Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Metabolites ; 13(10)2023 Oct 17.
Article in English | MEDLINE | ID: mdl-37887411

ABSTRACT

Growing evidence indicates that metabolites and energy metabolism play an active rather than consequential role in regulating cellular fate. Cardiac development requires dramatic metabolic remodeling from relying primarily on glycolysis in pluripotent stem cells (PSCs) to oxidizing a wide array of energy substrates to match the high bioenergetic demands of continuous contraction in the developed heart. However, a detailed analysis of how remodeling of energy metabolism contributes to human cardiac development is lacking. Using dynamic multiple reaction monitoring metabolomics of central carbon metabolism, we evaluated temporal changes in energy metabolism during human PSC 3D cardiac lineage specification. Significant metabolic remodeling occurs during the complete differentiation, yet temporal analysis revealed that most changes occur during transitions from pluripotency to mesoderm (day 1) and mesoderm to early cardiac (day 5), with limited maturation of cardiac metabolism beyond day 5. Real-time metabolic analysis demonstrated that while hPSC cardiomyocytes (hPSC-CM) showed elevated rates of oxidative metabolism compared to PSCs, they still retained high glycolytic rates, confirming an immature metabolic phenotype. These observations support the opportunity to metabolically optimize the differentiation process to support lineage specification and maturation of hPSC-CMs.

2.
Stem Cells Dev ; 31(17-18): 521-528, 2022 09.
Article in English | MEDLINE | ID: mdl-35726436

ABSTRACT

Lineage-specific differentiation of human-induced pluripotent stem cells (hiPSCs) into cardiomyocytes (CMs) offers a patient-specific model to dissect development and disease pathogenesis in a dish. However, challenges exist with this model system, such as the relative immaturity of iPSC-derived CMs, which evoke the question of whether this model faithfully recapitulates in vivo cardiac development. As in vivo cardiac developmental stage is intimately linked with the proliferative capacity (or maturation is inversely correlated to proliferative capacity), we sought to understand how proliferation is regulated during hiPSC CM differentiation and how it compares with in vivo mouse cardiac development. Using standard Chemically Defined Media 3 differentiation, gene expression profiles demonstrate that hiPSC-derived cardiomyocytes (hiPSC-CMs) do not progress past the equivalent of embryonic day 14.5 of murine cardiac development. Throughout differentiation, overall DNA synthesis rapidly declines with <5% of hiPSC-CMs actively synthesizing DNA at the end of the differentiation period despite their immaturity. Bivariate cell cycle analysis demonstrated that hiPSC-CMs have a cell cycle profile distinct from their non-cardiac counterparts from the same differentiation, with significantly fewer cells within G1 and a marked accumulation of cells in G2/M than their non-cardiac counterparts throughout differentiation. Pulse-chase analysis demonstrated that non-cardiac cells progressed completely through the cell cycle within a 24-h period, whereas hiPSC-CMs had restricted progression with only a small proportion of cells undergoing cytokinesis with the remainder stalling in late S-phase or G2/M. This cell cycle arrest phenotype is associated with abbreviated expression of cell cycle promoting genes compared with expression throughout murine embryonic cardiac development. In summary, directed differentiation of hiPSCs into CMs uncouples the developmental stage from cell cycle regulation compared with in vivo mouse cardiac development, leading to a premature exit of hiPSC-CMs from the cell cycle despite their relative immaturity.


Subject(s)
Induced Pluripotent Stem Cells , Animals , Cell Differentiation/physiology , Cells, Cultured , Humans , Mice , Myocytes, Cardiac
3.
Methods Mol Biol ; 2429: 85-102, 2022.
Article in English | MEDLINE | ID: mdl-35507157

ABSTRACT

Mitochondrial function and energy metabolism are increasingly recognized not only as regulators of pluripotent stem cell function and fate, but also as critical targets in disease pathogenesis and aging. Therefore across the downstream applications of pluripotent stem cells, including development and disease modeling, drug screening, and cell-based therapies, it is crucial to be able to measure mitochondrial function and metabolism in a high-throughput, real-time and label-free manner. Here we describe the application of Seahorse extracellular flux analysis to measure mitochondrial function in pluripotent stem cells and their derivatives. Specifically, we highlight two assays, the Mitochondrial Stress Test, which quantifies overall mitochondrial function including basal, maximal and ATP-couple oxygen consumption rates, and the Electron Transport Chain Complex Specific assay, that quantifies function of individual complexes within the electron transport chain.


Subject(s)
Pluripotent Stem Cells , Energy Metabolism , Mitochondria/metabolism , Oxygen Consumption , Pluripotent Stem Cells/metabolism
4.
Front Cell Dev Biol ; 8: 87, 2020.
Article in English | MEDLINE | ID: mdl-32181250

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.

5.
Cell Stem Cell ; 18(5): 625-36, 2016 05 05.
Article in English | MEDLINE | ID: mdl-27151456

ABSTRACT

The genetic integrity of iPSCs is an important consideration for therapeutic application. In this study, we examine the accumulation of somatic mitochondrial genome (mtDNA) mutations in skin fibroblasts, blood, and iPSCs derived from young and elderly subjects (24-72 years). We found that pooled skin and blood mtDNA contained low heteroplasmic point mutations, but a panel of ten individual iPSC lines from each tissue or clonally expanded fibroblasts carried an elevated load of heteroplasmic or homoplasmic mutations, suggesting that somatic mutations randomly arise within individual cells but are not detectable in whole tissues. The frequency of mtDNA defects in iPSCs increased with age, and many mutations were non-synonymous or resided in RNA coding genes and thus can lead to respiratory defects. Our results highlight a need to monitor mtDNA mutations in iPSCs, especially those generated from older patients, and to examine the metabolic status of iPSCs destined for clinical applications.


Subject(s)
Aging/genetics , DNA, Mitochondrial/genetics , Induced Pluripotent Stem Cells/metabolism , Mutation/genetics , Adult , Aged , Blood Cells/metabolism , Fibroblasts/metabolism , Human Embryonic Stem Cells/metabolism , Humans , Skin/cytology
6.
Semin Cell Dev Biol ; 52: 68-75, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26868758

ABSTRACT

Energy metabolism is traditionally considered a reactive homeostatic system addressing stage-specific cellular energy needs. There is however growing appreciation of metabolic pathways in the active control of vital cell functions. Case in point, the stem cell lifecycle--from maintenance and acquisition of stemness to lineage commitment and specification--is increasingly recognized as a metabolism-dependent process. Indeed, metabolic reprogramming is an early contributor to the orchestrated departure from or reacquisition of stemness. Recent advances in metabolomics have helped decipher the identity and dynamics of metabolic fluxes implicated in fueling cell fate choices by regulating the epigenetic and transcriptional identity of a cell. Metabolic cues, internal and/or external to the stem cell niche, facilitate progenitor pool restitution, long-term tissue renewal or ensure adoption of cytoprotective behavior. Convergence of energy metabolism with stem cell fate regulation opens a new avenue in understanding primordial developmental biology principles with future applications in regenerative medicine practice.


Subject(s)
Embryonic Stem Cells/metabolism , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Energy Metabolism/physiology , Hematopoietic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology
7.
Nature ; 524(7564): 234-8, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26176921

ABSTRACT

Mitochondria have a major role in energy production via oxidative phosphorylation, which is dependent on the expression of critical genes encoded by mitochondrial (mt)DNA. Mutations in mtDNA can cause fatal or severely debilitating disorders with limited treatment options. Clinical manifestations vary based on mutation type and heteroplasmy (that is, the relative levels of mutant and wild-type mtDNA within each cell). Here we generated genetically corrected pluripotent stem cells (PSCs) from patients with mtDNA disease. Multiple induced pluripotent stem (iPS) cell lines were derived from patients with common heteroplasmic mutations including 3243A>G, causing mitochondrial encephalomyopathy and stroke-like episodes (MELAS), and 8993T>G and 13513G>A, implicated in Leigh syndrome. Isogenic MELAS and Leigh syndrome iPS cell lines were generated containing exclusively wild-type or mutant mtDNA through spontaneous segregation of heteroplasmic mtDNA in proliferating fibroblasts. Furthermore, somatic cell nuclear transfer (SCNT) enabled replacement of mutant mtDNA from homoplasmic 8993T>G fibroblasts to generate corrected Leigh-NT1 PSCs. Although Leigh-NT1 PSCs contained donor oocyte wild-type mtDNA (human haplotype D4a) that differed from Leigh syndrome patient haplotype (F1a) at a total of 47 nucleotide sites, Leigh-NT1 cells displayed transcriptomic profiles similar to those in embryo-derived PSCs carrying wild-type mtDNA, indicative of normal nuclear-to-mitochondrial interactions. Moreover, genetically rescued patient PSCs displayed normal metabolic function compared to impaired oxygen consumption and ATP production observed in mutant cells. We conclude that both reprogramming approaches offer complementary strategies for derivation of PSCs containing exclusively wild-type mtDNA, through spontaneous segregation of heteroplasmic mtDNA in individual iPS cell lines or mitochondrial replacement by SCNT in homoplasmic mtDNA-based disease.


Subject(s)
DNA, Mitochondrial/genetics , Induced Pluripotent Stem Cells/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Profiling , Haplotypes/genetics , Humans , Leigh Disease/genetics , Leigh Disease/metabolism , Leigh Disease/pathology , Mice , Mitochondria/pathology , Mitochondrial Diseases/pathology , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Encephalomyopathies/metabolism , Mitochondrial Encephalomyopathies/pathology , Mutation/genetics , Nuclear Transfer Techniques , Nucleotides/genetics , Oxygen Consumption , Polymorphism, Single Nucleotide/genetics , Sequence Analysis, RNA , Skin/cytology
8.
Reprod Fertil Dev ; 27(1): 82-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25472047

ABSTRACT

Decoding stem cell metabolism has implicated a tight linkage between energy metabolism and cell fate regulation, a dynamic interplay vital in the execution of developmental and differentiation programs. The inherent plasticity in energy metabolism enables prioritisation of metabolic pathways in support of stage-specific demands. Beyond traditional support of energetic needs, intermediate metabolism may also dictate cell fate choices through regulation of cellular signalling and epigenetic regulation of gene expression. The notion of a 'metabolism-centric' control of stem cell differentiation has been informed by developmental embryogenesis based upon an on-demand paradigm paramount in defining diverse developmental behaviours, from a post-fertilisation nascent zygote to complex organogenesis leading to adequate tissue formation and maturation. Monitored through natural or bioengineered stem cell surrogates, nutrient-responsive metabolites are identified as mediators of cross-talk between metabolic flux, cell signalling and epigenetic regulation charting, collectively, whether a cell will self-renew to maintain progenitor pools, lineage specify to ensure tissue (re)generation or remain quiescent to curb stress damage. Thus, bioenergetics are increasingly recognised as integral in governing stemness and associated organogenic decisions, paving the way for metabolism-defined targets in control of embryology, stem cell biology and tissue regeneration.


Subject(s)
Cell Differentiation/physiology , Embryonic Development/physiology , Embryonic Stem Cells/metabolism , Energy Metabolism/physiology , Epigenesis, Genetic/physiology , Metabolome/physiology , Signal Transduction/physiology , Animals , Humans , Models, Biological , Regeneration/physiology
9.
Cell Metab ; 20(3): 389-91, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25185944

ABSTRACT

Nutrient availability and intermediate metabolism are increasingly recognized to govern stem cell behavior. Oburoglu et al. (2014) now demonstrate that glutamine- and glucose-dependent nucleotide synthesis segregate erythroid versus myeloid differentiation during hematopoietic stem cell specification, implicating a metabolism-centric regulation of lineage choices.


Subject(s)
Amino Acid Transport System ASC/metabolism , Cell Lineage , Gene Expression Regulation , Glucose/metabolism , Glutamine/metabolism , Hematopoietic Stem Cells/cytology , Animals , Humans , Minor Histocompatibility Antigens
10.
Antioxid Redox Signal ; 21(11): 1648-59, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-24949895

ABSTRACT

SIGNIFICANCE: Metabolism-dependent generation of reactive oxygen species (ROS) and associated oxidative damage have been traditionally linked to impaired homeostasis and cellular death. Beyond the adverse effects of ROS accumulation, increasing evidence implicates redox status as a regulator of vital cellular processes. RECENT ADVANCES: Emerging studies on the molecular mechanisms guiding stem cell fate decisions indicate a role for energy metabolism in regulating the fundamental ability of maintaining stemness versus undergoing lineage-specific differentiation. Stem cells have evolved protective metabolic phenotypes to minimize reactive oxygen generation through oxidative metabolism and support antioxidant scavenging through glycolysis and the pentose phosphate pathway. CRITICAL ISSUES: While the dynamics in ROS generation has been correlated with stem cell function, the intimate mechanisms by which energy metabolism regulates ROS to impact cellular fate remain to be deciphered. FUTURE DIRECTIONS: Decoding the linkage between nutrient sensing, energy metabolism, and ROS in regulating cell fate decisions would offer a redox-dependent strategy to regulate stemness and lineage specification.


Subject(s)
Oxidation-Reduction , Stem Cells/metabolism , Animals , Cell Differentiation , Homeostasis , Humans , Stem Cells/cytology
11.
Croat Med J ; 54(4): 319-29, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23986272

ABSTRACT

Development of innovative high throughput technologies has enabled a variety of molecular landscapes to be interrogated with an unprecedented degree of detail. Emergence of next generation nucleotide sequencing methods, advanced proteomic techniques, and metabolic profiling approaches continue to produce a wealth of biological data that captures molecular frameworks underlying phenotype. The advent of these novel technologies has significant translational applications, as investigators can now explore molecular underpinnings of developmental states with a high degree of resolution. Application of these leading-edge techniques to patient samples has been successfully used to unmask nuanced molecular details of disease vs healthy tissue, which may provide novel targets for palliative intervention. To enhance such approaches, concomitant development of algorithms to reprogram differentiated cells in order to recapitulate pluripotent capacity offers a distinct advantage to advancing diagnostic methodology. Bioinformatic deconvolution of several "-omic" layers extracted from reprogrammed patient cells, could, in principle, provide a means by which the evolution of individual pathology can be developmentally monitored. Significant logistic challenges face current implementation of this novel paradigm of patient treatment and care, however, several of these limitations have been successfully addressed through continuous development of cutting edge in silico archiving and processing methods. Comprehensive elucidation of genomic, transcriptomic, proteomic, and metabolomic networks that define normal and pathological states, in combination with reprogrammed patient cells are thus poised to become high value resources in modern diagnosis and prognosis of patient disease.


Subject(s)
Gene Expression Profiling/methods , Molecular Diagnostic Techniques , Point-of-Care Systems , Proteomics/methods , Stem Cell Transplantation , Delivery of Health Care/methods , Humans
12.
Stem Cells ; 31(7): 1298-308, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23553816

ABSTRACT

Mitochondrial diseases display pathological phenotypes according to the mixture of mutant versus wild-type mitochondrial DNA (mtDNA), known as heteroplasmy. We herein examined the impact of nuclear reprogramming and clonal isolation of induced pluripotent stem cells (iPSC) on mitochondrial heteroplasmy. Patient-derived dermal fibroblasts with a prototypical mitochondrial deficiency diagnosed as mitochondrial encephalomyopathy with lactic acidosis and stroke-like episodes (MELAS) demonstrated mitochondrial dysfunction with reduced oxidative reserve due to heteroplasmy at position G13513A in the ND5 subunit of complex I. Bioengineered iPSC clones acquired pluripotency with multilineage differentiation capacity and demonstrated reduction in mitochondrial density and oxygen consumption distinguishing them from the somatic source. Consistent with the cellular mosaicism of the original patient-derived fibroblasts, the MELAS-iPSC clones contained a similar range of mtDNA heteroplasmy of the disease-causing mutation with identical profiles in the remaining mtDNA. High-heteroplasmy iPSC clones were used to demonstrate that extended stem cell passaging was sufficient to purge mutant mtDNA, resulting in isogenic iPSC subclones with various degrees of disease-causing genotypes. On comparative differentiation of iPSC clones, improved cardiogenic yield was associated with iPSC clones containing lower heteroplasmy compared with isogenic clones with high heteroplasmy. Thus, mtDNA heteroplasmic segregation within patient-derived stem cell lines enables direct comparison of genotype/phenotype relationships in progenitor cells and lineage-restricted progeny, and indicates that cell fate decisions are regulated as a function of mtDNA mutation load. The novel nuclear reprogramming-based model system introduces a disease-in-a-dish tool to examine the impact of mutant genotypes for MELAS patients in bioengineered tissues and a cellular probe for molecular features of individual mitochondrial diseases.


Subject(s)
DNA, Mitochondrial/genetics , Induced Pluripotent Stem Cells/metabolism , MELAS Syndrome/genetics , MELAS Syndrome/pathology , Mitochondria/genetics , Cellular Reprogramming/genetics , Cellular Reprogramming/physiology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/pathology , MELAS Syndrome/enzymology , MELAS Syndrome/metabolism , Mitochondria/pathology
13.
Cell Metab ; 17(2): 153-5, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23395162

ABSTRACT

Metabolic plasticity is increasingly postulated to be vital in the transition between stemness maintenance and lineage specification. Knobloch et al. (2012) now demonstrate that regulation of lipogenesis by fatty acid synthase and Spot14-dependent malonyl-CoA supply determines the proliferative activity of resident neural stem cells, contributing to adult neurogenesis.

14.
J Cardiovasc Transl Res ; 6(1): 10-21, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23247633

ABSTRACT

Reprogramming strategies influence the differentiation capacity of derived induced pluripotent stem (iPS) cells. Removal of the reprogramming factor c-Myc reduces tumorigenic incidence and increases cardiogenic potential of iPS cells. c-Myc is a regulator of energy metabolism, yet the impact on metabolic reprogramming underlying pluripotent induction is unknown. Here, mitochondrial and metabolic interrogation of iPS cells derived with (4F) and without (3F) c-Myc demonstrated that nuclear reprogramming consistently reverted mitochondria to embryonic-like immature structures. Metabolomic profiling segregated derived iPS cells from the parental somatic source based on the attained pluripotency-associated glycolytic phenotype and discriminated between 3F versus 4F clones based upon glycolytic intermediates. Real-time flux analysis demonstrated a greater glycolytic capacity in 4F iPS cells, in the setting of equivalent oxidative capacity to 3F iPS cells. Thus, inclusion of c-Myc potentiates the pluripotent glycolytic behavior of derived iPS cells, supporting c-Myc-free reprogramming as a strategy to facilitate oxidative metabolism-dependent lineage engagement.


Subject(s)
Cellular Reprogramming , Glycolysis , Induced Pluripotent Stem Cells/metabolism , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Line , Cell Lineage , Gene Expression Regulation , Glycolysis/drug effects , Metabolomics/methods , Mice , Oxidation-Reduction , Proto-Oncogene Proteins c-myc/genetics , Time Factors , Transfection
15.
Cell Stem Cell ; 11(5): 596-606, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-23122287

ABSTRACT

Plasticity in energy metabolism allows stem cells to match the divergent demands of self-renewal and lineage specification. Beyond a role in energetic support, new evidence implicates nutrient-responsive metabolites as mediators of crosstalk between metabolic flux, cellular signaling, and epigenetic regulation of cell fate. Stem cell metabolism also offers a potential target for controlling tissue homeostasis and regeneration in aging and disease. In this Perspective, we cover recent progress establishing an emerging relationship between stem cell metabolism and cell fate control.


Subject(s)
Cell Differentiation , Stem Cells/cytology , Animals , Energy Metabolism , Epigenesis, Genetic , Glycolysis , Humans , Signal Transduction , Stem Cells/metabolism
16.
PLoS One ; 7(5): e37703, 2012.
Article in English | MEDLINE | ID: mdl-22629445

ABSTRACT

OBJECTIVE: Diet-induced obesity (DIO) leads to an accumulation of intra-myocardial lipid metabolites implicated in causing cardiac insulin resistance and contractile dysfunction. One such metabolite is ceramide, and our aim was to determine the effects of inhibiting de novo ceramide synthesis on cardiac function and insulin stimulated glucose utilization in mice subjected to DIO. MATERIALS AND METHODS: C57BL/6 mice were fed a low fat diet or subjected to DIO for 12 weeks, and then treated for 4 weeks with either vehicle control or the serine palmitoyl transferase I (SPT I) inhibitor, myriocin. In vivo cardiac function was assessed via ultrasound echocardiography, while glucose metabolism was assessed in isolated working hearts. RESULTS: DIO was not associated with an accumulation of intra-myocardial ceramide, but rather, an accumulation of intra-myocardial DAG (2.63±0.41 vs. 4.80±0.97 nmol/g dry weight). Nonetheless, treatment of DIO mice with myriocin decreased intra-myocardial ceramide levels (50.3±7.7 vs. 26.9±2.7 nmol/g dry weight) and prevented the DIO-associated increase in intra-myocardial DAG levels. Interestingly, although DIO impaired myocardial glycolysis rates (7789±1267 vs. 2671±326 nmol/min/g dry weight), hearts from myriocin treated DIO mice exhibited an increase in glycolysis rates. CONCLUSIONS: Our data reveal that although intra-myocardial ceramide does not accumulate following DIO, inhibition of de novo ceramide synthesis nonetheless reduces intra-myocardial ceramide levels and prevents the accumulation of intra-myocardial DAG. These effects improved the DIO-associated impairment of cardiac glycolysis rates, suggesting that SPT I inhibition increases cardiac glucose utilization.


Subject(s)
Ceramides/metabolism , Fatty Acids, Monounsaturated/therapeutic use , Glycolysis/drug effects , Heart/drug effects , Insulin Resistance/physiology , Obesity/drug therapy , Serine C-Palmitoyltransferase/antagonists & inhibitors , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Echocardiography , Fatty Acids, Monounsaturated/pharmacology , Glycolysis/physiology , Insulin/metabolism , Mice , Myocardium/metabolism , Obesity/diagnostic imaging , Obesity/metabolism
17.
Ann N Y Acad Sci ; 1254: 82-89, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22548573

ABSTRACT

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.


Subject(s)
Energy Metabolism , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Animals , Cell Dedifferentiation , Cell Lineage , Cell Transdifferentiation , Humans , Induced Pluripotent Stem Cells/cytology , Regeneration , Regenerative Medicine/methods , Regenerative Medicine/trends
18.
Circ Res ; 110(4): 526-9, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22343555
19.
Biomark Med ; 5(6): 715-29, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22103608

ABSTRACT

Nuclear reprogramming with stemness factors enables resetting of somatic differentiated tissue back to the pluripotent ground state. Recent evidence implicates mitochondrial restructuring and bioenergetic plasticity as key components underlying execution of orchestrated dedifferentiation and derivation of induced pluripotent stem cells. Aerobic to anaerobic transition of somatic oxidative energy metabolism into a glycolytic metabotype promotes proficient reprogramming, establishing a novel regulator of acquired stemness. Metabolomic profiling has further identified specific metabolic remodeling traits defining lineage redifferentiation of pluripotent cells. Therefore, mitochondrial biogenesis and energy metabolism comprise a vital axis for biomarker discovery, intimately reflecting the molecular dynamics fundamental for the resetting and redirection of cell fate.


Subject(s)
Cellular Reprogramming , Energy Metabolism/genetics , Biomarkers/metabolism , Glycolysis , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Metabolomics , Mitochondria/metabolism , Mitochondria/ultrastructure
20.
Cell Metab ; 14(2): 264-71, 2011 Aug 03.
Article in English | MEDLINE | ID: mdl-21803296

ABSTRACT

The bioenergetics of somatic dedifferentiation into induced pluripotent stem cells remains largely unknown. Here, stemness factor-mediated nuclear reprogramming reverted mitochondrial networks into cristae-poor structures. Metabolomic footprinting and fingerprinting distinguished derived pluripotent progeny from parental fibroblasts according to elevated glucose utilization and production of glycolytic end products. Temporal sampling demonstrated glycolytic gene potentiation prior to induction of pluripotent markers. Functional metamorphosis of somatic oxidative phosphorylation into acquired pluripotent glycolytic metabolism conformed to an embryonic-like archetype. Stimulation of glycolysis promoted, while blockade of glycolytic enzyme activity blunted, reprogramming efficiency. Metaboproteomics resolved upregulated glycolytic enzymes and downregulated electron transport chain complex I subunits underlying cell fate determination. Thus, the energetic infrastructure of somatic cells transitions into a required glycolytic metabotype to fuel induction of pluripotency.


Subject(s)
Cellular Reprogramming , Energy Metabolism , Glycolysis/physiology , Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Electron Transport , Gene Expression Regulation, Developmental , Glucose/metabolism , Metabolomics , Mice , Mitochondria/metabolism , Oxidative Phosphorylation , Oxygen Consumption
SELECTION OF CITATIONS
SEARCH DETAIL
...