Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Nature ; 598(7881): 483-488, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34599305

RESUMEN

The prefrontal cortex (PFC) and its connections with the mediodorsal thalamus are crucial for cognitive flexibility and working memory1 and are thought to be altered in disorders such as autism2,3 and schizophrenia4,5. Although developmental mechanisms that govern the regional patterning of the cerebral cortex have been characterized in rodents6-9, the mechanisms that underlie the development of PFC-mediodorsal thalamus connectivity and the lateral expansion of the PFC with a distinct granular layer 4 in primates10,11 remain unknown. Here we report an anterior (frontal) to posterior (temporal), PFC-enriched gradient of retinoic acid, a signalling molecule that regulates neural development and function12-15, and we identify genes that are regulated by retinoic acid in the neocortex of humans and macaques at the early and middle stages of fetal development. We observed several potential sources of retinoic acid, including the expression and cortical expansion of retinoic-acid-synthesizing enzymes specifically in primates as compared to mice. Furthermore, retinoic acid signalling is largely confined to the prospective PFC by CYP26B1, a retinoic-acid-catabolizing enzyme, which is upregulated in the prospective motor cortex. Genetic deletions in mice revealed that retinoic acid signalling through the retinoic acid receptors RXRG and RARB, as well as CYP26B1-dependent catabolism, are involved in proper molecular patterning of prefrontal and motor areas, development of PFC-mediodorsal thalamus connectivity, intra-PFC dendritic spinogenesis and expression of the layer 4 marker RORB. Together, these findings show that retinoic acid signalling has a critical role in the development of the PFC and, potentially, in its evolutionary expansion.


Asunto(s)
Organogénesis , Corteza Prefrontal/embriología , Corteza Prefrontal/metabolismo , Tretinoina/metabolismo , Animales , Axones/metabolismo , Corteza Cerebral , Regulación hacia Abajo , Femenino , Humanos , Macaca mulatta , Masculino , Ratones , Pan troglodytes , Corteza Prefrontal/anatomía & histología , Corteza Prefrontal/citología , Receptores de Ácido Retinoico/deficiencia , Receptor gamma X Retinoide/deficiencia , Transducción de Señal , Sinapsis/metabolismo , Tálamo/anatomía & histología , Tálamo/citología , Tálamo/metabolismo
2.
Development ; 147(12)2020 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-32467243

RESUMEN

Retinoic acid (RA) signaling is essential for multiple developmental processes, including appropriate pancreas formation from the foregut endoderm. RA is also required to generate pancreatic progenitors from human pluripotent stem cells. However, the role of RA signaling during endocrine specification has not been fully explored. In this study, we demonstrate that the disruption of RA signaling within the NEUROG3-expressing endocrine progenitor population impairs mouse ß cell differentiation and induces ectopic expression of crucial δ cell genes, including somatostatin. In addition, the inhibition of the RA pathway in hESC-derived pancreatic progenitors downstream of NEUROG3 induction impairs insulin expression. We further determine that RA-mediated regulation of endocrine cell differentiation occurs through Wnt pathway components. Together, these data demonstrate the importance of RA signaling in endocrine specification and identify conserved mechanisms by which RA signaling directs pancreatic endocrine cell fate.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Páncreas/metabolismo , Transducción de Señal , Tretinoina/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Embrión de Mamíferos/metabolismo , Proteínas de Homeodominio/genética , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Páncreas/citología , Receptores de Ácido Retinoico/deficiencia , Receptores de Ácido Retinoico/genética , Somatostatina/genética , Somatostatina/metabolismo , Células Secretoras de Somatostatina/citología , Células Secretoras de Somatostatina/metabolismo , Células Madre/citología , Células Madre/metabolismo , Transactivadores/deficiencia , Transactivadores/genética , Proteínas Wnt/metabolismo
3.
J Neurosci ; 38(49): 10454-10466, 2018 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-30355624

RESUMEN

Homeostatic synaptic plasticity is a synaptic mechanism through which the nervous system adjusts synaptic excitation and inhibition to maintain network stability. Retinoic acid (RA) and its receptor RARα have been established as critical mediators of homeostatic synaptic plasticity. In vitro studies reveal that RA signaling enhances excitatory synaptic strength and decreases inhibitory synaptic strength. However, it is unclear whether RA-mediated homeostatic synaptic plasticity occurs in vivo, and if so, whether it operates at specific types of synapses. Here, we examine the impact of RA/RARα signaling in the monocular zone of primary visual cortex (V1m) in mice of either sex. Exogenous RA treatment in acute cortical slices resulted in a reduction in mIPSCs of layer 2/3 pyramidal neurons, an effect mimicked by visual deprivation induced by binocular enucleation in postcritical period animals. Postnatal deletion of RARα blocked RA's effect on mIPSCs. Cell type-specific deletion of RARα revealed that RA acted specifically on parvalbumin (PV)-expressing interneurons. RARα deletion in PV+ interneurons blocked visual deprivation-induced changes in mIPSCs, demonstrating the critical involvement of RA signaling in PV+ interneurons in vivo Moreover, visual deprivation- or RA-induced downregulation of synaptic inhibition was absent in the visual cortical circuit of constitutive and PV-specific Fmr1 KO mice, strongly suggesting a functional interaction between fragile X mental retardation protein and RA signaling pathways. Together, our results demonstrate that RA/RARα signaling acts as a key component for homeostatic regulation of synaptic transmission at the inhibitory synapses of the visual cortex.SIGNIFICANCE STATEMENTIn vitro studies established that retinoic acid (RA) and its receptor RARα play key roles in homeostatic synaptic plasticity, a mechanism by which synaptic excitation/inhibition balance and network stability are maintained. However, whether synaptic RA signaling operates in vivo remains undetermined. Here, using a conditional RARα KO mouse and cell type-specific Cre-driver lines, we showed that RARα signaling in parvalbumin-expressing interneurons is crucial for visual deprivation-induced homeostatic synaptic plasticity at inhibitory synapses in visual cortical circuits. Importantly, this form of synaptic plasticity is absent when fragile X mental retardation protein is selectively deleted in parvalbumin-expressing interneurons, suggesting a functional connection between RARα and fragile X mental retardation protein signaling pathways in vivo Thus, dysfunction of RA-dependent homeostatic plasticity may contribute to cortical circuit abnormalities in fragile X syndrome.


Asunto(s)
Homeostasis/fisiología , Plasticidad Neuronal/fisiología , Receptores de Ácido Retinoico/deficiencia , Sinapsis/fisiología , Corteza Visual/patología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Inhibición Neural/fisiología , Receptores de Ácido Retinoico/genética
4.
J Endocrinol ; 236(3): 151-165, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29371236

RESUMEN

Excessive and/or persistent activation of calcium-calmodulin protein kinase II (CaMKII) is detrimental in acute and chronic cardiac injury. However, intrinsic regulators of CaMKII activity are poorly understood. We find that cellular retinoic acid-binding protein 1 (CRABP1) directly interacts with CaMKII and uncover a functional role for CRABP1 in regulating CaMKII activation. We generated Crabp1-null mice (CKO) in C57BL/6J background for pathophysiological studies. CKO mice develop hypertrophy as adults, exhibiting significant left ventricular dilation with reduced ejection fraction at the baseline cardiac function. Interestingly, CKO mice have elevated basal CaMKII phosphorylation at T287, and phosphorylation on its substrate phospholamban (PLN) at T17. Acute isoproterenol (ISO) challenge (80 mg/kg two doses in 1 day) causes more severe apoptosis and necrosis in CKO hearts, and treatment with a CaMKII inhibitor KN-93 protects CKO mice from this injury. Chronic (30 mg/kg/day) ISO challenge also significantly increases hypertrophy and fibrosis in CKO mice as compared to WT. In wild-type mice, CRABP1 expression is increased in early stages of ISO challenge and eventually reduces to the basal level. Mechanistically, CRABP1 directly inhibits CaMKII by competing with calmodulin (CaM) for CaMKII interaction. This study demonstrates increased susceptibility of CKO mice to ISO-induced acute and chronic cardiac injury due to, at least in part, elevated CaMKII activity. Deleting Crabp1 results in reduced baseline cardiac function and aggravated damage challenged with acute and persistent ß-adrenergic stimulation. This is the first report of a physiological role of CRABP1 as an endogenous regulator of CaMKII, which protects the heart from ISO-induced damage.


Asunto(s)
Corazón/efectos de los fármacos , Corazón/fisiopatología , Isoproterenol/administración & dosificación , Receptores de Ácido Retinoico/fisiología , Animales , Apoptosis/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Cardiomegalia/inducido químicamente , Cardiomegalia/patología , Cardiotónicos , Activación Enzimática/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/patología , Necrosis/inducido químicamente , Receptores de Ácido Retinoico/deficiencia , Receptores de Ácido Retinoico/metabolismo , Remodelación Ventricular/fisiología
5.
Int J Gynecol Cancer ; 27(4): 643-650, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28375930

RESUMEN

OBJECTIVE: Several studies have reported that retinoic acid (RA) might be used to treat malignancies. The effects of RA are mediated by the RA receptor (RAR), and RARα/RARß especially acts as a tumor suppressor. However, little is known about its role in human endometrial cancer. MATERIALS AND METHODS: In this study, we examined the effects of all-trans RA (ATRA) on progression of human endometrial cancer cell line, RL95-2 and Hec1A. We then examined the expression of RARα and RARß in 50 endometrial cancer tissues by using immunohistochemistry. RESULTS: We found inhibitory effects of ATRA on cell proliferation, apoptosis, and migration in RL95-2 cells, but not in Hec1A cells. RARα or RARß knockdown individually could not cancel out the inhibition of cell proliferation by ATRA in RL95-2 cells, but simultaneous knockdown of RARα and RARß could block its effect on proliferation. RARα and RARß knockdown dose dependently reduced the inhibition of migration by ATRA, but the effect was more pronounced with RARß knockdown than with RARα knockdown. We confirmed that RARß gene was directly regulated by ATRA in microarray and real-time reverse transcription polymerase chain reaction. Furthermore, the RARß agonist (BMS453) significantly suppressed proliferation of RL95-2 cells. In immunohistochemical analysis, RARα expression was positively correlated with tumor grade, and RARß showed the opposite tendency in endometrial cancer. CONCLUSIONS: Retinoic acid might have multiple antitumor effects, and RARß may be a potent therapeutic target in RA treatment for endometrial cancers.


Asunto(s)
Neoplasias Endometriales/tratamiento farmacológico , Receptores de Ácido Retinoico/metabolismo , Tretinoina/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Inmunohistoquímica , Terapia Molecular Dirigida , Receptores de Ácido Retinoico/biosíntesis , Receptores de Ácido Retinoico/deficiencia , Receptores de Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico/biosíntesis , Receptor alfa de Ácido Retinoico/deficiencia , Receptor alfa de Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico/metabolismo
6.
Nucl Recept Signal ; 12: e004, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25422594

RESUMEN

Retinoic acid receptor ß (RARß) has been proposed to act as a tumor suppressor in breast cancer. In contrast, recent data have shown that RARß promotes ERBB2-induced mammary gland tumorigenesis through remodeling of the stromal compartment and activation of cancer-associated fibroblasts. However, it is currently unknown whether RARß oncogenic activity is specific to ERBB2-induced tumors, or whether it influences the initiation and progression of other breast cancer subtypes. Accordingly, we set out to investigate the involvement of RARß in basal-like breast cancer using mouse mammary tumor virus (MMTV)-wingless-related integration site 1 (Wnt1)-induced mammary gland tumorigenesis as a model system. We found that compared with wild type mice, inactivation of Rarb resulted in a lengthy delay in Wnt1-induced mammary gland tumorigenesis and in a significantly slower tumor growth rate. Ablation of Rarb altered the composition of the stroma, repressed the activation of cancer-associated fibroblasts, and reduced the recruitment of inflammatory cells and angiogenesis. Reduced expression of IGF-1 and activity of its downstream signaling pathway contribute to attenuate EMT in the Rarb-null tumors. Our results show that, in the absence of retinoid signaling via RARß, reduced IGF-1 signaling results in suppression of epithelial-mesenchymal transition and delays tumorigenesis induced by the Wnt1 oncogene. Accordingly, our work reinforces the concept that antagonizing RARß-dependent retinoid signaling could provide a therapeutic avenue to treat poor outcome breast cancers.


Asunto(s)
Carcinogénesis/genética , Transición Epitelial-Mesenquimal/genética , Eliminación de Gen , Neoplasias Mamarias Experimentales/patología , Receptores de Ácido Retinoico/deficiencia , Receptores de Ácido Retinoico/genética , Proteína Wnt1/metabolismo , Animales , Línea Celular Tumoral , Regulación hacia Abajo/genética , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Neoplasias Mamarias Experimentales/genética , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células del Estroma/patología
7.
J Immunol ; 192(7): 3336-44, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24610012

RESUMEN

Vitamin A deficiency leads to increased susceptibility to a spectrum of infectious diseases. The studies presented dissect the intrinsic role of each of the retinoic acid receptor (RAR) isoforms in the clonal expansion, differentiation, and survival of pathogen-specific CD8 T cells in vivo. The data show that RARα is required for the expression of gut-homing receptors on CD8(+) T cells and survival of CD8(+) T cells in vitro. Furthermore, RARα is essential for survival of CD8(+) T cells in vivo following Listeria monocytogenes infection. In contrast, RARß deletion leads to modest deficiency in Ag-specific CD8(+) T cell expansion during infection. The defective survival of RARα-deficient CD8(+) T cells leads to a deficiency in control of L. monocytogenes expansion in the spleen. To our knowledge, these are the first comparative studies of the role of RAR isoforms in CD8(+) T cell immunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Receptores de Ácido Retinoico/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Proliferación Celular , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Citometría de Flujo , Expresión Génica/inmunología , Interacciones Huésped-Patógeno/inmunología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-2/inmunología , Interleucina-2/metabolismo , Listeria monocytogenes/fisiología , Listeriosis/microbiología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Ácido Retinoico/deficiencia , Receptores de Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/inmunología , Bazo/metabolismo , Bazo/microbiología , Receptor de Ácido Retinoico gamma
8.
Br J Haematol ; 164(2): 280-5, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24383846

RESUMEN

Vitamin A and its derivatives (retinoids) are important regulators of haematopoiesis, acting via retinoic acid receptors (RARs). Epidemiological studies indicated an association of vitamin A deficiency with anaemia in humans. To define the requirements of RARs in erythropoiesis, we evaluated erythroid parameters in RAR germ-line deficient and conditional knock out mice with erythroid specific deletion of RARs. Adult RARγ(-/-) mice were anaemic, however, Epor-Cre Rara(fl/fl) , Epor-Cre Rarg(fl/fl) and Epor-Cre Rara(fl/fl) g(fl/fl) mice were normal, indicating a lack of an erythroid intrinsic RAR function. Therefore, erythroid-specific RAR function is dispensable for erythropoiesis and RARγ plays an erythroid extrinsic role in erythropoiesis.


Asunto(s)
Eritropoyesis/fisiología , Receptores de Ácido Retinoico/fisiología , Animales , Células de la Médula Ósea/metabolismo , Eritroblastos/metabolismo , Inmunofenotipificación , Ratones , Ratones Noqueados , Fenotipo , Receptores de Eritropoyetina/genética , Receptores de Ácido Retinoico/deficiencia , Receptores de Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico , Receptor de Ácido Retinoico gamma
9.
PLoS Genet ; 9(8): e1003689, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23990796

RESUMEN

Normal embryonic development and tissue homeostasis require precise levels of retinoic acid (RA) signaling. Despite the importance of appropriate embryonic RA signaling levels, the mechanisms underlying congenital defects due to perturbations of RA signaling are not completely understood. Here, we report that zebrafish embryos deficient for RA receptor αb1 (RARαb1), a conserved RAR splice variant, have enlarged hearts with increased cardiomyocyte (CM) specification, which are surprisingly the consequence of increased RA signaling. Importantly, depletion of RARαb2 or concurrent depletion of RARαb1 and RARαb2 also results in increased RA signaling, suggesting this effect is a broader consequence of RAR depletion. Concurrent depletion of RARαb1 and Cyp26a1, an enzyme that facilitates degradation of RA, and employment of a novel transgenic RA sensor line support the hypothesis that the increases in RA signaling in RAR deficient embryos are the result of increased embryonic RA coupled with compensatory RAR expression. Our results support an intriguing novel mechanism by which depletion of RARs elicits a previously unrecognized positive feedback loop that can result in developmental defects due to teratogenic increases in embryonic RA.


Asunto(s)
Receptores de Ácido Retinoico/genética , Transducción de Señal , Teratogénesis/genética , Tretinoina/metabolismo , Animales , Animales Modificados Genéticamente , Sistema Enzimático del Citocromo P-450/metabolismo , Embrión no Mamífero , Desarrollo Embrionario/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Receptores de Ácido Retinoico/deficiencia , Ácido Retinoico 4-Hidroxilasa , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra
10.
J Lipid Res ; 54(9): 2458-74, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23833249

RESUMEN

All-trans retinoic acid (ATRA) has a key role in dendritic cells (DCs) and affects T cell subtype specification and gut homing. However, the identity of the permissive cell types and the required steps of conversion of vitamin A to biologically active ATRA bringing about retinoic acid receptor-regulated signaling remains elusive. Here we present that only a subset of murine and human DCs express the necessary enzymes, including RDH10, RALDH2, and transporter cellular retinoic acid binding protein (CRABP)2, to produce ATRA and efficient signaling. These permissive cell types include CD103(+) DCs, granulocyte-macrophage colony-stimulating factor, and interleukin-4-treated bone marrow-derived murine DCs and human monocyte-derived DCs (mo-DCs). Importantly, in addition to RDH10 and RALDH2, CRABP2 also appears to be regulated by the fatty acid-sensing nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ) and colocalize in human gut-associated lymphoid tissue DCs. In our model of human mo-DCs, all three proteins (RDH10, RALDH2, and CRABP2) appeared to be required for ATRA production induced by activation of PPARγ and therefore form a linear pathway. This now functionally validated PPARγ-regulated ATRA producing and signaling axis equips the cells with the capacity to convert precursors to active retinoids in response to receptor-activating fatty acids and is potentially amenable to intervention in diseases involving or affecting mucosal immunity.


Asunto(s)
Oxidorreductasas de Alcohol/metabolismo , Células Dendríticas/metabolismo , PPAR gamma/metabolismo , Receptores de Ácido Retinoico/metabolismo , Retinal-Deshidrogenasa/metabolismo , Transducción de Señal , Tretinoina/metabolismo , Oxidorreductasas de Alcohol/deficiencia , Oxidorreductasas de Alcohol/genética , Familia de Aldehído Deshidrogenasa 1 , Animales , Células Dendríticas/citología , Células Dendríticas/enzimología , Proteínas de Unión al GTP/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Intestinos/citología , Masculino , Ratones , Monocitos/citología , Células T Asesinas Naturales/citología , Células T Asesinas Naturales/metabolismo , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transporte de Proteínas , Receptores de Ácido Retinoico/deficiencia , Receptores de Ácido Retinoico/genética , Retinal-Deshidrogenasa/deficiencia , Retinal-Deshidrogenasa/genética , Transglutaminasas/metabolismo
11.
Med Oncol ; 30(2): 532, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23504373

RESUMEN

The aim of the study was to investigate the influence of allelic imbalance (AI) in several loci of tumor suppressor genes in 3p region on the non-small cell lung cancer (NSCLC) development. We evaluated the frequency of loss of heterozygosity and/or microsatellite imbalance (LOH/MSI) and assessed their association with patients' characteristics (age, gender, tobacco addiction) and NSCLC classification according to TNM/AJCC staging. To analyze the potential role of AI involved in NSCLC pathogenesis, we allelotyped a group of 74 NSCLC patients using 7 microsatellite markers. The highest frequency of LOH/MSI, however, not statistically significant, was observed in RARß and MLH1 (p = 0.104 and p = 0.216, respectively) loci. The association between high LOH/MSI frequency in 3p region with male gender (p = 0.041) as well as with age (especially >60 years) for RARß and MLH1 genes (p = 0.0001 and p = 0.020, respectively) was documented. Statistically significant increased frequency of MLH1 allelic loss in squamous cell carcinoma (SCC) versus non-squamous cell carcinoma (non-SCC) was observed (p = 0.01). Significant increase in LOH/MSI frequency in 3p region (mainly in FHIT and MLH1 loci) in correlation with cigarette addiction in a lifetime (≥40 years and ≥40 Pack Years) was also documented (p < 0.05). The highest LOH/MSI was revealed in RARß locus in IA tumors (p = 0.0001), while the similarly high allelic loss of MLH1 correlated with III A/B tumors (p = 0.0002), according to AJCC staging. The obtained results demonstrate that AI is influenced by tobacco smoking and seems to be vital in the molecular diagnosis of NSCLC, especially of SCC subtype.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Desequilibrio Alélico/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Sitios Genéticos/genética , Neoplasias Pulmonares/genética , Proteínas Nucleares/genética , Receptores de Ácido Retinoico/genética , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/deficiencia , Biomarcadores de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Femenino , Frecuencia de los Genes/genética , Humanos , Neoplasias Pulmonares/diagnóstico , Masculino , Persona de Mediana Edad , Homólogo 1 de la Proteína MutL , Proteínas Nucleares/deficiencia , Receptores de Ácido Retinoico/deficiencia
12.
Immunol Lett ; 146(1-2): 15-24, 2012 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-22546502

RESUMEN

Different studies have shown that retinoids and their receptors [retinoic acid receptors (RARs) and retinoid X receptors (RXRs)] have crucial effects on the differentiation and function of myeloid cells such as Dendritic cells (DCs) and the development of lymphoid tissue. However, the relationship between RARß expression and DCs has not been previously studied in vivo. This work examined the effect of decreased RARß expression on the number (and probably on differentiation) of splenic DCs and the structure of spleen using a conditional mouse that partially ablates floxed RARß gene (RARß(L-/L-) mice). Our results showed that RARß is expressed mainly in cells of the splenic White Pulp (WP) zone of Wild type mice. As expected, low levels of RARß expression were detected in the spleen of RARß(L-/L-) conditional mice. These results were consistent with a decrease in the population of splenic CD11c(+)MHC-II(+) cells. Histopathological analyses of conditional mice spleen indicated defects in cell organization and structure. The expression of Toll-like receptor 2 was also down-regulated in the spleen of these mice. These results suggest that RARß is involved in splenic cell organization as well as in the maintenance of splenic DCs population, indicating that RARß expression is important in homeostasis of immune system components.


Asunto(s)
Linaje de la Célula/inmunología , Células Dendríticas/inmunología , Expresión Génica/inmunología , Receptores de Ácido Retinoico/inmunología , Bazo/patología , Animales , Antígenos CD11/genética , Antígenos CD11/inmunología , Linaje de la Célula/genética , Células Dendríticas/metabolismo , Células Dendríticas/patología , Femenino , Citometría de Flujo , Homeostasis/genética , Homeostasis/inmunología , Inmunidad/genética , Inmunohistoquímica , Recuento de Linfocitos , Ratones , Ratones Noqueados , Receptores de Ácido Retinoico/deficiencia , Receptores de Ácido Retinoico/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/inmunología , Bazo/metabolismo , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/inmunología
13.
Endocrinology ; 153(1): 438-49, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22045663

RESUMEN

Vitamin A is instrumental to mammalian reproduction. Its metabolite, retinoic acid (RA), acts in a hormone-like manner through binding to and activating three nuclear receptor isotypes, RA receptor (RAR)α (RARA), RARß, and RARγ (RARG). Here, we show that 1) RARG is expressed by A aligned (A(al)) spermatogonia, as well as during the transition from A(al) to A(1) spermatogonia, which is known to require RA; and 2) ablation of Rarg, either in the whole mouse or specifically in spermatogonia, does not affect meiosis and spermiogenesis but impairs the A(al) to A(1) transition in the course of some of the seminiferous epithelium cycles. Upon ageing, this phenomenon yields seminiferous tubules containing only spermatogonia and Sertoli cells. Altogether, our findings indicate that RARG cell-autonomously transduces, in undifferentiated spermatogonia of adult testes, a RA signal critical for spermatogenesis. During the prepubertal spermatogenic wave, the loss of RARG function can however be compensated by RARA, as indicated by the normal timing of appearance of meiotic cells in Rarg-null testes. Accordingly, RARG- and RARA-selective agonists are both able to stimulate Stra8 expression in wild-type prepubertal testes. Interestingly, inactivation of Rarg does not impair expression of the spermatogonia differentiation markers Kit and Stra8, contrary to vitamin A deficiency. This latter observation supports the notion that the RA-signaling pathway previously shown to operate in Sertoli cells also participates in spermatogonia differentiation.


Asunto(s)
Receptores de Ácido Retinoico/metabolismo , Espermatogénesis/fisiología , Espermatogonias/citología , Espermatogonias/metabolismo , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Secuencia de Bases , Cartilla de ADN/genética , Regulación del Desarrollo de la Expresión Génica , Masculino , Meiosis , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Ácido Retinoico/deficiencia , Receptores de Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Espermatogénesis/genética , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Testículo/patología , Deficiencia de Vitamina A/metabolismo , Deficiencia de Vitamina A/patología , Receptor de Ácido Retinoico gamma
14.
Nat Med ; 17(4): 454-60, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21460849

RESUMEN

Heterotopic ossification consists of ectopic bone formation within soft tissues after surgery or trauma. It can have debilitating consequences, but there is no definitive cure. Here we show that heterotopic ossification was essentially prevented in mice receiving a nuclear retinoic acid receptor-γ (RAR-γ) agonist. Side effects were minimal, and there was no significant rebound effect. To uncover the mechanisms of these responses, we treated mouse mesenchymal stem cells with an RAR-γ agonist and transplanted them into nude mice. Whereas control cells formed ectopic bone masses, cells that had been pretreated with the RAR-γ agonist did not, suggesting that they had lost their skeletogenic potential. The cells became unresponsive to rBMP-2 treatment in vitro and showed decreases in phosphorylation of Smad1, Smad5 and Smad8 and in overall levels of Smad proteins. In addition, an RAR-γ agonist blocked heterotopic ossification in transgenic mice expressing activin receptor-like kinase-2 (ALK2) Q207D, a constitutively active form of the receptor that is related to ALK2 R206H found in individuals with fibrodysplasia ossificans progressiva. The data indicate that RAR-γ agonists are potent inhibitors of heterotopic ossification in mouse models and, thus, may also be effective against injury-induced and congenital heterotopic ossification in humans.


Asunto(s)
Osificación Heterotópica/tratamiento farmacológico , Receptores de Ácido Retinoico/agonistas , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Humanos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Noqueados , Ratones Mutantes , Ratones Desnudos , Ratones Transgénicos , Osificación Heterotópica/metabolismo , Osificación Heterotópica/patología , Receptores de Ácido Retinoico/deficiencia , Receptores de Ácido Retinoico/genética , Transducción de Señal/efectos de los fármacos , Receptor de Ácido Retinoico gamma
15.
Kidney Int ; 79(6): 624-634, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21150871

RESUMEN

All-trans retinoic acid protects against the development of HIV-associated nephropathy (HIVAN) in HIV-1 transgenic mice (Tg26). In vitro, all-trans retinoic acid inhibits HIV-induced podocyte proliferation and restores podocyte differentiation markers by activating its receptor-α (RARα). Here, we report that Am580, a water-soluble RARα-specific agonist, attenuated proteinuria, glomerosclerosis, and podocyte proliferation, and restored podocyte differentiation markers in kidneys of Tg26 mice. Furthermore, RARα-/- Tg26 mice developed more severe kidney and podocyte injury than did RARα+/- Tg26 mice. Am580 failed to ameliorate kidney injury in RARα-/- Tg26 mice, confirming our hypothesis that Am580 acts through RARα. Although the expression of RARα-target genes was suppressed in the kidneys of Tg26 mice and of patients with HIVAN, the expression of RARα in the kidney was not different between patients with HIVAN and minimal change disease. However, the tissue levels of retinoic acid were reduced in the kidney cortex and isolated glomeruli of Tg26 mice. Consistent with this, the expression of two key enzymes in the retinoic acid synthetic pathway, retinol dehydrogenase type 1 and 9, and the overall enzymatic activity for retinoic acid synthesis were significantly reduced in the glomeruli of Tg26 mice. Thus, a defect in the endogenous synthesis of retinoic acid contributes to loss of the protection by retinoic acid in HIVAN. Hence, RARα agonists may be potential agents for the treatment of HIVAN.


Asunto(s)
Nefropatía Asociada a SIDA/metabolismo , VIH-1/genética , Podocitos/metabolismo , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal , Nefropatía Asociada a SIDA/genética , Nefropatía Asociada a SIDA/patología , Nefropatía Asociada a SIDA/prevención & control , Nefropatía Asociada a SIDA/virología , Oxidorreductasas de Alcohol/metabolismo , Animales , Benzoatos/farmacología , Diferenciación Celular , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Glomerulonefritis/metabolismo , Glomerulonefritis/prevención & control , Glomerulonefritis/virología , Humanos , Hidroxiesteroide Deshidrogenasas/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Podocitos/efectos de los fármacos , Podocitos/patología , Podocitos/virología , Proteinuria/metabolismo , Proteinuria/prevención & control , Proteinuria/virología , Receptores de Ácido Retinoico/agonistas , Receptores de Ácido Retinoico/deficiencia , Receptores de Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico , Retinoides/metabolismo , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos , Tetrahidronaftalenos/farmacología , Factores de Tiempo
16.
Neuron ; 66(6): 908-20, 2010 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-20620876

RESUMEN

Abnormal signaling by retinoids or n-3 polyunsaturated fatty acids has been implicated in clinical depression. The converging point in activities of these two classes of molecules is transcriptional activation of retinoid X receptors (Rxr). We show here that ablation of Rxrgamma in mice leads to depressive-like behaviors including increased despair and anhedonia, which were accompanied by reduced expression of dopamine D2 receptor in the shell of nucleus accumbens (NAc) and altered serotonin signaling. While abnormal serotonin signaling is not sufficient to generate the depressive behaviors, increasing D2r expression by chronic fluoxetine (Prozac) treatment or adenoassociated virus type2 (AAV2) mediated expression of Rxrgamma or D2r in the NAc of Rxrgamma(-/-) mice normalizes depressive-like behaviors in Rxrgamma(-/-) animals. Conversely, NAc infusion of raclopride, a D2r antagonist prevents AAV2-Rxrgamma-mediated rescue of despair behaviors in Rxrgamma(-/-) mice. Combined, our data argue that control of NAc D2r expression is critical for Rxrgamma-mediated modulation of affective behaviors.


Asunto(s)
Dopamina/metabolismo , Regulación de la Expresión Génica/genética , Trastornos del Humor/genética , Receptor gamma X Retinoide/fisiología , Transducción de Señal/genética , Animales , Antidepresivos/uso terapéutico , Conducta Animal/fisiología , Recuento de Células/métodos , Dependovirus/fisiología , Modelos Animales de Enfermedad , Dopamina/genética , Antagonistas de Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Fluoxetina/uso terapéutico , Preferencias Alimentarias/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Haloperidol/farmacología , Pérdida de Tono Postural/fisiología , Ratones , Ratones Noqueados , Trastornos del Humor/tratamiento farmacológico , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de Ácido Retinoico/deficiencia , Receptor gamma X Retinoide/deficiencia , Serotonina/metabolismo
17.
Dev Cell ; 18(3): 480-5, 2010 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-20230754

RESUMEN

In many experimental models and clinical examples, defects in the differentiation of the second heart field (SHF) and heart outflow tract septation defects are combined, although the mechanistic basis for this relationship has been unclear. We found that as the initial SHF population incorporates into the outflow tract, it is replenished from the surrounding progenitor territory. In retinoic acid (RA) receptor mutant mice, this latter process fails, and the outflow tract is shortened and misaligned as a result. As an additional consequence, the outflow tract is misspecified along its proximal-distal axis, which results in ectopic expression of TGFbeta2 and ectopic mesenchymal transformation of the endocardium. Reduction of TGFbeta2 gene dosage in the RA receptor-deficient background restores septation but does not rescue alignment defects, indicating that excess TGFbeta causes septation defects. This may be a common pathogenic pathway when second heart field and septation defects are coupled.


Asunto(s)
Corazón/embriología , Miocardio/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo , Tretinoina/metabolismo , Animales , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Femenino , Dosificación de Gen , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Embarazo , Receptores de Ácido Retinoico/deficiencia , Receptores de Ácido Retinoico/genética , Transducción de Señal , Factor de Crecimiento Transformador beta2/genética
18.
Microsc Res Tech ; 73(6): 583-96, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19937743

RESUMEN

Retinoic acid receptor alpha (RARalpha)-deficient mice are sterile, with abnormalities in the progression of spermatogenesis and spermiogenesis. In this study, we investigated whether defective retinoid signaling involved at least in part, disrupted cell-cell interactions. Hypertonic fixation approaches revealed defects in the integrity of the Sertoli-cell barrier in the tubules of RARalpha-deficient testes. Dye transfer experiments further revealed that coupling between cells from the basal to adluminal compartments was aberrant. There were also differences in the expression of several known retinoic acid (RA)-responsive genes encoding structural components of tight junctions and gap junctions. Immunostaining demonstrated a delay in the incorporation of zonula occludens (ZO-1), a peripheral component protein of tight junctions, into the Sertoli cell tight junctions. Markedly reduced expression of connexin-40 in mutant pachytene spermatocytes and round spermatids was found by in situ hybridization. An ectopic distribution of vimentin and disrupted cyclic expression of vimentin, which is usually tightly regulated during spermiogenesis, was found in RARalpha-deficient testes at all ages examined. Thus, the specific defects in spermiogenesis in RARalpha-deficient testes may correlate with a disrupted cyclic expression of RA-responsive structural components, including vimentin, a downregulation of connexin-40 in spermatogenic cells, and delayed assembly of ZO-1 into Sertoli cell tight junctions. Interestingly, bioinformatic analysis revealed that many genes that are components of tight junctions and gap junctions contained potential retinoic acid response element binding sites.


Asunto(s)
Conexinas/metabolismo , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Receptores de Ácido Retinoico/deficiencia , Espermatogénesis , Animales , Regulación de la Expresión Génica , Masculino , Ratones , Receptor alfa de Ácido Retinoico , Células de Sertoli/química , Espermatocitos/química , Vimentina/metabolismo , Proteína de la Zonula Occludens-1 , Proteína alfa-5 de Unión Comunicante
19.
J Immunol ; 183(4): 2217-21, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19635901

RESUMEN

During fetal life, CD4(+)CD3(-) lymphoid tissue inducer (LTi) cells are required for lymph node and Peyer's patch development in mice. In adult animals, CD4(+)CD3(-) cells are found in low numbers in lymphoid organs. Whether adult CD4(+)CD3(-) cells are LTi cells and are generated and maintained through cytokine signals has not been directly addressed. In this study we show that adult CD4(+)CD3(-) cells adoptively transferred into neonatal CXCR5(-/-) mice induced the formation of intestinal lymphoid tissues, demonstrating for the first time their bona fide LTi function. Increasing IL-7 availability in wild-type mice either by IL-7 transgene expression or treatment with IL-7/anti-IL-7 complexes increased adult LTi cell numbers through de novo generation from bone marrow cells and increased the survival and proliferation of LTi cells. Our observations demonstrate that adult CD4(+)lineage(-) cells are LTi cells and that the availability of IL-7 determines the size of the adult LTi cell pool.


Asunto(s)
Diferenciación Celular/inmunología , Interleucina-7/fisiología , Linfangiogénesis/inmunología , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Receptores de Ácido Retinoico/biosíntesis , Receptores de Hormona Tiroidea/biosíntesis , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Animales , Complejo CD3/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/genética , Células Cultivadas , Feto/citología , Feto/inmunología , Feto/metabolismo , Interleucina-7/biosíntesis , Interleucina-7/deficiencia , Linfangiogénesis/genética , Tejido Linfoide/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares , Receptores de Ácido Retinoico/deficiencia , Receptores de Ácido Retinoico/fisiología , Receptores de Hormona Tiroidea/deficiencia , Receptores de Hormona Tiroidea/fisiología , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Subgrupos de Linfocitos T/citología
20.
Blood ; 114(3): 600-7, 2009 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-19458357

RESUMEN

Specific genetic alterations in multiple myeloma (MM) may cause more aggressive diseases. Paired gene array analysis on 51 samples showed that retinoic acid (RA) receptor alpha (RARalpha) expression significantly increased at relapse compared with diagnosis. RARalpha encodes 2 major isoforms: RARalpha1 and RARalpha2. In this study, we examined the function of RARalpha2 in MM. Reverse transcription-polymerase chain reaction (RT-PCR) revealed ubiquitous RARalpha1 expression in MM cells, but RARalpha2 was expressed in 26 (32%) of 80 newly diagnosed patients and 10 (28%) of 36 MM cell lines. Patients with RARalpha2 expression had a significantly shorter overall survival on identical treatments. The presence of RARalpha2 remained significant on multivariate analysis. Knockdown of RARalpha2 but not RARalpha1 induced significant MM cell death and growth inhibition, and overexpressing RARalpha2 activated STAT3 and mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) signaling pathways. Interestingly, all-trans retinoic acid (ATRA) treatment induced potent cell death and growth inhibition in RARalpha2(+) but not RARalpha2(-) MM cells; overexpressing RARalpha2 in RARalpha2-deficient MM cells restored sensitivity to ATRA. Furthermore, ATRA treatment significantly inhibited the growth of RARalpha2-overexpressing MM tumors in severe combined immunodeficiency (SCID) mouse model. These findings provide a rationale for RA-based therapy in aggressive RARalpha2(+) MM.


Asunto(s)
Mieloma Múltiple/química , Mieloma Múltiple/tratamiento farmacológico , Valor Predictivo de las Pruebas , Receptores de Ácido Retinoico/genética , Tretinoina/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Resistencia a Antineoplásicos/genética , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones SCID , Mieloma Múltiple/mortalidad , Mieloma Múltiple/patología , Isoformas de Proteínas , Receptores de Ácido Retinoico/análisis , Receptores de Ácido Retinoico/deficiencia , Recurrencia , Receptor alfa de Ácido Retinoico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Tasa de Supervivencia , Tretinoina/uso terapéutico , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...