Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Blood ; 142(6): 519-532, 2023 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-37339578

RESUMO

Developmental hematopoiesis consists of multiple, partially overlapping hematopoietic waves that generate the differentiated blood cells required for embryonic development while establishing a pool of undifferentiated hematopoietic stem cells (HSCs) for postnatal life. This multilayered design in which active hematopoiesis migrates through diverse extra and intraembryonic tissues has made it difficult to define a roadmap for generating HSCs vs non-self-renewing progenitors, especially in humans. Recent single-cell studies have helped in identifying the rare human HSCs at stages when functional assays are unsuitable for distinguishing them from progenitors. This approach has made it possible to track the origin of human HSCs to the unique type of arterial endothelium in the aorta-gonad-mesonephros region and document novel benchmarks for HSC migration and maturation in the conceptus. These studies have delivered new insights into the intricate process of HSC generation and provided tools to inform the in vitro efforts to replicate the physiological developmental journey from pluripotent stem cells via distinct mesodermal and endothelial intermediates to HSCs.


Assuntos
Embrião de Mamíferos , Células-Tronco Hematopoéticas , Feminino , Gravidez , Humanos , Hematopoese/fisiologia , Mesonefro
2.
Nature ; 604(7906): 534-540, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35418685

RESUMO

The ontogeny of human haematopoietic stem cells (HSCs) is poorly defined owing to the inability to identify HSCs as they emerge and mature at different haematopoietic sites1. Here we created a single-cell transcriptome map of human haematopoietic tissues from the first trimester to birth and found that the HSC signature RUNX1+HOXA9+MLLT3+MECOM+HLF+SPINK2+ distinguishes HSCs from progenitors throughout gestation. In addition to the aorta-gonad-mesonephros region, nascent HSCs populated the placenta and yolk sac before colonizing the liver at 6 weeks. A comparison of HSCs at different maturation stages revealed the establishment of HSC transcription factor machinery after the emergence of HSCs, whereas their surface phenotype evolved throughout development. The HSC transition to the liver marked a molecular shift evidenced by suppression of surface antigens reflecting nascent HSC identity, and acquisition of the HSC maturity markers CD133 (encoded by PROM1) and HLA-DR. HSC origin was tracked to ALDH1A1+KCNK17+ haemogenic endothelial cells, which arose from an IL33+ALDH1A1+ arterial endothelial subset termed pre-haemogenic endothelial cells. Using spatial transcriptomics and immunofluorescence, we visualized this process in ventrally located intra-aortic haematopoietic clusters. The in vivo map of human HSC ontogeny validated the generation of aorta-gonad-mesonephros-like definitive haematopoietic stem and progenitor cells from human pluripotent stem cells, and serves as a guide to improve their maturation to functional HSCs.


Assuntos
Células Endoteliais , Células-Tronco Hematopoéticas , Diferenciação Celular , Endotélio , Feminino , Hematopoese , Humanos , Mesonefro , Gravidez
3.
J Exp Med ; 219(3)2022 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-35201267

RESUMO

Atkins et al. (2022. J. Exp. Med.https://doi.org/10.1084/jem.20211924) create a PSC differentiation model for human yolk sac hematopoiesis and discover multipotent progenitors with erythro-myeloid and T lymphoid potential. The multipotent progenitors emerge via hemogenic endothelium and share origin with primitive erythroid wave in KDR+CD235a/b+ mesoderm.


Assuntos
Hematopoese , Saco Vitelino , Humanos , Mesoderma
4.
Adv Sci (Weinh) ; 8(4): 2002500, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33643791

RESUMO

Ischemia impacts multiple organ systems and is the major cause of morbidity and mortality in the developed world. Ischemia disrupts tissue homeostasis, driving cell death, and damages tissue structure integrity. Strategies to heal organs, like the infarcted heart, or to replace cells, as done in pancreatic islet ß-cell transplantations, are often hindered by ischemic conditions. Here, it is discovered that the basement membrane glycoprotein nidogen-1 attenuates the apoptotic effect of hypoxia in cardiomyocytes and pancreatic ß-cells via the αvß3 integrin and beneficially modulates immune responses in vitro. It is shown that nidogen-1 significantly increases heart function and angiogenesis, while reducing fibrosis, in a mouse postmyocardial infarction model. These results demonstrate the protective and regenerative potential of nidogen-1 in ischemic conditions.

5.
Blood ; 136(16): 1871-1883, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32842144

RESUMO

Hematopoietic stem cells (HSCs) reside in the bone marrow (BM) stem cell niche, which provides a vital source of HSC regulatory signals. Radiation and chemotherapy disrupt the HSC niche, including its sinusoidal vessels and perivascular cells, contributing to delayed hematopoietic recovery. Thus, identification of factors that can protect the HSC niche during an injury could offer a significant therapeutic opportunity to improve hematopoietic regeneration. In this study, we identified a critical function for vascular endothelial growth factor-C (VEGF-C), that of maintaining the integrity of the BM perivascular niche and improving BM niche recovery after irradiation-induced injury. Both global and conditional deletion of Vegfc in endothelial or leptin receptor-positive (LepR+) cells led to a disruption of the BM perivascular niche. Furthermore, deletion of Vegfc from the microenvironment delayed hematopoietic recovery after transplantation by decreasing endothelial proliferation and LepR+ cell regeneration. Exogenous administration of VEGF-C via an adenoassociated viral vector improved hematopoietic recovery after irradiation by accelerating endothelial and LepR+ cell regeneration and by increasing the expression of hematopoietic regenerative factors. Our results suggest that preservation of the integrity of the perivascular niche via VEGF-C signaling could be exploited therapeutically to enhance hematopoietic regeneration.


Assuntos
Células da Medula Óssea/metabolismo , Medula Óssea/metabolismo , Células Endoteliais/metabolismo , Nicho de Células-Tronco , Fator C de Crescimento do Endotélio Vascular/genética , Animais , Biomarcadores , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos da radiação , Expressão Gênica , Hematopoese/genética , Hematopoese/efeitos da radiação , Imunofenotipagem , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Ligação Proteica , RNA Mensageiro , Receptores para Leptina/metabolismo , Nicho de Células-Tronco/genética , Nicho de Células-Tronco/efeitos da radiação , Fator C de Crescimento do Endotélio Vascular/metabolismo
6.
Nature ; 576(7786): 281-286, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31776511

RESUMO

Limited knowledge of the mechanisms that govern the self-renewal of human haematopoietic stem cells (HSCs), and why this fails in culture, have impeded the expansion of HSCs for transplantation1. Here we identify MLLT3 (also known as AF9) as a crucial regulator of HSCs that is highly enriched in human fetal, neonatal and adult HSCs, but downregulated in culture. Depletion of MLLT3 prevented the maintenance of transplantable human haematopoietic stem or progenitor cells (HSPCs) in culture, whereas stabilizing MLLT3 expression in culture enabled more than 12-fold expansion of transplantable HSCs that provided balanced multilineage reconstitution in primary and secondary mouse recipients. Similar to endogenous MLLT3, overexpressed MLLT3 localized to active promoters in HSPCs, sustained levels of H3K79me2 and protected the HSC transcriptional program in culture. MLLT3 thus acts as HSC maintenance factor that links histone reader and modifying activities to modulate HSC gene expression, and may provide a promising approach to expand HSCs for transplantation.


Assuntos
Autorrenovação Celular , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas Nucleares/metabolismo , Animais , Células Cultivadas , Regulação da Expressão Gênica , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos , Proteínas Nucleares/genética , Ligação Proteica
7.
Nat Commun ; 9(1): 3634, 2018 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-30194383

RESUMO

Tissue-specific gene expression defines cellular identity and function, but knowledge of early human development is limited, hampering application of cell-based therapies. Here we profiled 5 distinct cell types at a single fetal stage, as well as chondrocytes at 4 stages in vivo and 2 stages during in vitro differentiation. Network analysis delineated five tissue-specific gene modules; these modules and chromatin state analysis defined broad similarities in gene expression during cartilage specification and maturation in vitro and in vivo, including early expression and progressive silencing of muscle- and bone-specific genes. Finally, ontogenetic analysis of freshly isolated and pluripotent stem cell-derived articular chondrocytes identified that integrin alpha 4 defines 2 subsets of functionally and molecularly distinct chondrocytes characterized by their gene expression, osteochondral potential in vitro and proliferative signature in vivo. These analyses provide new insight into human musculoskeletal development and provide an essential comparative resource for disease modeling and regenerative medicine.


Assuntos
Condrócitos/metabolismo , Condrogênese , Mioblastos/metabolismo , Osteoblastos/metabolismo , Tenócitos/metabolismo , Animais , Biomarcadores/metabolismo , Epigênese Genética , Desenvolvimento Fetal , Perfilação da Expressão Gênica , Código das Histonas , Humanos , Camundongos , Análise de Sequência de RNA , Suínos , Transcrição Gênica , Transcriptoma
8.
Exp Hematol ; 65: 1-16, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29981365

RESUMO

Hematopoietic stem cells (HSCs) are multipotent cells responsible for the maintenance of the hematopoietic system throughout life. Dysregulation of the balance in HSC self-renewal, death, and differentiation can have serious consequences such as myelodysplastic syndromes or leukemia. All-trans retinoic acid (ATRA), the biologically active metabolite of vitamin A/RA, has been shown to have pleiotropic effects on hematopoietic cells, enhancing HSC self-renewal while also increasing differentiation of more mature progenitors. Furthermore, ATRA has been shown to have key roles in regulating the specification and formation of hematopoietic cells from pluripotent stem cells including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). Here, we summarize the known roles of vitamin A and RA receptors in the regulation of hematopoiesis from HSCs, ES, and iPSCs.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Pluripotentes/citologia , Retinoides/metabolismo , Hematopoese , Células-Tronco Hematopoéticas/fisiologia , Humanos , Modelos Biológicos , Receptores do Ácido Retinoico/fisiologia , Transdução de Sinais
9.
Circulation ; 138(12): 1224-1235, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-29950403

RESUMO

BACKGROUND: Genetic diversity and the heterogeneous nature of cardiac fibroblasts (CFbs) have hindered characterization of the molecular mechanisms that regulate cardiac fibrosis. The Hybrid Mouse Diversity Panel offers a valuable tool to examine genetically diverse cardiac fibroblasts and their role in fibrosis. METHODS: Three strains of mice (C57BL/6J, C3H/HeJ, and KK/HlJ) were selected from the Hybrid Mouse Diversity Panel and treated with either isoproterenol (ISO) or saline by an intraperitoneally implanted osmotic pump. After 21 days, cardiac function and levels of fibrosis were measured by echocardiography and trichrome staining, respectively. Activation and proliferation of CFbs were measured by in vitro and in vivo assays under normal and injury conditions. RNA sequencing was done on isolated CFbs from each strain. Results were analyzed by Ingenuity Pathway Analysis and validated by reverse transcription-qPCR, immunohistochemistry, and ELISA. RESULTS: ISO treatment in C57BL/6J, C3H/HeJ, and KK/HlJ mice resulted in minimal, moderate, and extensive levels of fibrosis, respectively (n=7-8 hearts per condition). Isolated CFbs treated with ISO exhibited strain-specific increases in the levels of activation but showed comparable levels of proliferation. Similar results were found in vivo, with fibroblast activation, and not proliferation, correlating with the differential levels of cardiac fibrosis after ISO treatment. RNA sequencing revealed that CFbs from each strain exhibit unique gene expression changes in response to ISO. We identified Ltbp2 as a commonly upregulated gene after ISO treatment. Expression of LTBP2 was elevated and specifically localized in the fibrotic regions of the myocardium after injury in mice and in human heart failure patients. CONCLUSIONS: This study highlights the importance of genetic variation in cardiac fibrosis by using multiple inbred mouse strains to characterize CFbs and their response to ISO treatment. Our data suggest that, although fibroblast activation is a response that parallels the extent of scar formation, proliferation may not necessarily correlate with levels of fibrosis. In addition, by comparing CFbs from multiple strains, we identified pathways as potential therapeutic targets and LTBP2 as a marker for fibrosis, with relevance to patients with underlying myocardial fibrosis.


Assuntos
Cardiomiopatias/genética , Cardiomiopatias/patologia , Proliferação de Células , Fibroblastos/patologia , Variação Genética , Proteínas de Ligação a TGF-beta Latente/genética , Animais , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Feminino , Fibroblastos/metabolismo , Fibrose , Predisposição Genética para Doença , Isoproterenol , Proteínas de Ligação a TGF-beta Latente/metabolismo , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Fenótipo , Especificidade da Espécie , Transcriptoma
10.
Nat Commun ; 9(1): 754, 2018 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-29467410

RESUMO

The cellular mechanisms driving cardiac tissue formation remain poorly understood, largely due to the structural and functional complexity of the heart. It is unclear whether newly generated myocytes originate from cardiac stem/progenitor cells or from pre-existing cardiomyocytes that re-enter the cell cycle. Here, we identify the source of new cardiomyocytes during mouse development and after injury. Our findings suggest that cardiac progenitors maintain proliferative potential and are the main source of cardiomyocytes during development; however, the onset of αMHC expression leads to reduced cycling capacity. Single-cell RNA sequencing reveals a proliferative, "progenitor-like" population abundant in early embryonic stages that decreases to minimal levels postnatally. Furthermore, cardiac injury by ligation of the left anterior descending artery was found to activate cardiomyocyte proliferation in neonatal but not adult mice. Our data suggest that clonal dominance of differentiating progenitors mediates cardiac development, while a distinct subpopulation of cardiomyocytes may have the potential for limited proliferation during late embryonic development and shortly after birth.


Assuntos
Traumatismos Cardíacos/patologia , Coração/crescimento & desenvolvimento , Miócitos Cardíacos/citologia , Animais , Animais Recém-Nascidos , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Células-Tronco Embrionárias/citologia , Feminino , Coração Fetal/citologia , Coração Fetal/crescimento & desenvolvimento , Traumatismos Cardíacos/genética , Masculino , Camundongos , Camundongos Transgênicos , Mioblastos Cardíacos/citologia , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Pericárdio/citologia , Pericárdio/embriologia , Pericárdio/crescimento & desenvolvimento , Gravidez , Análise de Sequência de RNA
11.
Cell Rep ; 21(12): 3514-3523, 2017 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-29262330

RESUMO

The transcription factor hepatic leukemia factor (HLF) is strongly expressed in hematopoietic stem cells (HSCs) and is thought to influence both HSC self-renewal and leukemogenesis. However, the physiological role of HLF in hematopoiesis and HSC function is unclear. Here, we report that mice lacking Hlf are viable with essentially normal hematopoietic parameters, including an intact HSC pool during steady-state hematopoiesis. In contrast, when challenged through transplantation, Hlf-deficient HSCs showed an impaired ability to reconstitute hematopoiesis and became gradually exhausted upon serial transplantation. Transcriptional profiling of Hlf-deficient HSCs revealed changes associated with enhanced cellular activation, and cell-cycle analysis demonstrated a significant reduction of quiescent HSCs. Accordingly, toxic insults targeting dividing cells completely eradicated the HSC pool in Hlf-deficient mice. In summary, our findings point to HLF as a critical regulator of HSC quiescence and as an essential factor for maintaining the HSC pool during regeneration.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Proliferação de Células , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Animais , Apoptose , Fatores de Transcrição de Zíper de Leucina Básica/genética , Células Cultivadas , Dano ao DNA , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Transcriptoma
12.
Cell Rep ; 17(9): 2286-2298, 2016 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-27880904

RESUMO

The contribution of the different waves and sites of developmental hematopoiesis to fetal and adult blood production remains unclear. Here, we identify lymphatic vessel endothelial hyaluronan receptor-1 (LYVE1) as a marker of yolk sac (YS) endothelium and definitive hematopoietic stem and progenitor cells (HSPCs). Endothelium in mid-gestation YS and vitelline vessels, but not the dorsal aorta and placenta, were labeled by Lyve1-Cre. Most YS HSPCs and erythro-myeloid progenitors were Lyve1-Cre lineage traced, but primitive erythroid cells were not, suggesting that they represent distinct lineages. Fetal liver (FL) and adult HSPCs showed 35%-40% Lyve1-Cre marking. Analysis of circulation-deficient Ncx1-/- concepti identified the YS as a major source of Lyve1-Cre labeled HSPCs. FL proerythroblast marking was extensive at embryonic day (E) 11.5-13.5, but decreased to hematopoietic stem cell (HSC) levels by E16.5, suggesting that HSCs from multiple sources became responsible for erythropoiesis. Lyve1-Cre thus marks the divergence between YS primitive and definitive hematopoiesis and provides a tool for targeting YS definitive hematopoiesis and FL colonization.


Assuntos
Linhagem da Célula , Células Eritroides/citologia , Células Eritroides/metabolismo , Hemangioblastos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Saco Vitelino/metabolismo , Envelhecimento , Animais , Eritropoese , Feminino , Feto/citologia , Deleção de Genes , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Integrases/metabolismo , Fígado/embriologia , Camundongos Endogâmicos C57BL , Gravidez , Fatores de Tempo
13.
Nat Biotechnol ; 34(11): 1168-1179, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27748754

RESUMO

The ability to generate hematopoietic stem cells from human pluripotent cells would enable many biomedical applications. We find that hematopoietic CD34+ cells in spin embryoid bodies derived from human embryonic stem cells (hESCs) lack HOXA expression compared with repopulation-competent human cord blood CD34+ cells, indicating incorrect mesoderm patterning. Using reporter hESC lines to track the endothelial (SOX17) to hematopoietic (RUNX1C) transition that occurs in development, we show that simultaneous modulation of WNT and ACTIVIN signaling yields CD34+ hematopoietic cells with HOXA expression that more closely resembles that of cord blood. The cultures generate a network of aorta-like SOX17+ vessels from which RUNX1C+ blood cells emerge, similar to hematopoiesis in the aorta-gonad-mesonephros (AGM). Nascent CD34+ hematopoietic cells and corresponding cells sorted from human AGM show similar expression of cell surface receptors, signaling molecules and transcription factors. Our findings provide an approach to mimic in vitro a key early stage in human hematopoiesis for the generation of AGM-derived hematopoietic lineages from hESCs.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/metabolismo , Mesonefro/citologia , Mesonefro/embriologia , Neovascularização Fisiológica/fisiologia , Aorta/citologia , Aorta/embriologia , Aorta/crescimento & desenvolvimento , Diferenciação Celular/fisiologia , Células Cultivadas , Células-Tronco Embrionárias/fisiologia , Gônadas/citologia , Gônadas/embriologia , Gônadas/crescimento & desenvolvimento , Células-Tronco Hematopoéticas/fisiologia , Humanos , Mesonefro/crescimento & desenvolvimento
14.
Nat Commun ; 7: 12376, 2016 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-27507714

RESUMO

DNA double strand break (DSB) repair is critical for generation of B-cell receptors, which are pre-requisite for B-cell progenitor survival. However, the transcription factors that promote DSB repair in B cells are not known. Here we show that MEF2C enhances the expression of DNA repair and recombination factors in B-cell progenitors, promoting DSB repair, V(D)J recombination and cell survival. Although Mef2c-deficient mice maintain relatively intact peripheral B-lymphoid cellularity during homeostasis, they exhibit poor B-lymphoid recovery after sub-lethal irradiation and 5-fluorouracil injection. MEF2C binds active regulatory regions with high-chromatin accessibility in DNA repair and V(D)J genes in both mouse B-cell progenitors and human B lymphoblasts. Loss of Mef2c in pre-B cells reduces chromatin accessibility in multiple regulatory regions of the MEF2C-activated genes. MEF2C therefore protects B lymphopoiesis during stress by ensuring proper expression of genes that encode DNA repair and B-cell factors.


Assuntos
Quebras de DNA de Cadeia Dupla , Hematopoese/fisiologia , Células Precursoras de Linfócitos B/fisiologia , Recombinação V(D)J/fisiologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Sobrevivência Celular/efeitos da radiação , Cromatina/metabolismo , Feminino , Fluoruracila/farmacologia , Hematopoese/efeitos dos fármacos , Hematopoese/efeitos da radiação , Fatores de Transcrição MEF2/fisiologia , Masculino , Camundongos , Células Precursoras de Linfócitos B/efeitos dos fármacos , Células Precursoras de Linfócitos B/efeitos da radiação , Irradiação Corporal Total/efeitos adversos
15.
Blood ; 128(5): 710-20, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27343251

RESUMO

Vascular endothelial growth factor C (VEGF-C) is a major driver of lymphangiogenesis in embryos and adults. Vegfc gene deletion in mouse embryos results in failure of lymphangiogenesis, fluid accumulation in tissues, and lethality. The VEGF-C receptors VEGFR3 and VEGFR2 are required for embryonic blood vessel formation. The related VEGF is essential for both blood vessel formation and embryonic hematopoiesis, whereas the possible involvement of VEGF-C in hematopoiesis is unknown. Here we unveil a novel hematopoietic function of VEGF-C in fetal erythropoiesis. Deletion of Vegfc in embryonic day 7.5 (E7.5) embryos in the C57BL6 mouse genetic background led to defective fetal erythropoiesis, characterized by anemia and lack of enucleated red blood cells in blood circulation. Macrophages and erythroid cells in the fetal liver (FL) were also decreased after midgestation because of decreased cell proliferation and increased apoptosis. However, the Lin(-)Sca-1(+)c-Kit(+) stem cell compartment in E14.5 FL was not affected by Vegfc deletion. VEGF-C loss did not disrupt the generation of primitive erythroid cells or erythro-myeloid progenitors (EMPs) in the yolk sac, but it decreased the expression of α4-integrin on EMPs and compromised EMP colonization of the FL. The distribution, maturation, and enucleation of primitive erythroblasts were also impaired by Vegfc deletion. In contrast, Vegfc deletion from E10.5 onward did not compromise definitive hematopoiesis in the liver, and Vegfc deletion in adult mice did not cause anemia. These results reveal an unexpected role for VEGF-C, a major lymphangiogenic growth factor, in the transition to FL erythropoiesis.


Assuntos
Eritropoese , Feto/metabolismo , Fator C de Crescimento do Endotélio Vascular/metabolismo , Anemia/patologia , Animais , Apoptose , Linhagem da Célula , Proliferação de Células , Embrião de Mamíferos/metabolismo , Células Eritroides/metabolismo , Deleção de Genes , Hepatócitos/metabolismo , Integrina alfa4/metabolismo , Fígado/irrigação sanguínea , Fígado/embriologia , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Tamanho do Órgão
16.
Nat Cell Biol ; 18(6): 595-606, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27183470

RESUMO

Pluripotent stem cells (PSCs) may provide a potential source of haematopoietic stem/progenitor cells (HSPCs) for transplantation; however, unknown molecular barriers prevent the self-renewal of PSC-HSPCs. Using two-step differentiation, human embryonic stem cells (hESCs) differentiated in vitro into multipotent haematopoietic cells that had the CD34(+)CD38(-/lo)CD90(+)CD45(+)GPI-80(+) fetal liver (FL) HSPC immunophenotype, but exhibited poor expansion potential and engraftment ability. Transcriptome analysis of immunophenotypic hESC-HSPCs revealed that, despite their molecular resemblance to FL-HSPCs, medial HOXA genes remained suppressed. Knockdown of HOXA7 disrupted FL-HSPC function and caused transcriptome dysregulation that resembled hESC-derived progenitors. Overexpression of medial HOXA genes prolonged FL-HSPC maintenance but was insufficient to confer self-renewal to hESC-HSPCs. Stimulation of retinoic acid signalling during endothelial-to-haematopoietic transition induced the HOXA cluster and other HSC/definitive haemogenic endothelium genes, and prolonged HSPC maintenance in culture. Thus, medial HOXA gene expression induced by retinoic acid signalling marks the establishment of the definitive HSPC fate and controls HSPC identity and function.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula , Genes Homeobox/genética , Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/metabolismo , Células-Tronco Multipotentes/citologia , Antígenos CD34/metabolismo , Células Cultivadas , Perfilação da Expressão Gênica/métodos , Proteínas de Homeodomínio/genética , Humanos , Antígenos Comuns de Leucócito/metabolismo , Transcriptoma
17.
Dev Cell ; 36(5): 479-80, 2016 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-26954540

RESUMO

Reporting in Developmental Cell, Pereira et al. (2016) use in vitro lineage reprogramming insights to inform understanding of hematopoietic stem cell (HSC) development in vivo. They find Prom1(+)Sca1(+)CD34(+)CD45(-) hemogenic precursors, akin to fibroblast-derived hemato-vascular precursors, in mouse placenta and embryo. The cells mature into transplantable HSCs in culture.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Reprogramação Celular , Hematopoese/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Embrionárias Murinas/citologia , Animais , Feminino , Gravidez
18.
J Transl Med ; 13: 98, 2015 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-25889765

RESUMO

BACKGROUND: Dormant leukemia stem cells (LSC) promote therapeutic resistance and leukemic progression as a result of unbridled activation of stem cell gene expression programs. Thus, we hypothesized that 1) deregulation of the hedgehog (Hh) stem cell self-renewal and cell cycle regulatory pathway would promote dormant human LSC generation and 2) that PF-04449913, a clinical antagonist of the GLI2 transcriptional activator, smoothened (SMO), would enhance dormant human LSC eradication. METHODS: To test these postulates, whole transcriptome RNA sequencing (RNA-seq), microarray, qRT-PCR, stromal co-culture, confocal fluorescence microscopic, nanoproteomic, serial transplantation and cell cycle analyses were performed on FACS purified normal, chronic phase (CP) chronic myeloid leukemia (CML), blast crisis (BC) phase CML progenitors with or without PF-04449913 treatment. RESULTS: Notably, RNA-seq analyses revealed that Hh pathway and cell cycle regulatory gene overexpression correlated with leukemic progression. While lentivirally enforced GLI2 expression enhanced leukemic progenitor dormancy in stromal co-cultures, this was not observed with a mutant GLI2 lacking a transactivation domain, suggesting that GLI2 expression prevented cell cycle transit. Selective SMO inhibition with PF-04449913 in humanized stromal co-cultures and LSC xenografts reduced downstream GLI2 protein and cell cycle regulatory gene expression. Moreover, SMO inhibition enhanced cell cycle transit and sensitized BC LSC to tyrosine kinase inhibition in vivo at doses that spare normal HSC. CONCLUSION: In summary, while GLI2, forms part of a core HH pathway transcriptional regulatory network that promotes human myeloid leukemic progression and dormant LSC generation, selective inhibition with PF-04449913 reduces the dormant LSC burden thereby providing a strong rationale for clinical trials predicated on SMO inhibition in combination with TKIs or chemotherapeutic agents with the ultimate aim of obviating leukemic therapeutic resistance, persistence and progression.


Assuntos
Fatores de Transcrição Kruppel-Like/antagonistas & inibidores , Leucemia/patologia , Células-Tronco Neoplásicas/patologia , Proteínas Nucleares/antagonistas & inibidores , Animais , Sequência de Bases , Técnicas de Cocultura , Primers do DNA , Sangue Fetal/citologia , Proteínas Hedgehog/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcriptoma , Proteína Gli2 com Dedos de Zinco
19.
EMBO J ; 34(6): 759-77, 2015 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-25564442

RESUMO

Scl/Tal1 confers hemogenic competence and prevents ectopic cardiomyogenesis in embryonic endothelium by unknown mechanisms. We discovered that Scl binds to hematopoietic and cardiac enhancers that become epigenetically primed in multipotent cardiovascular mesoderm, to regulate the divergence of hematopoietic and cardiac lineages. Scl does not act as a pioneer factor but rather exploits a pre-established epigenetic landscape. As the blood lineage emerges, Scl binding and active epigenetic modifications are sustained in hematopoietic enhancers, whereas cardiac enhancers are decommissioned by removal of active epigenetic marks. Our data suggest that, rather than recruiting corepressors to enhancers, Scl prevents ectopic cardiogenesis by occupying enhancers that cardiac factors, such as Gata4 and Hand1, use for gene activation. Although hematopoietic Gata factors bind with Scl to both activated and repressed genes, they are dispensable for cardiac repression, but necessary for activating genes that enable hematopoietic stem/progenitor cell development. These results suggest that a unique subset of enhancers in lineage-specific genes that are accessible for regulators of opposing fates during the time of the fate decision provide a platform where the divergence of mutually exclusive fates is orchestrated.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/fisiologia , Elementos Facilitadores Genéticos/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Células-Tronco Hematopoéticas/citologia , Mesoderma/embriologia , Mioblastos Cardíacos/citologia , Proteínas Proto-Oncogênicas/metabolismo , Células Cultivadas , Imunoprecipitação da Cromatina , Perfilação da Expressão Gênica , Biblioteca Gênica , Células-Tronco Hematopoéticas/fisiologia , Humanos , Mesoderma/metabolismo , Análise em Microsséries , Modelos Biológicos , Dados de Sequência Molecular , Mioblastos Cardíacos/fisiologia , Análise de Sequência de RNA , Proteína 1 de Leucemia Linfocítica Aguda de Células T
20.
Cell Stem Cell ; 16(1): 80-7, 2015 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-25465114

RESUMO

Advances in pluripotent stem cell and reprogramming technologies have given us the hope of generating hematopoietic stem cells (HSCs) in culture. To succeed, greater understanding of the self-renewing HSC during human development is required. We discovered that the glycophosphatidylinositol-anchored surface protein GPI-80 defines a subpopulation of human fetal liver hematopoietic stem/progenitor cells (HSPCs) with self-renewal ability. CD34(+)CD38(lo/-)CD90(+)GPI-80(+) HSPCs were the sole population that maintained proliferative potential and an undifferentiated state in stroma coculture and engrafted in immunodeficient mice. GPI-80 expression also enabled tracking of HSPCs once they emerged from endothelium and migrated between human fetal hematopoietic niches. GPI-80 colocalized on the surface of HSPCs with Integrin alpha-M (ITGAM), which in leukocytes cooperates with GPI-80 to support migration. Knockdown of GPI-80 or ITGAM was sufficient to compromise HSPC expansion in culture and engraftment in vivo. These findings indicate that human fetal HSCs employ mechanisms used in leukocyte adhesion and migration to mediate HSC self-renewal.


Assuntos
Amidoidrolases/metabolismo , Moléculas de Adesão Celular/metabolismo , Desenvolvimento Embrionário , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Antígeno CD11b/metabolismo , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Feto/citologia , Citometria de Fluxo , Proteínas Ligadas por GPI/metabolismo , Transplante de Células-Tronco Hematopoéticas , Humanos , Imunofenotipagem , Fígado/citologia , Fígado/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...