Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Stem Cell Res ; 30: 201-205, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29960149

RESUMO

MERRF syndrome is predominantly caused by A8344G mutation in the mitochondrial DNA (mtDNA), affecting MT-TK gene, which impairs the mitochondrial electron transport chain function. Here, we report the generation of two isogenic induced pluripotent stem cell (iPSC) lines, TVGH-iPSC-MRF-Mlow and TVGH-iPSC-MRF-Mhigh, from the skin fibroblasts of a female MERRF patient harboring mtDNA A8344G mutation by using retrovirus transduction system. Both cell lines share the same genetic background except containing different proportions of mtDNA with the A8344G mutation. Both cell lines exhibited the pluripotency and capacity to differentiate into three germ layers.


Assuntos
DNA Mitocondrial/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Síndrome MERRF/genética , Adolescente , Animais , Feminino , Humanos , Camundongos , Mutação
2.
Sci Rep ; 6: 23661, 2016 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-27025901

RESUMO

Myoclonus epilepsy associated with ragged-red fibers (MERRF) is a mitochondrial disorder characterized by myoclonus epilepsy, generalized seizures, ataxia and myopathy. MERRF syndrome is primarily due to an A to G mutation at mtDNA 8344 that disrupts the mitochondrial gene for tRNA(Lys). However, the detailed mechanism by which this tRNA(Lys) mutation causes mitochondrial dysfunction in cardiomyocytes or neurons remains unclear. In this study, we generated human induced pluripotent stem cells (hiPSCs) that carry the A8344G genetic mutation from patients with MERRF syndrome. Compared with mutation-free isogenic hiPSCs, MERRF-specific hiPSCs (MERRF-hiPSCs) exhibited reduced oxygen consumption, elevated reactive oxygen species (ROS) production, reduced growth, and fragmented mitochondrial morphology. We sought to investigate the induction ability and mitochondrial function of cardiomyocyte-like cells differentiated from MERRF-hiPSCs. Our data demonstrate that that cardiomyocyte-like cells (MERRF-CMs) or neural progenitor cells (MERRF-NPCs) differentiated from MERRF-iPSCs also exhibited increased ROS levels and altered antioxidant gene expression. Furthermore, MERRF-CMs or -NPCs contained fragmented mitochondria, as evidenced by MitoTracker Red staining and transmission electron microscopy. Taken together, these findings showed that MERRF-hiPSCs and MERRF-CM or -NPC harboring the A8344G genetic mutation displayed contained mitochondria with an abnormal ultrastructure, produced increased ROS levels, and expressed upregulated antioxidant genes.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Síndrome MERRF/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Adolescente , Desdiferenciação Celular , Diferenciação Celular , Células Cultivadas , DNA Mitocondrial/genética , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Síndrome MERRF/patologia , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Forma das Organelas , Consumo de Oxigênio , Mutação Puntual
3.
Biochim Biophys Acta ; 1860(4): 686-93, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26779594

RESUMO

BACKGROUND: Nuclear reprogramming with pluripotency factors enables somatic cells to gain the properties of embryonic stem cells. Mitochondrial resetting and metabolic reprogramming are suggested to be key early events in the induction of human skin fibroblasts to induced pluripotent stem cells (iPSCs). SCOPE OF REVIEW: We review recent advances in the study of the molecular basis for mitochondrial resetting and metabolic reprogramming in the regulation of the formation of iPSCs. In particular, the recent progress in using iPSCs for mitochondrial disease modeling was discussed. MAJOR CONCLUSIONS: iPSCs rely on glycolysis rather than oxidative phosphorylation as a major supply of energy. Mitochondrial resetting and metabolic reprogramming thus play crucial roles in the process of generation of iPSCs from somatic cells. GENERAL SIGNIFICANCE: Neurons, myocytes, and cardiomyocytes are cells containing abundant mitochondria in the human body, which can be differentiated from iPSCs or trans-differentiated from fibroblasts. Generating these cells from iPSCs derived from skin fibroblasts of patients with mitochondrial diseases or by trans-differentiation with cell-specific transcription factors will provide valuable insights into the role of mitochondrial DNA heteroplasmy in mitochondrial disease modeling and serves as a novel platform for screening of drugs to treat patients with mitochondrial diseases.


Assuntos
Reprogramação Celular , Células-Tronco Pluripotentes Induzidas/metabolismo , Mitocôndrias/metabolismo , Doenças Mitocondriais/metabolismo , Modelos Biológicos , Animais , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Mitocôndrias/genética , Mitocôndrias/patologia , Doenças Mitocondriais/genética , Doenças Mitocondriais/patologia
4.
Stem Cells ; 31(12): 2607-19, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23940081

RESUMO

Tumor cells have long been observed to share several biological characteristics with normal stem/progenitor cells; however, the oncogenic mechanisms underlying the lung stem/progenitor cell signaling remain elusive. Here, we report that SOX2, a self-renewal factor in lung stem/progenitor cells, is highly expressed in a subclass of lung cancer cells, the proliferation, survival, and chemoresistance of which are dependent on SOX2 signaling. Overexpression of SOX2 promotes oncogenic phenotypes in lung cancer cells; knockdown of SOX2 attenuated cell proliferation. We observed that SOX2 increased the expression of epidermal growth factor receptor (EGFR), and EGFR activation further upregulated SOX2 levels, forming a positive feedback loop. SOX2 expression promoted chemoresistance, and silencing of SOX2 perturbed mitochondrial function, causing marked apoptosis and autophagy. SOX2 induced BCL2L1, the ectopic expression of which rescued the effects of SOX2 silencing on apoptosis, autophagy, and mitochondrial function. SOX2 promoted tumor formation, along with increased cell proliferation in a xenograft mouse model. SOX2 expression is associated with poor prognosis in lung cancer patients; moreover, SOX2, EGFR, and BCL2L1 expression levels were significantly correlated in lung tumors. Our findings support the emerging role of SOX2 in cell proliferation and survival by eliciting oncogenic EGFR and BCL2L1 signaling with potential applications as a prognosis marker and a therapeutic target in lung cancer.


Assuntos
Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Fatores de Transcrição SOXB1/metabolismo , Animais , Apoptose/fisiologia , Autofagia/fisiologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/biossíntese , Receptores ErbB/genética , Receptores ErbB/metabolismo , Técnicas de Silenciamento de Genes , Inativação Gênica , Xenoenxertos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Camundongos , Paclitaxel/farmacologia , Fatores de Transcrição SOXB1/genética , Transdução de Sinais , Análise de Sobrevida , Proteína bcl-X/metabolismo
5.
Stem Cells ; 31(12): 2779-88, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23733376

RESUMO

We previously demonstrated that metabolic switch and mitochondrial activation are required for osteogenic differentiation of human mesenchymal stem cells (hMSCs). However, stem cells in niches or transplanted into injured tissues constantly encounter hypoxic stress that hinders aerobic metabolism. Therefore, we investigated the effects of oxygen tension (1% vs. 21%) on metabolism and osteogenic differentiation of hMSCs. We found that hypoxia impaired osteogenic differentiation as indicated by attenuation of alkaline phosphatase activity and expression of osteogenic markers core binding factor a-1 and osteopontin. In addition, differentiation-induced mitochondrial activation was compromised as shown by the decrease in the expression of respiratory enzymes and oxygen consumption rate. On the contrary, anaerobic metabolism was augmented as revealed by the upregulation of glycolytic enzymes and increase of lactate production, rendering the cells to rely more on anaerobic glycolysis for energy supply. Moreover, administration of 2-deoxyglucose (a glycolytic inhibitor) but not antimycin A (a respiratory inhibitor) significantly decreased intracellular ATP levels of hMSCs differentiating under hypoxia. Treatment with cobalt chloride, a hypoxia-inducible factor-1α (HIF-1α) stabilizer, recapitulated the inhibitory effects of hypoxia, suggesting that HIF-1α is involved in the compromise of hMSCs differentiation. These results suggest that hypoxia inhibits metabolic switch and mitochondrial function and therefore suppresses osteogenic differentiation of hMSCs.


Assuntos
Hipóxia Celular/fisiologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese/fisiologia , Diferenciação Celular/fisiologia , Células Cultivadas , Glicólise , Humanos , Mitocôndrias/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Regulação para Cima
6.
Biochim Biophys Acta ; 1820(5): 571-6, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-21983491

RESUMO

BACKGROUND: The self-renewal ability and pluripotent differentiation potential of stem cells hold great promise for regenerative medicine. Many studies focus on the lineage-specific differentiation and expansion of stem cells, but little is known about the regulation of glycolysis and mitochondrial biogenesis and function during these processes. Recent studies have demonstrated a strong correlation between cellular metabolism and the pluripotency and differentiation potential of stem cells, which indicates the importance of bioenergetic function in the regulation of stem cell physiology. SCOPE OF REVIEW: We summarize recent findings in the control of stem cell competence through the regulation of bioenergetic function in embryonic, hematopoietic, mesenchymal, and induced pluripotent stem cells, and discuss the up-to-date understanding of the molecular mechanisms involved in these biological processes. MAJOR CONCLUSIONS: It is believed that the metabolic signatures are highly correlated with the stemness status (high glycolytic flux) and differentiation potential (mitochondrial function) of stem cells. Besides, mitochondrial rejuvenation has been observed to participate in the reprogramming process. GENERAL SIGNIFICANCE: Understanding the metabolic regulation of stem cells will have great value in the characterization and isolation of stem cells with better differentiation potential. It also provides novel strategies of metabolic manipulation to increase the efficiency of cellular reprogramming. This article is part of a Special Issue entitled Biochemistry of Mitochondria, Life and Intervention 2010.


Assuntos
Diferenciação Celular , Reprogramação Celular , Metabolismo Energético , Mitocôndrias/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Humanos
7.
J Pineal Res ; 49(3): 222-38, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20626586

RESUMO

The purpose of this study was to determine the critical time periods of melatonin treatment required to induce human mesenchymal stem cells (hAMSCs) into osteoblasts and to determine which osteogenic genes are involved in the process. The study design consisted of adding melatonin for different times (2, 5, 10, 14 or 21 days) toward the end of a 21-day treatment containing osteogenic (OS+) medium or at the beginning of the 21-day treatment and then withdrawn. The results show that a 21-day continuous melatonin treatment was required to induce both alkaline phosphatase (ALP) activity and calcium deposition and these effects were mediated through MT2Rs. Functional analysis revealed that peak ALP levels induced by melatonin were accompanied by attenuation of melatonin-mediated inhibition of forskolin-induced cAMP accumulation. Immunoprecipitation and western blot analyses, respectively, showed that MT2R/ß-arrestin scaffolds complexed to Gi, MEK1/2 and ERK1/2 formed in these differentiated hAMSCs (i.e., when ALP levels were highest) where ERK1/2 resided primarily in the cytosol. It is hypothesized that these complexes form to modulate the subcellular localization of ERK1/2 to affect osteogenic gene expression. Using real-time RT-PCR, chronic melatonin exposure induced the expression of osteogenic genes RUNX-2, osteocalcin and BMP-2, through MT2Rs. No melatonin-mediated changes in the mRNA expression of ALP, BMP-6 or in the oxidative enzymes MtTFA, PGC-1α, Polγ, NRF-1, PDH, PDK and LDH occurred. These data show that a continuous 21-day melatonin exposure is required to induce osteoblast differentiation from hAMSCs through the formation of MT2R/Gi/ß-arrestin/MEK/ERK1/2 complexes to induce osteogenesis.


Assuntos
Antioxidantes/farmacologia , Diferenciação Celular/efeitos dos fármacos , Melatonina/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Arrestinas/genética , Arrestinas/metabolismo , Proteína Morfogenética Óssea 6/genética , Proteína Morfogenética Óssea 6/metabolismo , Humanos , MAP Quinase Quinase 1/genética , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/genética , MAP Quinase Quinase 2/metabolismo , Osteogênese/genética , Receptor MT2 de Melatonina/genética , Receptor MT2 de Melatonina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , beta-Arrestinas
8.
Biochim Biophys Acta ; 1800(3): 257-63, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19747960

RESUMO

Stem cell research has received increasing attention due to their invaluable potentials in the clinical applications to cure degenerative diseases, genetic disorders and even cancers. A great number of studies have been conducted with an aim to elucidate the molecular mechanisms involved in the regulation of self-renewal of stem cells and the mysterious circuits guiding them to differentiate into all kinds of progenies that can replenish the cell pools. However, little effort has been made in studying the metabolic aspects of stem cells. Mitochondria play essential roles in mammalian cells in the generation of ATP, Ca(2+) homeostasis, compartmentalization of biosynthetic pathways and execution of apoptosis. Considering the metabolic roles of mitochondria, they must be also critical in stem cells. This review is primarily focused on the biogenesis and bioenergetic function of mitochondria in the differentiation process and metabolic features of stem cells. In addition, the involvement of reactive oxygen species and hypoxic signals in the regulation of stem cell pluripotency and differentiation is also discussed.


Assuntos
Mitocôndrias/fisiologia , Células-Tronco/citologia , Trifosfato de Adenosina/metabolismo , Animais , Diferenciação Celular , Hipóxia Celular/fisiologia , DNA Mitocondrial/genética , Mamíferos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Membranas Mitocondriais/fisiologia , Morfogênese , Biogênese de Organelas , Organelas/ultraestrutura , Osteogênese/fisiologia , Consumo de Oxigênio , Células-Tronco/fisiologia , Regulação para Cima
9.
FEBS Lett ; 583(4): 691-6, 2009 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-19166847

RESUMO

The helicase domain of dengue virus NS3 protein (DENV NS3H) contains RNA-stimulated nucleoside triphosphatase (NTPase), ATPase/helicase, and RNA 5'-triphosphatase (RTPase) activities that are essential for viral RNA replication and capping. Here, we show that DENV NS3H unwinds 3'-tailed duplex with an RNA but not a DNA loading strand, and the helicase activity is poorly processive. The substrate of the divalent cation-dependent RTPase activity is not restricted to viral RNA 5'-terminus, a protruding 5'-terminus made the RNA 5'-triphosphate readily accessible to DENV NS3H. DENV NS3H preferentially binds RNA to DNA, and the functional interaction with RNA is sensitive to ionic strength.


Assuntos
Hidrolases Anidrido Ácido/metabolismo , Vírus da Dengue/metabolismo , Nucleosídeo-Trifosfatase/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Hidrolases Anidrido Ácido/genética , Motivos de Aminoácidos , Sequência de Aminoácidos , Clonagem Molecular , Vírus da Dengue/genética , Escherichia coli/genética , Histidina/química , Dados de Sequência Molecular , Mutação , Nucleosídeo-Trifosfatase/genética , Estrutura Terciária de Proteína , RNA Helicases/química , RNA Helicases/classificação , RNA Helicases/genética , RNA Helicases/metabolismo , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Serina Endopeptidases/química , Serina Endopeptidases/classificação , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Sorotipagem , Proteínas não Estruturais Virais/classificação , Proteínas não Estruturais Virais/genética
10.
J Biomed Opt ; 13(5): 050505, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19021377

RESUMO

The metabolic changes of human mesenchymal stem cells (hMSCs) during osteogenic differentiation were accessed by reduced nicotinamide adenine dinucleotide (NADH) fluorescence lifetime. An increase in mean fluorescence lifetime and decrease in the ratio between free NADH and protein-bound NADH correlated with our previously reported increase in the adenosine triphosphate (ATP) level of hMSCs during differentiation. These findings suggest that NADH fluorescence lifetime may serve as a new optical biomarker for noninvasive selection of stem cells from differentiated progenies.


Assuntos
Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Microscopia de Fluorescência/métodos , NAD/análise , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese/fisiologia , Espectrometria de Fluorescência/métodos , Células Cultivadas , Humanos , Oxirredução
11.
J Biomed Opt ; 13(5): 054011, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19021391

RESUMO

Direct monitoring of cell death (i.e., apoptosis and necrosis) during or shortly after treatment is desirable in all cancer therapies to determine the outcome. Further differentiation of apoptosis from necrosis is crucial to optimize apoptosis-favored treatment protocols. We investigated the potential modality of using tissue intrinsic fluorescence chromophore, reduced nicotinamide adenine dinucleotide (NADH), for cell death detection. We imaged the fluorescence lifetime changes of NADH before and after staurosporine (STS)-induced mitochondria-mediated apoptosis and hydrogen peroxide (H2O2)-induced necrosis, respectively, using two-photon fluorescence lifetime imaging in live HeLa cells and 143B osteosarcoma. Time-lapsed lifetime images were acquired at the same site of cells. In untreated cells, the average lifetime of NADH fluorescence was approximately 1.3 ns. The NADH average fluorescence lifetime increased to approximately 3.5 ns within 15 min after 1 microM STS treatment and gradually decreased thereafter. The NADH fluorescence intensity increased within 15 min. In contrast, no significant dynamic lifetime change was found in cells treated with 1 mM H2O2. Our findings suggest that monitoring the NADH fluorescence lifetime may be a valuable noninvasive tool to detect apoptosis and distinguish apoptosis from necrosis for the optimization of apoptosis-favored treatment protocols and other clinical applications.


Assuntos
Apoptose/fisiologia , NAD/análise , Necrose/metabolismo , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Espectrometria de Fluorescência/métodos , Linhagem Celular Tumoral , Células HeLa , Humanos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
12.
Gastroenterology ; 134(7): 2111-21, 2121.e1-3, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18455168

RESUMO

BACKGROUND & AIMS: Liver transplantation is the primary treatment for various end-stage hepatic diseases but is hindered by the lack of donor organs and by complications associated with rejection and immunosuppression. There is increasing evidence to suggest the bone marrow is a transplantable source of hepatic progenitors. We previously reported that multipotent bone marrow-derived mesenchymal stem cells differentiate into functional hepatocyte-like cells with almost 100% induction frequency under defined conditions, suggesting the potential for clinical applications. The aim of this study was to critically analyze the various parameters governing the success of bone marrow-derived mesenchymal stem cell-based therapy for treatment of liver diseases. METHODS: Lethal fulminant hepatic failure in nonobese diabetic severe combined immunodeficient mice was induced by carbon tetrachloride gavage. Mesenchymal stem cell-derived hepatocytes and mesenchymal stem cells were then intrasplenically or intravenously transplanted at different doses. RESULTS: Both mesenchymal stem cell-derived hepatocytes and mesenchymal stem cells, transplanted by either intrasplenic or intravenous route, engrafted recipient liver, differentiated into functional hepatocytes, and rescued liver failure. Intravenous transplantation was more effective in rescuing liver failure than intrasplenic transplantation. Moreover, mesenchymal stem cells were more resistant to reactive oxygen species in vitro, reduced oxidative stress in recipient mice, and accelerated repopulation of hepatocytes after liver damage, suggesting a possible role for paracrine effects. CONCLUSIONS: Bone marrow-derived mesenchymal stem cells can effectively rescue experimental liver failure and contribute to liver regeneration and offer a potentially alternative therapy to organ transplantation for treatment of liver diseases.


Assuntos
Transplante de Medula Óssea , Diferenciação Celular , Hepatócitos/transplante , Falência Hepática Aguda/cirurgia , Regeneração Hepática , Fígado/cirurgia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Animais , Antioxidantes/metabolismo , Tetracloreto de Carbono , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Modelos Animais de Doenças , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Fígado/metabolismo , Fígado/patologia , Fígado/fisiopatologia , Falência Hepática Aguda/induzido quimicamente , Falência Hepática Aguda/metabolismo , Falência Hepática Aguda/fisiopatologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo
13.
Stem Cells ; 26(4): 960-8, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18218821

RESUMO

The multidifferentiation ability of mesenchymal stem cells holds great promise for cell therapy. Numerous studies have focused on the establishment of differentiation protocols, whereas little attention has been paid to the metabolic changes during the differentiation process. Mitochondria, the powerhouse of mammalian cells, vary in their number and function in different cell types with different energy demands, but how these variations are associated with cell differentiation remains elusive. In this study, we investigated the changes of mitochondrial biogenesis and bioenergetic function using human mesenchymal stem cells (hMSCs) because of their well-defined differentiation potentials. Upon osteogenic induction, the copy number of mitochondrial DNA, protein subunits of the respiratory enzymes, oxygen consumption rate, and intracellular ATP content were increased, indicating the upregulation of aerobic mitochondrial metabolism. On the other hand, undifferentiated hMSCs showed higher levels of glycolytic enzymes and lactate production rate, suggesting that hMSCs rely more on glycolysis for energy supply in comparison with hMSC-differentiated osteoblasts. In addition, we observed a dramatic decrease of intracellular reactive oxygen species (ROS) as a consequence of upregulation of two antioxidant enzymes, manganese-dependent superoxide dismutase and catalase. Finally, we found that exogenous H(2)O(2) and mitochondrial inhibitors could retard the osteogenic differentiation. These findings suggested an energy production transition from glycolysis to oxidative phosphorylation in hMSCs upon osteogenic induction. Meanwhile, antioxidant enzymes were concurrently upregulated to prevent the accumulation of intracellular ROS. Together, our findings suggest that coordinated regulation of mitochondrial biogenesis and antioxidant enzymes occurs synergistically during osteogenic differentiation of hMSCs.


Assuntos
Antioxidantes/fisiologia , Diferenciação Celular/fisiologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/enzimologia , Mitocôndrias/enzimologia , Osteogênese/fisiologia , Adulto , Células Cultivadas , Humanos , Pessoa de Meia-Idade , Mitocôndrias/genética , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA