Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
J Cell Mol Med ; 27(17): 2583-2593, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37525479

RESUMEN

Previous studies have demonstrated that mitogen-activated protein kinase 11 (MAPK11) functions as an important point of integration in signalling transduction pathways and controlling endocellular processes, including viability of cells, differentiation, proliferation and apoptosis, through the sequence phosphorylation of the substrate protein Ser/Thr kinase protein cascade. Though MAPK 11 plays an important role in various tumours, especially in the invasive and metastatic processes, its expression and molecular mechanism in clear cell renal cell carcinoma (ccRCC) remain unclear. Runt-associated transcription factor 2 (RUNX2), a main transcription factor for osteoblast differentiation and chondrocyte maturation, has high expression in a number of tumours. In this study, the mRNA and protein levels of targeted genes in ccRCC tissues and adjacent tissues are analysed using the Cancer Genome Atlas (TCGA) database and western blotting. The ccRCC cell proliferation was measured with colony formation and EdU assay, and cell migration was examined through transwell assay. The interactive behaviour between proteins was detected with immunoprecipitation. Half-life period of RUNX2 protein was measured with cycloheximide chase assay. The results of the study indicated overexpression of MAPK11 and RUNX2 in ccRCC tissues and cell lines. MAPK11 and RUNX2 promoted the ccRCC cell proliferation and migration. Additionally, physical interaction took place between RUNX2 and P-MAPK11, which functioned to sustain the stability of RUNX2 protein. The high expression of RUNX2 could neutralize the functional degradation in MAPK11. And the outcomes of the study suggest that the P-MAPK11/RUNX2 axis may be used as a potential therapeutic target of ccRCC.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Carcinoma de Células Renales/patología , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Neoplasias Renales/patología , Regulación Neoplásica de la Expresión Génica
2.
Plant Cell Environ ; 46(5): 1453-1471, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36691352

RESUMEN

High temperatures (HT) cause pollen abortion and poor floret fertility in rice, which is closely associated with excessive accumulation of reactive oxygen species (ROS) in the developing anthers. However, the relationships between accumulation of abscisic acid (ABA) and ROS, and their effects on tapetum-specific programmed cell death (PCD) in HT-stressed anthers are poorly characterised. Here, we determined the spatiotemporal changes in ABA and ROS levels, and their relationships with tapetal PCD under HT exposure. Mutants lacking ABA-activated protein kinase 2 (SAPK2) functions and exogenous ABA treatments were used to explore the effects of ABA signalling on the induction of PCD and ROS accumulation during pollen development. HT-induced pollen abortion was tightly associated with ABA accumulation and oxidative stress. The higher ABA level in HT-stressed anthers resulted in the earlier initiation of PCD induction and subsequently abnormal tapetum degeneration by activating ROS accumulation in developing anthers. Interactions between SAPK2 and DEAD-box ATP-dependent RNA helicase elF4A-1 (RH4) were required for ABA-induced ROS generation in developing anthers. The OsSAPK2 knockout mutants showed the impaired PCD responses in the absence of HT. However, the deficiency of SAPK2 functions did not suppress the ABA-mediated ROS generation in HT-stressed anthers.


Asunto(s)
Oryza , Especies Reactivas de Oxígeno/metabolismo , Oryza/fisiología , Ácido Abscísico/farmacología , Ácido Abscísico/metabolismo , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Polen/fisiología , Apoptosis/genética , Respuesta al Choque Térmico , Regulación de la Expresión Génica de las Plantas
3.
BMC Endocr Disord ; 21(1): 235, 2021 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-34814904

RESUMEN

BACKGROUND: Prolactinoma is a functional pituitary adenoma that secretes excessive prolactin. Dopamine agonists (DAs) such as bromocriptine (BRC) are the first-line treatment for prolactinomas, but the resistance rate is increasing year by year, creating a clinical challenge. Therefore, it is urgent to explore the molecular mechanism of bromocriptine resistance in prolactinomas. Activation of the P38 MAPK pathway affects multidrug resistance in tumours. Our previous studies have demonstrated that inhibiting MAPK14 can suppress the occurrence of prolactinoma, but the role of MAPK11/12/13/14 (p38 MAPK) signalling in dopamine agonist-resistant prolactinomas is still unclear. METHODS: A prolactinoma rat model was established to determine the effect of bromocriptine on MAPK11/12/13/14 signalling. DA-resistant GH3 cells and DA-sensitive MMQ cells were used, and the role of MAPK11/12/13/14 in bromocriptine-resistant prolactinomas was preliminarily verified by western blot, RT-qPCR, ELISA, flow cytometry and CCK-8 experiments. The effects of MAPK11 or MAPK14 on bromocriptine-resistant prolactinomas were further verified by siRNA transfection experiments. RESULTS: Bromocriptine was used to treat rat prolactinoma by upregulating DRD2 expression and downregulating the expression level of MAPK11/12/13/14 in vivo experiments. The in vitro experiments showed that GH3 cells are resistant to bromocriptine and that MMQ cells are sensitive to bromocriptine. Bromocriptine could significantly reduce the expression of MAPK12 and MAPK13 in GH3 cells and MMQ cells. Bromocriptine could significantly reduce the expression of MAPK11, MAPK14, NF-κB p65 and Bcl2 in MMQ but had no effect on MAPK11, MAPK14, NF-κB p65 and Bcl2 in GH3 cells. In addition, knockdown of MAPK11 and MAPK14 in GH3 cells by siRNA transfection reversed the resistance of GH3 cells to bromocriptine, and haloperidol (HAL) blocked the inhibitory effect of bromocriptine on MAPK14, MAPK11, and PRL in MMQ cells. Our findings show that MAPK11 and MAPK14 proteins are involved in bromocriptine resistance in prolactinomas. CONCLUSION: Bromocriptine reduces the expression of MAPK11/12/13/14 in prolactinomas, and MAPK11 and MAPK14 are involved in bromocriptine resistance in prolactinomas by regulating apoptosis. Reducing the expression of MAPK11 or MAPK14 can reverse bromocriptine resistance in prolactinomas.


Asunto(s)
Agonistas de Dopamina/uso terapéutico , Neoplasias Hipofisarias/tratamiento farmacológico , Neoplasias Hipofisarias/enzimología , Prolactinoma/tratamiento farmacológico , Prolactinoma/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Animales , Apoptosis , Bromocriptina/uso terapéutico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Resistencia a Medicamentos , Estradiol/administración & dosificación , Estradiol/análogos & derivados , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 12 Activada por Mitógenos/genética , Proteína Quinasa 13 Activada por Mitógenos/genética , Proteína Quinasa 14 Activada por Mitógenos/genética , Prolactina/genética , Prolactinoma/inducido químicamente , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D1/genética , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/genética
4.
Plant Cell Rep ; 40(3): 491-506, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33388892

RESUMEN

KEY MESSAGE: StMAPK11 overexpression promotes potato growth, physiological activities and photosynthesis under drought conditions. Mitogen-activated protein kinases (MAPKs) are import regulators of MAPK pathway in plants under drought condition. However, the critical role in potato (Solanum tuberosum L.) drought resistance is not fully understood. In this study, we aimed to explore the role of StMAPK11 under drought stress. The result of RT-qPCR for assay of StMAPKs expression demonstrated that 15 StMAPKs were differentially expressed in leaves, flowers, petioles, stamens, pistils, stems, stolons, roots, tubers and tuber peels of potato. StMAPKs was dynamically modulated by abiotic stresses and plant hormone treatments, and StMAPK11 was apparently up-regulated under drought conditions. Therefore, the vectors pCPB-StMAPK11 and pCPBI121-miRmapk11 for over-expression and down-regulation of StMAPK11 were constructed, respectively, and introduced into potato cultivar Atlantic. The result showed that StMAPK11 promoted potato growth under drought conditions, as well as the physiological activities evidenced by changes in SOD, CAT and POD activity and H2O2, proline and MDA content. StMAPK11 up-regulation intensified drought resistance of potato plant by elevating antioxidant activities and photosynthesis. Moreover, we consolidated the protective role of StMAPK11 in tobacco and Arabidopsis against drought stress. The result could provide new insights into the function of StMAPK11 in drought response and its possible mechanisms.


Asunto(s)
Sequías , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Fotosíntesis/fisiología , Proteínas de Plantas/metabolismo , Solanum tuberosum/fisiología , Arabidopsis/genética , Arabidopsis/crecimiento & desarrollo , Enzimas/metabolismo , Regulación de la Expresión Génica de las Plantas , Peróxido de Hidrógeno/metabolismo , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteínas de Plantas/genética , Plantas Modificadas Genéticamente , Prolina/metabolismo , Estrés Fisiológico , Nicotiana/genética , Nicotiana/crecimiento & desarrollo
5.
PLoS Pathog ; 14(5): e1007020, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29746571

RESUMEN

The fruit fly Drosophila melanogaster is a powerful model system for the study of innate immunity in vector insects as well as mammals. For vector insects, it is particularly important to understand all aspects of their antiviral immune defenses, which could eventually be harnessed to control the transmission of human pathogenic viruses. The immune responses controlling RNA viruses in insects have been extensively studied, but the response to DNA virus infections is poorly characterized. Here, we report that infection of Drosophila with the DNA virus Invertebrate iridescent Virus 6 (IIV-6) triggers JAK-STAT signaling and the robust expression of the Turandots, a gene family encoding small secreted proteins. To drive JAK-STAT signaling, IIV-6 infection more immediately induced expression of the unpaireds, a family of IL-6-related cytokine genes, via a pathway that required one of the three Drosophila p38 homologs, p38b. In fact, both Stat92E and p38b were required for the survival of IIV-6 infected flies. In addition, in vitro induction of the unpaireds required an NADPH-oxidase, and in vivo studies demonstrated Nox was required for induction of TotA. These results argue that ROS production, triggered by IIV-6 infection, leads to p38b activation and unpaired expression, and subsequent JAK-STAT signaling, which ultimately protects the fly from IIV-6 infection.


Asunto(s)
Drosophila melanogaster/inmunología , Drosophila melanogaster/virología , Iridovirus/patogenicidad , Transducción de Señal/inmunología , Animales , Animales Modificados Genéticamente , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Iridovirus/inmunología , Quinasas Janus/genética , Quinasas Janus/metabolismo , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo , Transducción de Señal/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
Int J Mol Sci ; 21(20)2020 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-33053909

RESUMEN

The p38 mitogen-activated protein kinase (MAPK) signaling pathway is implicated in cancer biology and has been widely studied over the past two decades as a potential therapeutic target. Most of the biological and pathological implications of p38MAPK signaling are often associated with p38α (MAPK14). Recently, several members of the p38 family, including p38γ and p38δ, have been shown to play a crucial role in several pathologies including cancer. However, the specific role of p38ß (MAPK11) in cancer is still elusive, and further investigation is needed. Here, we summarize what is currently known about the role of p38ß in different types of tumors and its putative implication in cancer therapy. All evidence suggests that p38ß might be a key player in cancer development, and could be an important therapeutic target in several pathologies, including cancer.


Asunto(s)
Susceptibilidad a Enfermedades , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Neoplasias/etiología , Neoplasias/metabolismo , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Proteína Quinasa 11 Activada por Mitógenos/genética , Familia de Multigenes , Neoplasias/patología , Transducción de Señal
7.
Apoptosis ; 24(5-6): 395-403, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30879166

RESUMEN

Lipoapoptosis of cardiomyocytes may underlie diabetic cardiomyopathy. Numerous forms of cardiomyopathies share a common end-pathway in which apoptotic loss of cardiomyocytes is mediated by p38α mitogen activated protein kinase (MAPK). Although we have previously shown that palmitic acid (PA), a saturated fatty acid (SFA) elevated in plasma of type 2 diabetes mellitus and morbid obesity, induces apoptosis in cardiomyocytes via p38α MAPK-dependent signaling, the downstream cascade events that cause cell death remain unknown. The objective of this study was to investigate mechanisms involved in palmitic acid-induced cardiomyocyte apoptosis. Human adult ventricular cardiomyocyte line (AC16 cells) exposed to high physiological levels of PA for 16 h showed enhanced transcription and phosphorylation of c-fos and c-jun subunits of AP-1 and transcription of caspase 8. When AC16 cells were transfected with small interfering RNA specific against p38α MAPK (si-p38α) for 24 or 48 h, the amplified phosphorylation of c-fos was dose-dependently attenuated, and procaspase 8 was dose-dependently reduced. With translational knockdown of c-fos, PA-induced apoptosis was diminished. Inhibition of caspase 8 for 24 h reduced apoptosis in PA-treated cardiomyocytes. These findings provide evidence for induction of apoptosis in cardiomyocytes exposed to high SFA by a novel pathway requiring activation of c-fos/AP-1 and caspase 8. These results demonstrate how elevated plasma SFA may lead to continual and cumulative loss of cardiomyocytes and potentially contribute to the development of diabetic cardiomyopathy.


Asunto(s)
Apoptosis , Caspasa 8/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Miocitos Cardíacos/patología , Ácido Palmítico/metabolismo , Factor de Transcripción AP-1/metabolismo , Apoptosis/efectos de los fármacos , Caspasa 8/genética , Inhibidores de Caspasas/farmacología , Línea Celular Transformada , Humanos , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/genética , Miocitos Cardíacos/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , ARN Interferente Pequeño , Factor de Transcripción AP-1/genética , Transcripción Genética
8.
J Biol Chem ; 292(5): 1762-1772, 2017 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-28011639

RESUMEN

The evolutionarily conserved protein kinase p38 mediates innate resistance to environmental stress and microbial infection. Four p38 isoforms exist in mammals and may have been co-opted for new roles in adaptive immunity. Murine T cells deficient in p38α, the ubiquitously expressed p38 isoform, showed no readily apparent cell-autonomous defects while expressing elevated amounts of another isoform, p38ß. Mice with T cells simultaneously lacking p38α and p38ß displayed lymphoid atrophy and elevated Foxp3+ regulatory T cell frequencies. Double deficiency of p38α and p38ß in naïve CD4+ T cells resulted in an attenuation of MAPK-activated protein kinase (MK)-dependent mTOR signaling after T cell receptor engagement, and enhanced their differentiation into regulatory T cells under appropriate inducing conditions. Pharmacological inhibition of the p38-MK-mTOR signaling module produced similar effects, revealing potential for therapeutic applications.


Asunto(s)
Sistema de Señalización de MAP Quinasas/inmunología , Proteína Quinasa 11 Activada por Mitógenos/inmunología , Proteína Quinasa 14 Activada por Mitógenos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Noqueados , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 14 Activada por Mitógenos/genética , Receptores de Antígenos de Linfocitos T/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/inmunología
9.
BMC Plant Biol ; 18(1): 203, 2018 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-30236054

RESUMEN

BACKGROUND: The sucrose non-fermenting-1-related protein kinase 2 family (SnRK2s) unifies different abiotic stress signals in plants. To date, the functions of two rice SnRK2s, osmotic stress/ABA-activated protein kinase 1 (SAPK1) and SAPK2, have been unknown. We investigated their roles in response to salt stress by generating loss-of-function lines using the CRISPR/Cas9 system and by overexpressing these proteins in transgenic rice plants. RESULTS: Expression profiling revealed that SAPK1 and SAPK2 expression were strongly induced by drought, NaCl, and PEG treatment, but not by ABA. SAPK2 expression was highest in the leaves, followed by the roots, whereas SAPK1 was highest expressed in roots followed by leaves. Both proteins were localized to the nucleus and the cytoplasm. Under salt stress, sapk1, sapk2 and, in particular, sapk1/2 mutants, exhibited reduced germination rates, more severe growth inhibition, more distinct chlorosis, reduced chlorophyll contents, and reduced survival rates in comparison with the wild-type plants. In contrast, SAPK1- and SAPK2-overexpression lines had increased germination rates and reduced sensitivities to salt; including mild reductions in growth inhibition, reduced chlorosis, increased chlorophyll contents and improved survival rates in comparison with the wild-type plants. These results suggest that SAPK1 and SAPK2 may function collaboratively as positive regulators of salt stress tolerance at the germination and seedling stages. We also found that SAPK1 and SAPK2 affected the osmotic potential following salt stress by promoting the generation of osmotically active metabolites such as proline. SAPK1 and SAPK2 also improved reactive oxygen species (ROS) detoxification following salt stress by promoting the generation of ROS scavengers such as ascorbic acid, and by increasing the expression levels of proteins such as superoxide dismutase (SOD) and catalase (CAT). SAPK1 and SAPK2 may function collaboratively in reducing Na+ toxicity by affecting the Na+ distribution between roots and shoots, Na+ exclusion from the cytoplasm, and Na+ sequestration into the vacuoles. These effects may be facilitated through the expression of Na+-and K+-homeostasis-related genes. CONCLUSION: SAPK1 and SAPK2 may function collaboratively as positive regulators of salt stress tolerance at the germination and seedling stages in rice. SAPK1 and SAPK2 may be useful to improve salt tolerance in crop plants.


Asunto(s)
Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Oryza/fisiología , Tolerancia a la Sal/fisiología , Clorofila/genética , Clorofila/metabolismo , Regulación de la Expresión Génica de las Plantas , Germinación , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 8 Activada por Mitógenos/genética , Mutación , Oryza/genética , Ósmosis , Plantas Modificadas Genéticamente/genética , Potasio/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Tolerancia a la Sal/genética , Plantones/fisiología , Sodio/metabolismo
10.
Brain Behav Immun ; 72: 34-44, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29128611

RESUMEN

p38 mitogen-activated protein kinase (MAPK) consists of two major isoforms: p38α and p38ß; however, it remains unclear which isoform is more important for chronic pain development. Recently, we developed potent, long-lasting, and p38 MAPK subtype-specific antisense oligonucleotides (ASOs). We examined the therapeutic effects of isoform-specific ASOs in several chronic pain models following single intrathecal injection (300 µg/10 µl) in CD1 mice. In the chronic constriction injury (CCI) model, p38α MAPK ASO, given on post-operative day 5, reduced CCI-induced mechanical allodynia in male but not female mice. In contrast, mechanical allodynia after CCI in both sexes was not affected by p38ß MAPK ASO. Intrathecal injection of p38α or p38ß ASO resulted in a partial reduction (≈ 50%) of spinal p38α or p38ß mRNA level, respectively, in both sexes at two weeks. In contrast, intrathecal injection of the ASOs did not affect p38α and p38ß MAPK mRNA levels in dorsal root ganglia. Intrathecal p38α ASO also reduced postoperative pain (mechanical and cold allodynia) in male mice after tibia fracture. However, intrathecal p38α ASO had no effect on mechanical allodynia in male mice after paclitaxel treatment. Intrathecal p38α MAPK ASO pre-treatment also prevented TLR4-mediated mechanical allodynia and downregulated levels of p38α MAPK and phosphorylated p38 MAPK following intrathecal treatment of lipopolysaccharide. In summary, our findings suggest that p38α MAPK is the major p38 MAPK isoform in the spinal cord and regulates chronic pain in a sex and model-dependent manner. Intrathecal p38α MAPK ASO may offer a new treatment for some chronic pain conditions.


Asunto(s)
Neuralgia/terapia , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Ganglios Espinales/metabolismo , Hiperalgesia/inducido químicamente , Hiperalgesia/terapia , Inyecciones Espinales , Masculino , Ratones , Microglía/metabolismo , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/genética , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Oligodesoxirribonucleótidos Antisentido/administración & dosificación , Oligodesoxirribonucleótidos Antisentido/genética , Dimensión del Dolor , Dolor Postoperatorio/terapia , Enfermedades del Sistema Nervioso Periférico/metabolismo , Fosforilación , Isoformas de Proteínas , Médula Espinal/metabolismo , Receptor Toll-Like 4/metabolismo
11.
Immunol Cell Biol ; 95(9): 814-823, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28611474

RESUMEN

The p38 mitogen-activated protein kinase (MAPK) pathway is involved in the regulation of immune and inflammatory processes. We used p38α-conditional, p38ß-deficient and p38α/ß double-null mouse models to address the role of these two p38 MAPK in CD4+ T cells, and found that p38α deficiency causes these cells to hyperproliferate. Our studies indicate that both p38α and p38ß are dispensable for T helper cell type 1 (Th1) differentiation but, by controlling interferon (IFN)γ and tumor necrosis factor (TNF)α production, are critical for normal Th1 effector function. We found that both p38α and p38ß modulate T-cell receptor-induced IFNγ and TNFα production, whereas only p38α regulates cytokine-induced IFNγ production. The lack of p38α and p38ß did not affect transcription and mRNA stability of Ifng. However, the absence of p38α in Th1 cells resulted in a decreased MNK1 phosphorylation after cytokine activation, and MNK1 inhibition blocked IFNγ production. Our results indicate that p38α regulates IFNγ secretion through the activation of the MNK1/eIF4E pathway of translation initiation and identify specific functions for p38α and p38ß in T-cell proliferation.


Asunto(s)
Factor 4E Eucariótico de Iniciación/metabolismo , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Células TH1/fisiología , Animales , Proliferación Celular/genética , Células Cultivadas , Interferón gamma/metabolismo , Activación de Linfocitos , Ratones , Ratones Noqueados , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 14 Activada por Mitógenos/genética , Fosforilación , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
12.
Mamm Genome ; 28(1-2): 38-46, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27913859

RESUMEN

Human infection with Salmonella is of global public health concern. In low- and middle-income countries, Salmonella infection is a major source of disease in terms of both mortality and morbidity, while in high-income nations, the pathogen is an ongoing threat to food security. The outcome of infection with Salmonella enterica serovar Typhimurium (Salmonella Typhimurium) in mouse models is dependent upon a coordinated and complex immune response. A panel of recombinant congenic strains (RCS) derived from the reciprocal double backcross of A/J and C57BL/6J mice has been screened for their susceptibility to Salmonella infection, and the RCS AcB60 was identified to be the most resistant strain to Salmonella infection, more resistant than the parental strain A/J. These mice are known to carry resistant alleles at three well-defined Salmonella susceptibility loci, Slc11a1 Ity (solute carrier family 11 member 1; Immunity to Typhimurium locus), Pklr Ity4 (pyruvate kinase liver and red blood cell; Ity4 locus), and Ity5. In the current study, we used interval mapping to validate a locus on Chr 15, named Ity8, linked to Salmonella resistance in AcB60 mice. Global gene expression analysis during infection identified AcB60-specific expression of genes involved in Ccr7 signaling, including downstream effector Mapk11 (mitogen-activated protein kinase 11), located within the Ity8 interval, and representing a potential positional candidate gene. An additional region on Chr 18 of C57BL/6J descent was shown to be associated with increase resistance in AcB60. These observations provide an opportunity to achieve new insight into the complex genetics of resistance to Salmonella infection in the context of mouse models of human infection with Salmonella Typhimurium.


Asunto(s)
Proteínas de Transporte de Catión/genética , Proteína Quinasa 11 Activada por Mitógenos/genética , Receptores CCR7/genética , Infecciones por Salmonella/genética , Animales , Mapeo Cromosómico , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Inmunidad Innata/genética , Hígado/metabolismo , Ratones , Infecciones por Salmonella/microbiología , Salmonella typhimurium/patogenicidad
13.
J Immunol ; 195(1): 277-88, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26019272

RESUMEN

Dual-specificity phosphatase (DUSP) 1 dephosphorylates and inactivates members of the MAPK superfamily, in particular, JNKs, p38α, and p38ß MAPKs. It functions as an essential negative regulator of innate immune responses, hence disruption of the Dusp1 gene renders mice extremely sensitive to a wide variety of experimental inflammatory challenges. The principal mechanisms behind the overexpression of inflammatory mediators by Dusp1(-/-) cells are not known. In this study, we use a genetic approach to identify an important mechanism of action of DUSP1, involving the modulation of the activity of the mRNA-destabilizing protein tristetraprolin. This mechanism is key to the control of essential early mediators of inflammation, TNF, CXCL1, and CXCL2, as well as the anti-inflammatory cytokine IL-10. The same mechanism also contributes to the regulation of a large number of transcripts induced by treatment of macrophages with LPS. These findings demonstrate that modulation of the phosphorylation status of tristetraprolin is an important physiological mechanism by which innate immune responses can be controlled.


Asunto(s)
Fosfatasa 1 de Especificidad Dual/inmunología , Lipopolisacáridos/farmacología , Macrófagos/inmunología , ARN Mensajero/inmunología , Tristetraprolina/inmunología , Animales , Quimiocina CXCL1/genética , Quimiocina CXCL1/inmunología , Quimiocina CXCL2/genética , Quimiocina CXCL2/inmunología , Fosfatasa 1 de Especificidad Dual/genética , Regulación de la Expresión Génica , Inmunidad Innata , Interleucina-10/genética , Interleucina-10/inmunología , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 11 Activada por Mitógenos/inmunología , Proteína Quinasa 14 Activada por Mitógenos/genética , Proteína Quinasa 14 Activada por Mitógenos/inmunología , Fosforilación , Cultivo Primario de Células , Estabilidad del ARN , ARN Mensajero/genética , Transducción de Señal , Tristetraprolina/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
14.
Tumour Biol ; 37(6): 7255-66, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26666822

RESUMEN

P38ß, p38γ, and p38δ have been sporadically and scarcely reported to be involved in the carcinogenesis of cancers, compared with p38α isoform. However, little has been known regarding their clinicopathological significance and biological roles in esophageal squamous cell carcinoma (ESCC). Expression status of p38ß, p38γ, and p38δ was assayed using immunohistochemistry with ESCC tissue microarray; ensuing clinicopathological significance was statistically analyzed. To define its biological roles on proliferation, migration and invasion of ESCC cell line Eca109 in vitro, MTT, wound healing, and Transwell assays were employed, respectively. As confirmation, athymic nude mice were taken to verify the effect over proliferation in vivo. It was found that both p38ß and p38δ expression, other than p38γ, were significantly higher in ESCC tissues compared with paired normal controls. In terms of prognosis, only p38ß expression was observed to be significantly associated with overall prognosis. Clinicopathologically, there was significant association between p38γ expression and clinical stage, lymph nodes metastases, and tumor volume. No significant association was found for p38ß and p38δ between its expression and other clinicopathological parameters other than significant difference of expression between ESCC versus normal control. In Eca109, it was observed that p38ß, p38γ, and p38δ can promote the cell growth and motility. As verification, over-expression of p38δ can promote, whereas knockdown of p38γ can prevent, the tumorigenesis in nude mice model xenografted with Eca109 cells whose basal level of p38δ was stably over-expressed and p38γ was stably knocked down. Together, our results demonstrate that p38ß, p38γ, and p38δ played oncogenic roles in ESCC.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Neoplasias Esofágicas/enzimología , Proteína Quinasa 11 Activada por Mitógenos/fisiología , Proteína Quinasa 12 Activada por Mitógenos/fisiología , Proteína Quinasa 13 Activada por Mitógenos/fisiología , Proteínas de Neoplasias/fisiología , Animales , Apoptosis , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Ciclo Celular , Línea Celular Tumoral , Movimiento Celular , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Femenino , Xenoinjertos , Humanos , Metástasis Linfática , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 12 Activada por Mitógenos/genética , Proteína Quinasa 13 Activada por Mitógenos/genética , Proteínas de Neoplasias/genética , Pronóstico , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas Recombinantes de Fusión/metabolismo , Carga Tumoral
15.
Mol Med ; 21: 626-36, 2015 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-26225830

RESUMEN

We recently confirmed that angiotensin II (Ang II) type 1 receptor (AT1R) was overexpressed in hepatocellular carcinoma tissue using a murine hepatoma model. Angiotensin(Ang)-(1-7) has been found beneficial in ameliorating lung cancer and prostate cancer. Which receptor of Ang-(1-7) is activated to mediate its effects is much speculated. This study was designed to investigate the effects of Ang-(1-7) on hepatocellular carcinoma, as well as the probable mechanisms. H22 hepatoma-bearing mice were randomly divided into five groups for treatment: mock group, low-dose Ang-(1-7), high-dose Ang-(1-7), high-dose Ang-(1-7) + A779 and high-dose Ang-(1-7) + PD123319. Ang-(1-7) treatment inhibited tumor growth time- and dose-dependently by arresting tumor proliferation and promoting tumor apoptosis as well as inhibiting tumor angiogenesis. The effects of Ang-(1-7) on tumor proliferation and apoptosis were reversed by coadministration with A779 or PD123319, whereas the effects on tumor angiogenesis were completely reversed by A779 but not by PD123319. Moreover, Ang-(1-7) downregulated AT1R mRNA, upregulated mRNA levels of Ang II type 2 receptor (AT2R) and Mas receptor (MasR) and p38-MAPK phosphorylation and suppressed H22 cell-endothelial cell communication. Thus, Ang-(1-7) administration suppresses hepatocellular carcinoma via complex interactions of AT1R, AT2R and MasR and may provide a novel and promising approach for the treatment of hepatocellular carcinoma.


Asunto(s)
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Neovascularización Patológica/genética , Receptor de Angiotensina Tipo 1/biosíntesis , Receptor de Angiotensina Tipo 2/biosíntesis , Angiotensina I/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Ratones , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Fragmentos de Péptidos/administración & dosificación , Fosforilación , Proto-Oncogenes Mas , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 2/genética , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Fish Shellfish Immunol ; 47(1): 331-43, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26363230

RESUMEN

The p38 kinases are one of the four subgroups of mitogen-activated protein kinase (MAPK) superfamily which are involved in the innate immunity. The p38 subfamily that includes four members namely p38α (MAPK14), p38ß (MAPK11), p38γ (MAPK12) and p38δ (MAPK13), regulates the activation of several transcription factors. In this study, a p38ß (OfMAPK11) homolog and a p38α (OfMAPK14) homolog of Oplegnathus fasciatus were identified at genomic level. Results clearly showed that both MAPK11 and MAPK14 are well-conserved at both genomic structural- and amino acid (aa)-levels. Genomic sequences of OfMAPK11 (∼ 15.6 kb) and OfMAPK14 (∼ 13.4 kb) had 12 exons. A comparison of exon-intron structural arrangement of these genes from different vertebrate lineages indicated that all the exon lengths are highly conserved, except their terminal exons. Full-length cDNAs of OfMAPK11 (3957 bp) and OfMAPK14 (2504 bp) encoded corresponding proteins of 361 aa and 360 aa, respectively. Both OfMAPK proteins harbored a Ser/Thr protein kinases catalytic domain (S_TKc domain) which includes an activation loop with a dual phosphorylation site (TGY motif) and several specific-binding sites for ATP and substrates. Molecular modeling of the activation loop and substrate binding sites of rock bream MAPKs revealed the conservation of crucial residues and their orientation in 3D space. Transcripts of OfMAPKs were ubiquitously detected in eleven tissues examined, however at different levels. The modulation of OfMAPKs' transcription upon pathogen-associated molecular patterns (PAMPs: flagellin, lipopolysaccharide and poly I:C) and pathogens (Edwardsiella tarda, Streptococcus iniae and rock bream iridovirus) was investigated. Among the seven examined tissues, the flagellin-challenge upregulated the mRNA level of both OfMAPKs in the head kidney. Meanwhile, modulation of OfMAPK mRNA expression in the liver upon other immune-challenges varied in a time-dependent manner. Collectively, these results suggest that OfMAPKs are true members of p38 subfamily, which might be induced by different immune stimuli.


Asunto(s)
Enfermedades de los Peces/genética , Proteínas de Peces/genética , Regulación de la Expresión Génica , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 14 Activada por Mitógenos/genética , Perciformes/genética , Perciformes/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Infecciones por Virus ADN/genética , Infecciones por Virus ADN/inmunología , Infecciones por Virus ADN/veterinaria , Infecciones por Virus ADN/virología , Edwardsiella tarda/fisiología , Infecciones por Enterobacteriaceae/genética , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/veterinaria , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/microbiología , Enfermedades de los Peces/virología , Proteínas de Peces/química , Proteínas de Peces/metabolismo , Flagelina/farmacología , Iridoviridae/fisiología , Lipopolisacáridos/farmacología , Proteína Quinasa 11 Activada por Mitógenos/química , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/química , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Especificidad de Órganos , Perciformes/clasificación , Filogenia , Poli I-C/farmacología , Alineación de Secuencia/veterinaria , Infecciones Estreptocócicas/genética , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/veterinaria , Streptococcus/fisiología
17.
J Biol Chem ; 288(23): 17030-17041, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23625915

RESUMEN

The Ca(2+) sensor STIM1 is crucial for activation of store-operated Ca(2+) entry (SOCE) through transient receptor potential canonical and Orai channels. STIM1 phosphorylation serves as an "off switch" for SOCE. However, the signaling pathway for STIM1 phosphorylation is unknown. Here, we show that SOCE activates AMP-activated protein kinase (AMPK); its effector p38ß mitogen-activated protein kinase (p38ß MAPK) phosphorylates STIM1, thus inhibiting SOCE in human lung microvascular endothelial cells. Activation of AMPK using 5-aminoimidazole-4-carboxamide-1-ß-d-ribofuranoside (AICAR) resulted in STIM1 phosphorylation on serine residues and prevented protease-activated receptor-1 (PAR-1)-induced Ca(2+) entry. Furthermore, AICAR pretreatment blocked PAR-1-induced increase in the permeability of mouse lung microvessels. Activation of SOCE with thrombin caused phosphorylation of isoform α1 but not α2 of the AMPK catalytic subunit. Moreover, knockdown of AMPKα1 augmented SOCE induced by thrombin. Interestingly, SB203580, a selective inhibitor of p38 MAPK, blocked STIM1 phosphorylation and led to sustained STIM1-puncta formation and Ca(2+) entry. Of the three p38 MAPK isoforms expressed in endothelial cells, p38ß knockdown prevented PAR-1-mediated STIM1 phosphorylation and potentiated SOCE. In addition, inhibition of the SOCE downstream target CaM kinase kinase ß (CaMKKß) or knockdown of AMPKα1 suppressed PAR-1-mediated phosphorylation of p38ß and hence STIM1. Thus, our findings demonstrate that SOCE activates CaMKKß-AMPKα1-p38ß MAPK signaling to phosphorylate STIM1, thereby suppressing endothelial SOCE and permeability responses.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Calcio/metabolismo , Células Endoteliales/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Proteínas de Neoplasias/metabolismo , Receptor PAR-1/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Canales de Calcio , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/fisiología , Células Cultivadas , Células Endoteliales/citología , Técnicas de Silenciamiento del Gen , Humanos , Hipoglucemiantes/farmacología , Pulmón/irrigación sanguínea , Pulmón/metabolismo , Glicoproteínas de Membrana/genética , Proteínas de la Membrana/genética , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteínas de Neoplasias/genética , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Receptor PAR-1/genética , Ribonucleótidos/farmacología , Molécula de Interacción Estromal 1
18.
Biochim Biophys Acta ; 1833(9): 2083-91, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23639288

RESUMEN

p27 is a cyclin-dependent kinase (CDK) inhibitor that suppresses a cell's transition from G0 to S phase, therefore acting as a tumor suppressor. Our most recent studies demonstrate that upon arsenite exposure, p27 suppresses Hsp27 and Hsp70 expressions through the JNK2/c-Jun- and HSF-1-dependent pathways, suggesting a novel molecular mechanism underlying the tumor suppressive function of p27 in a CDK-independent manner. We found that p27-deficiency (p27-/-) resulted in the elevation of cyclooxygenase-2 (COX-2) expression at transcriptional level, whereas the introduction of p27 brought back COX-2 expression to a level similar to that of p27+/+ cells, suggesting that p27 exhibits an inhibitory effect on COX-2 expression. Further studies identified that p27 inhibition of COX-2 expression was specifically due to phosphorylation of transcription factor cAMP response element binding (CREB) phosphorylation mediated by p38ß and p38δ. These results demonstrate a novel mechanism underlying tumor suppression effect of p27 and will contribute to the understanding of the overall mechanism of p27 tumor suppression in a CDK-independent manner.


Asunto(s)
Arsenitos/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Ciclooxigenasa 2/biosíntesis , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Proteína Quinasa 13 Activada por Mitógenos/metabolismo , Teratógenos/farmacología , Animales , Línea Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Ciclooxigenasa 2/genética , Regulación Enzimológica de la Expresión Génica/genética , Ratones , Ratones Noqueados , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 13 Activada por Mitógenos/genética , Fosforilación/efectos de los fármacos , Fosforilación/genética , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética
19.
FASEB J ; 27(7): 2893-901, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23568773

RESUMEN

The N-end rule pathway contributes significantly to accelerated muscle proteolysis mediated by the ubiquitin-proteasome pathway in various catabolic conditions. UBR2 (aka E3α-II) is the only known E3 ubiquitin ligase of the N-end rule pathway that is up-regulated by cachectic stimuli including proinflammatory cytokines and tumors. However, the signaling mechanism through which UBR2 is up-regulated remains undetermined. Here we identify a signaling pathway that mediates tumor cell-induced up-regulation of UBR2. UBR2 expression in C2C12 myotubes was up-regulated by conditioned medium from Lewis lung carcinoma cells or C26 colon adenocarcinoma cells, which was blocked by a pharmacological inhibitor of p38α/ß mitogen-activated protein kinase (MAPK), SB202190. Similarly, SB202190 administration (i.p.) abolished UBR2 up-regulation in the tibialis anterior of LLC tumor-bearing mice. Genetic gain and loss of function assays in C2C12 myotubes indicated that tumor-induced activation of the p38ß isoform is sufficient and necessary for UBR2 up-regulation. In addition, UBR2 up-regulation required p38ß-mediated phosphorylation of CCAAT/enhancer binding protein (C/EBP)-ß Thr-188, which was critical to C/EBPß binding to the UBR2 promoter. Furthermore, luciferase reporter assay revealed that the C/EBPß binding motif in the UBR2 promoter is a functional C/EBPß-responsive cis-element that enhances the promoter activity on activation by p38ß. Finally, genetic ablation of C/EBPß blocked UBR2 up-regulation in LLC tumor-bearing mice. These results suggest that UBR2 up-regulation in cachectic muscle is mediated by the p38ß-C/EBPß signaling pathway responsible for the bulk of tumor-induced muscle proteolysis.


Asunto(s)
Carcinoma Pulmonar de Lewis/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Regulación hacia Arriba , Animales , Western Blotting , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patología , Línea Celular , Línea Celular Tumoral , Medios de Cultivo Condicionados/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Imidazoles/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 11 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Mioblastos/citología , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo , Fosforilación , Regiones Promotoras Genéticas/genética , Unión Proteica , Piridinas/farmacología , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ubiquitina-Proteína Ligasas/genética
20.
Proc Natl Acad Sci U S A ; 108(31): 12764-9, 2011 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-21768366

RESUMEN

p38α MAPK is an important regulator of cellular responses induced by external cues, but the elucidation of physiological functions for p38α has been complicated by the possible functional redundancy in vivo with the related family member p38ß. We found that mice with combined deletion of p38α and p38ß display diverse developmental defects at midgestation, including major cardiovascular abnormalities, which are observed neither in single knockout nor in double heterozygous embryos. Expression analysis indicates specific functions of p38α and p38ß in the regulation of cardiac gene expression during development. By using knock-in animals that express p38ß under control of the endogenous p38α promoter, we also found that p38ß cannot perform all of the functions of p38α during embryogenesis. Our results identify essential roles for p38α and p38ß during development and suggest that some specific functions may be explained by differences in expression patterns.


Asunto(s)
Embrión de Mamíferos/metabolismo , Perfilación de la Expresión Génica , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 14 Activada por Mitógenos/genética , Animales , Animales Recién Nacidos , Apoptosis/genética , Apoptosis/fisiología , Ciclo Celular/genética , Ciclo Celular/fisiología , Proliferación Celular , Embrión de Mamíferos/anomalías , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Immunoblotting , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Miocardio/metabolismo , Miocardio/patología , Regiones Promotoras Genéticas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA