Energy metabolism plasticity enables stemness programs

Engineering pluripotency through nuclear reprogramming and directing stem cells into defined lineages underscores cell fate plasticity. Acquisition of and departure from stemness are governed by genetic and epigenetic controllers, with modulation of energy metabolism and associated signaling increasingly implicated in cell identity determination. Transition from oxidative metabolism, typical of somatic tissues, into glycolysis is a prerequisite to fuel‐proficient reprogramming, directing a differentiated cytotype back to the pluripotent state. The glycolytic metabotype supports the anabolic and catabolic requirements of pluripotent cell homeostasis. Conversely, redirection of pluripotency into defined lineages requires mitochondrial biogenesis and maturation of efficient oxidative energy generation and distribution networks to match demands. The vital function of bioenergetics in regulating stemness and lineage specification implicates a broader role for metabolic reprogramming in cell fate decisions and determinations of tissue regenerative potential.

[1]  Timothy J. Nelson,et al.  Mitochondria in control of cell fate. , 2012, Circulation research.

[2]  Juan Carlos Izpisua Belmonte,et al.  The metabolome of induced pluripotent stem cells reveals metabolic changes occurring in somatic cell reprogramming , 2011, Cell Research.

[3]  Laurent Vergnes,et al.  UCP2 regulates energy metabolism and differentiation potential of human pluripotent stem cells , 2011, The EMBO journal.

[4]  A. Terzic,et al.  Energy metabolism in nuclear reprogramming. , 2011, Biomarkers in medicine.

[5]  A. Terzic,et al.  Cardiovascular Health: The Global Challenge , 2011, Clinical pharmacology and therapeutics.

[6]  E. Marbán,et al.  The Stuttering Progress of Cell Therapy for Heart Disease , 2011, Clinical pharmacology and therapeutics.

[7]  K. Thangaraj,et al.  Cellular Model of Warburg Effect Identifies Tumor Promoting Function of UCP2 in Breast Cancer and Its Suppression by Genipin , 2011, PloS one.

[8]  J. Drenckhahn Heart development: mitochondria in command of cardiomyocyte differentiation. , 2011, Developmental cell.

[9]  K. L. de Mesy Bentley,et al.  The permeability transition pore controls cardiac mitochondrial maturation and myocyte differentiation. , 2011, Developmental cell.

[10]  Hans Lehrach,et al.  Human Induced Pluripotent Stem Cells Harbor Homoplasmic and Heteroplasmic Mitochondrial DNA Mutations While Maintaining Human Embryonic Stem Cell–like Metabolic Reprogramming , 2011, Stem cells.

[11]  Andre Terzic,et al.  Somatic oxidative bioenergetics transitions into pluripotency-dependent glycolysis to facilitate nuclear reprogramming. , 2011, Cell metabolism.

[12]  J. I. Izpisúa Belmonte,et al.  Anaerobicizing into pluripotency. , 2011, Cell metabolism.

[13]  A. Terzic,et al.  Regenerative medicine: on the vanguard of health care. , 2011, Mayo Clinic proceedings.

[14]  K. McCreath,et al.  Mitochondria Determine the Differentiation Potential of Cardiac Mesoangioblasts , 2011, Stem cells.

[15]  G. Schatten,et al.  Energy Metabolism in Human Pluripotent Stem Cells and Their Differentiated Counterparts , 2011, PloS one.

[16]  Andre Terzic,et al.  Chronic Diseases: The Emerging Pandemic , 2011, Clinical and translational science.

[17]  A. Nagy,et al.  Human Induced Pluripotent Stem Cells: The Past, Present, and Future , 2011, Clinical pharmacology and therapeutics.

[18]  Timothy J. Nelson,et al.  Induced Pluripotent Stem Cells: An Emerging Theranostics Platform , 2011, Clinical pharmacology and therapeutics.

[19]  S. Yamanaka,et al.  The Use of Induced Pluripotent Stem Cells in Drug Development , 2011, Clinical pharmacology and therapeutics.

[20]  A. Terzic,et al.  Developmental Enhancement of Adenylate Kinase-AMPK Metabolic Signaling Axis Supports Stem Cell Cardiac Differentiation , 2011, PloS one.

[21]  Michael D. Schneider,et al.  Cardiac muscle regeneration: lessons from development. , 2011, Genes & development.

[22]  J. C. Belmonte,et al.  Dedifferentiation, transdifferentiation and reprogramming: three routes to regeneration , 2011, Nature Reviews Molecular Cell Biology.

[23]  Aristide C. Chikando,et al.  Mitofusin-2 Maintains Mitochondrial Structure and Contributes to Stress-Induced Permeability Transition in Cardiac Myocytes , 2011, Molecular and Cellular Biology.

[24]  J. Adjaye,et al.  Modulation of mitochondrial biogenesis and bioenergetic metabolism upon in vitro and in vivo differentiation of human ES and iPS cells. , 2010, The International journal of developmental biology.

[25]  Sheng Ding,et al.  Reprogramming of human primary somatic cells by OCT4 and chemical compounds. , 2010, Cell stem cell.

[26]  Andre Terzic,et al.  Induced pluripotent stem cells: developmental biology to regenerative medicine , 2010, Nature Reviews Cardiology.

[27]  Mark Ellisman,et al.  Mitochondrial Rejuvenation After Induced Pluripotency , 2010, PloS one.

[28]  J. Rehman Empowering self-renewal and differentiation: the role of mitochondria in stem cells , 2010, Journal of Molecular Medicine.

[29]  A. Terzic,et al.  Guided cardiopoiesis enhances therapeutic benefit of bone marrow human mesenchymal stem cells in chronic myocardial infarction. , 2010, Journal of the American College of Cardiology.

[30]  E. Marbán,et al.  Boot camp for mesenchymal stem cells. , 2010, Journal of the American College of Cardiology.

[31]  Alfredo Quiñones-Hinojosa,et al.  Oxygen in stem cell biology: a critical component of the stem cell niche. , 2010, Cell stem cell.

[32]  K. McCreath,et al.  Mitochondrial Reactive Oxygen Species Mediate Cardiomyocyte Formation from Embryonic Stem Cells in High Glucose , 2010, Stem cells.

[33]  H. Blau,et al.  Nuclear reprogramming to a pluripotent state by three approaches , 2010, Nature.

[34]  A. Terzic,et al.  Regenerative medicine advancing health care 2020. , 2010, Journal of the American College of Cardiology.

[35]  H. Schöler,et al.  Induced pluripotent stem cells at nanoscale. , 2010, Stem cells and development.

[36]  A. Terzic,et al.  Cells as biologics for cardiac repair in ischaemic heart failure , 2010, Heart.

[37]  D. K. Arrell,et al.  Glycolytic network restructuring integral to the energetics of embryonic stem cell cardiac differentiation. , 2010, Journal of molecular and cellular cardiology.

[38]  M. Stojkovic,et al.  Human Induced Pluripotent Stem Cell Lines Show Stress Defense Mechanisms and Mitochondrial Regulation Similar to Those of Human Embryonic Stem Cells , 2010, Stem cells.

[39]  Sunia A Trauger,et al.  Metabolic oxidation regulates embryonic stem cell differentiation , 2010, Nature chemical biology.

[40]  Hans Lehrach,et al.  The Senescence‐Related Mitochondrial/Oxidative Stress Pathway is Repressed in Human Induced Pluripotent Stem Cells , 2010, Stem cells.

[41]  P. Pasdois,et al.  The role of the mitochondrial permeability transition pore in heart disease. , 2009, Biochimica et biophysica acta.

[42]  Timothy J. Nelson,et al.  iPS Programmed Without c-MYC Yield Proficient Cardiogenesis for Functional Heart Chimerism , 2009, Circulation research.

[43]  C. Pereira,et al.  Senescence impairs successful reprogramming to pluripotent stem cells. , 2009, Genes & development.

[44]  T. Ichisaka,et al.  Hypoxia enhances the generation of induced pluripotent stem cells. , 2009, Cell stem cell.

[45]  A. Ben-Yehudah,et al.  Enhancement of human embryonic stem cell pluripotency through inhibition of the mitochondrial respiratory chain. , 2009, Stem cell research.

[46]  Manuel Serrano,et al.  A p53-mediated DNA damage response limits reprogramming to ensure iPS cell genomic integrity , 2009, Nature.

[47]  M. Blasco,et al.  The Ink4/Arf locus is a barrier for iPS cell reprogramming , 2009, Nature.

[48]  T. Ichisaka,et al.  Suppression of induced pluripotent stem cell generation by the p53–p21 pathway , 2009, Nature.

[49]  Timothy J. Nelson,et al.  Repair of acute myocardial infarction by human stemness factors induced pluripotent stem cells. , 2009, Circulation.

[50]  R. Hammer,et al.  Dependence of Mouse Embryonic Stem Cells on Threonine Catabolism , 2009, Science.

[51]  Andre Terzic,et al.  Stem Cell Platforms for Regenerative Medicine , 2009, Clinical and translational science.

[52]  L. Cantley,et al.  Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation , 2009, Science.

[53]  F. Bouillaud UCP2, not a physiologically relevant uncoupler but a glucose sparing switch impacting ROS production and glucose sensing. , 2009, Biochimica et biophysica acta.

[54]  J. S. John,et al.  The Relationship Between Pluripotency and Mitochondrial DNA Proliferation During Early Embryo Development and Embryonic Stem Cell Differentiation , 2009, Stem Cell Reviews and Reports.

[55]  A. Terzic,et al.  Induced Pluripotent Reprogramming from Promiscuous Human Stemness‐Related Factors , 2009, Clinical and translational science.

[56]  M. Andreeff,et al.  Mitochondrial uncoupling and the Warburg effect: molecular basis for the reprogramming of cancer cell metabolism. , 2009, Cancer research.

[57]  K. Hochedlinger,et al.  Epigenetic reprogramming and induced pluripotency , 2009, Development.

[58]  B. Gersh,et al.  Cardiac cell repair therapy: a clinical perspective. , 2009, Mayo Clinic proceedings.

[59]  Breakthrough of the year. Areas to watch. , 2009, Science.

[60]  K. Hochedlinger,et al.  Guidelines and techniques for the generation of induced pluripotent stem cells. , 2008, Cell stem cell.

[61]  A. Terzic,et al.  Developmental Restructuring of the Creatine Kinase System Integrates Mitochondrial Energetics with Stem Cell Cardiogenesis , 2008, Annals of the New York Academy of Sciences.

[62]  J. P. McCoy,et al.  Mitochondrial Metabolism Modulates Differentiation and Teratoma Formation Capacity in Mouse Embryonic Stem Cells* , 2008, Journal of Biological Chemistry.

[63]  D. K. Arrell,et al.  Guided stem cell cardiopoiesis: discovery and translation. , 2008, Journal of molecular and cellular cardiology.

[64]  Ryan B. Huang,et al.  Effects of oxygen on mouse embryonic stem cell growth, phenotype retention, and cellular energetics. , 2008, Biotechnology and bioengineering.

[65]  R. Roberts,et al.  Identification of oxygen-sensitive transcriptional programs in human embryonic stem cells. , 2008, Stem cells and development.

[66]  T. Mikkelsen,et al.  Dissecting direct reprogramming through integrative genomic analysis , 2008, Nature.

[67]  R. Jaenisch,et al.  Neurons derived from reprogrammed fibroblasts functionally integrate into the fetal brain and improve symptoms of rats with Parkinson's disease , 2008, Proceedings of the National Academy of Sciences.

[68]  O. Lee,et al.  Coordinated Changes of Mitochondrial Biogenesis and Antioxidant Enzymes During Osteogenic Differentiation of Human Mesenchymal Stem Cells , 2008, Stem cells.

[69]  K. Plath,et al.  Generation of human induced pluripotent stem cells from dermal fibroblasts , 2008, Proceedings of the National Academy of Sciences.

[70]  C. Thompson,et al.  Uncoupling protein‐2 controls proliferation by promoting fatty acid oxidation and limiting glycolysis‐derived pyruvate utilization , 2008, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[71]  R. Deberardinis,et al.  The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. , 2008, Cell metabolism.

[72]  Shulan Tian,et al.  Induced Pluripotent Stem Cell Lines Derived from Human Somatic Cells , 2007, Science.

[73]  S. Egginton,et al.  Mitochondrial DNA replication during differentiation of murine embryonic stem cells , 2007, Journal of Cell Science.

[74]  B. Bavister,et al.  Mitochondria in stem cells. , 2007, Mitochondrion.

[75]  Marius Wernig,et al.  Direct reprogramming of genetically unmodified fibroblasts into pluripotent stem cells , 2007, Nature Biotechnology.

[76]  T. Ichisaka,et al.  Generation of germline-competent induced pluripotent stem cells , 2007, Nature.

[77]  R. Jaenisch,et al.  In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state , 2007, Nature.

[78]  Sabine S. Lange,et al.  Stimulation of ES-cell-derived cardiomyogenesis and neonatal cardiac cell proliferation by reactive oxygen species and NADPH oxidase , 2007, Journal of Cell Science.

[79]  Andre Terzic,et al.  Mitochondrial oxidative metabolism is required for the cardiac differentiation of stem cells , 2007, Nature Clinical Practice Cardiovascular Medicine.

[80]  D. K. Arrell,et al.  Stem cells transform into a cardiac phenotype with remodeling of the nuclear transport machinery , 2007, Nature Clinical Practice Cardiovascular Medicine.

[81]  Sujin Kwon,et al.  Dynamic changes in mitochondrial biogenesis and antioxidant enzymes during the spontaneous differentiation of human embryonic stem cells. , 2006, Biochemical and biophysical research communications.

[82]  H. Baharvand,et al.  Ultrastructural comparison of developing mouse embryonic stem cell‐ and in vivo‐derived cardiomyocytes , 2006, Cell biology international.

[83]  S. Yamanaka,et al.  Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors , 2006, Cell.

[84]  Eyal Gottlieb,et al.  TIGAR, a p53-Inducible Regulator of Glycolysis and Apoptosis , 2006, Cell.

[85]  B. Bavister,et al.  Differentiation‐related changes in mitochondrial properties as indicators of stem cell competence , 2006, Journal of cellular physiology.

[86]  A. Terzic,et al.  Derivation of a cardiopoietic population from human mesenchymal stem cells yields cardiac progeny , 2006, Nature Clinical Practice Cardiovascular Medicine.

[87]  M. Pucéat Role of Rac-GTPase and reactive oxygen species in cardiac differentiation of stem cells. , 2005, Antioxidants & redox signaling.

[88]  G. Schatten,et al.  The expression of mitochondrial DNA transcription factors during early cardiomyocyte in vitro differentiation from human embryonic stem cells. , 2005, Cloning and stem cells.

[89]  A. Tabilio,et al.  Characterization of Mitochondrial and Extra-mitochondrial Oxygen Consuming Reactions in Human Hematopoietic Stem Cells , 2005, Journal of Biological Chemistry.

[90]  R. Roberts,et al.  Low O2 tensions and the prevention of differentiation of hES cells. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[91]  B. Fleischmann,et al.  Activity of complex III of the mitochondrial electron transport chain is essential for early heart muscle cell differentiation , 2004, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[92]  M. Pucéat,et al.  A dual role of the GTPase Rac in cardiac differentiation of stem cells. , 2003, Molecular biology of the cell.

[93]  A. Terzic,et al.  Structural Adaptation of the Nuclear Pore Complex in Stem Cell–Derived Cardiomyocytes , 2003, Circulation research.

[94]  H. Nau,et al.  Anticonvulsant valproic acid inhibits cardiomyocyte differentiation of embryonic stem cells by increasing intracellular levels of reactive oxygen species. , 2003, Birth defects research. Part A, Clinical and molecular teratology.

[95]  H. Baharvand,et al.  The ultrastructure of mouse embryonic stem cells. , 2003, Reproductive biomedicine online.

[96]  E. Couplan,et al.  No Evidence for a Basal, Retinoic, or Superoxide-induced Uncoupling Activity of the Uncoupling Protein 2 Present in Spleen or Lung Mitochondria* , 2002, The Journal of Biological Chemistry.

[97]  S. Bonhoeffer,et al.  Cooperation and Competition in the Evolution of ATP-Producing Pathways , 2001, Science.

[98]  B. Miroux,et al.  Uncoupling Protein 2, in Vivo Distribution, Induction upon Oxidative Stress, and Evidence for Translational Regulation* , 2001, The Journal of Biological Chemistry.

[99]  J. Hescheler,et al.  Effects of electrical fields on cardiomyocyte differentiation of embryonic stem cells , 1999, Journal of cellular biochemistry.