Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.121
Filtrar
1.
Learn Mem ; 29(12): 435-446, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36446603

RESUMEN

Empirical and computational methods were combined to examine whether individual or dual-drug treatments can restore the deficit in long-term synaptic facilitation (LTF) of the Aplysia sensorimotor synapse observed in a cellular model of Coffin-Lowry syndrome (CLS). The model was produced by pharmacological inhibition of p90 ribosomal S6 kinase (RSK) activity. In this model, coapplication of an activator of the mitogen-activated protein kinase (MAPK) isoform ERK and an activator of protein kinase A (PKA) resulted in enhanced phosphorylation of RSK and enhanced LTF to a greater extent than either drug alone and also greater than their additive effects, which is termed synergism. The extent of synergism appeared to depend on another MAPK isoform, p38 MAPK. Inhibition of p38 MAPK facilitated serotonin (5-HT)-induced RSK phosphorylation, indicating that p38 MAPK inhibits activation of RSK. Inhibition of p38 MAPK combined with activation of PKA synergistically activated both ERK and RSK. Our results suggest that cellular models of disorders that affect synaptic plasticity and learning, such as CLS, may constitute a useful strategy to identify candidate drug combinations, and that combining computational models with empirical tests of model predictions can help explain synergism of drug combinations.


Asunto(s)
Síndrome de Coffin-Lowry , Proteínas Quinasas Dependientes de AMP Cíclico , Plasticidad Neuronal , Proteínas Quinasas p38 Activadas por Mitógenos , Humanos , Síndrome de Coffin-Lowry/fisiopatología , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Plasticidad Neuronal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Serotonina/farmacología
2.
Neuropharmacology ; 202: 108835, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34648772

RESUMEN

Peripheral nerve injuries (PNIs) often result in persistent neuropathic pain, seriously affecting quality of life. Existing therapeutic interventions for PNI-induced neuropathic pain are far from satisfactory. Extracellular signal-regulated kinases (ERKs) and p38 have been found to participate in triggering and maintaining PNI-induced neuropathic pain. However, ERK and p38 also contribute to axonal regeneration and motor function recovery after PNI, making it difficult to inhibit ERK and p38 for therapeutic purposes. In this study, we simultaneously characterized neuropathic pain and motor function recovery in a mouse sciatic nerve crush injury model to identify the time window for therapeutic interventions. We further demonstrated that delayed delivery of a combination of ERK and p38 inhibitors at three weeks after PNI could significantly alleviate PNI-induced neuropathic pain without affecting motor function recovery. Additionally, the combined use of these two inhibitors could suppress pain markedly better than either inhibitor alone, possibly reducing the required dose of each inhibitor and alleviating the side effects and risks of the inhibitors when used individually.


Asunto(s)
Butadienos/farmacología , Butadienos/uso terapéutico , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Imidazoles/farmacología , Imidazoles/uso terapéutico , Neuralgia/tratamiento farmacológico , Neuralgia/etiología , Nitrilos/farmacología , Nitrilos/uso terapéutico , Traumatismos de los Nervios Periféricos/complicaciones , Traumatismos de los Nervios Periféricos/fisiopatología , Piridinas/farmacología , Piridinas/uso terapéutico , Nervio Ciático/lesiones , Nervio Ciático/fisiopatología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Animales , Axones/fisiología , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Masculino , Ratones Endogámicos C57BL , Regeneración Nerviosa/genética , Neuralgia/genética , Recuperación de la Función , Resultado del Tratamiento , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
3.
Front Immunol ; 12: 767813, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34858424

RESUMEN

Viral infections seriously affect the health of organisms including humans. Now, more and more researchers believe that microRNAs (miRNAs), one of the members of the non-coding RNA family, play significant roles in cell biological function, disease occurrence, and immunotherapy. However, the roles of miRNAs in virus infection (entry and replication) and cellular immune response remain poorly understood, especially in low vertebrate fish. In this study, based on the established virus-cell infection model, Singapore grouper iridovirus (SGIV)-infected cells were used to explore the roles of miR-124 of Epinephelus coioides, an economically mariculture fish in southern China and Southeast Asia, in viral infection and host immune responses. The expression level of E. coioides miR-124 was significantly upregulated after SGIV infection; miR-124 cannot significantly affect the entry of SGIV, but the upregulated miR-124 could significantly promote the SGIV-induced cytopathic effects (CPEs), the viral titer, and the expressions of viral genes. The target genes of miR-124 were JNK3/p38α mitogen-activated protein kinase (MAPK). Overexpression of miR-124 could dramatically inhibit the activation of NF-κB/activating protein-1 (AP-1), the transcription of proinflammatory factors, caspase-9/3, and the cell apoptosis. And opposite results happen when the expression of miR-124 was inhibited. The results suggest that E. coioides miR-124 could promote viral replication and negatively regulate host immune response by targeting JNK3/p38α MAPK, which furthers our understanding of virus and host immune interactions.


Asunto(s)
Lubina/virología , Infecciones por Virus ADN/veterinaria , Enfermedades de los Peces/inmunología , Iridovirus/fisiología , MicroARNs/fisiología , Replicación Viral , Animales , Apoptosis , Infecciones por Virus ADN/inmunología , Inmunidad Innata , Proteína Quinasa 10 Activada por Mitógenos/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
4.
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
5.
Biomed Res Int ; 2021: 8328216, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34746310

RESUMEN

Although hypoxia has been shown to promote keratinocyte migration and reepithelialization, the underlying molecular mechanisms remain largely unknown. ADAM17, a member of the metalloproteinase superfamily, has been implicated in a variety of cellular behaviors such as proliferation, adhesion, and migration. ADAM17 is known to promote cancer cell migration under hypoxia, and whether or how ADAM17 plays a role in hypoxia-induced keratinocyte migration has not been identified. Here, we found that ADAM17 expression and activity were significantly promoted in keratinocytes under hypoxic condition, inhibition of ADAM17 by TAPI-2, or silencing of ADAM17 using small interfering RNA which suppressed the hypoxia-induced migration of keratinocytes significantly, indicating a pivotal role for ADAM17 in keratinocyte migration. Further, we showed that p38/MAPK was activated by hypoxia. SB203580, an inhibitor of p38/MAPK, significantly attenuated the upregulation of ADAM17 as well as the migration of keratinocytes induced by hypoxia. Activation of p38/MAPK by MKK6 (Glu) overexpression, however, had adverse effects. Taken together, our study demonstrated that hypoxia-induced keratinocyte migration requires the p38/MAPK-ADAM17 signal axis, which sheds new light on the regulatory mechanisms of keratinocyte migration. Our study might also help in developing therapeutic strategies to facilitate wound healing in vivo, where cells are migrated in a hypoxic microenvironment.


Asunto(s)
Proteína ADAM17/metabolismo , Queratinocitos/metabolismo , Hipoxia de la Célula/fisiología , Movimiento Celular/fisiología , Humanos , Ácidos Hidroxámicos/farmacología , Imidazoles/farmacología , Queratinocitos/citología , Piridinas/farmacología , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Cicatrización de Heridas/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
6.
Med Oncol ; 38(12): 138, 2021 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-34633592

RESUMEN

Tumor necrosis factor-related apoptosis-induced ligand (TRAIL) is a cytokine that initiates apoptosis upon binding to death receptor 5 (DR5) on cancer cells. Small molecule TRAIL mimetics have therefore been investigated as promising chemotherapeutic agents. Since anemia of chemotherapy is common, our goal is to investigate the hemolytic and eryptotic properties of novel DR5 agonist bioymifi (BMF) and identify the underlying molecular mechanisms. Whole blood (WB) was stimulated with 100 µM of BMF, whereas red blood cells (RBCs) were treated with 10-100 µM of BMF for 24 h at 37 °C. WB was analyzed for RBC, leukocyte, and platelet indices, while RBCs were examined for hemolysis by light absorbance of free hemoglobin, membrane scrambling by Annexin V-FITC, calcium by Fluo4/AM, cellular morphology by light scatter, and oxidative stress by 2',7'-dichlorodihydrofluorescein diacetate (H2DCFDA) using flow cytometry. Caspase inhibitor Z-VAD-FMK, p38 inhibitor SB203580, casein kinase 1α inhibitor D4476, receptor-interacting protein 1 inhibitor necrostatin-2, reduced glutathione, or cyclooxygenase (COX) inhibitor aspirin were added accordingly. BMF exerted dose-responsive, calcium-independent hemolysis, reduced RBC hemoglobin, significantly increased Annexin V-, Fluo4-, and DCF-positive cells, along with a dual effect on forward and side light scatter. Notably, the cytotoxic potential of BMF was significantly mitigated upon pharmacological inhibition of p38. Furthermore, BMF exhibited selective toxicity to eosinophils and significantly diminished reticulocyte hemoglobin content. Altogether, these novel findings highlight the adverse outcomes of BMF exposure on RBC physiology and provide the first toxicological assessment of BMF as an antitumor agent.


Asunto(s)
Eriptosis/efectos de los fármacos , Ftalimidas/toxicidad , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/agonistas , Tiazolidinas/toxicidad , Calcio/metabolismo , Eosinófilos/efectos de los fármacos , Eritrocitos/efectos de los fármacos , Hemólisis/efectos de los fármacos , Humanos , Especies Reactivas de Oxígeno/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
7.
Int J Mol Sci ; 22(17)2021 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-34502507

RESUMEN

p38 and c-Jun N-terninal kinase (JNK) are activated in response to acute stress and inflammatory signals. Through modification of a plethora of substrates, these kinases profoundly re-shape cellular physiology for the optimal response to a harmful environment and/or an inflammatory state. Here, we utilized phospho-proteomics to identify several hundred substrates for both kinases. Our results indicate that the scale of signaling from p38 and JNK are of a similar magnitude. Among the many new targets, we highlight the regulation of the transcriptional regulators grb10-interacting GYF protein 1 and 2 (GIGYF1/2) by p38-dependent MAP kinase-activated protein kinase 2 (MK2) phosphorylation and 14-3-3 binding. We also show that the Golgi apparatus contains numerous substrates, and is a major target for regulation by p38 and JNK. When activated, these kinases mediate structural rearrangement of the Golgi apparatus, which positively affects protein flux through the secretory system. Our work expands on our knowledge about p38 and JNK signaling with important biological ramifications.


Asunto(s)
MAP Quinasa Quinasa 4/metabolismo , Estrés Fisiológico/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Aparato de Golgi/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , MAP Quinasa Quinasa 4/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
8.
J Dairy Res ; 88(3): 293-301, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34425921

RESUMEN

Staphylococcus aureus is a common pathogen of bovine mastitis which can induce autophagy and inhibit autophagy flux, resulting in intracellular survival and persistent infection. The aim of the current study was to investigate the role of p38α in the autophagy induced by intracellular S. aureus in bovine mammary epithelial cells. An intracellular infection model of MAC-T cells was constructed, and activation of p38α was examined after S. aureus invasion. Through activating/inhibiting p38α by anisomycin/SB203580, the autophagosomes, LC3 and p62 level were analyzed by immunofluorescence and western blot. To further study the detailed mechanism of p38α, phosphorylation of ULK1ser757 was also detected. The results showed that intracellular S. aureus activated p38α, and the activation developed in a time-dependent manner. Inhibition of p38α promoted intracellular S. aureus-induced autophagy flow, up-regulated the ratio of LC3 II/I, reduced the level of p62 and inhibited the phosphorylation of ULK1ser757, whereas the above results were reversed after activation of p38α. The current study indicated that intracellular S. aureus can inhibit autophagy flow by activating p38α in bovine mammary epithelial cells.


Asunto(s)
Autofagia/fisiología , Células Epiteliales/microbiología , Mastitis Bovina/microbiología , Infecciones Estafilocócicas/veterinaria , Staphylococcus aureus/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Animales , Bovinos , Línea Celular , Activación Enzimática , Células Epiteliales/fisiología , Femenino , Glándulas Mamarias Animales/citología , Mastitis Bovina/fisiopatología
9.
Mol Hum Reprod ; 27(8)2021 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-34245298

RESUMEN

Hypoxia-induced oxidative stress and apoptosis of trophoblast are involved in the pathogenesis of preeclampsia (PE). Extensive research reports that the principal vagal neurotransmitter acetylcholine (ACh) shows anti-oxidative and anti-apoptotic effects in various diseases models. However, the role of ACh in hypoxic trophoblast remains unknown. Here, we examined the apoptotic levels of human placenta and explored the role(s) of ACh on cobalt chloride (CoCl2)-treated (trophoblast-derived) HTR-8/SVneo cells for mimicking hypoxic injuries. Cell counting kit-8 (CCK-8), dihydroethidium (DHE) probe, western blotting, immunofluorescence staining, migration and invasion assay were employed in the current study. Our data showed that placentas from PE women exhibited increased level of reactive oxygen species (ROS) and apoptotic index than those in normal pregnancy. Our in vitro study showed that CoCl2 enhanced ROS generation and apoptosis in HTR-8/SVneo cells through the activation of the p38 mitogen-activated protein kinase (p38 MAPK)/nuclear factor-κB (NF-κB) pathway. ACh significantly decreased hypoxia-induced ROS generation and the resulting apoptosis, accompanied by lowered phosphorylation of p38 MAPK and NF-κB. Western blotting analysis further confirmed that ACh decreased the ratio of pp38 MAPK/p38 MAPK, p-NF-κB/NF-κB, Bax/Bcl-2 and cleaved Caspase-3/Caspase-3. Besides, ACh promoted cell invasion and migration ability under hypoxic conditions. Atropine, the muscarinic receptor antagonist, abolished ACh's effects mentioned above. Overall, our data showed that ACh exerted protective effects on hypoxia-induced oxidative stress and apoptosis in trophoblast cells via muscarinic receptors, indicating that improved vagal activity may be of therapeutic value in PE management.


Asunto(s)
Acetilcolina/farmacocinética , Apoptosis/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Preeclampsia/metabolismo , Trofoblastos/efectos de los fármacos , Nervio Vago/fisiopatología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Adulto , Atropina/farmacología , Hipoxia de la Célula , Movimiento Celular/efectos de los fármacos , Cobalto/farmacología , Femenino , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Antagonistas Muscarínicos/farmacología , Preeclampsia/fisiopatología , Embarazo , Especies Reactivas de Oxígeno/metabolismo , Trofoblastos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Front Immunol ; 12: 675747, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34220822

RESUMEN

Chronic gastritis caused by Helicobacter pylori (H. pylori) infection has been widely recognized as the most important risk factor for gastric cancer. Analysis of the interaction between the key participants in gastric mucosal immunity and H. pylori infection is expected to provide important insights for the treatment of chronic gastritis and the prevention of gastric cancer. Heparanase is an endoglycosidase that degrades heparan sulfate, resulting in remodeling of the extracellular matrix thereby facilitating the extravasation and migration of immune cells towards sites of inflammation. Heparanase also releases heparan sulfate-bound cytokines and chemokines that further promote directed motility and recruitment of immune cells. Heparanase is highly expressed in a variety of inflammatory conditions and diseases, but its role in chronic gastritis has not been sufficiently explored. In this study, we report that H. pylori infection promotes up-regulation of heparanase in gastritis, which in turn facilitates the colonization of H. pylori in the gastric mucosa, thereby aggravating gastritis. By sustaining continuous activation, polarization and recruitment of macrophages that supply pro-inflammatory and pro-tumorigenic cytokines (i.e., IL-1, IL-6, IL-1ß, TNF-α, MIP-2, iNOS), heparanase participates in the generation of a vicious circle, driven by enhanced NFκB and p38-MAPK signaling, that supports the development and progression of gastric cancer. These results suggest that inhibition of heparanase may block this self-sustaining cycle, and thereby reduce the risk of gastritis and gastric cancer.


Asunto(s)
Mucosa Gástrica/microbiología , Gastritis/etiología , Glucuronidasa/fisiología , Helicobacter pylori/patogenicidad , Adulto , Animales , Polaridad Celular , Enfermedad Crónica , Femenino , Humanos , Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neoplasias Gástricas/etiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
11.
JCI Insight ; 6(12)2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34156029

RESUMEN

p38 MAPKs play a central role in orchestrating the cellular response to stress and inflammation and in the regulation of myogenesis. Potent inhibitors of p38 MAPKs have been pursued as potential therapies for several disease indications due to their antiinflammatory properties, although none have been approved to date. Here, we provide a brief overview of p38 MAPKs, including their role in regulating myogenesis and their association with disease progression. Finally, we discuss targeting p38 MAPKs as a therapeutic approach for treating facioscapulohumeral muscular dystrophy and other muscular dystrophies by addressing multiple pathological mechanisms in skeletal muscle.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Músculo Esquelético , Proteínas Quinasas p38 Activadas por Mitógenos , Animales , Humanos , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Desarrollo de Músculos/genética , Desarrollo de Músculos/fisiología , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiología , Distrofia Muscular Facioescapulohumeral/genética , Distrofia Muscular Facioescapulohumeral/metabolismo , Distrofia Muscular Facioescapulohumeral/fisiopatología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
12.
Commun Biol ; 4(1): 788, 2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-34172827

RESUMEN

Successful specification of the two mouse blastocyst inner cell mass (ICM) lineages (the primitive endoderm (PrE) and epiblast) is a prerequisite for continued development and requires active fibroblast growth factor 4 (FGF4) signaling. Previously, we identified a role for p38 mitogen-activated protein kinases (p38-MAPKs) during PrE differentiation, but the underlying mechanisms have remained unresolved. Here, we report an early blastocyst window of p38-MAPK activity that is required to regulate ribosome-related gene expression, rRNA precursor processing, polysome formation and protein translation. We show that p38-MAPK inhibition-induced PrE phenotypes can be partially rescued by activating the translational regulator mTOR. However, similar PrE phenotypes associated with extracellular signal-regulated kinase (ERK) pathway inhibition targeting active FGF4 signaling are not affected by mTOR activation. These data indicate a specific role for p38-MAPKs in providing a permissive translational environment during mouse blastocyst PrE differentiation that is distinct from classically reported FGF4-based mechanisms.


Asunto(s)
Blastocisto/fisiología , Endodermo/citología , Biosíntesis de Proteínas , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Animales , Diferenciación Celular , Linaje de la Célula , Proteínas de Unión al ADN/fisiología , Desarrollo Embrionario , Ratones , Proteínas de Unión al ARN/fisiología , Serina-Treonina Quinasas TOR/fisiología , Factores de Transcripción/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
13.
Commun Biol ; 4(1): 726, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34117353

RESUMEN

Argonaute 2 (AGO2) is an indispensable component of the RNA-induced silencing complex, operating at the translational or posttranscriptional level. It is compartmentalized into structures such as GW- and P-bodies, stress granules and adherens junctions as well as the midbody. Here we show using immunofluorescence, image and bioinformatic analysis and cytogenetics that AGO2 also resides in membrane protrusions such as open- and close-ended tubes. The latter are cytokinetic bridges where AGO2 colocalizes at the midbody arms with cytoskeletal components such as α-Τubulin and Aurora B, and various kinases. AGO2, phosphorylated on serine 387, is located together with Dicer at the midbody ring in a manner dependent on p38 MAPK activity. We further show that AGO2 is stress sensitive and important to ensure the proper chromosome segregation and cytokinetic fidelity. We suggest that AGO2 is part of a regulatory mechanism triggered by cytokinetic stress to generate the appropriate micro-environment for local transcript homeostasis.


Asunto(s)
Proteínas Argonautas/fisiología , División Celular , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Actinas/metabolismo , Proteínas Argonautas/metabolismo , Línea Celular , Citocinesis , Citoesqueleto/metabolismo , Técnica del Anticuerpo Fluorescente , Células HCT116 , Células Hep G2 , Humanos , Seudópodos/metabolismo , Tubulina (Proteína)/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
14.
Front Immunol ; 12: 676173, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34054865

RESUMEN

Increased interleukin (IL)-17A has been identified in joints affected by osteoarthritis (OA), but it is unclear how IL-17A, and its family members IL-17AF and IL-17F, can contribute to human OA pathophysiology. Therefore, we aimed to evaluate the gene expression and signalling pathway activation effects of the different IL-17 family members in chondrocytes and synovial fibroblasts derived from cartilage and synovium of patients with end-stage knee OA. Immunohistochemistry staining confirmed that IL-17 receptor A (IL-17RA) and IL-17RC are expressed in end-stage OA-derived cartilage and synovium. Chondrocytes and synovial fibroblasts derived from end-stage OA patients were treated with IL-17A, IL-17AF, or IL-17F, and gene expression was assessed with bulk RNA-Seq. Hallmark pathway analysis showed that IL-17 cytokines regulated several OA pathophysiology-related pathways including immune-, angiogenesis-, and complement-pathways in both chondrocytes and synovial fibroblasts derived from end-stage OA patients. While overall IL-17A induced the strongest transcriptional response, followed by IL-17AF and IL-17F, not all genes followed this pattern. Disease-Gene Network analysis revealed that IL-17A-related changes in gene expression in these cells are associated with experimental arthritis, knee arthritis, and musculoskeletal disease gene-sets. Western blot analysis confirmed that IL-17A significantly activates p38 and p65 NF-κB. Incubation of chondrocytes and synovial fibroblasts with anti-IL-17A monoclonal antibody secukinumab significantly inhibited IL-17A-induced gene expression. In conclusion, the association of IL-17-induced transcriptional changes with arthritic gene-sets supports a role for IL-17A in OA pathophysiology. Future studies should further investigate the role of IL-17A in the OA joint to establish whether anti-IL-17 treatment could be a potential therapeutic option in OA patients with an inflammatory phenotype.


Asunto(s)
Condrocitos/inmunología , Interleucina-17/fisiología , Osteoartritis de la Rodilla/etiología , Membrana Sinovial/inmunología , Anticuerpos Monoclonales Humanizados/farmacología , Células Cultivadas , Condrocitos/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/inmunología , Humanos , Interleucina-17/farmacología , FN-kappa B/fisiología , Osteoartritis de la Rodilla/inmunología , Receptores de Interleucina-17/análisis , Transducción de Señal/efectos de los fármacos , Membrana Sinovial/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
15.
Biomolecules ; 11(5)2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33922633

RESUMEN

Metastasis is a process by which cancer cells escape from the location of the primary tumor invading normal tissues at distant organs. Chromosomal instability (CIN) is a hallmark of human cancer, associated with metastasis and therapeutic resistance. The centrosome plays a major role in organizing the microtubule cytoskeleton in animal cells regulating cellular architecture and cell division. Loss of centrosome integrity activates the p38-p53-p21 pathway, which results in cell-cycle arrest or senescence and acts as a cell-cycle checkpoint pathway. Structural and numerical centrosome abnormalities can lead to aneuploidy and CIN. New findings derived from studies on cancer and rare genetic disorders suggest that centrosome dysfunction alters the cellular microenvironment through Rho GTPases, p38, and JNK (c-Jun N-terminal Kinase)-dependent signaling in a way that is favorable for pro-invasive secretory phenotypes and aneuploidy tolerance. We here review recent data on how centrosomes act as complex molecular platforms for Rho GTPases and p38 MAPK (Mitogen activated kinase) signaling at the crossroads of CIN, cytoskeleton remodeling, and immune evasion via both cell-autonomous and non-autonomous mechanisms.


Asunto(s)
Centrosoma/metabolismo , Inflamación/patología , Metástasis de la Neoplasia/patología , Aneuploidia , Animales , Ciclo Celular/fisiología , Puntos de Control del Ciclo Celular/fisiología , Centrosoma/fisiología , Inestabilidad Cromosómica/fisiología , Citoesqueleto/fisiología , Humanos , Inflamación/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Sistema de Señalización de MAP Quinasas/fisiología , Microtúbulos/metabolismo , Metástasis de la Neoplasia/genética , Neoplasias/metabolismo , Transducción de Señal , Microambiente Tumoral , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Proteínas de Unión al GTP rho/metabolismo
16.
Transl Res ; 235: 115-128, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33857660

RESUMEN

Sepsis represents a life-threatening event often mediated by the host's response to pathogens such as gram-negative organisms, which release the proinflammatory lipopolysaccharide (LPS). Within the endothelium, the mitogen-activated protein kinase (MAPK) pathway is an important driver of endothelial injury during sepsis, of which oxidant-sensitive apoptosis signal-regulating kinase 1 (ASK1) is postulated to be a critical upstream regulator. We hypothesized that ASK1 would play a key role in endothelial inflammation during bacterial challenge. Utilizing RNA sequencing data from patients and cultured human microvascular endothelial cells (HMVECs), ASK1 expression was increased in sepsis and after LPS challenge. Two ASK1 inhibitors, GS444217 and MSC2023964A, reduced cytokine production in HMVECs following LPS stimulation, but had no effect on permeability as measured by transendothelial electrical resistance and intercellular space. MAPKs are known to interact with endothelial nitric oxide synthase (eNOS) and ASK1 expression levels correlated with eNOS expression in patients with septic shock. In addition, eNOS physically interacted with ASK1, though this interaction was not altered by ASK1 inhibition, nor did inhibition alter MAPK p38 activity. Instead, among MAPKs, ASK1 inhibition only impaired LPS-induced JNK phosphorylation. The reduction in JNK activation caused by ASK1 inhibition impaired JNK-mediated cytokine production without affecting permeability. Thus, LPS triggers JNK-dependent cytokine production that requires ASK1 activation, but both its effects on permeability and activation of p38 are ASK1-independent. These data demonstrate how distinct MAPK signaling pathways regulate endothelial inflammatory outputs during acute infectious challenge.


Asunto(s)
Citocinas/biosíntesis , Células Endoteliales/metabolismo , MAP Quinasa Quinasa Quinasa 5/fisiología , Receptor Toll-Like 4/fisiología , Células Cultivadas , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , MAP Quinasa Quinasa Quinasa 5/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas/fisiología , Óxido Nítrico Sintasa de Tipo III/fisiología , Permeabilidad , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
17.
Drug Dev Res ; 82(8): 1131-1143, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33818788

RESUMEN

Circular RNA FAT atypical cadherin 1 (circFAT1) has been reported to play vital roles in the progression of some cancers. However, the regulatory role and underlying mechanisms of circFAT1 in cervical cancer (CC) remain largely unknown. The expression of circFAT1, microRNA (miR)-409-3p and cyclin-dependent kinase 8 (CDK8) was detected using qRT-PCR and Western blot assays. Cell proliferation, apoptosis, migration and invasion in vitro were investigated using cell counting kit-8, colony formation, flow cytometry, and transwell assays, respectively. Western blot assay was used to determine the activation of ERK1/2 and p38 MAPK pathway. The interaction miR-409-3p and circFAT1 or CDK8 was confirmed by dual-luciferase reporter, pull-down or RIP assays. The effects of circFAT1 in vivo were determined using xenograft models. CircFAT1 was highly expressed in CC, and closely associated with poor prognosis. CircFAT1 knockdown resulted in the suppression of proliferation, migration and invasion, and promotion of apoptosis in CC cells via the inactivation of ERK1/2 and p38 MAPK pathway; also, circFAT1 silencing could inactivate this pathway and repressed CC tumor growth in vivo. Mechanistic analysis showed that circFAT1 directly sponged miR-409-3p and then relieved the repressive effect of miR-409-3p on its target CDK8. Furthermore, miR-409-3p inhibition reversed the effects of circFAT1 silencing on CC cells. Whereas, miR-409-3p overexpression impeded CC cell growth and motility, which was attenuated by CDK8. CircFAT1 promoted CC progression via activating ERK1/2 and p38 MAPK pathway through the miR-409-3p/CDK8 axis, suggesting a promising prognostic biomarker and therapeutic target for CC.


Asunto(s)
Cadherinas/genética , Quinasa 8 Dependiente de Ciclina/fisiología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , MicroARNs/fisiología , ARN Circular/fisiología , Neoplasias del Cuello Uterino/etiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Animales , Línea Celular Tumoral , Quinasa 8 Dependiente de Ciclina/genética , Progresión de la Enfermedad , Femenino , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos BALB C
18.
FASEB J ; 35(3): e21393, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33570794

RESUMEN

UV irradiation can injure the epidermis, resulting in sunburn, inflammation, and cutaneous tissue disorders. Previous studies demonstrate that EGFR in keratinocytes can be activated by UVB and contributes to inflammation. Poly (ADP-ribose) polymerase-1 (PARP-1) is a nuclear enzyme and plays an essential role in DNA repair under moderate stress. In this study, we set out to understand how PARP-1 regulates UVB irradiation-induced skin injury and interplays with EGFR to mediate the inflammation response. We found that PARP-1 deficiency exacerbated the UVB-induced inflammation, water loss, and back skin damage in mice. In human primary keratinocytes, UVB can activate PARP-1 and enhance DNA damage upon PARP-1 gene silencing. Moreover, PARP-1 silencing and PARP inhibitor olaparib can suppress UVB-induced COX-2 and MMP-1 expression, but enhance TNF-α and IL-8 expression. In addition, EGFR silencing or EGFR inhibition by gefitinib can decrease UVB-induced COX-2, TNF-α, and IL-8 expression, suggesting EGFR activation via paracrine action can mediate UVB-induced inflammation responses. Immunoblotting data revealed that PARP-1 inhibition decreases UVB-induced EGFR and p38 activation. Pharmacological inhibition of p38 also dramatically led to the attenuation of UVB-induced inflammatory gene expression. Of note, genetic ablation of PARP-1 or EGFR can attenuate UVB-induced ROS production, and antioxidant NAC can attenuate UVB-induced EGFR-p38 signaling axis and PARP-1 activation. These data suggest the regulatory loops among EGFR, PARP-1, and ROS upon UVB stress. PARP-1 not only serves DNA repair function but also orchestrates interactions to EGFR transactivation and ROS production, leading to p38 signaling for inflammatory gene expression in keratinocytes.


Asunto(s)
Receptores ErbB/fisiología , Inflamación/etiología , Queratinocitos/efectos de la radiación , Poli(ADP-Ribosa) Polimerasa-1/fisiología , Especies Reactivas de Oxígeno/metabolismo , Piel/efectos de la radiación , Activación Transcripcional , Rayos Ultravioleta , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Animales , Células Cultivadas , Ciclooxigenasa 2/genética , Reparación del ADN , Receptores ErbB/genética , Humanos , Interleucina-8/genética , Ratones , Transducción de Señal/fisiología
19.
Front Immunol ; 12: 768813, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34975857

RESUMEN

Ventilator-induced lung injury (VILI) is one of the most common complications of mechanical ventilation and can severely affect health. VILI appears to involve excessive inflammatory responses, but its pathogenesis has not yet been clarified. Since interleukin-17 (IL-17) plays a critical role in the immune system and the development of infectious and inflammatory diseases, we investigated here whether it plays a role in VILI. In a mouse model of VILI, mechanical ventilation with high tidal volume promoted the accumulation of lung neutrophils, leading to increased IL-17 levels in the lung, which in turn upregulated macrophage chemoattractant protein-1 via p38 mitogen-activated protein kinase. Depletion of neutrophils decreases the production IL-17 in mice and inhibition of IL-17 significantly reduced HTV-induced lung injury and inflammatory response. These results were confirmed in vitro using RAW264.7 macrophage cultures. Our results suggest that IL-17 plays a pro-inflammatory role in VILI and could serve as a new target for its treatment.


Asunto(s)
Quimiocina CCL2/fisiología , Interleucina-17/fisiología , Neutrófilos/fisiología , Lesión Pulmonar Inducida por Ventilación Mecánica/etiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Animales , Interleucina-17/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Células RAW 264.7 , Respiración Artificial/efectos adversos , Transducción de Señal/fisiología
20.
Microbiol Immunol ; 65(1): 40-47, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33026667

RESUMEN

The ischemia-reperfusion-induced damage in human brain microvascular endothelial cells (BMECs) is associated with disruption of the blood-brain barrier. CXC chemokine ligand 5 (CXCL5) is reported to be up-regulated in ischemic stroke. However, the detailed function of CXCL5 in this pathological process remains largely unclear. To further analyze the function of CXCL5 in ischemic stroke, an oxygen-glucose deprivation model on human BMECs was constructed to mimic the ischemic stroke condition in vitro. Cell proliferation was analyzed using a cell counting kit-8 (CCK-8) assay. Quantitative real-time polymerase chain reaction and western blot were utilized to determine gene expression. The barrier function of BMECs was assessed using a fluorescently labeled dextran assay and a trans-epithelial/endothelial electrical resistance (TEER) technique. The results indicated that CXCL5 antibody (anti-CXCL5) promoted the proliferation of model cells, whereas it reduced the permeability. Moreover, the TEER value of model cells was enhanced in the presence of anti-CXCL5. Therefore, these findings demonstrated that CXCL5 silencing attenuated the ischemic/hypoxic-induced injury in human BMECs. Importantly, human recombinant protein CXCL5 (Re-CXCL5) deeply disrupted the function of BMECs in the normoxic condition. Furthermore, the p38 inhibitor SB203580 significantly abolished the function of CXCL5 in model cells. More importantly, similar results were also obtained in BMECs under normoxic conditions in the presence of Re-CXCL5. These results indicated that CXCL5 might regulate the function of BMECs by mediating the p38 pathway. This investigation not only enhanced the understanding of the biological effect of CXCL5 in human BMECs under ischemic/hypoxic conditions but also indicated its potential value as a therapeutic target for ischemic-induced brain disease.


Asunto(s)
Encéfalo , Quimiocina CXCL5/fisiología , Células Endoteliales , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Encéfalo/citología , Células Cultivadas , Células Endoteliales/citología , Humanos , Ligandos , Permeabilidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA