| | - Xiao An
- Department of Traditional Chinese Materia Medica, Shenyang Pharmaceutical UniversityShenyang, China
| | - Haifeng Wang
- Department of Traditional Chinese Materia Medica, Shenyang Pharmaceutical UniversityShenyang, China.,Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical UniversityShenyang, China
| | - Yue-Hu Pei
- Department of Traditional Chinese Materia Medica, Shenyang Pharmaceutical UniversityShenyang, China.,Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical UniversityShenyang, China
| |
Collapse
|
341
|
The mitochondrial respiratory chain is essential for haematopoietic stem cell function. Nat Cell Biol 2017; 19:614-625. [PMID: 28504706 PMCID: PMC5474760 DOI: 10.1038/ncb3529] [Citation(s) in RCA: 218] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 04/10/2017] [Indexed: 12/12/2022]
Abstract
Adult and fetal hematopoietic stem cells (HSCs) display a glycolytic phenotype, which is required for maintenance of stemness; however, whether mitochondrial respiration is required to maintain HSC function is not known. Here we report that loss of the mitochondrial complex III subunit Rieske iron sulfur protein (RISP) in fetal mouse HSCs allows them to proliferate but impairs their differentiation, resulting in anemia and prenatal death. RISP null fetal HSCs displayed impaired respiration resulting in a decreased NAD+/NADH ratio. RISP null fetal HSCs and progenitors exhibited an increase in both DNA and histone methylation associated with increases in 2-hydroxyglutarate (2-HG), a metabolite known to inhibit DNA and histone demethylases. RISP inactivation in adult HSCs also impaired respiration resulting in loss of quiescence concomitant with severe pancytopenia and lethality. Thus, respiration is dispensable for adult or fetal HSC proliferation, but essential for fetal HSC differentiation and maintenance of adult HSC quiescence.
Collapse
|
342
|
McElroy GS, Chandel NS. Mitochondria control acute and chronic responses to hypoxia. Exp Cell Res 2017; 356:217-222. [PMID: 28327410 DOI: 10.1016/j.yexcr.2017.03.034] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 03/16/2017] [Indexed: 12/30/2022]
Abstract
There are numerous mechanisms by which mammals respond to hypoxia. These include acute changes in pulmonary arterial tone due to smooth muscle cell contraction, acute increases in respiration triggered by the carotid body chemosensory cells, and chronic changes such as induction of red blood cell proliferation and angiogenesis by hypoxia inducible factor targets erythropoietin and vascular endothelial growth factor, respectively. Mitochondria account for the majority of oxygen consumption in the cell and have recently been appreciated to serve as signaling organelles required for the initiation or propagation of numerous homeostatic mechanisms. Mitochondria can influence cell signaling by production of reactive oxygen species and metabolites. Here we review recent evidence that mitochondrial signals can imitate acute and chronic hypoxia responses.
Collapse
Affiliation(s)
- G S McElroy
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| | - N S Chandel
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States.
| |
Collapse
|
343
|
Matilainen O, Quirós PM, Auwerx J. Mitochondria and Epigenetics - Crosstalk in Homeostasis and Stress. Trends Cell Biol 2017; 27:453-463. [PMID: 28274652 DOI: 10.1016/j.tcb.2017.02.004] [Citation(s) in RCA: 227] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 02/07/2017] [Accepted: 02/12/2017] [Indexed: 12/22/2022]
Abstract
Through epigenetic mechanisms cells integrate environmental stimuli to fine-tune gene expression levels. Mitochondrial function is essential to provide the intermediate metabolites necessary to generate and modify epigenetic marks in the nucleus, which in turn can regulate the expression of mitochondrial proteins. In this review we summarize the function of mitochondria in the regulation of epigenetic mechanisms as a new aspect of mitonuclear communication. We focus in particular on the most common epigenetic modifications - histone acetylation and histone and DNA methylation. We also discuss the emerging field of mitochondrial DNA (mtDNA) methylation, whose physiological role remains unknown. Finally, we describe the essential role of some histone modifications in regulating the mitochondrial unfolded protein response (UPRmt) and the mitochondrial stress-dependent lifespan extension.
Collapse
Affiliation(s)
- Olli Matilainen
- Laboratory for Integrative and Systems Physiology, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Pedro M Quirós
- Laboratory for Integrative and Systems Physiology, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory for Integrative and Systems Physiology, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland.
| |
Collapse
|
344
|
Srinivasan S, Guha M, Kashina A, Avadhani NG. Mitochondrial dysfunction and mitochondrial dynamics-The cancer connection. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2017; 1858:602-614. [PMID: 28104365 DOI: 10.1016/j.bbabio.2017.01.004] [Citation(s) in RCA: 268] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 01/03/2017] [Accepted: 01/05/2017] [Indexed: 02/07/2023]
Abstract
Mitochondrial dysfunction is a hallmark of many diseases. The retrograde signaling initiated by dysfunctional mitochondria can bring about global changes in gene expression that alters cell morphology and function. Typically, this is attributed to disruption of important mitochondrial functions, such as ATP production, integration of metabolism, calcium homeostasis and regulation of apoptosis. Recent studies showed that in addition to these factors, mitochondrial dynamics might play an important role in stress signaling. Normal mitochondria are highly dynamic organelles whose size, shape and network are controlled by cell physiology. Defective mitochondrial dynamics play important roles in human diseases. Mitochondrial DNA defects and defective mitochondrial function have been reported in many cancers. Recent studies show that increased mitochondrial fission is a pro-tumorigenic phenotype. In this paper, we have explored the current understanding of the role of mitochondrial dynamics in pathologies. We present new data on mitochondrial dynamics and dysfunction to illustrate a causal link between mitochondrial DNA defects, excessive fission, mitochondrial retrograde signaling and cancer progression. This article is part of a Special Issue entitled Mitochondria in Cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux.
Collapse
Affiliation(s)
- Satish Srinivasan
- The Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, #189E, Philadelphia, PA 19104, United States
| | - Manti Guha
- The Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, #189E, Philadelphia, PA 19104, United States
| | - Anna Kashina
- The Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, #189E, Philadelphia, PA 19104, United States
| | - Narayan G Avadhani
- The Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, #189E, Philadelphia, PA 19104, United States.
| |
Collapse
|
345
|
Zhang Y, Avalos JL. Traditional and novel tools to probe the mitochondrial metabolism in health and disease. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2017; 9. [PMID: 28067471 DOI: 10.1002/wsbm.1373] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/07/2016] [Accepted: 11/09/2016] [Indexed: 02/06/2023]
Abstract
Mitochondrial metabolism links energy production to other essential cellular processes such as signaling, cellular differentiation, and apoptosis. In addition to producing adenosine triphosphate (ATP) as an energy source, mitochondria are responsible for the synthesis of a myriad of important metabolites and cofactors such as tetrahydrofolate, α-ketoacids, steroids, aminolevulinic acid, biotin, lipoic acid, acetyl-CoA, iron-sulfur clusters, heme, and ubiquinone. Furthermore, mitochondria and their metabolism have been implicated in aging and several human diseases, including inherited mitochondrial disorders, cardiac dysfunction, heart failure, neurodegenerative diseases, diabetes, and cancer. Therefore, there is great interest in understanding mitochondrial metabolism and the complex relationship it has with other cellular processes. A large number of studies on mitochondrial metabolism have been conducted in the last 50 years, taking a broad range of approaches. In this review, we summarize and discuss the most commonly used tools that have been used to study different aspects of the metabolism of mitochondria: ranging from dyes that monitor changes in the mitochondrial membrane potential and pharmacological tools to study respiration or ATP synthesis, to more modern tools such as genetically encoded biosensors and trans-omic approaches enabled by recent advances in mass spectrometry, computation, and other technologies. These tools have allowed the large number of studies that have shaped our current understanding of mitochondrial metabolism. WIREs Syst Biol Med 2017, 9:e1373. doi: 10.1002/wsbm.1373 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Yanfei Zhang
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | - José L Avalos
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA.,Andlinger Center for Energy and the Environment, Princeton University, Princeton, NJ, USA.,Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| |
Collapse
|
346
|
Weyandt JD, Thompson CB, Giaccia AJ, Rathmell WK. Metabolic Alterations in Cancer and Their Potential as Therapeutic Targets. Am Soc Clin Oncol Educ Book 2017; 37:825-832. [PMID: 28561705 PMCID: PMC5954416 DOI: 10.1200/edbk_175561] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Otto Warburg's discovery in the 1920s that tumor cells took up more glucose and produced more lactate than normal cells provided the first clues that cancer cells reprogrammed their metabolism. For many years, however, it was unclear as to whether these metabolic alterations were a consequence of tumor growth or an adaptation that provided a survival advantage to these cells. In more recent years, interest in the metabolic differences in cancer cells has surged, as tumor proliferation and survival have been shown to be dependent upon these metabolic changes. In this educational review, we discuss some of the mechanisms that tumor cells use for reprogramming their metabolism to provide the energy and nutrients that they need for quick or sustained proliferation and discuss the potential for therapeutic targeting of these pathways to improve patient outcomes.
Collapse
Affiliation(s)
- Jamie D. Weyandt
- Department of Medicine, Division of Hemtology and Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Craig B. Thompson
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York, 10065
| | - Amato J. Giaccia
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305
| | - W. Kimryn Rathmell
- Department of Medicine, Division of Hemtology and Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
347
|
Tumour microenvironment factors shaping the cancer metabolism landscape. Br J Cancer 2016; 116:277-286. [PMID: 28006817 PMCID: PMC5294476 DOI: 10.1038/bjc.2016.412] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 10/31/2016] [Accepted: 11/02/2016] [Indexed: 02/07/2023] Open
Abstract
Cancer cells exhibit metabolic alterations that distinguish them from healthy tissues and make their metabolic processes susceptible to pharmacological targeting. Although typical cell-autonomous features of cancer metabolism have been emerging, it is increasingly appreciated that extrinsic factors also influence the metabolic properties of tumours. This review highlights evidence from the recent literature to discuss how conditions within the tumour microenvironment shape the metabolic character of tumours.
Collapse
|
348
|
Mi X, Tang W, Chen X, Liu F, Tang X. Mitofusin 2 attenuates the histone acetylation at collagen IV promoter in diabetic nephropathy. J Mol Endocrinol 2016; 57:233-249. [PMID: 27997345 DOI: 10.1530/jme-16-0031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 10/03/2016] [Indexed: 12/15/2022]
Abstract
Extracellular matrix (ECM) increase in diabetic nephropathy (DN) is closely related to mitochondrial dysfunction. The mechanism of protective function of mitofusin 2 (Mfn2) for mitochondria remains largely unknown. In this study, the molecular mechanisms for the effect of Mfn2 on mitochondria and subsequent collagen IV expression in DN were investigated. Ras-binding-deficient mitofusin 2 (Mfn2-Ras(Δ)) were overexpressed in rat glomerular mesangial cells, and then the cells were detected for mitochondrial morphology, cellular reactive oxygen species (ROS), mRNA and protein expression of collagen IV with advanced glycation end-product (AGE) stimulation. Preliminary results reveal that the mitochondrial dysfunction and the increased synthesis of collagen IV after AGE stimulation were reverted by Mfn2-Ras(Δ) overexpression. Bioinformatical computations were performed to search transcriptional factor motifs in the promoter region of collagen IV. Three specific regions for TFAP2A binding were identified, followed by validation with chromatin immunoprecipitation experiments. Knocking down TFAP2A significantly decreased the TF binding in the first two regions and the gene expression of collagen IV. Furthermore, results reveal that Mfn2-Ras(Δ) overexpression significantly mitigated TFAP2A binding and also reverted the histone acetylation at Regions 1 and 2 after AGE stimulation. In streptozotocin-induced diabetic rats, Mfn2-Ras(Δ) overexpression also ameliorated glomerular mesangial lesions with decreased collagen IV expression, accompanied by decreased acetylation and TFAP2A binding at Region 1. In conclusion, this study highlights the pathway by which mitochondria affect the histone acetylation of gene promoter and provides a new potential therapy approach for DN.
Collapse
Affiliation(s)
- Xuhua Mi
- Division of NephrologyWest China Hospital, Sichuan University, Chengdu, China
| | - Wanxin Tang
- Division of NephrologyWest China Hospital, Sichuan University, Chengdu, China
| | - Xiaolei Chen
- Division of NephrologyWest China Hospital, Sichuan University, Chengdu, China
| | - Fei Liu
- Division of NephrologyWest China Hospital, Sichuan University, Chengdu, China
| | - Xiaohong Tang
- Division of NephrologyWest China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
349
|
Abstract
Alterations in the epigenome and metabolism both affect molecular rewiring in cancer cells and facilitate cancer development and progression. However, recent evidence suggests the existence of important bidirectional regulatory mechanisms between metabolic remodelling and the epigenome (specifically methylation and acetylation of histones) in cancer. Most chromatin-modifying enzymes require substrates or cofactors that are intermediates of cell metabolism. Such metabolites, and often the enzymes that produce them, can transfer into the nucleus, directly linking metabolism to nuclear transcription. We discuss how metabolic remodelling can contribute to tumour epigenetic alterations, thereby affecting cancer cell differentiation, proliferation and/or apoptosis, as well as therapeutic responses.
Collapse
Affiliation(s)
- Adam Kinnaird
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
- Division of Urology, Department of Surgery, University of Alberta, Edmonton, Alberta T6G 2R7, Canada
| | - Steven Zhao
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Kathryn E Wellen
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | |
Collapse
|
350
|
Cuyàs E, Fernández-Arroyo S, Joven J, Menendez JA. Metformin targets histone acetylation in cancer-prone epithelial cells. Cell Cycle 2016; 15:3355-3361. [PMID: 27792453 DOI: 10.1080/15384101.2016.1249547] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The usage of metabolic intermediates as substrates for chromatin-modifying enzymes provides a direct link between the metabolic state of the cell and epigenetics. Because this metabolism-epigenetics axis can regulate not only normal but also diseased states, it is reasonable to suggest that manipulating the epigenome via metabolic interventions may improve the clinical manifestation of age-related diseases including cancer. Using a model of BRCA1 haploinsufficiency-driven accelerated geroncogenesis, we recently tested the hypothesis that: 1.) metabolic rewiring of the mitochondrial biosynthetic nodes that overproduce epigenetic metabolites such as acetyl-CoA should promote cancer-related acetylation of histone H3 marks; 2.) metformin-induced restriction of mitochondrial biosynthetic capacity should manifest in the epigenetic regulation of histone acetylation. We now provide one of the first examples of how metformin-driven metabolic shifts such as reduction of the 2-carbon epigenetic substrate acetyl-CoA is sufficient to correct specific histone H3 acetylation marks in cancer-prone human epithelial cells. The ability of metformin to regulate mitonuclear communication and modulate the epigenetic landscape in genomically unstable pre-cancerous cells might guide the development of new metabolo-epigenetic strategies for cancer prevention and therapy.
Collapse
Affiliation(s)
- Elisabet Cuyàs
- a ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism & Cancer Group, Catalan Institute of Oncology , Girona , Catalonia , Spain.,b Girona Biomedical Research Institute (IDIBGI) , Girona , Catalonia , Spain
| | - Salvador Fernández-Arroyo
- c Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain, The Campus of International Excellence Southern Catalonia , Tarragona , Spain
| | - Jorge Joven
- c Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain, The Campus of International Excellence Southern Catalonia , Tarragona , Spain
| | - Javier A Menendez
- a ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism & Cancer Group, Catalan Institute of Oncology , Girona , Catalonia , Spain.,b Girona Biomedical Research Institute (IDIBGI) , Girona , Catalonia , Spain
| |
Collapse
|