Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Leukemia ; 37(6): 1287-1297, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37100881

RESUMEN

Heterozygous mutation targeting proline 95 in Serine/Arginine-rich Splicing Factor 2 (SRSF2) is associated with V617F mutation in Janus Activated Kinase 2 (JAK2) in some myeloproliferative neoplasms (MPNs), most commonly primary myelofibrosis. To explore the interaction of Srsf2P95H with Jak2V617F, we generated Cre-inducible knock-in mice expressing these mutants under control of the stem cell leukemia (Scl) gene promoter. In transplantation experiments, Srsf2P95H unexpectedly delayed myelofibrosis induced by Jak2V617F and decreased TGFß1 serum level. Srsf2P95H reduced the competitiveness of transplanted Jak2V617F hematopoietic stem cells while preventing their exhaustion. RNA sequencing of sorted megakaryocytes identified an increased number of splicing events when the two mutations were combined. Focusing on JAK/STAT pathway, Jak2 exon 14 skipping was promoted by Srsf2P95H, an event detected in patients with JAK2V617F and SRSF2P95 co-mutation. The skipping event generates a truncated inactive JAK2 protein. Accordingly, Srsf2P95H delays myelofibrosis induced by the thrombopoietin receptor agonist Romiplostim in Jak2 wild-type animals. These results unveil JAK2 exon 14 skipping promotion as a strategy to reduce JAK/STAT signaling in pathological conditions.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Trastornos Mieloproliferativos , Mielofibrosis Primaria , Animales , Ratones , Janus Quinasa 2/genética , Quinasas Janus/genética , Mutación , Trastornos Mieloproliferativos/genética , Mielofibrosis Primaria/genética , Proteínas de Unión al ARN/genética , Transducción de Señal , Factores de Transcripción STAT/genética
2.
J Clin Invest ; 132(14)2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35587378

RESUMEN

Acute megakaryoblastic leukemia of Down syndrome (DS-AMKL) is a model of clonal evolution from a preleukemic transient myeloproliferative disorder requiring both a trisomy 21 (T21) and a GATA1s mutation to a leukemia driven by additional driver mutations. We modeled the megakaryocyte differentiation defect through stepwise gene editing of GATA1s, SMC3+/-, and MPLW515K, providing 20 different T21 or disomy 21 (D21) induced pluripotent stem cell (iPSC) clones. GATA1s profoundly reshaped iPSC-derived hematopoietic architecture with gradual myeloid-to-megakaryocyte shift and megakaryocyte differentiation alteration upon addition of SMC3 and MPL mutations. Transcriptional, chromatin accessibility, and GATA1-binding data showed alteration of essential megakaryocyte differentiation genes, including NFE2 downregulation that was associated with loss of GATA1s binding and functionally involved in megakaryocyte differentiation blockage. T21 enhanced the proliferative phenotype, reproducing the cellular and molecular abnormalities of DS-AMKL. Our study provides an array of human cell-based models revealing individual contributions of different mutations to DS-AMKL differentiation blockage, a major determinant of leukemic progression.


Asunto(s)
Síndrome de Down , Leucemia Megacarioblástica Aguda , Proteínas de Ciclo Celular/genética , Niño , Proteoglicanos Tipo Condroitín Sulfato/genética , Proteínas Cromosómicas no Histona/genética , Síndrome de Down/genética , Factor de Transcripción GATA1/genética , Hematopoyesis , Humanos , Leucemia Megacarioblástica Aguda/complicaciones , Leucemia Megacarioblástica Aguda/genética , Leucemia Megacarioblástica Aguda/metabolismo , Megacariocitos/metabolismo , Mutación , Trisomía
3.
Blood ; 138(22): 2231-2243, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34407546

RESUMEN

Classical BCR-ABL-negative myeloproliferative neoplasms (MPNs) are clonal disorders of hematopoietic stem cells (HSCs) caused mainly by recurrent mutations in genes encoding JAK2 (JAK2), calreticulin (CALR), or the thrombopoietin receptor (MPL). Interferon α (IFNα) has demonstrated some efficacy in inducing molecular remission in MPNs. To determine factors that influence molecular response rate, we evaluated the long-term molecular efficacy of IFNα in patients with MPN by monitoring the fate of cells carrying driver mutations in a prospective observational and longitudinal study of 48 patients over more than 5 years. We measured the clonal architecture of early and late hematopoietic progenitors (84 845 measurements) and the global variant allele frequency in mature cells (409 measurements) several times per year. Using mathematical modeling and hierarchical Bayesian inference, we further inferred the dynamics of IFNα-targeted mutated HSCs. Our data support the hypothesis that IFNα targets JAK2V617F HSCs by inducing their exit from quiescence and differentiation into progenitors. Our observations indicate that treatment efficacy is higher in homozygous than heterozygous JAK2V617F HSCs and increases with high IFNα dose in heterozygous JAK2V617F HSCs. We also found that the molecular responses of CALRm HSCs to IFNα were heterogeneous, varying between type 1 and type 2 CALRm, and a high dose of IFNα correlates with worse outcomes. Our work indicates that the long-term molecular efficacy of IFNα implies an HSC exhaustion mechanism and depends on both the driver mutation type and IFNα dose.


Asunto(s)
Células Madre Hematopoyéticas/efectos de los fármacos , Factores Inmunológicos/uso terapéutico , Interferón-alfa/uso terapéutico , Mutación/efectos de los fármacos , Trastornos Mieloproliferativos/tratamiento farmacológico , Calreticulina/genética , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Factores Inmunológicos/farmacología , Interferón-alfa/farmacología , Janus Quinasa 2/genética , Estudios Longitudinales , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Estudios Prospectivos , Receptores de Trombopoyetina/genética , Células Tumorales Cultivadas
5.
Cell ; 182(6): 1401-1418.e18, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32810439

RESUMEN

Blood myeloid cells are known to be dysregulated in coronavirus disease 2019 (COVID-19), caused by SARS-CoV-2. It is unknown whether the innate myeloid response differs with disease severity and whether markers of innate immunity discriminate high-risk patients. Thus, we performed high-dimensional flow cytometry and single-cell RNA sequencing of COVID-19 patient peripheral blood cells and detected disappearance of non-classical CD14LowCD16High monocytes, accumulation of HLA-DRLow classical monocytes (Human Leukocyte Antigen - DR isotype), and release of massive amounts of calprotectin (S100A8/S100A9) in severe cases. Immature CD10LowCD101-CXCR4+/- neutrophils with an immunosuppressive profile accumulated in the blood and lungs, suggesting emergency myelopoiesis. Finally, we show that calprotectin plasma level and a routine flow cytometry assay detecting decreased frequencies of non-classical monocytes could discriminate patients who develop a severe form of COVID-19, suggesting a predictive value that deserves prospective evaluation.


Asunto(s)
Infecciones por Coronavirus , Coronavirus , Pandemias , Neumonía Viral , Betacoronavirus , COVID-19 , Citometría de Flujo , Humanos , Complejo de Antígeno L1 de Leucocito , Monocitos , Células Mieloides , Estudios Prospectivos , SARS-CoV-2
6.
J Cachexia Sarcopenia Muscle ; 11(4): 1047-1069, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32157826

RESUMEN

BACKGROUND: Aldehyde dehydrogenases (ALDHs) are key players in cell survival, protection, and differentiation via the metabolism and detoxification of aldehydes. ALDH activity is also a marker of stem cells. The skeletal muscle contains populations of ALDH-positive cells amenable to use in cell therapy, whose distribution, persistence in aging, and modifications in myopathic context have not been investigated yet. METHODS: The Aldefluor® (ALDEF) reagent was used to assess the ALDH activity of muscle cell populations, whose phenotypic characterizations were deepened by flow cytometry. The nature of ALDH isoenzymes expressed by the muscle cell populations was identified in complementary ways by flow cytometry, immunohistology, and real-time PCR ex vivo and in vitro. These populations were compared in healthy, aging, or Duchenne muscular dystrophy (DMD) patients, healthy non-human primates, and Golden Retriever dogs (healthy vs. muscular dystrophic model, Golden retriever muscular dystrophy [GRMD]). RESULTS: ALDEF+ cells persisted through muscle aging in humans and were equally represented in several anatomical localizations in healthy non-human primates. ALDEF+ cells were increased in dystrophic individuals in humans (nine patients with DMD vs. five controls: 14.9 ± 1.63% vs. 3.6 ± 0.39%, P = 0.0002) and dogs (three GRMD dogs vs. three controls: 10.9 ± 2.54% vs. 3.7 ± 0.45%, P = 0.049). In DMD patients, such increase was due to the adipogenic ALDEF+ /CD34+ populations (11.74 ± 1.5 vs. 2.8 ± 0.4, P = 0.0003), while in GRMD dogs, it was due to the myogenic ALDEF+ /CD34- cells (3.6 ± 0.6% vs. 1.03 ± 0.23%, P = 0.0165). Phenotypic characterization associated the ALDEF+ /CD34- cells with CD9, CD36, CD49a, CD49c, CD49f, CD106, CD146, and CD184, some being associated with myogenic capacities. Cytological and histological analyses distinguished several ALDH isoenzymes (ALDH1A1, 1A2, 1A3, 1B1, 1L1, 2, 3A1, 3A2, 3B1, 3B2, 4A1, 7A1, 8A1, and 9A1) expressed by different cell populations in the skeletal muscle tissue belonging to multinucleated fibres, or myogenic, endothelial, interstitial, and neural lineages, designing them as potential new markers of cell type or of metabolic activity. Important modifications were noted in isoenzyme expression between healthy and DMD muscle tissues. The level of gene expression of some isoenzymes (ALDH1A1, 1A3, 1B1, 2, 3A2, 7A1, 8A1, and 9A1) suggested their specific involvement in muscle stability or regeneration in situ or in vitro. CONCLUSIONS: This study unveils the importance of the ALDH family of isoenzymes in the skeletal muscle physiology and homeostasis, suggesting their roles in tissue remodelling in the context of muscular dystrophies.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Envejecimiento Saludable/fisiología , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/fisiopatología , Homeostasis , Humanos
7.
Transl Lung Cancer Res ; 6(4): 444-453, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28904888

RESUMEN

In non-small cell lung cancer (NSCLC), diagnosis of predictive biomarkers for targeted therapies is currently done in small tumor biopsies. However, tumor biopsies can be invasive, in some cases associated with risk, and tissue adequacy, both in terms of quantity and quality is often insufficient. The development of efficient and non-invasive methods to identify genetic alterations is a key challenge which circulating tumor cells (CTCs) have the potential to be exploited for. CTCs are extremely rare and phenotypically diverse, two characteristics that impose technical challenges and impact the success of robust molecular analysis. Here we introduce the clinical needs in this disease that mainly consist of the diagnosis of epidermal growth factor receptor (EGFR) activating alterations and anaplastic lymphoma kinase (ALK) rearrangement. We present the proof-of-concept studies that explore the detection of these genetic alterations in CTCs from NSCLC patients. Finally, we discuss steps that are still required before CTCs are routinely used for diagnosis of EGFR-mutations and ALK-rearrangements in this disease.

8.
Adv Exp Med Biol ; 994: 169-179, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28560674

RESUMEN

Circulating tumor cells (CTCs) hold promise as biomarkers to aid in patient treatment stratification and disease monitoring. Because the number of cells is a critical parameter for exploiting CTCs for predictive biomarker's detection, we developed a FISH (fluorescent in situ hybridization) method for CTCs enriched on filters (filter-adapted FISH [FA-FISH]) that was optimized for high cell recovery. To increase the feasibility and reliability of the analyses, we combined fluorescent staining and FA-FISH and developed a semi-automated microscopy method for optimal FISH signal identification in filtration-enriched CTCs . Here we present these methods and their use for the detection and characterization of ALK-, ROS1-, RET-rearrangement in CTCs from non-small-cell lung cancer and ERG-rearrangements in CTCs from prostate cancer patients.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Reordenamiento Génico , Neoplasias Pulmonares , Células Neoplásicas Circulantes/metabolismo , Neoplasias de la Próstata , Proteínas Tirosina Quinasas , Proteínas Proto-Oncogénicas c-ret , Proteínas Proto-Oncogénicas , Proteínas Tirosina Quinasas Receptoras , Quinasa de Linfoma Anaplásico , Carcinoma de Pulmón de Células no Pequeñas/sangre , Carcinoma de Pulmón de Células no Pequeñas/genética , Femenino , Humanos , Hibridación Fluorescente in Situ/instrumentación , Hibridación Fluorescente in Situ/métodos , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/genética , Masculino , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/genética , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-ret/genética , Proteínas Proto-Oncogénicas c-ret/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Regulador Transcripcional ERG/genética , Regulador Transcripcional ERG/metabolismo
9.
Stem Cell Res ; 13(3 Pt A): 431-41, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25460604

RESUMEN

Understanding the role of Notch and its ligands within the different bone marrow niches could shed light on the mechanisms regulating haematopoietic progenitor cells (HPCs) maintenance and self-renewal. Here, we report that murine bone marrow HPCs activation by the vascular Notch Delta-4 ligand maintains a significant proportion of cells specifically in the G0 state. Furthermore, Delta-4/Notch pathway limits significantly the loss of the in vivo short-term reconstitutive potential upon transplantation of Delta-4 activated HPCs into lethally irradiated recipient mice. Both effects are directly correlated with the decrease of cell cycle genes transcription such as CYCLIN-D1, -D2, and -D3, and the upregulation of stemness related genes transcription such as BMI1, GATA2, HOXB4 and C-MYC. In addition, the transcriptional screening also highlights new downstream post-transcriptional factors, named PUMILIO1 and -2, as part of the stem signature associated with the Delta-4/Notch signalling pathway.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Notch/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Unión al Calcio , Células Cultivadas , Ciclina D/genética , Ciclina D/metabolismo , Regulación hacia Abajo , Redes Reguladoras de Genes , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Interfase , Ligandos , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Regulación hacia Arriba
10.
Mol Ther ; 21(5): 1064-75, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23439500

RESUMEN

The functional and architectural benefits of embryonic stem cells (ESC) and myoblasts (Mb) transplantations into infarcted myocardium have been investigated extensively. Whereas ESC repopulated fibrotic areas and contributed to myocardial regeneration, Mb exerted their effects through paracrine secretions and scar remodeling. This therapeutic perspective, however, has been less explored in the setting of nonischemic dilated cardiomyopathies (DCMs). Our aim was to compare the integration and functional efficacy of ESC committed to cardiac fate by bone morphogenic protein 2 (BMP-2) pretreatment and Mb used as gold standard following their transplantation into the myocardium of a mouse model of laminopathy exhibiting a progressive and lethal DCM. After 4 and 8 weeks of transplantation, stabilization was observed in Mb-transplanted mice (P = 0.008) but not in groups of ESC-transplanted or medium-injected animals, where the left ventricular fractional shortening (LVFS) decreased by 32 ± 8% and 41 ± 8% respectively. Engrafted differentiated cells were consistently detected in myocardia of mice receiving Mb, whereas few or no cells were detected in the hearts of mice receiving ESC, except in two cases where teratomas were formed. These data suggest that committed ESC fail to integrate in DCM where scar tissue is absent to provide the appropriate niche, whereas the functional benefits of Mb transplantation might extend to nonischemic cardiomyopathy.


Asunto(s)
Cardiomiopatía Dilatada/terapia , Células Madre Embrionarias/trasplante , Mioblastos/trasplante , Animales , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/mortalidad , Cardiomiopatía Dilatada/fisiopatología , Diferenciación Celular , Línea Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Modelos Animales de Enfermedad , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Femenino , Supervivencia de Injerto , Masculino , Ratones , Desarrollo de Músculos , Mioblastos/citología , Mioblastos/metabolismo , Contracción Miocárdica , Miocardio/metabolismo , Miocardio/patología , Fenotipo
11.
Curr Opin Organ Transplant ; 16(6): 640-9, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22001714

RESUMEN

PURPOSE OF REVIEW: Cell therapy is considered a potential therapeutic avenue for the treatment of skeletal muscle diseases. Heterologous and autologous approaches have been attempted in the context, respectively, of generalized degenerative disease and of localized repairs. Cell transplantation trials, however, have been hampered by poor survival and limited migratory ability of the cells. This article reviews recent problems including the identification of new putative cellular candidates, the combination of complementary genetic or pharmacological therapeutic approaches, and the set up of clinical trials. RECENT FINDINGS: Deeper investigations identified anoikis, oxidative stress, fusion inability and some administration methodologies as causes of early massive cell death. It was proposed to adapt the injection strategies or to combine them with genetic modifications of the cells or pharmacological interventions on the environment to improve the success of implantation. New myogenic cell types have been identified, mainly in the family of perivascular cells, which can be administered systemically. New concepts have emerged regarding the correction of gene expression (use of lentiviral vectors, set-up of exon skipping, direct DNA repair, etc.). SUMMARY: Initial cell transplantation trials dedicated to the repair of striated muscles in muscular dystrophies produced mitigated results and underlined some limitations of cellular candidates under study. The research and identification of new stem cell candidates, the invention of new molecular strategies for correction of gene expression, the development of complementary approaches to improve transplantation success, have been justified by the unmet medical needs. These efforts led to new preclinical and clinical trials based on these concepts.


Asunto(s)
Músculo Esquelético/cirugía , Distrofias Musculares/cirugía , Mioblastos Esqueléticos/trasplante , Trasplante de Células Madre , Animales , Terapia Combinada , Terapia Genética , Humanos , Desarrollo de Músculos , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofias Musculares/genética , Distrofias Musculares/patología , Distrofias Musculares/fisiopatología , Fármacos Neuromusculares/uso terapéutico , Recuperación de la Función , Trasplante de Células Madre/efectos adversos , Resultado del Tratamiento
12.
Blood ; 116(25): 5670-8, 2010 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-20829371

RESUMEN

The effects of Notch signaling on human megakaryocytic and erythroid differentiation were investigated by exposing human CD34(+) progenitor cells to an immobilized chimeric form of the Notch ligand, Delta-like4 (Dll4Fc). Exposure of human cord blood CD34(+) cells to Dll4Fc induced a modest enhancement of erythroid cell production. Conversely, under megakaryocytic culture conditions, Dll4Fc strongly impaired platelet production by reducing the generation of mature CD41a(+)CD42b(+) megakaryocytes (MKs) and platelet-forming cells. The inhibitory activity of Dll4 on terminal MK differentiation was confirmed by culturing CD34(+) cells onto Dll-4-expressing stroma cells (engineered to express the membrane-anchored form of Dll4). The reduced production of mature CD41a(+)CD42(+) cells was rescued by inhibiting Notch signaling either with the N-N-(3,5-difluorophenacetyl-L-alanyl)-S-phenylglycine t-butyl ester γ-secretase inhibitor or the dominant-negative version of Mastermind. Dll4 impaired the generation of mature CD41a(+)CD42b(+) cells and proplatelet formation without affecting earlier steps of MK differentiation, such as production of megakaryocytic/erythroid progenitors and colony-forming units-MKs. This blockade was accompanied by a modulation of the transcriptional program of megakaryocytic differentiation. All these results indicate that Dll4/Notch signaling inhibits human terminal MK differentiation.


Asunto(s)
Diferenciación Celular , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Megacariocitos/citología , Receptores Notch/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Unión al Calcio , Células Cultivadas , Técnicas de Cocultivo , Células Precursoras Eritroides/metabolismo , Sangre Fetal/citología , Sangre Fetal/metabolismo , Citometría de Flujo , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Megacariocitos/metabolismo , Ratones , ARN Mensajero/genética , Receptores Notch/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/metabolismo
13.
Mol Ther ; 17(11): 1948-58, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19738599

RESUMEN

Aldehyde dehydrogenase 1A1 (ALDH) activity is one hallmark of human bone marrow (BM), umbilical cord blood (UCB), and peripheral blood (PB) primitive progenitors presenting high reconstitution capacities in vivo. In this study, we have identified ALDH(+) cells within human skeletal muscles, and have analyzed their phenotypical and functional characteristics. Immunohistofluorescence analysis of human muscle tissue sections revealed rare endomysial cells. Flow cytometry analysis using the fluorescent substrate of ALDH, Aldefluor, identified brightly stained (ALDH(br)) cells with low side scatter (SSC(lo)), in enzymatically dissociated muscle biopsies, thereafter abbreviated as SMALD(+) (for skeletal muscle ALDH(+)) cells. Phenotypical analysis discriminated two sub-populations according to CD34 expression: SMALD(+)/CD34(-) and SMALD(+)/CD34(+) cells. These sub-populations did not initially express endothelial (CD31), hematopoietic (CD45), and myogenic (CD56) markers. Upon sorting, however, whereas SMALD(+)/CD34(+) cells developed in vitro as a heterogeneous population of CD56(-) cells able to differentiate in adipoblasts, the SMALD(+)/CD34(-) fraction developed in vitro as a highly enriched population of CD56(+) myoblasts able to form myotubes. Moreover, only the SMALD(+)/CD34(-) population maintained a strong myogenic potential in vivo upon intramuscular transplantation. Our results suggest that ALDH activity is a novel marker for a population of new human skeletal muscle progenitors presenting a potential for cell biology and cell therapy.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Células Musculares/citología , Desarrollo de Músculos/fisiología , Músculo Esquelético/citología , Adipogénesis/fisiología , Diferenciación Celular/fisiología , Células Cultivadas , Citometría de Flujo , Humanos , Inmunohistoquímica , Técnicas In Vitro , Fenotipo
14.
Stem Cells ; 26(3): 621-9, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18055448

RESUMEN

Although Notch ligands are considered to activate signaling through direct cell-cell contact, the existence of soluble forms has been demonstrated. However, their roles remain controversial: soluble forms have been reported to mimic the biological activity of membrane-bound form, whereas other studies rather suggested an antagonistic activity toward their full-length counterparts. We previously observed that membrane-bound Delta4-expressing S17 stroma (mbD4/S17) reduced human CD34+CD38(low) cell proliferation and favored self-renewal. Here, we assessed the effects of a soluble form of Delta4 (solD4) by exposing CD34+CD38(low) cells to S17 feeders engineered to express solD4 (solD4/S17). In contrast to mbD4/S17, (a) solD4/S17 increased 10-fold cell production after 2 weeks, through enhanced cell proliferation, and (b) it did not preserve colony-forming cell and long-term culture-initiating cell potential of output CD34+ cells. mbD4 and solD4 appeared to also differ in their signaling. Indeed, mbD4, but not solD4, strongly activated both CSL (the nuclear mediator of Notch signaling) in Hela cells overexpressing Notch1 and transcription of some classic Notch target genes in CD34+CD38(low) cells. Furthermore, both biological effects and CSL activation elicited by mbD4 were strictly dependent upon the gamma-secretase complex, whereas solD4 enhanced cell expansion in a partially gamma-secretase-independent manner. Altogether, these results suggest that part of solD4 activity did not rely upon canonical Notch pathway.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Antígenos CD34/metabolismo , Diferenciación Celular , Membrana Celular/metabolismo , Sangre Fetal/citología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Receptores Notch/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Unión al Calcio , Núcleo Celular/metabolismo , Proliferación Celular , Técnicas de Cocultivo , Regulación de la Expresión Génica , Células HeLa , Humanos , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Solubilidad
15.
Stem Cells ; 23(4): 550-60, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15790776

RESUMEN

We investigated whether Notch signaling pathways have a role in human developmental hematopoiesis. In situ histochemistry analysis revealed that Notch1, 2, and 4 and Notch ligand (Delta1-4, and Jagged1) proteins were not expressed in the yolk sac blood islands, the para-aortic splanchnopleure, the hematopoietic aortic clusters, and at the early stages of embryonic liver hematopoiesis. Notch1-2, and Delta4 were eventually detected in the embryonic liver, from 34 until 38 days postconception. Fluorescence-activated cell sorter analysis showed that first-trimester embryonic liver CD34(+)CD38(low) cells expressed both Notch1 and Notch2. When these cells were cultured on S17 stroma stably expressing Delta4, a 2.6-fold increase in BFU-E number was observed at day 7, as compared with cultures with control stroma, and this effect was maintained for 2 weeks. Importantly, exposure of these cells to Delta4 under these conditions maintained the original frequency and quality of long-term culture-initiating cells (LTC-ICs), while control cultures quickly resulted in the extinction of this LTC-IC potential. Furthermore, short-term exposure of embryonic liver adherent cells to erythropoietin resulted in a dose-dependent increase in Delta4 expression, almost doubling the expression observed with untreated stroma. This suggests that Delta4 has a role in the regulation of hematopoiesis after a hypoxic stress in the fetus.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Antígenos CD34/metabolismo , Proteínas Sanguíneas/metabolismo , Células Precursoras Eritroides/citología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Hígado/citología , Receptores Notch/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Proteínas de Unión al Calcio , Adhesión Celular , Línea Celular , Técnicas de Cocultivo , Células Precursoras Eritroides/metabolismo , Eritropoyetina/metabolismo , Citometría de Flujo , Hematopoyesis , Humanos , Hígado/embriología , Hígado/metabolismo , Células del Estroma/citología , Células del Estroma/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...