1
|
Hossain T, Eckmann DM. Hyperoxic exposure alters intracellular bioenergetics distribution in human pulmonary cells. Life Sci 2023:121880. [PMID: 37356749 DOI: 10.1016/j.lfs.2023.121880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/24/2023] [Accepted: 06/22/2023] [Indexed: 06/27/2023]
Abstract
AIMS Pulmonary oxygen toxicity is caused by exposure to a high fraction of inspired oxygen, which damages multiple cell types within the lung. The cellular basis for pulmonary oxygen toxicity includes mitochondrial dysfunction. The aim of this study was to identify the effects of hyperoxic exposure on mitochondrial bioenergetic and dynamic functions in pulmonary cells. MAIN METHODS Mitochondrial respiration, inner membrane potential, dynamics (including motility), and distribution of mitochondrial bioenergetic capacity in two intracellular regions were quantified using cultured human lung microvascular endothelial cells, human pulmonary artery endothelial cells and A549 cells. Hyperoxic (95 % O2) exposures lasted 24, 48 and 72 h, durations relevant to mechanical ventilation in intensive care settings. KEY FINDINGS Mitochondrial motility was altered following all hyperoxic exposures utilized in experiments. Inhomogeneities in inner membrane potential and respiration parameters were present in each cell type following hyperoxia. The partitioning of ATP-linked respiration was also hyperoxia-duration and cell type dependent. Hyperoxic exposure lasting 48 h or longer provoked the largest alterations in mitochondrial motility and the greatest decreases in ATP-linked respiration, with a suggestion of decreases in respiration complex protein levels. SIGNIFICANCE Hyperoxic exposures of different durations produce intracellular inhomogeneities in mitochondrial dynamics and bioenergetics in pulmonary cells. Oxygen therapy is utilized commonly in clinical care and can induce undesirable decrements in bioenergy function needed to maintain pulmonary cell function and viability. There may be adjunctive or prophylactic measures that can be employed during hyperoxic exposures to prevent the mitochondrial dysfunction that signals the presence of oxygen toxcity.
Collapse
Affiliation(s)
- Tanvir Hossain
- Department of Anesthesiology, The Ohio State University, Columbus, OH 43210, United States of America
| | - David M Eckmann
- Department of Anesthesiology, The Ohio State University, Columbus, OH 43210, United States of America; Center for Medical and Engineering Innovation, The Ohio State University, Columbus, OH 43210, United States of America.
| |
Collapse
|
2
|
Chen PH, Tjong WY, Yang HC, Liu HY, Stern A, Chiu DTY. Glucose-6-Phosphate Dehydrogenase, Redox Homeostasis and Embryogenesis. Int J Mol Sci 2022; 23:ijms23042017. [PMID: 35216131 PMCID: PMC8878822 DOI: 10.3390/ijms23042017] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 12/04/2022] Open
Abstract
Normal embryogenesis requires complex regulation and precision, which depends on multiple mechanistic details. Defective embryogenesis can occur by various mechanisms. Maintaining redox homeostasis is of importance during embryogenesis. NADPH, as produced from the action of glucose-6-phosphate dehydrogenase (G6PD), has an important role in redox homeostasis, serving as a cofactor for glutathione reductase in the recycling of glutathione from oxidized glutathione and for NADPH oxidases and nitric oxide synthases in the generation of reactive oxygen (ROS) and nitrogen species (RNS). Oxidative stress differentially influences cell fate and embryogenesis. While low levels of stress (eustress) by ROS and RNS promote cell growth and differentiation, supra-physiological concentrations of ROS and RNS can lead to cell demise and embryonic lethality. G6PD-deficient cells and organisms have been used as models in embryogenesis for determining the role of redox signaling in regulating cell proliferation, differentiation and migration. Embryogenesis is also modulated by anti-oxidant enzymes, transcription factors, microRNAs, growth factors and signaling pathways, which are dependent on redox regulation. Crosstalk among transcription factors, microRNAs and redox signaling is essential for embryogenesis.
Collapse
Affiliation(s)
- Po-Hsiang Chen
- Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan; (P.-H.C.); (W.-Y.T.); (D.T.-Y.C.)
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan
| | - Wen-Ye Tjong
- Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan; (P.-H.C.); (W.-Y.T.); (D.T.-Y.C.)
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan
| | - Hung-Chi Yang
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu 30015, Taiwan
- Correspondence: ; Tel.: +886-3-6108175; Fax: +886-3-6102327
| | - Hui-Ya Liu
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Arnold Stern
- Grossman School of Medicine, New York University, New York, NY 10016, USA;
| | - Daniel Tsun-Yee Chiu
- Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan; (P.-H.C.); (W.-Y.T.); (D.T.-Y.C.)
| |
Collapse
|
3
|
Winship A, Donoghue J, Houston BJ, Martin JH, Lord T, Adwal A, Gonzalez M, Desroziers E, Ahmad G, Richani D, Bromfield EG. Reproductive health research in Australia and New Zealand: highlights from the Annual Meeting of the Society for Reproductive Biology, 2019. Reprod Fertil Dev 2021; 32:637-647. [PMID: 32234188 DOI: 10.1071/rd19449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 12/13/2019] [Indexed: 12/19/2022] Open
Abstract
The 2019 meeting of the Society for Reproductive Biology (SRB) provided a platform for the dissemination of new knowledge and innovations to improve reproductive health in humans, enhance animal breeding efficiency and understand the effect of the environment on reproductive processes. The effects of environment and lifestyle on fertility and animal behaviour are emerging as the most important modern issues facing reproductive health. Here, we summarise key highlights from recent work on endocrine-disrupting chemicals and diet- and lifestyle-induced metabolic changes and how these factors affect reproduction. This is particularly important to discuss in the context of potential effects on the reproductive potential that may be imparted to future generations of humans and animals. In addition to key summaries of new work in the male and female reproductive tract and on the health of the placenta, for the first time the SRB meeting included a workshop on endometriosis. This was an important opportunity for researchers, healthcare professionals and patient advocates to unite and provide critical updates on efforts to reduce the effect of this chronic disease and to improve the welfare of the women it affects. These new findings and directions are captured in this review.
Collapse
Affiliation(s)
- Amy Winship
- Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Stem Cells and Development Program, Monash University, Vic. 3800, Australia
| | - Jacqueline Donoghue
- The University of Melbourne, Department of Obstetrics and Gynaecology, Gynaecology Research Centre, Royal Women's Hospital, Parkville, Vic. 3052, Australia
| | - Brendan J Houston
- School of Biological Sciences, Monash University, Vic. 3800, Australia
| | - Jacinta H Martin
- Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW 2305, Australia
| | - Tessa Lord
- Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW 2305, Australia; and Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2300, Australia
| | - Alaknanda Adwal
- The University of Adelaide Robinson Research Institute, Adelaide Medical School, North Adelaide, SA 5005, Australia
| | - Macarena Gonzalez
- The University of Adelaide Robinson Research Institute, School of Medicine, Faculty of Health and Medical Sciences, Adelaide, SA 5005, Australia
| | - Elodie Desroziers
- Department of Physiology and Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
| | - Gulfam Ahmad
- The University of Sydney Medical School, Discipline of Pathology, School of Medical Sciences, Sydney, NSW 2006, Australia
| | - Dulama Richani
- School of Women's and Children's Health, Fertility and Research Centre, University of New South Wales, Sydney, NSW 2052 Australia
| | - Elizabeth G Bromfield
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2300, Australia; and Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Netherlands; and Corresponding author:
| |
Collapse
|
4
|
Tsogtbaatar E, Landin C, Minter-Dykhouse K, Folmes CDL. Energy Metabolism Regulates Stem Cell Pluripotency. Front Cell Dev Biol 2020; 8:87. [PMID: 32181250 PMCID: PMC7059177 DOI: 10.3389/fcell.2020.00087] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/31/2020] [Indexed: 12/19/2022] Open
Abstract
Pluripotent stem cells (PSCs) are characterized by their unique capacity for both unlimited self-renewal and their potential to differentiate to all cell lineages contained within the three primary germ layers. While once considered a distinct cellular state, it is becoming clear that pluripotency is in fact a continuum of cellular states, all capable of self-renewal and differentiation, yet with distinct metabolic, mitochondrial and epigenetic features dependent on gestational stage. In this review we focus on two of the most clearly defined states: “naïve” and “primed” PSCs. Like other rapidly dividing cells, PSCs have a high demand for anabolic precursors necessary to replicate their genome, cytoplasm and organelles, while concurrently consuming energy in the form of ATP. This requirement for both anabolic and catabolic processes sufficient to supply a highly adapted cell cycle in the context of reduced oxygen availability, distinguishes PSCs from their differentiated progeny. During early embryogenesis PSCs adapt their substrate preference to match the bioenergetic requirements of each specific developmental stage. This is reflected in different mitochondrial morphologies, membrane potentials, electron transport chain (ETC) compositions, and utilization of glycolysis. Additionally, metabolites produced in PSCs can directly influence epigenetic and transcriptional programs, which in turn can affect self-renewal characteristics. Thus, our understanding of the role of metabolism in PSC fate has expanded from anabolism and catabolism to include governance of the pluripotent epigenetic landscape. Understanding the roles of metabolism and the factors influencing metabolic pathways in naïve and primed pluripotent states provide a platform for understanding the drivers of cell fate during development. This review highlights the roles of the major metabolic pathways in the acquisition and maintenance of the different states of pluripotency.
Collapse
Affiliation(s)
- Enkhtuul Tsogtbaatar
- Stem Cell and Regenerative Metabolism Laboratory, Departments of Cardiovascular Diseases and Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, United States
| | - Chelsea Landin
- Stem Cell and Regenerative Metabolism Laboratory, Departments of Cardiovascular Diseases and Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, United States
| | - Katherine Minter-Dykhouse
- Stem Cell and Regenerative Metabolism Laboratory, Departments of Cardiovascular Diseases and Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, United States
| | - Clifford D L Folmes
- Stem Cell and Regenerative Metabolism Laboratory, Departments of Cardiovascular Diseases and Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, United States
| |
Collapse
|
5
|
Prieto J, Ponsoda X, Izpisua Belmonte JC, Torres J. Mitochondrial dynamics and metabolism in induced pluripotency. Exp Gerontol 2020; 133:110870. [PMID: 32045634 DOI: 10.1016/j.exger.2020.110870] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/20/2019] [Accepted: 02/05/2020] [Indexed: 12/15/2022]
Abstract
Somatic cells can be reprogrammed to pluripotency by either ectopic expression of defined factors or exposure to chemical cocktails. During reprogramming, somatic cells undergo dramatic changes in a wide range of cellular processes, such as metabolism, mitochondrial morphology and function, cell signaling pathways or immortalization. Regulation of these processes during cell reprograming lead to the acquisition of a pluripotent state, which enables indefinite propagation by symmetrical self-renewal without losing the ability of reprogrammed cells to differentiate into all cell types of the adult. In this review, recent data from different laboratories showing how these processes are controlled during the phenotypic transformation of a somatic cell into a pluripotent stem cell will be discussed.
Collapse
Affiliation(s)
- Javier Prieto
- Departamento Biología Celular, Biología Funcional y Antropología Física, Universitat de València, Calle Dr. Moliner 50, 46100 Burjassot, Spain; Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Xavier Ponsoda
- Departamento Biología Celular, Biología Funcional y Antropología Física, Universitat de València, Calle Dr. Moliner 50, 46100 Burjassot, Spain; Instituto de Investigación Sanitaria (INCLIVA), Avenida de Menéndez y Pelayo 4, 46010, Valencia, Spain
| | - Juan Carlos Izpisua Belmonte
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Josema Torres
- Departamento Biología Celular, Biología Funcional y Antropología Física, Universitat de València, Calle Dr. Moliner 50, 46100 Burjassot, Spain; Instituto de Investigación Sanitaria (INCLIVA), Avenida de Menéndez y Pelayo 4, 46010, Valencia, Spain.
| |
Collapse
|
6
|
Lees JG, Gardner DK, Harvey AJ. Nicotinamide adenine dinucleotide induces a bivalent metabolism and maintains pluripotency in human embryonic stem cells. Stem Cells 2020; 38:624-638. [PMID: 32003519 DOI: 10.1002/stem.3152] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 12/27/2019] [Indexed: 12/19/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD+ ) and its precursor metabolites are emerging as important regulators of both cell metabolism and cell state. Interestingly, the role of NAD+ in human embryonic stem cell (hESC) metabolism and the regulation of pluripotent cell state is unresolved. Here we show that NAD+ simultaneously increases hESC mitochondrial oxidative metabolism and partially suppresses glycolysis and stimulates amino acid turnover, doubling the consumption of glutamine. Concurrent with this metabolic remodeling, NAD+ increases hESC pluripotent marker expression and proliferation, inhibits BMP4-induced differentiation and reduces global histone 3 lysine 27 trimethylation, plausibly inducing an intermediate naïve-to-primed bivalent metabolism and pluripotent state. Furthermore, maintenance of NAD+ recycling via malate aspartate shuttle activity is identified as an absolute requirement for hESC self-renewal, responsible for 80% of the oxidative capacity of hESC mitochondria. Our findings implicate NAD+ in the regulation of cell state, suggesting that the hESC pluripotent state is dependent upon cellular NAD+ .
Collapse
Affiliation(s)
- Jarmon G Lees
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia.,O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,Department of Medicine at St Vincent's Hospital, Melbourne Medical School, The University of Melbourne, Fitzroy, Victoria, Australia
| | - David K Gardner
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Alexandra J Harvey
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
7
|
End-to-End Platform for Human Pluripotent Stem Cell Manufacturing. Int J Mol Sci 2019; 21:ijms21010089. [PMID: 31877727 PMCID: PMC6981419 DOI: 10.3390/ijms21010089] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/16/2019] [Accepted: 12/20/2019] [Indexed: 12/20/2022] Open
Abstract
Industrialization of stem-cell based therapies requires innovative solutions to close the gap between research and commercialization. Scalable cell production platforms are needed to reliably deliver the cell quantities needed during the various stages of development and commercial supply. Human pluripotent stem cells (hPSCs) are a key source material for generating therapeutic cell types. We have developed a closed, automated and scalable stirred tank bioreactor platform, capable of sustaining high fold expansion of hPSCs. Such a platform could facilitate the in-process monitoring and integration of online monitoring systems, leading to significantly reduced labor requirements and contamination risk. hPSCs are expanded in a controlled bioreactor using perfused xeno-free media. Cell harvest and concentration are performed in closed steps. The hPSCs can be cryopreserved to generate a bank of cells, or further processed as needed. Cryopreserved cells can be thawed into a two-dimensional (2D) tissue culture platform or a three-dimensional (3D) bioreactor to initiate a new expansion phase, or be differentiated to the clinically relevant cell type. The expanded hPSCs express hPSC-specific markers, have a normal karyotype and the ability to differentiate to the cells of the three germ layers. This end-to-end platform allows a large scale expansion of high quality hPSCs that can support the required cell demand for various clinical indications.
Collapse
|
8
|
Zhao DC, Li YM, Ma JL, Yi N, Yao ZY, Li YP, Quan Y, Li XN, Xu CL, Qiu Y, Wu LQ. Single-cell RNA sequencing reveals distinct gene expression patterns in glucose metabolism of human preimplantation embryos. Reprod Fertil Dev 2019; 31:237-247. [PMID: 30017025 DOI: 10.1071/rd18178] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 06/19/2018] [Indexed: 12/21/2022] Open
Abstract
Precise regulation of glucose metabolism-related genes is essential for early embryonic development. Although previous research has yielded detailed information on the biochemical processes, little is yet known of the dynamic gene expression profiles in glucose metabolism of preimplantation embryos at a single-cell resolution. In the present study, we performed integrated analysis of single-cell RNA sequencing (scRNA-seq) data of human preimplantation embryos that had been cultured in sequential medium. Different cells in the same embryo have similar gene expression patterns in glucose metabolism. During the switch from the cleavage to morula stage, the expression of glycolysis-related genes, such as glucose transporter genes (solute carrier family 2 (facilitated glucose transporter), member 1 (SLC2A1) and solute carrier family 2 (facilitated glucose transporter), member 3 (SLC2A3) and genes encoding hexokinase, phosphofructokinase, pyruvate kinase and lactate dehydrogenase, is increased. The genes involved in the pentose phosphate pathway are highly expressed at the cleavage stage, generating the reducing power to balance oxidative stress derived from biosynthesis. Expression of the genes involved in the biosynthesis of glycerophospholipids is increased after the morula stage. Nevertheless, the expression of tricarboxylic acid-related genes remains relatively unchanged during the preimplantation stages. In conclusion, we discovered that the gene expression profiles are dynamic according to glucose utilisation in the embryos at different stages, which contributes to our understanding of regulatory mechanisms of glucose metabolism-related genes in human preimplantation embryos.
Collapse
Affiliation(s)
- Di-Cheng Zhao
- The State Key Laboratory of Medical Genetics of China, Central South University, 72 Xiangya Road, Changsha, 410008, China
| | - Yu-Mei Li
- The Reproductive Medical Center of Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
| | - Jie-Liang Ma
- Translational Center for Stem Cell Research, Tongji Hospital, Department of Regenerative Medicine, Tongji University School of Medicine, 1239 Siping Road, Shanghai 200065, China
| | - Ning Yi
- Translational Center for Stem Cell Research, Tongji Hospital, Department of Regenerative Medicine, Tongji University School of Medicine, 1239 Siping Road, Shanghai 200065, China
| | - Zhong-Yuan Yao
- The State Key Laboratory of Medical Genetics of China, Central South University, 72 Xiangya Road, Changsha, 410008, China
| | - Yan-Ping Li
- The Reproductive Medical Center of Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
| | - Yi Quan
- The State Key Laboratory of Medical Genetics of China, Central South University, 72 Xiangya Road, Changsha, 410008, China
| | - Xin-Ning Li
- The State Key Laboratory of Medical Genetics of China, Central South University, 72 Xiangya Road, Changsha, 410008, China
| | - Chang-Long Xu
- The Reproductive Medical Center of Nanning Second People's Hospital, Guangxi Medical University, 13 Dancun Road, Nanning, 530031, China
| | - Ying Qiu
- The Reproductive Medical Center of Nanning Second People's Hospital, Guangxi Medical University, 13 Dancun Road, Nanning, 530031, China
| | - Ling-Qian Wu
- The State Key Laboratory of Medical Genetics of China, Central South University, 72 Xiangya Road, Changsha, 410008, China
| |
Collapse
|
9
|
Harvey A, Caretti G, Moresi V, Renzini A, Adamo S. Interplay between Metabolites and the Epigenome in Regulating Embryonic and Adult Stem Cell Potency and Maintenance. Stem Cell Reports 2019; 13:573-589. [PMID: 31597110 PMCID: PMC6830055 DOI: 10.1016/j.stemcr.2019.09.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/29/2019] [Accepted: 09/06/2019] [Indexed: 12/15/2022] Open
Abstract
The environment surrounding stem cells has the ability to elicit profound, heritable epigenetic changes orchestrated by multiple epigenetic mechanisms, which can be modulated by the level of specific metabolites. In this review, we highlight the significance of metabolism in regulating stem cell homeostasis, cell state, and differentiation capacity, using metabolic regulation of embryonic and adult muscle stem cells as examples, and cast light on the interaction between cellular metabolism and epigenetics. These new regulatory networks, based on the dynamic interplay between metabolism and epigenetics in stem cell biology, are important, not only for understanding tissue homeostasis, but to determine in vitro culture conditions which accurately support normal cell physiology.
Collapse
Affiliation(s)
- Alexandra Harvey
- School of BioSciences, University of Melbourne, Parkville, VIC 2010, Australia
| | - Giuseppina Caretti
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Viviana Moresi
- Department of Anatomy, Histology, Forensic Medicine & Orthopedics, Histology & Medical Embryology Section, Sapienza University of Rome and Interuniversity Institute of Myology, Rome, Italy.
| | - Alessandra Renzini
- Department of Anatomy, Histology, Forensic Medicine & Orthopedics, Histology & Medical Embryology Section, Sapienza University of Rome and Interuniversity Institute of Myology, Rome, Italy
| | - Sergio Adamo
- Department of Anatomy, Histology, Forensic Medicine & Orthopedics, Histology & Medical Embryology Section, Sapienza University of Rome and Interuniversity Institute of Myology, Rome, Italy
| |
Collapse
|
10
|
Spyrou J, Gardner DK, Harvey AJ. Metabolomic and Transcriptional Analyses Reveal Atmospheric Oxygen During Human Induced Pluripotent Stem Cell Generation Impairs Metabolic Reprogramming. Stem Cells 2019; 37:1042-1056. [PMID: 31042329 DOI: 10.1002/stem.3029] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 04/08/2019] [Accepted: 04/20/2019] [Indexed: 12/19/2022]
Abstract
The transition to pluripotency invokes profound metabolic restructuring; however, reprogramming is accompanied by the retention of somatic cell metabolic and epigenetic memory. Modulation of metabolism during reprogramming has been shown to improve reprogramming efficiency, yet it is not known how metabolite availability during reprogramming affects the physiology of resultant induced pluripotent stem cells (iPSCs). Metabolic analyses of iPSCs generated under either physiological (5%; P-iPSC) or atmospheric (20%; A-iPSC) oxygen conditions revealed that they retained aspects of somatic cell metabolic memory and failed to regulate carbohydrate metabolism with A-iPSC acquiring different metabolic characteristics. A-iPSC exhibited a higher mitochondrial membrane potential and were unable to modulate oxidative metabolism in response to oxygen challenge, contrasting with P-iPSC. RNA-seq analysis highlighted that A-iPSC displayed transcriptomic instability and a reduction in telomere length. Consequently, inappropriate modulation of metabolism by atmospheric oxygen during reprogramming significantly impacts the resultant A-iPSC metabolic and transcriptional landscape. Furthermore, retention of partial somatic metabolic memory in P-iPSC derived under physiological oxygen suggests that metabolic reprogramming remains incomplete. As the metabolome is a regulator of the epigenome, these observed perturbations of iPSC metabolism will plausibly have downstream effects on cellular function and physiology, both during and following differentiation, and highlight the need to optimize nutrient availability during the reprogramming process. Stem Cells 2019;37:1042-1056.
Collapse
Affiliation(s)
- James Spyrou
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia.,Stem Cells Australia, Melbourne, Victoria, Australia
| | - David K Gardner
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia.,Stem Cells Australia, Melbourne, Victoria, Australia
| | - Alexandra J Harvey
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia.,Stem Cells Australia, Melbourne, Victoria, Australia
| |
Collapse
|
11
|
Oxygen Regulates Human Pluripotent Stem Cell Metabolic Flux. Stem Cells Int 2019; 2019:8195614. [PMID: 31236115 PMCID: PMC6545818 DOI: 10.1155/2019/8195614] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 02/27/2019] [Indexed: 02/07/2023] Open
Abstract
Metabolism has been shown to alter cell fate in human pluripotent stem cells (hPSC). However, current understanding is almost exclusively based on work performed at 20% oxygen (air), with very few studies reporting on hPSC at physiological oxygen (5%). In this study, we integrated metabolic, transcriptomic, and epigenetic data to elucidate the impact of oxygen on hPSC. Using 13C-glucose labeling, we show that 5% oxygen increased the intracellular levels of glycolytic intermediates, glycogen, and the antioxidant response in hPSC. In contrast, 20% oxygen increased metabolite flux through the TCA cycle, activity of mitochondria, and ATP production. Acetylation of H3K9 and H3K27 was elevated at 5% oxygen while H3K27 trimethylation was decreased, conforming to a more open chromatin structure. RNA-seq analysis of 5% oxygen hPSC also indicated increases in glycolysis, lysine demethylases, and glucose-derived carbon metabolism, while increased methyltransferase and cell cycle activity was indicated at 20% oxygen. Our findings show that oxygen drives metabolite flux and specifies carbon fate in hPSC and, although the mechanism remains to be elucidated, oxygen was shown to alter methyltransferase and demethylase activity and the global epigenetic landscape.
Collapse
|
12
|
Spyrou J, Gardner DK, Harvey AJ. Metabolism Is a Key Regulator of Induced Pluripotent Stem Cell Reprogramming. Stem Cells Int 2019; 2019:7360121. [PMID: 31191682 PMCID: PMC6525803 DOI: 10.1155/2019/7360121] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/15/2019] [Accepted: 04/02/2019] [Indexed: 12/14/2022] Open
Abstract
Reprogramming to pluripotency involves drastic restructuring of both metabolism and the epigenome. However, induced pluripotent stem cells (iPSC) retain transcriptional memory, epigenetic memory, and metabolic memory from their somatic cells of origin and acquire aberrant characteristics distinct from either other pluripotent cells or parental cells, reflecting incomplete reprogramming. As a critical link between the microenvironment and regulation of the epigenome, nutrient availability likely plays a significant role in the retention of somatic cell memory by iPSC. Significantly, relative nutrient availability impacts iPSC reprogramming efficiency, epigenetic regulation and cell fate, and differentially alters their ability to respond to physiological stimuli. The significance of metabolites during the reprogramming process is central to further elucidating how iPSC retain somatic cell characteristics and optimising culture conditions to generate iPSC with physiological phenotypes to ensure their reliable use in basic research and clinical applications. This review serves to integrate studies on iPSC reprogramming, memory retention and metabolism, and identifies areas in which current knowledge is limited.
Collapse
Affiliation(s)
- James Spyrou
- School of BioSciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - David K. Gardner
- School of BioSciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Alexandra J. Harvey
- School of BioSciences, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
13
|
Relevance of Oxygen Concentration in Stem Cell Culture for Regenerative Medicine. Int J Mol Sci 2019; 20:ijms20051195. [PMID: 30857245 PMCID: PMC6429522 DOI: 10.3390/ijms20051195] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 02/28/2019] [Accepted: 03/04/2019] [Indexed: 01/10/2023] Open
Abstract
The key hallmark of stem cells is their ability to self-renew while keeping a differentiation potential. Intrinsic and extrinsic cell factors may contribute to a decline in these stem cell properties, and this is of the most importance when culturing them. One of these factors is oxygen concentration, which has been closely linked to the maintenance of stemness. The widely used environmental 21% O2 concentration represents a hyperoxic non-physiological condition, which can impair stem cell behaviour by many mechanisms. The goal of this review is to understand these mechanisms underlying the oxygen signalling pathways and their negatively-associated consequences. This may provide a rationale for culturing stem cells under physiological oxygen concentration for stem cell therapy success, in the field of tissue engineering and regenerative medicine.
Collapse
|
14
|
Tobias I, Isaac R, Dierolf J, Khazaee R, Cumming R, Betts D. Metabolic plasticity during transition to naïve-like pluripotency in canine embryo-derived stem cells. Stem Cell Res 2018; 30:22-33. [DOI: 10.1016/j.scr.2018.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/01/2018] [Accepted: 05/13/2018] [Indexed: 12/11/2022] Open
|
15
|
Harvey AJ, O’Brien C, Lambshead J, Sheedy JR, Rathjen J, Laslett AL, Gardner DK. Physiological oxygen culture reveals retention of metabolic memory in human induced pluripotent stem cells. PLoS One 2018; 13:e0193949. [PMID: 29543848 PMCID: PMC5854358 DOI: 10.1371/journal.pone.0193949] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 02/21/2018] [Indexed: 12/31/2022] Open
Abstract
Reprogramming somatic cells to a pluripotent cell state (induced Pluripotent Stem (iPS) cells) requires reprogramming of metabolism to support cell proliferation and pluripotency, most notably changes in carbohydrate turnover that reflect a shift from oxidative to glycolytic metabolism. Some aspects of iPS cell metabolism differ from embryonic stem (ES) cells, which may reflect a parental cell memory, or be a consequence of the reprogramming process. In this study, we compared the metabolism of 3 human iPS cell lines to assess the fidelity of metabolic reprogramming. When challenged with reduced oxygen concentration, ES cells have been shown to modulate carbohydrate use in a predictably way. In the same model, 2 of 3 iPS cell lines failed to regulate carbohydrate metabolism. Oxygen is a well-characterized regulator of cell function and embryo viability, and an inability of iPS cells to modulate metabolism in response to oxygen may indicate poor metabolic fidelity. As metabolism is linked to the regulation of the epigenome, assessment of metabolic responses of iPS cells to physiological stimuli during characterization is warranted to ensure complete cell reprogramming and as a measure of cell quality.
Collapse
Affiliation(s)
- Alexandra J. Harvey
- School of BioSciences, University of Melbourne, Parkville, Victoria, Australia
- ARC Special Research Initiative, Stem Cells Australia, Melbourne, Victoria, Australia
| | - Carmel O’Brien
- ARC Special Research Initiative, Stem Cells Australia, Melbourne, Victoria, Australia
- CSIRO Manufacturing, and Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Jack Lambshead
- ARC Special Research Initiative, Stem Cells Australia, Melbourne, Victoria, Australia
- CSIRO Manufacturing, and Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - John R. Sheedy
- School of BioSciences, University of Melbourne, Parkville, Victoria, Australia
| | - Joy Rathjen
- School of BioSciences, University of Melbourne, Parkville, Victoria, Australia
- ARC Special Research Initiative, Stem Cells Australia, Melbourne, Victoria, Australia
- School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Andrew L. Laslett
- School of BioSciences, University of Melbourne, Parkville, Victoria, Australia
- ARC Special Research Initiative, Stem Cells Australia, Melbourne, Victoria, Australia
- CSIRO Manufacturing, and Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - David K. Gardner
- School of BioSciences, University of Melbourne, Parkville, Victoria, Australia
- ARC Special Research Initiative, Stem Cells Australia, Melbourne, Victoria, Australia
- * E-mail:
| |
Collapse
|
16
|
Lees JG, Gardner DK, Harvey AJ. Mitochondrial and glycolytic remodeling during nascent neural differentiation of human pluripotent stem cells. Development 2018; 145:dev.168997. [DOI: 10.1242/dev.168997] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/18/2018] [Indexed: 12/29/2022]
Abstract
As human pluripotent stem cells (hPSC) exit pluripotency, they reportedly switch from glycolytic energy production to primarily mitochondrial metabolism. Here we show that upon ectoderm differentiation to neural precursor cells (NPC), hPSC increase glycolytic rate, ultimately producing more carbon as lactate than consumed as glucose. However, glucose, lactate, and pyruvate utilization decrease to half their PSC levels by the NPC stage, establishing a more quiescent metabolic state. Furthermore, we characterize a metabolic exit event within the first 24 hours of differentiation, plausibly necessary to transition hPSC out of the pluripotent state. Contrary to the current thinking, mitochondrial mass does not increase during NPC induction. Instead, mitochondrial DNA copies and mitochondrial activity decrease suggesting that mitochondrial metabolism either requires suppression, or is not required, for nascent ectoderm differentiation. Our work, therefore, contrasts with the dogma that the hPSC state is primarily glycolytic, transitioning to an oxidative metabolism upon the loss of the pluripotent state. Instead, we show that a heightened glycolytic metabolism is acquired, indicating that metabolic modulation of both glycolysis and mitochondrial metabolism occurs during exit from pluripotency in hPSC.
Collapse
Affiliation(s)
- Jarmon G. Lees
- School of BioSciences, University of Melbourne, Parkville 3010, Victoria, Australia
| | - David K. Gardner
- School of BioSciences, University of Melbourne, Parkville 3010, Victoria, Australia
| | - Alexandra J. Harvey
- School of BioSciences, University of Melbourne, Parkville 3010, Victoria, Australia
| |
Collapse
|
17
|
Pluripotent Stem Cell Metabolism and Mitochondria: Beyond ATP. Stem Cells Int 2017; 2017:2874283. [PMID: 28804500 PMCID: PMC5540363 DOI: 10.1155/2017/2874283] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 05/07/2017] [Indexed: 12/19/2022] Open
Abstract
Metabolism is central to embryonic stem cell (ESC) pluripotency and differentiation, with distinct profiles apparent under different nutrient milieu, and conditions that maintain alternate cell states. The significance of altered nutrient availability, particularly oxygen, and metabolic pathway activity has been highlighted by extensive studies of their impact on preimplantation embryo development, physiology, and viability. ESC similarly modulate their metabolism in response to altered metabolite levels, with changes in nutrient availability shown to have a lasting impact on derived cell identity through the regulation of the epigenetic landscape. Further, the preferential use of glucose and anaplerotic glutamine metabolism serves to not only support cell growth and proliferation but also minimise reactive oxygen species production. However, the perinuclear localisation of spherical, electron-poor mitochondria in ESC is proposed to sustain ESC nuclear-mitochondrial crosstalk and a mitochondrial-H2O2 presence, to facilitate signalling to support self-renewal through the stabilisation of HIFα, a process that may be favoured under physiological oxygen. The environment in which a cell is grown is therefore a critical regulator and determinant of cell fate, with metabolism, and particularly mitochondria, acting as an interface between the environment and the epigenome.
Collapse
|
18
|
Energy Metabolism Plays a Critical Role in Stem Cell Maintenance and Differentiation. Int J Mol Sci 2016; 17:253. [PMID: 26901195 PMCID: PMC4783982 DOI: 10.3390/ijms17020253] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 01/29/2016] [Accepted: 02/15/2016] [Indexed: 12/19/2022] Open
Abstract
Various stem cells gradually turned to be critical players in tissue engineering and regenerative medicine therapies. Current evidence has demonstrated that in addition to growth factors and the extracellular matrix, multiple metabolic pathways definitively provide important signals for stem cell self-renewal and differentiation. In this review, we mainly focus on a detailed overview of stem cell metabolism in vitro. In stem cell metabolic biology, the dynamic balance of each type of stem cell can vary according to the properties of each cell type, and they share some common points. Clearly defining the metabolic flux alterations in stem cells may help to shed light on stemness features and differentiation pathways that control the fate of stem cells.
Collapse
|
19
|
Nerenz RD. Omics in Reproductive Medicine: Application of Novel Technologies to Improve the IVF Success Rate. Adv Clin Chem 2016; 76:55-95. [PMID: 27645816 DOI: 10.1016/bs.acc.2016.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Treatment for many infertile couples often consists of in vitro fertilization (IVF) but an estimated 70% of IVF cycles fail to produce a live birth. In an attempt to improve the live birth rate, the vast majority of IVF cycles performed in the United States involve the transfer of multiple embryos, a practice that increases the risk of multiple gestation pregnancy. This is a concern because multiple gestation pregnancies are associated with an increased incidence of maternal and fetal complications and significant cost associated with the care of preterm infants. As the ideal outcome of each IVF cycle is the birth of a single healthy baby, significant effort has focused on identifying embryos with the greatest developmental potential. To date, selection of euploid embryos using comprehensive chromosome screening (CCS) is the most promising approach while metabolomic and proteomic assessment of spent culture medium have the potential to noninvasively assess embryo viability. Endometrial gene expression profiling may help determine the optimal time to perform embryo transfer. While CCS has been implemented in some clinics, further development and optimization will be required before analysis of spent culture medium and endometrial gene expression profiling make the transition to clinical use. This review will describe efforts to identify embryos with the greatest potential to result in a healthy, live birth, with a particular emphasis on detection of embryo aneuploidy and metabolic profiling of spent embryo culture medium. Assessment of endometrial receptivity to identify the optimal time to perform embryo transfer will also be discussed.
Collapse
Affiliation(s)
- R D Nerenz
- Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States.
| |
Collapse
|
20
|
Harvey AJ, Rathjen J, Gardner DK. Metaboloepigenetic Regulation of Pluripotent Stem Cells. Stem Cells Int 2015; 2016:1816525. [PMID: 26839556 PMCID: PMC4709785 DOI: 10.1155/2016/1816525] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 09/29/2015] [Indexed: 12/19/2022] Open
Abstract
The differentiation of pluripotent stem cells is associated with extensive changes in metabolism, as well as widespread remodeling of the epigenetic landscape. Epigenetic regulation is essential for the modulation of differentiation, being responsible for cell type specific gene expression patterns through the modification of DNA and histones, thereby establishing cell identity. Each cell type has its own idiosyncratic pattern regarding the use of specific metabolic pathways. Rather than simply being perceived as a means of generating ATP and building blocks for cell growth and division, cellular metabolism can directly influence cellular regulation and the epigenome. Consequently, the significance of nutrients and metabolites as regulators of differentiation is central to understanding how cells interact with their immediate environment. This review serves to integrate studies on pluripotent stem cell metabolism, and the regulation of DNA methylation and acetylation and identifies areas in which current knowledge is limited.
Collapse
Affiliation(s)
- Alexandra J. Harvey
- Stem Cells Australia, Parkville, VIC 3010, Australia
- School of BioSciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Joy Rathjen
- Stem Cells Australia, Parkville, VIC 3010, Australia
- School of BioSciences, The University of Melbourne, Parkville, VIC 3010, Australia
- School of Medicine, University of Tasmania, Hobart, TAS 7000, Australia
| | - David K. Gardner
- Stem Cells Australia, Parkville, VIC 3010, Australia
- School of BioSciences, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
21
|
Wale PL, Gardner DK. The effects of chemical and physical factors on mammalian embryo culture and their importance for the practice of assisted human reproduction. Hum Reprod Update 2015. [PMID: 26207016 DOI: 10.1093/humupd/dmv034] [Citation(s) in RCA: 174] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Although laboratory procedures, along with culture media formulations, have improved over the past two decades, the issue remains that human IVF is performed in vitro (literally 'in glass'). METHODS Using PubMed, electronic searches were performed using keywords from a list of chemical and physical factors with no limits placed on time. Examples of keywords include oxygen, ammonium, volatile organics, temperature, pH, oil overlays and incubation volume/embryo density. Available clinical and scientific evidence surrounding physical and chemical factors have been assessed and presented here. RESULTS AND CONCLUSIONS Development of the embryo outside the body means that it is constantly exposed to stresses that it would not experience in vivo. Sources of stress on the human embryo include identified factors such as pH and temperature shifts, exposure to atmospheric (20%) oxygen and the build-up of toxins in the media due to the static nature of culture. However, there are other sources of stress not typically considered, such as the act of pipetting itself, or the release of organic compounds from the very tissue culture ware upon which the embryo develops. Further, when more than one stress is present in the laboratory, there is evidence that negative synergies can result, culminating in significant trauma to the developing embryo. It is evident that embryos are sensitive to both chemical and physical signals within their microenvironment, and that these factors play a significant role in influencing development and events post transfer. From the viewpoint of assisted human reproduction, a major concern with chemical and physical factors lies in their adverse effects on the viability of embryos, and their long-term effects on the fetus, even as a result of a relatively brief exposure. This review presents data on the adverse effects of chemical and physical factors on mammalian embryos and the importance of identifying, and thereby minimizing, them in the practice of human IVF. Hence, optimizing the in vitro environment involves far more than improving culture media formulations.
Collapse
Affiliation(s)
- Petra L Wale
- School of BioSciences, University of Melbourne, Parkville, Victoria, Australia Melbourne IVF, Melbourne, Victoria, Australia
| | - David K Gardner
- School of BioSciences, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
22
|
Lees JG, Rathjen J, Sheedy JR, Gardner DK, Harvey AJ. Distinct profiles of human embryonic stem cell metabolism and mitochondria identified by oxygen. Reproduction 2015; 150:367-82. [PMID: 26159831 DOI: 10.1530/rep-14-0633] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 07/09/2015] [Indexed: 12/20/2022]
Abstract
Oxygen is a powerful regulator of cell function and embryonic development. It has previously been determined that oxygen regulates human embryonic stem (hES) cell glycolytic and amino acid metabolism, but the effects on mitochondria are as yet unknown. Two hES cell lines (MEL1, MEL2) were analyzed to determine the role of 5% (physiological) and 20% (atmospheric) oxygen in regulating mitochondrial activity. In response to extended physiological oxygen culture, MEL2 hES cells displayed reduced mtDNA content, mitochondrial mass and expression of metabolic genes TFAM, NRF1, PPARa and MT-ND4. Furthermore, MEL2 hES cell glucose consumption, lactate production and amino acid turnover were elevated under physiological oxygen. In stark contrast, MEL1 hES cell amino acid and carbohydrate use and mitochondrial function were relatively unaltered in response to oxygen. Furthermore, differentiation kinetics were delayed in the MEL1 hES cell line following BMP4 treatment. Here we report the first incidence of metabolic dysfunction in a hES cell population, defined as a failure to respond to oxygen concentration through the modulation of metabolism, demonstrating that hES cells can be perturbed during culture despite exhibiting the defining characteristics of pluripotent cells. Collectively, these data reveal a central role for oxygen in the regulation of hES cell metabolism and mitochondrial function, whereby physiological oxygen promotes glucose flux and suppresses mitochondrial biogenesis and gene expression.
Collapse
Affiliation(s)
- Jarmon G Lees
- School of BiosciencesUniversity of Melbourne, Parkville 3010, Victoria, AustraliaMenzies Institute of Medical ResearchUniversity of Tasmania, Hobart 7000, Tasmania, Australia
| | - Joy Rathjen
- School of BiosciencesUniversity of Melbourne, Parkville 3010, Victoria, AustraliaMenzies Institute of Medical ResearchUniversity of Tasmania, Hobart 7000, Tasmania, Australia School of BiosciencesUniversity of Melbourne, Parkville 3010, Victoria, AustraliaMenzies Institute of Medical ResearchUniversity of Tasmania, Hobart 7000, Tasmania, Australia
| | - John R Sheedy
- School of BiosciencesUniversity of Melbourne, Parkville 3010, Victoria, AustraliaMenzies Institute of Medical ResearchUniversity of Tasmania, Hobart 7000, Tasmania, Australia
| | - David K Gardner
- School of BiosciencesUniversity of Melbourne, Parkville 3010, Victoria, AustraliaMenzies Institute of Medical ResearchUniversity of Tasmania, Hobart 7000, Tasmania, Australia
| | - Alexandra J Harvey
- School of BiosciencesUniversity of Melbourne, Parkville 3010, Victoria, AustraliaMenzies Institute of Medical ResearchUniversity of Tasmania, Hobart 7000, Tasmania, Australia
| |
Collapse
|