Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.494
Filtrar
1.
J Pharm Pharmacol ; 74(8): 1180-1192, 2022 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-35704284

RESUMEN

OBJECTIVES: To explore clonidine (Clon) nephroprotective effects as an inhibitor of organic cationic transporter 2 (OCT2) and p38 mitogen-activated protein kinase (p38 MAPK) against cisplatin (CP)-induced nephrotoxicity. OCT2 is mainly responsible for renal accumulation of CP. Clon has been recently recognized as an OCT2 inhibitor and exerts beneficial effects on renal function and p38 MAPK. This study further investigates its underlying anti-inflammatory, antioxidative and antiapoptotic effects. METHODS: Rats were randomly assigned into five groups: (I) CON, (II) CP, (III) CP + Clon 0.125, (IV) CP + Clon 0.25, (V) CP + Clon 0.5, and (VI) Clon 0.5 alone. Clon was administered orally at 0.125, 0.25 and 0.5 mg/kg/day dosages for 10 days. On day 7, rats in groups from (II) to (V) received a single intraperitoneal injection of CP (10 mg/kg). KEY FINDINGS: Clon 0.25 mg/kg displayed the best nephroprotective outcomes, justified by the significant amelioration of parameters like renal function, oxidative stress, and inflammatory status, as well as modulated the OCT2 expression, phosphorylation of p38 and p53, compared with Clon 0.125 and 0.5 mg/kg. CONCLUSION: This study suggests the promising nephroprotective impact of Clon as an OCT2 inhibitor against CP nephrotoxicity and its proficient role in attenuating oxidative stress, inflammatory status and apoptotic status.


Asunto(s)
Antineoplásicos , Cisplatino , Clonidina , Transportador 2 de Cátion Orgánico , Proteínas Quinasas p38 Activadas por Mitógenos , Animales , Antineoplásicos/toxicidad , Cisplatino/toxicidad , Clonidina/farmacología , Riñón/efectos de los fármacos , Transportador 2 de Cátion Orgánico/antagonistas & inhibidores , Estrés Oxidativo , Ratas , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
2.
Int J Mol Sci ; 23(3)2022 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-35163030

RESUMEN

c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK) family members integrate signals that affect proliferation, differentiation, survival, and migration in a cell context- and cell type-specific way. JNK and p38 MAPK activities are found upregulated in nasopharyngeal carcinoma (NPC). Studies have shown that activation of JNK and p38 MAPK signaling can promote NPC oncogenesis by mechanisms within the cancer cells and interactions with the tumor microenvironment. They regulate multiple transcription activities and contribute to tumor-promoting processes, ranging from cell proliferation to apoptosis, inflammation, metastasis, and angiogenesis. Current literature suggests that JNK and p38 MAPK activation may exert pro-tumorigenic functions in NPC, though the underlying mechanisms are not well documented and have yet to be fully explored. Here, we aim to provide a narrative review of JNK and p38 MAPK pathways in human cancers with a primary focus on NPC. We also discuss the potential therapeutic agents that could be used to target JNK and p38 MAPK signaling in NPC, along with perspectives for future works. We aim to inspire future studies further delineating JNK and p38 MAPK signaling in NPC oncogenesis which might offer important insights for better strategies in diagnosis, prognosis, and treatment decision-making in NPC patients.


Asunto(s)
Antineoplásicos/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Carcinoma Nasofaríngeo/tratamiento farmacológico , Neoplasias Nasofaríngeas/tratamiento farmacológico , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Humanos , Carcinoma Nasofaríngeo/enzimología , Carcinoma Nasofaríngeo/patología , Neoplasias Nasofaríngeas/enzimología , Neoplasias Nasofaríngeas/patología
3.
Anticancer Res ; 42(2): 745-757, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35093873

RESUMEN

BACKGROUND/AIM: Tumour repopulation is a major obstacle for successful cancer treatment. This study investigated whether anticancer agents contribute to tumour repopulation in TP53-mutated bile duct cancer cells. MATERIALS AND METHODS: TP53-mutated HuCCT1 and HuH28 cells were exposed to anticancer agents, and recipient cells were exposed to their conditioned media or exosomes. The effect of inhibitors and siRNA-mediated gene silencing of p38 mitogen-activated protein kinase (MAPK) and of TP53 was analyzed by cell proliferation assays and western blotting. RESULTS: Conditioned media from genotoxic agent-treated cells promoted proliferation of recipient cells (p<0.05), and this effect was abrogated by exosome inhibitors. Exosomes from gemcitabine- or cisplatin-treated cells increased cell proliferation by 1.6- to 2.2-fold (p<0.05) through p38 MAPK signalling. These effects of exosomes were inhibited by inhibition/silencing of p38 MAPK but not by TP53 silencing. CONCLUSION: Exosomal p38 MAPK plays a pivotal role in tumour repopulation in a TP53-independent manner.


Asunto(s)
Neoplasias de los Conductos Biliares/metabolismo , Neoplasias de los Conductos Biliares/patología , Exosomas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Antineoplásicos/farmacología , Neoplasias de los Conductos Biliares/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados , Exosomas/efectos de los fármacos , Silenciador del Gen , Humanos , Modelos Biológicos , Mutación , Estrés Oxidativo/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética
4.
Anticancer Res ; 42(1): 555-563, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34969765

RESUMEN

BACKGROUND: Epstein-Barr virus (EBV)-associated gastric cancer has been identified as a cancer subtype with definitive clinical and molecular characteristics. Although olaparib, a poly ADP ribose polymerase (PARP) inhibitor, is considered a potential effective agent for gastric cancer, the effect and underlying mechanism of olaparib on gastric cancer depending on EBV infection is not fully understood. MATERIALS AND METHODS: EBV-positive SNU719 and EBV-negative SNU638 gastric cancer cell lines were used to identify the effects of olaparib using the trypan blue exclusion method and annexin V staining assay. To observe the underlying cellular signaling mechanisms of olaparib-induced cell death, Epstein-Barr virus nuclear antigen 1 (EBNA1) and signaling related molecule expression were assessed using transfection, silencing of specific genes using small interfering RNA (siRNA), western blotting and signaling inhibition assay. RESULTS: Olaparib decreased the cell viability of EBV-positive SNU719 gastric cancer cells through caspase-3-dependent apoptosis in a dose dependent manner, whereas EBV-negative SNU638 gastric cancer cells showed drug resistance to olaparib. EBNA1 was expressed in SUN719 gastric cancer cells; however, ataxia telangiectasia and Rad3 related (ATR) and phosphorylated ATR kinase were expressed in SNU638 gastric cancer cells. EBNA1 transfection decreased ATR phosphorylation through p38 mitogen-activated protein kinase (MAPK) phosphorylation in SUN638 gastric cancer cells, and silencing of ATR kinase increased the susceptibility of these cells to olaparib treatment. Moreover, VE-821, an ATR kinase specific inhibitor, also increased the sensitivity of SNU638 cells to olaparib. In contrast, SB203580, a p38 MAPK inhibitor, inhibited this increase in sensitivity to olaparib by EBNA1 transfection. CONCLUSION: Olaparib treatment led to different cellular responses depending on EBV infection in gastric cancer cell lines. These results provide new insights into the mechanism of olaparib-induced apoptosis in gastric cancer cells and suggest that EBV infection should be considered when developing new potential therapeutic agents for gastric cancer.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/genética , Antígenos Nucleares del Virus de Epstein-Barr/genética , Ftalazinas/farmacología , Piperazinas/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Infecciones por Virus de Epstein-Barr/tratamiento farmacológico , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/patología , Infecciones por Virus de Epstein-Barr/virología , Antígenos Nucleares del Virus de Epstein-Barr/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Herpesvirus Humano 4/aislamiento & purificación , Herpesvirus Humano 4/patogenicidad , Humanos , Transducción de Señal/efectos de los fármacos , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Neoplasias Gástricas/virología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
5.
J Biomol Struct Dyn ; 40(13): 6114-6127, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-33522434

RESUMEN

The hallmark of the Alzheimer's disease (AD) is the accumulation of aggregated, misfolded proteins. The cause for this accumulation is increased production of misfolded proteins and impaired clearance of them. Amyloid aggregation and tau hyperphosphorylation are the two proteinopathies which accomplish deprivation of cell and tissue hemostasis during neuropathological process of the AD, as a result of which progressive neuronal degeneration and the loss of cognitive functions. p38 mitogen-activated protein kinase (p38 MAPK) has been implicated in both the events associated with AD: tau protein phosphorylation and inflammation. p38α MAPK pathway is activated by a dual phosphorylation at Thr180 and Tyr182 residues. Clinical and preclinical evidence implicates the stress related kinase p38α MAPK as a potential neurotherapeutic target. Drug design of p38α MAPK inhibitors is mainly focused on small molecules that compete for Adenosine triphosphate in the catalytic site. Here we have carried out the synthesis of phenyl sulfonamide derivatives Sulfo (I) and Sulfo (II). Crystal structures of Sulfo (I) and Sulfo (II) were solved by direct methods using SHELXS-97. Sulfo (I) and Sulfo (II) have Rint values of 0.0283 and 0.0660, respectively, indicating good quality of crystals and investigated their ability against p38α MAPK. Docking studies revealed that the Sulfo (I) had better binding affinity (-62.24 kcal/mol) as compared to Sulfo (II) and cocrystal having binding affinity of -54.61 kcal/mol and -59.84 kcal/mol, respectively. Molecular dynamics simulation studies of Sulfo (I) and cocrystal of p38α MAPK suggest that during the course of 30 ns simulation run, compound Sulfo (I) attained stability, substantiating the consistency of its binding to p38α MAPK compared to cocrystal. Binding free energy analysis suggests that the compound Sulfo (I) is better than the cocrystal. Thus, this study corroborates the therapeutic potential of synthesized Sulfo (I) in combatting AD.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Enfermedad de Alzheimer , Inhibidores de Proteínas Quinasas , Proteínas Quinasas p38 Activadas por Mitógenos , Enfermedad de Alzheimer/tratamiento farmacológico , Humanos , Simulación de Dinámica Molecular , Inhibidores de Proteínas Quinasas/química , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
6.
J Med Chem ; 65(2): 1225-1242, 2022 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-33974419

RESUMEN

Stress-induced p38α mitogen-activated protein (MAP) kinase activation modulates cytokine overproduction and is associated with neuroinflammation and neurodegeneration. As a potential therapeutic approach, novel Skepinone-based p38α MAP kinase inhibitors were optimized to cross the blood-brain barrier via either amino acid transporters or hydrophobic diffusion. To enhance absorption from the oral route, we used methyl ester prodrugs of the active carboxy analogs. Of these, 3-(8-((2,4-difluorophenyl)amino)-5-oxo-10,11-dihydro-5H-dibenzo[a,d][7]annulene-3-carboxamido)propanoic acid (43; p38α, IC50 = 5.5 nM) and 4-(8-((2,4-difluorophenyl)amino)-5-oxo-10,11-dihydro-5H-dibenzo[a,d][7]annulene-3-carboxamido)butanoic acid (44; p38α, IC50 = 12 nM) had brain-to-plasma ratios of 1.4 and 4.4, respectively. Compound 70, 3-(8-((2-aminophenyl)amino)-5-oxo-10,11-dihydro-5H-dibenzo[a,d][7]annulene-3-carboxamido)propanoic acid (p38α, IC50 = 1.0 nM), the Skepinone-N counterpart of 43, was most present in the mouse brain (brain-to-plasma ratio of 4.7; 0.4 mg/kg p.o., 2 h, 580 nmol/kg). Compounds 43, 44, and 70 were p38α-MAP-kinase-selective, metabolically stable, hERG nonbinding, and able to modulate IL-6 and TNF-α production in cell-based assays.


Asunto(s)
Encéfalo/metabolismo , Diseño de Fármacos , Simulación del Acoplamiento Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/química , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Encéfalo/efectos de los fármacos , Femenino , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Estructura Molecular , Proteínas Oncogénicas/metabolismo , Regulador Transcripcional ERG/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
7.
J Ovarian Res ; 14(1): 162, 2021 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-34789301

RESUMEN

BACKGROUND: Resistance to platinum-based chemotherapy is one of the crucial problems in ovarian cancer treatment. Ghrelin, a widely distributed peptide hormone, participates in a series of cancer progression. The aim of this study is to determine whether ghrelin influences the sensitivity of ovarian cancer to cisplatin, and to demonstrate the underlying mechanism. METHODS: The anti-tumor effects of ghrelin and cisplatin were evaluated with human ovarian cancer cells HO-8910 PM in vitro or in vivo. Cell apoptosis and cell cycle were analyzed via flow cytometry assay. The signaling pathway and the expression of cell cycle protein were analyzed with Western Blot. RESULTS: Our results showed that treatment with ghrelin specifically inhibited cell proliferation of HO-8910 PM and sensitized these cells to cisplatin via S phase cell cycle arrest, and enhanced the inhibitory effect of cisplatin on tumor growth of HO-8910 PM derived xenografts in vivo. Treatment with ghrelin inhibited the expression of p-Erk1/2 and p-p38, which was opposite the effect of cisplatin. However, under the treatment of ghrelin, cisplatin treatment exhibited a stronger effect on inhibiting P21 expression, upregulating p-CDK1 and cyclin B1 expression, and blocking cell cycle progression. Mechanistically, ghrelin promoted S phase cell cycle arrest and upregulated p-CDK1 and cyclin B1 expression induced by cisplatin via inhibition of p38. CONCLUSION: This study revealed a specifically inhibitory effect of ghrelin on platinum-resistance via suppressing p-P38 and subsequently promoting p-CDK1 mediated cell cycle arrest in HO-8910 PM.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Ghrelina/análogos & derivados , Neoplasias Ováricas/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Proteína Quinasa CDC2/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/uso terapéutico , Inhibidores Enzimáticos/farmacología , Femenino , Ghrelina/farmacología , Ghrelina/uso terapéutico , Humanos , Ratones , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
8.
Exp Cell Res ; 409(2): 112938, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34800541

RESUMEN

In lactating mammary glands, alveolar mammary epithelial cells (MECs) produce milk and form less-permeable tight junctions (TJs). However, alveolar TJs are weakened with a reduction in milk production in mammary glands due to mastitis or weaning in the presence of high levels of IL-1ß, IL-6, or TNF-α. In this study, using in vitro cultured model of MECs with milk-producing ability and lactation-specific TJs, we investigated whether the aforementioned cytokines affect MEC TJs. The results showed that TNF-α, IL-1ß, and IL-6 affected lactation-specific TJs in different ways. In particular, upon activation of p38 and JNK signalling, IL-1ß caused rapid disruption of TJs at tricellular contact points. IL-1ß treatment led to decreased CLDN3, CLDN4, and OCLN levels and a weakened TJ barrier. The adverse effects of IL-1ß on TJs were mimicked by anisomycin, which is an activator of p38 and JNK signalling, and were blocked by MEC pretreatment with a p38 inhibitor but not a JNK inhibitor. The mislocalization of tricellulin at tricellular contact areas was confirmed in MECs treated with IL-1ß or anisomycin. These results indicate that IL-1ß is a key cytokine that adversely affects the TJs between MECs by activating p38.


Asunto(s)
Anisomicina/farmacología , Claudina-3/metabolismo , Claudina-4/metabolismo , Interleucina-1beta/farmacología , Lactancia , Glándulas Mamarias Animales/patología , Uniones Estrechas/patología , Animales , Claudina-3/genética , Claudina-4/genética , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Glándulas Mamarias Animales/metabolismo , Ratones , Leche/química , Inhibidores de la Síntesis de la Proteína/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
9.
Sci Rep ; 11(1): 21604, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34732784

RESUMEN

Oxidative stress-induced dopaminergic neuronal loss and apoptosis play a crucial role in the pathogenesis of Parkinson's disease (PD), and as a vital antioxidant protein, thioredoxin (Trx) exerts neuroprotection against PD. In this study, we investigated the effect of Schisanhenol (Sal), an active component from a traditional Chinese herb Schisandra rubriflora (Franch.), on MPP+-induced apoptosis and its association with thioredoxin-1 (Trx1) in SH-SY5Y cells. The protein levels of Trx1 and apoptosis-related proteins were detected by Western blot, the expression of Trx1 mRNA by real time qPCR, and apoptosis was detected by fluorescence microscopy and flow cytometry. Pretreatment with Sal (1 µM, 10 µM, and 50 µM) dose-dependently ameliorated MPP+-induced neuronal injury, confirmed by the improvement of the viability and morphological changes. Sal decreased the apoptosis rate of cells, suppressed the production of DNA ladder and sub-G1 peak, inhibited the Caspase-3 activity and the expression of apoptosis-related proteins. Sal enhanced the expression of Trx1 both in the protein and mRNA levels. However, the Trx1 inhibitor PX-12 suppressed the protective effects of Sal. In addition, Sal inhibited NF-κB translocation and activation. These results suggest that Sal has a protective effect against MPP+-induced apoptosis in SH-SY5Y cells via up-regulation of Trx1 expression and suppression of ASK1-P38-NF-κB pathway.


Asunto(s)
1-Metil-4-fenilpiridinio/efectos adversos , Ciclooctanos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , MAP Quinasa Quinasa Quinasa 5/antagonistas & inhibidores , FN-kappa B/antagonistas & inhibidores , Neuroblastoma/patología , Compuestos Policíclicos/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Herbicidas/efectos adversos , Humanos , MAP Quinasa Quinasa Quinasa 5/genética , MAP Quinasa Quinasa Quinasa 5/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroprotección , Células Tumorales Cultivadas , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Toxicol Lett ; 352: 61-69, 2021 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-34624459

RESUMEN

Mitomycin treatment induces pulmonary toxicity, and alveolar epithelial cell senescence is crucial in the pathogenesis of the latter. However, the mechanism by which mitomycin induces alveolar epithelial cell senescence has yet to be elucidated. In this work, different doses (37.5-300 nM) of mitomycin induced the senescence of human alveolar type II-like epithelial cells and enhanced the phosphorylation of GSK3ß (S9). The GSK3ß (S9A) mutant reversed the senescence of mitomycin-treated alveolar epithelial cells. Pharmacological inhibition and gene deletion of Akt1, a kinase that regulates the phosphorylation of GSK3ß (S9), suppressed mitomycin-induced alveolar epithelial cell senescence. The knockdown of p53, a downstream effector of GSK3ß and an important regulator of cell senescence, repressed mitomycin-induced alveolar epithelial cell senescence. Treatment with baicalein weakened the phosphorylation of GSK3ß (S9) and alleviated the senescence of alveolar epithelial cells brought about by mitomycin treatment. GSK3ß (S9) phosphorylation appears to be the first signal involved in the mitomycin-induced senescence of alveolar epithelial cells and may present a potential target for attenuating mitomycin-induced pulmonary toxicity.


Asunto(s)
Alquilantes/toxicidad , Regulación hacia Abajo/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Mitomicina/toxicidad , Alveolos Pulmonares/efectos de los fármacos , Células A549 , Senescencia Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Flavanonas/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/genética , Humanos , Imidazoles/farmacología , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Alveolos Pulmonares/citología , Piridinas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Life Sci ; 287: 120092, 2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34715142

RESUMEN

AIMS: Transforming growth factor-ß (TGF-ß) mediates fibrotic manifestations of diabetic nephropathy. We demonstrated proteasomal degradation of anti-fibrotic protein, nuclear factor-erythroid derived 2 (NF-E2), in TGF-ß treated human renal proximal tubule (HK-11) cells and in diabetic mouse kidneys. The current study examined the role of mitogen-activated protein kinase (MAPK) pathways in mediating NF-E2 proteasomal degradation and stimulating profibrotic signaling in HK-11 cells. MAIN METHODS: HK-11 cells were pretreated with vehicle or appropriate proteasome and MAPK inhibitors, MG132 (0.5 µM), SB203580 (1 µM), PD98059 (25 µM) and SP600125 (10 µM), respectively, followed by treatment with/without TGF-ß (10 ng/ml, 24 h). Cell lysates and kidney homogenates from FVB and OVE26 mice treated with/without MG132 were immunoblotted with appropriate antibodies. pUse vector and pUse-NF-E2 cDNA were transfected in HK-11 cells and effects of TGF-ß on JNK MAPK phosphorylation (pJNK) was examined. KEY FINDINGS: We demonstrated activation of p38, ERK, and JNK MAPK pathways in TGF-ß treated HK-11 cells. Dual p38 and ERK MAPK blockade prevented TGF-ß-induced pSer82Hsp27, fibronectin and connective tissue growth factor (CTGF) expression while preserving NF-E2 expression. Blockade of JNK MAPK inhibited TGF-ß-induced CTGF expression without preserving NF-E2 expression. MG132 treatment prevented TGF-ß-induced pJNK in HK-11 cells and in type 1 diabetic OVE26 mouse kidneys, demonstrating that TGF-ß- and diabetes-induced pJNK occurs downstream of proteasome activation. A direct role for NF-E2 in modulating pJNK activation was demonstrated by NF-E2 over-expression. SIGNIFICANCE: ERK and p38 MAPK promotes NF-E2 proteasomal degradation while proteasome activation promotes pJNK and profibrotic signaling in renal proximal tubule cells.


Asunto(s)
Túbulos Renales Proximales/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Subunidad p45 del Factor de Transcripción NF-E2/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Antracenos/farmacología , Línea Celular Transformada , Inhibidores de Cisteína Proteinasa/farmacología , Femenino , Fibrosis , Humanos , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Leupeptinas/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Transgénicos
12.
Neurochem Int ; 150: 105190, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34537318

RESUMEN

Epidermal growth factor receptor (EGFR) activation is involved in blood spinal cord barrier (BSCB) disruption and secondary injury after spinal cord injury (SCI). However, the underlying mechanisms of EGFR activation mediating BSCB disruption and secondary injury after SCI remain unclear. An in vitro model of oxygen and glucose deprivation/reoxygenation (OGD/R) induced BSCB damage and in vivo rat SCI model were employed to define the role of EGFR/p38/NF-κB signal pathway activation and its induced inflammatory injury in main cellular components of BSCB. Genetic regulation (lentivirus delivered shRNA and overexpression system) or chemical intervention (agonist or inhibitor) were applied to activate or inactivate EGFR and p38 in astrocytes and microvascular endothelial cells (MEC) under which conditions, the expression of pro-inflammatory factors (TNF-α, iNOS, COX-2, and IL-1ß), tight junction (TJ) protein (ZO-1 and occludin), nuclear translocation of NF-κB and permeability of BSCB were analyzed. The pEGFR was increased in astrocytes and MEC which induced the activation of EGFR and p38 and NF-κB nuclear translocation. The activation of EGFR and p38 increased the TNF-α, iNOS, COX-2, and IL-1ß responsible for the inflammatory injury and reduced the ZO-1 and occludin which caused BSCB disruption. While EGFR or p38 inactivation inhibited NF-κB nuclear translocation, and markedly attenuated the production of pro-inflammatory factors and the loss of TJ protein. This study suggests that the EGFR activation in main cellular components of BSCB after SCI mediates BSCB disruption and secondary inflammatory injury via the EGFR/p38/NF-κB pathway.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Mediadores de Inflamación/antagonistas & inhibidores , FN-kappa B/antagonistas & inhibidores , Traumatismos de la Médula Espinal/prevención & control , Médula Espinal/irrigación sanguínea , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Células Cultivadas , Endotelio Vascular/metabolismo , Receptores ErbB/metabolismo , Femenino , Mediadores de Inflamación/metabolismo , FN-kappa B/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Vértebras Torácicas/lesiones , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Brain Tumor Pathol ; 38(3): 189-200, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34231121

RESUMEN

Current conventional treatment strategies for glioblastoma (GBM) have limited efficacy due to the rapid development of resistance to temozolomide (TMZ). It is particularly urgent to develop novel therapeutic strategies that can overcome TMZ resistance and provide patients with better prognoses. Here, a TMZ-resistant GBM cell strain and a mouse model of TMZ resistance are established as valuable tools to explore novel therapeutic strategies against TMZ resistance. Experimentally, p38MAPK inhibitor reduces the accumulation of F4/80+/CD11b+ macrophages/microglia in glioma and prolongs the survivals of glioma-bearing mice. Glioma-associated macrophages/microglia have a significanct expression of PD-L1. p38MAPK inhibitor in combination with PD-L1 antibody can effectively prolongs the survivals of TMZ-resistant GBM-bearing hosts, and differentially reduces the accumulation of circulating monocytes-derived tumor-associated macrophages and PD-L1 abundances of resident glioma-associated microglia. This combination therapy could be a treatment option for patients at the recurrence or chronic TMZ maintenance stages. A clinical study to confirm the safety and effectiveness of this combination therapy is warranted.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Alquilantes/farmacología , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Glioblastoma/genética , Glioblastoma/patología , Microglía/metabolismo , Temozolomida/farmacología , Macrófagos Asociados a Tumores/patología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Antineoplásicos Alquilantes/uso terapéutico , Antígeno B7-H1/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/genética , Quimioterapia Combinada , Glioblastoma/tratamiento farmacológico , Glioblastoma/mortalidad , Humanos , Ratones , Tasa de Supervivencia
14.
J Gen Virol ; 102(7)2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34328830

RESUMEN

The 5' capped, message-sense RNA genome of Chikungunya virus (CHIKV) utilizes the host cell machinery for translation. Translation is regulated by eIF2 alpha at the initiation phase and by eIF4F at cap recognition. Translational suppression by eIF2 alpha phosphorylation occurs as an early event in many alphavirus infections. We observe that in CHIKV-infected HEK293 cells, this occurs as a late event, by which time the viral replication has reached an exponential phase, implying its minimal role in virus restriction. The regulation by eIF4F is mediated through the PI3K-Akt-mTOR, p38 MAPK and RAS-RAF-MEK-ERK pathways. A kinetic analysis revealed that CHIKV infection did not modulate AKT phosphorylation, but caused a significant reduction in p38 MAPK phosphorylation. It caused degradation of phospho-ERK 1/2 by increased autophagy, leaving the PI3K-Akt-mTOR and p38 MAPK pathways for pharmacological targeting. mTOR inhibition resulted in moderate reduction in viral titre, but had no effect on CHIKV E2 protein expression, indicating a minimal role of the mTOR complex in virus replication. Inhibition of p38 MAPK using SB202190 caused a significant reduction in viral titre and CHIKV E2 and nsP3 protein expression. Furthermore, inhibiting the two pathways together did not offer any synergism, indicating that inhibiting the p38 MAPK pathway alone is sufficient to cause restriction of CHIKV replication. Meanwhile, in uninfected cells the fully functional RAS-RAF-MEK-ERK pathway can circumvent the effect of p38 MAPK inhibition on cap-dependent translation. Thus, our results show that host-directed antiviral strategies targeting cellular p38 MAPK are worth exploring against Chikungunya as they could be selective against CHIKV-infected cells with minimal effects on uninfected host cells.


Asunto(s)
Autofagia , Virus Chikungunya/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Imidazoles/farmacología , Biosíntesis de Proteínas , Piridinas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Apoptosis , Línea Celular Tumoral , Virus Chikungunya/genética , Virus Chikungunya/fisiología , Regulación hacia Abajo , Inhibidores Enzimáticos/farmacología , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Caperuzas de ARN , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Replicación Viral/efectos de los fármacos
15.
Cell Rep ; 36(4): 109421, 2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34320342

RESUMEN

Mitogen-activated protein kinases (MAPKs) are inactivated by dual-specificity phosphatases (DUSPs), the activities of which are tightly regulated during cell differentiation. Using knockdown screening and single-cell transcriptional analysis, we demonstrate that DUSP4 is the phosphatase that specifically inactivates p38 kinase to promote megakaryocyte (Mk) differentiation. Mechanistically, PRMT1-mediated methylation of DUSP4 triggers its ubiquitinylation by an E3 ligase HUWE1. Interestingly, the mechanistic axis of the DUSP4 degradation and p38 activation is also associated with a transcriptional signature of immune activation in Mk cells. In the context of thrombocytopenia observed in myelodysplastic syndrome (MDS), we demonstrate that high levels of p38 MAPK and PRMT1 are associated with low platelet counts and adverse prognosis, while pharmacological inhibition of p38 MAPK or PRMT1 stimulates megakaryopoiesis. These findings provide mechanistic insights into the role of the PRMT1-DUSP4-p38 axis on Mk differentiation and present a strategy for treatment of thrombocytopenia associated with MDS.


Asunto(s)
Diferenciación Celular , Fosfatasas de Especificidad Dual , Megacariocitos , Fosfatasas de la Proteína Quinasa Activada por Mitógenos , Adulto , Animales , Niño , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven , Arginina/metabolismo , Línea Celular , Fosfatasas de Especificidad Dual/metabolismo , Estabilidad de Enzimas , Células HEK293 , Sistema de Señalización de MAP Quinasas , Megacariocitos/citología , Megacariocitos/enzimología , Metilación , Ratones Endogámicos C57BL , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Síndromes Mielodisplásicos/enzimología , Síndromes Mielodisplásicos/patología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Poliubiquitina/metabolismo , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteolisis , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Ubiquitinación
16.
Mol Med Rep ; 24(2)2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34080021

RESUMEN

Colorectal cancer (CRC) ranks third in incidence and second in mortality among all types of cancer, and due to its insidious onset and lack of early symptoms, it is usually diagnosed at a later stage. Saponins, a class of compounds abundant in plants, have been reported to possess prominent anti­tumour properties. The use of ginsenoside Rg3 in the clinical setting was authorized by the National Medicinal Products Administration of China. In the present study, total saponins from Rhizoma Panacis Majoris (RPMTG) were prepared, and the pharmacological mechanisms underlying the anti­CRC effects of RPMTG were investigated. The effect of RPMTG on the proliferation, cell cycle progression and apoptosis of HCT116 and SW620 cells were detected by MTT, flow cytometry and western blotting assays, and it was demonstrated that RPMTG could inhibit the proliferation of HCT116 and SW620 cells with IC50 values of 315.8 and 355.1 µg/ml, respectively, induce cell cycle arrest in the S and G0/G1 phase, and trigger apoptosis by downregulating the expression of the anti­apoptotic proteins Bcl­2, Bcl­xL and induced myeloid leukaemia cell differentiation protein Mcl­1, and increasing the expression of the pro­apoptotic proteins Bax and Bad, cleaved caspased­3 and poly(ADP)­ribose polymerase. These findings suggested that RPMTG induced apoptosis through mitochondrial­related pathways. In addition, RPMTG also decreased the expression of phosphorylated (p)­extracellular signal­regulated kinase and increased p­c­Jun N­terminal kinase (p­JNK) and p­p38. Moreover, the effects of RPMTG on cell proliferation and apoptosis were partially reversed when the JNK and p38 mitogen­activated protein kinase (MAPK) pathways were inhibited, indicating that RPMTG triggered apoptosis mainly via regulating JNK and p38 MAPK signalling. Therefore, RPMTG may have potential as an anti­CRC agent, and further evaluations are needed.


Asunto(s)
Neoplasias Colorrectales/patología , Medicamentos Herbarios Chinos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Panax/química , Rizoma/química , Saponinas/farmacología , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Medicamentos Herbarios Chinos/aislamiento & purificación , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Mitocondriales/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Saponinas/aislamiento & purificación , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
17.
Int Immunopharmacol ; 98: 107848, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34126342

RESUMEN

The efficacy of adoptive cell therapy (ACT) relies on the abilities of T cells in self-expansion, survival and the secretion of effector molecules. Here, we presented an optimized method to generate T cells with improved functions by supplementing the culture medium with p38 inhibitor and the combination of IL-7 and IL-15 or IL-2 alone. The addition of p38 inhibitor, Doramapimod or SB202190, to IL-7 and IL-15 culture largely increased the capacity of T cells in the proliferation with enrichment of the naïve-like subsets and expression of CD62L. Importantly, we found this regimen has generated complete T cell resistance to TGF-ß-induced functional suppression, with sustained levels of the IFN-γ and Granzyme-B productions. Such findings were also validated in the melanoma-associated antigen recognized by T cells (MART-1) specific T cell receptor (TCR) engineered T cells, which were expanded in Doramapimod and IL-7 + IL-15 added media. In conclusion, we have established and optimized a protocol with the combination of p38 inhibitor, IL-7 and IL-15, rather than IL-2, for the generation of functionally enhanced T cells applicable for ACT.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Naftalenos/farmacología , Compuestos de Fenilurea/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Línea Celular , Ingeniería Genética , Granzimas/metabolismo , Humanos , Tolerancia Inmunológica , Interferón gamma/metabolismo , Activación de Linfocitos , Antígeno MART-1/inmunología , Linfocitos T/trasplante , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
18.
Inflammopharmacology ; 29(4): 1139-1155, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34115226

RESUMEN

Catechin, a flavonol belonging to the flavonoid group of polyphenols is present in many plant foods. The present study was done to evaluate the effect of catechin on various inflammatory mediators using lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. The effect of catechin on total cyclooxygenase (COX) activity, 5-lipoxygenase (5-LOX), myeloperoxidase, nitrite and inducible nitric oxide synthase (iNOS) level, secretion of tumor necrosis factor-α (TNF-α) and interleukin-10 (IL-10) were assessed in LPS-stimulated RAW 264.7 cells. The expression of COX-2, iNOS, TNF-α, nuclear factor-ĸB (NF-κB) and p38 mitogen-activated protein kinase (MAPK) genes were also investigated. The effect was further analyzed using human PBMCs by assessing the level of TNF-α and IL-10. The study demonstrated that the inflammatory mediators such as COX, 5-LOX, nitrite, iNOS, and TNF-α were significantly inhibited by catechin in a concentration-dependent manner whereas IL-10 production was up-regulated in RAW 264.7 cells. Moreover, catechin down-regulated the mRNA level expression of COX-2, iNOS, TNF-α, NF-κB and p38 MAPK. The current study ratifies the beneficial effect of catechin as a dietary component in plant foods to provide protection against inflammatory diseases.


Asunto(s)
Antiinflamatorios/farmacología , Catequina/farmacología , Mediadores de Inflamación/antagonistas & inhibidores , Lipopolisacáridos/toxicidad , FN-kappa B/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Humanos , Mediadores de Inflamación/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Ratones , FN-kappa B/metabolismo , Células RAW 264.7 , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Eur J Pharmacol ; 904: 174155, 2021 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-33971178

RESUMEN

The methylglyoxal elicits diverse adverse effects on the body. Uridine diphosphate, an extracellular nucleotide, plays an important role as a signaling molecule controlling vascular tone. This study aimed to evaluate the relationship between methylglyoxal and uridine diphosphate-induced carotid arterial contraction in rats. Additionally, we examined whether p38 mitogen-activated protein kinase (MAPK) would involve such responses. Organ baths were conducted to determine vascular reactivity in isolated carotid arterial rings, and western blotting was used for protein analysis. Treatment with methylglyoxal to carotid arterial rings showed concentration-dependent augmentation to uridine diphosphate-induced contraction in the absence and presence of NG-nitro-L-arginine, which is a nitric oxide synthase inhibitor, whereas, methylglyoxal did not affect serotonin- or isotonic high K+-induced contraction in the presence of a nitric oxide synthase inhibitor. Under nitric oxide synthase inhibition, SB203580, which is a selective p38 MAPK inhibitor, suppressed uridine diphosphate-induced contraction in both the control and methylglyoxal-treated groups, and the difference in uridine diphosphate-induced contraction was abolished by SB203580 treatment. The levels of phosphorylated p38 MAPK were increased by methylglyoxal in carotid arteries, not only under the basal condition but also under uridine diphosphate stimulation. The suppression of uridine diphosphate-induced contraction by a highly selective cell-permeable protein kinase C inhibitor bisindolylmaleimide I was observed in the methylglyoxal-treated group but not in the controls. Moreover, methylglyoxal-induced augmentation of uridine diphosphate-induced contraction was prevented by N-acetyl-L-cysteine. These results suggest that methylglyoxal could enhance uridine diphosphate-induced contraction in rat carotid arteries and may be caused by activation of p38 MAPK and protein kinase C and increased oxidative stress.


Asunto(s)
Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/metabolismo , Contracción Muscular/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Piruvaldehído/farmacología , Uridina Difosfato/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Acetilcisteína/farmacología , Animales , Depuradores de Radicales Libres/farmacología , Imidazoles/farmacología , Masculino , Músculo Esquelético/irrigación sanguínea , Estrés Oxidativo/efectos de los fármacos , Potasio/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Ratas Wistar , Serotonina/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
20.
Sci Rep ; 11(1): 9832, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33972601

RESUMEN

We recently found that tumor necrosis factor-α (TNF-α) may be involved in neuronal cell death induced by methylmercury in the mouse brain. Here, we examined the cells involved in the induction of TNF-α expression by methylmercury in the mouse brain by in situ hybridization. TNF-α-expressing cells were found throughout the brain and were identified as microglia by immunostaining for ionized calcium binding adaptor molecule 1 (Iba1). Methylmercury induced TNF-α expression in mouse primary microglia and mouse microglial cell line BV2. Knockdown of apoptosis signal-regulating kinase 1 (ASK1), an inflammatory cytokine up-regulator that is responsible for reactive oxygen species (ROS), decreased methylmercury-induced TNF-α expression through decreased phosphorylation of p38 MAP kinase in BV2 cells. Suppression of methylmercury-induced reactive oxygen species (ROS) by antioxidant treatment largely abolished the induction of TNF-α expression and phosphorylation of p38 by methylmercury in BV2 cells. Finally, in mouse brain slices, the TNF-α antagonist (WP9QY) inhibited neuronal cell death induced by methylmercury, as did the p38 inhibitor SB203580 and liposomal clodronate (a microglia-depleting agent). These results indicate that methylmercury induces mitochondrial ROS that are involved in activation of the ASK1/p38 pathway in microglia and that this is associated with induction of TNF-α expression and neuronal cell death.


Asunto(s)
Encéfalo/patología , Intoxicación del Sistema Nervioso por Mercurio/patología , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Encéfalo/citología , Línea Celular , Ácido Clodrónico/farmacología , Modelos Animales de Enfermedad , Contaminantes Ambientales/administración & dosificación , Contaminantes Ambientales/toxicidad , Técnicas de Silenciamiento del Gen , Humanos , Imidazoles/farmacología , MAP Quinasa Quinasa Quinasa 5/genética , MAP Quinasa Quinasa Quinasa 5/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Intoxicación del Sistema Nervioso por Mercurio/etiología , Compuestos de Metilmercurio/administración & dosificación , Compuestos de Metilmercurio/toxicidad , Ratones , Microglía/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neuronas/patología , Péptidos Cíclicos/farmacología , Fosforilación/efectos de los fármacos , Cultivo Primario de Células , Piridinas/farmacología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA