Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
Mais filtros

País/Região como assunto
Intervalo de ano de publicação
1.
Exp Cell Res ; 440(1): 114103, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38848951

RESUMO

Elevated homocysteine (Hcy) levels have been recognized as significant risk factor for cardiovascular and cerebrovascular diseases, closely related to endothelial injury. While expression of Ciliary Neurotrophic Factor (CNTF) significantly increases during Hcy-induced vascular endothelial cell injury, the precise molecular pathways through which CNTF operates remain to be clarified. To induce vascular endothelial cell injury, human umbilical vein endothelial cells (HUVECs) were treated with Hcy. Cell viability and apoptosis in HUVECs were assessed using the CCK-8 assay and flow cytometry. Western blot analysis determined the expression levels of the JAK2-STAT3 pathway, inflammation-related factors (IL-1ß, NLRP3, ICAM-1, VCAM-1), and apoptosis-related factors (cleaved Caspase-3 and Bax). Immunofluorescence staining and western blotting were employed to examine CD31 and α-SMA expression. Knockdown of CNTF was achieved using lentiviral interference, and its effects on inflammation and cell injury were evaluated. Chromatin immunoprecipitation (ChIP) and dual luciferase reporter analysis were conducted to investigate the interaction between the MAFK and CNTF promoters. Our results indicated that Hcy induced high expression of CNTF and activated the JAK2-STAT3 signaling pathway, thereby upregulating factors associated with inflammation and cell apoptosis. Inhibiting CNTF alleviated Hcy-induced inflammation and cell injury. MAFK was identified as a transcription factor promoting CNTF transcription, and its overexpression exacerbated inflammation and cell injury in Hcy-treated HUVECs through the CNTF-JAK2-STAT3 axis, which could be reversed by knocking down CNTF. Activation of MAFK leads to CNTF upregulation, which activates the JAK2-STAT3 signaling pathway, regulating inflammation and inducing injury in Hcy-exposed vascular endothelial cells. Targeting CNTF or its upstream regulator MAFK may represent potential therapeutic strategies for mitigating endothelial dysfunction associated with hyperhomocysteinemia and cardiovascular diseases.


Assuntos
Apoptose , Fator Neurotrófico Ciliar , Homocisteína , Células Endoteliais da Veia Umbilical Humana , Inflamação , Janus Quinase 2 , Fator de Transcrição STAT3 , Transdução de Sinais , Janus Quinase 2/metabolismo , Humanos , Fator de Transcrição STAT3/metabolismo , Homocisteína/farmacologia , Homocisteína/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Fator Neurotrófico Ciliar/metabolismo , Fator Neurotrófico Ciliar/genética , Apoptose/efeitos dos fármacos , Células Cultivadas , Sobrevivência Celular/efeitos dos fármacos
2.
Drug Resist Updat ; 68: 100957, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36990047

RESUMO

Resistance to epidermal growth factor receptor (EGFR) inhibitors, from the first-generation erlotinib to the third generation osimertinib, is a clinical challenge in the treatment of patients with EGFR-mutant lung adenocarcinoma. Our previous work found that a novel allosteric inhibitor of phosphoglycerate mutase 1 (PGAM1), HKB99, restrains erlotinib resistance in lung adenocarcinoma cells. However, the role of HKB99 in osimertinib resistance and its underlying molecular mechanism remains to be elucidated. Herein, we found that IL-6/JAK2/STAT3 signaling pathway is aberrantly activated in both erlotinib and osimertinib resistant cells. Importantly, HKB99 significantly blocks the interaction of PGAM1 with JAK2 and STAT3 via the allosteric sites of PGAM1, which leads to inactivation of JAK2/STAT3 and thereby disrupts IL-6/JAK2/STAT3 signaling pathway. Consequently, HKB99 remarkably restores EGFR inhibitor sensitivity and exerts synergistic tumoricidal effect. Additionally, HKB99 alone or in combination with osimertinib down-regulated the level of p-STAT3 in xenograft tumor models. Collectively, this study identifies PGAM1 as a key regulator in IL-6/JAK2/STAT3 axis in the development of resistance to EGFR inhibitors, which could serve as a therapeutic target in lung adenocarcinoma with acquired resistance to EGFR inhibitors.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Humanos , Cloridrato de Erlotinib/farmacologia , Cloridrato de Erlotinib/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Interleucina-6/genética , Interleucina-6/farmacologia , Interleucina-6/uso terapêutico , Fosfoglicerato Mutase/metabolismo , Fosfoglicerato Mutase/farmacologia , Resistencia a Medicamentos Antineoplásicos , Adenocarcinoma de Pulmão/tratamento farmacológico , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/patologia , Receptores ErbB , Transdução de Sinais , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Mutação , Linhagem Celular Tumoral , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Janus Quinase 2/farmacologia
3.
Ecotoxicol Environ Saf ; 272: 116069, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38340601

RESUMO

Di-(2-ethylhexyl) phthalate (DEHP), a common endocrine-disrupting chemical (EDC), is widely used in daily articles, early exposure to DEHP is associated with many behavioral changes in pups. This study aimed to investigate the effects and underlying mechanisms of maternal exposure to DEHP on the impaired social interaction in pups. Pregnant rats were administered 0, 30, 300, or 750 mg/kg/d DEHP daily by oral gavage. Highly aggressive proliferating immortalized (HAPI) cells were treated with mono-(2-ethylhexyl) phthalate (MEHP) and tyrosine phosphorylation inhibitor (AG490). Our results showed that DEHP exposure induced the activation of microglias (MGs) via activating the janus kinase 2 / signal transducer and activator of transcription 3 (JAK2/STAT3) signaling pathway, and increased the level of pro-inflammatory factors, then impaired the social behavior in male pups, but not female pups. Moreover, MEHP exposure could also activate HAPI via activating this signaling pathway, and AG490 could inhibit the activation of this signaling pathway caused by MEHP. Therefore, we indicated that maternal exposure to DEHP could cause the gender-specific impaired social interaction in pups that might be related to the activation of MGs.


Assuntos
Dietilexilftalato , Dietilexilftalato/análogos & derivados , Ácidos Ftálicos , Tirfostinas , Humanos , Gravidez , Feminino , Masculino , Ratos , Animais , Dietilexilftalato/toxicidade , Dietilexilftalato/metabolismo , Exposição Materna/efeitos adversos , Microglia/metabolismo , Interação Social
4.
Curr Issues Mol Biol ; 45(11): 8687-8703, 2023 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-37998723

RESUMO

Autophagy is a vital cellular process that functions to degrade and recycle damaged organelles into basic metabolites. This allows a cell to adapt to a diverse range of challenging conditions. Autophagy assists in maintaining homeostasis, and it is tightly regulated by the cell. The disruption of autophagy has been associated with many diseases, such as neurodegenerative disorders and cancer. This review will center its discussion on providing an in-depth analysis of the current molecular understanding of autophagy and its relevance to brain tumors. We will delve into the current literature regarding the role of autophagy in glioma pathogenesis by exploring the major pathways of JAK2/STAT3 and PI3K/AKT/mTOR and summarizing the current therapeutic interventions and strategies for glioma treatment. These treatments will be evaluated on their potential for autophagy induction and the challenges associated with their utilization. By understanding the mechanism of autophagy, clinical applications for future therapeutics in treating gliomas can be better targeted.

5.
Biochem Genet ; 2023 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-37828348

RESUMO

Adenomyosis (ADS) is a common benign gynecological disease. Abnormal proliferation at the endometrial-myometrial interface (EMI) plays a crucial role in the occurrence and progression of ADS. miR-141-3p is associated with cell proliferation and apoptosis. However, the specific mechanism of miR-141-3p in the etiology of ADS is still unknown. In this study, we explored the effects of miR-141-3p on the proliferation and apoptosis of ADS EMI smooth muscle cells (SMCs). We collected EMI tissues for the primary culture of SMCs from 25 patients diagnosed with ADS and 20 without ADS. Real-time quantitative polymerase chain reaction and western blot were used to measure the mRNA and protein expression levels of miR-141-3p, JAK2, STAT3, phospho-JAK2, and phospho-STAT3 in ADS EMI SMCs. The cell counting kit 8 assay and flow cytometry analysis were used to evaluate the proliferation and apoptosis of EMI SMCs. The miR-141-3p mimic/inhibitor was used to increase or decrease the expression level of miR-141-3p. We added WP1066 to block the phosphorylation of JAK2/STAT3 pathway components. The miR-141-3p levels were decreased, while JAK2 and STAT3 levels were increased in ADS EMI SMCs. miR-141-3p overexpression significantly inhibited the proliferation and enhanced the apoptosis of EMI SMCs, whereas a decrease in miR-141-3p expression level was connected to the opposite results. Meanwhile, inactivated JAK2/STAT3 pathway decreased proliferation and enhanced apoptosis of EMI SMCs after WP1066 treatment. Furthermore, rescue experiments confirmed that the JAK2/STAT3 pathway was the downstream pathway of miR-141-3p and reduced the effect of miR-141-3p on the proliferation and apoptosis of EMI SMCs. These results demonstrate that miR-141-3p regulates the proliferation and apoptosis of ADS EMI SMCs by modulating the JAK2/STAT3 pathway.

6.
Int J Mol Sci ; 24(21)2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37958803

RESUMO

The aim of this study was to discover new biomarkers to detect breast cancer (BC), which is an aggressive cancer with a high mortality rate. In this study, bioinformatic analyses (differential analysis, weighted gene co-expression network analysis, and machine learning) were performed to identify potential candidate genes for BC to study their molecular mechanisms. Furthermore, Quantitative Real-time PCR and immunohistochemistry assays were used to examine the protein and mRNA expression levels of a particular candidate gene (DLGAP5). And the effects of DLGAP5 on cell proliferation, migration, invasion, and cell cycle were further assessed using the Cell Counting Kit-8 assay, colony formation, Transwell, wound healing, and flow cytometry assays. Moreover, the changes in the JAK2/STAT3 signaling-pathway-related proteins were detected by Western Blot. A total of 44 overlapping genes were obtained by differential analysis and weighted gene co-expression network analysis, of which 25 genes were found in the most tightly connected cluster. Finally, NEK2, CKS2, UHRF1, DLGAP5, and FAM83D were considered as potential biomarkers of BC. Moreover, DLGAP5 was highly expressed in BC. The down-regulation of DLGAP5 may inhibit the proliferation, migration, invasion, and cell cycle of BC cells, and the opposite was true for DLGAP5 overexpression. Correspondingly, silencing or overexpression of the DLGAP5 gene inhibited or activated the JAK2/STAT3 signaling pathway, respectively. DLGAP5, as a potential biomarker of BC, may impact the cell proliferation, migration, invasion, cell cycle, and BC development by modulating the JAK2/STAT3 signaling pathway.


Assuntos
Neoplasias da Mama , Quinases relacionadas a CDC2 e CDC28 , Humanos , Feminino , Linhagem Celular Tumoral , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Movimento Celular/genética , Transdução de Sinais , Ciclo Celular/genética , Proliferação de Células/genética , Biomarcadores , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Regulação Neoplásica da Expressão Gênica , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Ciclo Celular/metabolismo , Quinases relacionadas a CDC2 e CDC28/genética
7.
Zhongguo Zhong Yao Za Zhi ; 48(12): 3199-3206, 2023 Jun.
Artigo em Zh | MEDLINE | ID: mdl-37382003

RESUMO

Based on the Janus kinase 2/signal transducer and activator of transcription 3(JAK2/STAT3) signaling pathway, this study investigated the effect of medicated serum of Sparganii Rhizoma(SR) and Curcumae Rhizoma(CR) on the proliferation, apoptosis, migration, and secretion of inflammatory factors of ectopic endometrial stromal cells(ESCs). Specifically, human ESCs were primary-cultured. The effect of different concentration(5%, 10%, 20%) of SR-, CR-, and SR-CR combination-medicated serum, and AG490 solution(50 µmol·L~(-1)) on the proliferation of ESCs was detected by methyl thiazolyl tetrazolium(MTT) assay, and the optimal dose was selected accordingly for further experiment. The cells were classified into normal serum(NS) group, SR group(10%), CR group(10%), combination(CM) group(10%), and AG490 group. The apoptosis level of ESCs was detected by flow cytometry, and the migration ability was examined by wound healing assay. The secretion of interleukin(IL)-1ß, IL-6, and tumor necrosis factor(TNF)-α was determined by enzyme-linked immunosorbent assay(ELISA). The protein levels of cysteinyl aspartate specific protei-nase-3(caspase-3), B-cell lymphoma(Bcl-2), and Bcl-2-associated X protein(Bax) and the levels of phosphorylated(p)-JAK2 and p-STAT3 were detected by Western blot. The results showed that the viability of ESCs cells was lowered in the administration groups compared with the blank serum group(P<0.01), especially the 10% drug-medicated serum, which was selected for further experiment. The 10% SR-medicated serum, 10% CR-medicated serum, and 10% CM-medicated serum could increase the apoptosis rate(P<0.01), up-regulate the protein expression of caspase-3 and Bax in cells(P<0.05 or P<0.01), down-regulate the expression of Bcl-2(P<0.01), decrease the cell migration rate(P<0.05 or P<0.01), and reduce the secretion levels of IL-1ß, IL-6, and TNF-α(P<0.05 or P<0.01), and levels of p-JAK2 and p-STAT3(P<0.05 or P<0.01). Compared with the SR and CR groups, CM group showed low cell viability(P<0.01), high protein expression of caspase-3 and Bax(P<0.05 or P<0.01), and low protein expression of Bcl-2 and p-JAK2(P<0.05). After incubation with CM, the apoptosis rate was higher(P<0.05) and the migration rate was lower(P<0.01) than that of the CR group. The p-STAT3 protein level of CM group was lower than that of the RS group(P<0.05). The mechanism of SR, CR, and the combination underlying the improvement of endometriosis may be that they blocked JAK2/STAT3 signaling pathway, inhibited ESC proliferation, promoted apoptosis, weakened cell migration, and reduced the secretion of inflammatory factors. The effect of the combination was better than that of RS alone and CR alone.


Assuntos
Interleucina-6 , Janus Quinase 2 , Feminino , Humanos , Caspase 3 , Proteína X Associada a bcl-2 , Interleucina-6/genética , Apoptose , Transdução de Sinais , Proliferação de Células , Fator de Transcrição STAT3/genética
8.
Mol Pain ; 18: 17448069221135743, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-36227008

RESUMO

Bone cancer pain (BCP) is a clinically intractable mixed pain, involving inflammation and neuropathic pain, and its mechanisms remain unclear. CXC chemokine receptor 1 (CXCR1, IL-8RA) and 2 (CXCR2, IL-8RB) are high-affinity receptors for interleukin 8 (IL8). According to previous studies, CXCR2 plays a crucial role in BCP between astrocytes and neurons, while the role of CXCR1 remains unclear. The objective of this study was to investigate the role of CXCR1 in BCP. We found that CXCR1 expression increased in the spinal dorsal horn. Intrathecal injection of CXCR1 siRNA effectively attenuated mechanical allodynia and pain-related behaviors in rats. It was found that CXCR1 was predominantly co-localized with neurons. Intrathecal injection of CXCR1-siRNA reduced phosphorylated JAK2/STAT3 protein levels and the NLRP3 inflammasome (NLRP3, caspase1, and IL-1ß) levels. Furthermore, in vitro cytological experiments confirmed this conclusion. The study results suggest that the spinal chemokine receptor CXCR1 activation mediates BCP through JAK2/STAT3 signaling pathway and NLRP3 inflammasome (NLRP3, caspase1, and IL-1ß).


Assuntos
Neoplasias Ósseas , Dor do Câncer , Neuralgia , Ratos , Feminino , Animais , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8A/metabolismo , Dor do Câncer/etiologia , Dor do Câncer/metabolismo , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , RNA Interferente Pequeno/metabolismo , Neoplasias Ósseas/complicações , Neoplasias Ósseas/metabolismo , Receptores de Interleucina-8B/metabolismo , Neuralgia/metabolismo , Medula Espinal/metabolismo
9.
Cancer Cell Int ; 22(1): 228, 2022 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-35818076

RESUMO

BACKGROUND: Prostate cancer (PCa) is one of the most common malignant tumors in the male urinary system. In recent years, the morbidity and mortality of PCa have been increasing due to the limited effects of existing treatment strategies. Long non-coding RNA (lncRNA) LINC00893 was reported to inhibit the proliferation and metastasis of papillary thyroid cancer cells, but its role in PCa has not been reported. This study aims to investigate the role and underlying mechanism of LINC00893 in regulating the progression of PCa cells. METHODS: We first compared LINC00893 expression levels between PCa tissues and normal prostate tissues through TCGA database. The relative LINC00893 expression levels were further validated in 66 pairs of PCa tissues and para-cancerous normal tissues, as well as in PCa cell lines. Gain-of-function experiment was performed by transfecting PCa cell with LINC00893 expression vector, and CCK (Cell count kit)-8, 5-Ethynyl-2'-deoxyuridine (EdU) incorporation, colony information and transwell assays were conducted to assess the functional phenotypes. Dual-luciferase reporter, RNA-binding protein immunoprecipitation (RIP) and RNA pull-down assays were performed to evaluate the molecular interactions. RESULTS: LINC00893 was downregulated in PCa tissues and cell lines, and patients with low expression of LINC00893 were associated with a poorer overall survival rate. LINC00893 overexpression hindered the proliferation, epithelial-mesenchymal transition (EMT) as well as the migratory ability of PCa cells, and suppressed the tumorigenesis of PCa cells in nude mice. We further demonstrated that LINC00893 acted as a sponge for miR-3173-5p and inhibited its activity, which in turn regulated the suppressor of cytokine signaling 3 (SOCS3)/Janus Kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling axis. CONCLUSIONS: Our study demonstrated that LINC00893 suppresses the progression of PCa cells through targeting miR-3173-5p/SOCS3/JAK2/STAT3 axis. Our data uncovers a novel tumor-suppressor role of LINC00893 in PCa, which may serve as a potential strategy for targeted therapy in PCa.

10.
J Clin Lab Anal ; 36(9): e24536, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35837990

RESUMO

OBJECTIVE: This study aimed to clarify the function of miR-630 on non-small cell lung cancer (NSCLC) cells. METHODS: Quantitative real-time PCR was utilized to detect the mRNA expression of miR-630 and vimentin (VIM) in NSCLC tissues and cells. The protein expression of VIM, P53, Caspase-3, Bcl-2, Bax and JAK2/STAT3 was evaluated via Western blot. Dual-luciferase reporter assay was applied to evaluate whether VIM is the target gene of miR-630. The migration, invasion, proliferation and apoptosis of NSCLC cells were examined by wound-healing assay, transwell assay, CCK-8 assay, and flow cytometry, respectively. RESULTS: MiR-630 was lowly expressed in NSCLC tissues and cells, while VIM was highly expressed in NSCLC cells. Dual-luciferase reporter assay data validated that miR-630 directly targeted VIM. MiR-630 overexpression inhibited VIM expression, but the inhibition of miR-630 upregulated VIM expression. Besides, miR-630 mimics restrained cell migration, invasion, and proliferation, and promoted NSCLC cell apoptosis. Whereas, VIM overexpression partly attenuated the inhibitory effect of miR-630 on NSCLC cells. Moreover, miR-630 mimics impeded p-JAK2 and p-STAT3 protein expression; and miR-630 inhibitor upregulated p-STAT3 and VIM protein expression, which was reversed after the addition of STAT3 inhibitor C188-9. CONCLUSION: MiR-630 constrained the progression of NSCLC by inhibiting JAK2/STAT3 pathway and downregulating VIM expression.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , MicroRNAs , Apoptose/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Vimentina/genética , Vimentina/metabolismo , Vimentina/farmacologia
11.
Zhongguo Zhong Yao Za Zhi ; 47(20): 5473-5480, 2022 Oct.
Artigo em Zh | MEDLINE | ID: mdl-36471962

RESUMO

Urticaria is an immune-mediated allergic disease. This study explored the effect of Jingfang Mixture on spleen T lymphocyte subsets of urticaria mice. A total of 50 Kunming mice were randomized into normal group(C), model group(V), and low-(JF-L, 0.5 g·kg~(-1)), medium-(JF-M, 1 g·kg~(-1)) and high-dose(JF-H, 2 g·kg~(-1)) Jingfang Mixture groups, with 10 mice in each group. The mixture of ovalbumin and aluminum hydroxide(0.1 mg + 0.1 mL) was used(intraperitoneal injection) to induce urticaria in mice. The administration began 6 days after the first immunization, and the second immunization was carried out 10 days after the first immunization. The pruritus index was detected within 30 min after the second immunization. The administration lasted 21 days. After 21 days, the serum was taken to detect the total IgE level. Based on hematoxylin and eosin(HE) staining, the pathological changes of skin tissue were observed, and Western blot was used to detect the levels of p-Janus kinase 2(JAK2)/JAK2 and p-signal transducer and activator of transcription 3(STAT3)/STAT3 in skin tissue. The spleen was taken to detect the spleen index, and flow cytometry was employed to determine the expression of lymphocyte subsets. The results showed that group V had obvious pathological changes in skin tissue compared with group C. Moreover, group V showed more scratches, higher spleen index, and higher level of total serum IgE than group C. In addition, higher levels of p-JAK2 and p-STAT3, lower proportions of CD4~+T, Th1, and Treg, higher proportions of CD8~+T, Th2, and Th17, and lower ratios of CD4~+/CD8~+, Th1/Th2, and Terg/Th17 were observed in group V than in group C. Compared with group V, each administration group showed alleviation of the pathological morphology of skin tissue, obvious epidermal thickening, relatively intact collagen fiber structure of dermal reticular layer, alleviated edema, and relief of vasodilation and peripheral inflammatory cell infiltration. Moreover, less scratching, lower spleen index, lower p-JAK2/JAK2 and p-STAT3/STAT3 were observed in the administration groups than in group V. JF-M group and JF-H group demonstrated lower levels of total IgE, larger proportions of CD4~+T, Th1, and Treg, smaller proportions of CD8~+ T, Th2, and Th17, and higher ratios of CD4~+/CD8~+, Th1/Th2, and Terg/Th17. In conclusion, Jingfang Mixture may improve the symptoms of urticaria mice by regulating the balance of spleen T lymphocyte subsets through JAK2-STAT3 signaling pathway.


Assuntos
Janus Quinase 2 , Urticária , Camundongos , Animais , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Janus Quinase 2/farmacologia , Baço , Subpopulações de Linfócitos T/metabolismo , Transdução de Sinais , Imunoglobulina E
12.
Neurochem Res ; 46(4): 945-956, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33515352

RESUMO

Spinal cord injury (SCI) is a serious neurological disease. Long non-coding RNA (lncRNA) small nucleolar RNA host gene (SNHG1) and microRNA-362-3p (miR-362-3p) were confirmed to be related to neurological disorders. However, it is unclear whether SNHG1 was involved in the development of SCI via regulating miR-362-3p. PC12 cells were treated with lipopolysaccharide (LPS) to imitate the in vitro cell model of SCI. Cell ciability and apoptosis rate were detected by cell counting kit-8 (CCK-8) assay and flow cytometry assay. The levels of SNHG1, miR-362-3p, and Janus kinase-2 (Jak2) were examined by quantitative real-time polymerase chain reaction (qRT-PCR). The dual-luciferase reporter assay, RNA pull-down assay, and RNA immunoprecipitation (RIP) assay were performed to verify the interaction between miR-362-3p and SNHG1 or Jak2. Besides, the levels of apoptosis- and autophagy- related proteins were detected by western blot assay. In present research, LPS suppressed cell viability, and induced apoptosis and autophagy in PC12 cells. SNHG1 knockdown could affect cell viability, and suppress cell apoptosis and autophagy in LPS-treated PC12 cells. Moreover, miR-362-3p was a target of SNHG1, miR-362-3p targeted Jak2 and negatively regulated Jak2/stat3 pathway. Our data also demonstrated that SNHG1 depletion inactivated Jak2/stat3 pathway to affect cell viability and confine apoptosis, autophagy in LPS-treated PC12 cells. Taken together, SNHG1 regulated cell viability, apoptosis and autophagy in LPS-treated PC12 cells by activating Jak2/stat3 pathway via sponging miR-362-3p.


Assuntos
Apoptose/fisiologia , Autofagia/fisiologia , RNA Longo não Codificante/metabolismo , Transdução de Sinais/fisiologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Autofagia/efeitos dos fármacos , Autofagia/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Técnicas de Silenciamento de Genes , Janus Quinase 2/metabolismo , Lipopolissacarídeos/toxicidade , MicroRNAs/metabolismo , Células PC12 , RNA Longo não Codificante/genética , Ratos , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/genética
13.
J Appl Toxicol ; 41(12): 2042-2054, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34081793

RESUMO

PM2.5 is a well-known air pollutant threatening public health. Studies confirmed that exposure to the particles could impair pulmonary function, cause chronic obstructive pulmonary disease, and increase the incidence of lung cancer. The characteristic of PM2.5 varies across regions. The toxic function of PM2.5 in southwest China remains to be elucidated. This study aimed to investigate lung injury and its mechanisms induced by PM2.5 collected in Chengdu. Rats were administered with PM2.5 by intratracheal instillation for 4 weeks. Biochemical, cell count, and inflammation-related parameters were measured. Lung tissues were obtained for hematoxylin and eosin and Masson's trichrome staining. The expression levels of vascular endothelial growth factor (VEGF), Janus tyrosine protein kinase-2 (JAK-2), and signal transducer and activator of transcription-3 (STAT-3) were detected by immunohistochemistry assays. Meanwhile, A549 cells were treated with the PM2.5. The cell cycle, and apoptosis were measured by flow cytometry. mRNA and protein expressions of JAK-2, STAT-3, p-STAT-3, and VEGFA were detected using qPCR and Western blot analysis respectively. Results of in vivo study showed that PM2.5 induced lung pathological injury, aggravated the accumulation of inflammatory cells, and increased the serum levels of inflammatory factors. In vitro experiments showed that PM2.5 disrupted the cell growth cycle and increased cell apoptosis through the activation of the JAK-2/STAT-3 signaling pathway. Taken together, this study provided convincing experimental evidence that PM2.5 collected in southwest China could induce pulmonary injury as manifested by inflammatory response and lung fibrosis, possibly through the modulation of the JAK-2/STAT-3 signaling pathway.


Assuntos
Poluentes Atmosféricos/toxicidade , Janus Quinase 2/genética , Material Particulado/toxicidade , Fator de Transcrição STAT3/genética , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/genética , Células A549 , Animais , China , Humanos , Janus Quinase 2/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Fator de Transcrição STAT3/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
Ecotoxicol Environ Saf ; 226: 112872, 2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34624536

RESUMO

BACKGROUND: Abnormal lipid accumulation in macrophages may lead to macrophages foaming, which is the most important pathological process of atherosclerosis. Atmospheric PM2.5 could enter the blood circulation and further affect the lipid metabolism of macrophages. But the underlying mechanism is not unclear. This study was undertaken to clarify the effect of PM2.5 on lipid metabolism in macrophages, and to explore the role of inflammatory reaction and JAK2/STAT3 signaling pathway in this process. METHOD: Macrophages derived from THP-1 cells were exposed to PM2.5 (0,100,200,400 µg/mL) for 6 h and 12 h. STAT3 agonist ColivelinTFA is used to specifically excite STAT3. The survival rate of macrophages was detected by CCK-8. The lipid levels in macrophages were detected by colorimetry. The levels of inflammatory factors secreted by macrophages were detected by ELISA. Q-PCR was used to detect the mRNA expression levels, and Western Blot was used to detect the protein expression levels of JAK2/STAT3 pathway genes. RESULT: The survival rate of macrophages was reduced by PM2.5, and the levels of TG, T-CHO and LDL-C of macrophages exposed to PM2.5 were increased. PM2.5 led to the increasing level of IL-6 and the decreasing level of IL-4, and the JAK2/STAT3 signaling pathway was inhibited by PM2.5. Colivelin TFA significantly decreased the increasing levels of TG, T-CHO and LDL-C levels, and increased the decreasing mRNA levels of IL-4, and LPL induced by PM2.5 (p < 0.05). DISCUSSION: PM2.5 could cause the lipid accumulation of macrophages by inhibiting the JAK2/STAT3 signaling pathway, and inflammatory responses may be involved in this process.


Assuntos
Macrófagos , Transdução de Sinais , Humanos , Inflamação/induzido quimicamente , Janus Quinase 2/genética , Lipídeos , Material Particulado/toxicidade , Fator de Transcrição STAT3/genética
15.
Microvasc Res ; 132: 104060, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32818511

RESUMO

BACKGROUND: Cardiovascular diseases are associated with proliferation and phenotypic switch. Platelet-derived growth factor-BB (PDGF-BB) is a major initiating factor for proliferative vascular diseases, such as neointimal lesion formation, restenosis after angioplasty, and atherosclerosis. Ruxolitinib, a potent Janus kinase (JAK) 1 and 2 inhibitor, has been reported to significantly block the proliferation-related signaling pathway of JAK2/signal transducers and activators of transcription 3 (STAT3) and harbor a broad spectrum of anti-cancer activities, including proliferation inhibition, apoptosis induction, and anti-inflammation. However, the role of ruxolitinib in regulating PDGF-BB-induced VSMC proliferation remains to be elucidated. Thus, this study investigates the role of ruxolitinib in regulating PDGF-BB-induced VSMC proliferation and its underlying mechanisms. METHODS: In vivo, the medial thickness of the carotid artery was evaluated using a mouse carotid ligation model, ruxolitinib was administered orally to the mice every other day, and the mice were euthanized on day 28 to evaluate the therapeutic effects of ruxolitinib. Cell proliferation markers were measured using real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blotting. In vitro, VSMCs were treated with ruxolitinib with or without PDGF-BB at an indicated time and concentration. Cell proliferation and apoptosis were measured using Cell Counting Kit-8 assay, MTS assays and flow cytometry. The JAK2/STAT3 signaling pathway involved in the effects of ruxolitinib on VSMCs was detected by western blotting with the specific pathway inhibitor AG490. RESULTS: In vivo, ruxolitinib significantly decreased the ratio-of-intima ratio (I/M ratio) by inhibiting the expression of PCNA and cyclinD1 (p <0.05). In vitro, ruxolitinib inhibited PDGF-BB-induced VSMC proliferation compared with the PDGF-BB treatment group (p <0.05). In addition, ruxolitinib inhibited the PDGF-BB-induced activation of the JAK2/STAT3 signaling pathway and decreased the expression of proliferation related-proteins cyclinD1 and PCNA in VSMCs (p <0.05). CONCLUSION: Our findings suggest that ruxolitinib inhibits VSMC proliferation in vivo and in vitro by suppressing the activation of the JAK2/STAT3 signaling pathway. Therefore, ruxolitinib has a therapeutic potential for proliferative vascular diseases.


Assuntos
Becaplermina/farmacologia , Estenose das Carótidas/prevenção & controle , Janus Quinase 2/metabolismo , Inibidores de Janus Quinases/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Neointima , Pirazóis/farmacologia , Fator de Transcrição STAT3/metabolismo , Animais , Artéria Carótida Primitiva/efeitos dos fármacos , Artéria Carótida Primitiva/enzimologia , Artéria Carótida Primitiva/patologia , Estenose das Carótidas/enzimologia , Estenose das Carótidas/patologia , Células Cultivadas , Ciclina D1/metabolismo , Modelos Animais de Doenças , Hiperplasia , Masculino , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/patologia , Nitrilas , Antígeno Nuclear de Célula em Proliferação/metabolismo , Pirimidinas , Transdução de Sinais
16.
FASEB J ; 33(4): 5112-5125, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30652956

RESUMO

Peritoneal fibrosis (PF) represents a well-recognized complication associated with continuous ambulatory peritoneal dialysis therapy, characterized by a reversible epithelial-to-mesenchymal transition (EMT) at the early stage. The aim of the current study was to investigate the effects linked with the long noncoding RNA (lncRNA) AK089579 on the EMT of peritoneal mesothelial cells (PMCs) as well as the associated regulatory mechanisms of AK089579 downstream of tyrosine kinase 2 (DOK2) and microRNA-296-3p (miR-296-3p). Enrichment analysis, gene intersection association analysis, and a gene-gene intersection network were initially constructed to ascertain whether AK089579 regulated the expression of DOK2 through the mediation of miR-296-3p via the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway in PF. After the PF mouse model had been constructed, the expression of the proteins associated with the JAK2/STAT3 signaling pathway and EMT and PMC migration and invasion were all determined accordingly. Based on the obtained results, AK089579 was determined to function as a competing endogenous RNA for miR-296-3p while acting to up-regulate the expression of DOK2, which is a target gene of miR-296-3p. AK089579 was detected to confer an inhibitory effect on the activation of the JAK2/STAT3 signaling pathway, whereby the migration and invasion of PMCs among the mice models were suppressed. Meanwhile, up-regulated miR-296-3p and down-regulated DOK2 produced contrasting effects when compared with the aforementioned findings. Treatment with wp10066, a JAK2/STAS3 signaling pathway inhibitor, was shown to reverse the effects exerted by up-regulated miR-296-3p. Taken together, the central findings of the current study present evidence highlighting the capability of the lncRNA AK089579 to bind competitively to miR-296-3p and indirectly enhance the expression of DOK2, which in turn suppresses the activation of the JAK2/STAT3 signaling pathway, whereby the EMT, migration, and invasion of PMCs was inhibited in PF.-Zhang, X. W., Wang, L., Ding, H. Long noncoding RNA AK089579 inhibits epithelial-to-mesenchymal transition of peritoneal mesothelial cells by competitively binding to microRNA-296-3p via DOK2 in peritoneal fibrosis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Movimento Celular/genética , Movimento Celular/fisiologia , Células Cultivadas , Transição Epitelial-Mesenquimal/genética , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/genética , Análise em Microsséries , Fibrose Peritoneal/genética , Fibrose Peritoneal/metabolismo , RNA Longo não Codificante/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
17.
Mol Cell Biochem ; 463(1-2): 175-187, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31712941

RESUMO

Calycosin-7-O-ß-D-glucoside (CG) is the component of Astragali Radix, and the aim of the present study is to investigate whether CG protects myocardium from I/R-induced damage by the regulation of IL-10/JAK2/STAT3 signaling pathway. H9C2 cells were subjected to I/R treatment and pretreated with 1 µm CG in vitro. In addition, a mouse model of myocardial I/R injury was induced by left anterior descending (LAD) coronary artery ligation and administrated with 30 mg/kg CG by intravenous injection before I/R surgery. In vitro and in vivo results showed that CG up-regulated IL-10 level, activated the JAK2/STAT3 pathway, and protected myocardial cells from I/R-induced apoptosis. The hemodynamic measurement, TTC staining, TUNEL staining, and western blot results in vivo showed that the protective effects of CG on myocardial function and cell apoptosis were all reversed by the IL-10R α neutralizing antibody. CG-induced phosphorylation activation of JAK2/STAT3 signaling pathway was also suppressed by the blocking of IL-10. In summary, these findings suggest that CG might alleviate myocardial I/R injury by activating the JAK2/STAT3 signaling pathway via up-regulation of IL-10 secretion, which provides us insights into the mechanism underlying the protective effect of CG on myocardial I/R injury.


Assuntos
Glucosídeos/farmacologia , Interleucina-10/metabolismo , Isoflavonas/farmacologia , Janus Quinase 2/metabolismo , Traumatismo por Reperfusão Miocárdica , Miocárdio/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular , Camundongos , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/patologia , Ratos
18.
J Biochem Mol Toxicol ; 34(5): e22464, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32020710

RESUMO

Paroxetine, a representative of serotonin reuptake inhibitors, has recently gained attention due to its anti-inflammatory properties. However, underlying mechanisms responsible for its immunosuppressive effects remain to be unveiled. To understand the responsible signaling mechanisms, we examined paroxetine's effect on the Janus kinase 2-signal transducer and activator of transcription 3 (JAK2-STAT3) signaling pathway on lipopolysaccharide + phytohemagglutinin-induced human peripheral blood mononuclear cells culture. We also evaluate the possible dependency of paroxetine immunomodulation effects on the 5-HT system of immune cells. Our results indicated that paroxetine attenuates proinflammatory cytokine production (interleukin-1ß [IL-1ß], IL-17, and tumor necrosis factor-α) and increases expression of IL-10 and JAK2/STAT3 evidence for macrophages polarization to M2 subset and functional dendritic cells depletion. In conclusion, paroxetine can exert its anti-inflammatory effects via both the 5-HT systems present in immune cells and the JAK2-STAT3 pathway. Our results also suggest that paroxetine exerted its immunosuppressive effects partially via serotonin. Nonetheless, JAK2/STAT3-modulated paroxetine effects were independent of serotonin, hence sufficiently applicable for inflammation repression.


Assuntos
Anti-Inflamatórios/farmacologia , Imunidade/efeitos dos fármacos , Imunossupressores/farmacologia , Janus Quinase 2/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Paroxetina/farmacologia , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Adulto , Doadores de Sangue , Polaridade Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/metabolismo , Humanos , Leucócitos Mononucleares/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Fito-Hemaglutininas/farmacologia , Serotonina/metabolismo , Adulto Jovem
19.
BMC Cardiovasc Disord ; 20(1): 85, 2020 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-32066388

RESUMO

BACKGROUND: Coronary microembolization (CME) has a poor prognosis, with ventricular arrhythmia being the most serious consequence. Understanding the underlying mechanisms could improve its management. We investigated the effects of granulocyte colony-stimulating factor (G-CSF) on connexin-43 (Cx43) expression and ventricular arrhythmia susceptibility after CME. METHODS: Forty male rabbits were randomized into four groups (n = 10 each): Sham, CME, G-CSF, and AG490 (a JAK2 selective inhibitor). Rabbits in the CME, G-CSF, and AG490 groups underwent left anterior descending (LAD) artery catheterization and CME. Animals in the G-CSF and AG490 groups received intraperitoneal injection of G-CSF and G-CSF + AG490, respectively. The ventricular structure was assessed by echocardiography. Ventricular electrical properties were analyzed using cardiac electrophysiology. The myocardial interstitial collagen content and morphologic characteristics were evaluated using Masson and hematoxylin-eosin staining, respectively. RESULTS: Western blot and immunohistochemistry were employed to analyze the expressions of Cx43, G-CSF receptor (G-CSFR), JAK2, and STAT3. The ventricular effective refractory period (VERP), VERP dispersion, and inducibility and lethality of ventricular tachycardia/fibrillation were lower in the G-CSF than in the CME group (P < 0.01), indicating less severe myocardial damage and arrhythmias. The G-CSF group showed higher phosphorylated-Cx43 expression (P < 0.01 vs. CME). Those G-CSF-induced changes were reversed by A490, indicating the involvement of JAK2. G-CSFR, phosphorylated-JAK2, and phosphorylated-STAT3 protein levels were higher in the G-CSF group than in the AG490 (P < 0.01) and Sham (P < 0.05) groups. CONCLUSION: G-CSF might attenuate myocardial remodeling via JAK2-STAT3 signaling and thereby reduce ventricular arrhythmia susceptibility after CME.


Assuntos
Arritmias Cardíacas/prevenção & controle , Doença da Artéria Coronariana/tratamento farmacológico , Fator Estimulador de Colônias de Granulócitos/farmacologia , Frequência Cardíaca/efeitos dos fármacos , Janus Quinase 2/metabolismo , Infarto do Miocárdio/prevenção & controle , Miocárdio/enzimologia , Função Ventricular Esquerda/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Animais , Arritmias Cardíacas/enzimologia , Arritmias Cardíacas/patologia , Arritmias Cardíacas/fisiopatologia , Conexina 43/metabolismo , Doença da Artéria Coronariana/enzimologia , Doença da Artéria Coronariana/patologia , Doença da Artéria Coronariana/fisiopatologia , Modelos Animais de Doenças , Fibrose , Masculino , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Fosforilação , Coelhos , Receptores de Fator Estimulador de Colônias de Granulócitos/metabolismo , Período Refratário Eletrofisiológico/efeitos dos fármacos , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais
20.
J Cell Physiol ; 234(5): 6182-6193, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30370578

RESUMO

MicroRNAs (miRs) were involved in numerous cardiovascular diseases, especially ischemic heart diseases, but the miR changes during cardiac ischemia-reperfusion (I/R) injury following sevoflurane (SEV) preconditioning are still unknown. This study aims to investigate the effect of miR-874 on cardiac I/R injury in mouse models pretreated with SEV. Following establishment of mouse models with myocardial I/R injury, mice were pretreated with SEV. The functional mechanism of miR-874 in I/R injury was explored when miR-874 and the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway were inhibited. Terminal deoxynucleotidyl transferase (TdT)-mediated dUTP biotin nick-end labeling (TUNEL) staining was used to detect cardiomyocyte apoptosis and dual luciferase reporter gene assay to identify the targeting relationship between miR-874 and STAT3. Expression of the JAK2/STAT3 signaling pathway and apoptosis-related genes was determined. Initially, upregulated miR-874 was observed in I/R mice. Then, miR-874 inhibition improved cardiac function of I/R mice, inhibited cardiomyocyte apoptosis (also shown as decreased Bcl-2 associated X protein B [Bax] and increased B-cell lymphoma-2 [Bcl-2]), and activated the JAK2/STAT3 signaling pathway. STAT3, a target gene of miR-874, was upregulated following miR-874 inhibition. Finally, we also observed that the effect of miR-874 was lost when the JAK2/STAT3 signaling pathway was blocked. The findings indicate miR-874 as a contributory role in cardiac I/R injury, with miR-874 inhibition alleviating cardiac I/R injury in mice following SEV pretreatment by targeting STAT3 through the JAK2/STAT3 signaling pathway.


Assuntos
Apoptose/fisiologia , MicroRNAs/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Modelos Animais de Doenças , Masculino , Camundongos , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA