Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
J Cell Biol ; 222(11)2023 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-37672657

RESUMEN

Stress granules (SGs) are formed in the cytoplasm in response to various toxic agents and are believed to play a critical role in the regulation of mRNA metabolism during stress. In SGs, mRNAs are stored in an abortive translation initiation complex that can be routed to either translation initiation or degradation. Here, we show that G3BP, a phosphorylation-dependent endoribonuclease that interacts with RasGAP, is recruited to SGs in cells exposed to arsenite. G3BP may thus determine the fate of mRNAs during cellular stress. Remarkably, SG assembly can be either dominantly induced by G3BP overexpression, or on the contrary, inhibited by expressing a central domain of G3BP. This region binds RasGAP and contains serine 149 whose dephosphorylation is induced by arsenite treatment. Critically, a non-phosphorylatable G3BP mutant (S149A) oligomerizes and assembles SG. These results suggest that G3BP is an effector of SG assembly and that Ras signaling contributes to this process by regulating G3BP dephosphorylation.


Asunto(s)
Endorribonucleasas , Gránulos de Estrés , Proteínas Activadoras de ras GTPasa , Arsenitos/farmacología , Endorribonucleasas/genética , Proteínas Activadoras de GTPasa/genética , ARN Mensajero/genética
4.
J Clin Invest ; 131(4)2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33332285

RESUMEN

To clarify the function of cyclin A2 in colon homeostasis and colorectal cancer (CRC), we generated mice deficient for cyclin A2 in colonic epithelial cells (CECs). Colons of these mice displayed architectural changes in the mucosa and signs of inflammation, as well as increased proliferation of CECs associated with the appearance of low- and high-grade dysplasias. The main initial events triggering those alterations in cyclin A2-deficient CECs appeared to be abnormal mitoses and DNA damage. Cyclin A2 deletion in CECs promoted the development of dysplasia and adenocarcinomas in a murine colitis-associated cancer model. We next explored the status of cyclin A2 expression in clinical CRC samples at the mRNA and protein levels and found higher expression in tumors of patients with stage 1 or 2 CRC compared with those of patients with stage 3 or 4 CRC. A meta-analysis of 11 transcriptome data sets comprising 2239 primary CRC tumors revealed different expression levels of CCNA2 (the mRNA coding for cyclin A2) among the CRC tumor subtypes, with the highest expression detected in consensus molecular subtype 1 (CMS1) and the lowest in CMS4 tumors. Moreover, we found high expression of CCNA2 to be a new, independent prognosis factor for CRC tumors.


Asunto(s)
Colon/metabolismo , Neoplasias Colorrectales/metabolismo , Ciclina A2/metabolismo , Homeostasis , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/metabolismo , Animales , Colon/patología , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Ciclina A2/genética , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Estadificación de Neoplasias , Neoplasias Experimentales/diagnóstico , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Pronóstico
5.
Life Sci Alliance ; 3(3)2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32041848

RESUMEN

Microbiota and chronic infections can affect not only immune status, but also the overall physiology of animals. Here, we report that chronic infections dramatically modify the phenotype of Cxcr2 KO mice, impairing in particular, their reproduction ability. We show that exposure of Cxcr2 KO females to multiple types of chronic infections prevents their ability to cycle, reduces the development of the mammary gland and alters the morphology of the uterus due to an impairment of ovary function. Mammary gland and ovary transplantation demonstrated that the hormonal contexture was playing a crucial role in this phenomenon. This was further evidenced by alterations to circulating levels of sex steroid and pituitary hormones. By analyzing at the molecular level the mechanisms of pituitary dysfunction, we showed that in the absence of Cxcr2, bystander infections affect leukocyte migration, adhesion, and function, as well as ion transport, synaptic function behavior, and reproduction pathways. Taken together, these data reveal that a chemokine receptor plays a direct role in pituitary function and reproduction in the context of chronic infections.


Asunto(s)
Hipófisis/fisiología , Receptores de Interleucina-8B/metabolismo , Animales , Femenino , Hormonas Esteroides Gonadales/metabolismo , Infecciones/microbiología , Infecciones/fisiopatología , Masculino , Ratones , Ratones Endogámicos BALB C , Ovario/metabolismo , Enfermedades de la Hipófisis/metabolismo , Hipófisis/metabolismo , Receptores de Interleucina-8B/genética , Reproducción , Útero/metabolismo
6.
Sci Data ; 4: 170150, 2017 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-29039845

RESUMEN

RNA-Seq enables the generation of extensive transcriptome information providing the capability to characterize transcripts (including alternative isoforms and polymorphism), to quantify expression and to identify differential regulation in a single experiment. To reveal the capacity of new anti-HIV ABX464 candidate in modulating the expression of genes, datasets were generated and validated using RNA-seq approach. This comprehensive dataset will be useful to deepen the comprehensive understanding of the progression of human immunodeficiency virus (HIV) associated with mucosal damage in the gastrointestinal (GI) tract and subsequent inflammation, providing an opportunity to generate new therapies, diagnoses, and preventive strategies.


Asunto(s)
Fármacos Anti-VIH/efectos adversos , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/genética , Quinolinas/efectos adversos , Gastroenteritis/complicaciones , Gastroenteritis/tratamiento farmacológico , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico , Humanos , Análisis de Secuencia de ARN
7.
Sci Rep ; 7(1): 4860, 2017 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-28687795

RESUMEN

The progression of human immunodeficiency virus (HIV) is associated with mucosal damage in the gastrointestinal (GI) tract. This damage enables bacterial translocation from the gut and leads to subsequent inflammation. Dextran sulfate sodium (DSS-exposure) is an established animal model for experimental colitis that was recently shown to recapitulate the link between GI-tract damage and pathogenic features of SIV infection. The current study tested the protective properties of ABX464, a first-in-class anti-HIV drug candidate currently in phase II clinical trials. ABX464 treatment strongly attenuated DSS-induced colitis in mice and produced a long-term protection against prolonged DSS-exposure after drug cessation. Consistently, ABX464 reduced the colonic production of the inflammatory cytokines IL-6 and TNFα as well as that of the chemoattractant MCP-1. However, RNA profiling analysis revealed the capacity of ABX464 to induce the expression of IL-22, a cytokine involved in colitis tissue repair, both in DSS-treated mice and in LPS-stimulated bone marrow-derived macrophages. Importantly, anti-IL-22 antibodies significantly reduced the protective effect of ABX464 on colitis in DSS-treated mice. Because reduced IL-22 production in the gut mucosa is an established factor of HIV and DSS-induced immunopathogenesis, our data suggest that the anti-inflammatory properties of ABX464 warrant exploration in both HIV and inflammatory ulcerative colitis (UC) disease.


Asunto(s)
Fármacos Anti-VIH/administración & dosificación , Antiinflamatorios/administración & dosificación , Colitis/tratamiento farmacológico , Interleucinas/metabolismo , Macrófagos/inmunología , Quinolinas/administración & dosificación , Animales , Fármacos Anti-VIH/farmacología , Antiinflamatorios/farmacología , Colitis/inducido químicamente , Colitis/patología , Citocinas/análisis , Sulfato de Dextran/administración & dosificación , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Interleucinas/genética , Mucosa Intestinal/patología , Macrófagos/efectos de los fármacos , Ratones , Quinolinas/farmacología , Resultado del Tratamiento , Interleucina-22
8.
J Neurochem ; 139(3): 349-368, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27513819

RESUMEN

Neuronal granules play an important role in the localization and transport of translationally silenced messenger ribonucleoproteins in neurons. Among the factors associated with these granules, the RNA-binding protein G3BP1 (stress-granules assembly factor) is involved in neuronal plasticity and is induced in Alzheimer's disease. We immunopurified a stable complex containing G3BP1 from mouse brain and performed high-throughput sequencing and cross-linking immunoprecipitation to identify the associated RNAs. The G3BP-complex contained the deubiquitinating protease USP10, CtBP1 and the RNA-binding proteins Caprin-1, G3BP2a and splicing factor proline and glutamine rich, or PSF. The G3BP-complex binds preferentially to transcripts that retain introns, and to non-coding sequences like 3'-untranslated region and long non-coding RNAs. Specific transcripts with retained introns appear to be enriched in the cerebellum compared to the rest of the brain and G3BP1 depletion decreased this intron retention in the cerebellum of G3BP1 knockout mice. Among the enriched transcripts, we found an overrepresentation of genes involved in synaptic transmission, especially glutamate-related neuronal transmission. Notably, G3BP1 seems to repress the expression of the mature Grm5 (metabotropic glutamate receptor 5) transcript, by promoting the retention of an intron in the immature transcript in the cerebellum. Our results suggest that G3BP is involved in a new functional mechanism to regulate non-coding RNAs including intron-retaining transcripts, and thus have broad implications for neuronal gene regulation, where intron retention is widespread.


Asunto(s)
Química Encefálica/genética , Proteínas Portadoras/metabolismo , Cerebelo/metabolismo , Intrones/genética , Regiones no Traducidas 3'/genética , Oxidorreductasas de Alcohol/metabolismo , Animales , Reactivos de Enlaces Cruzados , Gránulos Citoplasmáticos/metabolismo , ADN Helicasas , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas de Unión a Poli-ADP-Ribosa , ARN/biosíntesis , ARN/genética , ARN Helicasas , Proteínas con Motivos de Reconocimiento de ARN , ARN Largo no Codificante/genética , Transcripción Genética , Ubiquitina Tiolesterasa/metabolismo
9.
EMBO Rep ; 15(5): 529-39, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24639560

RESUMEN

Alternative RNA processing of LMNA pre-mRNA produces three main protein isoforms, that is, lamin A, progerin, and lamin C. De novo mutations that favor the expression of progerin over lamin A lead to Hutchinson-Gilford progeria syndrome (HGPS), providing support for the involvement of LMNA processing in pathological aging. Lamin C expression is mutually exclusive with the splicing of lamin A and progerin isoforms and occurs by alternative polyadenylation. Here, we investigate the function of lamin C in aging and metabolism using mice that express only this isoform. Intriguingly, these mice live longer, have decreased energy metabolism, increased weight gain, and reduced respiration. In contrast, progerin-expressing mice show increased energy metabolism and are lipodystrophic. Increased mitochondrial biogenesis is found in adipose tissue from HGPS-like mice, whereas lamin C-only mice have fewer mitochondria. Consistently, transcriptome analyses of adipose tissues from HGPS and lamin C-only mice reveal inversely correlated expression of key regulators of energy expenditure, including Pgc1a and Sfrp5. Our results demonstrate that LMNA encodes functionally distinct isoforms that have opposing effects on energy metabolism and lifespan in mammals.


Asunto(s)
Tejido Adiposo/fisiología , Metabolismo Energético/genética , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Adipocitos/citología , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Envejecimiento , Empalme Alternativo , Animales , Células Cultivadas , Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Lamina Tipo A/biosíntesis , Longevidad/genética , Ratones , Ratones Transgénicos , Mitocondrias , Proteínas Nucleares/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Progeria/genética , Isoformas de Proteínas , Precursores de Proteínas/genética , Transducción de Señal , Factores de Transcripción/metabolismo
10.
J Neurochem ; 125(2): 175-84, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23373770

RESUMEN

Ras-GAP SH3-domain-binding protein, G3BP, is an important component in the assembly of stress granules (SGs), which are cytoplasmic aggregates assembled following translational stress. To assess the physiological function of G3BP, we generated viable G3bp1-knockout (KO) mice, which demonstrated behavioral defects linked to the CNS-associated with ataxia phenotype. Immunohistochemistry pinpointed high expression of G3BP in the cytoplasm of hippocampal neurons and Purkinje cells of the cerebellum of wild-type mice. Also, electrophysiological measurements revealed that the absence of G3BP1 leads to an enhancement of short-term potentiation (STP) and long-term depression in the CA1 area of G3bp1 KO mice compared with wild-type mice. Consistently, G3BP1 deficiency in neurons leads to an increase in intracellular calcium and calcium release in response to (S)-3,5-Dihydroxyphenylglycine, a selective agonist of group I metabotropic glutamate receptors. These results show, for the first time, a requirement for G3BP1 in the control of neuronal plasticity and calcium homeostasis and further establish a direct link between SG formation and neurodegenerative diseases.


Asunto(s)
Encéfalo/metabolismo , Proteínas Portadoras/metabolismo , Homeostasis/fisiología , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Animales , Western Blotting , Calcio/metabolismo , Gránulos Citoplasmáticos/metabolismo , Femenino , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados
11.
Blood ; 121(11): 2144-53, 2013 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-23305740

RESUMEN

Donor hematopoietic stem cells (HSCs) can correct T-cell deficiencies in patients with severe combined immunodeficiency by replacing resident thymus cells. However, as those progenitors that naturally migrate to the thymus are not capable of supporting long-term thymopoiesis, a successful transplant is thought to require the ongoing migration of donor progenitors. We previously showed that the forced intrathymic administration of histocompatible HSCs can sustain long-term thymopoiesis in ZAP-70-immunodeficient mice. However, it is not known whether T-cell reconstitution across histocompatibility barriers is modulated by intrathymic vs intravenous administration of HSCs. In the absence of conditioning, long-term thymopoiesis by semiallogeneic progenitors was detected in mice transplanted via the intrathymic, but not the intravenous, route. In intrathymic-transplanted mice, ongoing thymopoiesis was associated with a 10-fold higher level of early thymic progenitors (ETPs). The enhanced reconstitution capacity of these intrathymic-derived ETPs was corroborated by their significantly augmented myeloid lineage potential compared with endogenous ETPs. Notably, though, myeloablative conditioning resulted in a reduced expansion of intrathymic-administered donor ETPs. Thus, in the absence of conditioning, the forced thymic entry of HSCs results in a sustained T-cell development across histocompatibility barriers, highlighting the capacity of the thymus to support cells with long-term renewal potential.


Asunto(s)
Diferenciación Celular/inmunología , Supervivencia de Injerto , Trasplante de Células Madre Hematopoyéticas/métodos , Histocompatibilidad/fisiología , Células Progenitoras Linfoides/fisiología , Linfocitos T/fisiología , Timo , Animales , Células Cultivadas , Supervivencia de Injerto/inmunología , Supervivencia de Injerto/fisiología , Hematopoyesis/inmunología , Hematopoyesis/fisiología , Histocompatibilidad/inmunología , Prueba de Histocompatibilidad , Infusiones Intravenosas , Células Progenitoras Linfoides/citología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología , Timo/citología , Acondicionamiento Pretrasplante/métodos , Proteína Tirosina Quinasa ZAP-70/deficiencia , Proteína Tirosina Quinasa ZAP-70/genética , Proteína Tirosina Quinasa ZAP-70/inmunología
12.
EMBO J ; 31(3): 606-15, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22117218

RESUMEN

Genomic imprinting in mammals is controlled by DNA methylation imprints that are acquired in the gametes, at essential sequence elements called 'imprinting control regions' (ICRs). What signals paternal imprint acquisition in male germ cells remains unknown. To address this question, we explored histone methylation at ICRs in mouse primordial germ cells (PGCs). By 13.5 days post coitum (d.p.c.), H3 lysine-9 and H4 lysine-20 trimethylation are depleted from ICRs in male (and female) PGCs, indicating that these modifications do not signal subsequent imprint acquisition, which initiates at ∼15.5 d.p.c. Furthermore, during male PGC development, H3 lysine-4 trimethylation becomes biallelically enriched at 'maternal' ICRs, which are protected against DNA methylation, and whose promoters are active in the male germ cells. Remarkably, high transcriptional read-through is detected at the paternal ICRs H19-DMR and Ig-DMR at the time of imprint establishment, from one of the strands predominantly. Combined, our data evoke a model in which differential histone modification states linked to transcriptional events may signal the specificity of imprint acquisition during spermatogenesis.


Asunto(s)
Metilación de ADN , Impresión Genómica , Histonas/metabolismo , Transcripción Genética , Animales , Separación Celular , Inmunoprecipitación de Cromatina , Drosophila melanogaster , Femenino , Citometría de Flujo , Masculino
13.
Proc Natl Acad Sci U S A ; 107(49): 21076-81, 2010 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-21088222

RESUMEN

A growing body of evidence suggests that the multifunctional protein E4F1 is involved in signaling pathways that play essential roles during normal development and tumorigenesis. We generated E4F1 conditional knockout mice to address E4F1 functions in vivo in newborn and adult skin. E4F1 inactivation in the entire skin or in the basal compartment of the epidermis induces skin homeostasis defects, as evidenced by transient hyperplasia in the interfollicular epithelium and alteration of keratinocyte differentiation, followed by loss of cellularity in the epidermis and severe skin ulcerations. E4F1 depletion alters clonogenic activity of epidermal stem cells (ESCs) ex vivo and ends in exhaustion of the ESC pool in vivo, indicating that the lesions observed in the E4F1 mutant skin result, at least in part, from cell-autonomous alterations in ESC maintenance. The clonogenic potential of E4F1 KO ESCs is rescued by Bmi1 overexpression or by Ink4a/Arf or p53 depletion. Skin phenotype of E4F1 KO mice is also delayed in animals with Ink4a/Arf and E4F1 compound gene deficiencies. Our data identify a regulatory axis essential for ESC-dependent skin homeostasis implicating E4F1 and the Bmi1-Arf-p53 pathway.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Células Epidérmicas , Homeostasis , Células Madre/fisiología , Factores de Transcripción/fisiología , Factores de Edad , Animales , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Ratones , Ratones Noqueados , Proteínas Nucleares/metabolismo , Fenotipo , Complejo Represivo Polycomb 1 , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Células Madre/citología , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas
14.
PLoS One ; 5(6): e11235, 2010 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-20574536

RESUMEN

BACKGROUND: The c-fos proto-oncogene is an archetype for rapid and integrative transcriptional activation. Innumerable studies have focused on the canonical promoter, located upstream from the transcriptional start site. However, several regulatory sequences have been found in the first intron. METHODOLOGY/PRINCIPAL FINDINGS: Here we describe an extremely conserved region in c-fos first intron that contains a putative TATA box, and functional TRE and CRE sites. This fragment drives reporter gene activation in fibroblasts, which is enhanced by increasing intracellular calcium and cAMP and by cotransfection of CREB or c-Fos/c-Jun expression vectors. We produced transgenic mice expressing a lacZ reporter controlled by the intronic promoter. Lac Z expression of this promoter is restricted to the developing central nervous system (CNS) and the mesenchyme of developing mammary buds in embryos 12.5 days post-conception, and to brain tissue in adults. RT-QPCR analysis of tissue mRNA, including the anlage of the mammary gland and the CNS, confirms the existence of a novel, nested mRNA initiated in the first intron. CONCLUSIONS/SIGNIFICANCE: Our results provide evidence for a novel, developmentally regulated promoter in the first intron of the c-fos gene.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Intrones/genética , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-fos/genética , Factor de Transcripción AP-1/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Secuencia Conservada , Ensayo de Cambio de Movilidad Electroforética , Evolución Molecular , Femenino , Humanos , Luciferasas/genética , Masculino , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Especificidad de Órganos , Proto-Oncogenes Mas , ARN Mensajero/genética , TATA Box/genética , Xenopus , beta-Galactosidasa/genética
15.
J Leukoc Biol ; 84(2): 380-8, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18483203

RESUMEN

The TNF family member, a proliferation-inducing ligand (APRIL), has been suggested to act as a costimulatory molecule in T cell responses. However, studies addressing this role in vivo are largely lacking. Here, we evaluated the effects of APRIL on physiological T cell responses in vivo. Although receptors for APRIL are expressed on a subset of T cells, neither TCR transgenic (Tg) T cell responses nor endogenous TCR responses were affected by Tg APRIL expression in vivo. Moreover, APRIL did not significantly enhance the induction of T cell lymphomas upon Moloney murine leukemia virus (MLV) infection. This clearly contrasts current belief and indicates that APRIL does not serve a major role in T cell immunity or lymphomagenesis. However, we did observe a strong increase in erythroleukemia formation after MLV inoculation of APRIL Tg mice. Strikingly, this erythroleukemia-facilitating property of APRIL was confirmed using the erythroleukemogenic Friend-MLV. Erythroleukemia in APRIL Tg mice was characterized by low hematocrits and grossly enlarged spleens with an increased percentage of erythroid precursors. Altogether, these results unveil new proerythroleukemogenic properties of APRIL.


Asunto(s)
Leucemia Eritroblástica Aguda/inmunología , Leucemia Eritroblástica Aguda/virología , Linfoma de Células T/fisiopatología , Linfocitos T/inmunología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Animales , Autoinmunidad , Citometría de Flujo , Hematócrito , Heterocigoto , Homocigoto , Linfoma de Células T/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Virus de la Leucemia Murina de Moloney/inmunología , Virus de la Leucemia Murina de Moloney/patogenicidad , Ovalbúmina/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Células Madre/fisiología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
16.
Int J Dev Biol ; 52(4): 383-8, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18415939

RESUMEN

Regulation of migration and proliferation by calpain has been shown in various cell types; however, no data are available concerning calpain 2 (capn2) localization in embryonic tissues. Here, we report the expression pattern of capn2 during mouse embryonic development. Expression of the capn2 gene is observed throughout embryonic development. From ES cells and the 8-cell stage to late neurulation stages, CAPN2 is expressed in the cytoplasm and nuclear compartments, with a clear co-localisation with chromatin. Whole-mount in situ hybridization analysis from E8.5 to 14.5 stages indicates high levels of capn2 expression in the nervous system, heart and mesodermal tissues. Up-regulation is maintained during later developmental stages in proliferating cells and in precursor cells involved in muscle (myoblasts) or bone formation (chondrocytes). At later developmental stages, elevated mRNA levels coincided with CAPN2 nuclear localization in these cell types, while differentiated cells maintained cytoplasmic expression. This detailed analysis reveals dynamic expression: nuclear localization was associated either with active cell mitosis in embryonic stem cells and early developmental stages or with precursor cells later during organogenesis. Thus, these data indicate that CAPN2 may represent a key factor in development from the first cell division.


Asunto(s)
Calpaína/genética , Desarrollo Embrionario/genética , Animales , Calpaína/metabolismo , Desarrollo Embrionario/fisiología , Células Madre Embrionarias/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Hibridación in Situ , Ratones , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Somitos/metabolismo , Fracciones Subcelulares/metabolismo , Distribución Tisular
17.
Mol Cell Biol ; 28(7): 2314-23, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18212043

RESUMEN

The chemotropic guidance cue netrin-1 promotes neurite outgrowth through its receptor Deleted in Colorectal Cancer (DCC) via activation of Rac1. The guanine nucleotide exchange factor (GEF) linking netrin-1/DCC to Rac1 activation has not yet been identified. Here, we show that the RhoGEF Trio mediates Rac1 activation in netrin-1 signaling. We found that Trio interacts with the netrin-1 receptor DCC in mouse embryonic brains and that netrin-1-induced Rac1 activation in brain is impaired in the absence of Trio. Trio(-/-) cortical neurons fail to extend neurites in response to netrin-1, while they are able to respond to glutamate. Accordingly, netrin-1-induced commissural axon outgrowth is reduced in Trio(-/-) spinal cord explants, and the guidance of commissural axons toward the floor plate is affected by the absence of Trio. The anterior commissure is absent in Trio-null embryos, and netrin-1/DCC-dependent axonal projections that form the internal capsule and the corpus callosum are defective in the mutants. Taken together, these findings establish Trio as a GEF that mediates netrin-1 signaling in axon outgrowth and guidance through its ability to activate Rac1.


Asunto(s)
Conos de Crecimiento/fisiología , Factores de Intercambio de Guanina Nucleótido/fisiología , Factores de Crecimiento Nervioso/fisiología , Neuropéptidos/fisiología , Fosfoproteínas/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Receptores de Superficie Celular/fisiología , Proteínas Supresoras de Tumor/fisiología , Proteínas de Unión al GTP rac/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Encéfalo/anomalías , Encéfalo/citología , Encéfalo/embriología , Células COS , Línea Celular , Células Cultivadas/citología , Chlorocebus aethiops , Receptor DCC , Activación Enzimática , Femenino , Conos de Crecimiento/ultraestructura , Factores de Intercambio de Guanina Nucleótido/deficiencia , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Netrina-1 , Proteínas Oncogénicas/fisiología , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Mapeo de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Médula Espinal/citología , Médula Espinal/embriología , Quinasas p21 Activadas/fisiología , Proteína de Unión al GTP rac1
18.
Mol Cell Biol ; 28(3): 1104-13, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18039842

RESUMEN

Whereas DNA methylation is essential for genomic imprinting, the importance of histone methylation in the allelic expression of imprinted genes is unclear. Imprinting control regions (ICRs), however, are marked by histone H3-K9 methylation on their DNA-methylated allele. In the placenta, the paternal silencing along the Kcnq1 domain on distal chromosome 7 also correlates with the presence of H3-K9 methylation, but imprinted repression at these genes is maintained independently of DNA methylation. To explore which histone methyltransferase (HMT) could mediate the allelic H3-K9 methylation on distal chromosome 7, and at ICRs, we generated mouse conceptuses deficient for the SET domain protein G9a. We found that in the embryo and placenta, the differential DNA methylation at ICRs and imprinted genes is maintained in the absence of G9a. Accordingly, in embryos, imprinted gene expression was unchanged at the domains analyzed, in spite of a global loss of H3-K9 dimethylation (H3K9me2). In contrast, the placenta-specific imprinting of genes on distal chromosome 7 is impaired in the absence of G9a, and this correlates with reduced levels of H3K9me2 and H3K9me3. These findings provide the first evidence for the involvement of an HMT and suggest that histone methylation contributes to imprinted gene repression in the trophoblast.


Asunto(s)
Impresión Genómica , N-Metiltransferasa de Histona-Lisina/fisiología , Histonas/metabolismo , Placenta/enzimología , Animales , Embrión de Mamíferos , Femenino , Silenciador del Gen , Histona Metiltransferasas , Metilación , Ratones , Proteína Metiltransferasas , Trofoblastos/metabolismo
19.
Dev Cell ; 11(4): 535-46, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17011492

RESUMEN

The transcriptional mechanisms underlying lineage specification and differentiation of embryonic stem (ES) cells remain elusive. Oct-3/4 (POU5f1) is one of the earliest transcription factors expressed in the embryo. Both the pluripotency and the fate of ES cells depend upon a tight control of Oct-3/4 expression. We report that transgene- or TGFbeta-induced increase in Oct-3/4 mRNA and protein levels in undifferentiated ES cells and at early stages of differentiation triggers expression of mesodermal and cardiac specific genes through Smad2/4. cDNA antisense- and siRNA-mediated inhibition of upregulation of Oct-3/4 in ES cells prevent their specification toward the mesoderm and their differentiation into cardiomyocytes. Similarly, Oct-3/4 siRNA injected in the inner cell mass of blastocysts impairs cardiogenesis in early embryos. Thus, quantitative Oct-3/4 expression is regulated by a morphogen, pointing to a pivotal and physiological function of the POU factor in mesodermal and cardiac commitments of ES cells and of the epiblast.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Corazón/crecimiento & desarrollo , Miocitos Cardíacos/citología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre/citología , Células Madre/fisiología , Animales , Blastocisto/citología , Blastocisto/metabolismo , Linaje de la Célula , Inmunohistoquímica , Hibridación in Situ , Ratones , Microinyecciones , Miocitos Cardíacos/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Técnicas de Cultivo de Órganos , ARN Mensajero/biosíntesis , ARN Interferente Pequeño/metabolismo
20.
Clin Immunol ; 119(1): 38-50, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16426893

RESUMEN

A baculovirus-expressed chimeric recombinant IgG1 (rIgG1) antibody, with Cgamma1 and Ckappa human constant domains, was derived from the murine monoclonal antibody (mAb) 13B8.2, which is specific for the CDR3-like loop of the CD4 molecule and which inhibits HIV-1 replication. Chimeric rIgG1 antibody 13B8.2 blocked, in a dose-dependent manner, antigen presentation through inhibition of subsequent IL-2 secretion by stimulated T cells. The one-way mixed lymphocyte reaction was abrogated by previous addition of baculovirus-produced rIgG1 13B8.2 in the T-cell culture. Anti-proliferative activity of rIgG1 was demonstrated on CD3-activated CD4+ T lymphocytes from healthy donors, such effect being associated with reduced IL-2 secretion of activated T cells. On the other hand, no proliferation inhibition was observed on CD4+ T lymphocytes activated with phorbol ester plus ionomycin, suggesting that rIgG1 13B8.2 preferentially acts on a proximal TCR-induced signaling pathway. Treatment of DBA1/J human CD4-transgenic mice with 100 microg of recombinant antibody for three consecutive days led to in vivo recovery of rIgG1 antibody 13B8.2 both coated on murine T lymphocytes and free in mouse serum, without CD4 depletion or down-modulation. These findings predict that the baculovirus-expressed chimeric rIgG1 anti-CD4 antibody 13B8.2 is a promising candidate for immunotherapy.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígenos CD4/inmunología , Inmunoglobulina G/inmunología , Proteínas Recombinantes de Fusión/farmacología , Linfocitos T/efectos de los fármacos , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Formación de Anticuerpos/inmunología , Presentación de Antígeno/efectos de los fármacos , Reacciones Antígeno-Anticuerpo/inmunología , Baculoviridae/genética , Complejo CD3/inmunología , Antígenos CD4/genética , Linfocitos T CD4-Positivos/inmunología , Línea Celular , Proliferación Celular/efectos de los fármacos , Epítopos/genética , Epítopos/inmunología , Expresión Génica/efectos de los fármacos , Duplicado del Terminal Largo de VIH/genética , Células HeLa , Humanos , Inmunización Pasiva , Interleucina-2/metabolismo , Ionomicina/farmacología , Activación de Linfocitos/efectos de los fármacos , Prueba de Cultivo Mixto de Linfocitos , Ratones , Ratones Endogámicos DBA , Ratones Transgénicos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Acetato de Tetradecanoilforbol/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...