Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
J Endocrinol Invest ; 47(1): 149-166, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37477865

RESUMO

PURPOSE: To explore the key genes and molecular pathways in the progression of thyroid papillary carcinoma (PTC) promoted by testosterone using RNA-sequencing technology, and to provide new drug targets for improving the therapeutic effect of PTC. METHODS: Orchiectomy (ORX) was carried out to construct ORX mouse models. TPC-1 cells were subcutaneously injected for PTC formation in mice, and the tumor tissues were collected for RNA-seq. The key genes were screened by bioinformatics technology. Tnnt1 expression in PTC cells was knocked down or overexpressed by transfection. Cell counting kit-8 (CCK-8), colony formation assay, scratch assay and transwell assay were adopted, respectively, for the detection of cell proliferation, colony formation, migration and invasion. Besides, quantification real-time polymerase chain reaction (qRT-PCR) and western blot were utilized to determine the mRNA and protein expression levels of genes in tissues or cells. RESULTS: Both estradiol and testosterone promoted the growth of PTC xenografts. The key gene Tnnt1 was screened and obtained by bioinformatics technology. Functional analysis revealed that overexpression of Tnnt1 could markedly promote the proliferation, colony formation, migration, invasion, and epithelial-to-mesenchymal transition (EMT) process of PTC cells, as well as could activate p38/JNK pathway. In addition, si-Tnt1 was able to inhibit the cancer-promoting effect of testosterone. CONCLUSION: Based on the outcomes of bioinformatics and basic experiments, it is found that testosterone can promote malignant behaviors such as growth, migration, invasion and EMT process of PTC by up-regulating Tnnt1 expression. In addition, the function of testosterone may be achieved by activating p38/JNK signaling pathway.


Assuntos
MicroRNAs , Neoplasias da Glândula Tireoide , Humanos , Animais , Camundongos , Câncer Papilífero da Tireoide/patologia , Neoplasias da Glândula Tireoide/patologia , Testosterona/farmacologia , Proliferação de Células/genética , Linhagem Celular Tumoral , Movimento Celular/genética , MicroRNAs/genética , Regulação Neoplásica da Expressão Gênica
2.
Ecotoxicol Environ Saf ; 231: 113176, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35026588

RESUMO

Lead (Pb), as a toxic heavy metal pollutant, has been paid much attention. Pb is often discharged into the environment through the soot, wastewater and waste residue in industrial production, which poses a great threat to animal health. Selenium (Se) is a trace element known to antagonize the toxicity caused by heavy metals. However, the interaction between Se and Pb in chicken kidney and its specific biological mechanism are still unclear. So, we constructed chicken models of Pb exposure and Pb, Se co-exposure. Therefore, we used western blot and qRT-PCR to detect the expression of related genes. The results showed that Pb activated the MAPK signaling pathway by up-regulating the expression of MARK pathway genes to induce the expression of pro-apoptotic genes and necroptosis-related genes. Se can regulate the MARK signaling pathway and attenuated the expression of MAPK pathway genes altered by Pb to reduce apoptosis and necroptosis of chicken kidney cells. Our study gives new ideas for the specific mechanism of Pb nephrotoxicity and provides a reference for comparative medicine and clinical medication.


Assuntos
Selênio , Animais , Apoptose , Galinhas , Rim/metabolismo , Chumbo/metabolismo , Chumbo/toxicidade , Sistema de Sinalização das MAP Quinases , Necroptose , Selênio/metabolismo
3.
Ecotoxicol Environ Saf ; 237: 113558, 2022 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-35483150

RESUMO

Bisphenol A (BPA) is an endocrine disruptor, that can cause immune dysfunction. Cineole (CIN) has that effect of regulating immune function and resist oxidation. Neutrophil extracellular traps (NETs) are one of the ways to resist pathogen invasion. In order to explore the effects of BPA and CIN on the release of chicken NETs and the antagonistic effect of CIN, take chicken peripheral blood neutrophils as object of study, grouping as NC, CIN, BPA + CIN and BPA. SEM, flow cytometry, RT-PCR, Western-blot and other methods were used to detect related indicators. The results showed that BPA inhibited the activities of GPX, SOD and CAT, increased the contents of MDA and NO, increased the activity of iNOS. BPA exposure inhibited the expression of myeloperoxidase (MPO), neutrophil elastase (NE) and histone, and inhibited the release of NETs. BPA activated downstream apoptosis and necroptosis through the p38 mitogen-activated protein kinase (p38-MAPK) pathway, which increased the expression of cytochrome C (CytC), bcl-2 associated K protein gene (bak), cysteinyl aspartate specific proteinase 3 (caspase-3), cysteinyl aspartate specific proteinase 9 (caspase-9), receptor-interacting protein kinase 1 (RIPK1), receptor-interacting protein kinase 1 (RIPK3) and mixed lineage kinase domain-like protein (MLKL), decreased the expression of B-cell lymphoma-2 (bcl-2). However, the co-exposure of CIN and BPA partially recovered the release of NETs, alleviated BPA-induced oxidative stress, and inhibited the activation of p38-MAPK pathway, necroptosis, and mitochondrial apoptosis pathway. These results indicated that CIN modulated p38 pathway alleviated BPA-induced neutrophil necroptosis and apoptosis, and increased NETs formation.


Assuntos
Armadilhas Extracelulares , Apoptose/genética , Ácido Aspártico , Compostos Benzidrílicos , Eucaliptol/metabolismo , Armadilhas Extracelulares/metabolismo , Fenóis , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
4.
Int J Mol Sci ; 23(3)2022 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-35163786

RESUMO

Labor is a process of inflammation and hormonal changes involving both fetal and maternal compartments. MicroRNA-132-3p (miR-132-3p) has been reported to be involved in the development of inflammation-related diseases. However, little is known about its potential role in labor onset. This study aimed to explore the mechanism of miR-132-3p in amnion for labor initiation. In the mouse amnion membranes, the expression of miR-132-3p was found to increase gradually during late gestation. In human amniotic epithelial cell line (WISH), upregulation of miR-132-3p was found to increase proinflammatory cytokines and cyclooxygenase 2 (COX2) as well as prostaglandin E2 (PGE2), which was suppressed by miR-132-3p inhibitor. Dual-specificity phosphatase 9 (DUSP9) was identified as a novel target gene of miR-132-3p, which could be negatively regulated by miR-132-3p. DUSP9 was present in the mouse amnion epithelial cells, with a decrease in its abundance at 18.5 days post coitum (dpc) relative to 15.5 dpc. Silencing DUSP9 was found to facilitate the expression of proinflammatory cytokines and COX2 as well as PGE2 secretion in WISH cells, which could be attenuated by p38 inhibitor SB203580 or JNK inhibitor SP600125. Additionally, intraperitoneal injection of pregnant mice with miR-132-3p agomir not only caused preterm birth, but also promoted the abundance of COX2 as well as phosphorylated JNK and p38 levels, and decreased DUSP9 level in mouse amnion membranes. Collectively, miR-132-3p might participate in inflammation and PGE2 release via targeting DUSP9-dependent p38 and JNK signaling pathways to cause preterm birth.


Assuntos
Âmnio/imunologia , Fosfatases de Especificidade Dupla/genética , Inflamação/genética , Trabalho de Parto/genética , MicroRNAs/genética , Âmnio/citologia , Âmnio/metabolismo , Animais , Antracenos/farmacologia , Ciclo-Oxigenase 2/metabolismo , Células Epiteliais/citologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Feminino , Imidazóis/farmacologia , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Gravidez , Piridinas/farmacologia
5.
J Cell Mol Med ; 24(2): 1451-1459, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31769187

RESUMO

The beneficial function of gastrodin towards many inflammatory diseases has been identified. This study designed to see the influence of gastrodin in a cell model of chronic obstructive pulmonary disease (COPD). MRC-5 cells were treated by LPS, before which gastrodin was administrated. The effects of gastrodin were evaluated by conducting CCK-8, FITC-PI double staining, Western blot, qRT-PCR and ELISA. Besides this, the downstream effector and signalling were studied to decode how gastrodin exerted its function. And dual-luciferase assay was used to detect the targeting link between miR-103 and lipoprotein receptor-related protein 1 (LRP1). LPS induced apoptosis and the release of MCP-1, IL-6 and TNF-α in MRC-5 cells. Pre-treating MRC-5 cells with gastrodin attenuated LPS-induced cell damage. Meanwhile, p38/JNK and NF-κB pathways induced by LPS were repressed by gastrodin. miR-103 expression was elevated by gastrodin. Further, the protective functions of gastrodin were attenuated by miR-103 silencing. And LRP1 was a target of miR-103 and negatively regulated by miR-103. The in vitro data illustrated the protective function of gastrodin in LPS-injured MRC-5 cells. Gastrodin exerted its function possibly by up-regulating miR-103 and modulating p38/JNK and NF-κB pathways.

6.
Dev Neurosci ; 42(2-4): 83-93, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33130681

RESUMO

OBJECTIVE: Recent reports have corroborated that micro-RNAs (miRs) are related to the pathological changes of cerebral ischemia-reperfusion (CIR) induced injury. This work aimed to unearth the role and potential mechanism of miR-325-3p in regulating neuronal survival in CIR injury. METHODS: To conduct this investigation, we established an in vitro model of CIR injury by subjecting neurons to oxygen-glucose deprivation and reoxygenation (OGD/R). Gain and loss of function of miR-325-3p and receptor-interacting serine-threonine kinase 3 (RIP3) in neurons were performed to observe its effect on cell apoptosis and the release of lactate dehydrogenase. The levels of miR-325-3p and RIP3 in neurons were detected by qRT-PCR. Western blot was employed to inspect the levels of caspase3, Bax, and Bcl-2, as well as p38 and JNK phosphorylation. The relationship between miR-325-3p and RIP3 was detected by TargetScan and validated by dual-luciferase reporter assay. RESULTS: Firstly, miR-325-3p expression was obviously downregulated while RIP3 expression was upregulated in neurons following OGD/R treatment. Overexpressed miR-325-3p or downexpressed RIP3 ameliorated OGD/R-induced neuronal injury. Besides, RIP3 was a direct target mRNA of miR-325-3p. Additionally, Western blot revealed the mitogen-activated protein kinase (MAPK) pathway was involved in the regulation of miR-325-3p on OGD/R-induced neuronal injury. Furthermore, miR-325-3p was verified to hinder OGD/R-induced neuronal injury through downregulating RIP3. CONCLUSION: This study demonstrated that miR-325-3p targets RIP3 to inactivate the MAPK pathway, thereby protecting neurons against OGD/R-induced injury.


Assuntos
Isquemia Encefálica/metabolismo , MicroRNAs/metabolismo , Neurônios/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Isquemia Encefálica/patologia , Células Cultivadas , Glucose/deficiência , Neurônios/patologia , Oxigênio/metabolismo , Ratos , Ratos Sprague-Dawley
7.
Mar Drugs ; 17(5)2019 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-31086026

RESUMO

Flaccidoxide-13-acetate, an active compound isolated from cultured-type soft coral Sinularia gibberosa, has been shown to have inhibitory effects against invasion and cell migration of RT4 and T24 human bladder cancer cells. In our study, we used an 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT), colony formation assay, and flow cytometry to determine the mechanisms of the anti-tumor effect of flaccidoxide-13-acetate. The MTT and colony formation assays showed that the cytotoxic effect of flaccidoxide-13-acetate on T24 and RT4 cells was dose-dependent, and the number of colonies formed in the culture was reduced with increasing flaccidoxide-13-acetate concentration. Flow cytometry analysis revealed that flaccidoxide-13-acetate induced late apoptotic events in both cell lines. Additionally, we found that flaccidoxide-13-acetate treatment upregulated the expressions of cleaved caspase 3, cleaved caspase 9, Bax, and Bad, and down-regulated the expressions of Bcl-2, p-Bad, Bcl-x1, and Mcl-1. The results indicated that apoptotic events were mediated by mitochondrial dysfunction via the caspase-dependent pathway. Flaccidoxide-13-acetate also provoked endoplasmic reticulum (ER) stress and led to activation of the PERK-eIF2α-ATF6-CHOP pathway. Moreover, we examined the PI3K/AKT signal pathway, and found that the expressions of phosphorylated PI3K (p-PI3K) and AKT (p-AKT) were decreased with flaccidoxide-13-acetate concentrations. On the other hand, our results showed that the phosphorylated JNK and p38 were obviously activated. The results support the idea that flaccidoxide-13-acetate-induced apoptosis is mediated by mitochondrial dysfunction, ER stress, and activation of both the p38 and JNK pathways, and also relies on inhibition of PI3K/AKT signaling. These findings imply that flaccidoxide-13-acetate has potential in the development of chemotherapeutic agents for human bladder cancer.


Assuntos
Apoptose/efeitos dos fármacos , Diterpenos/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Neoplasias da Bexiga Urinária/tratamento farmacológico , Animais , Caspase 3/metabolismo , Caspase 9/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/uso terapêutico , Coelhos , Fator de Transcrição CHOP/metabolismo , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia
8.
Cell Physiol Biochem ; 46(5): 1779-1792, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29705796

RESUMO

BACKGROUND/AIMS: The intestinal mucosa forms a physical and metabolic barrier against the diffusion of pathogens, toxins, and allergens from the lumen into the circulatory system. Early weaning, a critical phase in swine production, can compromise intestinal barrier function through mucosal damage and alteration of tight junction integrity Maintenance of intestinal barrier function plays a pivotal role in optimum gastrointestinal health. In this study, we investigated the effects of Clostridium tyrobutyricum (C.t) on intestinal barrier dysfunction induced by lipopolysaccharide (LPS) and the underlying mechanisms involved in intestinal barrier protection. METHODS: A Transwell model of IPEC-J2 cells was used to imitate the intestinal barrier. Fluorescence microscopy and flow cytometry were used to evaluate apoptosis. Real-time PCR was used to detect apoptosis-related genes and the downstream genes of the p38/c-Jun N-terminal kinase (JNK) signaling pathways. Western blotting was used to measure the expressions of tight junction proteins and mitogen-activated protein kinases. RESULTS: C.t efficiently maintained trans-epithelium electrical resistance values and intestinal permeability after LPS-induced intestinal barrier disruption. The expressions of tight junction proteins (ZO-1, claudin-1, and occludin) were promoted when IPEC-J2 cells were treated with C.t. Fluorescence imaging and flow cytometry revealed that C.t qualitatively and quantitatively inhibited LPS-induced cell apoptosis. C.t also increased the relative expression of the anti-apoptotic gene Bcl-2 and decreased that of the apoptotic genes Bax and caspase-3/-8. Moreover, the protective effect of C.t on damaged intestinal cell models was associated with suppression of p38 and JNK phosphorylation, negative regulation of the relative expressions of downstream genes including AP-1, ATF-2, ELK-1, and p53, and activation of Stat3 expression. CONCLUSIONS: These findings indicate that C.t may promote intestinal integrity, suggesting a novel probiotic effect on intestinal barrier function.


Assuntos
Apoptose , Clostridium tyrobutyricum/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/microbiologia , Lipopolissacarídeos/metabolismo , Sistema de Sinalização das MAP Quinases , Probióticos/metabolismo , Animais , Linhagem Celular , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Permeabilidade , Suínos
9.
Biochem Biophys Res Commun ; 505(3): 858-864, 2018 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-30301525

RESUMO

Chronic myeloid leukemia (CML) responds well to BCR-ABL tyrosine kinase inhibitors (TKI), such as imatinib and dasatinib. However, these inhibitors have been less effective as single agents in the blast phase-CML. In this work, we show that anisomycin, a clinically available drug, targets CML cells at all stages of development and enhances BCR-ABL TKIs' efficacy. Anisomycin at nanomolar concentration inhibits proliferation and induces apoptosis in a panel of CML cell lines in a dose-dependent manner. It induces apoptosis CD34 stem/progenitor cells isolated from patients with blast phase CML. Using colony formation and serial replating assays, we further show that anisomycin inhibits CML CD34 cell differentiation, proliferation and self-renewal. Additionally, anisomycin is less effective in normal bone marrow (NBM) CD34 cells, suggesting the selective anti-leukemia activity of anisomycin. Combination of anisomycin with imatinib or dasatinib achieves significantly better efficacy than TKI alone in leukemia cell lines and patient samples while sparing normal counterparts. Mechanistically, we demonstrate that p38 MAPK/JNK activation is not required for anti-leukemia activities of anisomycin. Instead, anisomycin displays preferential inhibitory effects to Wnt/ß-catenin-mediated signaling in CML. Our work provides the preclinical evidence on the potent efficacy of anisomycin in leukemia and its mechanisms of action. Our work suggests that anisomycin is a potential drug to overcome resistance to BCR-ABL TKI treatment in blast phase CML.


Assuntos
Anisomicina/farmacologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Antibacterianos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Crise Blástica/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dasatinibe , Resistencia a Medicamentos Antineoplásicos , Humanos , Mesilato de Imatinib , Células K562 , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Células-Tronco Neoplásicas , Inibidores de Proteínas Quinases , Via de Sinalização Wnt/efeitos dos fármacos
10.
Biochem Biophys Res Commun ; 502(3): 409-414, 2018 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-29852169

RESUMO

Hepatic ischemia and reperfusion (I/R) injury is a major cause of liver damage during liver transplantation, resection surgery, shock, and trauma. It has been reported that TXNIP expression was upregulated in a rat model of hepatic I/R injury. However, the role of TXNIP in the hepatic I/R injury is little known. In our study, we investigated the biological role of TXNIP and its potential molecular mechanism in the human hepatic cell line (HL7702 cells). Using oxygen-glucose deprivation and reoxygenation (OGD/R) to create a cell model of hepatic I/R injury, we found that the mRNA and protein expression levels of TXNIP were upregulated in HL7702 cells exposed to OGD/R. TXNIP overexpression remarkably promoted OGD/R-induced cell apoptosis and lactate dehydrogenase (LDH) release, both of which were significantly decreased by TXNIP knockdown. The production of malondialdehyde (MDA) was also increased by TXNIP overexpression, but was reduced by TXNIP knockdown. Moreover, TXNIP overexpression significantly upregulated the phosphorylation of p38 and JNK, which was remarkably inhibited by TXNIP knockdown. Additionally, p38-specific inhibitor SB203580 abrogated the effect of TXNIP overexpression on OGD/R-induced cell injury. Taken together, these results indicated that TXNIP knockdown alleviated hepatocyte I/R injury through preventing p38/JNK pathway activation. Thus, TXNIP might offer a novel potential therapeutic target for the treatment of hepatic I/R injury.


Assuntos
Proteínas de Transporte/metabolismo , Hepatócitos/metabolismo , Sistema de Sinalização das MAP Quinases , Traumatismo por Reperfusão/metabolismo , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Linhagem Celular , Técnicas de Silenciamento de Genes , Hepatócitos/efeitos dos fármacos , Humanos , Imidazóis/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Malondialdeído/metabolismo , Modelos Biológicos , Piridinas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Traumatismo por Reperfusão/terapia , Regulação para Cima
11.
Biochem Biophys Res Commun ; 503(2): 467-473, 2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-29680658

RESUMO

Unc-51 like autophagy activating kinase 1 (Ulk1) is a serine/threonine kinase that plays a key role in regulating autophagy processes. We attempted to investigate the effects of Ulk1 on traumatic brain injury (TBI) progression by using wild type (WT) mice and Ulk1-knockout (KO) mice suffered with or not TBI. The results were verified using LPS-treated primary astrocyte (AST). Here, Ulk1 was over-expressed in hippocampus of WT mice after TBI, as well as in lipopolysaccharide (LPS)-stimulated AST. Ulk1-deletion improved cognitive ability and hippocampus histological changes in TBI mice. Nissl and neuronal nuclei (NeuN) staining indicated that Ulk1-deletion increased the number of surviving neurons in hippocampus of TBI mice. Ulk1-ablation alleviated neuroinflammation, as evidenced by the reduced expression of hippocampus pro-inflammatory cytokines in TBI mice. TBI-induced apoptosis was also ameliorated by Ulk1-ablation, as proved by the reduced number of TUNEL-staining cells, and cleaved Caspase-3 and poly (ADP-ribose) polymerase (PARP) expressions. Moreover, Ulk1-knockout suppressed TBI-stimulated activation of astrocytes and microglia cells. Additionally, hippocampus autophagy induced by TBI was attenuated by Ulk1-knockout. Further, TBI-activated p38/c-Jun N-terminal Kinase (JNK) pathway was repressed by Ulk1-deletion in hippocampus of mice. The findings above were confirmed in LPS-stimulated AST with or without Ulk1 siRNA transfection. Intriguingly, pre-treatment of p38 or JNK activator markedly abolished the anti-inflammation, anti-apoptosis and anti-autophagy effects of Ulk1-knockdown on LPS-incubated AST. In conclusion, our results demonstrated that Ulk1 might be a potential target for developing therapeutic strategy against TBI in future.


Assuntos
Proteína Homóloga à Proteína-1 Relacionada à Autofagia/genética , Lesões Encefálicas Traumáticas/genética , Lesões Encefálicas Traumáticas/patologia , Deleção de Genes , Sistema de Sinalização das MAP Quinases , Animais , Apoptose , Astrócitos/metabolismo , Astrócitos/patologia , Autofagia , Lesões Encefálicas Traumáticas/metabolismo , Células Cultivadas , Hipocampo/metabolismo , Hipocampo/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
12.
J Neuroinflammation ; 15(1): 119, 2018 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-29679979

RESUMO

BACKGROUND: Melatonin is a well-known potent endogenous antioxidant pharmacological agent with significant neuroprotective actions. Here in the current study, we explored the nuclear factor erythroid 2-related factor 2 (Nrf2) gene-dependent antioxidant mechanism underlying the neuroprotective effects of the acute melatonin against acute ethanol-induced elevated reactive oxygen species (ROS)-mediated neuroinflammation and neurodegeneration in the developing rodent brain. METHODS: In vivo rat pups were co-treated with a single dose of acute ethanol (5 g/kg, subcutaneous (S.C.)) and a single dose of acute melatonin (20 mg/kg, intraperitoneal (I.P.)). Four hours after a single S.C. and I.P. injections, all of the rat pups were sacrificed for further biochemical (Western blotting, ROS- assay, LPO-assay, and immunohistochemical) analyses. In order to corroborate the in vivo results, we used the in vitro murine-hippocampal HT22 and microglial BV2 cells, which were subjected to knockdown with small interfering RNA (siRNA) of Nrf2 genes and exposed with melatonin (100 µM) and ethanol (100 mM) and proceed for further biochemical analyses. RESULTS: Our biochemical, immunohistochemical, and immunofluorescence results demonstrate that acute melatonin significantly upregulated the master endogenous antioxidant Nrf2 and heme oxygenase-1, consequently reversing the acute ethanol-induced elevated ROS and oxidative stress in the developing rodent brain, and in the murine-hippocampal HT22 and microglial BV2 cells. In addition, acute melatonin subsequently reduced the activated MAPK-p-P38-JNK pathways and attenuated neuroinflammation by decreasing the expression of activated gliosis and downregulated the p-NF-K-B/p-IKKß pathway and decreased the expression levels of other inflammatory markers in the developing rodent brain and BV2 cells. Of note, melatonin acted through the Nrf2-dependent mechanism to attenuate neuronal apoptosis in the postnatal rodent brain and HT22 cells. Immunohistofluorescence results also showed that melatonin prevented ethanol-induced neurodegeneration in the developing rodent brain. The in vitro results indicated that melatonin induced neuroprotection via Nrf2-dependent manner and reduced ethanol-induced neurotoxicity. CONCLUSIONS: The pleiotropic and potent neuroprotective antioxidant characteristics of melatonin, together with our in vivo and in vitro findings, suppose that acute melatonin could be beneficial to prevent and combat the acute ethanol-induced neurotoxic effects, such as elevated ROS, neuroinflammation, and neurodegeneration in the developing rodent brain.


Assuntos
Antioxidantes , Melatonina , Fator 2 Relacionado a NF-E2 , Síndromes Neurotóxicas , Animais , Feminino , Masculino , Animais Recém-Nascidos , Antioxidantes/uso terapêutico , Proteínas de Ligação ao Cálcio/metabolismo , Linhagem Celular Transformada , Depressores do Sistema Nervoso Central/toxicidade , Deficiências do Desenvolvimento/tratamento farmacológico , Deficiências do Desenvolvimento/etiologia , Modelos Animais de Doenças , Etanol/toxicidade , Heme Oxigenase-1/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Melatonina/uso terapêutico , Proteínas dos Microfilamentos/metabolismo , Síndromes Neurotóxicas/complicações , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/prevenção & controle , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos
13.
Biochem Biophys Res Commun ; 487(2): 201-208, 2017 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-28392397

RESUMO

Diabetic nephropathy (DN) is one of the most common microvascular complication of diabetes mellitus (DM) as well as the main reason resulting in chronic renal failure. Transmembrane protein 16A (TMEM16A) plays an important role in multiple physiological actions. Here we found that it was up-regulated in high-fat diet (HFD)/streptozotocin (STZ)-induced diabetic mice. Moreover, reverse transcription-polymerase chain reaction (RT-PCR) amplification, Western blot detection, Periodic Acid Schiff (PAS) staining and immunohistochemical analysis confirmed that TMEM16A deficiency alleviated renal injury in diabetic mice and TMEM16A knockout diabetic mice were protected from the HFD-induced reduction in Nephrin expression. To understand further the molecular mechanism of its function, podocytes treated with high glucose (HG, 30 mmol/L glucose) in vitro was chosen as a model to study its signal transduction pathway. Nephrin expression level in siRNA-TMEM16A group was significantly higher than that of the HG group (also called Model group). Flow cytometric analysis revealed that podocyte apoptosis in siRNA-TMEM16A group was significantly lower than that of the Model group. RT-PCR and Western blot exhibited that apoptosis-related genes including apoptosis-inducing factor (AIF) and cystinylaspartate specific protease-3/-9 (caspase-3/-9) were dramatically down regulated in siRNA-TMEM16A group, compared with Model group. Phosphorylation levels of P38 and JNK in siRNA-TMEM16A group were lower than that of the Model group. Thus, TMEM16A is one of the critical components of a signal transduction pathway that links renal injury to podocyte apoptosis in DN.


Assuntos
Apoptose , Canais de Cloreto/metabolismo , Nefropatias Diabéticas/metabolismo , Rim/metabolismo , Sistema de Sinalização das MAP Quinases , Podócitos/metabolismo , Animais , Anoctamina-1 , Nefropatias Diabéticas/patologia , Progressão da Doença , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Podócitos/patologia
14.
Zhongguo Zhong Yao Za Zhi ; 42(4): 731-738, 2017 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-28959845

RESUMO

To investigate the effect and mechanism of oxymatrine(OMT) on hippocampal neurons apoptosis. Effect of OMT on survival of hippocampal neurons was measured by MTT.Effect of OMT on LPS-induced lactate dehydrogenase(LDH) release rate in hippocampal neurons was measured by biochemical methods. Hoechst 33342 staining was used to observe the apoptotic morphology of hippocampal neurons.The mRNA expression levels of Bax, Bcl-2, and Caspase-3 were detected by Real-time quantitative PCR(RT-qPCR), and the protein expression levels of p38, p-p38, JNK, p-JNK, Bax, Bcl-2 and Caspase-3 were detected by Western blot.The results showed that, hippocampal neurons all grew well after treatment by different doses (0.37-6.0 g•L⁻¹) of OMT for 24 h. Stimulation from LPS increased the release of LDH(P<0.01), improved the JNK and p38 phosphorylation levels(P<0.01), increased the proportion of Bax/Bcl-2 and the expression of Caspase-3(P<0.01), and promoted the apoptosis of hippocampal neurons. OMT pretreatment could significantly reduce the release of LDH induced by LPS stimulation(P<0.05 or P<0.01), reduce the p38 and JNK phosphorylation, decrease the expression of Caspase-3 and Bax/Bcl-2(P<0.01), and diminish the apoptosis of hippocampal neurons.In conclusion, OMT could reduce the LPS-induced phosphorylation of p38 and JNK, down-regulate the Bax/Bcl-2 ratio and expression of Caspase-3, thus inhibiting apoptosis of hippocampal neurons. The mechanism may be associated with p38/JNK signaling pathway.


Assuntos
Alcaloides/farmacologia , Apoptose , Sistema de Sinalização das MAP Quinases , Neurônios/efeitos dos fármacos , Quinolizinas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Caspase 3/metabolismo , Hipocampo/citologia , Neurônios/citologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo
15.
Toxicol Appl Pharmacol ; 303: 79-89, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27174766

RESUMO

Arsenic is a widely distributed toxic metalloid all over the world. Inorganic arsenic species are supposed to affect astrocytic functions and to cause neuron apoptosis in CNS. Microglias are the key cell type involved in innate immune responses in CNS, and microglia activation has been linked to inflammation and neurotoxicity. In this study, using ELISA, we showed that Arsenic trioxide up-regulated the expression and secretion of IL-1ß in a dose-dependent manner and a time-dependent manner in cultured HAPI microglia cells. The secretion of IL-1ß caused the apoptosis of SH-SY5Y. These pro-inflammatory responses were inhibited by the STAT3 blocker, AG490 and P38/JNK MAPK blockers SB202190, SP600125. Further, Arsenic trioxide exposure could induce phosphorylation and activation of STAT3, and the translocation of STAT3 from the cytosol to the nucleus in this HAPI microglia cell line. Thus, the STAT3 signaling pathway can be activated after Arsenic trioxide treatment. However, P38/JNK MAPK blockers SB202190, SP600125 also obviously attenuated STAT3 activation and transnuclear transport induced by Arsenic trioxide. In concert with these results, we highlighted that the secretion of IL-1ß and STAT3 activation induced by Arsenic trioxide can be mediated by elevation of P38/JNK MAPK in HAPI microglia cells and then induced the toxicity of neurons.


Assuntos
Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Óxidos/toxicidade , Fator de Transcrição STAT3/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Antracenos/farmacologia , Apoptose/efeitos dos fármacos , Trióxido de Arsênio , Arsenicais , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Imidazóis/farmacologia , Inflamação , Interleucina-1beta/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Microglia/metabolismo , Neurônios/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Ratos , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
16.
Naunyn Schmiedebergs Arch Pharmacol ; 397(9): 6611-6621, 2024 09.
Artigo em Inglês | MEDLINE | ID: mdl-38472369

RESUMO

Myocardial fibrosis (MF), which is an inevitable pathological manifestation of many cardiovascular diseases in the terminal stage, often contributes to severe cardiac dysfunction and sudden death. Morroniside (MOR) is the main active component of Cornus officinalis with a variety of biological activities. This study was designed to explore the efficacy of MOR in MF and to investigate its pharmacological mechanism. The viability of MOR-treated human cardiac fibroblast (HCF) cells with or without Angiotensin II (AngII) induction was assessed with Cell Counting Kit-8 (CCK-8). The migration of AngII-induced HCF cells was appraised with a transwell assay. Gelatin zymography analysis was adopted to evaluate the activities of MMP2 and MMP9, while immunofluorescence assay was applied for the estimation of Collagen I and Collagen III. By means of western blot, the expressions of migration-, fibrosis-, and p38/c-Jun N-terminal kinase (JNK) signal pathway-related proteins were resolved. The transfection efficacy of oe-Kruppel-like factor 5 (KLF5) was examined with reverse transcription-quantitative PCR (RT-qPCR) and western blot. In this study, it was found that MOR treatment inhibited AngII-induced hyperproliferation, migration, and fibrosis of HCF cells, accompanied with decreased activities of matrix metalloproteinase 2 (MMP2), matrix metalloproteinase 9 (MMP9), connective tissue growth factor (CTGF), Fibronectin, and α-SMA, which were all reversed by KLF5 overexpression. Collectively, MOR exerted protective effects on MF by blocking p38/JNK signal pathway through the downregulation of KLF5.


Assuntos
Angiotensina II , Movimento Celular , Proliferação de Células , Regulação para Baixo , Matriz Extracelular , Fibroblastos , Fatores de Transcrição Kruppel-Like , Sistema de Sinalização das MAP Quinases , Proteínas Quinases p38 Ativadas por Mitógeno , Angiotensina II/farmacologia , Humanos , Proliferação de Células/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Regulação para Baixo/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Miocárdio/patologia , Miocárdio/citologia , Miocárdio/metabolismo , Fibrose , Células Cultivadas , Transdução de Sinais/efeitos dos fármacos
17.
Discov Oncol ; 15(1): 8, 2024 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-38216781

RESUMO

Glutamate dehydrogenase 1 (GLUD1) is an important enzyme in glutamine metabolism. Previously, we found GLUD1 was down-regulated in tumor tissues of hepatocellular carcinoma (HCC) patients by proteomics study. To explore its role in the progression of HCC, the expressional level of GLUD1 was firstly examined and presented as that both the protein and mRNA levels were down-regulated in tumor tissues compared to the normal liver tissues. GLUD1 overexpression significantly inhibited HCC cells proliferation, migration, invasion and tumor growth both in vitro and in vivo, while GLUD1 knocking-down promoted HCC progression. Metabolomics study of GLUD1 overexpressing and control HCC cells showed that 129 differentially expressed metabolites were identified, which mainly included amino acids, bases, and phospholipids. Moreover, metabolites in mitochondrial oxidative phosphorylation system (OXPHOS) were differentially expressed in GLUD1 overexpressing cells. Mechanistic studies showed that GLUD1 overexpression enhanced mitochondrial respiration activity and reactive oxygen species (ROS) production. Excessive ROS lead to mitochondrial apoptosis that was characterized by increased expression levels of p53, Cytochrome C, Bax, Caspase 3 and decreased expression level of Bcl-2. Furthermore, we found that the p38/JNK MAPK pathway was activated in GLUD1 overexpressing cells. N-acetylcysteine (NAC) treatment eliminated cellular ROS and blocked p38/JNK MAPK pathway activation, as well as cell apoptosis induced by GLUD1 overexpression. Taken together, our findings suggest that GLUD1 inhibits HCC progression through regulating cellular metabolism and oxidative stress state, and provide that ROS generation and p38/JNK MAPK pathway activation as promising methods for HCC treatment.

18.
Sci Rep ; 14(1): 7747, 2024 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-38565569

RESUMO

6-Gingerol, the main bioactive compound of ginger, has antioxidant, anti-inflammatory, anti-cancer and neuroprotective effects. However, it is unclear whether 6-Gingerol has protective effects against hepatic ischemia/reperfusion (I/R) injury. In this study, the mouse liver I/R injury model and the mouse AML12 cell hypoxia/reoxygenation (H/R) model were established by pretreatment with 6-Gingerol at different concentrations to explore the potential effects of 6-Gingerol. Serum transaminase levels, liver necrotic area, cell viability, inflammatory response, and cell apoptosis were used to assess the effect of 6-Gingerol on hepatic I/R or cell H/R injury. Quantitative polymerase chain reaction (qPCR) and Western blotting were used to detect the mRNA and protein expression. The results show that 6-Gingerol decreased serum alanine aminotransferase (ALT), aspartate aminotransferase (AST) levels, liver necrosis, inflammatory cytokines IL-1ß, IL-6, MCP-1, TNF-α expression, Ly6g+ inflammatory cell infiltration, protein phosphorylation of NF-κB signaling pathway, Terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) positive cells, cell apoptosis rate, the protein expression of pro-apoptotic protein BAX and C-Caspase3, increased cell viability, and expression of anti-apoptotic protein BCL-2. Moreover, 6-Gingerol could increase the mRNA and protein expression of mitogen activated protein kinase phosphatase 5 (MKP5) and inhibit the activation of P38/JNK signaling pathway. In MKP5 knockout (KO) mice, the protective effect of 6-gingerol and the inhibition of P38/JNK pathway were significantly weakened. Therefore, our results suggest that 6-Gingerol exerts anti-inflammatory and anti-apoptotic effects to attenuate hepatic I/R injury by regulating the MKP5-mediated P38/JNK signaling pathway.


Assuntos
Catecóis , Álcoois Graxos , Sistema de Sinalização das MAP Quinases , Traumatismo por Reperfusão , Camundongos , Animais , Traumatismo por Reperfusão/tratamento farmacológico , Fígado , Isquemia , Anti-Inflamatórios/farmacologia , Proteínas Reguladoras de Apoptose/farmacologia , Apoptose , RNA Mensageiro/farmacologia
19.
Int J Nanomedicine ; 19: 1097-1108, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38327597

RESUMO

Introduction: Osteosarcoma is a prevalent and highly malignant primary bone tumor. However, current clinical therapeutic drugs for osteosarcoma are not suitable for long-term use due to significant side effects. Therefore, there is an urgent need to develop new drugs with fewer side effects. Dipsacus asperoides C. Y. Cheng et T. M. Ai, a traditional Chinese medicine, is commonly used for its anti-inflammatory, anti-pain, bone fracture healing, and anti-tumor effects. In this study, we investigated the effects of exosome-like nanoparticles derived from Dipsacus asperoides (DAELNs) on osteosarcoma cells in vitro and in vivo. Methods: DAELNs were isolated and purified from Dipsacus asperoides and their physical and chemical properties were characterized using transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). The cellular uptake of DAELNs in osteosarcoma cells was analyzed by PKH26 staining. The proliferation, invasion, migration, and apoptosis of osteosarcoma cells were assessed using CCK8 assay, EdU assay, colony-formation assay, transwell assay, wound healing assay, and mitochondrial membrane potential measurement, respectively. The regulatory mechanism of DAELNs inhibiting the progression of osteosarcoma via activating P38/JNK signaling pathway was investigated using Western blotting and immunohistochemistry. Moreover, the therapeutic effects of DAELNs were evaluated using in vivo small animal imaging assay, HE staining, and immunohistochemistry. Results: Our results showed that DAELNs inhibited the proliferation, invasion, migration, and fostered the apoptosis of osteosarcoma cells in vitro and suppressed the tumor growth of osteosarcoma cells in a xenograft nude mouse model. Furthermore, the bio-distribution of DiD-labeled DAELNs showed preferential targeting of osteosarcoma tumors and excellent biosafety in histological analysis of the liver and kidney. Mechanistically, DAELNs activated the P38/JNK signaling pathway-induced apoptosis. Conclusion: Taken together, DAELNs are novel, natural, and osteosarcoma-targeted agents that can serve as safe and effective therapeutic approaches for the treatment of osteosarcoma.


Assuntos
Neoplasias Ósseas , Dipsacaceae , Exossomos , Osteossarcoma , Humanos , Camundongos , Animais , Sistema de Sinalização das MAP Quinases , Dipsacaceae/química , Exossomos/metabolismo , Apoptose , Osteossarcoma/patologia , Linhagem Celular Tumoral , Neoplasias Ósseas/patologia , Modelos Animais de Doenças , Proliferação de Células , Movimento Celular
20.
In Vivo ; 38(3): 1094-1103, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38688591

RESUMO

BACKGROUND/AIM: The underlying processes of renal cell carcinoma (RCC), one of the deadliest malignancies of the urinary system, are still poorly understood. HECT domain E3 ubiquitin protein ligase 2 (HECTD2) is an E3 ubiquitin ligase implicated in the pulmonary inflammatory response. This study investigated the impact of HECTD2 on regulating inflammation in RCC cells and its potential mechanisms. MATERIALS AND METHODS: HECTD2 expression in RCC tissues was examined. Immunoprecipitation and western blot (WB) analysis confirmed that HECTD2 up-regulated euchromatic histone lysine methyltransferase 2 (EHMT2) protein degradation. ChIP experiments validated tumor necrosis factor α Inducing protein 1 (TNFAIP1) as a direct target of EHMT2. qRT-PCR determined HECTD2 and TNFAIP1 expression in RCC cells. Cell viability was assayed via CCK-8. ELISA was employed to measure the expression of IL-6, TNF-α, IL-8, and IL-1ß. WB analysis was conducted to test p38/JNK pathway-related protein (p38, p-p38, JNK, and p-JNK) expression. RESULTS: HECTD2 and TNFAIP1 were significantly up-regulated in RCC patient tissues and cells. Subsequent investigations revealed that HECTD2 promoted an inflammatory response in RCC cells. Additionally, HECTD2 up-regulated TNFAIP1 expression, and high TNFAIP1 expression could reverse the repressive impact of low HECTD2 expression on the inflammatory response in RCC cells. Rescue experiments demonstrated that the addition of p38/JNK pathway inhibitors attenuated the impact of TNFAIP1 overexpression on the RCC inflammatory response. CONCLUSION: Our findings establish a new mechanism by which HECTD2 exerts a pro-inflammatory role in RCC cells and present a prospective method for an anti-inflammatory intervention targeting the HECTD2/TNFAIP1 axis in malignancies.


Assuntos
Carcinoma de Células Renais , Inflamação , Neoplasias Renais , Sistema de Sinalização das MAP Quinases , Ubiquitina-Proteína Ligases , Humanos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Inflamação/metabolismo , Inflamação/genética , Inflamação/patologia , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Neoplasias Renais/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Transdução de Sinais , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa