Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 18(7): 762-770, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28504698

RESUMEN

Trafficking of tissue dendritic cells (DCs) via lymph is critical for the generation of cellular immune responses in draining lymph nodes (LNs). In the current study we found that DCs docked to the basolateral surface of lymphatic vessels and transited to the lumen through hyaluronan-mediated interactions with the lymph-specific endothelial receptor LYVE-1, in dynamic transmigratory-cup-like structures. Furthermore, we show that targeted deletion of the gene Lyve1, antibody blockade or depletion of the DC hyaluronan coat not only delayed lymphatic trafficking of dermal DCs but also blunted their capacity to prime CD8+ T cell responses in skin-draining LNs. Our findings uncovered a previously unknown function for LYVE-1 and show that transit through the lymphatic network is initiated by the recognition of leukocyte-derived hyaluronan.


Asunto(s)
Células Dendríticas/inmunología , Células Endoteliales/metabolismo , Glicoproteínas/genética , Ácido Hialurónico/metabolismo , Vasos Linfáticos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animales , Movimiento Celular/inmunología , Células Dendríticas/metabolismo , Endotelio Linfático/citología , Endotelio Linfático/metabolismo , Citometría de Flujo , Glicoproteínas/metabolismo , Humanos , Inmunidad Celular/inmunología , Ganglios Linfáticos/inmunología , Proteínas de Transporte de Membrana , Ratones , Ratones Noqueados , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología
2.
Hum Mol Genet ; 29(21): 3516-3531, 2021 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-33105479

RESUMEN

Neurodevelopmental disorder with microcephaly, hypotonia and variable brain anomalies (NMIHBA) is an autosomal recessive neurodevelopmental and neurodegenerative disorder characterized by global developmental delay and severe intellectual disability. Microcephaly, progressive cortical atrophy, cerebellar hypoplasia and delayed myelination are neurological hallmarks in affected individuals. NMIHBA is caused by biallelic variants in PRUNE1 encoding prune exopolyphosphatase 1. We provide in-depth clinical description of two affected siblings harboring compound heterozygous variant alleles, c.383G > A (p.Arg128Gln), c.520G > T (p.Gly174*) in PRUNE1. To gain insights into disease biology, we biochemically characterized missense variants within the conserved N-terminal aspartic acid-histidine-histidine (DHH) motif and provide evidence that they result in the destabilization of protein structure and/or loss of exopolyphosphatase activity. Genetic ablation of Prune1 results in midgestational lethality in mice, associated with perturbations to embryonic growth and vascular development. Our findings suggest that NMIHBA results from hypomorphic variant alleles in humans and underscore the potential key role of PRUNE1 exopolyphoshatase activity in neurodevelopment.


Asunto(s)
Ácido Anhídrido Hidrolasas/deficiencia , Discapacidad Intelectual/patología , Microcefalia/patología , Hipotonía Muscular/patología , Mutación , Trastornos del Neurodesarrollo/patología , Monoéster Fosfórico Hidrolasas/genética , Alelos , Animales , Preescolar , Femenino , Humanos , Lactante , Discapacidad Intelectual/etiología , Discapacidad Intelectual/metabolismo , Masculino , Ratones , Microcefalia/etiología , Microcefalia/metabolismo , Hipotonía Muscular/etiología , Hipotonía Muscular/metabolismo , Trastornos del Neurodesarrollo/etiología , Trastornos del Neurodesarrollo/metabolismo , Linaje , Fenotipo
3.
Angiogenesis ; 23(2): 179-192, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31754927

RESUMEN

Angiogenesis is largely driven by motile endothelial tip-cells capable of invading avascular tissue domains and enabling new vessel formation. Highly responsive to Vascular Endothelial Growth-Factor-A (VEGFA), endothelial tip-cells also suppress angiogenic sprouting in adjacent stalk cells, and thus have been a primary therapeutic focus in addressing neovascular pathologies. Surprisingly, however, there remains a paucity of specific endothelial tip-cell markers. Here, we employ transcriptional profiling and a lacZ reporter allele to identify Kcne3 as an early and selective endothelial tip-cell marker in multiple angiogenic contexts. In development, Kcne3 expression initiates during early phases of angiogenesis (E9) and remains specific to endothelial tip-cells, often adjacent to regions expressing VEGFA. Consistently, Kcne3 activation is highly responsive to exogenous VEGFA but maintains tip-cell specificity throughout normal retinal angiogenesis. We also demonstrate endothelial tip-cell selectivity of Kcne3 in several injury and tumor models. Together, our data show that Kcne3 is a unique marker of sprouting angiogenic tip-cells and offers new opportunities for investigating and targeting this cell type.


Asunto(s)
Células Endoteliales/fisiología , Neovascularización Patológica/genética , Neovascularización Fisiológica/genética , Canales de Potasio con Entrada de Voltaje/genética , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Animales Recién Nacidos , Diferenciación Celular/genética , Células Cultivadas , Retinopatía Diabética/genética , Retinopatía Diabética/patología , Embrión de Mamíferos , Células Endoteliales/patología , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Morfogénesis/genética , Neovascularización Patológica/metabolismo , Embarazo , Retina/metabolismo , Retina/patología , Vasos Retinianos/metabolismo , Vasos Retinianos/patología
4.
Biol Reprod ; 100(3): 686-696, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30289441

RESUMEN

The Adisintegrin and metalloprotease domain-containing (ADAM) family of proteins is involved in cell adhesion, migration, proteolysis, and signaling. Many ADAMs are required for reproduction; however, the role of Adam6 has remained largely unknown. In the course of humanizing the mouse immunoglobulin heavy chain (IgH) locus, we generated Adam6-deficient mice that demonstrate severe subfertility. We decided to elucidate the role of ADAM6 in fertility and explore the underlying mechanisms. Despite normal sperm development and motility, Adam6-deficient mice display diminished male fertility, have abnormal sperm adhesion, and most importantly cannot transition from uterus to oviduct. To test whether ADAM6 is required for sperm's binding to extracellular matrix (ECM) components, we used a panel of ECM components and showed that unlike normal sperm, Adam6-deficient sperm cannot bind fibronectin, laminin, and tenascin. Reintroduction of Adam6 into these deficient mice repaired sperm interaction with ECM, restored male fertility, and corrected the sperm transport deficit. Together, our data suggest that ADAM6, either alone or in complex with other proteins, aids sperm transport through the female reproductive tract by providing a temporary site of attachment of sperm to ECM components prior to ascent into the oviduct.


Asunto(s)
Proteínas ADAM/metabolismo , Infertilidad Masculina/genética , Motilidad Espermática/fisiología , Espermatozoides/fisiología , Proteínas ADAM/genética , Animales , Femenino , Masculino , Ratones , Ratones Noqueados , Oviductos , Motilidad Espermática/genética
5.
Immunity ; 30(1): 67-79, 2009 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-19110448

RESUMEN

Notch1 signaling is required for T cell development and has been implicated in fate decisions in the thymus. We showed that Notch1 deletion in progenitor T cells (pro-T cells) revealed their latent developmental potential toward becoming conventional and plasmacytoid dendritic cells. In addition, Notch1 deletion in pro-T cells resulted in large numbers of thymic B cells, previously explained by T-to-B cell fate conversion. Single-cell genotyping showed, however, that the majority of these thymic B cells arose from Notch1-sufficient cells by a cell-extrinsic pathway. Fate switching nevertheless exists for a subset of thymic B cells originating from Notch1-deleted pro-T cells. Chimeric mice lacking the Notch ligand delta-like 4 (Dll4) in thymus epithelium revealed an essential role for Dll4 in T cell development. Thus, Notch1-Dll4 signaling fortifies T cell commitment by suppressing non-T cell lineage potential in pro-T cells, and normal Notch1-driven T cell development repels excessive B cells in the thymus.


Asunto(s)
Linfocitos B/inmunología , Células Dendríticas/inmunología , Eliminación de Gen , Receptor Notch1/genética , Linfocitos T/inmunología , Timo/citología , Animales , Linaje de la Célula , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Receptor Notch1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Proc Natl Acad Sci U S A ; 112(6): 1845-9, 2015 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-25624481

RESUMEN

G protein-coupled receptor 17 (GPR17) was recently reported to be a Foxo1 target in agouti-related peptide (AGRP) neurons. Intracerebroventricular injection of GPR17 agonists induced food intake, whereas administration of an antagonist to the receptor reduced feeding. These data lead to the conclusion that pharmacological modulation of GPR17 has therapeutic potential to treat obesity. Here we report that mice deficient in Gpr17 (Gpr17(-/-)) have similar food intake and body weight compared with their wild-type littermates. Gpr17(-/-) mice have normal hypothalamic Agrp mRNA expression, AGRP plasma levels, and metabolic rate. GPR17 deficiency in mice did not affect glucose homeostasis or prevent fat-induced insulin resistance. These data do not support a role for GPR17 in the control of food intake, body weight, or glycemic control.


Asunto(s)
Ingestión de Alimentos/genética , Glucosa/metabolismo , Proteínas del Tejido Nervioso/genética , Receptores Acoplados a Proteínas G/genética , Proteína Relacionada con Agouti/metabolismo , Análisis de Varianza , Animales , Secuencia de Bases , Composición Corporal/efectos de los fármacos , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Neuronas/metabolismo , Análisis de Secuencia de ARN , Factores de Tiempo , Microtomografía por Rayos X
7.
Proc Natl Acad Sci U S A ; 110(34): E3179-88, 2013 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-23918385

RESUMEN

Conditional mutagenesis is becoming a method of choice for studying gene function, but constructing conditional alleles is often laborious, limited by target gene structure, and at times, prone to incomplete conditional ablation. To address these issues, we developed a technology termed conditionals by inversion (COIN). Before activation, COINs contain an inverted module (COIN module) that lies inertly within the antisense strand of a resident gene. When inverted into the sense strand by a site-specific recombinase, the COIN module causes termination of the target gene's transcription and simultaneously provides a reporter for tracking this event. COIN modules can be inserted into natural introns (intronic COINs) or directly into coding exons as part of an artificial intron (exonic COINs), greatly simplifying allele design and increasing flexibility over previous conditional KO approaches. Detailed analysis of over 20 COIN alleles establishes the reliability of the method and its broad applicability to any gene, regardless of exon-intron structure. Our extensive testing provides rules that help ensure success of this approach and also explains why other currently available conditional approaches often fail to function optimally. Finally, the ability to split exons using the COIN's artificial intron opens up engineering modalities for the generation of multifunctional alleles.


Asunto(s)
Alelos , Silenciador del Gen , Ingeniería Genética/métodos , Mutagénesis Insercional/métodos , Inversión de Secuencia/genética , ADN Nucleotidiltransferasas/metabolismo
8.
Dev Biol ; 383(1): 90-105, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23994639

RESUMEN

WNT signaling is critical in most aspects of skeletal development and homeostasis, and antagonists of WNT signaling are emerging as key regulatory proteins with great promise as therapeutic agents for bone disorders. Here we show that Sost and its paralog Sostdc1 emerged through ancestral genome duplication and their expression patterns have diverged to delineate non-overlapping domains in most organ systems including musculoskeletal, cardiovascular, nervous, digestive, reproductive and respiratory. In the developing limb, Sost and Sostdc1 display dynamic expression patterns with Sost being restricted to the distal ectoderm and Sostdc1 to the proximal ectoderm and the mesenchyme. While Sostdc1(-/-) mice lack any obvious limb or skeletal defects, Sost(-/-) mice recapitulate the hand defects described for Sclerosteosis patients. However, elevated WNT signaling in Sost(-/-); Sostdc1(-/-) mice causes misregulation of SHH signaling, ectopic activation of Sox9 in the digit 1 field and preaxial polydactyly in a Gli1- and Gli3-dependent manner. In addition, we show that the syndactyly documented in Sclerosteosis is present in both Sost(-/-) and Sost(-/-); Sostdc1(-/-) mice, and is driven by misregulation of Fgf8 in the AER, a region lacking Sost and Sostdc1 expression. This study highlights the complexity of WNT signaling in skeletal biology and disease and emphasizes how redundant mechanism and non-cell autonomous effects can synergize to unveil new intricate phenotypes caused by elevated WNT signaling.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Ectodermo/embriología , Extremidades/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Glicoproteínas/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Vía de Señalización Wnt/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Morfogenéticas Óseas/genética , Biología Computacional , Ectodermo/metabolismo , Evolución Molecular , Glicoproteínas/genética , Proteínas Hedgehog/metabolismo , Hibridación in Situ , Péptidos y Proteínas de Señalización Intercelular , Ratones , Ratones Noqueados , Análisis por Micromatrices , Factor de Transcripción SOX9/metabolismo , Proteína Gli3 con Dedos de Zinc
9.
Proc Natl Acad Sci U S A ; 108(7): 2807-12, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21282641

RESUMEN

The vasculature of the CNS is structurally and functionally distinct from that of other organ systems and is particularly prone to developmental abnormalities and hemorrhage. Although other embryonic tissues undergo primary vascularization, the developing nervous system is unique in that it is secondarily vascularized by sprouting angiogenesis from a surrounding perineural plexus. This sprouting angiogenesis requires the TGF-ß and Wnt pathways because ablation of these pathways results in aberrant sprouting and hemorrhage. We have genetically deleted Gpr124, a member of the large family of long N-terminal group B G protein-coupled receptors, few members of which have identified ligands or well-defined biologic functions in mammals. We show that, in the developing CNS, Gpr124 is specifically expressed in the vasculature and is absolutely required for proper angiogenic sprouting into the developing neural tube. Embryos lacking Gpr124 exhibit vascular defects characterized by delayed vascular penetration, formation of pathological glomeruloid tufts within the CNS, and hemorrhage. In addition, they display defects in palate and lung development, two processes in which TGF-ß and/or Wnt pathways also play important roles. We also show that TGF-ß stimulates Gpr124 expression, and ablation of Gpr124 results in perturbed TGF-ß pathway activation, suggesting roles for Gpr124 in modulating TGF-ß signaling. These results represent a unique function attributed to a long N-terminal group B-type G protein-coupled receptor in a mammalian system.


Asunto(s)
Sistema Nervioso Central/irrigación sanguínea , Sistema Nervioso Central/embriología , Neovascularización Fisiológica/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Embrión de Mamíferos , Ingeniería Genética , Técnicas Histológicas , Inmunohistoquímica , Hibridación in Situ , Pulmón/embriología , Pulmón/metabolismo , Ratones , Análisis por Micromatrices , Hueso Paladar/embriología , Hueso Paladar/metabolismo , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Wnt/metabolismo
10.
Nat Med ; 12(2): 235-9, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16462802

RESUMEN

The angiopoietins Ang-1 and Ang-2 have been identified as ligands of the receptor tyrosine kinase Tie-2 (refs. 1,2). Paracrine Ang-1-mediated activation of Tie-2 acts as a regulator of vessel maturation and vascular quiescence. In turn, the antagonistic ligand Ang-2 acts by an autocrine mechanism and is stored in endothelial Weibel-Palade bodies from where it can be rapidly released upon stimulation. The rapid release of Ang-2 implies functions of the angiopoietin-Tie system beyond its established role during vascular morphogenesis as a regulator of rapid vascular responses. Here we show that mice deficient in Ang-2 (encoded by the gene Angpt2) cannot elicit an inflammatory response in thioglycollate-induced or Staphylococcus aureus-induced peritonitis, or in the dorsal skinfold chamber model. Recombinant Ang-2 restores the inflammation defect in Angpt2(-/-) mice. Intravital microscopy showed normal TNF-alpha-induced leukocyte rolling in the vasculature of Angpt2(-/-)mice, but rolling cells did not firmly adhere to activated endothelium. Cellular experiments showed that Ang-2 promotes adhesion by sensitizing endothelial cells toward TNF-alpha and modulating TNF-alpha-induced expression of endothelial cell adhesion molecules. Together, these findings identify Ang-2 as an autocrine regulator of endothelial cell inflammatory responses. Ang-2 thereby acts as a switch of vascular responsiveness exerting a permissive role for the activities of proinflammatory cytokines.


Asunto(s)
Angiopoyetina 2/fisiología , Inflamación/etiología , Factor de Necrosis Tumoral alfa/fisiología , Angiopoyetina 1/fisiología , Angiopoyetina 2/deficiencia , Angiopoyetina 2/genética , Animales , Citocinas/fisiología , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Humanos , Inflamación/patología , Inflamación/fisiopatología , Mediadores de Inflamación/fisiología , Ratones , Ratones Noqueados , Modelos Biológicos , Neovascularización Patológica , Transducción de Señal
11.
Nature ; 449(7160): 346-50, 2007 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-17882221

RESUMEN

Adult mammalian testis is a source of pluripotent stem cells. However, the lack of specific surface markers has hampered identification and tracking of the unrecognized subset of germ cells that gives rise to multipotent cells. Although embryonic-like cells can be derived from adult testis cultures after only several weeks in vitro, it is not known whether adult self-renewing spermatogonia in long-term culture can generate such stem cells as well. Here, we show that highly proliferative adult spermatogonial progenitor cells (SPCs) can be efficiently obtained by cultivation on mitotically inactivated testicular feeders containing CD34+ stromal cells. SPCs exhibit testicular repopulating activity in vivo and maintain the ability in long-term culture to give rise to multipotent adult spermatogonial-derived stem cells (MASCs). Furthermore, both SPCs and MASCs express GPR125, an orphan adhesion-type G-protein-coupled receptor. In knock-in mice bearing a GPR125-beta-galactosidase (LacZ) fusion protein under control of the native Gpr125 promoter (GPR125-LacZ), expression in the testis was detected exclusively in spermatogonia and not in differentiated germ cells. Primary GPR125-LacZ SPC lines retained GPR125 expression, underwent clonal expansion, maintained the phenotype of germline stem cells, and reconstituted spermatogenesis in busulphan-treated mice. Long-term cultures of GPR125+ SPCs (GSPCs) also converted into GPR125+ MASC colonies. GPR125+ MASCs generated derivatives of the three germ layers and contributed to chimaeric embryos, with concomitant downregulation of GPR125 during differentiation into GPR125- cells. MASCs also differentiated into contractile cardiac tissue in vitro and formed functional blood vessels in vivo. Molecular bookmarking by GPR125 in the adult mouse and, ultimately, in the human testis could enrich for a population of SPCs for derivation of GPR125+ MASCs, which may be employed for genetic manipulation, tissue regeneration and revascularization of ischaemic organs.


Asunto(s)
Células Madre Adultas/citología , Células Madre Multipotentes/citología , Receptores Acoplados a Proteínas G/metabolismo , Espermatogonias/citología , Espermatogonias/metabolismo , Células Madre Adultas/metabolismo , Envejecimiento , Animales , Vasos Sanguíneos/citología , Busulfano , Diferenciación Celular , Línea Celular , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Células Madre Multipotentes/metabolismo , Miocardio/citología , Regeneración , Testículo/citología , Testículo/metabolismo
12.
Nature ; 444(7122): 1032-7, 2006 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-17183313

RESUMEN

Tumour growth requires accompanying expansion of the host vasculature, with tumour progression often correlated with vascular density. Vascular endothelial growth factor (VEGF) is the best-characterized inducer of tumour angiogenesis. We report that VEGF dynamically regulates tumour endothelial expression of Delta-like ligand 4 (Dll4), which was previously shown to be absolutely required for normal embryonic vascular development. To define Dll4 function in tumour angiogenesis, we manipulated this pathway in murine tumour models using several approaches. Here we show that blockade resulted in markedly increased tumour vascularity, associated with enhanced angiogenic sprouting and branching. Paradoxically, this increased vascularity was non-productive-as shown by poor perfusion and increased hypoxia, and most importantly, by decreased tumour growth-even for tumours resistant to anti-VEGF therapy. Thus, VEGF-induced Dll4 acts as a negative regulator of tumour angiogenesis; its blockade results in a striking uncoupling of tumour growth from vessel density, presenting a novel therapeutic approach even for tumours resistant to anti-VEGF therapies.


Asunto(s)
Proteínas de la Membrana/metabolismo , Neoplasias/irrigación sanguínea , Neoplasias/patología , Neovascularización Patológica , Adenoviridae/genética , Animales , Hipoxia de la Célula , Línea Celular Tumoral , Regulación de la Expresión Génica , Genes Reporteros/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Ratones , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Ratas , Receptores Notch/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
Proc Natl Acad Sci U S A ; 106(52): 22399-404, 2009 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-20018779

RESUMEN

Inhibiting angiogenesis has become an effective approach for treating cancer and other diseases. However, our understanding of signaling pathways in tumor angiogenesis has been limited by the embryonic lethality of many gene knockouts. To overcome this limitation, we used the plasticity of embryonic stem (ES) cells to develop a unique approach to study tumor angiogenesis. Murine ES cells can be readily manipulated genetically; in addition, ES cells implanted subcutaneously in mice develop into tumors that contain a variety of cell types (teratomas). We show that ES cells differentiate into bona fide endothelial cells within the teratoma, and that these ES-derived endothelial cells form part of the functional tumor vasculature. Using this powerful and flexible system, the Angiopoietin/Tie2 system is shown to have a key role in the regulation of tumor vessel size. Endothelial differentiation in the ES teratoma model allows gene-targeting methods to be used in the study of tumor angiogenesis.


Asunto(s)
Células Madre Embrionarias/enzimología , Células Madre Embrionarias/patología , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/enzimología , Neovascularización Patológica , Proteínas Tirosina Quinasas Receptoras/fisiología , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/fisiología , Angiopoyetinas/antagonistas & inhibidores , Animales , Diferenciación Celular , Línea Celular , Modelos Animales de Enfermedad , Células Endoteliales/enzimología , Células Endoteliales/patología , Ratones , Ratones SCID , Neoplasias Experimentales/etiología , Receptor TIE-2 , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/deficiencia , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Teratoma/irrigación sanguínea , Teratoma/enzimología , Teratoma/etiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología
14.
J Clin Invest ; 118(6): 2111-20, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18497886

RESUMEN

Colon cancer stem cells are believed to originate from a rare population of putative CD133+ intestinal stem cells. Recent publications suggest that a small subset of colon cancer cells expresses CD133, and that only these CD133+ cancer cells are capable of tumor initiation. However, the precise contribution of CD133+ tumor-initiating cells in mediating colon cancer metastasis remains unknown. Therefore, to temporally and spatially track the expression of CD133 in adult mice and during tumorigenesis, we generated a knockin lacZ reporter mouse (CD133lacZ/+), in which the expression of lacZ is driven by the endogenous CD133 promoters. Using this model and immunostaining, we discovered that CD133 expression in colon is not restricted to stem cells; on the contrary, CD133 is ubiquitously expressed on differentiated colonic epithelium in both adult mice and humans. Using Il10-/-CD133lacZ mice, in which chronic inflammation in colon leads to adenocarcinomas, we demonstrated that CD133 is expressed on a full gamut of colonic tumor cells, which express epithelial cell adhesion molecule (EpCAM). Similarly, CD133 is widely expressed by human primary colon cancer epithelial cells, whereas the CD133- population is composed mostly of stromal and inflammatory cells. Conversely, CD133 expression does not identify the entire population of epithelial and tumor-initiating cells in human metastatic colon cancer. Indeed, both CD133+ and CD133- metastatic tumor subpopulations formed colonospheres in in vitro cultures and were capable of long-term tumorigenesis in a NOD/SCID serial xenotransplantation model. Moreover, metastatic CD133- cells form more aggressive tumors and express typical phenotypic markers of cancer-initiating cells, including CD44 (CD44+CD24-), whereas the CD133+ fraction is composed of CD44lowCD24+ cells. Collectively, our data suggest that CD133 expression is not restricted to intestinal stem or cancer-initiating cells, and during the metastatic transition, CD133+ tumor cells might give rise to the more aggressive CD133(- )subset, which is also capable of tumor initiation in NOD/SCID mice.


Asunto(s)
Antígenos CD/biosíntesis , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Regulación Neoplásica de la Expresión Génica , Glicoproteínas/biosíntesis , Células Madre/metabolismo , Antígeno AC133 , Animales , Células Epiteliales/metabolismo , Inflamación , Ratones , Ratones SCID , Ratones Transgénicos , Modelos Biológicos , Modelos Genéticos , Metástasis de la Neoplasia , Péptidos , Fenotipo , Regiones Promotoras Genéticas , Células Madre/citología
15.
Circ Res ; 105(2): 201-8, 2009 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-19542015

RESUMEN

Blood vessel formation is controlled by the balance between pro- and antiangiogenic pathways. Although much is known about the factors that drive sprouting of neovessels, the factors that stabilize and pattern neovessels are undefined. The expression of angiomodulin (AGM), a vascular endothelial growth factor (VEGF)-A binding protein, was increased in the vasculature of several human tumors as compared to normal tissue, raising the hypothesis that AGM may modulate VEGF-A-dependent vascular patterning. To elucidate the expression pattern of AGM, we developed an AGM knockin reporter mouse (AGM(lacZ/+)), with which we demonstrate that AGM is predominantly expressed in the vasculature of developing embryos and adult organs. During physiological and pathological angiogenesis, AGM is upregulated in the angiogenic vasculature. Using the zebrafish model, we found that AGM is restricted to developing vasculature by 17 to 22 hours postfertilization. Blockade of AGM activity with morpholino oligomers results in prominent angiogenesis defects in vascular sprouting and remodeling. Concurrent knockdown of both AGM and VEGF-A results in synergistic angiogenesis defects. When VEGF-A is overexpressed, the compensatory induction of the VEGF-A receptor, VEGFR2/flk-1, is blocked by the simultaneous injection of AGM morpholino oligomers. These results demonstrate that the vascular-specific marker AGM modulates vascular remodeling in part by temporizing the proangiogenic effects of VEGF-A.


Asunto(s)
Proteínas de Neoplasias/metabolismo , Neoplasias/irrigación sanguínea , Neovascularización Patológica/metabolismo , Neovascularización Fisiológica , Neovascularización Retiniana/metabolismo , Piel/irrigación sanguínea , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genotipo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Morfolinas/metabolismo , Proteínas de Neoplasias/genética , Neovascularización Patológica/genética , Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/genética , Oligonucleótidos Antisentido/metabolismo , Fenotipo , Regiones Promotoras Genéticas , Neovascularización Retiniana/genética , Neovascularización Retiniana/fisiopatología , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas , Pez Cebra/embriología , Proteínas de Pez Cebra/genética
16.
Mol Cell Biol ; 27(2): 595-604, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17101772

RESUMEN

The hyaluronan receptor LYVE-1 is expressed abundantly on the surfaces of lymphatic vessels and lymph node sinus endothelial cells from early development, where it has been suggested to function both in cell adhesion/transmigration and as a scavenger for hyaluronan turnover. To investigate the physiological role(s) of LYVE-1, we generated mice in which the gene for the receptor was inactivated by replacement with a beta-galactosidase reporter. LYVE-1(-/-) mice displayed an apparently normal phenotype, with no obvious alteration in lymphatic vessel ultrastructure or function and no apparent change in secondary lymphoid tissue structure or cellularity. In addition, the levels of hyaluronan in tissue and blood were unchanged. LYVE-1(-/-) mice also displayed normal trafficking of cutaneous CD11c(+) dendritic cells to draining lymph nodes via afferent lymphatics and normal resolution of oxazolone-induced skin inflammation. Finally, LYVE-1(-/-) mice supported normal growth of transplanted B16F10 melanomas and Lewis lung carcinomas. These results indicate that LYVE-1 is not obligatory for normal lymphatic development and function and suggest either the existence of compensatory receptors or a role more specific than that previously envisaged.


Asunto(s)
Glicoproteínas/fisiología , Ácido Hialurónico/metabolismo , Ganglios Linfáticos/fisiología , Vasos Linfáticos/fisiología , Animales , Antígeno CD11c/metabolismo , Carcinoma Pulmonar de Lewis/patología , Movimiento Celular , Células Dendríticas/fisiología , Dermatitis por Contacto/etiología , Dermatitis por Contacto/inmunología , Glicoproteínas/genética , Ácido Hialurónico/sangre , Inflamación/inducido químicamente , Inflamación/inmunología , Ganglios Linfáticos/citología , Ganglios Linfáticos/metabolismo , Vasos Linfáticos/citología , Vasos Linfáticos/metabolismo , Melanoma/patología , Proteínas de Transporte de Membrana , Ratones , Ratones Noqueados , Trasplante de Neoplasias , Oxazolona , beta-Galactosidasa/genética
17.
Nat Biotechnol ; 25(1): 91-9, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17187059

RESUMEN

A useful approach for exploring gene function involves generating mutant mice from genetically modified embryonic stem (ES) cells. Recent advances in genetic engineering of ES cells have shifted the bottleneck in this process to the generation of mice. Conventional injections of ES cells into blastocyst hosts produce F0 generation chimeras that are only partially derived from ES cells, requiring additional breeding to obtain mutant mice that can be phenotyped. The tetraploid complementation approach directly yields mice that are almost entirely derived from ES cells, but it is inefficient, works only with certain hybrid ES cell lines and suffers from nonspecific lethality and abnormalities, complicating phenotypic analyses. Here we show that laser-assisted injection of either inbred or hybrid ES cells into eight cell-stage embryos efficiently yields F0 generation mice that are fully ES cell-derived and healthy, exhibit 100% germline transmission and allow immediate phenotypic analysis, greatly accelerating gene function assignment.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/trasplante , Marcación de Gen/métodos , Terapia por Láser/métodos , Ratones Transgénicos/genética , Microinyecciones/métodos , Trasplante de Células Madre/métodos , Animales , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos/anatomía & histología , Ratones Transgénicos/cirugía , Microcirugia/métodos , Fenotipo
18.
Dev Cell ; 3(3): 411-23, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12361603

RESUMEN

VEGF and Angiopoietin-1 requisitely collaborate during blood vessel development. While Angiopoietin-1 obligately activates its Tie2 receptor, Angiopoietin-2 can activate Tie2 on some cells, while it blocks Tie2 activation on others. Our analysis of mice lacking Angiopoietin-2 reveals that Angiopoietin-2 is dispensable for embryonic vascular development but is requisite for subsequent angiogenic remodeling. Unexpectedly, mice lacking Angiopoietin-2 also exhibit major lymphatic vessel defects. Genetic rescue with Angiopoietin-1 corrects the lymphatic, but not the angiogenesis, defects, suggesting that Angiopoietin-2 acts as a Tie2 agonist in the former setting, but as an antagonist in the latter setting. Our studies define a vascular growth factor whose primary role is in postnatal angiogenic remodeling and also demonstrate that members of the VEGF and Angiopoietin families collaborate during development of the lymphatic vasculature.


Asunto(s)
Inductores de la Angiogénesis/fisiología , Tipificación del Cuerpo , Sistema Linfático/crecimiento & desarrollo , Glicoproteínas de Membrana/fisiología , Neovascularización Fisiológica/fisiología , Angiopoyetina 1 , Angiopoyetina 2 , Animales , Ascitis Quilosa/genética , Ascitis Quilosa/patología , ADN Complementario/genética , Edema/genética , Edema/patología , Ojo/irrigación sanguínea , Regulación del Desarrollo de la Expresión Génica , Marcación de Gen , Homocigoto , Sistema Linfático/patología , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Vasos Retinianos/patología
19.
Neuron ; 39(6): 919-35, 2003 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-12971893

RESUMEN

In animals with binocular vision, retinal ganglion cell (RGC) axons either cross or avoid the midline at the optic chiasm. Here, we show that ephrin-Bs in the chiasm region direct the divergence of retinal axons through the selective repulsion of a subset of RGCs that express EphB1. Ephrin-B2 is expressed at the mouse chiasm midline as the ipsilateral projection is generated and is selectively inhibitory to axons from ventrotemporal (VT) retina, where ipsilaterally projecting RGCs reside. Moreover, blocking ephrin-B2 function in vitro rescues the inhibitory effect of chiasm cells and eliminates the ipsilateral projection in the semiintact mouse visual system. A receptor for ephrin-B2, EphB1, is found exclusively in regions of retina that give rise to the ipsilateral projection. EphB1 null mice exhibit a dramatically reduced ipsilateral projection, suggesting that this receptor contributes to the formation of the ipsilateral retinal projection, most likely through its repulsive interaction with ephrin-B2.


Asunto(s)
Axones/metabolismo , Efrina-B1/deficiencia , Efrina-B2/biosíntesis , Quiasma Óptico/metabolismo , Retina/metabolismo , Animales , Células Cultivadas , Efrina-B1/biosíntesis , Efrina-B1/genética , Efrina-B2/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Quiasma Óptico/embriología , Embarazo , Retina/embriología , Células Ganglionares de la Retina/metabolismo , Xenopus
20.
J Neurosci ; 26(4): 1179-89, 2006 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-16436605

RESUMEN

Peripheral myelin protein 22 (PMP22) is a tetraspan membrane glycoprotein, the misexpression of which is associated with hereditary demyelinating neuropathies. Myelinating Schwann cells (SCs) produce the highest levels of PMP22, yet the function of the protein in peripheral nerve biology is unresolved. To investigate the potential roles of PMP22, we engineered a novel knock-out (-/-) mouse line by replacing the first two coding exons of pmp22 with the lacZ reporter. PMP22-deficient mice show strong beta-galactosidase reactivity in peripheral nerves, cartilage, intestines, and lungs, whereas phenotypically they display the characteristics of tomaculous neuropathy. In the absence of PMP22, myelination of peripheral nerves is delayed, and numerous axon-SC profiles show loose basal lamina, suggesting altered interactions of the glial cells with the extracellular matrix. The levels of beta4 integrin, a molecule involved in the linkage between SCs and the basal lamina, are severely reduced in nerves of PMP22-deficient mice. During early stages of myelination, PMP22 and beta4 integrin are coexpressed at the cell surface and can be coimmunoprecipitated together with laminin and alpha6 integrin. In agreement, in clone A colonic carcinoma cells, epitope-tagged PMP22 forms a complex with beta4 integrin. Together, these data indicate that PMP22 is a binding partner in the integrin/laminin complex and is involved in mediating the interaction of SCs with the extracellular environment.


Asunto(s)
Integrina alfa6beta4/metabolismo , Proteínas de la Mielina/metabolismo , Células de Schwann/metabolismo , Adenocarcinoma/química , Adenocarcinoma/patología , Animales , Membrana Basal/metabolismo , Membrana Basal/ultraestructura , Sitios de Unión , Línea Celular Tumoral/química , Técnicas de Cocultivo , Neoplasias del Colon/química , Neoplasias del Colon/patología , Exones/genética , Ganglios Espinales/citología , Humanos , Integrina alfa6beta4/química , Operón Lac , Laminina/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Complejos Multiproteicos , Proteínas de la Mielina/química , Proteínas de la Mielina/deficiencia , Proteínas de la Mielina/genética , Proteínas de la Mielina/fisiología , Especificidad de Órganos , Enfermedades del Sistema Nervioso Periférico/genética , Enfermedades del Sistema Nervioso Periférico/metabolismo , Enfermedades del Sistema Nervioso Periférico/patología , Fenotipo , Unión Proteica , Mapeo de Interacción de Proteínas , Ratas , Células de Schwann/ultraestructura , Nervio Ciático/metabolismo , Nervio Ciático/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA