Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(22)2021 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-34830455

RESUMEN

Chronic myeloid leukemia (CML) is a hematopoietic malignancy characterized by the presence of the BCR-ABL oncogene. Therapeutic regimens with tyrosine kinase inhibitors (TKIs) specifically targeting BCR-ABL have greatly improved overall survival of CML. However, drug intolerance and related toxicity remain. Combined therapy is effective in reducing drug magnitude while increasing therapeutic efficacy and, thus, lowers undesired adverse side effects. The p38 MAPK activity is critically linked to the pathogenesis of a number of diseases including hematopoietic diseases; however, the role of each isozyme in CML and TKI-mediated effects is still elusive. In this study, we used specific gene knockdown to clearly demonstrate that the deficiency of p38α greatly enhanced the therapeutic efficacy in growth suppression and cytotoxicity of TKIs, first-generation imatinib, and second generation dasatinib by approximately 2.5-3.0-fold in BCR-ABL-positive CML-derived leukemia K562 and KMB5 cells. Knockdown of p38ß, which displays the most sequence similarity to p38α, exerted distinct and opposite effects on the TKI-mediated therapeutic efficacy. These results show the importance of isotype-specific intervention in enhancing the therapeutic efficacy of TKI. A highly specific p38α inhibitor, TAK715, also significantly enhanced the imatinib- and dasatinib-mediated therapeutic efficacy, supporting the feasibility of p38α deficiency in future clinic application. Taken together, our results demonstrated that p38α is a promising target for combined therapy with BCR-ABL-targeting tyrosine kinase inhibitors for future application to increase therapeutic efficacy.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Proteínas de Fusión bcr-abl/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Proteína Quinasa 14 Activada por Mitógenos/genética , Terapia Combinada , Dasatinib/farmacología , Resistencia a Antineoplásicos/genética , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Técnicas de Silenciamiento del Gen , Terapia Genética , Humanos , Mesilato de Imatinib/farmacología , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Inhibidores de Proteínas Quinasas/farmacología
2.
J Clin Invest ; 130(10): 5287-5301, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32573492

RESUMEN

In the mammalian heart, the left ventricle (LV) rapidly becomes more dominant in size and function over the right ventricle (RV) after birth. The molecular regulators responsible for this chamber-specific differential growth are largely unknown. We found that cardiomyocytes in the neonatal mouse RV had lower proliferation, more apoptosis, and a smaller average size compared with the LV. This chamber-specific growth pattern was associated with a selective activation of p38 mitogen-activated protein kinase (MAPK) activity in the RV and simultaneous inactivation in the LV. Cardiomyocyte-specific deletion of both the Mapk14 and Mapk11 genes in mice resulted in loss of p38 MAPK expression and activity in the neonatal heart. Inactivation of p38 activity led to a marked increase in cardiomyocyte proliferation and hypertrophy but diminished cardiomyocyte apoptosis, specifically in the RV. Consequently, the p38-inactivated hearts showed RV-specific enlargement postnatally, progressing to pulmonary hypertension and right heart failure at the adult stage. Chamber-specific p38 activity was associated with differential expression of dual-specific phosphatases (DUSPs) in neonatal hearts, including DUSP26. Unbiased transcriptome analysis revealed that IRE1α/XBP1-mediated gene regulation contributed to p38 MAPK-dependent regulation of neonatal cardiomyocyte proliferation and binucleation. These findings establish an obligatory role of DUSP/p38/IRE1α signaling in cardiomyocytes for chamber-specific growth in the postnatal heart.


Asunto(s)
Corazón/crecimiento & desarrollo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Miocardio/enzimología , Animales , Animales Recién Nacidos , Apoptosis , Proliferación Celular , Tamaño de la Célula , Activación Enzimática , Femenino , Perfilación de la Expresión Génica , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/enzimología , Ventrículos Cardíacos/crecimiento & desarrollo , Masculino , Ratones , Ratones Noqueados , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Proteína Quinasa 14 Activada por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/deficiencia , Proteínas Quinasas Activadas por Mitógenos/genética , Miocardio/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/enzimología , Especificidad de Órganos , Remodelación Vascular/genética , Remodelación Vascular/fisiología
3.
FASEB J ; 34(7): 9628-9649, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32475008

RESUMEN

Alzheimer's disease (AD) is the leading cause of dementia with very limited therapeutic options. Amyloid ß (Aß) and phosphorylated Tau (p-Tau) are key pathogenic molecules in AD. P38α-MAPK is specifically activated in AD lesion sites. However, its effects on AD pathogenesis, especially on p-Tau-associated brain pathology, and the underlying molecular mechanisms remain unclear. We mated human APP-transgenic mice and human P301S Tau-transgenic mice with mapk14-floxed and neuron-specific Cre-knock-in mice. We observed that deletion of p38α-MAPK specifically in neurons improves the cognitive function of both 9-month-old APP and Tau-transgenic AD mice, which is associated with decreased Aß and p-Tau load in the brain. We further used next-generation sequencing to analyze the gene transcription in brains of p38α-MAPK deficient and wild-type APP-transgenic mice, which indicated that deletion of p38α-MAPK regulates the transcription of calcium homeostasis-related genes, especially downregulates the expression of grin2a, a gene encoding NMDAR subunit NR2A. Cell culture experiments further verified that deletion of p38α-MAPK inhibits NMDA-triggered calcium influx and neuronal apoptosis. Our systemic studies of AD pathogenic mechanisms using both APP- and Tau-transgenic mice suggested that deletion of neuronal p38α-MAPK attenuates AD-associated brain pathology and protects neurons in AD pathogenesis. This study supports p38α-MAPK as a novel target for AD therapy.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Trastornos del Conocimiento/prevención & control , Modelos Animales de Enfermedad , Inflamación/prevención & control , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Neuronas/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/patología , Femenino , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Proteínas tau/genética
4.
J Neuroinflammation ; 16(1): 98, 2019 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-31077217

RESUMEN

Traumatic brain injury (TBI) is a significant cause of morbidity and mortality in the USA and other developed countries worldwide. Following the initial mechanical insult, the brain's primary innate immune effector, microglia, initiate inflammatory signaling cascades and pathophysiological responses that can lead to chronic neuroinflammation and neurodegenerative sequelae. The p38α MAPK signaling pathway in microglia is a key contributor to inflammatory responses to diverse disease-relevant stressors and injury conditions. Therefore, we tested here whether microglia p38α contributes to acute and persistent inflammatory responses induced by a focal TBI. We generated conditional cell-specific knockout of p38α in microglia using a CX3CR1 Cre-lox system, subjected the p38α knockout and wild-type mice to a controlled cortical impact TBI, and measured inflammatory responses at acute (1-day) and subacute (7-day) post-injury time points. We found that deletion of p38α in microglia only was sufficient to attenuate multiple pro-inflammatory responses following TBI, notably reducing pro-inflammatory cytokine/chemokine production and recruitment of inflammatory monocytes into the brain and preventing the persistent microglial morphological activation. These data provide strong evidence supporting a role for microglial p38α in propagation of a chronic and potentially neurotoxic pro-inflammatory environment in the brain following TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/prevención & control , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Microglía/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Animales , Lesiones Traumáticas del Encéfalo/patología , Inflamación/metabolismo , Inflamación/patología , Inflamación/prevención & control , Ratones , Ratones Noqueados , Ratones Transgénicos , Microglía/patología , Proteína Quinasa 14 Activada por Mitógenos/genética
5.
Cell Death Dis ; 10(6): 376, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31092814

RESUMEN

Apoptosis and senescence are two mutually exclusive cell fate programs that can be activated by stress. The factors that instruct cells to enter into senescence or apoptosis are not fully understood, but both programs can be regulated by the stress kinase p38α. Using an inducible system that specifically activates this pathway, we show that sustained p38α activation suffices to trigger massive autophagosome formation and to enhance the basal autophagic flux. This requires the concurrent effect of increased mitochondrial reactive oxygen species production and the phosphorylation of the ULK1 kinase on Ser-555 by p38α. Moreover, we demonstrate that macroautophagy induction by p38α signaling determines that cancer cells preferentially enter senescence instead of undergoing apoptosis. In agreement with these results, we present evidence that the induction of autophagy by p38α protects cancer cells from chemotherapy-induced apoptosis by promoting senescence. Our results identify a new mechanism of p38α-regulated basal autophagy that controls the fate of cancer cells in response to stress.


Asunto(s)
Autofagia , Senescencia Celular , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Homólogo de la Proteína 1 Relacionada con la Autofagia/antagonistas & inhibidores , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Línea Celular Tumoral , Senescencia Celular/efectos de los fármacos , Doxorrubicina/farmacología , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , MAP Quinasa Quinasa 6/antagonistas & inhibidores , MAP Quinasa Quinasa 6/genética , MAP Quinasa Quinasa 6/metabolismo , Mitocondrias/metabolismo , Proteína Quinasa 12 Activada por Mitógenos/deficiencia , Proteína Quinasa 12 Activada por Mitógenos/genética , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Proteína Quinasa 14 Activada por Mitógenos/genética , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína Sequestosoma-1/genética , Proteína Sequestosoma-1/metabolismo , Transducción de Señal
6.
FEBS J ; 284(24): 4200-4215, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29052963

RESUMEN

Growing evidence suggests that hepatic macrophages play an important role in tissue repair after liver injury by coordinating the induction and resolution of inflammation, removing apoptotic cells, and promoting hepatocyte proliferation. Understanding the role of macrophages in the pathogenesis of liver injury will help pave the way to future therapeutics. Here, we investigated whether macrophage p38α plays a regulatory role in the tissue repair following d-galactosamine (GalN)/tumor necrosis factor-α (TNF-α)-induced acute liver injury. We found that macrophage p38α-deficient mice displayed decreased mortality and relieved liver injury as evident from less apoptosis, accelerated regeneration, decreased granulocytes recruitment, monocytes infiltration, and cytokine production after GalN/TNF-α treatment. Mechanistically, we found that p38 signaling was activated by lipopolysaccharide/interferon-γ treatment but not by inteleukin-4 stimulation, while pharmaceutical inhibition of p38α induced a shift in polarization from M1 macrophages to M2 macrophages. Together, our results indicated that macrophage p38α signaling is involved in the pathogenesis of liver injury induced by GalN/TNF-α, and inhibition of p38α signaling in macrophage could ameliorate liver injury and accelerate regeneration, probably by promoting the polarization of macrophages from the M1 phenotype to the M2 phenotype.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/enzimología , Galactosamina/toxicidad , Regeneración Hepática/fisiología , Macrófagos/enzimología , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Factor de Necrosis Tumoral alfa/toxicidad , Animales , Antígenos de Diferenciación/biosíntesis , Antígenos de Diferenciación/genética , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Citocinas/biosíntesis , Citocinas/genética , Células Hep G2 , Hepatocitos/efectos de los fármacos , Humanos , Inmunofenotipificación , Interferón gamma/farmacología , Dosificación Letal Mediana , Lipopolisacáridos/farmacología , Regeneración Hepática/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/clasificación , Masculino , Ratones , Ratones Noqueados , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 14 Activada por Mitógenos/fisiología , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Células RAW 264.7
7.
Arterioscler Thromb Vasc Biol ; 37(12): e185-e196, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28982666

RESUMEN

OBJECTIVE: MAPKs (mitogen-activated protein kinases), especially p38, play detrimental roles in cardiac diseases and cardiac remodeling post-myocardial infarction. However, the activation and function of MAPKs in coronary thrombosis in vivo and its relationship with clinical outcomes remain poorly understood. APPROACH AND RESULTS: Here, we showed that p38α was the major isoform expressed in human and mouse platelets. Platelet-specific p38α-deficient mice presented impaired thrombosis and hemostasis but had improved cardiac function, reduced infarct size, decreased inflammatory response, and microthrombus in a left anterior descending artery ligation model. Signaling analysis revealed that p38 activation was one of the earliest events in platelets after treatment with receptor agonists or reactive oxygen species. p38α/MAPK-activated protein kinase 2/heat shock protein 27 and p38α/cytosolic phospholipases A2 were the major pathways regulating receptor-mediated or hydrogen peroxide-induced platelet activation in an ischemic environment. Moreover, the distinct roles of ERK1/2 (extracellular signal-regulated kinase) in receptor- or reactive oxygen species-induced p38-mediated platelet activation reflected the complicated synergistic relationships among MAPKs. Analysis of clinical samples revealed that MAPKs were highly phosphorylated in platelets from preoperative patients with ST-segment-elevation myocardial infarction, and increased phosphorylation of p38 was associated with no-reflow outcomes. CONCLUSIONS: We conclude that p38α serves as a critical regulator of platelet activation and potential indicator of highly thrombotic lesions and no-reflow, and inhibition of platelet p38α may improve clinical outcomes in subjects with ST-segment-elevation myocardial infarction.


Asunto(s)
Plaquetas/enzimología , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Activación Plaquetaria , Infarto del Miocardio con Elevación del ST/enzimología , Función Ventricular Izquierda , Adulto , Anciano , Anciano de 80 o más Años , Animales , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Activación Enzimática , Femenino , Genotipo , Proteínas de Choque Térmico HSP27/metabolismo , Hemostasis , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Proteína Quinasa 14 Activada por Mitógenos/sangre , Proteína Quinasa 14 Activada por Mitógenos/genética , Contracción Miocárdica , Miocardio/metabolismo , Miocardio/patología , Fenómeno de no Reflujo/sangre , Fenómeno de no Reflujo/enzimología , Fenómeno de no Reflujo/fisiopatología , Fenotipo , Fosfolipasas A2 Citosólicas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Infarto del Miocardio con Elevación del ST/sangre , Infarto del Miocardio con Elevación del ST/patología , Infarto del Miocardio con Elevación del ST/fisiopatología , Transducción de Señal , Volumen Sistólico , Trombosis/sangre , Trombosis/enzimología
8.
BMC Bioinformatics ; 18(Suppl 7): 260, 2017 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-28617232

RESUMEN

BACKGROUND: Transcription factor (TF) networks play a key role in controlling the transfer of genetic information from gene to mRNA. Much progress has been made on understanding and reverse-engineering TF network topologies using a range of experimental and theoretical methodologies. Less work has focused on using these models to examine how TF networks respond to changes in the cellular environment. METHODS: In this paper, we have developed a simple, pragmatic methodology, TIGERi (Transcription-factor-activity Illustrator for Global Explanation of Regulatory interaction), to model the response of an inferred TF network to changes in cellular environment. The methodology was tested using publicly available data comparing gene expression profiles of a mouse p38α (Mapk14) knock-out line to the original wild-type. RESULTS: Using the model, we have examined changes in the TF network resulting from the presence or absence of p38α. A part of this network was confirmed by experimental work in the original paper. Additional relationships were identified by our analysis, for example between p38α and HNF3, and between p38α and SOX9, and these are strongly supported by published evidence. FXR and MYC were also discovered in our analysis as two novel links of p38α. To provide a computational methodology to the biomedical communities that has more user-friendly interface, we also developed a standalone GUI (graphical user interface) software for TIGERi and it is freely available at https://github.com/namshik/tigeri/ . CONCLUSIONS: We therefore believe that our computational approach can identify new members of networks and new interactions between members that are supported by published data but have not been integrated into the existing network models. Moreover, ones who want to analyze their own data with TIGERi could use the software without any command line experience. This work could therefore accelerate researches in transcriptional gene regulation in higher eukaryotes.


Asunto(s)
Aprendizaje Automático , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Redes Reguladoras de Genes , Ratones , Ratones Noqueados , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Proteína Quinasa 14 Activada por Mitógenos/genética , Factores de Transcripción/química , Factores de Transcripción/genética , Transcriptoma
9.
Sci Rep ; 7: 45306, 2017 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-28361984

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by a severe and progressive neuronal loss leading to cognitive dysfunctions. Previous reports, based on the use of chemical inhibitors, have connected the stress kinase p38α to neuroinflammation, neuronal death and synaptic dysfunction. To explore the specific role of neuronal p38α signalling in the appearance of pathological symptoms, we have generated mice that combine expression of the 5XFAD transgenes to induce AD symptoms with the downregulation of p38α only in neurons (5XFAD/p38α∆-N). We found that the neuronal-specific deletion of p38α improves the memory loss and long-term potentiation impairment induced by 5XFAD transgenes. Furthermore, 5XFAD/p38α∆-N mice display reduced amyloid-ß accumulation, improved neurogenesis, and important changes in brain cytokine expression compared with 5XFAD mice. Our results implicate neuronal p38α signalling in the synaptic plasticity dysfunction and memory impairment observed in 5XFAD mice, by regulating both amyloid-ß deposition in the brain and the relay of this accumulation to mount an inflammatory response, which leads to the cognitive deficits.


Asunto(s)
Enfermedad de Alzheimer/psicología , Péptidos beta-Amiloides/metabolismo , Disfunción Cognitiva/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/genética , Neuronas/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Eliminación de Gen , Humanos , Ratones , Ratones Transgénicos , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Plasticidad Neuronal , Neuronas/citología , Transducción de Señal
10.
Sci Rep ; 7: 45964, 2017 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-28382965

RESUMEN

Bone mass is determined by the balance between bone formation, carried out by mesenchymal stem cell-derived osteoblasts, and bone resorption, carried out by monocyte-derived osteoclasts. Here we investigated the potential roles of p38 MAPKs, which are activated by growth factors and cytokines including RANKL and BMPs, in osteoclastogenesis and bone resorption by ablating p38α MAPK in LysM+monocytes. p38α deficiency promoted monocyte proliferation but regulated monocyte osteoclastic differentiation in a cell-density dependent manner, with proliferating p38α-/- cultures showing increased differentiation. While young mutant mice showed minor increase in bone mass, 6-month-old mutant mice developed osteoporosis, associated with an increase in osteoclastogenesis and bone resorption and an increase in the pool of monocytes. Moreover, monocyte-specific p38α ablation resulted in a decrease in bone formation and the number of bone marrow mesenchymal stem/stromal cells, likely due to decreased expression of PDGF-AA and BMP2. The expression of PDGF-AA and BMP2 was positively regulated by the p38 MAPK-Creb axis in osteoclasts, with the promoters of PDGF-AA and BMP2 having Creb binding sites. These findings uncovered the molecular mechanisms by which p38α MAPK regulates osteoclastogenesis and coordinates osteoclastogenesis and osteoblastogenesis.


Asunto(s)
Envejecimiento/metabolismo , Remodelación Ósea , Diferenciación Celular , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Osteoclastos/citología , Células Madre/citología , Animales , Proteína Morfogenética Ósea 2/metabolismo , Resorción Ósea/patología , Recuento de Células , Proliferación Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Integrasas/metabolismo , Masculino , Ratones , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Monocitos/citología , Osteogénesis , Osteoporosis/diagnóstico por imagen , Osteoporosis/enzimología , Osteoporosis/patología , Fenotipo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Microtomografía por Rayos X
11.
Circulation ; 136(6): 549-561, 2017 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-28356446

RESUMEN

BACKGROUND: In the heart, acute injury induces a fibrotic healing response that generates collagen-rich scarring that is at first protective but if inappropriately sustained can worsen heart disease. The fibrotic process is initiated by cytokines, neuroendocrine effectors, and mechanical strain that promote resident fibroblast differentiation into contractile and extracellular matrix-producing myofibroblasts. The mitogen-activated protein kinase p38α (Mapk14 gene) is known to influence the cardiac injury response, but its direct role in orchestrating programmed fibroblast differentiation and fibrosis in vivo is unknown. METHODS: A conditional Mapk14 allele was used to delete the p38α encoding gene specifically in cardiac fibroblasts or myofibroblasts with 2 different tamoxifen-inducible Cre recombinase-expressing gene-targeted mouse lines. Mice were subjected to ischemic injury or chronic neurohumoral stimulation and monitored for survival, cardiac function, and fibrotic remodeling. Antithetically, mice with fibroblast-specific transgenic overexpression of activated mitogen-activated protein kinase kinase 6, a direct inducer of p38, were generated to investigate whether this pathway can directly drive myofibroblast formation and the cardiac fibrotic response. RESULTS: In mice, loss of Mapk14 blocked cardiac fibroblast differentiation into myofibroblasts and ensuing fibrosis in response to ischemic injury or chronic neurohumoral stimulation. A similar inhibition of myofibroblast formation and healing was also observed in a dermal wounding model with deletion of Mapk14. Transgenic mice with fibroblast-specific activation of mitogen-activated protein kinase kinase 6-p38 developed interstitial and perivascular fibrosis in the heart, lung, and kidney as a result of enhanced myofibroblast numbers. Mechanistic experiments show that p38 transduces cytokine and mechanical signals into myofibroblast differentiation through the transcription factor serum response factor and the signaling effector calcineurin. CONCLUSIONS: These findings suggest that signals from diverse modes of injury converge on p38α mitogen-activated protein kinase within the fibroblast to program the fibrotic response and myofibroblast formation in vivo, suggesting a novel therapeutic approach with p38 inhibitors for future clinical application.


Asunto(s)
Fibroblastos/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/genética , Actinas/metabolismo , Alelos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular , Células Cultivadas , Citocinas/metabolismo , Fibroblastos/citología , Fibrosis , Ventrículos Cardíacos/diagnóstico por imagen , Isquemia/etiología , Isquemia/metabolismo , Isquemia/patología , Riñón/metabolismo , Riñón/patología , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Miocardio/metabolismo , Miocardio/patología , Miofibroblastos/citología , Miofibroblastos/metabolismo , Transducción de Señal
12.
Behav Brain Res ; 322(Pt B): 212-222, 2017 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-27765672

RESUMEN

A major aspect of mammalian aging is the decline in functional competence of many self-renewing cell types, including adult-born neuronal precursors. Since age-related senescence of self-renewal occurs simultaneously with chronic up-regulation of the p38MAPKalpha (p38α) signaling pathway, we used the dominant negative mouse model for attenuated p38α activity (DN-p38αAF/+) in which Thr180 and Tyr182 are mutated (T→A/Y→F) to prevent phosphorylation activation (DN-p38αAF/+) and kinase activity. As a result, aged DN-p38αAF/+ mice are resistant to age-dependent decline in proliferation and regeneration of several peripheral tissue progenitors when compared to wild-type littermates. Aging is the major risk factor for non-inherited forms of Alzheimer's disease (AD); environmental and genetic risk factors that accelerate the senescence phenotype are thought to contribute to an individual's relative risk. In the present study, we evaluated aged DN-p38αAF/+ and wildtype littermates in a series of behavioral paradigms to test if p38α mutant mice exhibit altered baseline abnormalities in neurological reflexes, locomotion, anxiety-like behavior, and age-dependent cognitive decline. While aged DN-p38αAF/+ and wildtype littermates appear equal in all tested baseline neurological and behavioral parameters, DN-p38αAF/+ exhibit superior context discrimination fear conditioning. Context discrimination is a cognitive task that is supported by proliferation and differentiation of adult-born neurons in the dentate gyrus of the hippocampus. Consistent with enhanced context discrimination in aged DN-p38αAF/+, we discovered enhanced production of adult-born neurons in the dentate gyrus of DN-p38αAF/+ mice compared to wildtype littermates. Our findings support the notion that p38α inhibition has therapeutic utility in aging diseases that affect cognition, such as AD.


Asunto(s)
Envejecimiento/metabolismo , Envejecimiento/psicología , Discriminación en Psicología/fisiología , Miedo/fisiología , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Neurogénesis/fisiología , Envejecimiento/patología , Análisis de Varianza , Animales , Ansiedad/enzimología , Ansiedad/patología , Condicionamiento Psicológico/fisiología , Electrochoque , Conducta Exploratoria/fisiología , Miedo/psicología , Femenino , Reacción Cataléptica de Congelación/fisiología , Hipocampo/enzimología , Hipocampo/patología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Quinasa 14 Activada por Mitógenos/genética , Neuronas/enzimología , Neuronas/patología , Pruebas Psicológicas
13.
Skelet Muscle ; 6: 9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26981231

RESUMEN

BACKGROUND: Extracellular stimuli induce gene expression responses through intracellular signaling mediators. The p38 signaling pathway is a paradigm of the mitogen-activated protein kinase (MAPK) family that, although originally identified as stress-response mediator, contributes to establishing stem cell differentiation fates. p38α is central for induction of the differentiation fate of the skeletal muscle stem cells (satellite cells) through not fully characterized mechanisms. METHODS: To investigate the global gene transcription program regulated by p38α during satellite cell differentiation (myogenesis), and to specifically address whether this regulation occurs through direct action of p38α on gene promoters, we performed a combination of microarray gene expression and genome-wide binding analyses. For experimental robustness, two myogenic cellular systems with genetic and chemical loss of p38α function were used: (1) satellite cells derived from mice with muscle-specific deletion of p38α, and (2) the C2C12 murine myoblast cell line cultured in the absence or presence of the p38α/ß inhibitor SB203580. Analyses were performed at cell proliferation and early differentiation stages. RESULTS: We show that p38α binds to a large set of active promoters during the transition of myoblasts from proliferation to differentiation stages. p38α-bound promoters are enriched with binding motifs for several transcription factors, with Sp1, Tcf3/E47, Lef1, FoxO4, MyoD, and NFATc standing out in all experimental conditions. p38α association with chromatin correlates very well with high levels of transcription, in agreement with its classical function as an activator of myogenic differentiation. Interestingly, p38α also associates with genes repressed at the onset of differentiation, thus highlighting the relevance of p38-dependent chromatin regulation for transcriptional activation and repression during myogenesis. CONCLUSIONS: These results uncover p38α association and function on chromatin at novel classes of target genes during skeletal muscle cell differentiation. This is consistent with this MAPK isoform being a transcriptional regulator.


Asunto(s)
Diferenciación Celular , Inmunoprecipitación de Cromatina , Cromatina/metabolismo , Perfilación de la Expresión Génica , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Desarrollo de Músculos , Células Satélite del Músculo Esquelético/enzimología , Animales , Sitios de Unión , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Genotipo , Ratones Noqueados , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Proteína Quinasa 14 Activada por Mitógenos/genética , Desarrollo de Músculos/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Regiones Promotoras Genéticas , Inhibidores de Proteínas Quinasas/farmacología , Células Satélite del Músculo Esquelético/efectos de los fármacos , Transducción de Señal , Transcripción Genética
14.
FASEB J ; 29(4): 1414-25, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25550462

RESUMEN

The skeleton acts as an endocrine organ that regulates energy metabolism and calcium and phosphorous homeostasis through the secretion of osteocalcin (Oc) and fibroblast growth factor 23 (FGF23). However, evidence suggests that osteoblasts secrete additional unknown factors that contribute to the endocrine function of bone. To search for these additional factors, we generated mice with a conditional osteoblast-specific deletion of p38α MAPK known to display profound defects in bone homeostasis. Herein, we show that impaired osteoblast function is associated with a strong decrease in body weight and adiposity (P < 0.01). The differences in adiposity were not associated with diminished caloric intake, but rather reflected 20% increased energy expenditure and the up-regulation of uncoupling protein-1 (Ucp1) in white adipose tissue (WAT) and brown adipose tissue (BAT) (P < 0.05). These alterations in lipid metabolism and energy expenditure were correlated with a decrease in the blood levels of neuropeptide Y (NPY) (40% lower) rather than changes in the serum levels of insulin, Oc, or FGF23. Among all Npy-expressing tissues, only bone and primary osteoblasts showed a decline in Npy expression (P < 0.01). Moreover, the intraperitoneal administration of recombinant NPY partially restored the WAT weight and adipocyte size of p38α-deficient mice (P < 0.05). Altogether, these results further suggest that, in addition to Oc, other bone-derived signals affect WAT and energy expenditure contributing to the regulation of energy metabolism.


Asunto(s)
Tejido Adiposo/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Osteoblastos/enzimología , Adipocitos/citología , Adipocitos/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Adiposidad , Animales , Peso Corporal , Desarrollo Óseo , Tamaño de la Célula , Metabolismo Energético , Femenino , Factor-23 de Crecimiento de Fibroblastos , Regulación de la Expresión Génica , Homeostasis , Canales Iónicos/genética , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Noqueados , Proteínas Mitocondriales/genética , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Proteína Quinasa 14 Activada por Mitógenos/genética , Neuropéptido Y/sangre , Neuropéptido Y/genética , Obesidad/enzimología , Obesidad/prevención & control , Osteocalcina/metabolismo , Embarazo , Transducción de Señal , Proteína Desacopladora 1 , Regulación hacia Arriba
15.
J Hepatol ; 62(6): 1319-27, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25595884

RESUMEN

BACKGROUND & AIMS: It is proposed that p38 is involved in gluconeogenesis, however, the genetic evidence is lacking and precise mechanisms remain poorly understood. We sought to delineate the role of hepatic p38α in gluconeogenesis during fasting by applying a loss-of-function genetic approach. METHODS: We examined fasting glucose levels, performed pyruvate tolerance test, imaged G6Pase promoter activity, as well as determined the expression of gluconeogenic genes in mice with a targeted deletion of p38α in liver. Results were confirmed both in vivo and in vitro by using an adenoviral dominant-negative form of p38α (p38α-AF) and the constitutively active mitogen-activated protein kinase 6, respectively. Adenoviral dominant-negative form of AMP-activated protein kinase α (DN-AMPKα) was employed to test our proposed model. RESULTS: Mice lacking hepatic p38α exhibited reduced fasting glucose level and impaired gluconeogenesis. Interestingly, hepatic deficiency of p38α did not result in an alteration in CREB phosphorylation, but led to an increase in AMPKα phosphorylation. Adenoviral DN-AMPKα could abolish the effect of p38α-AF on gluconeogenesis. Knockdown of up-steam transforming growth factor ß-activated kinase 1 decreased the AMPKα phosphorylation induced by p38α-AF, suggesting a negative feedback loop. Consistently, inverse correlations between p38 and AMPKα phosphorylation were observed during fasting and in diabetic mouse models. Importantly, adenoviral p38α-AF treatment ameliorated hyperglycemia in diabetic mice. CONCLUSIONS: Our study provides evidence that hepatic p38α functions as a negative regulator of AMPK signaling in maintaining gluconeogenesis, dysregulation of this regulatory network contributes to unrestrained gluconeogenesis in diabetes, and hepatic p38α could be a drug target for hyperglycemia.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Gluconeogénesis/fisiología , Hígado/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/genética , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Ayuno/metabolismo , Gluconeogénesis/genética , Glucosa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Proteína Quinasa 14 Activada por Mitógenos/genética , Fosforilación , ARN Mensajero/genética , ARN Mensajero/metabolismo
16.
J Neurosci ; 33(14): 6143-53, 2013 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-23554495

RESUMEN

Neuropathology after traumatic brain injury (TBI) is the result of both the immediate impact injury and secondary injury mechanisms. Unresolved post-traumatic glial activation is a secondary injury mechanism that contributes to a chronic state of neuroinflammation in both animal models of TBI and human head injury patients. We recently demonstrated, using in vitro models, that p38α MAPK signaling in microglia is a key event in promoting cytokine production in response to diverse disease-relevant stressors and subsequent inflammatory neuronal dysfunction. From these findings, we hypothesized that the p38α signaling pathway in microglia could be contributing to the secondary neuropathologic sequelae after a diffuse TBI. Mice where microglia were p38α-deficient (p38α KO) were protected against TBI-induced motor deficits and synaptic protein loss. In wild-type (WT) mice, diffuse TBI produced microglia morphological activation that lasted for at least 7 d; however, p38α KO mice failed to activate this response. Unexpectedly, we found that the peak of the early, acute phase cytokine and chemokine levels was increased in injured p38α KO mice compared with injured WT mice. The increased cytokine levels in the p38α KO mice could not be accounted for by more infiltration of macrophages or neutrophils, or increased astrogliosis. By 7 d after injury, the cytokine and chemokine levels remained elevated in injured WT mice but not in p38α KO mice. Together, these data suggest that p38α balances the inflammatory response by acutely attenuating the early proinflammatory cytokine surge while perpetuating the chronic microglia activation after TBI.


Asunto(s)
Lesiones Encefálicas/patología , Encéfalo/patología , Citocinas/metabolismo , Regulación de la Expresión Génica/genética , Microglía/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Análisis de Varianza , Animales , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/genética , Lesiones Encefálicas/metabolismo , Proteínas de Unión al Calcio/metabolismo , Citocinas/genética , Modelos Animales de Enfermedad , Gliosis/etiología , Gliosis/genética , Antígenos Comunes de Leucocito/metabolismo , Macrófagos/patología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Microglía/patología , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Actividad Motora , Trastornos del Movimiento/etiología , Trastornos del Movimiento/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neutrófilos/patología , Método Simple Ciego , Factores de Tiempo
17.
Hepatology ; 57(5): 1950-61, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23354775

RESUMEN

UNLABELLED: p38α mitogen-activated protein kinases (MAPK) may be essential in the up-regulation of proinflammatory cytokines and can be activated by transforming growth factor ß, tumor necrosis factor-α, interleukin-1ß, and oxidative stress. p38 MAPK activation results in hepatocyte growth arrest, whereas increased proliferation has been considered a hallmark of p38α-deficient cells. Our aim was to assess the role of p38α in the progression of biliary cirrhosis induced by chronic cholestasis as an experimental model of chronic inflammation associated with hepatocyte proliferation, apoptosis, oxidative stress, and fibrogenesis. Cholestasis was induced in wildtype and liver-specific p38α knockout mice by bile duct ligation and animals were sacrificed at 12 and 28 days. p38α knockout mice exhibited a 50% decrease in mean life-span after cholestasis induction. MK2 phosphorylation was markedly reduced in liver of p38α-deficient mice upon chronic cholestasis. Hepatocyte growth was reduced and hepatomegaly was absent in p38α-deficient mice during chronic cholestasis through down-regulation of both AKT and mammalian target of rapamycin. Cyclin D1 and cyclin B1 were up-regulated in liver of p38α-deficient mice upon chronic cholestasis, but unexpectedly proliferating cell nuclear antigen was down-regulated at 12 days after cholestasis induction and the mitotic index was very high upon cholestasis in p38α-deficient mice. p38α-knockout hepatocytes exhibited cytokinesis failure evidenced by an enhanced binucleation rate. As chronic cholestasis evolved the binucleation rate decreased in wildtype animals, whereas it remained high in p38α-deficient mice. CONCLUSION: Our results highlight a key role of p38α in hepatocyte proliferation, in the development of hepatomegaly, and in survival during chronic inflammation such as biliary cirrhosis.


Asunto(s)
Proliferación Celular , Citocinesis , Cirrosis Hepática Biliar/metabolismo , Cirrosis Hepática Biliar/patología , Hígado/metabolismo , Hígado/patología , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Animales , Apoptosis/fisiología , Enfermedad Crónica , Ciclina B1/metabolismo , Ciclina D1/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Hepatocitos/metabolismo , Hepatocitos/patología , Cirrosis Hepática Biliar/mortalidad , MAP Quinasa Quinasa 2/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 14 Activada por Mitógenos/genética , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Estrés Oxidativo/fisiología , Transducción de Señal/fisiología , Tasa de Supervivencia
18.
Dev Cell ; 23(5): 1020-31, 2012 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-23102580

RESUMEN

Loss of the kinase MAP3K4 causes mouse embryonic gonadal sex reversal due to reduced expression of the testis-determining gene, Sry. However, because of widespread expression of MAP3K4, the cellular basis of this misregulation was unclear. Here, we show that mice lacking Gadd45γ also exhibit XY gonadal sex reversal caused by disruption to Sry expression. Gadd45γ is expressed in a dynamic fashion in somatic cells of the developing gonads from 10.5 days postcoitum (dpc) to 12.5 dpc. Gadd45γ and Map3k4 genetically interact during sex determination, and transgenic overexpression of Map3k4 rescues gonadal defects in Gadd45γ-deficient embryos. Sex reversal in both mutants is associated with reduced phosphorylation of p38 MAPK and GATA4. In addition, embryos lacking both p38α and p38ß also exhibit XY gonadal sex reversal. Taken together, our data suggest a requirement for GADD45γ in promoting MAP3K4-mediated activation of p38 MAPK signaling in embryonic gonadal somatic cells for testis determination in the mouse.


Asunto(s)
Proteínas Portadoras/metabolismo , MAP Quinasa Quinasa Quinasa 4/metabolismo , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Proteína de la Región Y Determinante del Sexo/genética , Testículo/embriología , Testículo/metabolismo , Animales , Proteínas Portadoras/genética , Metilación de ADN , Femenino , Factor de Transcripción GATA4/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes sry , Disgenesia Gonadal 46 XY/embriología , Disgenesia Gonadal 46 XY/genética , Disgenesia Gonadal 46 XY/metabolismo , Péptidos y Proteínas de Señalización Intracelular , MAP Quinasa Quinasa Quinasa 4/deficiencia , MAP Quinasa Quinasa Quinasa 4/genética , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 11 Activada por Mitógenos/deficiencia , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Proteína Quinasa 14 Activada por Mitógenos/genética , Modelos Biológicos , Procesos de Determinación del Sexo/genética , Procesos de Determinación del Sexo/fisiología
19.
Cell Res ; 22(3): 539-50, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21946500

RESUMEN

Enucleation of erythroblasts during terminal differentiation is unique to mammals. Although erythroid enucleation has been extensively studied, only a few genes, including retinoblastoma protein (Rb), have been identified to regulate nuclear extrusion. It remains largely undefined by which signaling molecules, the extrinsic stimuli, such as erythropoietin (Epo), are transduced to induce enucleation. Here, we show that p38α, a mitogen-activated protein kinase (MAPK), is required for erythroid enucleation. In an ex vivo differentiation system that contains high Epo levels and mimics stress erythropoiesis, p38α is activated during erythroid differentiation. Loss of p38α completely blocks enucleation of primary erythroblasts. Moreover, p38α regulates erythroblast enucleation in a cell-autonomous manner in vivo during fetal and anemic stress erythropoiesis. Markedly, loss of p38α leads to downregulation of p21, and decreased activation of the p21 target Rb, both of which are important regulators of erythroblast enucleation. This study demonstrates that p38α is a key signaling molecule for erythroblast enucleation during stress erythropoiesis.


Asunto(s)
Eritroblastos/metabolismo , Eritropoyesis , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Proteína de Retinoblastoma/metabolismo , Transducción de Señal , Animales , Diferenciación Celular , Eritroblastos/citología , Ratones , Proteína Quinasa 14 Activada por Mitógenos/deficiencia
20.
J Neurosci ; 31(34): 12059-67, 2011 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-21865449

RESUMEN

Although the p38 mitogen-activated protein kinases are active in many neuronal populations in the peripheral and central nervous systems, little is known about the physiological functions of p38 in postmitotic neurons. We report that p38 activity determines in vitro and in vivo the switch from noradrenergic to cholinergic neurotransmission that occurs in sympathetic neurons on exposure to the neuropoietic cytokines CNTF and LIF. This transdifferentiation serves as a model for the plastic mechanisms that enable mature neurons to change some of their central functions without passing through the cell cycle. We demonstrate that in postmitotic neurons, p38 and STAT pathways are concurrently activated by neuropoietic cytokine treatment for at least 12 h overlapping with changes in neurotransmitter marker gene expression. Inhibition of p38 blocks the upregulation of the nuclear matrix protein Satb2 and of cholinergic markers by CNTF without affecting STAT3 phosphorylation. Conversely, overexpression of p38α or ß in the absence of cytokines stimulates cholinergic marker expression. The neurotransmitter switch in vitro is impaired in neurons isolated from p38ß(-/-) mice. Consistent with these in vitro results, a substantial loss of cells expressing cholinergic properties is observed in vivo in the stellate ganglion of mature mice deficient in the p38ß isoform.


Asunto(s)
Acetilcolina/fisiología , Transdiferenciación Celular/genética , Neuronas Colinérgicas/enzimología , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 14 Activada por Mitógenos/genética , Ganglio Estrellado/enzimología , Animales , Animales Recién Nacidos , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Transdiferenciación Celular/efectos de los fármacos , Células Cultivadas , Neuronas Colinérgicas/citología , Neuronas Colinérgicas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 11 Activada por Mitógenos/deficiencia , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Neurotransmisores/genética , Neurotransmisores/fisiología , Ratas , Ratas Sprague-Dawley , Factores de Transcripción STAT/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Ganglio Estrellado/citología , Ganglio Estrellado/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...