Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Cell ; 161(7): 1553-65, 2015 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-26073944

RESUMO

Hematopoietic stem cells (HSCs) reside in hypoxic niches within bone marrow and cord blood. Yet, essentially all HSC studies have been performed with cells isolated and processed in non-physiologic ambient air. By collecting and manipulating bone marrow and cord blood in native conditions of hypoxia, we demonstrate that brief exposure to ambient oxygen decreases recovery of long-term repopulating HSCs and increases progenitor cells, a phenomenon we term extraphysiologic oxygen shock/stress (EPHOSS). Thus, true numbers of HSCs in the bone marrow and cord blood are routinely underestimated. We linked ROS production and induction of the mitochondrial permeability transition pore (MPTP) via cyclophilin D and p53 as mechanisms of EPHOSS. The MPTP inhibitor cyclosporin A protects mouse bone marrow and human cord blood HSCs from EPHOSS during collection in air, resulting in increased recovery of transplantable HSCs. Mitigating EPHOSS during cell collection and processing by pharmacological means may be clinically advantageous for transplantation.


Assuntos
Medula Óssea , Sangue Fetal/citologia , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Animais , Peptidil-Prolil Isomerase F , Ciclofilinas/metabolismo , Feminino , Transplante de Células-Tronco Hematopoéticas/instrumentação , Células-Tronco Hematopoéticas/citologia , Humanos , Hipóxia , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio/metabolismo , Proteína Supressora de Tumor p53/metabolismo
2.
Curr Opin Hematol ; 22(4): 273-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26049746

RESUMO

PURPOSE OF REVIEW: Hematopoietic stem (HSCs) and progenitor (HPCs) cells reside in a hypoxic (lowered oxygen tension) environment, in vivo. We review literature on growth of HSCs and HPCs under hypoxic and normoxic (ambient air) conditions with a focus on our recent work demonstrating the detrimental effects of collecting and processing cells in ambient air through a phenomenon termed extra physiologic oxygen shock/stress (EPHOSS), and we describe means to counteract EPHOSS for enhanced collection of HSCs. RECENT FINDINGS: Collection and processing of bone marrow and cord blood cells in ambient air cause rapid differentiation and loss of HSCs, with increases in HPCs. This apparently irreversible EPHOSS phenomenon results from increased mitochondrial reactive oxygen species, mediated by a p53-cyclophilin D-mitochondrial permeability transition pore axis, and involves hypoxia inducing factor-1α and micro-RNA 210. EPHOSS can be mitigated by collecting and processing cells in lowered (3%) oxygen, or in ambient air in the presence of, cyclosporine A which effects the mitochondrial permeability transition pore, resulting in increased HSC collections. SUMMARY: Our recent findings may be advantageous for HSC collection for hematopoietic cell transplantation, and likely for enhanced collection of other stem cell types. EPHOSS should be considered when ex-vivo cell analysis is utilized for personalized medicine, as metabolism of cells and their response to targeted drug treatment ex vivo may not mimic what occurs in vivo.


Assuntos
Coleta de Amostras Sanguíneas/métodos , Células da Medula Óssea/metabolismo , Sangue Fetal/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Hipóxia/metabolismo , Células da Medula Óssea/citologia , Diferenciação Celular , Ciclofilinas/genética , Ciclofilinas/metabolismo , Sangue Fetal/citologia , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Humanos , Hipóxia/genética , Hipóxia/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
3.
Blood ; 119(24): 5731-41, 2012 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-22535665

RESUMO

In the present study, surface CD1d, which is involved in immune cell interactions, was assessed for effects on hematopoiesis. Mouse BM hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) express CD1d. The numbers and cycling status of HPCs in the BM and spleen of different strains of cd1d(-/-) mice were enhanced significantly, suggesting that CD1d is a negative regulator of HPCs. In support of this, CD1d was required for the SCF and Flt3 ligand synergistic enhancement of CSF induction of HPC colony formation and for HPC response to myelosuppressive chemokines. Colony formation by immature subsets of HPCs was greatly enhanced when normal, but not cd1d(-/-), BM cells were pretreated with CD1d Abs in vitro. These effects required the full CD1d cytoplasmic tail. In contrast, long-term, but not short-term, repopulating HSC engraftment was impaired significantly, an effect that was minimally influenced by the presence of a truncated CD1d cytoplasmic tail. Pretreatment of normal BM cells with CD1d Abs greatly enhanced their engraftment of HSCs. The results of the present study implicate CD1d in a previously unrecognized regulatory role of normal and stressed hematopoiesis.


Assuntos
Antígenos CD1d/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Animais , Anticorpos/farmacologia , Antígenos CD1d/química , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Contagem de Células , Proliferação de Células/efeitos dos fármacos , Quimiocinas/farmacologia , Ensaio de Unidades Formadoras de Colônias , Galactosilceramidas/farmacologia , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Interferon gama/farmacologia , Proteínas de Membrana/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células Mieloides/citologia , Células Mieloides/efeitos dos fármacos , Células Mieloides/metabolismo , Fenótipo , Estrutura Terciária de Proteína , Fator de Células-Tronco/farmacologia , Fator de Necrose Tumoral alfa/farmacologia
4.
Blood ; 120(13): 2589-99, 2012 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-22665934

RESUMO

Nuclear transcription factor Stat3 is important for proper regulation of hematopoietic stem cell (HSC) and hematopoietic progenitor cell (HPC) proliferation, survival, and cytokine signaling responses. A new, noncanonical role for Stat3 in mitochondrial function has been discovered recently. However, there is little information on the role(s) of mitochondrial Stat3 in HSC/HPC function, especially potential effects of Stat3/mitochondrial dysregulation in human diseases. We investigated hematopoietic cell-targeted deletion of the STAT3 gene in HSCs/HPCs with a focus on mitochondrial function. We found that STAT3(-/-) mice, which have a very shortened lifespan, dysfunctional/dysregulated mitochondrial function and excessive reactive oxygen species production in HSCs/HPCs that coincides with pronounced defects in function. These animals have a blood phenotype with similarities to premature aging and to human diseases of myelodysplastic syndrome and myeloproliferative neoplasms such as erythroid dysplasia, anemia, excessive myeloproliferation, and lymphomyeloid ratio shifts. We show herein that the lifespan of STAT3(-/-) animals is lengthened by treatment with a reactive oxygen species scavenger, which lessened the severity of the blood phenotype. These data suggest a need for more detailed studies of role(s) of Stat3 in HSC/HPC mitochondrial function in human diseases and raise the idea that mitochondrial Stat3 could be used as a potential therapeutic target.


Assuntos
Envelhecimento/patologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Mitocôndrias/patologia , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT3/fisiologia , Acetilcisteína/farmacologia , Anemia , Animais , Medula Óssea/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Células Eritroides/citologia , Células Eritroides/efeitos dos fármacos , Feminino , Sequestradores de Radicais Livres/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Consumo de Oxigênio/efeitos dos fármacos , Fenótipo , Deleção de Sequência
5.
Stem Cells ; 31(4): 666-81, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23255147

RESUMO

While most somatic cells undergoing induced pluripotent stem (iPS) cell reprogramming with Yamanaka factors accumulate at stable partially reprogrammed stages, the molecular mechanisms required to achieve full reprogramming are unknown. MicroRNAs (miRNAs) fine-tune mRNA translation and are implicated in reprogramming, but miRNA functional targets critical for complete iPS cell reprogramming remain elusive. We identified methyl-DNA binding domain protein 2 (MBD2) as an epigenetic suppressor, blocking full reprogramming of somatic to iPS cells through direct binding to NANOG promoter elements preventing transcriptional activation. When we overexpressed miR-302 cluster we observed a significant increase in conversion of partial to fully reprogrammed iPS cells by suppressing MBD2 expression, thereby increasing NANOG expression. Thus, expression of exogenous miR-302 cluster (without miR-367) is efficient in attaining a fully reprogrammed iPS state in partially reprogrammed cells by relieving MBD2-mediated inhibition of NANOG expression. Our studies provide a direct molecular mechanism involved in generating complete human iPS cell reprogramming to study disease pathogenesis, drug screening, and for potential cell-based therapies.


Assuntos
Reprogramação Celular/fisiologia , Epigênese Genética/genética , Proteínas de Homeodomínio/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , MicroRNAs/genética , Células Cultivadas , Reprogramação Celular/genética , Humanos , Imunoprecipitação , Células-Tronco Pluripotentes Induzidas/citologia , Proteína Homeobox Nanog
6.
Blood ; 117(21): 5643-51, 2011 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-21447833

RESUMO

Intracellular factors are involved in and essential for hematopoiesis. HIV-1 Tat-interacting protein of 110 kDa (TIP110; p110(nrb)/SART3/p110) is an RNA-binding nuclear protein implicated in the regulation of HIV-1 gene and host gene transcription, pre-mRNA splicing, and cancer immunology. In the present study, we demonstrate a role for TIP110 in the regulation of hematopoiesis. TIP110 was expressed in human CD34(+) cells and decreased with differentiation of CD34(+) cells. TIP110 mRNA was also expressed in phenotyped mouse marrow hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs). Using TIP110 transgenic (TIP110(TG)) and haploinsufficient (TIP110(+/-)) mice, we found that increased TIP110 expression enhanced HPC numbers, survival, and cell cycling, whereas decreased TIP110 expression had the opposite effects. Moreover, TIP110(+/-) bone marrow HPCs responded more effectively, and TIP110(TG) HPCs less effectively, than those of wild-type control mice to recovery from the cell-cycle-active drug 5-fluorouracil (5-FU). Unexplained sex differences were noted in HSC competitive repopulating ability, but not HPC numbers, in TIP110(TG) mice. Intracellularly, TIP110 regulated CMYC and GATA2 expression at the transcriptional level, and TIP110 and CMYC reciprocally regulated the expression of each other. These results demonstrate a role for TIP110 in the regulation of hematopoiesis, effects that are likely linked to TIP110 regulation of CMYC.


Assuntos
Antígenos de Neoplasias/fisiologia , Medula Óssea/metabolismo , Regulação da Expressão Gênica , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas de Ligação a RNA/fisiologia , Animais , Antimetabólitos Antineoplásicos/farmacologia , Western Blotting , Medula Óssea/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Sangue Fetal/metabolismo , Fluoruracila/farmacologia , Fator de Transcrição GATA2/genética , Fator de Transcrição GATA2/metabolismo , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Curr Opin Hematol ; 18(4): 208-13, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21537169

RESUMO

PURPOSE OF REVIEW: Reactive oxygen species (ROS) have an important function in blood cell homeostasis and hematopoietic diseases. Recent discoveries concerning how ROS are generated and regulated in mitochondria via the mitochondrial permeability transition pore (mPTP) and the new phenomenon, superoxide flashes, and ROS-induced ROS release, have not been investigated in hematopoietic stem and progenitor cells, but likely have important implications for their regulation and survival. Here we relate our opinions about these potential implications. RECENT FINDINGS: The mPTP has been recently implicated in ROS generation via binding of Stat3 transcription factor to a central component of the pore. SUMMARY: The implications of this new information for hematopoiesis regulation and transplantation methodologies could prove to be important, especially as they relate to myeloid neoplasm oncogenesis and potentially new therapeutic targets. New details about ROS production suggest that techniques for bone marrow and umbilical cord blood harvest may benefit from means to downmodulate ROS.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Superóxidos/metabolismo , Animais , Apoptose/fisiologia , Humanos , Proteínas de Transporte da Membrana Mitocondrial/fisiologia , Poro de Transição de Permeabilidade Mitocondrial
8.
Stem Cells ; 26(1): 30-4, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17932422

RESUMO

Understanding survival/antiapoptosis of murine embryonic stem (ES) cells may enhance their clinical potential. We hypothesized that Oct-4 might be involved in survival of undifferentiated ES cells under stress. The Oct-4 tetracycline conditional knockout cell line ZHBtc4 was used to test this possibility, and apoptosis was induced by either etoposide, heat shock, or UV exposure. Apoptosis in Oct-4 knocked-down ES cells was significantly increased in response to all stress situations compared with parental cells. Oct-4 knockdown was not associated with changes in morphology or expression of Nanog, SSEA-1, KLF-4, or Sox2 within the time frame and culture conditions used, suggesting that enhanced sensitivity of these cells to apoptosis was not due to an overtly differentiated state of the cells. To address potential intracellular mediators, we focused on the inhibitor of apoptosis proteins family member Survivin, an antiapoptosis protein. The Survivin promoter was transfected into ES cells after knockdown of Oct-4. Survivin promoter activity was dramatically decreased in the Oct-4 knockdown cells. Western blots substantiated that Oct-4 knockdown ES cells had decreased Survivin protein expression. Since the Survivin promoter does not have binding sites for Oct-4, this suggested an indirect effect of Oct-4 on expression of Survivin. Leukemia inhibitory factor-induced signal transducer and activator of transcription-3 (STAT3) is responsible for ES cell survival, and STAT3 regulates Survivin expression in breast cancer cells. Western blot analysis showed that downregulated Oct-4 was associated with decreased phosphorylation of STAT3. Our results suggest that Oct-4 is essential for antiapoptosis of ES cells in response to stress, effects that may be mediated through the STAT3/Survivin pathway.


Assuntos
Apoptose/fisiologia , Células-Tronco Embrionárias/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Antineoplásicos Fitogênicos/toxicidade , Western Blotting , Sobrevivência Celular , Células Cultivadas , Proteínas de Ligação a DNA/biossíntese , Etoposídeo/toxicidade , Citometria de Fluxo , Proteínas de Homeodomínio/biossíntese , Temperatura Alta , Proteínas Inibidoras de Apoptose , Camundongos , Camundongos Knockout , Proteína Homeobox Nanog , Fosforilação , Regiões Promotoras Genéticas , RNA Mensageiro/análise , Proteínas Repressoras , Survivina , Transfecção , Raios Ultravioleta
9.
Curr Opin Hematol ; 15(4): 326-31, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18536570

RESUMO

PURPOSE OF REVIEW: New discoveries focused on mitochondrial metabolism and gene silencing and their regulation by the sirtuin family of protein deacetylases is stimulating new ideas on how to improve geriatric medicine. Information about sertuins in stem cell biology is scarce. We consider recent information on sirtuin 1, its role in aging and metabolism in several species and tissues, and attempt to anticipate how it might influence stem cell aging. RECENT FINDINGS: Calorie restriction lengthens lifespan, in part, due to mitochondrial metabolism reorganization through sirtuin 1/peroxisome proliferator-activated receptor gamma-coactivator-1alpha-regulated mitochondrial biogenesis. This reduces radical oxygen species levels that cause macromolecule damage, a major contributor to aging. Little is known about these processes in stem cells, whose longevity is implicated in human aging. Recent work indicates that sirtuin 1 influences growth-factor responses and maintenance of stem cells. Sirtuin 1 is required for calorie restriction-induced lifespan extension in mice, and calorie restriction upregulates sirtuin 1 in humans. Sirtuin 1 also appears to influence lineage/cell-fate decisions of stem cells via redox status. SUMMARY: The same thermodynamic and biochemical mechanisms linked to aging in somatic cells may also work in stem cells. Developments in mitochondrial biology and new drug development based on this knowledge are finding their way into the clinic (i.e. diabetes) and may illuminate new ways of manipulating and using stem cells in medicine.


Assuntos
Envelhecimento , Senescência Celular , Sirtuínas/fisiologia , Células-Tronco/citologia , Humanos , Longevidade , Sirtuína 1
10.
Exp Hematol ; 35(4 Suppl 1): 78-86, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17379091

RESUMO

OBJECTIVE: Interleukin (IL)-31 is a recently discovered helical cytokine. Its receptor consists of a ligand-specific IL-31 receptor (IL-31R) subunit and a receptor chain that is shared with Oncostatin M (OSM), called OSM-Rbeta. Because OSM-Rbeta-deficient animals have reduced hematopoietic progenitor cells (HPC) and OSM has effects on and is involved in homeostasis of HPC, we studied whether IL-31 and IL-31R play a role in hematopoiesis. MATERIALS AND METHODS: IL-31R(-/-) mice and their littermate wild-type (WT) controls were assessed for absolute numbers and cycling status of bone marrow and spleen HPC (colony-forming unit granulocyte macrophage [CFU-GM], burst-forming unit erythroid [BFU-E], colony-forming unit granulocyte, erythrocyte, macrophage, megakaryocyte). Recombinant IL-31 was evaluated for stimulation, enhancement, or inhibition of colony formation by HPC, and for survival-enhancing effects on HPC subjected to growth-factor withdrawal and delayed addition of grown factors. Hematopoietic stem cells (HSC) from WT and IL-31R(-/-) mice were compared for competitive repopulating capacity in lethally irradiated congenic mice. RESULTS: IL-31R(-/-) mice demonstrated significantly decreased absolute numbers and cycling status of immature subsets of HPC in bone marrow bone and spleen compared to WT mice. There were no significant differences in absolute numbers of more mature subsets of WT and IL-31R(-/-) bone marrow CFU-GM. WT but not IL-31R(-/-) bone marrow CFU-GM responded to synergistic stimulation by combinations of cytokines. While IL-31 had neither colony-stimulating, -enhancing, or -inhibiting activity for bone marrow HPC, it did enhance survival of these HPC in the context of delayed addition of growth factors. No significant differences were detected in competitive repopulating HSC activity between WT and IL-31R(-/-) bone marrow cells. CONCLUSION: IL-31R is involved in positive regulation of absolute numbers and cycling status of immature subsets of HPC in vivo. While IL-31 in vitro does not modulate proliferation of HPC, it does enhance their survival, which may contribute to effects on cycling and numbers of HPC in vivo. Under steady-state conditions, loss of IL-31R on HPC does not appear to influence the activity of competitive repopulating HSC. These results with HPC may be of future utility for manipulation of hematopoiesis in a preclinical setting.


Assuntos
Proliferação de Células , Células Precursoras Eritroides/metabolismo , Células Precursoras de Granulócitos/metabolismo , Hematopoese , Interleucinas/metabolismo , Receptores de Interleucina/metabolismo , Animais , Sobrevivência Celular , Células Precursoras Eritroides/citologia , Células Precursoras de Granulócitos/citologia , Hematopoese/efeitos dos fármacos , Hematopoese/genética , Transplante de Células-Tronco Hematopoéticas , Humanos , Interleucinas/deficiência , Interleucinas/farmacologia , Camundongos , Camundongos Knockout
11.
Ann N Y Acad Sci ; 1106: 1-19, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17360804

RESUMO

The chemokine stromal cell-derived factor-1 (SDF-1/CXCL12) and its receptor, CXCR4, are involved in a number of facets of the regulation of hematopoiesis at the level of hematopoietic stem (HSCs) and progenitor (HPCs) cells. Modulation of this ligand-receptor interaction may be of clinical utility. We now report that: (1) the CC chemokine, macrophage inflammatory protein-1alpha (MIP-1alpha/CCL3) synergizes with AMD3100 (an antagonist of the binding of SDF-1/CXCL12 to CXCR4) to rapidly mobilize HPCs to the blood of mice; moreover, the combination of granulocyte colony-stimulating factor (G-CSF) with AMD3100 and MIP-1alpha/CCL3, given in a specific sequence, mobilizes the greatest number of HPCs compared to any combination of two of these mobilizing agents; (2) pretreatment of recipient mice with Diprotin A, an inhibitor of CD26/Dipeptidylpeptidase IV (DPPIV), enhances the competitive HSCs repopulating capacity of untreated donor cells; (3) the survival-enhancing effects of SDF-1/CXCL12 on HPCs subjected in vitro to delayed addition of growth factors (GFs) are mediated in part through the cell cycle-related proteins p21(cip1/waf1) (as assessed using p21(cip1/waf1) -/- and +/+ mice) and Mad2 (using Mad2 +/- and +/+ mice); and (4) deletion of CD26/DPPIV on mouse bone marrow cells increases the survival-enhancing effects of SDF-1/CXCL12 on HPCs. These results demonstrate the means to increase the mobilization of HPCs, the engrafting capability of HSCs, and responsiveness of HPCs to the survival-enhancing activity of SDF-1/CXCL12, effects that may be of practical value.


Assuntos
Quimiocinas CXC/metabolismo , Dipeptidil Peptidase 4/metabolismo , Compostos Heterocíclicos/farmacologia , Receptores CXCR4/metabolismo , Animais , Fármacos Anti-HIV/farmacologia , Antígenos CD34/biossíntese , Benzilaminas , Células da Medula Óssea/citologia , Linhagem da Célula , Quimiocina CXCL12 , Quimiocinas/metabolismo , Ciclamos , Dipeptidil Peptidase 4/química , Fator Estimulador de Colônias de Granulócitos/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ligação Proteica
12.
Stem Cells Dev ; 26(10): 734-742, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28346802

RESUMO

Acquisition of proper metabolomic fate is required to convert somatic cells toward fully reprogrammed pluripotent stem cells. The majority of induced pluripotent stem cells (iPSCs) are partially reprogrammed and have a transcriptome different from that of the pluripotent stem cells. The metabolomic profile and mitochondrial metabolic functions required to achieve full reprogramming of somatic cells to iPSC status have not yet been elucidated. Clarification of the metabolites underlying reprogramming mechanisms should enable further optimization to enhance the efficiency of obtaining fully reprogrammed iPSCs. In this study, we characterized the metabolites of human fully reprogrammed iPSCs, partially reprogrammed iPSCs, and embryonic stem cells (ESCs). Using capillary electrophoresis time-of-flight mass spectrometry-based metabolomics, we found that 89% of analyzed metabolites were similarly expressed in fully reprogrammed iPSCs and human ESCs (hESCs), whereas partially reprogrammed iPSCs shared only 74% similarly expressed metabolites with hESCs. Metabolomic profiling analysis suggested that converting mitochondrial respiration to glycolytic flux is critical for reprogramming of somatic cells into fully reprogrammed iPSCs. This characterization of metabolic reprogramming in iPSCs may enable the development of new reprogramming parameters for enhancing the generation of fully reprogrammed human iPSCs.


Assuntos
Reprogramação Celular , Células-Tronco Embrionárias/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Metaboloma , Linhagem Celular , Células Cultivadas , Células-Tronco Embrionárias/citologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia
13.
Stem Cell Reports ; 7(1): 1-10, 2016 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-27346679

RESUMO

Metabolism is remodeled when somatic cells are reprogrammed into induced pluripotent stem cells (iPSCs), but the majority of iPSCs are not fully reprogrammed. In a shift essential for reprogramming, iPSCs use less mitochondrial respiration but increased anaerobic glycolysis for bioenergetics. We found that microRNA 31 (miR-31) suppressed succinate dehydrogenase complex subunit A (SDHA) expression, vital for mitochondrial electron transport chain (ETC) complex II. MiR-31 overexpression in partially reprogrammed iPSCs lowered SDHA expression levels and oxygen consumption rates to that of fully reprogrammed iPSCs, but did not increase the proportion of fully reprogrammed TRA1-60(+) cells in colonies unless miR-31 was co-transduced with Yamanaka factors, which resulted in a 2.7-fold increase in full reprogramming. Thus switching from mitochondrial respiration to glycolytic metabolism through regulation of the miR-31/SDHA axis is critical for lowering the reprogramming threshold. This is supportive of multi-stage reprogramming whereby metabolic remodeling is fundamental.


Assuntos
Diferenciação Celular/genética , Complexo II de Transporte de Elétrons/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , MicroRNAs/genética , Mitocôndrias/metabolismo , Animais , Linhagem Celular , Reprogramação Celular/genética , Complexo II de Transporte de Elétrons/metabolismo , Metabolismo Energético/genética , Humanos , Camundongos , Mitocôndrias/genética , Transdução de Sinais
14.
J Leukoc Biol ; 73(5): 630-8, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12714578

RESUMO

Stromal cell-derived factor-1 (SDF-1/CXCL12) enhances survival of myeloid progenitor cells. The two main questions addressed by us were whether these effects on the progenitors were direct-acting and if SDF-1/CXCL12 enhanced engrafting capability of competitive, repopulating mouse stem cells subjected to short-term ex vivo culture with other growth factors. SDF-1/CXCL12 had survival-enhancing/antiapoptosis effects on human bone marrow (BM) and cord blood (CB) and mouse BM colony-forming units (CFU)-granulocyte macrophage, burst-forming units-erythroid, and CFU-granulocyte-erythroid-macrophage-megakaryocyte with similar dose responses. The survival effects were direct-acting, as assessed on colony formation by single isolated human BM and CB CD34(+++) cells. Effects were mediated through CXCR4 and G(alpha)i proteins. Moreover, SDF-1/CXCL12 greatly enhanced the engrafting capability of mouse long-term, marrow-competitive, repopulating stem cells cultured ex vivo with interleukin-6 and steel factor for 48 h. These results extend information on the survival effects mediated through the SDF-1/CXCL12-CXCR4 axis and may be of relevance for ex vivo expansion and gene-transduction procedures.


Assuntos
Quimiocinas CXC/farmacologia , Citocinas/farmacologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Células Mieloides/efeitos dos fármacos , Proteínas Proto-Oncogênicas/fisiologia , Receptores CXCR4/efeitos dos fármacos , Animais , Animais Congênicos , Apoptose/efeitos dos fármacos , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Quimiocina CXCL12 , Ensaio de Unidades Formadoras de Colônias , Feminino , Sangue Fetal/citologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP , Humanos , Recém-Nascido , Interleucina-6/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/citologia , Toxina Pertussis/farmacologia , Quimera por Radiação , Receptores CXCR4/fisiologia , Proteínas Recombinantes/farmacologia , Fator de Células-Tronco/farmacologia
15.
Cell Cycle ; 1(5): 327-36, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12461295

RESUMO

Microtubule-disruption (MTD) is often thought to arrest the mammalian cell cycle only during mitosis. However, MTD has also been demonstrated to arrest cells during interphase at a G(1)-phase point we call G(1)MTA. Microtubule integrity is now shown to be required for progression past G(1)MTA and the mammalian restriction-point. Neither p21(waf1) nor p27(kip1) are required for MTD-induced G(1)-arrest. Only p21(waf1) is crucial for normal G(1)MTA passage. The p21(waf1)-Chk1-cdc25C-cdc2-checkpoint-pathway is implicated in monitoring this passage. P21(waf1) deletion deregulates G(1)MTA transition and decreases MTD-G(1) arrest, possibly via Chk1 disregulation. Oncogene-induced overexpression of p21(waf1) produced opposite effects on the Chk1-cdc25C-cdc2 pathway and enhanced MTD-G(1) arrest. G(1)MTA thus represents a novel facet of mammalian G(1)/S checkpoint.


Assuntos
Ciclinas/fisiologia , Fase G1/fisiologia , Microtúbulos/fisiologia , Proteínas Quinases/fisiologia , Animais , Proteínas de Ciclo Celular/análise , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Quinase 1 do Ponto de Checagem , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Etoposídeo/farmacologia , Fase G1/efeitos dos fármacos , Deleção de Genes , Humanos , Interfase , Camundongos , Camundongos Knockout , Nocodazol/farmacologia , Fosforilação , Proteínas Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Fase S , Transdução de Sinais , Fosfatases cdc25/análise , Fosfatases cdc25/metabolismo
16.
Exp Mol Med ; 35(3): 222-6, 2003 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-12858022

RESUMO

Stem cell factor (SCF) is an early-acting cytokine inducing proliferative synergy with other cytokines in hematopoietic cells. We earlier showed that p21 was synergistically induced in SCF synergy and the p44/42 MAPK pathway was essential for the transcriptional control of p21. SCF synergy accompanies protein synthesis. p70S6K implicated in translational control in many other systems has not been shown in SCF synergy induced system. GM-CSF dependent human cell line MO7e was stimulated with GM-CSF with SCF, and investigated activation of p70S6K by using phospho-specific antibody. A possible contribution of p70S6K to SCF synergy was examined by measuring p21 induction as a model system. p70S6K was slightly activated by GM-CSF alone and markedly activated by SCF alone. Combined stimulation with these two cytokines synergistically activated p70S6K resulting in persistent activation. Addition of the pathway-specific inhibitors for P13K or FRAP/TOR, two up-stream pathways of p70S6K resulted in abolishment of p70S6K phosphorylation and also significant reduction of p21 protein level. These data suggest that synergistically activated p70S6K by GM-CSF plus SCF involves, at least in part, protein translational control including regulation of p21 protein.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Células-Tronco Hematopoéticas/enzimologia , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Fator de Células-Tronco/farmacologia , Sinergismo Farmacológico , Ativação Enzimática , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/antagonistas & inibidores , Proteína 1A de Ligação a Tacrolimo/metabolismo , Quinases Ativadas por p21
17.
Stem Cells Dev ; 22(20): 2706-13, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23746070

RESUMO

In this study, we isolated and characterized spontaneously differentiated human embryonic stem cells (SD-hESCs) found in hESC colonies in comparison to the morphologically premature ESCs in the colonies to investigate the potential role of SD-hESCs in embryogenesis. SD-hESCs were distinguished from undifferentiated hESCs by their higher expression of GATA6, a marker for primitive endoderm and transthyretin, a marker visceral endoderm in embryoid bodies (EBs). SD-hESCs expressed OCT4 and NANOG, markers for pluripotent stem cells, at significantly lower levels than undifferentiated hESCs. EBs derived from isolated SD-hESCs were morphologically distinct from cells directly derived from the undifferentiated hESCs; they contained higher number of cysts compared to EBs from undifferentiated hESC-derived EBs (42% vs. 20%). Furthermore, the extracellular signal molecule, BMP2/4, induced a higher GATA4/6 expression and cystic EB formation than control and noggin-treated EBs. Since cystic formation in EBs play a role in primitive endoderm formation during embryogenesis, the SD-hESC may be a relevant cell type equipped to differentiate into primitive endoderm. Our results suggest that SD-ESCs generated during routine hESC culture are not just an artifact of in vitro culture and these cells could serve as a useful model to study the process of embryogenesis.


Assuntos
Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias/citologia , Endoderma/citologia , Fator de Transcrição GATA6/genética , Regulação da Expressão Gênica no Desenvolvimento , Biomarcadores/metabolismo , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Proteínas de Transporte/farmacologia , Diferenciação Celular , Linhagem Celular , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Endoderma/efeitos dos fármacos , Endoderma/metabolismo , Fator de Transcrição GATA6/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Pré-Albumina/genética , Pré-Albumina/metabolismo
19.
Stem Cells Dev ; 21(10): 1597-603, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21936707

RESUMO

Cyclin dependent kinase inhibitors (CDKIs) influence proliferation of hematopoietic progenitor cells (HPCs), but little is known of how they influence proliferative responsiveness of HPCs to colony stimulating factors (CSFs), alone and in combination with other hematopoietically active factors, such as the potent co-stimulating cytokine stem cell factor (SCF), or inhibition by myelosuppressive chemokines. Using mice with deletions in p18(INK4c), p21(CIP1/WAF1), or p27(KIP1) genes, and in mice with double gene deletions for either p18/p21 or p18/p27, we determined effects of absence of these CDKIs and their interactions on functional HPC numbers in vivo, and HPC proliferative responsiveness in vitro. There is a decrease in bone marrow HPC proliferation in p18(-/-) mice commensurate with decreased numbers of HPC, suggesting a positive role for p18 on HPC in vivo, similar to that for p21. These positive effects of p18 dominate negative effects of p27 gene deletion. Moreover, the CDKIs differentially regulate responsiveness of granulocyte macrophage (GM) progenitors to synergistic cell proliferation in response to GM-CSF plus SCF, which is considered important for normal hematopoiesis. Responsiveness of HPCs to inhibition by myelosuppressive chemokines is directly related to the capacity of HPCs to respond to synergistic stimulation, and their cell cycle status. P18(INK4c) gene deletion rescued the loss of chemokine suppression of synergistic proliferation due to deletion of p21(CIP1/WAF1). These findings underscore the complex interplay of cell cycle regulators in HPC, and demonstrate that loss of one can sometimes be compensated by loss of another CDKI in both, a pro- or anti-proliferative context.


Assuntos
Proteínas Inibidoras de Quinase Dependente de Ciclina/fisiologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Fator de Células-Tronco/fisiologia , Animais , Células da Medula Óssea/fisiologia , Proliferação de Células , Células Cultivadas , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Proteínas Inibidoras de Quinase Dependente de Ciclina/metabolismo , Citocinas/fisiologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Células Progenitoras de Granulócitos e Macrófagos/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Interleucina-6/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fase S , Baço/citologia , Fator de Células-Tronco/farmacologia
20.
Nat Med ; 18(12): 1786-96, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23160239

RESUMO

Enhancement of hematopoietic recovery after radiation, chemotherapy, or hematopoietic stem cell (HSC) transplantation is clinically relevant. Dipeptidylpeptidase (DPP4) cleaves a wide variety of substrates, including the chemokine stromal cell-derived factor-1 (SDF-1). In the course of experiments showing that inhibition of DPP4 enhances SDF-1-mediated progenitor cell survival, ex vivo cytokine expansion and replating frequency, we unexpectedly found that DPP4 has a more general role in regulating colony-stimulating factor (CSF) activity. DPP4 cleaved within the N-termini of the CSFs granulocyte-macrophage (GM)-CSF, G-CSF, interleukin-3 (IL-3) and erythropoietin and decreased their activity. Dpp4 knockout or DPP4 inhibition enhanced CSF activities both in vitro and in vivo. The reduced activity of DPP4-truncated versus full-length human GM-CSF was mechanistically linked to effects on receptor-binding affinity, induction of GM-CSF receptor oligomerization and signaling capacity. Hematopoiesis in mice after radiation or chemotherapy was enhanced in Dpp4(-/-) mice or mice receiving an orally active DPP4 inhibitor. DPP4 inhibition enhanced engraftment in mice without compromising HSC function, suggesting the potential clinical utility of this approach.


Assuntos
Quimiocina CXCL12/metabolismo , Dipeptidil Peptidase 4/metabolismo , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Hematopoese/fisiologia , Radioterapia/efeitos adversos , Transdução de Sinais/fisiologia , Animais , Linhagem Celular , Primers do DNA/genética , Dipeptidil Peptidase 4/genética , Hematopoese/efeitos dos fármacos , Hematopoese/efeitos da radiação , Humanos , Imunofenotipagem , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA