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

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Blood ; 138(14): 1249-1257, 2021 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-34166485

RESUMEN

The incidence and prognosis of clonal hematopoiesis in patients with isolated neutropenia among patients with idiopathic cytopenia of undetermined significance (ICUS), known as ICUS-N or chronic idiopathic neutropenia (CIN) patients, is poorly defined. The current study sought to investigate the frequency and clinical significance of mutations of genes implicated in myeloid malignancies using next-generation sequencing in patients with CIN (n = 185) with a long follow-up. We found that 21 (11.35%) of 185 patients carried a total of 25 somatic mutations in 6 genes with a median variant allele frequency of 12.75%. The most frequently mutated genes were DNMT3A and TET2 involving >80% of patients, followed by IDH1/2, SRSF2, and ZRSR2. The frequency of transformation to a myeloid malignancy was low in the total group of patients (5 of 185 patients [2.70%]). However, from the transformed patients, 4 belonged to the clonal group (4 of 21 [19.05%]) and 1 to the nonclonal group (1 of 164 [0.61%]), indicating that the presence of mutation(s) confers a relative risk for transformation of 31.24 (P = .0017). The variant allele frequency of the mutant clones in the transformed patients was >10% in all cases, and the genes most frequently associated with malignant transformation were SRSF2 and IDH1. No significant differences were identified between the clonal and nonclonal groups in the severity of neutropenia. Patients with clonal disease were older compared with nonclonal patients. These data contribute to the better understanding of the heterogeneous entities underlying ICUS and highlight the importance of mutation analysis for the diagnosis and prognosis of patients with unexplained neutropenias.


Asunto(s)
Hematopoyesis Clonal , Neutropenia/genética , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Estudios de Seguimiento , Frecuencia de los Genes , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Mutación , Neutropenia/diagnóstico , Pronóstico , Adulto Joven
4.
Br J Haematol ; 157(3): 299-311, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22324374

RESUMEN

The bone marrow contains specific microenvironmental stem cell niches that maintain haemopoiesis. CXCL12-expressing mesenchymal stromal cells are closely associated with the bone marrow sinusoidal endothelia, forming key elements of the haemopoietic stem cell niche, yet their ability to regulate endothelial function is not clearly defined. Given that the murine nestin(+) cell line, MS-5, provides a clonal surrogate bone marrow stromal niche capable of regulating both murine and human primitive haemopoietic stem/progenitor cell (HSC/HPC) fate in vitro, we hypothesized that MS-5 cells might also support new blood vessel formation and function. Here, for the first time, we demonstrate that this is indeed the case. Using proteome arrays, we identified HSC/HPC active angiogenic factors that are preferentially secreted by haemopoietic supportive nestin(+) MS-5 cells, including CXCL12 (SDF-1), NOV (CCN3), HGF, Angiopoietin-1 and CCL2 (MCP-1). Concentrating on CXCL12, we confirmed its presence in MS-5 conditioned media and demonstrated that its antagonist in receptor binding, AMD-3100, which mobilizes HSC/HPCs and endothelial progenitors from bone marrow, could significantly reduce MS-5 mediated human vasculogenesis in vitro, principally by regulating human endothelial cell migration. Thus, the clonal nestin(+) MS-5 murine bone marrow stromal cell line not only promotes human haemopoiesis but also induces human vasculogenesis, with CXCL12 playing important roles in both processes.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Neovascularización Fisiológica/fisiología , Inductores de la Angiogénesis/metabolismo , Animales , Células de la Médula Ósea/fisiología , Comunicación Celular/fisiología , Línea Celular , Movimiento Celular/fisiología , Proliferación Celular , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/fisiología , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Células Endoteliales/fisiología , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Humanos , Ratones , Proteómica/métodos
5.
Angiogenesis ; 14(3): 381-92, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21720855

RESUMEN

As umbilical cord blood (UCB) is a rich source of endothelial colony-forming cells (ECFC), our aim was twofold: (1) to examine potential obstetric selection criteria for achieving the highest ECFC yields from UCB units, and (2) to determine whether transient storage temperatures of fresh UCB and cryopreservation of UCB units affected ECFC yield and function. ECFC quality was assessed before and after cryopreservation by their clonogenic proliferative potential. Of the 20 factors examined, placental weight was the only statistically significant obstetric factor that predicted ECFC frequency in UCB. Studies on the effects of storage revealed that transient storage of fresh UCB at 4°C reduced ECFC yield compared with storage at 22°C, while cryopreservation of UCB MNCs significantly reduced ECFC recoveries. To our knowledge, this is the first demonstration that placental weight and temperature of storage prior to processing or culture have significant effects on ECFC frequency in UCB. Our studies further support the evidence that cryopreservation of UCB MNCs compromises ECFC recovery.


Asunto(s)
Conservación de la Sangre , Criopreservación , Parto Obstétrico , Células Endoteliales/citología , Sangre Fetal/citología , Células Madre/citología , Células Endoteliales/metabolismo , Femenino , Sangre Fetal/metabolismo , Humanos , Recién Nacido , Masculino , Tamaño de los Órganos , Placenta/citología , Placenta/metabolismo , Embarazo , Células Madre/metabolismo , Factores de Tiempo
6.
Cells ; 9(5)2020 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-32365526

RESUMEN

P0-related protein (PZR), a Noonan and Leopard syndrome target, is a member of the transmembrane Immunoglobulin superfamily. Its cytoplasmic tail contains two immune-receptor tyrosine-based inhibitory motifs (ITIMs), implicated in adhesion-dependent signaling and regulating cell adhesion and motility. PZR promotes cell migration on the extracellular matrix (ECM) molecule, fibronectin, by interacting with SHP-2 (Src homology-2 domain-containing protein tyrosine phosphatase-2), a molecule essential for skeletal development and often mutated in Noonan and Leopard syndrome patients sharing overlapping musculoskeletal abnormalities and cardiac defects. To further explore the role of PZR, we assessed the expression of PZR and its ITIM-less isoform, PZRb, in human bone marrow mesenchymal stromal cells (hBM MSC), and its ability to facilitate adhesion to and spreading and migration on various ECM molecules. Furthermore, using siRNA knockdown, confocal microscopy, and immunoprecipitation assays, we assessed PZR and PZRb interactions with ß1 integrins. PZR was the predominant isoform in hBM MSC. Migrating hBM MSCs interacted most effectively with fibronectin and required the association of PZR, but not PZRb, with the integrin, VLA-5(α5ß1), leading to modulation of focal adhesion kinase phosphorylation and vinculin levels. This raises the possibility that dysregulation of PZR function may modify hBM MSC migratory behavior, potentially contributing to skeletal abnormalities.


Asunto(s)
Movimiento Celular/fisiología , Fibronectinas/metabolismo , Integrina alfa5beta1/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteínas Portadoras/genética , Humanos , Fosfoproteínas/metabolismo , Transducción de Señal/fisiología , Tirosina/metabolismo
7.
Sci Rep ; 7: 44137, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28276476

RESUMEN

Circulating endothelial colony forming cells (ECFCs) contribute to vascular repair where they are a target for therapy. Since ECFC proliferative potential is increased in cord versus peripheral blood and to define regulatory factors controlling this proliferation, we compared the miRNA profiles of cord blood and peripheral blood ECFC-derived cells. Of the top 25 differentially regulated miRNAs selected, 22 were more highly expressed in peripheral blood ECFC-derived cells. After validating candidate miRNAs by q-RT-PCR, we selected miR-193a-3p for further investigation. The miR-193a-3p mimic reduced cord blood ECFC-derived cell proliferation, migration and vascular tubule formation, while the miR-193a-3p inhibitor significantly enhanced these parameters in peripheral blood ECFC-derived cells. Using in silico miRNA target database analyses combined with proteome arrays and luciferase reporter assays of miR-193a-3p mimic treated cord blood ECFC-derived cells, we identified 2 novel miR-193a-3p targets, the high mobility group box-1 (HMGB1) and the hypoxia upregulated-1 (HYOU1) gene products. HMGB1 silencing in cord blood ECFC-derived cells confirmed its role in regulating vascular function. Thus, we show, for the first time, that miR-193a-3p negatively regulates human ECFC vasculo/angiogenesis and propose that antagonising miR-193a-3p in less proliferative and less angiogenic ECFC-derived cells will enhance their vasculo/angiogenic function.


Asunto(s)
Movimiento Celular/fisiología , Proliferación Celular/fisiología , Regulación hacia Abajo/fisiología , Células Endoteliales/metabolismo , Proteína HMGB1/biosíntesis , MicroARNs/metabolismo , Células Endoteliales/citología , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/metabolismo , Proteína HMGB1/genética , Humanos , MicroARNs/genética
8.
Stem Cells Dev ; 23(22): 2730-43, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24940843

RESUMEN

Proangiogenic factors, vascular endothelial growth factor (VEGF), and fibroblast growth factor-2 (FGF-2) prime endothelial cells to respond to "hematopoietic" chemokines and cytokines by inducing/upregulating expression of the respective chemokine/cytokine receptors. Coculture of human endothelial colony forming cell (ECFC)-derived cells with human stromal cells in the presence of VEGF and FGF-2 for 14 days resulted in upregulation of the "hematopoietic" chemokine CXCL12 and its CXCR4 receptor by day 3 of coculture. Chronic exposure to the CXCR4 antagonist AMD3100 in this vasculo/angiogenesis assay significantly reduced vascular tubule formation, an observation recapitulated by delayed AMD3100 addition. While AMD3100 did not affect ECFC-derived cell proliferation, it did demonstrate a dual action. First, over the later stages of the 14-day cocultures, AMD3100 delayed tubule organization into maturing vessel networks, resulting in enhanced endothelial cell retraction and loss of complexity as defined by live cell imaging. Second, at earlier stages of cocultures, we observed that AMD3100 significantly inhibited the integration of exogenous ECFC-derived cells into established, but immature, vascular networks. Comparative proteome profiler array analyses of ECFC-derived cells treated with AMD3100 identified changes in expression of potential candidate molecules involved in adhesion and/or migration. Blocking antibodies to CD31, but not CD146 or CD166, reduced the ECFC-derived cell integration into these extant vascular networks. Thus, CXCL12 plays a key role not only in endothelial cell sensing and guidance, but also in promoting the integration of ECFC-derived cells into developing vascular networks.


Asunto(s)
Vasos Sanguíneos/fisiología , Quimiocina CXCL12/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Sistema Hematopoyético/metabolismo , Sistema Hematopoyético/fisiología , Bencilaminas , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo/métodos , Ciclamas , Células Endoteliales/efectos de los fármacos , Sistema Hematopoyético/efectos de los fármacos , Compuestos Heterocíclicos/farmacología , Humanos , Receptores CXCR4/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
PLoS One ; 8(1): e54747, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23359810

RESUMEN

Human amniotic fluid obtained at amniocentesis, when cultured, generates at least two morphologically distinct mesenchymal stem/stromal cell (MSC) subsets. Of these, the spindle shaped amniotic fluid MSCs (SS-AF-MSCs) contain multipotent cells with enhanced adipogenic, osteogenic and chondrogenic capacity. Here, we demonstrate, for the first time, the capacity of these SS-AF-MSCs to support neovascularization by umbilical cord blood (UCB) endothelial colony forming cell (ECFC) derived cells in both in vitro and in vivo models. Interestingly, although the kinetics of vascular tubule formation in vitro were similar when the supporting SS-AF-MSCs were compared with the best vasculogenic supportive batches of bone marrow MSCs (BMSCs) or human dermal fibroblasts (hDFs), SS-AF-MSCs supported vascular tubule formation in vivo more effectively than BMSCs. In NOD/SCID mice, the human vessels inosculated with murine vessels demonstrating their functionality. Proteome profiler array analyses revealed both common and distinct secretion profiles of angiogenic factors by the SS-AF-MSCs as opposed to the hDFs and BMSCs. Thus, SS-AF-MSCs, which are considered to be less mature developmentally than adult BMSCs, and intermediate between adult and embryonic stem cells in their potentiality, have the additional and very interesting potential of supporting increased neovascularisation, further enhancing their promise as vehicles for tissue repair and regeneration.


Asunto(s)
Líquido Amniótico/citología , Células Madre Mesenquimatosas/citología , Neovascularización Fisiológica , Proliferación Celular , Medios de Cultivo Condicionados , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Inmunofenotipificación
10.
Nat Med ; 19(3): 351-7, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23435171

RESUMEN

Previous attempts to identify neuroprotective targets by studying the ischemic cascade and devising ways to suppress it have failed to translate to efficacious therapies for acute ischemic stroke. We hypothesized that studying the molecular determinants of endogenous neuroprotection in two well-established paradigms, the resistance of CA3 hippocampal neurons to global ischemia and the tolerance conferred by ischemic preconditioning (IPC), would reveal new neuroprotective targets. We found that the product of the tuberous sclerosis complex 1 gene (TSC1), hamartin, is selectively induced by ischemia in hippocampal CA3 neurons. In CA1 neurons, hamartin was unaffected by ischemia but was upregulated by IPC preceding ischemia, which protects the otherwise vulnerable CA1 cells. Suppression of hamartin expression with TSC1 shRNA viral vectors both in vitro and in vivo increased the vulnerability of neurons to cell death following oxygen glucose deprivation (OGD) and ischemia. In vivo, suppression of TSC1 expression increased locomotor activity and decreased habituation in a hippocampal-dependent task. Overexpression of hamartin increased resistance to OGD by inducing productive autophagy through an mTORC1-dependent mechanism.


Asunto(s)
Autofagia , Región CA1 Hipocampal/fisiología , Región CA3 Hipocampal/fisiología , Hipoxia-Isquemia Encefálica/prevención & control , Fármacos Neuroprotectores/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Animales , Autofagia/efectos de los fármacos , Región CA1 Hipocampal/metabolismo , Región CA3 Hipocampal/metabolismo , Células Cultivadas , Hipoxia , Hipoxia-Isquemia Encefálica/metabolismo , Precondicionamiento Isquémico , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos , Prosencéfalo/irrigación sanguínea , Prosencéfalo/lesiones , Proteínas/antagonistas & inhibidores , Proteínas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Ratas , Ratas Wistar , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética
11.
Tissue Eng Part C Methods ; 18(2): 143-55, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21943098

RESUMEN

Skin has a remarkable capacity for regeneration, but age- and diabetes-related vascular problems lead to chronic non-healing wounds for many thousands of U.K. patients. There is a need for new therapeutic approaches to treat these resistant wounds. Donor mesenchymal stem/stromal cells (MSCs) have been shown to assist cutaneous wound healing by accelerating re-epithelialization. The aim of this work was to devise a low risk and convenient delivery method for transferring these cells to wound beds. Plasma polymerization was used to functionalize the surface of medical-grade silicone with acrylic acid. Cells attached well to these carriers, and culture for up to 3 days on the carriers did not significantly affect their phenotype or ability to support vascular tubule formation. These carriers were then used to transfer MSCs onto human dermis. Cell transfer was confirmed using an MTT assay to assess viable cell numbers and enhanced green fluorescent protein-labeled MSCs to demonstrate that the cells post-transfer attached to the dermis. We conclude that this synthetic carrier membrane is a promising approach for delivery of therapeutic MSCs and opens the way for future studies to evaluate its impact on repairing difficult skin wounds.


Asunto(s)
Acrilatos/farmacología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Siliconas/farmacología , Piel/patología , Cicatrización de Heridas/efectos de los fármacos , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Dermis/efectos de los fármacos , Epitelio/efectos de los fármacos , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Fenotipo , Espectroscopía de Fotoelectrones , Piel/efectos de los fármacos
12.
Burns ; 38(5): 691-701, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22360956

RESUMEN

Forming a microcirculation is critical for vascularisation of artificial skin substitutes. One strategy to improve speed of grafting is to pre-form microvascular networks in the substitute before applying to a wound. For clinical application, this requires sufficient functional endothelial cell numbers. In vitro endothelial colony forming cells (ECFCs) derived cells were expanded from cord and adult blood donations and co-cultured with human dermal fibroblasts or bone marrow mesenchymal stem/stromal cells to form microvascular networks in the presence or absence of dermal substitutes which are in clinical use. The number of endothelial cells generated ranged from 1.03×10(9) to 2.18×10(11) from 10 adult blood donations and 1×10(12) to 1.76×10(13) from 6 cord blood units after 50 days in culture. Two adult donations failed to generate ECFCs. Both cord and adult blood cells formed 2D microvascular networks in vitro, although there was a significant difference in the functional capacity of adult and cord blood ECFCs. While co-culture of the latter within dermal substitutes Matriderm or Integra demonstrated the formation of 3D microvascular networks penetrating 100µm, enhanced expansion, while maintaining functional capacity, of adult blood cells is required for fully pre-vascularising the clinical grade acellular dermal substitutes used here prior to applying these to burns.


Asunto(s)
Quemaduras/terapia , Endotelio Vascular/citología , Microvasos/crecimiento & desarrollo , Piel Artificial , Piel/irrigación sanguínea , Adulto , Células Cultivadas , Técnicas de Cocultivo , Estudios de Factibilidad , Femenino , Sangre Fetal/citología , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Persona de Mediana Edad , Células Madre/citología , Células del Estroma/citología , Ingeniería de Tejidos/métodos
13.
J R Soc Interface ; 7 Suppl 6: S731-51, 2010 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-20843839

RESUMEN

Neovascularization or new blood vessel formation is of utmost importance not only for tissue and organ development and for tissue repair and regeneration, but also for pathological processes, such as tumour development. Despite this, the endothelial lineage, its origin, and the regulation of endothelial development and function either intrinsically from stem cells or extrinsically by proangiogenic supporting cells and other elements within local and specific microenvironmental niches are still not fully understood. There can be no doubt that for most tissues and organs, revascularization represents the holy grail for tissue repair, with autologous endothelial stem/progenitor cells, their proangiogenic counterparts and the products of these cells all being attractive targets for therapeutic intervention. Historically, a great deal of controversy has surrounded the identification and origin of cells and factors that contribute to revascularization, the use of such cells or their products as biomarkers to predict and monitor tissue damage and repair or tumour progression and therapeutic responses, and indeed their efficacy in revascularizing and repairing damaged tissues. Here, we will review the role of endothelial progenitor cells and of supporting proangiogenic cells and their products, principally in humans, as diagnostic and therapeutic agents for wound repair and tissue regeneration.


Asunto(s)
Células Endoteliales/citología , Neovascularización Fisiológica/fisiología , Células Madre/fisiología , Inductores de la Angiogénesis/farmacología , Animales , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Medicina Regenerativa , Células Madre/metabolismo , Ingeniería de Tejidos , Cicatrización de Heridas
14.
Stem Cells Dev ; 18(2): 359-75, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18662129

RESUMEN

Revascularization of the damaged tissue is pivotal to tissue repair. Here, by bringing together two in vitro model systems, we have been able to examine (1) the ability of human umbilical vein endothelial cells (HUVEC) containing a complete hierarchy of endothelial progenitors derived from the human umbilical cord to generate vascular tubules within a human stromal niche in vitro and (2) the effects of exposure to low oxygen tensions on endothelial progenitor cell proliferation and tubule formation in vitro. Our results demonstrate that high proliferative potential endothelial colony forming cells (HPP-ECFC) from cultured HUVEC preferentially contribute to vascular tubule formation in vitro and that these progenitor cells are concentrated in the CD34(lo/-) fraction. HUVEC were initially resistant when exposed to hypoxia (1.5% O(2)) for short periods (1-2 days), but sustained chronic hypoxia (4-14 days) inhibited their ability to proliferate. This was reflected by a loss in their ability to form tubules in cocultures of human dermal fibroblasts (hDFs). In contrast, an acute exposure to low oxygen tensions (1.5% O(2) for 24 h) followed by reoxygenation did not adversely affect the capacity of these cells to both proliferate and form vascular tubules in vitro.These studies therefore provide a model system to study the influences of the microenvironmental niche and modification of this niche on vascular tubule formation in vitro from HPP-ECFC.


Asunto(s)
Células Endoteliales/citología , Neovascularización Fisiológica , Células Madre/citología , Cordón Umbilical/citología , Antígenos CD34/metabolismo , Apoptosis , Recuento de Células , Hipoxia de la Célula , Membrana Celular/metabolismo , Proliferación Celular , Células Cultivadas , Células Clonales , Técnicas de Cocultivo , Ensayo de Unidades Formadoras de Colonias , Dermis/citología , Células Endoteliales/ultraestructura , Fibroblastos/citología , Humanos , Necrosis , Células Madre/ultraestructura , Venas Umbilicales/citología
15.
J Cell Biochem ; 102(4): 955-69, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17440960

RESUMEN

The human P zero-related protein (hPZR) has a unique function in regulating cell migration. This activity is dependent on both its cytoplasmic immunoreceptor tyrosine inhibitory motif (ITIM) and its interaction with the tyrosine protein phosphatase, src homology phosphatase-2 (SHP-2). Here, using in silico and cDNA cloning approaches, we identify the murine ITIM-containing hPZR ortholog, mPZR, together with its ITIM-less isoform, mPZRb. We demonstrate that, like hPZR, these type 1 integral murine transmembrane isoforms are derived by differential splicing from a single gene transcription unit on mouse chromosome 1, and differ only in the sequence of their cytoplasmic domains. Importantly, mPZR mimicks hPZR functionally by accelerating SHP-2-mediated cell migration on fibronectin. Interestingly, we further demonstrate that although neither mPZR nor mPZRb is expressed in murine pluripotent embryonic stem cells, they first appear at approximately day 3 of blastocyst formation in vivo and of embryoid body formation in vitro. These studies thus provide the basis for defining the function of the mPZR isoforms in vivo, particularly with respect to their roles in regulating SHP-2-dependent cell migration during development.


Asunto(s)
Movimiento Celular , Desarrollo Embrionario , Fibronectinas , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/fisiología , Fosfoproteínas/fisiología , Secuencia de Aminoácidos , Animales , Blastocisto/enzimología , Biología Computacional , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Fosfoproteínas/genética , Isoformas de Proteínas/química , Proteína Tirosina Fosfatasa no Receptora Tipo 11
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA