Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Stem Cells Dev ; 26(10): 734-742, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28346802

ABSTRACT

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.


Subject(s)
Cellular Reprogramming , Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Metabolome , Cell Line , Cells, Cultured , Embryonic Stem Cells/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Induced Pluripotent Stem Cells/cytology
2.
Stem Cell Reports ; 7(1): 1-10, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27346679

ABSTRACT

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.


Subject(s)
Cell Differentiation/genetics , Electron Transport Complex II/genetics , Induced Pluripotent Stem Cells/metabolism , MicroRNAs/genetics , Mitochondria/metabolism , Animals , Cell Line , Cellular Reprogramming/genetics , Electron Transport Complex II/metabolism , Energy Metabolism/genetics , Humans , Mice , Mitochondria/genetics , Signal Transduction
3.
Cell ; 161(7): 1553-65, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26073944

ABSTRACT

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.


Subject(s)
Bone Marrow , Fetal Blood/cytology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Peptidyl-Prolyl Isomerase F , Cyclophilins/metabolism , Female , Hematopoietic Stem Cell Transplantation/instrumentation , Hematopoietic Stem Cells/cytology , Humans , Hypoxia , Mice , Mice, Inbred C57BL , Oxygen/metabolism , Tumor Suppressor Protein p53/metabolism
4.
Curr Opin Hematol ; 22(4): 273-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26049746

ABSTRACT

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.


Subject(s)
Blood Specimen Collection/methods , Bone Marrow Cells/metabolism , Fetal Blood/metabolism , Hematopoietic Stem Cells/metabolism , Hypoxia/metabolism , Bone Marrow Cells/cytology , Cell Differentiation , Cyclophilins/genetics , Cyclophilins/metabolism , Fetal Blood/cytology , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Humans , Hypoxia/genetics , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Oxidative Stress , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
5.
Stem Cells Dev ; 22(20): 2706-13, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23746070

ABSTRACT

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.


Subject(s)
Embryonic Development/genetics , Embryonic Stem Cells/cytology , Endoderm/cytology , GATA6 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Biomarkers/metabolism , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Carrier Proteins/pharmacology , Cell Differentiation , Cell Line , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Endoderm/drug effects , Endoderm/metabolism , GATA6 Transcription Factor/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Prealbumin/genetics , Prealbumin/metabolism
6.
Stem Cells ; 31(4): 666-81, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23255147

ABSTRACT

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.


Subject(s)
Cellular Reprogramming/physiology , Epigenesis, Genetic/genetics , Homeodomain Proteins/genetics , Induced Pluripotent Stem Cells/metabolism , MicroRNAs/genetics , Cells, Cultured , Cellular Reprogramming/genetics , Humans , Immunoprecipitation , Induced Pluripotent Stem Cells/cytology , Nanog Homeobox Protein
7.
Nat Med ; 18(12): 1786-96, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23160239

ABSTRACT

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.


Subject(s)
Chemokine CXCL12/metabolism , Dipeptidyl Peptidase 4/metabolism , Drug-Related Side Effects and Adverse Reactions , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hematopoiesis/physiology , Radiotherapy/adverse effects , Signal Transduction/physiology , Animals , Cell Line , DNA Primers/genetics , Dipeptidyl Peptidase 4/genetics , Hematopoiesis/drug effects , Hematopoiesis/radiation effects , Humans , Immunophenotyping , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics
9.
Blood ; 120(13): 2589-99, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22665934

ABSTRACT

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.


Subject(s)
Aging/pathology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mitochondria/pathology , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/physiology , Acetylcysteine/pharmacology , Anemia , Animals , Bone Marrow/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Erythroid Cells/cytology , Erythroid Cells/drug effects , Female , Free Radical Scavengers/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxygen Consumption/drug effects , Phenotype , Sequence Deletion
10.
Blood ; 119(24): 5731-41, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22535665

ABSTRACT

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.


Subject(s)
Antigens, CD1d/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Animals , Antibodies/pharmacology , Antigens, CD1d/chemistry , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Count , Cell Proliferation/drug effects , Chemokines/pharmacology , Colony-Forming Units Assay , Galactosylceramides/pharmacology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Interferon-gamma/pharmacology , Membrane Proteins/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/cytology , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Phenotype , Protein Structure, Tertiary , Stem Cell Factor/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
11.
Stem Cells Dev ; 21(10): 1597-603, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-21936707

ABSTRACT

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.


Subject(s)
Cyclin-Dependent Kinase Inhibitor Proteins/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Hematopoietic Stem Cells/physiology , Stem Cell Factor/physiology , Animals , Bone Marrow Cells/physiology , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Cytokines/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Progenitor Cells/physiology , Intercellular Signaling Peptides and Proteins/physiology , Interleukin-6/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , S Phase , Spleen/cytology , Stem Cell Factor/pharmacology
12.
Curr Opin Hematol ; 18(4): 208-13, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21537169

ABSTRACT

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.


Subject(s)
Hematopoietic Stem Cells/physiology , Mitochondrial Membrane Transport Proteins/metabolism , Reactive Oxygen Species/metabolism , Superoxides/metabolism , Animals , Apoptosis/physiology , Humans , Mitochondrial Membrane Transport Proteins/physiology , Mitochondrial Permeability Transition Pore
13.
Blood ; 117(21): 5643-51, 2011 May 26.
Article in English | MEDLINE | ID: mdl-21447833

ABSTRACT

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.


Subject(s)
Antigens, Neoplasm/physiology , Bone Marrow/metabolism , Gene Expression Regulation , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Proto-Oncogene Proteins c-myc/genetics , RNA-Binding Proteins/physiology , Animals , Antimetabolites, Antineoplastic/pharmacology , Blotting, Western , Bone Marrow/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Female , Fetal Blood/metabolism , Fluorouracil/pharmacology , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
14.
Cell Cycle ; 9(10): 2008-17, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20495374

ABSTRACT

Oxidative damage by reactive oxygen species generated in mitochondria is a potential cause of stem-cell dysregulation. Little is known about how hematopoietic stem cells mitigate/lessen this risk in the face of upregulated mitochondrial biogenesis/function necessary for the energy needs of differentiation and progenitor expansion. Here we report that upregulation of mitochondrial mass in mouse hematopoietic stem cells is closely linked to the appearance of CD34 on their surface, a marker indicating loss of long-term repopulating ability. These mitochondria have low membrane potential initially, but become active before exiting the primitive LSK compartment. Steady-state hematopoiesis perturbed by global expression of SDF-1/CXCL12 transgene causes a shift in ratios of these mitochondrialy-distinct LSK populations. Based on known effects of SDF-1 and signaling by it's receptor, CXCR4, along with finding primitive progenitors with high mitochondrial mass but low activity, we suggest a model of asymmetric self-renewing stem cell division that could lessen stem cell exposure to oxidative damage.


Subject(s)
Antigens, CD34/metabolism , Hematopoietic Stem Cells/metabolism , Animals , Cell Division/physiology , Cells, Cultured , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Membrane Potential, Mitochondrial/physiology , Mice , Models, Biological , Oxidative Stress/physiology , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism
15.
Cell Cycle ; 7(18): 2821-5, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18797187

ABSTRACT

Almost all complex multicellular organisms on earth utilize oxygen for the production of energy. This strategy carries the risk for damaging ROS to be generated and so these biochemical pathways must be highly regulated. Because of this, regulation of oxidative-phosphorylation is tightly coordinated with every aspect of cellular physiology, including stem cell regulation during embryonic development and in adult organisms. The protein-deacetylase, SIRT1, has received much attention because of its roles in oxygen metabolism, cellular stress response, aging, and has been investigated in various species and cell types including embryonic stem cells. However, there is a dearth of information on SIRT1 in adult stem cells, which have a pivotal role in adult aging processes. Here, we discuss the potential relationships between SIRT1 and the surface receptor protein, Notch, with stem cell self-renewal, asymmetric cell division, signaling and stem cell aging.


Subject(s)
Cellular Senescence , Histone Deacetylases/metabolism , Receptors, Notch/metabolism , Stem Cells/cytology , Stem Cells/enzymology , Animals , Cell Division , Cell Proliferation , Humans , Models, Biological
16.
Stem Cells Dev ; 17(5): 1017-20, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18788999

ABSTRACT

Previously, we reported that the spindle assembly checkpoint (SAC), which is coupled in somatic cells, is uncoupled from apoptosis-initiation in mouse and human embryonic stem cells (ESCs). This condition allows ESCs to tolerate and proliferate as polyploidy/aneuploid cells. Proper function of the SAC is vital to prevent polyploidy/aneuploidy during ex vivo hematopoietic stem cell (HSC) expansion. Here we address, for the first time, whether HSCs are more like ESCs or somatic cells with respect to SAC-apoptosis coupling. Using multiparametric permeablized cell flow-cytometric analysis to identify and analyze the mouse sca 1(+)/c-kit(+)/lin(-) (LSK) population, we found the mitotic spindle checkpoint to be functional in primary murine LSK cells, a population enriched in primitive hematopoietic stem/progenitor cells, after prolonged activation of the SAC by microtubule-depolymerizing agents such as nocodazole. HSCs can efficiently initiate apoptosis after activation of the SAC in LSK cells as indicated by increased hypodiploidy and increased levels of activated caspase 3, suggesting that HSCs behave more like somatic cells instead of ESCs with respect to this important cell cycle checkpoint. We conclude that mouse HSCs are not subject to the same kinds of chromosomal instability as are ESCs, knowledge that might aid in optimizing in vitro culture and expansion of human bone marrow or cord blood HSC for clinical applications.


Subject(s)
Apoptosis , Cell Cycle , Embryonic Stem Cells/cytology , Hematopoietic Stem Cells/cytology , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Embryonic Stem Cells/drug effects , Hematopoietic Stem Cells/drug effects , Humans , Mice , Mice, Inbred C57BL , Microtubules/drug effects , Microtubules/metabolism , Mitosis/drug effects , Nocodazole/pharmacology , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism
17.
Curr Opin Hematol ; 15(4): 326-31, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18536570

ABSTRACT

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.


Subject(s)
Aging , Cellular Senescence , Sirtuins/physiology , Stem Cells/cytology , Humans , Longevity , Sirtuin 1
18.
Cell Stem Cell ; 2(3): 241-51, 2008 Mar 06.
Article in English | MEDLINE | ID: mdl-18371449

ABSTRACT

Nuclear tumor suppressor p53 transactivates proapoptotic genes or antioxidant genes depending on stress severity, while cytoplasmic p53 induces mitochondrial-dependent apoptosis without gene transactivation. Although SIRT1, a p53 deacetylase, inhibits p53-mediated transactivation, how SIRT1 regulates these p53 multifunctions is unclear. Here we show that SIRT1 blocks nuclear translocation of cytoplasmic p53 in response to endogenous reactive oxygen species (ROS) and triggers mitochondrial-dependent apoptosis in mouse embryonic stem (mES) cells. ROS generated by antioxidant-free culture caused p53 translocation into mitochondria in wild-type mES cells but induced p53 translocation into the nucleus in SIRT1(-/-) mES cells. Endogenous ROS triggered apoptosis of wild-type mES through mitochondrial translocation of p53 and BAX but inhibited Nanog expression of SIRT1(-/-) mES, indicating that SIRT1 makes mES cells sensitive to ROS and inhibits p53-mediated suppression of Nanog expression. Our results suggest that endogenous ROS control is important for mES cell maintenance in culture.


Subject(s)
Apoptosis/physiology , Cell Nucleus/metabolism , Embryonic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Sirtuins/metabolism , Tumor Suppressor Protein p53/metabolism , Active Transport, Cell Nucleus/physiology , Animals , Cell Nucleus/genetics , Cells, Cultured , Cytoplasm/genetics , Cytoplasm/metabolism , Embryonic Stem Cells/cytology , Homeodomain Proteins/genetics , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Nanog Homeobox Protein , Reactive Oxygen Species/metabolism , Sirtuin 1 , Sirtuins/genetics , Transcriptional Activation/physiology , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
19.
Cell Cycle ; 7(4): 484-92, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18235235

ABSTRACT

It is widely accepted that mammalian cells enter the next G(1)-phase (G(1)) with 4N DNA after slippage from prolonged drug-induced mitotic block caused by activation of the transient spindle checkpoint. Understanding cell fate after mitotic slippage (MS) has significant clinical importance. The conclusion the MS cells enter 4N-G(1) is based on morphology and mitotic cyclin destruction. Definitive biochemical evidence for G(1) is scarce or unconvincing, in part because of methods of protein extraction required for immunoblot analysis that cannot take into account the cell cycle heterogeneity of cell cultures. We used single-cell-intracellular-flow-cytometric analysis to further define important factors determining cell fate after MS. Results from human and mouse embryonic stem cells (ESC) that reenter polyploid cell cycles are compared to human somatic cells that die after MS. We conclude that phosphorylation status of pRb, p53, CDK1, and especially cyclin B1 levels are important for cell fate decision in MS cells, which occur in a unique, intervening, non-G(1), tetraploid subphase.


Subject(s)
Embryonic Stem Cells/cytology , G1 Phase/physiology , Mitosis/physiology , Animals , CDC2 Protein Kinase/metabolism , Cyclin B/metabolism , Cyclin B1 , Flow Cytometry , Humans , Mice , Phosphorylation , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism
20.
Stem Cells ; 26(1): 30-4, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17932422

ABSTRACT

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.


Subject(s)
Apoptosis/physiology , Embryonic Stem Cells/metabolism , Microtubule-Associated Proteins/metabolism , Octamer Transcription Factor-3/metabolism , STAT3 Transcription Factor/metabolism , Animals , Antineoplastic Agents, Phytogenic/toxicity , Blotting, Western , Cell Survival , Cells, Cultured , DNA-Binding Proteins/biosynthesis , Etoposide/toxicity , Flow Cytometry , Homeodomain Proteins/biosynthesis , Hot Temperature , Inhibitor of Apoptosis Proteins , Mice , Mice, Knockout , Nanog Homeobox Protein , Phosphorylation , Promoter Regions, Genetic , RNA, Messenger/analysis , Repressor Proteins , Survivin , Transfection , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...