Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Stem Cells ; 37(3): 306-317, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30471152

RESUMO

Directed differentiation of human induced pluripotent stem cells (iPSCs) toward hepatobiliary lineages has been increasingly used as models of human liver development/diseases. As protein kinases are important components of signaling pathways regulating cell fate changes, we sought to define the key molecular mediators regulating human liver development using inhibitors targeting tyrosine kinases during hepatic differentiation of human iPSCs. A library of tyrosine kinase inhibitors was used for initial screening during the multistage differentiation of human iPSCs to hepatic lineage. Among the 80 kinase inhibitors tested, only Src inhibitors suppressed endoderm formation while none had significant effect on later stages of hepatic differentiation. Transient inhibition of c-Src during endodermal induction of human iPSCs reduced endodermal commitment and expression of endodermal markers, including SOX17 and FOXA2, in a dose-dependent manner. Interestingly, the transiently treated cells later developed into profibrogenic cholangiocyte-like cells expressing both cholangiocyte markers, such as CK7 and CK19, and fibrosis markers, including Collagen1 and smooth muscle actin. Further analysis of these cells revealed colocalized expression of collagen and yes-associated protein (YAP; a marker associated with bile duct proliferation/fibrosis) and an increased production of interleukin-6 and tumor necrosis factor-α. Moreover, treatment with verteporfin, a YAP inhibitor, significantly reduced expression of fibrosis markers. In summary, these results suggest that c-Src has a critical role in cell fate determination during endodermal commitment of human iPSCs, and its alteration in early liver development in human may lead to increased production of abnormal YAP expressing profibrogenic proinflammatory cholangiocytes, similar to those seen in livers of patients with biliary fibrosis. Stem Cells 2019;37:306-317.


Assuntos
Proteína Tirosina Quinase CSK/antagonistas & inibidores , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Endoderma/enzimologia , Inibidores de Proteínas Quinases/farmacologia , Ductos Biliares/enzimologia , Ductos Biliares/patologia , Proteína Tirosina Quinase CSK/metabolismo , Endoderma/patologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/enzimologia , Células-Tronco Pluripotentes Induzidas/patologia , Fígado/enzimologia , Fígado/patologia
2.
Cell Stem Cell ; 11(3): 346-58, 2012 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-22958930

RESUMO

Internal tandem duplication (ITD) mutations within the FMS-like tyrosine kinase-3 (FLT3) render the receptor constitutively active driving proliferation and survival in leukemic blasts. Expression of FLT3-ITD from the endogenous promoter in a murine knockin model results in progenitor expansion and a myeloproliferative neoplasm. In this study, we show that this expansion begins with overproliferation within a compartment of normally quiescent long-term hematopoietic stem cells (LT-HSCs), which become rapidly depleted. This depletion is reversible upon treatment with the small molecule inhibitor Sorafenib, which also ablates the disease. Although the normal LT-HSC has been defined as FLT3(-) by flow cytometric detection, we demonstrate that FLT3 is capable of playing a role within this compartment by examining the effects of constitutively activated FLT3-ITD. This indicates an important link between stem cell quiescence/homeostasis and myeloproliferative disease while also giving novel insight into the emergence of FLT3-ITD mutations in the evolution of leukemic transformation.


Assuntos
Neoplasias da Medula Óssea/patologia , Duplicação Gênica , Técnicas de Introdução de Genes , Células-Tronco Hematopoéticas/patologia , Homeostase , Sequências de Repetição em Tandem/genética , Tirosina Quinase 3 Semelhante a fms/genética , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Medula Óssea/patologia , Neoplasias da Medula Óssea/genética , Compartimento Celular/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Feminino , Duplicação Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Homeostase/efeitos dos fármacos , Homeostase/genética , Masculino , Camundongos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Fenótipo , Compostos de Fenilureia/farmacologia , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células da Side Population/efeitos dos fármacos , Células da Side Population/metabolismo , Células da Side Population/patologia , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Sorafenibe , Tirosina Quinase 3 Semelhante a fms/metabolismo
3.
Cancer Cell ; 21(3): 430-46, 2012 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-22439938

RESUMO

Reversal of promoter DNA hypermethylation and associated gene silencing is an attractive cancer therapy approach. The DNA methylation inhibitors decitabine and azacitidine are efficacious for hematological neoplasms at lower, less toxic, doses. Experimentally, high doses induce rapid DNA damage and cytotoxicity, which do not explain the prolonged time to response observed in patients. We show that transient exposure of cultured and primary leukemic and epithelial tumor cells to clinically relevant nanomolar doses, without causing immediate cytotoxicity, produce an antitumor "memory" response, including inhibition of subpopulations of cancer stem-like cells. These effects are accompanied by sustained decreases in genomewide promoter DNA methylation, gene reexpression, and antitumor changes in key cellular regulatory pathways. Low-dose decitabine and azacitidine may have broad applicability for cancer management.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Metilação de DNA/efeitos dos fármacos , Metilases de Modificação do DNA/antagonistas & inibidores , Leucemia/genética , Animais , Apoptose/efeitos dos fármacos , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/patologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Dano ao DNA , Decitabina , Inativação Gênica , Humanos , Leucemia/tratamento farmacológico , Leucemia/patologia , Camundongos , Dados de Sequência Molecular , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Regiões Promotoras Genéticas , Transdução de Sinais , Células Tumorais Cultivadas
4.
Sci Transl Med ; 4(127): 127ps9, 2012 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-22461639

RESUMO

The development of induced pluripotent stem cell (iPSC) technology has generated enthusiasm about the therapeutic potential of these cells for treating a variety of diseases. However, the evidence that they actually will be clinically useful is limited. Here, we discuss the potential therapeutic applications of iPSCs for treating cancer and other diseases and highlight the current barriers restricting their use.


Assuntos
Neoplasias/terapia , Células-Tronco Pluripotentes/citologia , Transplante de Células-Tronco , Células-Tronco Adultas/citologia , Animais , Neoplasias Hematológicas/terapia , Humanos , Pesquisa Translacional Biomédica
5.
Blood ; 119(15): 3571-7, 2012 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-22262762

RESUMO

Relapse of acute myeloid leukemia (AML) is thought to reflect the failure of current therapies to adequately target leukemia stem cells (LSCs), the rare, resistant cells presumed responsible for maintenance of the leukemia and typically enriched in the CD34(+)CD38(-) cell population. Despite the considerable research on LSCs over the past 2 decades, the clinical significance of these cells remains uncertain. However, if clinically relevant, it is expected that LSCs would be enriched in minimal residual disease and predictive of relapse. CD34(+) subpopulations from AML patients were analyzed by flow cytometry throughout treatment. Sorted cell populations were analyzed by fluorescence in situ hybridization for leukemia-specific cytogenetic abnormalities (when present) and by transplantation into immunodeficient mice to determine self-renewal capacity. Intermediate (int) levels of aldehyde dehydrogenase (ALDH) activity reliably distinguished leukemic CD34(+)CD38(-) cells capable of engrafting immunodeficient mice from residual normal hematopoietic stem cells that exhibited relatively higher ALDH activity. Minimal residual disease detected during complete remission was enriched for the CD34(+)CD38(-)ALDH(int) leukemic cells, and the presence of these cells after therapy highly correlated with subsequent clinical relapse. ALDH activity appears to distinguish normal from leukemic CD34(+)CD38(-) cells and identifies those AML cells associated with relapse.


Assuntos
Antígenos CD34/metabolismo , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/patologia , Células-Tronco Neoplásicas/patologia , ADP-Ribosil Ciclase 1/metabolismo , Adulto , Idoso , Animais , Separação Celular/métodos , Células Cultivadas , Feminino , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Pessoa de Meia-Idade , Células-Tronco Neoplásicas/metabolismo , Prognóstico , Recidiva , Adulto Jovem
6.
Mutat Res ; 713(1-2): 8-17, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21718709

RESUMO

To maintain the integrity of the organism, embryonic stem cells (ESC) need to maintain their genomic integrity in response to DNA damage. DNA double strand breaks (DSBs) are one of the most lethal forms of DNA damage and can have disastrous consequences if not repaired correctly, leading to cell death, genomic instability and cancer. How human ESC (hESC) maintain genomic integrity in response to agents that cause DSBs is relatively unclear. Adult somatic cells can be induced to "dedifferentiate" into induced pluripotent stem cells (iPSC) and reprogram into cells of all three germ layers. Whether iPSC have reprogrammed the DNA damage response is a critical question in regenerative medicine. Here, we show that hESC demonstrate high levels of endogenous reactive oxygen species (ROS) which can contribute to DNA damage and may arise from high levels of metabolic activity. To potentially counter genomic instability caused by DNA damage, we find that hESC employ two strategies: First, these cells have enhanced levels of DNA repair proteins, including those involved in repair of DSBs, and they demonstrate elevated nonhomologous end-joining (NHEJ) activity and repair efficacy, one of the main pathways for repairing DSBs. Second, they are hypersensitive to DNA damaging agents, as evidenced by a high level of apoptosis upon irradiation. Importantly, iPSC, unlike the parent cells they are derived from, mimic hESC in their ROS levels, cell cycle profiles, repair protein expression and NHEJ repair efficacy, indicating reprogramming of the DNA repair pathways. Human iPSC however show a partial apoptotic response to irradiation, compared to hESC. We suggest that DNA damage responses may constitute important markers for the efficacy of iPSC reprogramming.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Células-Tronco Embrionárias/fisiologia , Células-Tronco Pluripotentes/fisiologia , Ciclo Celular , Dano ao DNA , Instabilidade Genômica , Humanos , Espécies Reativas de Oxigênio/metabolismo
7.
Blood ; 118(7): 1801-5, 2011 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-21628406

RESUMO

EBV-immortalized B lymphocyte cell lines have been widely banked for studying a variety of diseases, including rare genetic disorders. These cell lines represent an important resource for disease modeling with the induced pluripotent stem cell (iPSC) technology. Here we report the generation of iPSCs from EBV-immortalized B-cell lines derived from multiple inherited disease patients via a nonviral method. The reprogramming method for the EBV cell lines involves a distinct protocol compared with that of patient fibroblasts. The B-cell line-derived iPSCs expressed pluripotency markers, retained the inherited mutation and the parental V(D)J rearrangement profile, and differentiated into all 3 germ layer cell types. There was no integration of the reprogramming-related transgenes or the EBV-associated genes in these iPSCs. The ability to reprogram the widely banked patient B-cell lines will offer an unprecedented opportunity to generate human disease models and provide novel drug therapies.


Assuntos
Linfócitos B/citologia , Linfócitos B/virologia , Herpesvirus Humano 4/fisiologia , Células-Tronco Pluripotentes Induzidas/citologia , Linfócitos B/metabolismo , Diferenciação Celular , Linhagem Celular , Transformação Celular Viral , Células Cultivadas , Reprogramação Celular , Fibroblastos/citologia , Fibroblastos/metabolismo , Herpesvirus Humano 4/genética , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Transgenes
8.
Sci Transl Med ; 3(82): 82ra39, 2011 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-21562231

RESUMO

Human induced pluripotent stem cells (iPSCs) are a potential source of hepatocytes for liver transplantation to treat end-stage liver disease. In vitro differentiation of human iPSCs into hepatic cells has been achieved using a multistage differentiation protocol, but whether these cells are functional and capable of engrafting and regenerating diseased liver tissue is not clear. We show that human iPSC-derived hepatic cells at various differentiation stages can engraft the liver in a mouse transplantation model. Using the same differentiation and transplantation protocols, we also assessed the ability of human iPSCs derived from each of the three developmental germ layer tissues (that is, ectoderm, mesoderm, and endoderm) to regenerate mouse liver. These iPSC lines, with similar but distinct global DNA methylation patterns, differentiated into multistage hepatic cells with an efficiency similar to that of human embryonic stem cells. Human hepatic cells at various differentiation stages derived from iPSC lines of different origins successfully repopulated the liver tissue of mice with liver cirrhosis. They also secreted human-specific liver proteins into mouse blood at concentrations comparable to that of proteins secreted by human primary hepatocytes. Our results demonstrate the engraftment and liver regenerative capabilities of human iPSC-derived multistage hepatic cells in vivo and suggest that human iPSCs of distinct origins and regardless of their parental epigenetic memory can efficiently differentiate along the hepatic lineage.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Regeneração Hepática/fisiologia , Animais , Diferenciação Celular , Linhagem Celular , Metilação de DNA , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Epigênese Genética , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fígado/citologia , Fígado/metabolismo , Camundongos , Transplante de Células-Tronco
9.
Am J Hematol ; 86(1): 31-7, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21132730

RESUMO

Although tyrosine kinase inhibitors have redefined the care of chronic myeloid leukemia (CML), these agents have not proved curative, likely due to resistance of the leukemia stem cells (LSC). While a number of potential therapeutic targets have emerged in CML, their expression in the LSC remains largely unknown. We therefore isolated subsets of CD34(+) stem/progenitor cells from normal donors and from patients with chronic phase or blast crisis CML. These cell subsets were then characterized based on ability to engraft immunodeficient mice and expression of candidate therapeutic targets. The CD34(+)CD38(-) CML cell population with high aldehyde dehydrogenase (ALDH) activity was the most enriched for immunodeficient mouse engrafting capacity. The putative targets: PROTEINASE 3, SURVIVIN, and hTERT were expressed only at relatively low levels by the CD34(+)CD38(-)ALDH(high) CML cells, similar to the normal CD34(+)CD38(-)ALDH(high) cells and less than in the total CML CD34(+) cells. In fact, the highest expression of these antigens was in normal, unfractionated CD34(+) cells. In contrast, PRAME and WT1 were more highly expressed by all CML CD34(+) subsets than their normal counterparts. Thus, ALDH activity appears to enrich for CML stem cells, which display an expression profile that is distinct from normal stem/progenitor cells and even the CML progenitors. Indeed, expression of a putative target by the total CD34(+) population in CML does not guarantee expression by the LSC. These expression patterns suggest that PROTEINASE 3, SURVIVIN, and hTERT are not optimal therapeutic targets in CML stem cells; whereas PRAME and WT1 seem promising.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , ADP-Ribosil Ciclase 1/biossíntese , Adulto , Idoso , Aldeído Desidrogenase/biossíntese , Animais , Antígenos CD34/biossíntese , Antígenos de Neoplasias/biossíntese , Antígenos de Neoplasias/genética , Feminino , Células-Tronco Hematopoéticas/imunologia , Humanos , Proteínas Inibidoras de Apoptose , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/imunologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Associadas aos Microtúbulos/genética , Pessoa de Meia-Idade , Mieloblastina/biossíntese , Mieloblastina/genética , Transplante de Neoplasias , RNA Mensageiro/biossíntese , Survivina , Telomerase/biossíntese , Telomerase/genética , Proteínas WT1/biossíntese , Proteínas WT1/genética
10.
PLoS One ; 5(6): e11251, 2010 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-20582166

RESUMO

BACKGROUND: Pulmonary hypertension (PH) is a disease of multiple etiologies with several common pathological features, including inflammation and pulmonary vascular remodeling. Recent evidence has suggested a potential role for the recruitment of bone marrow-derived (BMD) progenitor cells to this remodeling process. We recently demonstrated that hypoxia-induced mitogenic factor (HIMF/FIZZ1/RELM alpha) is chemotactic to murine bone marrow cells in vitro and involved in pulmonary vascular remodeling in vivo. METHODOLOGY/PRINCIPAL FINDINGS: We used a mouse bone marrow transplant model in which lethally irradiated mice were rescued with bone marrow transplanted from green fluorescent protein (GFP)(+) transgenic mice to determine the role of HIMF in recruiting BMD cells to the lung vasculature during PH development. Exposure to chronic hypoxia and pulmonary gene transfer of HIMF were used to induce PH. Both models resulted in markedly increased numbers of BMD cells in and around the pulmonary vasculature; in several neomuscularized small (approximately 20 microm) capillary-like vessels, an entirely new medial wall was made up of these cells. We found these GFP(+) BMD cells to be positive for stem cell antigen-1 and c-kit, but negative for CD31 and CD34. Several of the GFP(+) cells that localized to the pulmonary vasculature were alpha-smooth muscle actin(+) and localized to the media layer of the vessels. This finding suggests that these cells are of mesenchymal origin and differentiate toward myofibroblast and vascular smooth muscle. Structural location in the media of small vessels suggests a functional role in the lung vasculature. To examine a potential mechanism for HIMF-dependent recruitment of mesenchymal stem cells to the pulmonary vasculature, we performed a cell migration assay using cultured human mesenchymal stem cells (HMSCs). The addition of recombinant HIMF induced migration of HMSCs in a phosphoinosotide-3-kinase-dependent manner. CONCLUSIONS/SIGNIFICANCE: These results demonstrate HIMF-dependent recruitment of BMD mesenchymal-like cells to the remodeling pulmonary vasculature.


Assuntos
Vasos Sanguíneos/citologia , Células da Medula Óssea/citologia , Hipóxia/fisiopatologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Pulmão/irrigação sanguínea , Animais , Western Blotting , Transplante de Medula Óssea , Quimiotaxia , Dependovirus/genética , Feminino , Vetores Genéticos , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência
11.
Hepatology ; 51(5): 1810-9, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20432258

RESUMO

UNLABELLED: Recent advances in induced pluripotent stem (iPS) cell research have significantly changed our perspective on regenerative medicine. Patient-specific iPS cells have been derived not only for disease modeling but also as sources for cell replacement therapy. However, there have been insufficient data to prove that iPS cells are functionally equivalent to human embryonic stem (hES) cells or are safer than hES cells. There are several important issues that need to be addressed, and foremost are the safety and efficacy of human iPS cells of different origins. Human iPS cells have been derived mostly from cells originating from mesoderm and in a few cases from ectoderm. So far, there has been no report of endoderm-derived human iPS cells, and this has prevented comprehensive comparative investigations of the quality of human iPS cells of different origins. Here we show for the first time reprogramming of human endoderm-derived cells (i.e., primary hepatocytes) to pluripotency. Hepatocyte-derived iPS cells appear indistinguishable from hES cells with respect to colony morphology, growth properties, expression of pluripotency-associated transcription factors and surface markers, and differentiation potential in embryoid body formation and teratoma assays. In addition, these cells are able to directly differentiate into definitive endoderm, hepatic progenitors, and mature hepatocytes. CONCLUSION: The technology to develop endoderm-derived human iPS cell lines, together with other established cell lines, will provide a foundation for elucidating the mechanisms of cellular reprogramming and for studying the safety and efficacy of differentially originated human iPS cells for cell therapy. For the study of liver disease pathogenesis, this technology also provides a potentially more amenable system for generating liver disease-specific iPS cells.


Assuntos
Endoderma/citologia , Hepatócitos/citologia , Células-Tronco Pluripotentes Induzidas , Ativinas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/genética , Reprogramação Celular , Fator de Crescimento Epidérmico/farmacologia , Fator de Crescimento de Hepatócito/farmacologia , Humanos
12.
Exp Hematol ; 37(4): 423-434.e2, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19302917

RESUMO

OBJECTIVE: To investigate the mechanism of glycosylphosphatidylinositol (GPI) anchor deficiency in Burkitt lymphoma cell lines. METHODS: We identified a large GPI anchor protein deficient population in three different Burkitt lymphoma cell lines through proaerolysin treatment of the cells and flow cytometry analysis using a proaerolysin variant (FLAER). The mechanism of GPI anchor protein deficiency was studied by DNA gene sequencing, a cell-free assay to investigate the GPI anchor biosynthetic pathway, microarray analysis, and quantitative real-time polymerase chain reaction. RESULTS: Burkitt lymphoma cell lines harbor large populations of FLAER(neg) cells, which are resistant to proaerolysin. In all three cell lines, silencing of a gene involved in an early step in GPI-anchor biosynthesis was responsible for the lack of GPI-anchored proteins on the cell surface. Quantitative polymerase chain reaction and microarray analysis demonstrate that the level of mRNA for PIGL and PIGY is lower in the FLAER(neg) Ramos cells and that mRNA levels of PIGY are reduced in the Akata and Daudi cells. Hypermethylation of these genes was associated with the low levels of mRNA and treatment of the cells with 5-aza-2' deoxycytidine restored cell surface GPI-anchored proteins to the FLAER(neg) cells. CONCLUSION: GPI-anchored protein deficiency in Burkitt lymphoma cells is not due to a genetic mutation (e.g., PIGA); rather, the lack of GPI-anchored proteins results from transcriptional silencing of PIGL and PIGY.


Assuntos
Linfoma de Burkitt/metabolismo , Inativação Gênica , Glicosilfosfatidilinositóis/biossíntese , Linfoma de Burkitt/genética , Linhagem Celular Tumoral , Citometria de Fluxo , Glicosilfosfatidilinositóis/deficiência , Glicosilfosfatidilinositóis/genética , Sistema Hematopoético/citologia , Humanos , Imuno-Histoquímica , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Methods Mol Biol ; 430: 21-30, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18370289

RESUMO

A major challenge facing investigators working in the field of hematopoietic stem cell (HSC) biology has been to develop a strategy to purify rare primitive HSCs from bone marrow. Several methods have been available including the commonly used technique of isolating HSCs based on a specific cell-surface phenotype. As surface marker expression is dynamic and may fluctuate depending on the proliferative or activation state of the cell, our laboratory has established a unique functional in vivo assay (the 2-day homing assay) to isolate murine HSCs. This protocol selects for HSCs on the basis of their ability to home to bone marrow and yields a population that can reconstitute the murine hematopoietic system with the transplantation of a single cell. In contrast to other methods that use specific cell-surface antigens to acquire HSCs, our functional assay aids in obtaining a primitive HSC that exhibits both hematopoietic and epithelial engraftment capabilities. The 2-day homing protocol involves harvesting whole bone marrow and performing a physical separation method (elutriation) to acquire a fraction of small-sized cells (fraction 25). Fraction 25 cells are then depleted of later progenitors and differentiated hematopoietic cells, labeled with a fluorescent tracking dye and transplanted into lethally irradiated recipient mice. Two days after transplantation, the bone marrow is harvested from the primary recipient, and HSCs that have homed to the bone marrow are collected by fluorescence-activated cell sorting. In addition to the traditional 2-day homing protocol, we have included in this chapter our recently developed method of using density gradient centrifugation to replace the elutriation step that also selects for a primitive HSC.


Assuntos
Células-Tronco Hematopoéticas/citologia , Animais , Separação Celular , Transplante de Células-Tronco Hematopoéticas , Masculino , Camundongos , Camundongos Endogâmicos C57BL
14.
Cell ; 130(3): 403-4, 2007 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-17693249

RESUMO

What controls the inherent differences between fetal and adult hematopoietic stem cells (HSCs)? In this issue of Cell, Kim et al. (2007) demonstrate in mice that the endodermal transcription factor Sox17 is required for the maintenance of fetal and neonatal but not adult HSCs.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Células-Tronco Fetais/citologia , Células-Tronco Fetais/metabolismo , Proteínas HMGB/deficiência , Proteínas HMGB/genética , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Animais , Diferenciação Celular/genética , Proteínas HMGB/fisiologia , Camundongos , Fatores de Transcrição SOXF , Fatores de Transcrição/fisiologia
15.
Blood ; 110(8): 3056-63, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17595331

RESUMO

A low-oxygenic niche in bone marrow limits reactive oxygen species (ROS) production, thus providing long-term protection for hematopoietic stem cells (HSCs) from ROS stress. Although many approaches have been used to enrich HSCs, none has been designed to isolate primitive HSCs located within the low-oxygenic niche due to difficulties of direct physical access. Here we show that an early HSC population that might reside in the niche can be functionally isolated by taking advantage of the relative intracellular ROS activity. Many attributes of primitive HSCs in the low-oxygenic osteoblastic niche, such as quiescence, and calcium receptor, N-cadherin, Notch1, and p21 are higher in the ROS(low) population. Intriguingly, the ROS(low) population has a higher self-renewal potential. In contrast, significant HSC exhaustion in the ROS(high) population was observed following serial transplantation, and expression of activated p38 mitogen-activated protein kinase (MAPK) and mammalian target of rapamycin (mTOR) was higher in this population. Importantly, treatment with an antioxidant, a p38 inhibitor, or rapamycin was able to restore HSC function in the ROS(high) population. Thus, more potent HSCs associated with the low-oxygenic niche can be isolated by selecting for the low level of ROS expression. The ROS-related signaling pathways together with specific characteristics of niche HSCs may serve as targets for beneficial therapies.


Assuntos
Técnicas Citológicas/métodos , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Oxigênio , Espécies Reativas de Oxigênio , Animais , Western Blotting , Transplante de Medula Óssea , Adesão Celular/fisiologia , Diferenciação Celular/fisiologia , Feminino , Citometria de Fluxo , Fluoresceínas , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Masculino , Camundongos , Reação em Cadeia da Polimerase , Proteínas Quinases/metabolismo , Serina-Treonina Quinases TOR , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
Nat Genet ; 39(2): 237-42, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17211412

RESUMO

Adult cancers may derive from stem or early progenitor cells. Epigenetic modulation of gene expression is essential for normal function of these early cells but is highly abnormal in cancers, which often show aberrant promoter CpG island hypermethylation and transcriptional silencing of tumor suppressor genes and pro-differentiation factors. We find that for such genes, both normal and malignant embryonic cells generally lack the hypermethylation of DNA found in adult cancers. In embryonic stem cells, these genes are held in a 'transcription-ready' state mediated by a 'bivalent' promoter chromatin pattern consisting of the repressive mark, histone H3 methylated at Lys27 (H3K27) by Polycomb group proteins, plus the active mark, methylated H3K4. However, embryonic carcinoma cells add two key repressive marks, dimethylated H3K9 and trimethylated H3K9, both associated with DNA hypermethylation in adult cancers. We hypothesize that cell chromatin patterns and transient silencing of these important regulatory genes in stem or progenitor cells may leave these genes vulnerable to aberrant DNA hypermethylation and heritable gene silencing during tumor initiation and progression.


Assuntos
Cromatina/metabolismo , Metilação de DNA , Genes Supressores de Tumor , Células-Tronco/metabolismo , Adulto , Proliferação de Células , Células-Tronco Embrionárias/metabolismo , Inativação Gênica , Histonas/metabolismo , Humanos , Proteínas do Grupo Polycomb , Regiões Promotoras Genéticas , Proteínas Repressoras/metabolismo , Células Tumorais Cultivadas
17.
Exp Hematol ; 35(2): 335-41, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17258082

RESUMO

OBJECTIVE: Our laboratory has established two unique methods to isolate murine hematopoietic stem cells on the basis of functional characteristics such as the ability of stem cells to home to bone marrow and aldehyde dehydrogenase (ALDH) activity. An essential component of both protocols is the separation of whole bone marrow into small-sized cells by counter-flow elutriation. We sought to provide the scientific community with an alternate approach to acquire our stem cells by replacing elutriation with the use of density-gradient centrifugation. METHODS: The elutriated fraction 25 population was characterized based on density using a discontinuous gradient. The long-term reconstituting potential of whole bone marrow cells collected at each density interface was determined by subjecting the fractions to the two-day homing protocol, transplanting them into lethally irradiated recipient mice, and assessing peripheral blood chimerism. We also investigated the ability of high-density bone marrow cells isolated in conjunction with the ALDH protocol to repopulate the hematopoietic system of myeloablated recipients. RESULTS: Bone marrow cells collected at the high-density interface of 1.081/1.087 g/mL (fraction 3) had the capacity for homing to marrow and the ability to provide long-term hematopoietic reconstitution. Fraction three lineage-depleted ALDH-bright cells could also engraft and provide long-term hematopoiesis at limiting dilutions. CONCLUSIONS: Density-gradient centrifugation can be used in conjunction with either of our stem cell isolation protocols to obtain cells with long-term reconstitution ability. We anticipate that this strategy will encourage and enable investigators to study the biology of HSCs isolated using functional characteristics.


Assuntos
Células da Medula Óssea/citologia , Separação Celular/métodos , Células-Tronco Hematopoéticas/citologia , Aldeído Desidrogenase/imunologia , Animais , Células da Medula Óssea/enzimologia , Células da Medula Óssea/efeitos da radiação , Centrifugação com Gradiente de Concentração/métodos , Ativação Enzimática , Feminino , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/enzimologia , Células-Tronco Hematopoéticas/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Taxa de Sobrevida , Condicionamento Pré-Transplante , Transplante Homólogo , Irradiação Corporal Total
18.
J Urol ; 176(2): 813-8, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16813953

RESUMO

PURPOSE: Using various nonphysiological tissue injury/repair models numerous studies have demonstrated the capacity of bone marrow derived cells to contribute to the repopulation of epithelial tissues following damage. To investigate whether this phenomenon might also occur during periods of physiological tissue degeneration/regeneration we compared the ability of bone marrow derived cells to rejuvenate the prostate gland in mice that were castrated and then later treated with dihydrotestosterone vs mice with prostate epithelium that had been damaged by lytic virus infection. MATERIALS AND METHODS: Using allogenic bone marrow grafts from female donor transgenic mice expressing green fluorescent protein transplanted into lethally irradiated males we were able to assess the contributions of bone marrow derived cells to recovery of the prostatic epithelium in 2 distinct systems, including 1) surgical castration followed 1 week later by dihydrotestosterone replacement and 2) intraprostatic viral injection. Eight to 10-week-old male C57/Bl6 mice were distributed among bone marrow donor-->recipient/prostate injury groups, including 5 with C57/Bl6-->C57/Bl6/no injury, 3 with green fluorescent protein-->C57/Bl6/no injury, 3 with green fluorescent protein-->C57/Bl6/vehicle injection, 4 with green fluorescent protein-->C57/Bl6/virus injection and 3 each with green fluorescent protein-->C57/Bl6/castration without and with dihydrotestosterone, respectively. Prostate tissues were harvested 3 weeks after dihydrotestosterone replacement or 14 days following intraprostatic viral injection. Prostate tissue immunofluorescence was performed with antibodies against the epithelial marker cytokeratin 5/8, the hematopoietic marker CD45 and green fluorescent protein. RESULTS: Mice that sustained prostate injury from vaccinia virus infection with concomitant severe inflammation and glandular disruption showed evidence of bone marrow derived cell reconstitution of prostate epithelium, that is approximately 4% of all green fluorescent protein positive cells in the epithelial compartment 14 days after injury expressed cytokeratin 5/8, similar to the proportion of green fluorescent protein positive cells in the prostate that no longer expressed the hematopoietic marker CD45. When prostatic degeneration/regeneration was triggered by androgen deprivation and reintroduction, no green fluorescent protein positive prostate epithelial cells were detected. CONCLUSIONS: These findings are consistent with a requirement for inflammation associated architectural destruction for the bone marrow derived cell contribution to the regeneration of prostate epithelium.


Assuntos
Células da Medula Óssea/fisiologia , Próstata/patologia , Próstata/fisiologia , Regeneração , Animais , Epitélio/patologia , Epitélio/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL
19.
Stem Cells ; 24(6): 1476-86, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16456129

RESUMO

Human embryonic stem cells (hESCs) self-renew indefinitely while maintaining pluripotency. The molecular mechanism underlying hESCs self-renewal and pluripotency is poorly understood. To identify the signaling pathway molecules that maintain the proliferation of hESCs, we performed a microarray analysis comparing an aneuploid H1 hESC line (named H1T) versus euploid H1 hESC line because the H1T hESC line demonstrates a self-renewal advantage while maintaining pluripotency. We find differential gene expression for the Nodal/Activin, fibroblast growth factor (FGF), Wnt, and Hedgehog (Hh) signaling pathways in the H1T line, which implicates each of these molecules in maintaining the undifferentiated state, whereas the bone morphogenic protein (BMP) and Notch pathways could promote hESCs differentiation. Experimentally, we find that Activin A is necessary and sufficient for the maintenance of self-renewal and pluripotency of hESCs and supports long-term feeder and serum-free growth of hESCs. We show that Activin A induces the expression of Oct4, Nanog, Nodal, Wnt3, basic FGF, and FGF8 and suppresses the BMP signal. Our data indicates Activin A as a key regulator in maintenance of the stemness in hESCs. This finding will help elucidate the complex signaling network that maintains the hESC phenotype and function.


Assuntos
Ativinas/farmacologia , Proteínas Morfogenéticas Ósseas/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Subunidades beta de Inibinas/farmacologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Proteínas Wnt/metabolismo , Aneuploidia , Animais , Proteínas Morfogenéticas Ósseas/genética , Técnicas de Cultura de Células , Diferenciação Celular , Linhagem Celular , Embrião de Mamíferos/citologia , Fatores de Crescimento de Fibroblastos/genética , Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Pluripotentes/citologia , Transdução de Sinais/efeitos dos fármacos , Transplante de Células-Tronco/efeitos adversos , Teratoma/etiologia , Transplante Heterólogo , Proteínas Wnt/genética
20.
Curr Stem Cell Res Ther ; 1(1): 85-94, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18220857

RESUMO

The continuing interest in the biology of stem cells is enhanced by new discoveries surrounding developmental plasticity of both embryonic and adult stem cells. Adoptive transfer of concepts and definitions from the hematopoietic system to other tissue stem cells suggests inclusion of characteristics such as ability to self-renew and differentiate to functionally reconstitute a tissue/organ of origin. How adequate and accurate are these definitions? Within the great unknown of how these cells function, modulate their gene expression patterns and respond to extrinsic signals, it is apparent that there are numerous levels of stemness. We may envision a scale of developmental flexibility. At one end of the scale are positioned the embryonic stem cells, and at the other end are positioned partially-differentiated, differentiation restricted (committed) tissue/organ stem cells. There is evidence that some stem cells in the adult are pluripotent, thus positioned close to the embryonic end of the stem scale. It is uncertain yet to what extent stem cells can move back and forth along the stem scale.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Plasticidade Neuronal/fisiologia , Células-Tronco/fisiologia , Antígenos CD/análise , Ciclo Celular , Diferenciação Celular , Divisão Celular , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Humanos , Fenótipo , Células-Tronco/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA