Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Haematologica ; 105(4): 987-9998, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31289201

RESUMO

Targeting chemoresistant malignant cells is one of the current major challenges in oncology. Therefore, it is mandatory to refine the characteristics of these cells to monitor their survival and develop adapted therapies. This is of particular interest in acute myeloid leukemia (AML), for which the 5-year survival rate only reaches 30%, regardless of the prognosis. The role of the microenvironment is increasingly reported to be a key regulator for blast survival. In this context, we demonstrate that contact with mesenchymal stromal cells promotes a better survival of blasts in culture in the presence of anthracycline through the activation of ABC transporters. Stroma-dependent ABC transporter activation leads to the induction of a Side Population (SP) phenotype in a subpopulation of primary leukemia blasts through alpha (α)4 engagement. The stroma-promoting effect is reversible and is observed with stromal cells isolated from either healthy donors or leukemia patients. Blasts expressing an SP phenotype are mostly quiescent and are chemoresistant in vitro and in vivo in patient-derived xenograft mouse models. At the transcriptomic level, blasts from the SP are specifically enriched in the drug metabolism program. This detoxification signature engaged in contact with mesenchymal stromal cells represents promising ways to target stroma-induced chemoresistance of AML cells.


Assuntos
Leucemia Mieloide Aguda , Células-Tronco Mesenquimais , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Camundongos , Células Estromais , Microambiente Tumoral
2.
Stem Cells ; 35(3): 694-704, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27734598

RESUMO

The molecular mechanisms by which heterogeneity, a major characteristic of stem cells, is achieved are yet unclear. We here study the expression of the membrane stem cell antigen-1 (Sca-1) in mouse bone marrow mesenchymal stem cell (MSC) clones. We show that subpopulations with varying Sca-1 expression profiles regenerate the Sca-1 profile of the mother population within a few days. However, after extensive replication in vitro, the expression profiles shift to lower values and the regeneration time increases. Study of the promoter of Ly6a unravels that the expression level of Sca-1 is related to the promoter occupancy by the activating histone mark H3K4me3. We demonstrate that these findings can be consistently explained by a computational model that considers positive feedback between promoter H3K4me3 modification and gene transcription. This feedback implicates bistable epigenetic states which the cells occupy with an age-dependent frequency due to persistent histone (de-)modification. Our results provide evidence that MSC heterogeneity, and presumably that of other stem cells, is associated with bistable epigenetic states and suggest that MSCs are subject to permanent state fluctuations. Stem Cells 2017;35:694-704.


Assuntos
Envelhecimento/genética , Epigênese Genética , Células-Tronco Mesenquimais/metabolismo , Animais , Antígenos Ly/metabolismo , Células da Medula Óssea/citologia , Diferenciação Celular/genética , Proliferação de Células , Células Clonais , Perfilação da Expressão Gênica , Proteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos Endogâmicos C57BL , Modelos Biológicos , Modelos Genéticos , Regiões Promotoras Genéticas
3.
Blood ; 123(2): 191-202, 2014 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-24277075

RESUMO

In addition to its well-known effect on migration and homing of hematopoietic stem/progenitor cells (HSPCs), CXCL12 chemokine also exhibits a cell cycle and survival-promoting factor for human CD34(+) HSPCs. CXCR4 was suggested to be responsible for CXCL12-induced biological effects until the recent discovery of its second receptor, CXCR7. Until now, the participation of CXCR7 in CXCL12-induced HSPC cycling and survival is unknown. We show here that CXCL12 was capable of binding CXCR7 despite its scarce expression at CD34(+) cell surface. Blocking CXCR7 inhibited CXCL12-induced Akt activation as well as the percentage of CD34(+) cells in cycle, colony formation, and survival, demonstrating its participation in CXCL12-induced functional effects in HSPCs. At steady state, CXCR7 and ß-arrestin2 co-localized near the plasma membrane of CD34(+) cells. After CXCL12 treatment, ß-arrestin2 translocated to the nucleus, and this required both CXCR7 and CXCR4. Silencing ß-arrestin expression decreased CXCL12-induced Akt activation in CD34(+) cells. Our results demonstrate for the first time the role of CXCR7, complementary to that played by CXCR4, in the control of HSPC cycling, survival, and colony formation induced by CXCL12. We also provide evidence for the involvement of ß-arrestins as signaling hubs downstream of both CXCL12 receptors in primary human HSPCs.


Assuntos
Arrestinas/metabolismo , Ciclo Celular , Quimiocina CXCL12/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores CXCR/metabolismo , Antígenos CD34/metabolismo , Sobrevivência Celular , Ensaio de Unidades Formadoras de Colônias , Ativação Enzimática , Humanos , Espaço Intracelular/metabolismo , Fosforilação , Ligação Proteica , Transporte Proteico , beta-Arrestinas
4.
Haematologica ; 100(6): 757-67, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25840601

RESUMO

Primary myelofibrosis is characterized by clonal myeloproliferation, dysmegakaryopoiesis, extramedullary hematopoiesis associated with myelofibrosis and altered stroma in the bone marrow and spleen. The expression of CD9, a tetraspanin known to participate in megakaryopoiesis, platelet formation, cell migration and interaction with stroma, is deregulated in patients with primary myelofibrosis and is correlated with stage of myelofibrosis. We investigated whether CD9 participates in the dysmegakaryopoiesis observed in patients and whether it is involved in the altered interplay between megakaryocytes and stromal cells. We found that CD9 expression was modulated during megakaryocyte differentiation in primary myelofibrosis and that cell surface CD9 engagement by antibody ligation improved the dysmegakaryopoiesis by restoring the balance of MAPK and PI3K signaling. When co-cultured on bone marrow mesenchymal stromal cells from patients, megakaryocytes from patients with primary myelofibrosis displayed modified behaviors in terms of adhesion, cell survival and proliferation as compared to megakaryocytes from healthy donors. These modifications were reversed after antibody ligation of cell surface CD9, suggesting the participation of CD9 in the abnormal interplay between primary myelofibrosis megakaryocytes and stroma. Furthermore, silencing of CD9 reduced CXCL12 and CXCR4 expression in primary myelofibrosis megakaryocytes as well as their CXCL12-dependent migration. Collectively, our results indicate that CD9 plays a role in the dysmegakaryopoiesis that occurs in primary myelofibrosis and affects interactions between megakaryocytes and bone marrow stromal cells. These results strengthen the "bad seed in bad soil" hypothesis that we have previously proposed, in which alterations of reciprocal interactions between hematopoietic and stromal cells participate in the pathogenesis of primary myelofibrosis.


Assuntos
Megacariócitos/metabolismo , Mielofibrose Primária/metabolismo , Células Estromais/metabolismo , Tetraspanina 29/fisiologia , Trombopoese/fisiologia , Técnicas de Cocultura , Humanos , Megacariócitos/patologia , Mielofibrose Primária/patologia , Células Estromais/patologia
5.
BMC Biol ; 11: 86, 2013 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-23870169

RESUMO

BACKGROUND: Human pluripotent stem cells (hPSCs) hold great promise for applications in regenerative medicine. However, the safety of cell therapy using differentiated hPSC derivatives must be improved through methods that will permit the transplantation of homogenous populations of a specific cell type. To date, purification of progenitors and mature cells generated from either embryonic or induced pluripotent stem cells remains challenging with use of conventional methods. RESULTS: We used lentivectors encoding green fluorescent protein (GFP) driven by the liver-specific apoliprotein A-II (APOA-II) promoter to purify human hepatic progenitors. We evaluated both integrating and integration-defective lentivectors in combination with an HIV integrase inhibitor. A human embryonic stem cell line was differentiated into hepatic progenitors using a chemically defined protocol. Subsequently, cells were transduced and sorted at day 16 of differentiation to obtain a cell population enriched in hepatic progenitor cells. After sorting, more than 99% of these APOA-II-GFP-positive cells expressed hepatoblast markers such as α-fetoprotein and cytokeratin 19. When further cultured for 16 days, these cells underwent differentiation into more mature cells and exhibited hepatocyte properties such as albumin secretion. Moreover, they were devoid of vector DNA integration. CONCLUSIONS: We have developed an effective strategy to purify human hepatic cells from cultures of differentiating hPSCs, producing a novel tool that could be used not only for cell therapy but also for in vitro applications such as drug screening. The present strategy should also be suitable for the purification of a broad range of cell types derived from either pluripotent or adult stem cells.


Assuntos
Diferenciação Celular , Separação Celular/métodos , Células-Tronco Embrionárias/citologia , Vetores Genéticos/genética , Hepatócitos/citologia , Lentivirus/genética , Integração Viral/fisiologia , Apolipoproteína A-II/genética , Biomarcadores/metabolismo , Linhagem Celular , Citocromo P-450 CYP3A/metabolismo , DNA Viral/metabolismo , Citometria de Fluxo , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Hepatócitos/metabolismo , Humanos , Fígado/citologia , Especificidade de Órgãos , Regiões Promotoras Genéticas/genética , Transdução Genética
6.
Am J Hematol ; 88(5): 343-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23417980

RESUMO

Lentiviral modification combined with ex vivo erythroid differentiation was used to stably inhibit RhAG expression, a critical component of the Rh(rhesus) membrane complex defective in the Rh(null) syndrome. The cultured red cells generated recapitulate the major alterations of native Rh(null) cells regarding antigen expression, membrane deformability, and gas transport function, providing the proof of principle for their use as model of Rh(null) syndrome and to investigate Rh complex biogenesis in human primary erythroid cells. Using this model, we were able to reveal for the first time that RhAG extinction alone is sufficient to explain ICAM-4 and CD47 loss observed on native Rh(null) RBCs. Together with the effects of RhAG forced expression in Rh(null) progenitors, this strongly strengthens the hypothesis that RhAG is critical to Rh complex formation. The strategy is also promising for diagnosis purpose in order to overcome the supply from rare blood donors and is applicable to other erythroid defects and rare phenotypes, providing models to dissect membrane biogenesis of multicomplex proteins in erythroid cells, with potential clinical applications in transfusion medicine.


Assuntos
Proteínas Sanguíneas/metabolismo , Antígeno CD47/metabolismo , Moléculas de Adesão Celular/metabolismo , Células Eritroides/metabolismo , Doenças Genéticas Inatas/metabolismo , Glicoproteínas de Membrana/metabolismo , Sistema do Grupo Sanguíneo Rh-Hr/metabolismo , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Anemia Hemolítica Congênita/metabolismo , Anemia Hemolítica Congênita/patologia , Anemia Hipoplástica Congênita/metabolismo , Anemia Hipoplástica Congênita/patologia , Proteínas Sanguíneas/antagonistas & inibidores , Proteínas Sanguíneas/genética , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Células Eritroides/patologia , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/metabolismo , Feminino , Sangue Fetal , Células-Tronco Fetais/citologia , Células-Tronco Fetais/metabolismo , Doenças Genéticas Inatas/sangue , Doenças Genéticas Inatas/patologia , Humanos , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/genética , Porfiria Eritropoética/metabolismo , Porfiria Eritropoética/patologia , Gravidez , Interferência de RNA , RNA Interferente Pequeno , Reticulócitos/metabolismo , Reticulócitos/patologia , Sistema do Grupo Sanguíneo Rh-Hr/sangue
7.
Exp Hematol ; 124: 22-35.e3, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37331423

RESUMO

Generating hematopoietic stem cells (HSCs) from pluripotent stem cells (PSCs) has been a long-lasting quest in the field of hematopoiesis. Previous studies suggested that enforced expression of BCR-ABL, the unique oncogenic driver of chronic myelogeneous leukemia (CML), in embryonic stem cells (ESCs)-derived hematopoietic cells is sufficient to confer long-term in vivo repopulating potential. To precisely uncover the molecular events regulated by the tyrosine kinase activity of BCR-ABL1 (p210) during the course of hematopoietic differentiation, we engineered a Tet-ON inducible system to modulate its expression in murine ESCs (mESCs). We showed in unique site-directed knock-in ESC model that BCR-ABL expression tightly regulated by doxycycline (dox) controls the formation and the maintenance of immature hematopoietic progenitors. Interestingly, these progenitors can be expanded in vitro for several passages in the presence of dox. Our analysis of cell surface markers and transcriptome compared with wild-type fetal and adult HSCs unraveled a similar molecular signature. Long-term culture initiating cell (LTC-IC) assay confirmed their self-renewal capacities albeit with a differentiation bias toward erythroid and myeloid cells. Collectively, our novel Tet-ON system represents a unique in vitro model to shed lights on ESC-derived hematopoiesis, CML initiation, and maintenance.


Assuntos
Leucemia Mielogênica Crônica BCR-ABL Positiva , Camundongos , Animais , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Proteínas de Fusão bcr-abl/genética , Proteínas de Fusão bcr-abl/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Diferenciação Celular , Células-Tronco Embrionárias/metabolismo , Doxiciclina/farmacologia , Doxiciclina/metabolismo
8.
Blood ; 116(22): 4444-55, 2010 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-20693433

RESUMO

Edification of the human hematopoietic system during development is characterized by the production of waves of hematopoietic cells separated in time, formed in distinct embryonic sites (ie, yolk sac, truncal arteries including the aorta, and placenta). The embryonic liver is a major hematopoietic organ wherein hematopoietic stem cells (HSCs) expand, and the future, adult-type, hematopoietic cell hierarchy becomes established. We report herein the identification of a new, transient, and rare cell population in the human embryonic liver, which coexpresses VE-cadherin, an endothelial marker, CD45, a pan-hematopoietic marker, and CD34, a common endothelial and hematopoietic marker. This population displays an outstanding self-renewal, proliferation, and differentiation potential, as detected by in vitro and in vivo hematopoietic assays compared with its VE-cadherin negative counterpart. Based on VE-cadherin expression, our data demonstrate the existence of 2 phenotypically and functionally separable populations of multipotent HSCs in the human embryo, the VE-cadherin(+) one being more primitive than the VE-cadherin(-) one, and shed a new light on the hierarchical organization of the embryonic liver HSC compartment.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Hematopoéticas/citologia , Sistema Hematopoético/embriologia , Fígado/citologia , Fígado/embriologia , Animais , Antígenos CD/genética , Antígenos CD34/metabolismo , Caderinas/genética , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Células-Tronco Embrionárias/metabolismo , Feminino , Expressão Gênica , Hematopoese , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Sistema Hematopoético/citologia , Humanos , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Camundongos SCID , Gravidez
9.
Blood ; 115(18): 3686-94, 2010 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-20223923

RESUMO

The mitogen-activated protein kinases (MAPKs) extracellular signal-regulated kinase 1 (ERK1) and ERK2 are among the main signal transduction molecules, but little is known about their isoform-specific functions in vivo. We have examined the role of ERK1 in adult hematopoiesis with ERK1(-/-) mice. Loss of ERK1 resulted in an enhanced splenic erythropoiesis, characterized by an accumulation of erythroid progenitors in the spleen, without any effect on the other lineages or on bone marrow erythropoiesis. This result suggests that the ablation of ERK1 induces a splenic stress erythropoiesis phenotype. However, the mice display no anemia. Deletion of ERK1 did not affect erythropoietin (EPO) serum levels or EPO/EPO receptor signaling and was not compensated by ERK2. Splenic stress erythropoiesis response has been shown to require bone morphogenetic protein 4 (BMP4)-dependent signaling in vivo and to rely on the expansion of a resident specialized population of erythroid progenitors, termed stress erythroid burst-forming units (BFU-Es). A great expansion of stress BFU-Es and increased levels of BMP4 mRNA were found in ERK1(-/-) spleens. The ERK1(-/-) phenotype can be transferred by bone marrow cells. These findings show that ERK1 controls a BMP4-dependent step, regulating the steady state of splenic erythropoiesis.


Assuntos
Anemia/patologia , Células Precursoras Eritroides/fisiologia , Eritropoese/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Baço/metabolismo , Anemia/induzido quimicamente , Animais , Apoptose , Western Blotting , Transplante de Medula Óssea , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Ensaio de Unidades Formadoras de Colônias , Células Precursoras Eritroides/citologia , Eritropoetina/metabolismo , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Oxidantes/toxicidade , Fenil-Hidrazinas/toxicidade , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores da Eritropoetina/genética , Receptores da Eritropoetina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Baço/citologia
10.
Stem Cell Res Ther ; 13(1): 254, 2022 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-35715824

RESUMO

BACKGROUND: hPSC-derived endothelial and hematopoietic cells (ECs and HCs) are an interesting source of cells for tissue engineering. Despite their close spatial and temporal embryonic development, current hPSC differentiation protocols are specialized in only one of these lineages. In this study, we generated a hematoendothelial population that could be further differentiated in vitro to both lineages. METHODS: Two hESCs and one hiPSC lines were differentiated into a hematoendothelial population, hPSC-ECs and blast colonies (hPSC-BCs) via CD144+-embryoid bodies (hPSC-EBs). hPSC-ECs were characterized by endothelial colony-forming assay, LDL uptake assay, endothelial activation by TNF-α, nitric oxide detection and Matrigel-based tube formation. Hematopoietic colony-forming cell assay was performed from hPSC-BCs. Interestingly, we identified a hPSC-BC population characterized by the expression of both CD144 and CD45. hPSC-ECs and hPSC-BCs were analyzed by flow cytometry and RT-qPCR; in vivo experiments have been realized by ischemic tissue injury model on a mouse dorsal skinfold chamber and hematopoietic reconstitution in irradiated immunosuppressed mouse from hPSC-ECs and hPSC-EB-CD144+, respectively. Transcriptomic analyses were performed to confirm the endothelial and hematopoietic identity of hESC-derived cell populations by comparing them against undifferentiated hESC, among each other's (e.g. hPSC-ECs vs. hPSC-EB-CD144+) and against human embryonic liver (EL) endothelial, hematoendothelial and hematopoietic cell subpopulations. RESULTS: A hematoendothelial population was obtained after 84 h of hPSC-EBs formation under serum-free conditions and isolated based on CD144 expression. Intrafemorally injection of hPSC-EB-CD144+ contributed to the generation of CD45+ human cells in immunodeficient mice suggesting the existence of hemogenic ECs within hPSC-EB-CD144+. Endothelial differentiation of hPSC-EB-CD144+ yields a population of > 95% functional ECs in vitro. hPSC-ECs derived through this protocol participated at the formation of new vessels in vivo in a mouse ischemia model. In vitro, hematopoietic differentiation of hPSC-EB-CD144+ generated an intermediate population of > 90% CD43+ hPSC-BCs capable to generate myeloid and erythroid colonies. Finally, the transcriptomic analyses confirmed the hematoendothelial, endothelial and hematopoietic identity of hPSC-EB-CD144+, hPSC-ECs and hPSC-BCs, respectively, and the similarities between hPSC-BC-CD144+CD45+, a subpopulation of hPSC-BCs, and human EL hematopoietic stem cells/hematopoietic progenitors. CONCLUSION: The present work reports a hPSC differentiation protocol into functional hematopoietic and endothelial cells through a hematoendothelial population. Both lineages were proven to display characteristics of physiological human cells, and therefore, they represent an interesting rapid source of cells for future cell therapy and tissue engineering.


Assuntos
Células-Tronco Embrionárias Humanas , Células-Tronco Pluripotentes Induzidas , Animais , Diferenciação Celular/fisiologia , Corpos Embrioides , Células Endoteliais/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Camundongos
11.
Bone Res ; 10(1): 22, 2022 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-35217633

RESUMO

The cells of origin of neurogenic heterotopic ossifications (NHOs), which develop frequently in the periarticular muscles following spinal cord injuries (SCIs) and traumatic brain injuries, remain unclear because skeletal muscle harbors two progenitor cell populations: satellite cells (SCs), which are myogenic, and fibroadipogenic progenitors (FAPs), which are mesenchymal. Lineage-tracing experiments using the Cre recombinase/LoxP system were performed in two mouse strains with the fluorescent protein ZsGreen specifically expressed in either SCs or FAPs in skeletal muscles under the control of the Pax7 or Prrx1 gene promoter, respectively. These experiments demonstrate that following muscle injury, SCI causes the upregulation of PDGFRα expression on FAPs but not SCs and the failure of SCs to regenerate myofibers in the injured muscle, with reduced apoptosis and continued proliferation of muscle resident FAPs enabling their osteogenic differentiation into NHOs. No cells expressing ZsGreen under the Prrx1 promoter were detected in the blood after injury, suggesting that the cells of origin of NHOs are locally derived from the injured muscle. We validated these findings using human NHO biopsies. PDGFRα+ mesenchymal cells isolated from the muscle surrounding NHO biopsies could develop ectopic human bones when transplanted into immunocompromised mice, whereas CD56+ myogenic cells had a much lower potential. Therefore, NHO is a pathology of the injured muscle in which SCI reprograms FAPs to undergo uncontrolled proliferation and differentiation into osteoblasts.

12.
Stem Cells ; 27(8): 1750-9, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19544443

RESUMO

Embryoid bodies (EBs) generated during differentiation of human embryonic stem cells (hESCs) contain vascular-like structures, suggesting that commitment of mesoderm progenitors into endothelial cells occurs spontaneously. We showed that bone morphogenetic protein 4 (BMP4), an inducer of mesoderm, accelerates the peak expression of CD133/kinase insert domain-containing receptor (KDR) and CD144/KDR. Because the CD133(+)KDR(+) population could represent endothelial progenitors, we sorted them at day 7 and cultured them in endothelial medium. These cells were, however, unable to differentiate into endothelial cells. Under standard conditions, the CD144(+)KDR(+) population represents up to 10% of the total cells at day 12. In culture, these cells, if sorted, give rise to a homogeneous population with a morphology typical of endothelial cells and express endothelial markers. These endothelial cells derived from the day 12 sorted population were functional, as assessed by different in vitro assays. When EBs were stimulated by BMP4, the CD144(+)KDR(+) peak was shifted to day 7. Most of these cells, however, were CD31(-), becoming CD31(+) in culture. They then expressed von Willebrand factor and were functional. This suggests that, initially, the BMP4-boosted day 7, CD144(+)KDR(+)CD31(-) population represents immature endothelial cells that differentiate into mature endothelial cells in culture. The expression of OCT3/4, a marker of immaturity for hESCs decreases during EB differentiation, decreasing faster following BMP4 induction. We also show that BMP4 inhibits the global expression of GATA2 and RUNX1, two transcription factors involved in hemangioblast formation, at day 7 and day 12.


Assuntos
Proteína Morfogenética Óssea 4/farmacologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Antígenos CD/biossíntese , Caderinas/biossíntese , Proteínas de Transporte/farmacologia , Diferenciação Celular/efeitos dos fármacos , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/fisiologia , Células Cultivadas , Citocinas/farmacologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Citometria de Fluxo , Humanos , Cinética , Fatores de Transcrição/biossíntese
13.
Stem Cells ; 27(10): 2552-62, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19650038

RESUMO

Identification of prevalent specific markers is crucial to stem/progenitor cell purification. Determinants such as the surface antigens CD34 and CD38 are traditionally used to analyze and purify hematopoietic stem/progenitor cells (HSCs/HPCs). However, the variable expression of these membrane antigens poses some limitations to their use in HSC/HPC purification. Techniques based on drug/stain efflux through the ATP-binding cassette (ABC)G2 pump (side population [SP] phenotype) or on detection of aldehyde dehydrogenase (ALDH) activity have been independently developed and distinguish the SP and ALDH(Bright) (ALDH(Br)) cell subsets for their phenotype and proliferative capability. In this study, we developed a multiparametric flow cytometric method associating both SP and ALDH activities on human lineage negative (Lin(-)) bone marrow cells and sorted different cell fractions according to their SP/ALDH activity level. We find that Lin(-)CD34(+)CD38(Low/-) cells are found throughout the spectrum of ALDH expression and are enriched especially in ALDH(Br) cells when associated with SP functionality (SP/ALDH(Br) fraction). Furthermore, the SP marker identified G(0) cells in all ALDH fractions, allowing us to sort quiescent cells regardless of ALDH activity. Moreover, we show that, within the Lin(-)CD34(+)CD38(-)ALDH(Br) population, the SP marker identifies cells with higher primitive characteristics, in terms of stemness-related gene expression and in vitro and in vivo proliferative potential, than the Lin(-)CD34(+) CD38(-)ALDH(Br) main population cells. In conclusion, our study shows that the coexpression of SP and ALDH markers refines the Lin(-)CD34(+)CD38(-) hematopoietic compartment and identifies an SP/ALDH(Br) cell subset enriched in quiescent primitive HSCs/HPCs.


Assuntos
Aldeído Desidrogenase/metabolismo , Antígenos de Superfície/metabolismo , Separação Celular/métodos , Citometria de Fluxo/métodos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , ADP-Ribosil Ciclase 1/análise , ADP-Ribosil Ciclase 1/metabolismo , Aldeído Desidrogenase/análise , Animais , Antígenos CD34/análise , Antígenos CD34/metabolismo , Antígenos de Superfície/análise , Biomarcadores/análise , Biomarcadores/metabolismo , Ciclo Celular/fisiologia , Desdiferenciação Celular/genética , Linhagem da Célula , Células Cultivadas , Regulação da Expressão Gênica/fisiologia , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Camundongos , Nucleotidiltransferases/análise , Nucleotidiltransferases/metabolismo
14.
Antibodies (Basel) ; 9(3)2020 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-32630723

RESUMO

CD63, a member of the tetraspanin superfamily, is used as a marker of late endosomes and lysosome-related organelles, as well as a marker of exosomes. Here, we selected rare isotype variants of TS63 by sorting hybridoma cells on the basis of their high expression of surface immunoglobulins of the IgG2a and IgG2b subclass. Pure populations of cells secreting IgG2a and IgG2b variants of TS63 (referred to as TS63a and TS63b) were obtained using two rounds of cell sorting and one limited dilution cloning step. We validate that these new TS63 variants are suitable for co-labeling with mAb of the IgG1 subclass directed to other molecules, using anti mouse subclass antibodies, and for the labeling of exosomes through direct binding to protein A-coated gold particles. These mAbs will be useful to study the intracellular localization of various proteins and facilitate electron microscopy analysis of CD63 localization.

15.
Life Sci Alliance ; 3(1)2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31792032

RESUMO

ADAM10 is a transmembrane metalloprotease that is essential for development and tissue homeostasis. It cleaves the ectodomain of many proteins, including amyloid precursor protein, and plays an essential role in Notch signaling. ADAM10 associates with six members of the tetraspanin superfamily referred to as TspanC8 (Tspan5, Tspan10, Tspan14, Tspan15, Tspan17, and Tspan33), which regulate its exit from the endoplasmic reticulum and its substrate selectivity. We now show that ADAM10, Tspan5, and Tspan15 influence each other's expression level. Notably, ADAM10 undergoes faster endocytosis in the presence of Tspan5 than in the presence of Tspan15, and Tspan15 stabilizes ADAM10 at the cell surface yielding high expression levels. Reciprocally, ADAM10 stabilizes Tspan15 at the cell surface, indicating that it is the Tspan15/ADAM10 complex that is retained at the plasma membrane. Chimeric molecules indicate that the cytoplasmic domains of these tetraspanins contribute to their opposite action on ADAM10 trafficking and Notch signaling. In contrast, an unusual palmitoylation site at the end of Tspan15 C-terminus is dispensable. Together, these findings uncover a new level of ADAM10 regulation by TspanC8 tetraspanins.


Assuntos
Proteína ADAM10/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Endocitose/genética , Proteínas de Membrana/metabolismo , Transdução de Sinais/genética , Tetraspaninas/metabolismo , Proteína ADAM10/genética , Secretases da Proteína Precursora do Amiloide/genética , Membrana Celular/metabolismo , Estabilidade Enzimática/genética , Meia-Vida , Células HeLa , Humanos , Proteínas de Membrana/genética , Células PC-3 , Receptor Notch1/genética , Receptor Notch1/metabolismo , Especificidade por Substrato , Tetraspaninas/genética , Transfecção
16.
Stem Cells ; 26(12): 3150-61, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18757300

RESUMO

Cell cycle regulation plays a fundamental role in stem cell biology. A balance between quiescence and proliferation of hematopoietic stem cells in interaction with the microenvironment is critical for sustaining long-term hematopoiesis and for protection against stress. We analyzed the molecular mechanisms by which stromal cell-derived factor-1 (SDF-1) exhibited a cell cycle-promoting effect and interacted with transforming growth factor-beta (TGF-beta), which has negative effects on cell cycle orchestration of human hematopoietic CD34(+) progenitor cells. We demonstrated that a low concentration of SDF-1 modulated the expression of key cell cycle regulators such as cyclins, cyclin-dependent kinase inhibitors, and TGF-beta target genes, confirming its cell cycle-promoting effect. We showed that a cross-talk between SDF-1- and TGF-beta-related signaling pathways involving phosphatidylinositol 3-kinase (PI3K)/Akt phosphorylation participated in the control of CD34(+) cell cycling. We demonstrated a pivotal role of two downstream effectors of the PI3K/Akt pathway, FoxO3a and mammalian target of rapamycin, as connectors in the SDF-1-/TGF-beta-induced control of the cycling/quiescence switch and proposed a model integrating a dialogue between the two molecules in cell cycle progression. Our data shed new light on the signaling pathways involved in SDF-1 cell cycle-promoting activity and suggest that the balance between SDF-1- and TGF-beta-activated pathways is critical for the regulation of hematopoietic progenitor cell cycle status.


Assuntos
Quimiocina CXCL12/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Proteínas Quinases/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Antígenos CD34/biossíntese , Ciclo Celular , Proteína Forkhead Box O3 , Hematopoese , Células-Tronco Hematopoéticas/citologia , Humanos , Modelos Biológicos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Células-Tronco/citologia , Serina-Treonina Quinases TOR
17.
Stem Cells Dev ; 28(3): 165-185, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30426841

RESUMO

Despite advances to engineer transplantable hematopoietic stem and progenitor cells (HSPCs) for research and therapy, an in-depth characterization of the developing human hematopoietic system is still lacking. The human embryonic liver is at the crossroad of several hematopoietic sites and harbors a complex hematopoietic hierarchy, including the first actively dividing HSPCs that will further seed the definitive hematopoietic organs. However, few are known about the phenotypic and functional HSPC organization operating at these stages of development. In this study, using a combination of four endothelial and hematopoietic surface markers, that is, the endothelial-specific marker vascular endothelial-cadherin (Cdh5, CD144), the pan-leukocyte antigen CD45, the hemato-endothelial marker CD34, and the angiotensin-converting enzyme (ACE, CD143), we identified distinct HSPC subsets, and among them, a population co-expressing the four markers that uniquely harbored an outstanding proliferation potential both ex vivo and in vivo. Moreover, we traced back this population to the yolk sac (YS) and aorta-gonad-mesonephros (AGM) sites of hematopoietic emergence. Taken together, our data will help to identify human HSPC self-renewal and amplification mechanisms for future cell therapies.


Assuntos
Antígenos CD/genética , Caderinas/genética , Linhagem da Célula , Proliferação de Células , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Fígado/citologia , Peptidil Dipeptidase A/genética , Antígenos CD/metabolismo , Caderinas/metabolismo , Diferenciação Celular , Células Cultivadas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/fisiologia , Humanos , Fígado/embriologia , Peptidil Dipeptidase A/metabolismo
18.
Int J Cancer ; 122(9): 2130-41, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18183592

RESUMO

Multiple myeloma (MM) is a malignancy characterized by the accumulation of tumoral plasma cells in bone marrow. This disease remains incurable and the development of new therapeutic strategies is urgently required. We have studied the effects of 2 selective estrogen receptor disrupters (SERDs), RU 58668 (RU) and ICI 182,780 (ICI) or pure antiestrogens (AEs) on MM cell lines. Both compounds have antimyeloma activity through either cell cycle arrest or induction of apoptosis. To analyze the molecular mechanisms of SERD action, we choose 2 differently responding cell lines as models. In LP-1 cells, RU blocked cell cycle at the G1 phase. RU treatment induced a rapid decrease of c-Myc, an upregulation of p27(Kip1), and the subsequent decreased activity of cyclin-dependent kinase, CDK6 and associated cyclin D3, impairing the inactivation of the retinoblastoma protein (pRb). In RPMI 8226 cells, RU induced apoptosis by recruiting endoplasmic reticulum- as well as mitochondria-associated caspases. Moreover, RU interfered with the NF-kappaB survival pathway, often deregulated in MM malignancy. Antimyeloma activities were observed in dexamethasone (Dex)- and RU-resistant cells when RU was combined with bortezomib; Dex and bortezomib being frequently used in MM therapy. RU induced the death of CD138+ cells purified from MM patients but not CD19+ normal cells obtained from tonsils. Therefore, RU mediates the inhibition of survival, the activation of apoptosis and finally potentiates anticancer drug. Those combinatory effects provide a basis for the potential use of pure AEs in MM treatment.


Assuntos
Antineoplásicos Hormonais/farmacologia , Apoptose/efeitos dos fármacos , Ácidos Borônicos/farmacologia , Caspases/metabolismo , Quinase 6 Dependente de Ciclina/metabolismo , Ciclinas/metabolismo , Retículo Endoplasmático/metabolismo , Estradiol/análogos & derivados , Moduladores de Receptor Estrogênico/farmacologia , Fase G1/efeitos dos fármacos , Mieloma Múltiplo/tratamento farmacológico , Pirazinas/farmacologia , Western Blotting , Bortezomib , Proliferação de Células/efeitos dos fármacos , Colorimetria , Ciclina D3 , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Estradiol/farmacologia , Citometria de Fluxo , Humanos , Imunoprecipitação , Mitocôndrias/metabolismo , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Transdução de Sinais , Células Tumorais Cultivadas
19.
Stem Cell Reports ; 11(5): 1075-1091, 2018 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-30449320

RESUMO

Fanconi anemia (FA) causes bone marrow failure early during childhood, and recent studies indicate that a hematopoietic defect could begin in utero. We performed a unique kinetics study of hematopoiesis in Fancg-/- mouse embryos, between the early embryonic day 11.5 (E11.5) to E12.5 developmental window (when the highest level of hematopoietic stem cells [HSC] amplification takes place) and E14.5. This study reveals a deep HSC defect with exhaustion of proliferative and self-renewal capacities very early during development, together with severe FA clinical and biological manifestations, which are mitigated at E14.5 due to compensatory mechanisms that help to ensure survival of Fancg-/- embryos. It also reports that a deep HSC defect is also observed during human FA development, and that human FA fetal liver (FL) HSCs present a transcriptome profile similar to that of mouse E12.5 Fancg-/- FL HSCs. Altogether, our results highlight that early mouse FL could represent a good alternative model for studying Fanconi pathology.


Assuntos
Desenvolvimento Embrionário , Anemia de Fanconi/patologia , Células-Tronco Hematopoéticas/patologia , Animais , Apoptose , Ciclo Celular , Dano ao DNA , Embrião de Mamíferos/patologia , Eritrócitos/metabolismo , Proteína do Grupo de Complementação G da Anemia de Fanconi/deficiência , Proteína do Grupo de Complementação G da Anemia de Fanconi/metabolismo , Feminino , Ontologia Genética , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Humanos , Fígado/embriologia , Fígado/metabolismo , Camundongos Endogâmicos C57BL , Fenótipo , Placenta/metabolismo , Gravidez , Transcriptoma/genética
20.
JCI Insight ; 2(21)2017 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-29093266

RESUMO

Neurogenic heterotopic ossification (NHO) is the formation of ectopic bone generally in muscles surrounding joints following spinal cord or brain injury. We investigated the mechanisms of NHO formation in 64 patients and a mouse model of spinal cord injury-induced NHO. We show that marrow from human NHOs contains hematopoietic stem cell (HSC) niches, in which mesenchymal stromal cells (MSCs) and endothelial cells provide an environment supporting HSC maintenance, proliferation, and differentiation. The transcriptomic signature of MSCs from NHOs shows a neuronal imprinting associated with a molecular network required for HSC support. We demonstrate that oncostatin M (OSM) produced by activated macrophages promotes osteoblastic differentiation and mineralization of human muscle-derived stromal cells surrounding NHOs. The key role of OSM was confirmed using an experimental model of NHO in mice defective for the OSM receptor (OSMR). Our results provide strong evidence that macrophages contribute to NHO formation through the osteogenic action of OSM on muscle cells within an inflammatory context and suggest that OSM/OSMR could be a suitable therapeutic target. Altogether, the evidence of HSCs in ectopic bones growing at the expense of soft tissue in spinal cord/brain-injured patients indicates that inflammation and muscle contribute to HSC regulation by the brain-bone-blood triad.


Assuntos
Macrófagos/metabolismo , Oncostatina M/metabolismo , Ossificação Heterotópica/imunologia , Ossificação Heterotópica/metabolismo , Animais , Antígenos CD34 , Lesões Encefálicas , Diferenciação Celular , Proliferação de Células , Células Endoteliais , Feminino , Hematopoese , Células-Tronco Hematopoéticas , Xenoenxertos , Humanos , Células-Tronco Mesenquimais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subunidade beta de Receptor de Oncostatina M , Ossificação Heterotópica/patologia , Osteogênese , Medula Espinal , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa