Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 209(1): 77-92, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35705252

RESUMO

The zinc-finger transcription factor GATA-3 plays a crucial role during early T cell development and also dictates later T cell differentiation outcomes. However, its role and collaboration with the Notch signaling pathway in the induction of T lineage specification and commitment have not been fully elucidated. We show that GATA-3 deficiency in mouse hematopoietic progenitors results in an early block in T cell development despite the presence of Notch signals, with a failure to upregulate Bcl11b expression, leading to a diversion along a myeloid, but not a B cell, lineage fate. GATA-3 deficiency in the presence of Notch signaling results in the apoptosis of early T lineage cells, as seen with inhibition of CDK4/6 (cyclin-dependent kinases 4 and 6) function, and dysregulated cyclin-dependent kinase inhibitor 2b (Cdkn2b) expression. We also show that GATA-3 induces Bcl11b, and together with Bcl11b represses Cdkn2b expression; however, loss of Cdkn2b failed to rescue the developmental block of GATA-3-deficient T cell progenitor. Our findings provide a signaling and transcriptional network by which the T lineage program in response to Notch signals is realized.


Assuntos
Fator de Transcrição GATA3/metabolismo , Transdução de Sinais , Linfócitos T , Animais , Diferenciação Celular , Linhagem da Célula , Proteínas Inibidoras de Quinase Dependente de Ciclina , Redes Reguladoras de Genes , Camundongos , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Linfócitos T/metabolismo , Proteínas Supressoras de Tumor/metabolismo
2.
Sci Rep ; 11(1): 6777, 2021 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-33762663

RESUMO

Gene expression analysis of individual cells enables characterization of heterogeneous and rare cell populations, yet widespread implementation of existing single-cell gene analysis techniques has been hindered due to limitations in scale, ease, and cost. Here, we present a novel microdroplet-based, one-step reverse-transcriptase polymerase chain reaction (RT-PCR) platform and demonstrate the detection of three targets simultaneously in over 100,000 single cells in a single experiment with a rapid read-out. Our customized reagent cocktail incorporates the bacteriophage T7 gene 2.5 protein to overcome cell lysate-mediated inhibition and allows for one-step RT-PCR of single cells encapsulated in nanoliter droplets. Fluorescent signals indicative of gene expressions are analyzed using a probabilistic deconvolution method to account for ambient RNA and cell doublets and produce single-cell gene signature profiles, as well as predict cell frequencies within heterogeneous samples. We also developed a simulation model to guide experimental design and optimize the accuracy and precision of the assay. Using mixtures of in vitro transcripts and murine cell lines, we demonstrated the detection of single RNA molecules and rare cell populations at a frequency of 0.1%. This low cost, sensitive, and adaptable technique will provide an accessible platform for high throughput single-cell analysis and enable a wide range of research and clinical applications.


Assuntos
Perfilação da Expressão Gênica/métodos , Reação em Cadeia da Polimerase Multiplex/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Análise de Célula Única/métodos , Biologia Computacional/métodos , Ensaios de Triagem em Larga Escala/métodos , Dispositivos Lab-On-A-Chip , Técnicas Analíticas Microfluídicas/métodos , Transcriptoma , Fluxo de Trabalho
3.
Biochim Biophys Acta Mol Cell Res ; 1864(12): 2449-2459, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28964849

RESUMO

Although Hematopoietic Stem and Progenitor Cell (HSPC) proliferation, survival and expansion have been shown to be supported by the cooperative action of different cytokines, little is known about the intracellular signaling pathways that are activated by cytokines upon binding to their receptors. Our study showed that Growth factor receptor-bound protein 2 (Grb2) mRNAs are preferentially expressed in HSC compared to progenitors and differentiated cells of the myeloid and erythroid lineages. Conditional deletion of Grb2 induced a rapid decline of erythroid and myeloid progenitors and a progressive decline of HSC numbers in steady state conditions. We showed that when transplanted, Grb2 deleted bone marrow cells could not reconstitute irradiated recipients. Strinkingly, Grb2 deletion did not modify HSPC quiescence, but impaired LT-HSC and progenitors ability to respond a proliferative signal induced by 5FU in vivo and by various cytokines in vitro. We showed finally that Grb2 links IL3 signaling to the ERK/MAPK proliferative pathway and that both SH2 and SH3 domains of Grb2 are crucial for IL3 signaling in progenitor cells. Our findings position Grb2 as a key adaptor that integrates various cytokines response in cycling HSPC.


Assuntos
Diferenciação Celular/genética , Linhagem da Célula/genética , Proteína Adaptadora GRB2/genética , Células-Tronco Hematopoéticas/metabolismo , Animais , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Proliferação de Células/genética , Células Eritroides/metabolismo , Técnicas de Inativação de Genes , Células-Tronco Hematopoéticas/citologia , Camundongos , Células Mieloides/metabolismo , Transdução de Sinais
4.
Cell Stem Cell ; 16(3): 302-13, 2015 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-25704240

RESUMO

Regulated blood production is achieved through the hierarchical organization of dormant hematopoietic stem cell (HSC) subsets that differ in self-renewal potential and division frequency, with long-term (LT)-HSCs dividing the least. The molecular mechanisms underlying this variability in HSC division kinetics are unknown. We report here that quiescence exit kinetics are differentially regulated within human HSC subsets through the expression level of CDK6. LT-HSCs lack CDK6 protein. Short-term (ST)-HSCs are also quiescent but contain high CDK6 protein levels that permit rapid cell cycle entry upon mitogenic stimulation. Enforced CDK6 expression in LT-HSCs shortens quiescence exit and confers competitive advantage without impacting function. Computational modeling suggests that this independent control of quiescence exit kinetics inherently limits LT-HSC divisions and preserves the HSC pool to ensure lifelong hematopoiesis. Thus, differential expression of CDK6 underlies heterogeneity in stem cell quiescence states that functionally regulates this highly regenerative system.


Assuntos
Divisão Celular/fisiologia , Simulação por Computador , Quinase 6 Dependente de Ciclina/biossíntese , Regulação Enzimológica da Expressão Gênica/fisiologia , Células-Tronco Hematopoéticas/enzimologia , Modelos Biológicos , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Humanos
5.
Science ; 345(6203): 1509-12, 2014 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-25237102

RESUMO

The small number of hematopoietic stem and progenitor cells in cord blood units limits their widespread use in human transplant protocols. We identified a family of chemically related small molecules that stimulates the expansion ex vivo of human cord blood cells capable of reconstituting human hematopoiesis for at least 6 months in immunocompromised mice. The potent activity of these newly identified compounds, UM171 being the prototype, is independent of suppression of the aryl hydrocarbon receptor, which targets cells with more-limited regenerative potential. The properties of UM171 make it a potential candidate for hematopoietic stem cell transplantation and gene therapy.


Assuntos
Sangue Fetal/efeitos dos fármacos , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Indóis/farmacologia , Pirimidinas/farmacologia , Receptores de Hidrocarboneto Arílico/antagonistas & inibidores , Regeneração/efeitos dos fármacos , Animais , Técnicas de Cultura de Células , Sangue Fetal/citologia , Sangue Fetal/fisiologia , Terapia Genética/métodos , Hematopoese/fisiologia , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/fisiologia , Humanos , Hospedeiro Imunocomprometido , Indóis/química , Camundongos , Pirimidinas/química , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia
6.
Nat Med ; 20(1): 29-36, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24292392

RESUMO

Tumor recurrence following treatment remains a major clinical challenge. Evidence from xenograft models and human trials indicates selective enrichment of cancer-initiating cells (CICs) in tumors that survive therapy. Together with recent reports showing that CIC gene signatures influence patient survival, these studies predict that targeting self-renewal, the key 'stemness' property unique to CICs, may represent a new paradigm in cancer therapy. Here we demonstrate that tumor formation and, more specifically, human colorectal CIC function are dependent on the canonical self-renewal regulator BMI-1. Downregulation of BMI-1 inhibits the ability of colorectal CICs to self-renew, resulting in the abrogation of their tumorigenic potential. Treatment of primary colorectal cancer xenografts with a small-molecule BMI-1 inhibitor resulted in colorectal CIC loss with long-term and irreversible impairment of tumor growth. Targeting the BMI-1-related self-renewal machinery provides the basis for a new therapeutic approach in the treatment of colorectal cancer.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Compostos Heterocíclicos com 2 Anéis/farmacologia , Recidiva Local de Neoplasia/metabolismo , Células-Tronco Neoplásicas/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Tiazóis/farmacologia , Animais , Western Blotting , Bromodesoxiuridina , Linhagem Celular Tumoral , Citometria de Fluxo , Vetores Genéticos/genética , Compostos Heterocíclicos com 2 Anéis/uso terapêutico , Humanos , Luciferases , Camundongos Endogâmicos NOD , Camundongos SCID , Complexo Repressor Polycomb 1/antagonistas & inibidores , Interferência de RNA , RNA Interferente Pequeno/genética , Tiazóis/uso terapêutico
7.
Proc Natl Acad Sci U S A ; 110(51): 20599-604, 2013 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-24297922

RESUMO

The Lnk (Sh2b3) adaptor protein dampens the response of hematopoietic stem cells and progenitors (HSPCs) to a variety of cytokines by inhibiting JAK2 signaling. As a consequence, Lnk(-/-) mice develop hematopoietic hyperplasia, which progresses to a phenotype resembling the nonacute phase of myeloproliferative neoplasm. In addition, Lnk mutations have been identified in human myeloproliferative neoplasms and acute leukemia. We find that Lnk suppresses the development of radiation-induced acute B-cell malignancies in mice. Lnk-deficient HSPCs recover more effectively from irradiation than their wild-type counterparts, and this resistance of Lnk(-/-) HSPCs to radiation underlies the subsequent emergence of leukemia. A search for the mechanism responsible for radiation resistance identified the cytokine IL-11 as being critical for the ability of Lnk(-/-) HSPCs to recover from irradiation and subsequently become leukemic. In IL-11 signaling, wild-type Lnk suppresses tyrosine phosphorylation of the Src homology region 2 domain-containing phosphatase-2/protein tyrosine phosphatase nonreceptor type 11 and its association with the growth factor receptor-bound protein 2, as well as activation of the Erk MAP kinase pathway. Indeed, Src homology region 2 domain-containing phosphatase-2 has a binding motif for the Lnk Src Homology 2 domain that is phosphorylated in response to IL-11 stimulation. IL-11 therefore drives a pathway that enhances HSPC radioresistance and radiation-induced B-cell malignancies, but is normally attenuated by the inhibitory adaptor Lnk.


Assuntos
Raios gama/efeitos adversos , Interleucina-11/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Leucemia de Células B/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos da radiação , Proteínas de Neoplasias/metabolismo , Neoplasias Induzidas por Radiação/metabolismo , Proteínas/metabolismo , Tolerância a Radiação/efeitos da radiação , Proteínas Adaptadoras de Transdução de Sinal , Motivos de Aminoácidos , Animais , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo , Humanos , Interleucina-11/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Leucemia de Células B/genética , Leucemia de Células B/patologia , Sistema de Sinalização das MAP Quinases/genética , Proteínas de Membrana , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/genética , Neoplasias Induzidas por Radiação/genética , Neoplasias Induzidas por Radiação/patologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteínas/genética , Tolerância a Radiação/genética
8.
Cell ; 155(1): 215-27, 2013 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-24074870

RESUMO

Hematopoietic stem cells (HSCs) develop from a specialized subpopulation of endothelial cells known as hemogenic endothelium (HE). Although the HE origin of HSCs is now well established in different species, the signaling pathways that control this transition remain poorly understood. Here, we show that activation of retinoic acid (RA) signaling in aorta-gonad-mesonephros-derived HE ex vivo dramatically enhanced its HSC potential, whereas conditional inactivation of the RA metabolizing enzyme retinal dehydrogenase 2 in VE-cadherin expressing endothelial cells in vivo abrogated HSC development. Wnt signaling completely blocked the HSC inductive effects of RA modulators, whereas inhibition of the pathway promoted the development of HSCs in the absence of RA signaling. Collectively, these findings position RA and Wnt signaling as key regulators of HSC development and in doing so provide molecular insights that will aid in developing strategies for their generation from pluripotent stem cells.


Assuntos
Células-Tronco Hematopoéticas/citologia , Tretinoína/metabolismo , Aldeído Oxirredutases/metabolismo , Animais , Aorta/citologia , Aorta/embriologia , Regulação para Baixo , Embrião de Mamíferos , Gônadas/citologia , Gônadas/embriologia , Células-Tronco Hematopoéticas/metabolismo , Mesonefro/citologia , Camundongos , Receptores do Ácido Retinoico/metabolismo , Via de Sinalização Wnt
9.
Nat Immunol ; 14(10): 1037-44, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23974957

RESUMO

The transcription factor GATA-3 is expressed and required for differentiation and function throughout the T lymphocyte lineage. Despite evidence it may also be expressed in multipotent hematopoietic stem cells (HSCs), any role for GATA-3 in these cells has remained unclear. Here we found GATA-3 was in the cytoplasm in quiescent long-term stem cells from steady-state bone marrow but relocated to the nucleus when HSCs cycled. Relocation depended on signaling via the mitogen-activated protein kinase p38 and was associated with a diminished capacity for long-term reconstitution after transfer into irradiated mice. Deletion of Gata3 enhanced the repopulating capacity and augmented the self-renewal of long-term HSCs in cell-autonomous fashion without affecting the cell cycle. Our observations position GATA-3 as a regulator of the balance between self-renewal and differentiation in HSCs that acts downstream of the p38 signaling pathway.


Assuntos
Fator de Transcrição GATA3/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Animais , Fator de Transcrição GATA3/genética , Deleção de Genes , Expressão Gênica , Hematopoese/genética , Células-Tronco Hematopoéticas/efeitos dos fármacos , Ligantes , Camundongos , Camundongos Knockout , Poli I-C/farmacologia , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Receptores Toll-Like/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
PLoS One ; 8(3): e58787, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23516556

RESUMO

Tachykinins are a large group of neuropeptides with both central and peripheral activity. Despite the increasing number of studies reporting a growth supportive effect of tachykinin peptides in various in vitro stem cell systems, it remains unclear whether these findings are applicable in vivo. To determine how neurokinin-1 receptor (NK-1R) deficient hematopoietic stem cells would behave in a normal in vivo environment, we tested their reconstitution efficiency using competitive bone marrow repopulation assays. We show here that bone marrow taken from NK-1R deficient mice (Tacr1(-/-)) showed lineage specific B and T cell engraftment deficits compared to wild-type competitor bone marrow cells, providing evidence for an involvement of NK-1R signalling in adult hematopoiesis. Tachykinin knockout mice lacking the peptides SP and/or HK-1 (Tac1 (-/-), Tac4 (-/-) and Tac1 (-/-)/Tac4 (-/-) mice) repopulated a lethally irradiated wild-type host with similar efficiency as competing wild-type bone marrow. The difference between peptide and receptor deficient mice indicates a paracrine and/or endocrine mechanism of action rather than autocrine signalling, as tachykinin peptides are supplied by the host environment.


Assuntos
Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Receptores da Neurocinina-1/metabolismo , Transdução de Sinais , Animais , Linfócitos B/citologia , Linfócitos B/metabolismo , Linhagem da Célula , Feminino , Técnicas de Inativação de Genes , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Precursores de Proteínas/deficiência , Receptores da Neurocinina-1/deficiência , Receptores da Neurocinina-1/genética , Substância P/deficiência , Linfócitos T/citologia , Linfócitos T/metabolismo , Taquicininas/deficiência
11.
PLoS One ; 8(1): e53161, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23326393

RESUMO

Hoxb4, a 3'-located Hox gene, enhances hematopoietic stem cell (HSC) activity, while a subset of 5'-located Hox genes is involved in hematopoiesis and leukemogenesis, and some of them are common translocation partners for Nucleoporin 98 (Nup98) in patients with leukemia. Although these Hox gene derivatives are believed to act as transcription regulators, the molecular involvement of the Hox gene derivatives in hematopoiesis and leukemogenesis remains largely elusive. Since we previously showed that Hoxb4 forms a complex with a Roc1-Ddb1-Cul4a ubiquitin ligase core component and functions as an E3 ubiquitin ligase activator for Geminin, we here examined the E3 ubiquitin ligase activities of the 5'-located Hox genes, Hoxa9 and Hoxc13, and Nup98-Hoxa9. Hoxa9 formed a similar complex with the Roc1-Ddb1-Cul4a component to induce ubiquitination of Geminin, but the others did not. Retroviral transduction-mediated overexpression or siRNA-mediated knock-down of Hoxa9 respectively down-regulated or up-regulated Geminin in hematopoietic cells. And Hoxa9 transduction-induced repopulating and clonogenic activities were suppressed by Geminin supertransduction. These findings suggest that Hoxa9 and Hoxb4 differ from Hoxc13 and Nup98-Hoxa9 in their molecular role in hematopoiesis, and that Hoxa9 induces the activity of HSCs and hematopoietic progenitors at least in part through direct down-regulation of Geminin.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Regulação para Baixo , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Homeodomínio/metabolismo , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/genética , Ensaio de Unidades Formadoras de Colônias/métodos , Proteínas Culina/genética , Proteínas Culina/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Geminina , Células HEK293 , Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/genética , Humanos , Immunoblotting , Camundongos , Camundongos Endogâmicos C57BL , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Ligação Proteica , Interferência de RNA , Retroviridae/genética , Células Sf9 , Transdução Genética , Ubiquitinação
12.
Blood ; 119(11): 2510-22, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22174158

RESUMO

The stem cell-intrinsic model of self-renewal via asymmetric cell division (ACD) posits that fate determinants be partitioned unequally between daughter cells to either activate or suppress the stemness state. ACD is a purported mechanism by which hematopoietic stem cells (HSCs) self-renew, but definitive evidence for this cellular process remains open to conjecture. To address this issue, we chose 73 candidate genes that function within the cell polarity network to identify potential determinants that may concomitantly alter HSC fate while also exhibiting asymmetric segregation at cell division. Initial gene-expression profiles of polarity candidates showed high and differential expression in both HSCs and leukemia stem cells. Altered HSC fate was assessed by our established in vitro to in vivo screen on a subcohort of candidate polarity genes, which revealed 6 novel positive regulators of HSC function: Ap2a2, Gpsm2, Tmod1, Kif3a, Racgap1, and Ccnb1. Interestingly, live-cell videomicroscopy of the endocytic protein AP2A2 shows instances of asymmetric segregation during HSC/progenitor cell cytokinesis. These results contribute further evidence that ACD is functional in HSC self-renewal, suggest a role for Ap2a2 in HSC activity, and provide a unique opportunity to prospectively analyze progeny from HSC asymmetric divisions.


Assuntos
Complexo 2 de Proteínas Adaptadoras/metabolismo , Subunidades alfa do Complexo de Proteínas Adaptadoras/metabolismo , Divisão Celular Assimétrica/fisiologia , Polaridade Celular/genética , Endocitose/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Neoplásicas/patologia , Células-Tronco/citologia , Complexo 2 de Proteínas Adaptadoras/antagonistas & inibidores , Complexo 2 de Proteínas Adaptadoras/genética , Subunidades alfa do Complexo de Proteínas Adaptadoras/antagonistas & inibidores , Subunidades alfa do Complexo de Proteínas Adaptadoras/genética , Animais , Biomarcadores/metabolismo , Western Blotting , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Citometria de Fluxo , Perfilação da Expressão Gênica , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/fisiologia , Leucemia/metabolismo , Leucemia/patologia , Camundongos , Células-Tronco Neoplásicas/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/fisiologia
14.
Blood ; 117(16): 4253-61, 2011 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-21398220

RESUMO

Src homology 2 domain-containing phosphatase 2 (Shp2), encoded by Ptpn11, is a member of the nonreceptor protein-tyrosine phosphatase family, and functions in cell survival, proliferation, migration, and differentiation in many tissues. Here we report that loss of Ptpn11 in murine hematopoietic cells leads to bone marrow aplasia and lethality. Mutant mice show rapid loss of hematopoietic stem cells (HSCs) and immature progenitors of all hematopoietic lineages in a gene dosage-dependent and cell-autonomous manner. Ptpn11-deficient HSCs and progenitors undergo apoptosis concomitant with increased Noxa expression. Mutant HSCs/progenitors also show defective Erk and Akt activation in response to stem cell factor and diminished thrombopoietin-evoked Erk activation. Activated Kras alleviates the Ptpn11 requirement for colony formation by progenitors and cytokine/growth factor responsiveness of HSCs, indicating that Ras is functionally downstream of Shp2 in these cells. Thus, Shp2 plays a critical role in controlling the survival and maintenance of HSCs and immature progenitors in vivo.


Assuntos
Medula Óssea/patologia , Deleção de Genes , Células-Tronco Hematopoéticas/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Animais , Ciclo Celular , Morte Celular , Epistasia Genética , Células-Tronco Hematopoéticas/citologia , Camundongos , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Células-Tronco/citologia , Células-Tronco/metabolismo
15.
Cell Stem Cell ; 7(1): 101-13, 2010 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-20621054

RESUMO

In this study, we describe an in vivo RNA interference functional genetics approach to evaluate the role of 20 different conserved polarity factors and fate determinants in mouse hematopoietic stem cell (HSC) activity. In total, this screen revealed three enhancers and one suppressor of HSC-derived reconstitution. Pard6a, Prkcz, and Msi2 shRNA-mediated depletion significantly impaired HSC repopulation. An in vitro promotion of differentiation was observed after the silencing of these genes, consistent with their function in regulating HSC self-renewal. Conversely, Prox1 knockdown led to in vivo accumulation of primitive and differentiated cells. HSC activity was also enhanced in vitro when Prox1 levels were experimentally reduced, identifying it as a potential antagonist of self-renewal. HSC engineered to overexpress Msi2 or Prox1 showed the reverse phenotype to those transduced with corresponding shRNA vectors. Gene expression profiling studies identified a number of known HSC and cell cycle regulators as potential downstream targets to Msi2 and Prox1.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Proteínas de Homeodomínio/metabolismo , Interferência de RNA/fisiologia , Proteínas de Ligação a RNA/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Ciclo Celular/genética , Ciclo Celular/fisiologia , Células Cultivadas , Citometria de Fluxo , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Proteínas de Ligação a RNA/genética , Retroviridae/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Supressoras de Tumor/genética
16.
Blood ; 116(19): 3792-801, 2010 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-20660792

RESUMO

Hemokinin-1 (HK-1), encoded by the TAC4 gene, is a tachykinin peptide that is predominantly expressed in non-neuronal cells, such as immune cells. We have disrupted the mouse TAC4 gene to obtain a better understanding of the actions of HK-1 during hematopoiesis. We demonstrate here that TAC4(-/-) mice exhibit an increase of CD19(+)CD117(+)HSA(+)BP.1(-) "fraction B" pro-B cells in the bone marrow, whereas pre-B, immature, and mature B cells are within the normal range. We show that in vitro cultures derived from TAC4(-/-) bone marrow, sorted "fraction B" pro-B cells or purified long-term reconstituting stem cells, contain significantly higher numbers of pro-B cells compared with controls, suggesting an inhibitory role for HK-1 on developing B cells. Supporting this idea, we show that addition of HK-1 to cultures established from long-term reconstituting stem cells and the newly described intermediate-term reconstituting stem cells leads to a significant decrease of de novo generated pro-B cells. Based on our studies, we postulate that HK-1 plays an inhibitory role in hematopoiesis, and we hypothesize that it may be part of the bone marrow microenvironment that supports and regulates the proliferation and differentiation of hematopoietic cells.


Assuntos
Linfopoese/genética , Linfopoese/fisiologia , Células Precursoras de Linfócitos B/citologia , Células Precursoras de Linfócitos B/imunologia , Precursores de Proteínas/deficiência , Precursores de Proteínas/genética , Taquicininas/deficiência , Taquicininas/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Primers do DNA/genética , Feminino , Expressão Gênica , Marcação de Genes , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Técnicas In Vitro , Linfopoese/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Imunológicos , Precursores de Proteínas/imunologia , Precursores de Proteínas/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores da Neurocinina-1/genética , Taquicininas/imunologia , Taquicininas/fisiologia
17.
Cell Stem Cell ; 6(1): 48-58, 2010 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-20074534

RESUMO

Sustained blood cell production depends on divisions by hematopoietic stem cells (HSCs) that yield both differentiating progeny as well as new HSCs via self-renewal. Differentiating progeny remain capable of self-renewal, but only HSCs sustain self-renewal through successive divisions securely enough to maintain clones that persist life-long. Until recently, the first identified next stage consisted of "short-term" reconstituting cells able to sustain clones of differentiating cells for only 4-6 weeks. Here we expand evidence for a numerically dominant "intermediate-term" multipotent HSC stage in mice whose clones persist for 6-8 months before becoming extinct and that are separable from both short-term as well as permanently reconstituting "long-term" HSCs. The findings suggest that the first step in stem cell differentiation consists not in loss of initial capacity for serial self-renewal divisions, but rather in loss of mechanisms that stabilize self-renewing behavior throughout successive future stem cell divisions.


Assuntos
Diferenciação Celular , Divisão Celular , Células-Tronco Hematopoéticas/citologia , Animais , Antígenos CD/genética , Antígenos CD34/genética , Linhagem da Célula , Separação Celular , Células Cultivadas , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Integrina alfa2/genética , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Superfície Celular/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Fatores de Tempo , Tirosina Quinase 3 Semelhante a fms/genética
18.
Blood ; 115(4): 792-803, 2010 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-19850742

RESUMO

The majority of long-term reconstituting hematopoietic stem cells (LT-HSCs) in the adult is in G(0), whereas a large proportion of progenitors are more cycling. We show here that the SCL/TAL1 transcription factor is highly expressed in LT-HSCs compared with short-term reconstituting HSCs and progenitors and that SCL negatively regulates the G(0)-G(1) transit of LT-HSCs. Furthermore, when SCL protein levels are decreased by gene targeting or by RNA interference, the reconstitution potential of HSCs is impaired in several transplantation assays. First, the mean stem cell activity of HSCs transplanted at approximately 1 competitive repopulating unit was 2-fold decreased when Scl gene dosage was decreased. Second, Scl(+/-) HSCs were at a marked competitive disadvantage with Scl(+/+) cells when transplanted at 4 competitive repopulating units equivalent. Third, reconstitution of the stem cell pool by adult HSCs expressing Scl-directed shRNAs was decreased compared with controls. At the molecular level, we found that SCL occupies the Cdkn1a and Id1 loci in primary hematopoietic cells and that the expression levels of these 2 regulators of HSC cell cycle and long-term functions are sensitive to Scl gene dosage. Together, our observations suggest that SCL impedes G(0)-G(1) transition in HSCs and regulates their long-term competence.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Animais , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Fase G1/fisiologia , Expressão Gênica/fisiologia , Sobrevivência de Enxerto , Células-Tronco Hematopoéticas/efeitos dos fármacos , Proteína 1 Inibidora de Diferenciação/genética , Interleucina-11/farmacologia , Interleucina-6/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Interferência de RNA , Fase de Repouso do Ciclo Celular/fisiologia , Fator de Células-Tronco/farmacologia , Proteína 1 de Leucemia Linfocítica Aguda de Células T
19.
Proc Natl Acad Sci U S A ; 105(51): 20286-90, 2008 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-19095794

RESUMO

The properties and biology of mRNA transcripts can be affected profoundly by the choice of alternative polyadenylation sites, making definition of the 3' ends of transcripts essential for understanding their regulation. Here we show that 22-52% of sequences in commonly used human and murine "full-length" transcript databases may not currently end at bona fide polyadenylation sites. To identify probable transcript termini over the entire murine and human genomes, we analyzed the EST databases for positional clustering of EST ends. The analysis yielded 58,282 murine- and 86,410 human-candidate polyadenylation sites, of which 75% mapped to 23,091 known murine transcripts and 22,891 known human transcripts. The murine dataset correctly predicted 97% of the 3' ends in a manually curated and experimentally supported benchmark transcript set. Of currently known genes, 15% had no associated prediction and 25% had only a single predicted termination site. The remaining genes had an average of 3-4 alternative polyadenylation sites predicted for each murine or human transcript, respectively. The results are made available in the form of tables and an interactive web site that can be mined for rapid assessment of the validity of 3' ends in existing collections, enumeration of potential alternative 3' polyadenylation sites of known transcripts, direct retrieval of terminal sequences for design of probes, and detection of polyadenylation sites not currently mapped to known genes.


Assuntos
Região 3'-Flanqueadora , Análise por Conglomerados , Etiquetas de Sequências Expressas , Animais , Humanos , Métodos , Camundongos , Poli A , Poliadenilação
20.
Cell Stem Cell ; 3(1): 109-18, 2008 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-18593563

RESUMO

Mature mammary epithelial cells are generated from undifferentiated precursors through a hierarchical process, but the molecular mechanisms involved, particularly in the human mammary gland, are poorly understood. To address this issue, we isolated highly purified subpopulations of primitive bipotent and committed luminal progenitor cells as well as mature luminal and myoepithelial cells from normal human mammary tissue and compared their transcriptomes obtained using three different methods. Elements unique to each subset of mammary cells were identified, and changes that accompany their differentiation in vivo were shown to be recapitulated in vitro. These include a stage-specific change in NOTCH pathway gene expression during the commitment of bipotent progenitors to the luminal lineage. Functional studies further showed NOTCH3 signaling to be critical for this differentiation event to occur in vitro. Taken together, these findings provide an initial foundation for future delineation of mechanisms that perturb primitive human mammary cell growth and differentiation.


Assuntos
Mama/citologia , Mama/fisiologia , Células Epiteliais/citologia , Antígenos CD/genética , Diferenciação Celular , Células Epiteliais/fisiologia , Feminino , Perfilação da Expressão Gênica , Humanos , Gravidez , Receptores Notch/genética , Valores de Referência , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA