Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Exp Hematol ; 124: 22-35.e3, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37331423

RESUMEN

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.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva , Ratones , Animales , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Células Madre Hematopoyéticas/metabolismo , Diferenciación Celular , Células Madre Embrionarias/metabolismo , Doxiciclina/farmacología , Doxiciclina/metabolismo
2.
Stem Cell Res Ther ; 13(1): 254, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35715824

RESUMEN

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.


Asunto(s)
Células Madre Embrionarias Humanas , Células Madre Pluripotentes Inducidas , Animales , Diferenciación Celular/fisiología , Cuerpos Embrioides , Células Endoteliales/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Humanos , Ratones
3.
Bone Res ; 10(1): 22, 2022 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-35217633

RESUMEN

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.

4.
Antibodies (Basel) ; 9(3)2020 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-32630723

RESUMEN

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.

5.
Haematologica ; 105(4): 987-9998, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31289201

RESUMEN

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.


Asunto(s)
Leucemia Mieloide Aguda , Células Madre Mesenquimatosas , Transportadoras de Casetes de Unión a ATP/genética , Animales , Resistencia a Antineoplásicos/genética , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Ratones , Células del Estroma , Microambiente Tumoral
6.
Life Sci Alliance ; 3(1)2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31792032

RESUMEN

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.


Asunto(s)
Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Endocitosis/genética , Proteínas de la Membrana/metabolismo , Transducción de Señal/genética , Tetraspaninas/metabolismo , Proteína ADAM10/genética , Secretasas de la Proteína Precursora del Amiloide/genética , Membrana Celular/metabolismo , Estabilidad de Enzimas/genética , Semivida , Células HeLa , Humanos , Proteínas de la Membrana/genética , Células PC-3 , Receptor Notch1/genética , Receptor Notch1/metabolismo , Especificidad por Sustrato , Tetraspaninas/genética , Transfección
7.
Stem Cells Dev ; 28(3): 165-185, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30426841

RESUMEN

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.


Asunto(s)
Antígenos CD/genética , Cadherinas/genética , Linaje de la Célula , Proliferación Celular , Células Madre Hematopoyéticas/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Hígado/citología , Peptidil-Dipeptidasa A/genética , Antígenos CD/metabolismo , Cadherinas/metabolismo , Diferenciación Celular , Células Cultivadas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/fisiología , Humanos , Hígado/embriología , Peptidil-Dipeptidasa A/metabolismo
8.
Stem Cell Reports ; 11(5): 1075-1091, 2018 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-30449320

RESUMEN

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.


Asunto(s)
Desarrollo Embrionario , Anemia de Fanconi/patología , Células Madre Hematopoyéticas/patología , Animales , Apoptosis , Ciclo Celular , Daño del ADN , Embrión de Mamíferos/patología , Eritrocitos/metabolismo , Proteína del Grupo de Complementación G de la Anemia de Fanconi/deficiencia , Proteína del Grupo de Complementación G de la Anemia de Fanconi/metabolismo , Femenino , Ontología de Genes , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Humanos , Hígado/embriología , Hígado/metabolismo , Ratones Endogámicos C57BL , Fenotipo , Placenta/metabolismo , Embarazo , Transcriptoma/genética
9.
JCI Insight ; 2(21)2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-29093266

RESUMEN

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.


Asunto(s)
Macrófagos/metabolismo , Oncostatina M/metabolismo , Osificación Heterotópica/inmunología , Osificación Heterotópica/metabolismo , Animales , Antígenos CD34 , Lesiones Encefálicas , Diferenciación Celular , Proliferación Celular , Células Endoteliales , Femenino , Hematopoyesis , Células Madre Hematopoyéticas , Xenoinjertos , Humanos , Células Madre Mesenquimatosas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subunidad beta del Receptor de Oncostatina M , Osificación Heterotópica/patología , Osteogénesis , Médula Espinal , Transcriptoma
10.
Stem Cells ; 35(3): 694-704, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27734598

RESUMEN

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.


Asunto(s)
Envejecimiento/genética , Epigénesis Genética , Células Madre Mesenquimatosas/metabolismo , Animales , Antígenos Ly/metabolismo , Células de la Médula Ósea/citología , Diferenciación Celular/genética , Proliferación Celular , Células Clonales , Perfilación de la Expresión Génica , Proteínas de la Membrana/metabolismo , Células Madre Mesenquimatosas/citología , Ratones Endogámicos C57BL , Modelos Biológicos , Modelos Genéticos , Regiones Promotoras Genéticas
11.
Exp Hematol ; 44(4): 297-302.e1, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26733047

RESUMEN

Thrombopoietin (TPO) and its receptor Mpl (CD110) play a crucial role in the regulation of hematopoietic stem cells (HSCs). Functional study of Mpl-expressing HSCs has, however, been hampered by the lack of efficient monoclonal antibodies, explaining the very few data available on Mpl(+) HSCs during human embryonic development and after birth. Investigating the main monoclonal antibodies used so far to sort CD110(+) cells from cord blood (CB) and adult bone marrow (BM), we found that only the recent monoclonal antibody 1.6.1 engineered by Immunex Corporation was specific. Using in vitro functional assays, we found that this antibody can be used to sort a CD34(+)CD38(-)CD110(+) population enriched in hematopoietic progenitor stem cells, both in CB and in adult BM. In vivo injection into NSG mice further indicated that the CB CD34(+)CD38(-)CD110(+) population is highly enriched in HSCs compared with both CD34(+)CD38(-)CD110(-) and CD34(+)CD38(-) populations. Together our results validate MAb1.6.1 as an important tool, which has so far been lacking, in the HSC field.


Asunto(s)
Anticuerpos Monoclonales , Células de la Médula Ósea , Sangre Fetal/citología , Células Madre Hematopoyéticas/metabolismo , Receptores de Trombopoyetina/metabolismo , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Especificidad de Anticuerpos/inmunología , Antígenos de Superficie/metabolismo , Técnicas de Cultivo de Célula , Línea Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Humanos , Fenotipo , Receptores de Trombopoyetina/antagonistas & inhibidores , Receptores de Trombopoyetina/inmunología
12.
Oncotarget ; 7(13): 15507-24, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-26551931

RESUMEN

As rapidly developing patient-derived xenografts (PDX) could represent potential sources of cancer stem cells (CSC), we selected and characterized non-cultured PDX cell suspensions from four different renal carcinomas (RCC). Only the cell suspensions from the serial xenografts (PDX-1 and PDX-2) of an undifferentiated RCC (RCC-41) adapted to the selective CSC medium. The cell suspension derived from the original tumor specimen (RCC-41-P-0) did not adapt to the selective medium and strongly expressed CSC-like markers (CD133 and CD105) together with the non-CSC tumor marker E-cadherin. In comparison, PDX-1 and PDX-2 cells exhibited evolution in their phenotype since PDX-1 cells were CD133high/CD105-/Ecadlow and PDX-2 cells were CD133low/CD105-/Ecad-. Both PDX subsets expressed additional stem cell markers (CD146/CD29/OCT4/NANOG/Nestin) but still contained non-CSC tumor cells. Therefore, using different cell sorting strategies, we characterized 3 different putative CSC subsets (RCC-41-PDX-1/CD132+, RCC-41-PDX-2/CD133-/EpCAMlow and RCC-41-PDX-2/CD133+/EpCAMbright). In addition, transcriptomic analysis showed that RCC-41-PDX-2/CD133- over-expressed the pluripotency gene ERBB4, while RCC-41-PDX-2/CD133+ over-expressed several tumor suppressor genes. These three CSC subsets displayed ALDH activity, formed serial spheroids and developed serial tumors in SCID mice, although RCC-41-PDX-1/CD132+ and RCC-41-PDX-2/CD133+ displayed less efficiently the above CSC properties. RCC-41-PDX-1/CD132+ tumors showed vessels of human origin with CSC displaying peri-vascular distribution. By contrast, RCC-41-PDX-2 originated tumors exhibiting only vessels of mouse origin without CSC peri-vascular distribution.Altogether, our results indicate that PDX murine microenvironment promotes a continuous redesign of CSC phenotype, unmasking CSC subsets potentially present in a single RCC or generating ex novo different CSC-like subsets.


Asunto(s)
Carcinoma de Células Renales/patología , Xenoinjertos , Neoplasias Renales/patología , Células Madre Neoplásicas/patología , Animales , Técnicas de Cultivo de Célula/métodos , Separación Celular/métodos , Citometría de Flujo/métodos , Humanos , Ratones , Ratones SCID , Células Tumorales Cultivadas
13.
Haematologica ; 100(6): 757-67, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25840601

RESUMEN

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.


Asunto(s)
Megacariocitos/metabolismo , Mielofibrosis Primaria/metabolismo , Células del Estroma/metabolismo , Tetraspanina 29/fisiología , Trombopoyesis/fisiología , Técnicas de Cocultivo , Humanos , Megacariocitos/patología , Mielofibrosis Primaria/patología , Células del Estroma/patología
14.
Stem Cells Dev ; 23(24): 2983-95, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24955741

RESUMEN

During human embryonic stem cell (ESC) hematopoietic differentiation, the description of the initial steps of lymphopoiesis remains elusive. Using a two-step culture procedure, we identified two original populations of ESC-derived hematopoietic progenitor cells (HPCs) with CD34(+)CD45RA(+)CD7(-) and CD34(+)CD45RA(+)CD7(+) phenotypes. Bulk cultures and limiting dilution assays, culture with MS5 cells in the presence of Notch ligand Delta-like-1 (DL-1), and ex vivo colonization tests using fetal thymic organ cultures showed that although CD34(+)CD45RA(+)CD7(-) HPCs could generate cells of the three lymphoid lineages, their potential was skewed toward the B cell lineages. In contrast, CD34(+)CD45RA(+)CD7(+) HPCs predominantly exhibited a T/natural killer (NK) cell differentiation potential. Furthermore these cells could differentiate equivalently into cells of the granulo-macrophagic lineage and dendritic cells and lacked erythroid potential. Expression profiling of 18 markers by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) revealed that CD34(+)CD45RA(+)CD7(-) and CD34(+)CD45RA(+)CD7(+) HPCs express genes of the lymphoid specification and that CD34(+)CD45RA(+)CD7(-) cells express B-cell-associated genes, while CD34(+)CD45RA(+)CD7(+) HPCs display a T-cell molecular profile. Altogether, these findings indicate that CD34(+)CD45RA(+)CD7(-) and CD34(+)CD45RA(+)CD7(+) HPCs correspond to candidate multipotent early lymphoid progenitors polarized toward either the B or T/NK lineage, respectively. This work should improve our understanding of the early steps of lymphopoiesis from pluripotent stem cells and pave the way for the production of lymphocytes for cell-based immunotherapy and lymphoid development studies.


Asunto(s)
Células Madre Embrionarias/citología , Hematopoyesis , Células Progenitoras Linfoides/citología , Células Madre Pluripotentes/citología , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Proteínas de Unión al Calcio , Línea Celular , Linaje de la Célula , Células Cultivadas , Células Madre Embrionarias/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células Progenitoras Linfoides/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos NOD , Células Madre Pluripotentes/metabolismo
15.
Blood ; 123(2): 191-202, 2014 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-24277075

RESUMEN

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.


Asunto(s)
Arrestinas/metabolismo , Ciclo Celular , Quimiocina CXCL12/metabolismo , Células Madre Hematopoyéticas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores CXCR/metabolismo , Antígenos CD34/metabolismo , Supervivencia Celular , Ensayo de Unidades Formadoras de Colonias , Activación Enzimática , Humanos , Espacio Intracelular/metabolismo , Fosforilación , Unión Proteica , Transporte de Proteínas , beta-Arrestinas
16.
BMC Biol ; 11: 86, 2013 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-23870169

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Separación Celular/métodos , Células Madre Embrionarias/citología , Vectores Genéticos/genética , Hepatocitos/citología , Lentivirus/genética , Integración Viral/fisiología , Apolipoproteína A-II/genética , Biomarcadores/metabolismo , Línea Celular , Citocromo P-450 CYP3A/metabolismo , ADN Viral/metabolismo , Citometría de Flujo , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Hepatocitos/metabolismo , Humanos , Hígado/citología , Especificidad de Órganos , Regiones Promotoras Genéticas/genética , Transducción Genética
17.
Am J Hematol ; 88(5): 343-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23417980

RESUMEN

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.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Antígeno CD47/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células Eritroides/metabolismo , Enfermedades Genéticas Congénitas/metabolismo , Glicoproteínas de Membrana/metabolismo , Sistema del Grupo Sanguíneo Rh-Hr/metabolismo , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Anemia Hemolítica Congénita/metabolismo , Anemia Hemolítica Congénita/patología , Anemia Hipoplástica Congénita/metabolismo , Anemia Hipoplástica Congénita/patología , Proteínas Sanguíneas/antagonistas & inhibidores , Proteínas Sanguíneas/genética , Diferenciación Celular , Línea Celular , Células Cultivadas , Células Eritroides/patología , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Femenino , Sangre Fetal , Células Madre Fetales/citología , Células Madre Fetales/metabolismo , Enfermedades Genéticas Congénitas/sangre , Enfermedades Genéticas Congénitas/patología , Humanos , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Porfiria Eritropoyética/metabolismo , Porfiria Eritropoyética/patología , Embarazo , Interferencia de ARN , ARN Interferente Pequeño , Reticulocitos/metabolismo , Reticulocitos/patología , Sistema del Grupo Sanguíneo Rh-Hr/sangre
18.
PLoS One ; 7(1): e30788, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22303456

RESUMEN

The mitogen-activated protein kinases (MAPK) ERK1 and ERK2 are among the major signal transduction molecules but little is known about their specific functions in vivo. ERK activity is provided by two isoforms, ERK1 and ERK2, which are ubiquitously expressed and share activators and substrates. However, there are not in vivo studies which have reported a role for ERK1 or ERK2 in HSCs and the bone marrow microenvironment. The present study shows that the ERK1-deficient mice present a mild osteopetrosis phenotype. The lodging and the homing abilities of the ERK1(-/-) HSC are impaired, suggesting that the ERK1(-/-)-defective environment may affect the engrafment of HSCs. Serial transplantations demonstrate that ERK1 is involved in the maintenance of an appropriate medullar microenvironment, but that the intrinsic properties of HSCs are not altered by the ERK1(-/-) defective microenvironment. Deletion of ERK1 impaired in vitro and in vivo osteoclastogenesis while osteoblasts were unaffected. As osteoclasts derive from precursors of the monocyte/macrophage lineage, investigation of the monocytic compartment was performed. In vivo analysis of the myeloid lineage progenitors revealed that the frequency of CMPs increased by approximately 1.3-fold, while the frequency of GMPs significantly decreased by almost 2-fold, compared with the respective WT compartments. The overall mononuclear-phagocyte lineage development was compromised in these mice due to a reduced expression of the M-CSF receptor on myeloid progenitors. These results show that the cellular targets of ERK1 are M-CSFR-responsive cells, upstream to osteoclasts. While ERK1 is well known to be activated by M-CSF, the present results are the first to point out an ERK1-dependent M-CSFR regulation on hematopoietic progenitors. This study reinforces the hypothesis of an active cross-talk between HSCs, their progeny and bone cells in the maintenance of the homeostasis of these compartments.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/enzimología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Nicho de Células Madre , Animales , Densidad Ósea , Médula Ósea/patología , Huesos/enzimología , Huesos/patología , Compartimento Celular , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Proliferación Celular , Microambiente Celular , Eliminación de Gen , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 3 Activada por Mitógenos/deficiencia , Monocitos , Osteoblastos/enzimología , Osteoblastos/patología , Osteoclastos/enzimología , Osteoclastos/patología , Osteogénesis
19.
J Natl Cancer Inst ; 103(24): 1884-98, 2011 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-22043039

RESUMEN

BACKGROUND: Many renal cancer patients experience disease recurrence after immunotherapy or combined treatments due to persistence of cancer stem cells (CSCs). The identification of reliable inducers of CSC differentiation may facilitate the development of efficient strategies for eliminating CSCs. We investigated whether interleukin 15 (IL-15), a regulator of kidney homeostasis, induces the differentiation of CD105-positive (CD105(+)) CSCs from human renal cancers. METHODS: CD105(+) CSCs were cultured to preserve their stem cell properties and treated with recombinant human IL-15 (rhIL-15) to evaluate their ability to differentiate, to acquire sensitivity to chemotherapeutic drugs, and to form spheroids in vitro and tumors in vivo. Expression of stem cell and epithelial markers were studied by flow cytometry, immunocytochemistry, and immunoblotting. Identification of a CSC side population fraction and its sensitivity to chemotherapy drugs and expression of ATP-binding cassette (ABC) transporters and aldehyde dehydrogenase (ALDH) activities were determined by flow cytometry. Spheroid formation was determined in limiting dilution assay. Xenograft tumors were generated in severe combined immunodeficient mice (n = 12-18 mice per group). All statistical tests were two-sided. RESULTS: CD105(+) CSCs treated with rhIL-15 at 10 pg/mL differentiated into cells expressing epithelial markers. rhIL-15 induced epithelial differentiation of all CD105(+) CSCs subsets and blocked CSC self-renewal (sphere-forming ability) and their tumorigenic properties in severe combined immunodeficient mice. Vinblastine and paclitaxel induced statistically significant higher levels of apoptosis in rhIL-15-differentiated epithelial cells compared with CD105(+) CSCs (mean percentage of apoptotic cells, vinblastine: 33% vs 16.5%, difference = 16.5%, 95% confidence interval = 12.25% to 20.74%, P = .0025; paclitaxel: 35% vs 11.6%, difference = 23.4%, 95% confidence interval = 22.5% to 24.24%, P = .0015). The higher sensitivity of rhIL-15-differentiated epithelial cells to chemotherapeutic drugs was associated with loss of detoxifying mechanisms such as ALDH and ABC transporter activities. CONCLUSION: IL-15 directs the epithelial differentiation of renal CSCs and meets the criteria for a treatment strategy: CSC pool depletion and generation of differentiated nontumorigenic cells that are sensitive to chemotherapeutic agents.


Asunto(s)
Antígenos CD/metabolismo , Antineoplásicos/farmacología , Carcinoma de Células Renales/tratamiento farmacológico , Interleucina-15/farmacología , Neoplasias Renales/tratamiento farmacológico , Recurrencia Local de Neoplasia/prevención & control , Células Madre Neoplásicas/efectos de los fármacos , Receptores de Superficie Celular/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Aldehído Deshidrogenasa/metabolismo , Animales , Antineoplásicos/uso terapéutico , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Endoglina , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Humanos , Immunoblotting , Inmunohistoquímica , Interleucina-15/uso terapéutico , Ratones , Ratones SCID , Células Madre Neoplásicas/metabolismo , Factor de Transcripción STAT5/efectos de los fármacos , Factor de Transcripción STAT5/metabolismo , Prevención Secundaria , Trasplante Heterólogo
20.
PLoS One ; 6(5): e19988, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21625521

RESUMEN

Human bone marrow mesenchymal stem cells (BM-MSC) are multipotent progenitor cells that have transient immunomodulatory properties on Natural Killer (NK) cells, Dendritic Cells (DC), and T cells. This study compared the use of MSC isolated from bone marrow and fetal liver (FL-MSC) to determine which displayed the most efficient immunosuppressive effects on T cell activation. Although both types of MSC exhibit similar phenotype profile, FL-MSC displays a much more extended in vitro life-span and immunomodulatory properties. When co-cultured with CD3/CD28-stimulated T cells, both BM-MSC and FL-MSC affected T cell proliferation by inhibiting their entry into the cell cycle, by inducing the down-regulation of phospho-retinoblastoma (pRb), cyclins A and D1, as well as up-regulating p27(kip1) expression. The T cell inhibition by MSC was not due to the soluble HLA-G5 isoform, but to the surface expression of HLA-G1, as shown by the need of cell-cell contact and by the use of neutralizing anti-HLA-G antibodies. To note, in a HLA-G-mediated fashion, MSC facilitated the expansion of a CD4(low)/CD8(low) T subset that had decreased secretion of IFN-γ, and an induced secretion of the immunomodulatory cytokine IL-10. Because of their longer lasting in vitro immunosuppressive properties, mainly mediated by HLA-G, and their more efficient induction of IL-10 production and T cell apoptosis, fetal liver MSC could be considered a new tool for MSC therapy to prevent allograft rejection.


Asunto(s)
Proliferación Celular , Hígado/embriología , Células Madre Mesenquimatosas/citología , Linfocitos T/citología , Antígenos CD/inmunología , Western Blotting , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Antígenos HLA/inmunología , Humanos , Hígado/citología , Activación de Linfocitos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...