Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Blood ; 129(4): 460-472, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-27683414

RESUMEN

Epithelial-to-mesenchymal-transition (EMT) is critical for normal embryogenesis and effective postnatal wound healing, but is also associated with cancer metastasis. SNAIL, ZEB, and TWIST families of transcription factors are key modulators of the EMT process, but their precise roles in adult hematopoietic development and homeostasis remain unclear. Here we report that genetic inactivation of Zeb2 results in increased frequency of stem and progenitor subpopulations within the bone marrow (BM) and spleen and that these changes accompany differentiation defects in multiple hematopoietic cell lineages. We found no evidence that Zeb2 is critical for hematopoietic stem cell self-renewal capacity. However, knocking out Zeb2 in the BM promoted a phenotype with several features that resemble human myeloproliferative disorders, such as BM fibrosis, splenomegaly, and extramedullary hematopoiesis. Global gene expression and intracellular signal transduction analysis revealed perturbations in specific cytokine and cytokine receptor-related signaling pathways following Zeb2 loss, especially the JAK-STAT and extracellular signal-regulated kinase pathways. Moreover, we detected some previously unknown mutations within the human Zeb2 gene (ZFX1B locus) from patients with myeloid disease. Collectively, our results demonstrate that Zeb2 controls adult hematopoietic differentiation and lineage fidelity through widespread modulation of dominant signaling pathways that may contribute to blood disorders.


Asunto(s)
Citocinas/genética , Transición Epitelial-Mesenquimal/genética , Hematopoyesis Extramedular/genética , Proteínas de Homeodominio/genética , Mielofibrosis Primaria/genética , Proteínas Represoras/genética , Esplenomegalia/genética , Adulto , Animales , Secuencia de Bases , Médula Ósea/metabolismo , Médula Ósea/patología , Diferenciación Celular , Linaje de la Célula/genética , Citocinas/metabolismo , Regulación de la Expresión Génica , Humanos , Quinasas Janus/genética , Quinasas Janus/metabolismo , Ratones , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Mutación , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/patología , Proteínas Represoras/deficiencia , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Bazo/metabolismo , Bazo/patología , Esplenomegalia/metabolismo , Esplenomegalia/patología , Células Madre/metabolismo , Células Madre/patología , Transcripción Genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc
2.
Development ; 141(4): 773-83, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24496615

RESUMEN

The activation of epidermal growth factor receptor (EGFR) affects multiple aspects of neural precursor behaviour, including proliferation and migration. Telencephalic precursors acquire EGF responsiveness and upregulate EGFR expression at late stages of development. The events regulating this process and its significance are still unclear. We here show that in the developing and postnatal hippocampus (HP), growth/differentiation factor (GDF) 15 and EGFR are co-expressed in primitive precursors as well as in more differentiated cells. We also provide evidence that GDF15 promotes responsiveness to EGF and EGFR expression in hippocampal precursors through a mechanism that requires active CXC chemokine receptor (CXCR) 4. Besides EGFR expression, GDF15 ablation also leads to decreased proliferation and migration. In particular, lack of GDF15 impairs both processes in the cornu ammonis (CA) 1 and only proliferation in the dentate gyrus (DG). Importantly, migration and proliferation in the mutant HP were altered only perinatally, when EGFR expression was also affected. These data suggest that GDF15 regulates migration and proliferation by promoting EGFR signalling in the perinatal HP and represent a first description of a functional role for GDF15 in the developing telencephalon.


Asunto(s)
Animales Recién Nacidos , Movimiento Celular/fisiología , Receptores ErbB/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Factor 15 de Diferenciación de Crecimiento/metabolismo , Hipocampo/crecimiento & desarrollo , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Bromodesoxiuridina , Carbocianinas , Proliferación Celular , Citometría de Flujo , Fluorescencia , Regulación del Desarrollo de la Expresión Génica/genética , Hipocampo/metabolismo , Inmunohistoquímica , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores CXCR4/metabolismo , beta-Galactosidasa/metabolismo
3.
Stem Cell Reports ; 19(3): 351-365, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38366596

RESUMEN

The expression of growth/differentiation factor (GDF) 15 increases in the ganglionic eminence (GE) late in neural development, especially in neural stem cells (NSCs). However, GDF15 function in this region remains unknown. We report that GDF15 receptor is expressed apically in the GE and that GDF15 ablation promotes proliferation and cell division in the embryonic GE and in the adult ventricular-subventricular zone (V-SVZ). This causes a transient generation of additional neuronal progenitors, compensated by cell death, and a lasting increase in the number of ependymal cells and apical NSCs. Finally, both GDF15 receptor and the epidermal growth factor receptor (EGFR) were expressed in progenitors and mutation of GDF15 affected EGFR signaling. However, only exposure to exogenous GDF15, but not to EGF, normalized proliferation and the number of apical progenitors. Thus, GDF15 regulates proliferation of apical progenitors in the GE, thereby affecting the number of ependymal cells and NSCs.


Asunto(s)
Ventrículos Laterales , Células-Madre Neurales , Receptores ErbB/metabolismo , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Recuento de Células , Proliferación Celular , Diferenciación Celular/fisiología
4.
J Med Chem ; 65(2): 1505-1524, 2022 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-34818008

RESUMEN

Restoring lost heart muscle is an attractive goal for cardiovascular regenerative medicine. One appealing strategy is the therapeutic stimulation of cardiomyocyte proliferation, which inter alia remains challenging due to available assay technologies capturing the complex biology. Here, a high-throughput-formatted phenotypic assay platform was established using rodent whole heart-derived cells to preserve the cellular environment of cardiomyocytes. Several readouts allowed the quantification of cycling cardiomyocytes, including a transgenic H2B-mCherry system for unequivocal, automated detection of cardiomyocyte nuclei. A chemical genetics approach revealed pronounced species differences and furnished pan-kinase inhibitors 5 and 36 as potent and robust inducers of endoreplication and acytokinetic mitosis. Combined profiling of the commonly used p38 MAPK inhibitors SB203580 (1), SB239063 (2) and a novel set of skepinone-L (6) derivatives pointed to off-target effects beyond p38 that might be critical for effective cardiomyocyte cytokinesis. Kinome-focused screening eventually furnished TG003 (38) as a novel candidate for stimulating cardiomyocyte proliferation.


Asunto(s)
Ciclo Celular , Proliferación Celular , Corazón , Ensayos Analíticos de Alto Rendimiento , Sondas Moleculares , Miocitos Cardíacos , Inhibidores de Proteínas Quinasas , Animales , Ratones , Ratas , Animales Recién Nacidos , Corazón/efectos de los fármacos , Corazón/crecimiento & desarrollo , Ensayos Analíticos de Alto Rendimiento/métodos , Ratones Endogámicos C57BL , Mitosis , Sondas Moleculares/química , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología
5.
Mol Cell Neurosci ; 44(4): 318-29, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20417282

RESUMEN

Precursors within the subventricular zone (SVZ) exhibit regional variations in the expression of transcription factors important for the regulation of their proliferation and differentiation. In the anterior SVZ (aSVZ) the homeobox transcription factor distalless (Dlx)2 modulates both processes by promoting neural stem cell (NSC) activation as well as neurogenesis. Activated NSCs and transit-amplifying precursors (TAPs) in the aSVZ both express high levels of epidermal growth factor receptor (EGFR(high)) and form clones in response to exogenous EGF. EGF-responsive cells are also present in the hippocampal subependyma (hSVZ). However, it is not clear whether they represent NSCs or TAPs and whether their proliferation and differentiation are regulated as in the aSVZ. Here we have purified EGFR(high) cells from both the aSVZ and hSVZ at different ages. When isolated from perinatal tissue both populations were enriched in multipotent clonogenic precursors, which generated GABAergic neurons. Although they differed in absolute expression levels, activated NSCs and TAPs in both regions displayed similar signatures of transcription factor expression. However, activated NSCs were less frequent in the hSVZ than in the aSVZ. Furthermore, increasing age had a greater inhibitory effect on NSC proliferation in the hSVZ than in the aSVZ. This suggests that NSC activation is differentially regulated in the two regions. Consistent with this hypothesis, we found that in hippocampal precursors Dlx2 promoted neurogenesis but not NSC activation. Thus, most clonogenic EGFR(high) precursors in the hSVZ represent TAPs and NSC proliferation in the aSVZ and hSVZ is regulated by different mechanisms.


Asunto(s)
Receptores ErbB/metabolismo , Proteínas de Homeodominio/metabolismo , Células Madre Multipotentes/fisiología , Neurogénesis/fisiología , Neuronas/fisiología , Nicho de Células Madre/fisiología , Factores de Transcripción/metabolismo , Factores de Edad , Animales , Diferenciación Celular/fisiología , Proliferación Celular , Células Cultivadas , Receptores ErbB/genética , Femenino , Ratones , Ratones Endogámicos C57BL , Células Madre Multipotentes/citología , Neuronas/citología , Especificidad de Órganos , Embarazo , Nicho de Células Madre/citología
6.
J Med Chem ; 63(1): 425-432, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31841335

RESUMEN

N-(4-Aminobutyl)-N'-(2-methoxyethyl)guanidine (8a) is a potent inhibitor targeting the hDDAH-1 active site (Ki = 18 µM) and derived from a series of guanidine- and amidine-based inhibitors. Its nonamino acid nature leads to high selectivities toward other enzymes of the nitric oxide-modulating system. Crystallographic data of 8a-bound hDDAH-1 illuminated a unique binding mode. Together with its developed N-hydroxyguanidine prodrug 11, 8a will serve as a most widely applicable, pharmacological tool to target DDAH-1-associated diseases.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Guanidinas/química , Amidohidrolasas/química , Amidohidrolasas/metabolismo , Dominio Catalítico/efectos de los fármacos , Cristalografía por Rayos X , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/metabolismo , Guanidinas/síntesis química , Guanidinas/metabolismo , Humanos , Unión Proteica
7.
Nat Commun ; 11(1): 5425, 2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-33110077

RESUMEN

Transcription factors are key protein effectors in the regulation of gene transcription, and in many cases their activity is regulated via a complex network of protein-protein interactions (PPI). The chemical modulation of transcription factor activity is a long-standing goal in drug discovery but hampered by the difficulties associated with the targeting of PPIs, in particular when extended and flat protein interfaces are involved. Peptidomimetics have been applied to inhibit PPIs, however with variable success, as for certain interfaces the mimicry of a single secondary structure element is insufficient to obtain high binding affinities. Here, we describe the design and characterization of a stabilized protein tertiary structure that acts as an inhibitor of the interaction between the transcription factor TEAD and its co-repressor VGL4, both playing a central role in the Hippo signalling pathway. Modification of the inhibitor with a cell-penetrating entity yielded a cell-permeable proteomimetic that activates cell proliferation via regulation of the Hippo pathway, highlighting the potential of protein tertiary structure mimetics as an emerging class of PPI modulators.


Asunto(s)
Peptidomiméticos , Factores de Transcripción/química , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Vía de Señalización Hippo , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Transducción de Señal , Factores de Transcripción de Dominio TEA , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Front Pharmacol ; 10: 1357, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31849643

RESUMEN

Extracts from the leaves and flowers of Crataegus spp. (i.e., hawthorn species) have been traditionally used with documented preclinical and clinical activities in cardiovascular medicine. Based on reported positive effects on heart muscle after ischemic injury and the overall cardioprotective profile, the present study addressed potential contributions of Crataegus extracts to cardiopoietic differentiation from stem cells. The quantified Crataegus extract WS®1442 stimulated cardiomyogenesis from murine and human embryonic stem cells (ESCs). Mechanistically, this effect was found to be induced by promoting differentiation of cardiovascular progenitor cell populations but not by proliferation. Bioassay-guided fractionation, phytochemical and analytical profiling suggested high-molecular weight ingredients as the active principle with at least part of the activity due to oligomeric procyanidines (OPCs) with a degree of polymerization between 3 and 6 (DP3-6). Transcriptome profiling in mESCs suggested two main, plausible mechanisms: These were early, stress-associated cellular events along with the modulation of distinct developmental pathways, including the upregulation of brain-derived neurotrophic factor (BDNF) and retinoic acid as well as the inhibition of transforming growth factor ß/bone morphogenetic protein (TGFß/BMP) and fibroblast growth factor (FGF) signaling. In addition, WS®1442 stimulated angiogenesis ex vivo in Sca-1+ progenitor cells from adult mice hearts. These in vitro data provide evidence for a differentiation promoting activity of WS®1442 on distinct cardiovascular stem/progenitor cells that could be valuable for therapeutic heart regeneration after myocardial infarction. However, the in vivo relevance of this new pharmacological activity of Crataegus spp. remains to be investigated and active ingredients from bioactive fractions will have to be further characterized.

9.
Anal Cell Pathol (Amst) ; 2018: 1754085, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30406001

RESUMEN

Cultivation of A549 non-small-cell lung carcinoma (NSCLC) cells in the presence of staurosporine (SSP) leads to a reduction or a lack of proliferation in a concentration-dependent manner. This inhibition of proliferation is accompanied by the generation of polyploid giant cancer cells (PGCCs) that are characterized by cell flattening, increased cell size, polyploidy, and polynucleation as determined by crystal violet staining, BrdU and DiI labelling, and flow cytometry as well as video time-lapse analysis. Continuous SSP treatment of A549 cells can preserve PGCCs for at least two months in a resting state. Upon removal of SSP, A549 PGCCs restart to divide and exhibit a proliferation pattern and cellular morphology indistinguishable from cells where PGCCs originally derived from. Thus, SSP-treated A549 cells represent a simple and reliable experimental model for the reversible generation of PGCCs and their subsequent experimental analysis.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Células Gigantes/patología , Neoplasias Pulmonares/patología , Poliploidía , Estaurosporina/efectos adversos , Células A549 , Bromodesoxiuridina/metabolismo , Proliferación Celular/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Células Gigantes/efectos de los fármacos , Humanos
10.
Sci Rep ; 8(1): 2833, 2018 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-29434282

RESUMEN

In adult mammals, hematopoietic stem cells (HSCs) reside in the bone marrow and are in part regulated by the bone marrow microenvironment, called the stem cell niche. We have previously identified the bone marrow morphogen osteopontin (OPN), which is abundantly present in the bone marrow extracellular matrix, as a negative regulator of the size of the HSC pool under physiological conditions. Here, we study the impact of OPN on HSC function during aging using an OPN-knockout mouse model. We show that during aging OPN deficiency is associated with an increase in lymphocytes and a decline in erythrocytes in peripheral blood. In a bone marrow transplantation setting, aged OPN-deficient stem cells show reduced reconstitution ability likely due to insufficient differentiation of HSCs into more mature cells. In serial bone marrow transplantation, aged OPN-/- bone marrow cells fail to adequately reconstitute red blood cells and platelets, resulting in severe anemia and thrombocytopenia as well as premature deaths of recipient mice. Thus, OPN has different effects on HSCs in aged and young animals and is particularly important to maintain stem cell function in aging mice.


Asunto(s)
Envejecimiento/genética , Anemia/genética , Células Madre Hematopoyéticas/citología , Osteopontina/genética , Trombocitopenia/genética , Envejecimiento/sangre , Envejecimiento/metabolismo , Anemia/sangre , Anemia/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Eritrocitos/metabolismo , Técnicas de Inactivación de Genes , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Linfocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Osteopontina/metabolismo , Nicho de Células Madre , Trombocitopenia/sangre , Trombocitopenia/metabolismo
11.
PLoS One ; 9(10): e109266, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25286245

RESUMEN

Specialized blood cells are generated through the entire life of an organism by differentiation of a small number of hematopoietic stem cells (HSC). There are strictly regulated mechanisms assuring a constant and controlled production of mature blood cells. Although such mechanisms are not completely understood, some factors regulating cell cycle and differentiation have been identified. We have previously shown that Caspase-3 is an important regulator of HSC homeostasis and cytokine responsiveness. p21cip1/waf1 is a known cell cycle regulator, however its role in stem cell homeostasis seems to be limited. Several reports indicate interactions between p21cip1/waf1 and Caspase-3 in a cell type dependent manner. Here we studied the impact of simultaneous depletion of both factors on HSC homeostasis. Depletion of both Caspase-3 and p21cip1/waf1 resulted in an even more pronounced increase in the frequency of hematopoietic stem and progenitor cells. In addition, simultaneous deletion of both genes revealed a further increase of cell proliferation compared to single knock-outs and WT control mice, while apoptosis or self-renewal ability were not affected in any of the genotypes. Upon transplantation, p21cip1/waf1-/- bone marrow did not reveal significant alterations in engraftment of lethally irradiated mice, while Caspase-3 deficient HSPC displayed a significant reduction of blood cell production. However, when both p21cip1/waf1 and Caspase-3 were eliminated this differentiation defect caused by Caspase-3 deficiency was abrogated.


Asunto(s)
Caspasa 3/deficiencia , Caspasa 3/genética , Diferenciación Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Eliminación de Gen , Células Madre Hematopoyéticas/citología , Animales , Apoptosis/genética , Proliferación Celular/genética , Autorrenovación de las Células/genética , Femenino , Técnicas de Inactivación de Genes , Trasplante de Células Madre Hematopoyéticas , Ratones , Transducción de Señal/genética
12.
PLoS One ; 9(2): e86910, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586258

RESUMEN

Small cell lung carcinomas (SCLCs) represent highly aggressive tumors with an overall five-year survival rate in the range of 5 to 10%. Here, we show that four out of five SCLC cell lines reversibly develop a neuron-like phenotype on extracellular matrix constituents such as fibronectin, laminin or thrombospondin upon staurosporine treatment in an RGD/integrin-mediated manner. Neurite-like processes extend rapidly with an average speed of 10 µm per hour. Depending on the cell line, staurosporine treatment affects either cell cycle arrest in G2/M phase or induction of polyploidy. Neuron-like conversion, although not accompanied by alterations in the expression pattern of a panel of neuroendocrine genes, leads to changes in protein expression as determined by two-dimensional gel electrophoresis. It is likely that SCLC cells already harbour the complete molecular repertoire to convert into a neuron-like phenotype. More extensive studies are needed to evaluate whether the conversion potential of SCLC cells is suitable for therapeutic interventions.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Estaurosporina/metabolismo , Western Blotting , Adhesión Celular/fisiología , Diferenciación Celular/fisiología , Línea Celular Tumoral , Proliferación Celular , Electroforesis en Gel Bidimensional , Citometría de Flujo , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Cell Stem Cell ; 10(5): 481-2, 2012 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-22560067

RESUMEN

Until recently, aging was viewed as a fixed and irreversible process. However, in this issue of Cell Stem Cell, Florian et al. (2012) reveal a link between increased activity of the RhoGTPase Cdc42, hematopoietic stem cell polarity, and aging that can be regulated by pharmacological inhibition of Cdc42.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA