Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oncogene ; 36(1): 1-12, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-27270432

RESUMEN

The inadequate transport of drugs into the tumor tissue caused by its abnormal vasculature is a major obstacle to the treatment of cancer. Anti-vascular endothelial growth factor (anti-VEGF) drugs can cause phenotypic alteration and maturation of the tumor's vasculature. However, whether this consistently improves delivery and subsequent response to therapy is still controversial. Clinical results indicate that not all patients benefit from antiangiogenic treatment, necessitating the development of criteria to predict the effect of these agents in individual tumors. We demonstrate that, in anti-VEGF-refractory murine tumors, vascular changes after VEGF ablation result in reduced delivery leading to therapeutic failure. In these tumors, the impaired response after anti-VEGF treatment is directly linked to strong deposition of fibrillar extracellular matrix (ECM) components and high expression of lysyl oxidases. The resulting condensed, highly crosslinked ECM impeded drug permeation, protecting tumor cells from exposure to small-molecule drugs. The reduced vascular density after anti-VEGF treatment further decreased delivery in these tumors, an effect not compensated by the improved vessel quality. Pharmacological inhibition of lysyl oxidases improved drug delivery in various tumor models and reversed the negative effect of VEGF ablation on drug delivery and therapeutic response in anti-VEGF-resistant tumors. In conclusion, the vascular changes after anti-VEGF therapy can have a context-dependent negative impact on overall therapeutic efficacy. A determining factor is the tumor ECM, which strongly influences the effect of anti-VEGF therapy. Our results reveal the prospect to revert a possible negative effect and to potentiate responsiveness to antiangiogenic therapy by concomitantly targeting ECM-modifying enzymes.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Matriz Extracelular/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Femenino , Humanos , Ratones , Modelos Biológicos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Permeabilidad , Proteína-Lisina 6-Oxidasa/metabolismo , Sarcoma/tratamiento farmacológico , Sarcoma/metabolismo , Sarcoma/patología , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Cytotherapy ; 9(1): 23-34, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17361485

RESUMEN

BACKGROUND: We have previously shown that xenogenic DNA vaccines encoding rat neu and melanosomal differentiation Ag induce tumor immunity. Others have developed vaccines targeting tumor neovasculature. Tumor endothelial marker 8 (TEM8) is expressed in the neovasculature of human tumors, and in the mouse melanoma B16, but its expression is limited in normal adult tissues. We describe a DNA vaccine combining xenogeneic tumor Ag and TEM8. METHODS: In-situ hybridization was used to detect TEM8 RNA in mouse tumors. Mice transgenic for the rat neu proto-oncogene were immunized with DNA vaccines encoding TEM8 and the extracellular domain of rat neu and challenged with the 233-VSGA1 breast cancer cell line. In parallel experiments, C57BL/6 mice were immunized with TEM8 and human tyrosinase-related protein 1 (hTYRP1/hgp75) and challenged with B16F10 melanoma. RESULTS: TEM8 was expressed in the stroma of transplantable mouse breast and melanoma tumors. In both model systems, TEM8 DNA had no activity as a single agent but significantly enhanced the anit-tumor immunity of neu and hTYRP1/hgp75 DNA vaccines when given in concert. The observed synergy was dependent upon CD8+ T cells, as depletion of this cell population just prior to tumor challenge obviated the effect of the TEM8 vaccine in both tumor models. DISCUSSION: A local immune responses to TEM8 may increase inflammation or tumor necrosis within the tumor, resulting in improved Ag presentation of HER2/neu and hTYRP1/hgp75. Alternatively, TEM8 expression in host APC may act more as an adjuvant than an immunologic target.


Asunto(s)
Antígenos de Diferenciación/inmunología , Vacunas contra el Cáncer/inmunología , Tolerancia Inmunológica/inmunología , Proteínas de la Membrana/inmunología , Proteínas de Neoplasias/inmunología , Receptores de Superficie Celular/inmunología , Animales , Western Blotting , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/genética , Diferenciación Celular/inmunología , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Tolerancia Inmunológica/genética , Inmunización , Hibridación in Situ , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/inmunología , Neoplasias Mamarias Animales/patología , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Glicoproteínas de Membrana/inmunología , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos , Proteínas de Neoplasias/genética , Oxidorreductasas/inmunología , Proto-Oncogenes Mas , Ratas , Receptores de Superficie Celular/genética , Proteínas Recombinantes/inmunología , Análisis de Supervivencia
3.
Cytotherapy ; 9(1): 23-34, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18236207

RESUMEN

BACKGROUND: We have previously shown that xenogeneic DNA vaccines encoding rat neu and melanosomal differentiation Ag induce tumor immunity. Others have developed vaccines targeting tumor neovasculature. Tumor endothelial marker 8 (TEM8) is expressed in the neovasculature of human tumors, and in the mouse melanoma B16, but its expression is limited in normal adult tissues. We describe a DNA vaccine combining xenogeneic tumor Ag and TEM8. METHODS: In-situ hybridization was used to detect TEM8 RNA in mouse tumors. Mice transgenic for the rat neu proto-oncogene were immunized with DNA vaccines encoding TEM8 and the extracellular domain of rat neu and challenged with the 233-VSGA1 breast cancer cell line. In parallel experiments, C57BL/6 mice were immunized with TEM8 and human tyrosinase-related protein 1 (hTYRP1/hgp75) and challenged with B16F10 melanoma. RESULTS: TEM8 was expressed in the stroma of transplantable mouse breast and melanoma tumors. In both model systems, TEM8 DNA had no activity as a single agent but significantly enhanced the anti-tumor immunity of neu and hTYRP1/hgp75 DNA vaccines when given in concert. The observed synergy was dependent upon CD8+ T cells, as depletion of this cell population just prior to tumor challenge obviated the effect of the TEM8 vaccine in both tumor models. DISCUSSION: A local immune response to TEM8 may increase inflammation or tumor necrosis within the tumor, resulting in improved Ag presentation of HER2/neu and hTYRP1/hgp75. Alternatively, TEM8 expression in host APC may alter T-cell interactions or homing. In this way, TEM8 may act more as an adjuvant than an immunologic target.


Asunto(s)
Antígenos de Diferenciación/inmunología , Vacunas contra el Cáncer/inmunología , Neoplasias/inmunología , Receptores de Superficie Celular/inmunología , Vacunación , Animales , Anticuerpos Antineoplásicos/inmunología , Linfocitos T CD8-positivos/inmunología , ADN de Neoplasias/inmunología , Femenino , Humanos , Neoplasias Mamarias Animales/inmunología , Neoplasias Mamarias Animales/patología , Melanoma/inmunología , Melanoma/patología , Glicoproteínas de Membrana/inmunología , Ratones , Oxidorreductasas/inmunología , Proto-Oncogenes Mas , Ratas , Receptor ErbB-2/inmunología , Vacunas de ADN/inmunología
4.
Mol Cell Biol ; 25(10): 4105-16, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15870281

RESUMEN

The docking protein FRS2alpha is a major mediator of fibroblast growth factor (FGF) signaling. However, the physiological role of FRS2alpha in vivo remains unknown. In this report, we show that Frs2alpha-null mouse embryos have a defect in anterior-posterior (A-P) axis formation and are developmentally retarded, resulting in embryonic lethality by embryonic day 8. We demonstrate that FRS2alpha is essential for the maintenance of self-renewing trophoblast stem (TS) cells in response to FGF4 in the extraembryonic ectoderm (ExE) that gives rise to tissues of the placenta. By analyzing chimeric embryos, we found that FRS2alpha also plays a role in cell movement through the primitive streak during gastrulation. In addition, experiments are presented demonstrating that Bmp4 expression in TS cells is controlled by mitogen-activated protein kinase-dependent FGF4 stimulation. Moreover, both the expression of Bmp4 in ExE and activation of Smad1/5 in epiblasts are reduced in Frs2alpha-null embryos. These experiments underscore the critical role of FRS2alpha in mediating multiple processes during embryonic development and reveal a potential new link between FGF and Bmp4 signaling pathways in early embryogenesis.


Asunto(s)
Embrión de Mamíferos/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/farmacología , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Tipificación del Cuerpo , Proteína Morfogenética Ósea 4 , Proteínas Morfogenéticas Óseas/deficiencia , Proteínas Morfogenéticas Óseas/genética , Movimiento Celular , Supervivencia Celular , Quimera/anomalías , Quimera/embriología , Quimera/genética , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor 4 de Crecimiento de Fibroblastos , Gástrula/efectos de los fármacos , Gástrula/patología , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteína Nodal , Regiones Promotoras Genéticas/genética , Proteínas Smad , Proteína Smad1 , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta/genética
5.
Nat Med ; 7(11): 1194-201, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11689883

RESUMEN

The role of bone marrow (BM)-derived precursor cells in tumor angiogenesis is not known. We demonstrate here that tumor angiogenesis is associated with recruitment of hematopoietic and circulating endothelial precursor cells (CEPs). We used the angiogenic defective, tumor resistant Id-mutant mice to show that transplantation of wild-type BM or vascular endothelial growth factor (VEGF)-mobilized stem cells restore tumor angiogenesis and growth. We detected donor-derived CEPs throughout the neovessels of tumors and Matrigel-plugs in an Id1+/-Id3-/- host, which were associated with VEGF-receptor-1-positive (VEGFR1+) myeloid cells. The angiogenic defect in Id-mutant mice was due to impaired VEGF-driven mobilization of VEGFR2+ CEPs and impaired proliferation and incorporation of VEGFR1+ cells. Although targeting of either VEGFR1 or VEGFR2 alone partially blocks the growth of tumors, inhibition of both VEGFR1 and VEGFR2 was necessary to completely ablate tumor growth. These data demonstrate that recruitment of VEGF-responsive BM-derived precursors is necessary and sufficient for tumor angiogenesis and suggest new clinical strategies to block tumor growth.


Asunto(s)
Células Madre Hematopoyéticas/patología , Proteínas de Neoplasias , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/patología , Neovascularización Patológica , Proteínas Represoras , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Endotelio Vascular/patología , Trasplante de Células Madre Hematopoyéticas , Proteína 1 Inhibidora de la Diferenciación , Proteínas Inhibidoras de la Diferenciación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Mutación , Neovascularización Patológica/genética , Pruebas de Neutralización , Proteínas Proto-Oncogénicas/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores de Factores de Crecimiento/fisiología , Receptores de Factores de Crecimiento Endotelial Vascular , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Receptor 1 de Factores de Crecimiento Endotelial Vascular
6.
Nat Genet ; 27(4): 412-6, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11279523

RESUMEN

Fate-mapping experiments in the mouse have revealed that the primitive streak can be divided into three functional regions: the proximal region gives rise to germ cells and the extra-embryonic mesoderm of the yolk sac; the distal region generates cardiac mesoderm and node-derived axial mesendoderm; and the middle streak region produces the paraxial, intermediate and lateral plate mesoderm of the trunk. To gain insight into the mechanisms that mediate the assembly of the primitive streak into these functional regions, we have cloned and functionally identified the gene disrupted in the amnionless (amn) mouse, which has a recessive, embryonic lethal mutation that interferes specifically with the formation and/or specification of the middle primitive streak region during gastrulation. Here we report that the gene Amn encodes a novel type I transmembrane protein that is expressed exclusively in the extra-embryonic visceral endoderm layer during gastrulation. The extracellular region of the Amn protein contains a cysteine-rich domain with similarity to bone morphogenetic protein (BMP)-binding cysteine-rich domains in chordin, its Drosophila melanogaster homolog (Short gastrulation) and procollagen IIA (ref. 3). Our findings indicate that Amn may direct the production of trunk mesoderm derived from the middle streak by acting in the underlying visceral endoderm to modulate a BMP signaling pathway.


Asunto(s)
Cisteína/metabolismo , Endodermo/metabolismo , Gástrula/citología , Proteínas de la Membrana/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Cartilla de ADN , Inmunohistoquímica , Hibridación in Situ , Proteínas de la Membrana/química , Ratones , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido
7.
Cancer Res ; 61(5): 1941-7, 2001 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11280750

RESUMEN

IgG polyclonal antiserum was generated in New Zealand White rabbits immunized with a 16-mer peptide consisting of a specific amino acid sequence at residues corresponding to the sixth to seventh predicted transmembrane domain of the human reduced folate carrier (RFC). Using Western immunoblotting to examine the cytosolic and membrane fractions of the human CCRF-CEM T-cell lymphoblastic leukemia cell line, polyclonal antihuman RFC antiserum recognized two bands in the cytosolic fraction (approximately 60 kDa and approximately 70 kDa) on 10% polyacrylamide gels. In the membrane fraction, an approximately 60-kDa protein was identified. Comparative studies of a panel of human tumor cell lines including the HT1080 fibrosarcoma, 8805 malignant fibrous histiocytoma, and the MCF breast cancer cell lines revealed similar findings. Likewise, a recombinant approximately 60-kDa membrane protein was identified after expression of baculovirus-infected Sf9 insect cells containing cDNA of the human RFC. In the CEM-7A cell line, a variant of the CCRF-CEM cell line that overexpresses the RFC, 21-fold overexpression of the approximately 60-kDa membrane protein (RFC) was shown by Western analysis. To characterize further the cellular distribution of the human RFC, immunohistochemical analyses were performed in CCRF-CEM T-cell lymphoblastic leukemia cells. Predominantly membrane localization of the antibody reacting sites was detected; however, a cytoplasmic component was noted as well. By confocal microscopy and by immunogold electron microscopy, the cytoplasmic expression was found to be largely of mitochondrial origin. These findings were corroborated by Western immunoblotting of mitochondrial membrane isolates from the CCRF-CEM cell line, which demonstrate an approximately 60-kDa protein. The localization of the human RFC to the mitochondrial membrane is a novel finding, and it suggests a role for the mitochondrial membrane in the transport of folates.


Asunto(s)
Proteínas Portadoras/metabolismo , Leucemia de Células T/metabolismo , Proteínas de la Membrana , Proteínas de Transporte de Membrana , Mitocondrias/metabolismo , Animales , Antígenos/inmunología , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/inmunología , Membrana Celular/metabolismo , Citosol/metabolismo , Humanos , Inmunoglobulina G/inmunología , Inmunohistoquímica , Microscopía Electrónica , Microscopía Fluorescente , Péptidos/inmunología , Conejos , Proteína Portadora de Folato Reducido , Células Tumorales Cultivadas
8.
Mol Cell ; 6(5): 989-98, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11106739

RESUMEN

Spo11, a protein first identified in yeast, is thought to generate the chromosome breaks that initiate meiotic recombination. We now report that disruption of mouse Spo11 leads to severe gonadal abnormalities from defective meiosis. Spermatocytes suffer apoptotic death during early prophase; oocytes reach the diplotene/dictyate stage in nearly normal numbers, but most die soon after birth. Consistent with a conserved function in initiating meiotic recombination, Dmc1/Rad51 focus formation is abolished. Spo11(-/-) meiocytes also display homologous chromosome synapsis defects, similar to fungi but distinct from flies and nematodes. We propose that recombination initiation precedes and is required for normal synapsis in mammals. Our results also support the view that mammalian checkpoint responses to meiotic recombination and/or synapsis defects are sexually dimorphic.


Asunto(s)
Proteínas de Ciclo Celular , Emparejamiento Cromosómico , Esterasas/deficiencia , Eliminación de Gen , Meiosis/genética , Proteínas , Caracteres Sexuales , Adenosina Trifosfatasas/metabolismo , Animales , Apoptosis , Cromosomas/ultraestructura , Proteínas de Unión al ADN/metabolismo , Endodesoxirribonucleasas , Esterasas/genética , Esterasas/metabolismo , Femenino , Gónadas/anomalías , Gónadas/citología , Gónadas/metabolismo , Gónadas/patología , Masculino , Ratones , Ratones Noqueados , Insuficiencia Ovárica Primaria/metabolismo , Insuficiencia Ovárica Primaria/patología , Recombinasa Rad51 , Recombinación Genética , Homología de Secuencia de Ácido Nucleico , Espermatocitos/citología , Espermatocitos/metabolismo , Espermatocitos/ultraestructura
9.
EMBO J ; 19(6): 1312-26, 2000 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-10716931

RESUMEN

The Kit receptor tyrosine kinase functions in hemato- poiesis, melanogenesis and gametogenesis. Kit receptor-mediated cellular responses include proliferation, survival, adhesion, secretion and differentiation. In mast cells, Kit-mediated recruitment and activation of phosphatidylinositol 3'-kinase (PI 3-kinase) produces phosphatidylinositol 3'-phosphates, plays a critical role in mediating cell adhesion and secretion and has contributory roles in mediating cell survival and proliferation. To investigate the consequences in vivo of blocking Kit-mediated PI 3-kinase activation we have mutated the binding site for the p85 subunit of PI 3-kinase in the Kit gene, using a knock-in strategy. Mutant mice have no pigment deficiency or impairment of steady-state hematopoiesis. However, gametogenesis is affected in several ways and tissue mast cell numbers are affected differentially. While primordial germ cells during embryonic development are not affected, Kit(Y719F)/Kit(Y719F) males are sterile due to a block at the premeiotic stages in spermatogenesis. Furthermore, adult males develop Leydig cell hyperplasia. The Leydig cell hyperplasia implies a role for Kit in Leydig cell differentiation and/or steroidogenesis. In mutant females follicle development is impaired at the cuboidal stages resulting in reduced fertility. Also, adult mutant females develop ovarian cysts and ovarian tubular hyperplasia. Therefore, a block in Kit receptor-mediated PI 3-kinase signaling may be compensated for in hematopoiesis, melanogenesis and primordial germ cell development, but is critical in spermatogenesis and oogenesis.


Asunto(s)
Oogénesis/genética , Mutación Puntual/genética , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transducción de Señal , Espermatogénesis/genética , Animales , Recuento de Células , Células Cultivadas , Femenino , Células Germinativas/citología , Hematopoyesis/genética , Hiperplasia , Infertilidad/genética , Células Intersticiales del Testículo/citología , Masculino , Mastocitos/citología , Mastocitos/enzimología , Meiosis/genética , Melanocitos/citología , Ratones , Ratones Endogámicos , Folículo Ovárico/crecimiento & desarrollo , Folículo Ovárico/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Pigmentación/genética , Proteínas Proto-Oncogénicas c-kit/química
10.
Genomics ; 70(3): 364-74, 2000 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11161787

RESUMEN

Megabase-scale mapping of the PML gene locus revealed the presence of a large-scale insertion-deletion polymorphism located 25 kb downstream of the PML gene. The polymorphism is organized as a head-to-tail tandem 25-kb repeat containing one to five units. Characterization of the first repeat unit downstream of PML revealed the presence of a gene with strong homology to a family of Golgin-related proteins. The gene, designated GLP (for Golgin linked to PML), is strongly expressed as a 6-kb transcript in normal human testis. In situ hybridization of normal human testis demonstrated that the expression of GLP was restricted to late meiotic germ cells. There was weak expression in late pachytene spermatocytes and strong expression in spermatids. GLP is 50% homologous to other Golgin-related proteins including the vesicle docking protein GM130. Southern blot hybridization of genomic DNA with a GLP probe demonstrated numerous homologous bands outside the PML locus. Three of these loci have been mapped by fluorescence in situ hybridization to chromosome loci 9q34.1, 15q11-q13, and 15q22-q24. Hybridization of a GLP cDNA probe to a zoo blot demonstrated multiple signals in nonhuman primates but not in other species and suggested the duplication of an ancestral locus around 20 million years ago.


Asunto(s)
Ligamiento Genético , Proteínas de Neoplasias/genética , Proteínas Nucleares , Polimorfismo Genético , Proteínas/genética , Factores de Transcripción/genética , Secuencia de Aminoácidos , Autoantígenos , Secuencia de Bases , Mapeo Cromosómico , Cromosomas Humanos Par 15 , Clonación Molecular , ADN Complementario , Electroforesis en Gel de Campo Pulsado , Exones , Proteínas de la Matriz de Golgi , Humanos , Hibridación in Situ , Intrones , Datos de Secuencia Molecular , Proteína de la Leucemia Promielocítica , Proteínas/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Proteínas Supresoras de Tumor
11.
Nature ; 401(6754): 670-7, 1999 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-10537105

RESUMEN

Id proteins may control cell differentiation by interfering with DNA binding of transcription factors. Here we show that targeted disruption of the dominant negative helix-loop-helix proteins Id1 and Id3 in mice results in premature withdrawal of neuroblasts from the cell cycle and expression of neural-specific differentiation markers. The Id1-Id3 double knockout mice also display vascular malformations in the forebrain and an absence of branching and sprouting of blood vessels into the neuroectoderm. As angiogenesis both in the brain and in tumours requires invasion of avascular tissue by endothelial cells, we examined the Id knockout mice for their ability to support the growth of tumour xenografts. Three different tumours failed to grow and/or metastasize in Id1+/- Id3-/- mice, and any tumour growth present showed poor vascularization and extensive necrosis. Thus, the Id genes are required to maintain the timing of neuronal differentiation in the embryo and invasiveness of the vasculature. Because the Id genes are expressed at very low levels in adults, they make attractive new targets for anti-angiogenic drug design.


Asunto(s)
Secuencias Hélice-Asa-Hélice , Neoplasias Experimentales/irrigación sanguínea , Neovascularización Fisiológica/fisiología , Neuronas/citología , Proteínas Represoras , Factores de Transcripción/fisiología , Animales , Antígenos CD/biosíntesis , Encéfalo/irrigación sanguínea , Encéfalo/embriología , Encéfalo/patología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proteína 1 Inhibidora de la Diferenciación , Integrina alfa5 , Metaloproteinasa 2 de la Matriz/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Trasplante de Neoplasias , Neovascularización Fisiológica/genética , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Factores de Transcripción/genética , Trasplante Heterólogo , Células Tumorales Cultivadas
12.
Genomics ; 61(2): 170-82, 1999 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-10534402

RESUMEN

The Saccharomyces cerevisiae Spo11 protein is thought to catalyze formation of the DNA double-strand breaks that initiate meiotic recombination. We have cloned cDNA and genomic DNA for a mouse gene encoding a protein with significant sequence similarity to conserved domains found in proteins of the Spo11p family. This putative mouse Spo11 gene maps to the distal region of chromosome 2 (homologous to human chromosome 20q13.2-q13.3) and comprises at least 12 exons, spanning approximately 15-18 kb. Strong expression of the Spo11 message is seen in juvenile and adult testis by RNA in situ hybridization, RT-PCR, and Northern blot, with much weaker expression in thymus and brain. In situ hybridization detects expression in oocytes of embryonic ovary, but not of adult ovary. RT-PCR and in situ hybridization analyses of a time course of juvenile testis development indicate that Spo11 expression begins in early meiotic Prophase I, prior to the pachytene stage, with increasing accumulation of mRNA through the pachytene stage. Taken together, these results strongly suggest that this gene encodes the functional homolog of yeast Spo11p, which in turn suggests that the mechanism of meiotic recombination initiation is conserved between yeast and mammals.


Asunto(s)
Esterasas/genética , Meiosis/genética , Recombinación Genética , Saccharomyces cerevisiae/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Mapeo Cromosómico , Clonación Molecular , Secuencia Conservada , ADN-Topoisomerasas de Tipo I/genética , Endodesoxirribonucleasas , Femenino , Células Germinativas/metabolismo , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Oocitos/metabolismo , Ovario/citología , Ovario/embriología , Ovario/metabolismo , Polimorfismo de Longitud del Fragmento de Restricción , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Testículo/citología , Testículo/metabolismo
13.
Dev Biol ; 204(1): 34-54, 1998 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-9851841

RESUMEN

The primitive streak is the defining feature of the gastrulating mouse embryo. Currently, little is known in the mouse about the mechanisms that mediate the assembly of the primitive streak or about the signaling pathways that specify the different types of mesoderm and endoderm generated from the streak. To gain insight into primitive streak assembly and function, we have conducted a detailed phenotypic characterization of amnionless, a transgene-induced insertional mouse mutation that arrests embryonic development during gastrulation. Our histological and molecular analyses, when examined in the context of the mouse gastrula fate map, lead to the model that middle streak formation is specifically impaired in the amnionless mutant. Significantly, these observations argue that the formation of the middle streak is mediated by a pathway that is genetically separable from those that direct the specification of the distal and proximal streak regions. Intriguingly, our findings from wt ES cell left and right arrow amnionless-/- blastocyst chimeras indicate that this proposed separate pathway for middle streak formation is dependent on amnionless gene functions in the visceral endoderm.


Asunto(s)
Desarrollo Embrionario y Fetal/genética , Gástrula/fisiología , Regulación del Desarrollo de la Expresión Génica , Proteínas de la Membrana/genética , Animales , Diferenciación Celular/genética , Ratones , Ratones Transgénicos , Mutación
14.
Dev Dyn ; 213(3): 293-308, 1998 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9825865

RESUMEN

The pattern of apoptotic cell death has been surveyed in prestreak and primitive streak embryos of four strains of mice and in three mutants affecting gastrulation. In C57BL/6 embryos, a high level of cell death occurs in the early egg cylinder stage at embryonic day 5 (E5) to E5.5. In all strains, cell death is elevated shortly before gastrulation, but the level varies four- to fivefold among strains. During gastrulation, cell death declines but is relatively more abundant in the distal and distal anterior regions. Early streak embryos cultured in media with reduced levels of growth factors show increased cell death mainly in the distal region. In three mutants with disturbed function of the proximal visceral endoderm and/or primitive streak, cell death is increased, and the regional pattern seen in normal embryos is intensified. The results strongly suggest that the proximal visceral endoderm and primitive streak region are the principal sites of synthesis of growth factors promoting cell survival. We conclude that localized growth factor supply has an important role in regulating the size of the embryo and of embryonic regions.


Asunto(s)
Apoptosis/fisiología , Desarrollo Embrionario y Fetal , Gástrula/citología , Animales , Apoptosis/efectos de los fármacos , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , División Celular/efectos de los fármacos , Proteínas de Unión al ADN/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Sustancias de Crecimiento/farmacología , Factor Nuclear 4 del Hepatocito , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Mutantes , Mutación/fisiología , Fosfoproteínas/genética , Embarazo , Factores de Tiempo , Factores de Transcripción/genética
15.
Proc Natl Acad Sci U S A ; 95(23): 13709-13, 1998 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-9811865

RESUMEN

RNA helicase A (RHA) is the human homologue of the Drosophila maleless protein, an essential factor for the development of male flies. Recently, it was shown that RHA cooperates with the cAMP-responsive element in mediating the cAMP-dependent transcriptional activation of a number of genes. Due to the participation of cAMP as a second messenger in a number of signaling pathways, we examined the function of RHA during mammalian embryogenesis. To examine the role(s) of RHA in mammalian development, RHA knockout mice were generated by homologous recombination. Homozygosity for the mutant RHA allele led to early embryonic lethality. Histological analysis, combined with terminal deoxynucleotidyltransferase-mediated UTP end labeling (TUNEL) reactions of RHA-null embryos, revealed marked apoptotic cell death specifically in embryonic ectodermal cells during gastrulation. RNA in situ analyses of the expression of HNF-3beta and Brachyury, two molecular markers for gastrulation, showed that RHA-null embryos at days 7.5 and 8.5 expressed both HNF-3beta and Brachyury in a pattern similar to those of pre- and early streak stages of embryos, respectively. These observations indicate that RHA is necessary for early embryonic development and suggest the requirement of RHA for the survival and differentiation of embryonic ectoderm.


Asunto(s)
Desarrollo Embrionario y Fetal/genética , Gástrula , ARN Helicasas/genética , Animales , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Noqueados , Mutación , Recombinación Genética
16.
Cell Growth Differ ; 9(9): 787-94, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9751122

RESUMEN

The involvement of cyclin-dependent kinase inhibitors in differentiation remains unclear: are the roles of cyclin-dependent kinase inhibitors restricted to cell cycle arrest; or also required for completion of the differentiation program; or both? Here, we report that differentiation of luteal cells can be uncoupled from growth arrest in p27-deficient mice. In these mice, female-specific infertility correlates with a failure of embryos to implant at embryonic day 4.5. We show by ovarian transplant and hormone reconstitution experiments that failure to regulate luteal cell estradiol is one physiological mechanism for infertility in these mice. This failure is not due to a failure of p27-deficient granulosa cells to differentiate after hormonal stimulation; P450scc, a marker for luteal progesterone biosynthesis, is expressed and granulosa cell-specific cyclin D2 expression is reduced. However, unlike their wild-type counterparts, p27-deficient luteal cells continue to proliferate for up to 3.5 days after hormonal stimulation. By day 5.5, however, these cells withdraw from the cell cycle, suggesting that p27 plays a role in the early events regulating withdrawal of cells from the cell cycle. We have further shown that in the absence of this timely withdrawal, estradiol regulation is perturbed, explaining in part how fertility is compromised at the level of implantation. These data support the interpretation of our previous observations on oligodendrocyte differentiation about a role for p27 in establishing the nonproliferative state, which in some cases (oligodendrocytes) is required for differentiation, whereas in other cases it is required for the proper functioning of a differentiated cell (luteal cell).


Asunto(s)
Proteínas de Ciclo Celular , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Fase G1 , Células de la Granulosa/citología , Células Lúteas/citología , Proteínas Asociadas a Microtúbulos/fisiología , Proteínas Supresoras de Tumor , Animales , Ciclo Celular/fisiología , Diferenciación Celular/fisiología , División Celular/fisiología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Femenino , Hormona Folículo Estimulante/farmacología , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Infertilidad/genética , Infertilidad/fisiopatología , Células Lúteas/efectos de los fármacos , Células Lúteas/metabolismo , Masculino , Ratones , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/genética , Ovariectomía , Ovario/citología , Ovario/efectos de los fármacos , Ovario/metabolismo , Embarazo
17.
Proc Natl Acad Sci U S A ; 95(13): 7485-90, 1998 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-9636176

RESUMEN

The genetic pathways that control development of the early mammalian embryo have remained poorly understood, in part because the systematic mutant screens that have been so successful in the identification of genes and pathways that direct embryonic development in Drosophila, Caenorhabditis elegans, and zebrafish have not been applied to mammalian embryogenesis. Here we demonstrate that chemical mutagenesis with ethylnitrosourea can be combined with the resources of mouse genomics to identify new genes that are essential for mammalian embryogenesis. A pilot screen for abnormal morphological phenotypes of midgestation embryos identified five mutant lines; the phenotypes of four of the lines are caused by recessive traits that map to single regions of the genome. Three mutant lines display defects in neural tube closure: one is caused by an allele of the open brain (opb) locus, one defines a previously unknown locus, and one has a complex genetic basis. Two mutations produce novel early phenotypes and map to regions of the genome not previously implicated in embryonic patterning.


Asunto(s)
Embrión de Mamíferos/anomalías , Genes Letales , Pruebas Genéticas/métodos , Mutación , Animales , Mapeo Cromosómico , Femenino , Genes Recesivos , Marcadores Genéticos , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Defectos del Tubo Neural/diagnóstico , Defectos del Tubo Neural/genética , Fenotipo
18.
Appl Environ Microbiol ; 63(9): 3707-10, 1997 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9293022

RESUMEN

The linB gene product (LinB), 1,3,4,6-tetrachloro-1,4-cyclohexadiene halidohydrolase, which is involved in the degradation of gamma-hexachlorocyclohexane in Sphingomonas paucimobilis UT26 (Y. Nagata, T. Nariya, R. Ohtomo, M. Fukuda, K. Yano, and M. Takagi, J. Bacteriol. 175:6403-6410, 1993), was overproduced in E. coli and purified to homogeneity. The molecular mass of LinB was deduced to be 30 kDa by gel filtration chromatography and 32 kDa by electrophoresis on sodium dodecyl sulfate-polyacrylamide gel, indicating that LiuB is a monomeric enzyme. The optimal pH for activity was 8.2. Not only monochloroalkanes (C3 to C10) but also dichloroalkanes, bromoalkanes, and chlorinated allphatic alcohols were good substrates for LinB, suggesting that LinB shares properties with another haloalkane dehalogenase, DhlA (S. Keuning, D.B. Janssen, and B. Witholt, J. Bacteriol. 163:635-639, 1985), which shows significant similarity to LinB in primary structure (D. B. Janssen, F. Pries, J. van der Ploeg, B. Kazemier, P. Terpstra, and B. Witholt, J. Bacteriol. 171:6791-6799, 1989) but not in substrate specificity. Principal component analysis of substrate activities of various haloalkane dehalogenases suggested that LinB probably constitutes a new substrate specificity class within this group of enzymes.


Asunto(s)
Bacterias Aerobias Gramnegativas/enzimología , Bacterias Aerobias Gramnegativas/metabolismo , Hexaclorociclohexano/metabolismo , Hidrolasas/aislamiento & purificación , Biodegradación Ambiental , Escherichia coli/genética , Genes Bacterianos , Bacterias Aerobias Gramnegativas/genética , Concentración de Iones de Hidrógeno , Hidrolasas/genética , Hidrolasas/metabolismo , Cinética , Peso Molecular , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
19.
Dev Dyn ; 208(1): 92-106, 1997 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8989524

RESUMEN

We have performed a detailed comparative in situ hybridization analysis to examine the patterns of expression of all the members of the Id gene family (Id1-4) during murine gastrulation and neurogenesis. During gastrulation, both Id1 and Id3 are expressed in the tissues derived from the inner cell mass from 5.5 dpc onward, whereas Id2 is expressed in tissues derived from trophoblasts. Id4 expression is absent during this period of development. Embryonic Id1 messages are detected during gastrulation on the proximal side of the embryonic ectoderm, which is the border between the embryo proper and the extraembryonic tissues, and the expression of Id3 is found throughout the entire embryo proper. This unique pattern of expression of the different members of the Id family suggests a nonredundant role for these genes in antagonizing the activity of bHLH transcription factors during very early mouse development. During neurogenesis, the expression of each member of the Id family is present in an unique pattern along the dorsal-ventral axis of the neural tube: In the early stages of spinal cord development, both Id1 and Id2 are expressed in the roof plate, whereas Id3 is expressed both in the roof and the floor plates. As development progresses, the expression of both Id1 and Id3 is detected in the dividing neuroblasts, whereas Id2 and 4 are expressed in presumptive neurons which are undergoing maturation. The expression patterns of all the members of the Id gene family persist throughout the entire CNS, both in the spinal cord and in the brain. In addition, the characteristic expression of Id2 and Id4 in more mature neurons is reiterated both in the PNS and in the neurons of some of the sensory organs. These data suggest that the expression of different subgroups of the Id gene family may have different physiological consequences and thereby contributes in unique ways to specify the differentiation state of neuronal cells during development.


Asunto(s)
Sistema Nervioso Central/embriología , Gástrula , Regulación del Desarrollo de la Expresión Génica , Proteínas Represoras , Factores de Transcripción/genética , Animales , Oído Interno/embriología , Ectodermo/química , Desarrollo Embrionario y Fetal/genética , Ojo/embriología , Proteína 1 Inhibidora de la Diferenciación , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Nariz/embriología , Especificidad de Órganos , Sistema Nervioso Periférico/embriología , ARN Mensajero/análisis
20.
Dev Dyn ; 207(3): 235-52, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8922523

RESUMEN

The murine dominant negative helix-loop-helix (dnHLH) proteins inhibit the activities of bHLH transcription factors in diverse cell lineages (Benezra et al. [1990] Cell 61:49-59; Christy et al [1991] Proc. Natl. Acad. Sci. U.S.A. 88:1815-1819; Sun et al [1991] Mol. Cell Biol. 11: 5603-5611; Riechmann et al. [1994] Nucleic Acids Res. 22:749-755). Currently, there are four members in the dnHLH family, Id1, Id2, Id3, and Id4. In this report, we have performed a detailed comparative in situ hybridization analysis to examine their expression pattern during post-gastrulational mouse development. Id1, 2, and 3 are expressed in multiple tissues, whereas Id4 expression can only be detected in neuronal tissues and in the ventral portion of the epithelium of the developing stomach. The regions where Id1-3 genes are expressed, such as gut, lung, kidney, tooth, whisker, and several glandular structures, are undergoing active morphogenetic activities. The expression patterns of Id1, 2, and 3 overlap in many organs, except in the tissue derived from primitive gut. In the latter, Id1 and Id3 signals are detected in the mesenchyme surrounding the epithelium, whereas Id2 is expressed within the epithelium. The difference in the patterns of expressions of Id2-3 and Id4 suggest that the dominant negative transcriptional activity of these two subclasses of the Id family may have different physiological consequences.


Asunto(s)
Expresión Génica , Secuencias Hélice-Asa-Hélice/genética , Animales , Huesos/embriología , Huesos/metabolismo , Cartílago/embriología , Cartílago/metabolismo , Sistema Digestivo/embriología , Sistema Digestivo/metabolismo , Glándulas Endocrinas/embriología , Glándulas Endocrinas/metabolismo , Epitelio/embriología , Epitelio/metabolismo , Glándulas Exocrinas/embriología , Glándulas Exocrinas/metabolismo , Corazón/embriología , Riñón/embriología , Riñón/metabolismo , Pulmón/embriología , Pulmón/metabolismo , Mesodermo/metabolismo , Ratones , Miocardio/metabolismo , Vísceras/embriología , Vísceras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA