Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell Commun Signal ; 22(1): 148, 2024 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-38395872

RESUMO

BACKGROUND: Tubulointerstitial kidney disease associated microenvironmental dysregulation, like acidification, inflammation and fibrosis, affects tubule cells and fibroblasts. Micromilieu homeostasis influences intracellular signaling and intercellular crosstalk. Cell-cell communication in turn modulates the interstitial microenvironment. We assessed the impact of acidosis on inflammatory and fibrotic responses in proximal tubule cells and fibroblasts as a function of cellular crosstalk. Furthermore, cellular signaling pathways involved were identified. METHODS: HK-2 (human proximal tubule) and CCD-1092Sk (human fibroblasts), in mono and coculture, were exposed to acidic or control media for 3 or 48 h. Protein expression of inflammation markers (TNF, TGF-ß and COX-2), dedifferentiation markers (N-cadherin, vinculin, ß-catenin and vimentin), fibrosis markers (collagen III and fibronectin) and phospho- as well as total MAPK levels were determined by western blot. Secreted collagen III and fibronectin were measured by ELISA. The impact of MAPK activation was assessed by pharmacological intervention. In addition, necrosis, apoptosis and epithelial permeability were determined. RESULTS: Independent of culture conditions, acidosis caused a decrease of COX-2, vimentin and fibronectin expression in proximal tubule cells. Only in monoculture, ß-Catenin expression decreased and collagen III expression increased in tubule cells during acidosis. By contrast, in coculture collagen III protein expression of tubule cells was reduced. In fibroblasts acidosis led to an increase of TNF, COX-2, vimentin, vinculin, N-cadherin protein expression and a decrease of TGF-ß expression exclusively in coculture. In monoculture, expression of COX-2 and fibronectin was reduced. Collagen III expression of fibroblasts was reduced by acidosis independent of culture conditions. In coculture, acidosis enhanced phosphorylation of ERK1/2, JNK1/2 and p38 transiently in proximal tubule cells. In fibroblasts, acidosis enhanced phosphorylation of p38 in a sustained and very strong manner. ERK1/2 and JNK1/2 were not affected in fibroblasts. Inhibition of JNK1/2 and p38 under coculture conditions reduced acidosis-induced changes in fibroblasts significantly. CONCLUSIONS: Our data show that the crosstalk between proximal tubule cells and fibroblasts is crucial for acidosis-induced dedifferentiation of fibroblasts into an inflammatory phenotype. This dedifferentiation is at least in part mediated by p38 and JNK1/2. Thus, cell-cell communication is essential for the pathophysiological impact of tubulointerstitial acidosis.


Assuntos
Acidose , Fibronectinas , Proteínas Quinases p38 Ativadas por Mitógeno , Humanos , Acidose/metabolismo , Caderinas/metabolismo , Cateninas/metabolismo , Colágeno/metabolismo , Ciclo-Oxigenase 2/metabolismo , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Fibrose , Inflamação/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Vimentina/metabolismo , Vinculina/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/metabolismo
2.
Neurochem Res ; 49(8): 2249-2270, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38837092

RESUMO

Morphine (Mor) has exhibited efficacy in safeguarding neurons against ischemic injuries by simulating ischemic/hypoxic preconditioning (I/HPC). Concurrently, autophagy plays a pivotal role in neuronal survival during IPC against ischemic stroke. However, the involvement of autophagy in Mor-induced neuroprotection and the potential mechanisms remain elusive. Our experiments further confirmed the effect of Mor in cellular and animal models of ischemic stroke and explored its potential mechanism. The findings revealed that Mor enhanced cell viability in a dose-dependent manner by augmenting autophagy levels and autophagic flux in neurons subjected to oxygen-glucose deprivation/reoxygenation (OGD/R). Pretreatment of Mor improved neurological outcome and reduced infarct size in mice with middle cerebral artery occlusion/reperfusion (MCAO/R) at 1, 7 and 14 days. Moreover, the use of autophagy inhibitors nullified the protective effects of Mor, leading to reactive oxygen species (ROS) accumulation, increased loss of mitochondrial membrane potential (MMP) and neuronal apoptosis in OGD/R neurons. Results further demonstrated that Mor-induced autophagy activation was regulated by mTOR-independent activation of the c-Jun NH2- terminal kinase (JNK)1/2 Pathway, both in vitro and in vivo. Overall, these findings suggested Mor-induced neuroprotection by activating autophagy, which were regulated by JNK1/2 pathway in ischemic stroke.


Assuntos
Autofagia , AVC Isquêmico , Morfina , Fármacos Neuroprotetores , Serina-Treonina Quinases TOR , Animais , Autofagia/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Masculino , AVC Isquêmico/tratamento farmacológico , AVC Isquêmico/metabolismo , AVC Isquêmico/patologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Morfina/farmacologia , Morfina/uso terapêutico , Camundongos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos Endogâmicos C57BL , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Neuroproteção/efeitos dos fármacos , Neuroproteção/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Espécies Reativas de Oxigênio/metabolismo
3.
J Cell Mol Med ; 27(9): 1250-1260, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36967712

RESUMO

Oral squamous cell carcinoma (OSCC) has a high recurrence rate and poor prognosis. Hispolon, a polyphenolic compound with antiviral, antioxidant, and anticancer activities, is a potential chemotherapy agent. However, few studies have investigated the anti-cancer mechanism of hispolon in oral cancer. This present study used the cell viability assay, clonogenic assay, fluorescent nuclear staining, and flow cytometry assay to analyse the apoptosis-inducing effects of hispolon in OSCC cells. After hispolon treatment, the apoptotic initiators, cleaved caspase-3, -8, and - 9, were upregulated, whereas the cellular inhibitor of apoptosis protein-1 (cIAP1) was downregulated. Furthermore, a proteome profile analysis using a human apoptosis array revealed the overexpression of heme oxygenase-1 (HO-1) by hispolon, which was determined to be involved in caspase-dependent apoptosis. Moreover, cotreatment with hispolon and mitogen-activated protein kinase (MAPK) inhibitors revealed that hispolon induces apoptosis in OSCC cells through activation of the c-Jun N-terminal kinase (JNK) pathway and not the extracellular signal-regulated kinase (ERK) or p38 pathway. These findings indicate that hispolon may exert an anticancer effect on oral cancer cells by upregulating HO-1 and inducing caspase-dependent apoptosis by activating the JNK pathway.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Neoplasias Bucais , Humanos , Sistema de Sinalização das MAP Quinases , Heme Oxigenase-1 , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas de Cabeça e Pescoço , Neoplasias Bucais/tratamento farmacológico , Apoptose , Proteínas Quinases JNK Ativadas por Mitógeno , Linhagem Celular Tumoral , Proteínas Quinases p38 Ativadas por Mitógeno
4.
FASEB J ; 36(4): e22243, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35224782

RESUMO

Osteoarthritis (OA) is a prevalent degenerative disease of the joint, featured by articular cartilage destruction and subchondral bone marrow lesions. Articular cartilage and subchondral bone constitute an osteochondral unit that guarantees joint homeostasis. During OA initiation, activated osteoclasts in subchondral bone ultimately result in impaired capacities of the subchondral bone in response to mechanical stress, followed by the degradation of overlying articular cartilage. Thus, targeting osteoclasts could be a potential therapeutic option for treating OA. Here, we observed that farnesoid X receptor (FXR) expression and osteoclast fusion and activity in subchondral bone were concomitantly changed during early-stage OA in the OA mouse model established by anterior cruciate ligament transection (ACLT). Then, we explored the therapeutic effects of FXR agonist GW4064 on the osteochondral pathologies in ACLT mice. We showed that GW4064 obviously ameliorated subchondral bone deterioration, associated with reduction in tartrate-resistant acid phosphatase (TRAP) positive multinuclear osteoclast number, as well as articular cartilage degradation, which were blocked by the treatment with FXR antagonist Guggulsterone. Mechanistically, GW4064 impeded osteoclastogenesis through inhibiting subchondral bone osteoclast fusion via suppressing c-Jun N-terminal kinase (JNK) 1/2/nuclear factor of activated T-cells 1 (NFATc1) pathway. Taken together, our results present evidence for the protective effects of GW4064 against OA by blunting osteoclast-mediated aberrant subchondral bone loss and subsequent cartilage deterioration. Therefore, GW4064 demonstrates the potential as an alternative therapeutic option against OA for further drug development.


Assuntos
Reabsorção Óssea/prevenção & controle , Regulação da Expressão Gênica/efeitos dos fármacos , Isoxazóis/farmacologia , Osteoartrite/prevenção & controle , Osteoclastos/efeitos dos fármacos , Osteogênese , Proteínas de Ligação a RNA/agonistas , Animais , Remodelação Óssea , Reabsorção Óssea/etiologia , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/genética , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Osteoartrite/etiologia , Osteoartrite/metabolismo , Osteoartrite/patologia , Osteoclastos/metabolismo , Osteoclastos/patologia
5.
J Cell Mol Med ; 26(8): 2273-2284, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35191177

RESUMO

Human oral squamous cell carcinoma (OSCC) is the common head and neck malignancy in the world. While surgery, radiotherapy and chemotherapy are emerging as the standard treatment for OSCC patients, the outcome is limited to the recurrence and side effects. Therefore, patients with OSCC require alternative strategies for treatment. In this study, we aimed to explore the therapeutic effect and the mode of action of the novel curcumin analog, HO-3867, against human OSCC cells. We analysed the cytotoxicity of HO-3867 using MTT assay. In vitro mechanic studies were performed to determine whether MAPK pathway is involved in HO-3867 induced cell apoptosis. As the results, we found HO-3867 suppressed OSCC cells growth effectively. The flow cytometry data indicate that HO-3867 induce the sub-G1 phase. Moreover, we found that HO-3867 induced cell apoptosis by triggering formation of activated caspase 3, caspase 8, caspase 9 and PARP. After dissecting MAPK pathway, we found HO-3867 induced cell apoptosis via the c-Jun N-terminal kinase (JNK)1/2 pathway. Our results suggest that HO-3867 is an effective anticancer agent as its induction of cell apoptosis through JNK1/2 pathway in human oral cancer cells.


Assuntos
Carcinoma de Células Escamosas , Curcumina , Neoplasias de Cabeça e Pescoço , Neoplasias Bucais , Apoptose , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Curcumina/uso terapêutico , Humanos , Neoplasias Bucais/patologia , Piperidonas , Carcinoma de Células Escamosas de Cabeça e Pescoço
6.
Cell Biol Int ; 46(1): 148-157, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34694031

RESUMO

Currently, the prevention of ischemic diseases such as myocardial infarction associated with ischemia/reperfusion (I/R) injury remains to be a challenge. Thus, this study was designed to explore the effects of tripartite motif protein 11 (TRIM11) on cardiomyocytes I/R injury and its underlying mechanism. Cardiomyocytes AC16 were used to establish an I/R injury cell model. After TRIM11 downregulation in I/R cells, cell proliferation (0, 12, 24, and 48 h) and apoptosis at 48 h as well as the related molecular changes in oxidative stress-related pathways was detected. Further, after the treatment of TRIM11 overexpression, SP600125, or DUSP1 overexpression, cell proliferation, apoptosis, and related genes were detected again. As per our findings, it was determined that TRIM11 was highly expressed in the cardiomyocytes AC16 after I/R injury. Downregulation of TRIM11 was determined to have significantly reduced I/R-induced proliferation suppression and apoptosis. Besides, I/R-activated c-Jun N-terminal kinase (JNK) signaling and cleaved caspase 3 and Bax expression were significantly inhibited by TRIM11 downregulation. In addition, the overexpression of TRIM11 significantly promoted apoptosis in AC16 cells, and JNK1/2 inhibition and DUSP1 overexpression potently counteracted the induction of TRIM11 overexpression in AC16 cells. These suggested that the downregulation of TRIM11 attenuates apoptosis in AC16 cells after I/R injury probably through the DUSP1-JNK1/2 pathways.


Assuntos
Apoptose , Fosfatase 1 de Especificidade Dupla/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Traumatismo por Reperfusão Miocárdica/enzimologia , Miócitos Cardíacos/enzimologia , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Hipóxia Celular , Linhagem Celular , Regulação para Baixo , Fosfatase 1 de Especificidade Dupla/genética , Humanos , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miócitos Cardíacos/patologia , Transdução de Sinais , Proteínas com Motivo Tripartido/genética , Ubiquitina-Proteína Ligases/genética
7.
J Biochem Mol Toxicol ; 36(7): e23051, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35315184

RESUMO

Psoralidin (PSO) is a natural coumarin isolated from the seeds of Psoralea corylifolia Linn. Previous studies have reported that PSO exerts numerous pharmacological bioactivities including antitumor. The present study aimed to investigate its anticancer effect using colon cancer cells. Cultured HT-29 and HCT-116 colon cancer cells were treated with different concentrations of PSO, and the cell viability, the intracellular reactive oxygen species (ROS), the protein expression, and the apoptosis were determined by MTT assay, DCFH2 -DA fluorescence probe, Western blotting, and Annexin V/7-AAD staining, respectively. The activities of caspase 3/7 were determined by a commercial kit. Our study found that PSO effectively induces apoptotic cell death mediated by caspase 3/7 in HT-29 and HCT-116 colon cancer cells. PSO treatment rapidly boosts the ROS generation, which is responsible for the PSO-triggered DNA damage, mitochondria membrane potential decrease and caspase 3/7 activation, and c-Jun N-terminal kinase 1/2 activation. Collectively, these results showed that PSO triggered oxidative damage mediated apoptosis in colon cancer cells.


Assuntos
Benzofuranos , Neoplasias do Colo , Cumarínicos , Psoralea , Apoptose , Benzofuranos/farmacologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Neoplasias do Colo/tratamento farmacológico , Cumarínicos/farmacologia , Humanos , Estresse Oxidativo , Psoralea/química , Espécies Reativas de Oxigênio/metabolismo
8.
Cell Biol Int ; 45(7): 1393-1403, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33595160

RESUMO

Farnesyl pyrophosphate synthase (FPPS)-catalyzed isoprenoid intermediates are involved in diabetic cardiomyopathy. This study investigated the specific role of FPPS in the development of diabetic cardiomyopathy. We demonstrated that FPPS expression was elevated in both in vivo and in vitro models of diabetic cardiomyopathy. FPPS inhibition decreased the expression of proteins related to cardiac fibrosis and cardiomyocytic hypertrophy, including collagen I, collagen III, connective tissue growth factor, natriuretic factor, brain natriuretic peptide, and ß-myosin heavy chain. Furthermore, FPPS inhibition and knockdown prevented phosphorylated c-Jun N-terminal kinase 1/2 (JNK1/2) activation in vitro. In addition, a JNK1/2 inhibitor downregulated high-glucose-induced responses to diabetic cardiomyopathy. Finally, immunofluorescence revealed that cardiomyocytic size was elevated by high glucose and was decreased by zoledronate, small-interfering farnesyl pyrophosphate synthase (siFPPS), and a JNK1/2 inhibitor. Taken together, our findings indicate that FPPS and JNK1/2 may be part of a signaling pathway that plays an important role in diabetic cardiomyopathy.


Assuntos
Cardiomiopatias Diabéticas/enzimologia , Geraniltranstransferase/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Animais , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos , Ratos , Ratos Sprague-Dawley
9.
J Cell Mol Med ; 24(22): 13383-13396, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33063955

RESUMO

This study aims to determine the efficacy of Zinc finger protein ZBTB20 in treatment of post-infarction cardiac remodelling. For this purpose, left anterior descending (LAD) ligation was operated on mice to induce myocardial infarction (MI) with sham control group as contrast and adeno-associated virus (AAV9) system was used to deliver ZBTB20 to mouse heart by myocardial injection with vehicle-injected control group as contrast two weeks before MI surgery. Then four weeks after MI, vehicle-treated mice with left ventricular (LV) remodelling underwent deterioration of cardiac function, with symptoms of hypertrophy, interstitial fibrosis, inflammation and apoptosis. The vehicle-injected mice also showed increase of infarct size and decrease of survival rate. Meanwhile, the ZBTB20-overexpressed mice displayed improvement after MI. Moreover, the anti-apoptosis effect of ZBTB20 was further confirmed in H9c2 cells subjected to hypoxia in vitro. Further study suggested that ZBTB20 exerts cardioprotection by inhibiting tumour necrosis factor α/apoptosis signal-regulating kinase 1 (ASK1)/c-Jun N-terminal kinase 1/2 (JNK1/2) signalling, which was confirmed by shRNA-JNK adenoviruses transfection or a JNK activator in vitro as well as ASK1 overexpression in vivo. In summary, our data suggest that ZBTB20 could alleviate cardiac remodelling post-MI. Thus, administration of ZBTB20 can be considered as a promising treatment strategy for heart failure post-MI. Significance Statement: ZBTB20 could alleviate cardiac remodelling post-MI via inhibition of ASK1/JNK1/2 signalling.


Assuntos
Regulação da Expressão Gênica , Infarto do Miocárdio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fatores de Transcrição/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Células Cultivadas , Vasos Coronários/cirurgia , Dependovirus/metabolismo , Fibroblastos/metabolismo , Insuficiência Cardíaca/metabolismo , Hipóxia , MAP Quinase Quinase Quinase 5/metabolismo , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Remodelação Ventricular/efeitos dos fármacos
10.
J Neurochem ; 153(4): 525-538, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31729763

RESUMO

Following peripheral nerve injury, remnant Schwann cells adopt a migratory phenotype and remodel the extracellular matrix allowing axonal regrowth. Although much evidence has demonstrated that TGF-ß1 promotes glioma cell motility and induces the expression of extracellular matrix proteins, the effects of TGF-ß1 on Schwann cell migration has not yet been studied. We therefore investigated the cellular effects and the signal transduction pathways evoked by TGF-ß1 in rattus norvegicus neuronal Schwann RSC96 cell. TGF-ß1 significantly increased migration and invasion of Schwann cells assessed by the wound-healing assay and by cell invasion assay. TGF-ß1-enhanced migration/invasion was blocked by inhibition of MMP-2 and MMP-9. Consistently, by real-time and western blot analyses, we demonstrated that TGF-ß1 increased MMP-2 and MMP-9 mRNA and protein levels. TGF-ß1 also increased MMPs activities in cell growth medium, as shown by gelatin zymography. The selective TGF-ß Type I receptor inhibitor SB431542 completely abrogated any effects by TGF-ß1. Indeed, TGF-ß1 Type I receptor activation provoked the cytosol-to-nucleus translocation of SMAD2 and SMAD3. SMAD2 knockdown by siRNA blocked MMP-2 induction and cell migration/invasion due to TGF-ß1. TGF-ß1 also provoked phosphorylation of MAPKs extracellular regulated kinase 1/2 and JNK1/2. Both MAPKs were upstream to p65/NF-kB inasmuch as both MAPKs' inhibitors PD98059 and SP600125 or their down-regulation by siRNA significantly blocked the TGF-ß1-induced nuclear translocation of p65/NF-kB. In addition, p65/NF-κB siRNA knockdown inhibited the effects of TGF-ß1 on both MMP-9 and cell migration/invasion. We conclude that TGF-ß1 controls RSC96 Schwann cell migration and invasion through MMP-2 and MMP-9 activities. MMP-2 is controlled by SMAD2 whilst MMP-9 is controlled via an ERK1/2-JNK1/2-NF-κB dependent pathway.


Assuntos
Movimento Celular/fisiologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Células de Schwann/enzimologia , Fator de Crescimento Transformador beta1/farmacologia , Animais , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ratos , Células de Schwann/efeitos dos fármacos
11.
Microb Pathog ; 139: 103891, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31783123

RESUMO

Previous study have shown that Talaromyces marneffei (T. marneffei) induced activation of autophagy. Therefore, we explore signaling pathway that regulates activation of autophagy by intracellular signaling mechanisms during T. marneffei infection. Further, we examine c-Jun N-terminal kinase 1 and 2 (JNK1/2) and p38 signaling pathways that regulate IL-1ß and IL-10 production and activation of autophagy during T. marneffei infection in human dendritic cells (DCs). We found that T. marneffei induced activation of JNK1/2 and p38 in human DCs. Furthermore, the inhibition of JNK1/2 and p38 increased activation of autophagy and decreased the replication of T. marneffei in T. marneffei-infected human DCs. Moreover, IL-1ß secretion in T. marneffei-infected human DCs was dependent on JNK1/2 and autophagy pathways, whereas IL-10 secretion was dependent on JNK1/2, p38 and autophagy pathways. These data suggest that JNK1/2 and p38 pathways play critical roles in activation of autophagy, the multiplication of T. marneffei and subsequent cytokine production during T. marneffei infection.


Assuntos
Autofagia , Células Dendríticas/metabolismo , Interleucina-10/biossíntese , Sistema de Sinalização das MAP Quinases , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Micoses/metabolismo , Micoses/microbiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Citocinas/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Interleucina-1beta/biossíntese , Talaromyces
12.
Microb Pathog ; 147: 104270, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32446872

RESUMO

BACKGROUND: The Mycoplasma pneumoniae (M.pneumoniae) was accounted to 3-10% of total pneumonia incidences. In recent decades, metallic nanoparticles were extensively examined as nano-antibiotics. OBJECTIVE: In this investigation, we intended to inspect the therapeutic potential of Zinc oxide nanoparticles (ZnONPs) from (Corydalis yanhusuo) C. yanhusuo against the mycoplasma infected pneumonia in mice. METHODOLOGY: The ZnONPs were formulated via green route technique and characterized by UV-vis spectroscopy, transmission electron microscopy, Fourier transform infrared technique, and atomic force microscopy. The antimicrobial activity of formulated ZnONPs was tested by well diffusion method. The total protein, interleukin-1 (IL-1), interleukin-6 (IL-6), interleukin-8 (IL-8), tumor necrosis factor alpha (TNF-α) and transforming growth factor (TGF) status in the BALF of M. pneumonia infected animals were investigated via kit method. The expressions of ERK1/2, JNK1/2, and NF-κB were examined through the Western blotting. The Histopathological analysis of lung tissues of experimental animals was done. RESULTS: The UV-vis spectroscopy and TEM examinations were proved the existence of CY-ZnONPs. The formulated CY-ZnONPs were displayed the potential antimicrobial activity. The supplementation of CY-ZnONPs were noticeably diminished the total protein and IL-6, IL-8, and TNF-α levels in the BALF of pneumonia mice. The ERK1/2, JNK1/2, and NF-κB expressions were appreciably diminished in the CY-ZnONPs supplemented mice. It also reduced the inflammatory cells penetration, and exhibited normal tissue arrangements in the lung tissues of pneumonia mice. CONCLUSION: The findings of this investigation were proved that the synthesized CY-ZnONPs has the potential to ameliorate the M. pneumoniae infected pneumonia in investigational mice.


Assuntos
Corydalis , Nanopartículas Metálicas , Nanopartículas , Pneumonia por Mycoplasma , Óxido de Zinco , Animais , Sistema de Sinalização das MAP Quinases , Camundongos , Pneumonia por Mycoplasma/tratamento farmacológico , Transdução de Sinais
13.
Mar Drugs ; 18(12)2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-33271756

RESUMO

Efficacy and mechanism of action of marine alkaloid 3,10-dibromofascaplysin (DBF) were investigated in human prostate cancer (PCa) cells harboring different levels of drug resistance. Anticancer activity was observed across all cell lines examined without signs of cross-resistance to androgen receptor targeting agents (ARTA) or taxane based chemotherapy. Kinome analysis followed by functional investigation identified JNK1/2 to be one of the molecular targets of DBF in 22Rv1 cells. In contrast, no activation of p38 and ERK1/2 MAPKs was observed. Inhibition of the drug-induced JNK1/2 activation or of the basal p38 activity resulted in increased cytotoxicity of DBF, whereas an active ERK1/2 was identified to be important for anticancer activity of the alkaloid. Synergistic effects of DBF were observed in combination with PARP-inhibitor olaparib most likely due to the induction of ROS production by the marine alkaloid. In addition, DBF intensified effects of platinum-based drugs cisplatin and carboplatin, and taxane derivatives docetaxel and cabazitaxel. Finally, DBF inhibited AR-signaling and resensitized AR-V7-positive 22Rv1 prostate cancer cells to enzalutamide, presumably due to AR-V7 down-regulation. These findings propose DBF to be a promising novel drug candidate for the treatment of human PCa regardless of resistance to standard therapy.


Assuntos
Alcaloides/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Oxindóis/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Humanos , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Células PC-3 , Fosforilação , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Espécies Reativas de Oxigênio/metabolismo , Receptores Androgênicos/metabolismo , Transdução de Sinais
14.
Ecotoxicol Environ Saf ; 192: 110266, 2020 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-32058163

RESUMO

Despite the well-known acknowledgement of both the toxicity of cadmium (Cd) and the ameliorative effect of selenium (Se), the mechanism of the protective effect of selenium on cadmium-induced Mouse Leydig (TM3) cell apoptosis remains unknown. In this study, we hypothesized that the reactive oxygen species (ROS)-mediated c-jun N-terminal kinase (JNK) signaling pathway is involved in anti-apoptosis of selenium against cadmium in TM3 cells. We found that exposure to cadmium caused evident cytotoxicity, in which cell viability was inhibited, followed by inducement of apoptosis. Moreover, the level of ROS generation was elevated, leading to the phosphorylation of JNK. In addition, following cadmium exposure, the nuclear transcription factor c-jun was significantly activated, which led to increased expression of downstream gene c-jun, resulting in downstream activation of the apoptosis-related protein Caspase3 and upregulation of Cleaved-PARP, as well as inhibition of the anti-apoptosis protein Bcl-2. However, pretreatment with selenium remarkably suppressed cadmium-induced TM3 cell apoptosis. Furthermore, the level of ROS declined, and the JNK signaling pathway was blocked. Following this, the gene expression of c-jun decreased while Bcl-2 increased, which was consistent with the effects on proteins, that Caspase3 activity and Cleaved-PARP were inhibited while Bcl-2 level was restored. In order to explain the relationship between molecules of the signaling pathway, N-acetyl-L-cysteine (NAC), the ROS inhibitor, and JNK1/2 siRNA were administered, which further indicated the mediatory role of the ROS/JNK/c-jun signaling pathway in regulating anti-apoptosis of selenium against cadmium-induced TM3 cell apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Cádmio/toxicidade , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Células Intersticiais do Testículo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Selênio/farmacologia , Acetilcisteína/farmacologia , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Intersticiais do Testículo/metabolismo , Células Intersticiais do Testículo/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Fosforilação , Transdução de Sinais/efeitos dos fármacos
15.
Int J Neurosci ; 130(7): 643-652, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31518515

RESUMO

Aim: The medial prefrontal context (mPFC) plays pivotal roles in initiation, development, and maintenance of chronic pain, whereas the underlying molecular mechanisms remain elusive, which invited investigation of potential involvement of MKP1 in mPFC in mice in neuropathic pain, and its cellular and molecular mechanisms.Materials and methods: Neuropathic pain model was established in adult male Kunming mice via chronic constrictive injury (CCI) of the sciatic nerve. Paw withdrawal latency (PWL) was measured at the plantar area by radiant heat test. Stereotaxic microinjection was applied in mice as per the atlas of Mouse Brain in Stereotaxic Coordinates. mRNA levels of MKP1 in mPFC in CCI mice were assessed by RT-PCR; protein expressions of MKP1, p-p38, p-JNK and p-ERK in mPFC in CCI mice were analyzed by Western blotting, and expressions of the c-Fos in mPFC in CCI mice evaluated by immunohistochemistry. Moreover, Lenti-MKP1 particles or BCI treatment was employed to inhibit MKP1 in mPFC contralateral to the injury.Results: MKP1 was activated and persistently upregulated in mPFC neurons in CCI mice. Inhibition of MKP1 in the mPFC contralateral to the injury could reverse CCI-induced pain behavior and neuronal activity either via employment of Lenti-MKP1 particles or BCI treatment. MKP1 in the mPFC modulated neuropathic pain via dephosphorization of p38 and JNK1/2.Conclusion: The findings demonstrated that MKP1 in mPFC could play a paramount role in the modulation of neuropathic pain, which might be associated to the increased neuronal excitability in the mPFC and downregulated p-p38 and p-JNK expression.


Assuntos
Fosfatase 1 de Especificidade Dupla/metabolismo , Neuralgia/metabolismo , Córtex Pré-Frontal/metabolismo , Transdução de Sinais , Animais , Masculino , Camundongos , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Medição da Dor , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
Cell Mol Neurobiol ; 39(2): 301-319, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30666520

RESUMO

Non-muscle myosin heavy chain IIA (NMMHC IIA), a member of Myosin II family, plays a critical role in various cellular physiological processes. Our previous research had suggested that NMMHC IIA could participate in regulating tight junction morphological changes induced by ischemia stroke. Thus, in the current study, we attempted to uncover the regulation pattern of NMMHC IIA on tight junction dysfunction in oxygen glucose-deprived (OGD) mouse brain bEND.3 endothelial cells. The regulation of NMMHC IIA on tight junction in OGD-stimulated bEND.3 cells was evaluated by western blotting assay. Morphologic change of occludin, claudin-5, and ZO-1 tight junction proteins was compared with pretreatment with NMMHC II inhibitor blebbistatin via immunohistochemical staining. Detection of activation of NMMHC IIA on OGD-mediated tight junction transduction pathway was investigated via Koch's postulate using corresponding protein inhibitor. Our results showed that NMMHC IIA was activated in OGD-stimulated bEND.3 endothelial cells. The inhibition of NMMHC IIA could attenuate the morphologic change of occludin, claudin-5, and ZO-1 tight junction proteins. NMMHC IIA participated in regulating downstream transduction pathway TLR4, phosphatidylinositol 3-kinase (PI3K), Akt, JNK1/2, 14-3-3ε, nuclear factor kappa B (NF-кB) and matrix metalloprotein 9 (MMP9). Blocking of these pathways using indicated inhibitors demonstrated that NMMHC IIA destroyed the connection of tight junction via the activation of TLR4/PI3K/Akt/JNK1/2/14-3-3ε/NF-κB/MMP9 pathway. Our study described the key role of NMMHC IIA in OGD-stimulated mouse brain bEND.3 endothelial cells, while also exhibited the molecule effect on tight junction dysfunction via TLR4/PI3K/Akt/JNK1/2/14-3-3ε/NF-κB/MMP9 signal transduction pathway.


Assuntos
Encéfalo/patologia , Células Endoteliais/metabolismo , Glucose/deficiência , Miosina não Muscular Tipo IIA/metabolismo , Oxigênio/metabolismo , Transdução de Sinais , Junções Íntimas/metabolismo , Proteínas 14-3-3/metabolismo , Animais , Isquemia Encefálica/complicações , Isquemia Encefálica/patologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos Endogâmicos C57BL , Modelos Biológicos , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/patologia , Receptor 4 Toll-Like/metabolismo
17.
Pharmacol Res ; 148: 104400, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31425749

RESUMO

Validation of a small molecular compound targeting the oncogenic pathways is the primary approach for the development of the anti-cancer drugs. In the present study, we employed the computational mimic drug targets prediction software to foresee the molecular targets of a series of spirooxindole-pyrrolizidine derivatives, which were synthesized by our laboratory viatargeted combinational chemistry. We found that CPHSP, a novel spirooxindole-pyrrolizidine derivative, can target the MDM2/p53 signaling that is essential for the tumorigenesis of hepatocellular carcinoma (HCC). To validate its anti-tumoral function, we firstly established the soluble receipt of CPHSP through 2-hydroxypropyl-ß-cyclodextrin (HBC) loading and showed that oral administration of HBC-loaded CPHSP significantly inhibited the tumor growth and prolonged the survival time of tumor-bearing mice in the subcutaneously human hepatoma cells-xenografted nude mouse model of HCC. Immunohistochemistry staining showed that HBC-loaded CPHSP treatment suppressed the proliferation and induced apoptosis of tumor cells in this model. Our mechanistic studies showed that CPHSP treatment inhibited MDM2 protein expression and up-regulated p53 activity and activated MKK4/MKK7/JNK1/2/C-Jun signaling pathway, which resulted in cell cycle arrest and apoptosis of HepG2 cells in vitro. Moreover, we showed that JNK1/2 activation could also up-regulate p53 expression in CPHSP-treated HepG2 cells. Finally, we documented the antitumor activities of oral administration of the HBC-loaded CPHSP in the ML-1 bearing orthotopic mouse model. In summary, this study suggests that oral administration of HBC-loaded CPHSP is a safe and effective treatment for HCC, of which the clinical potency for patients with HCC warrants further studies.


Assuntos
2-Hidroxipropil-beta-Ciclodextrina/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Antineoplásicos , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Células HCT116 , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Herz ; 44(3): 247-256, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-29101624

RESUMO

BACKGROUND: We investigated the effects and underlying mechanisms of genistein on pathological cardiac hypertrophy in vivo and in vitro. MATERIALS AND METHODS: A cardiac hypertrophy model was developed by aortic banding (AB). C57/BL6 mice were randomly assigned to sham + vehicle (CON), sham + genistein (GEN), AB + vehicle (AB), and AB + genistein groups. Genistein (40 mg/kg/day) was administered by gavage for 7 weeks. After assessing the echocardiographic and hemodynamic parameters, mouse hearts were harvested for histopathological and molecular biological analysis. In the in vitro experiments, neonatal rat cardiomyocytes (NRCM) were prepared to test whether genistein could prevent cardiomyocyte hypertrophy induced by phenylephrine (PE). RESULTS: Compared with the CON or GEN group, mice in the AB group exhibited a markedly increased cross-sectional area of cardiomyocytes, collagen volume fraction, and up-regulated expression of atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), beta-myosin heavy chain (ß-MHC) promoter, connective tissue growth factor (CTGF), collagen Iα, collagen III, fibronectin, transforming growth factor (TGF)-ß1, and vimentin mRNA, as well as down-regulated expression of SERCA2α and α­MHC mRNA. Genistein significantly attenuated the expression of these abnormal hypertrophic and fibrotic markers. This anti-hypertrophic effect might be associated with the inhibition of MAPK (ERK1/2, P38, and JNK1/2) and AKT/GSK-3ß signaling. However, in the in vitro experiment, genistein only inhibited hyperphosphorylation of JNK1/2 in the early stage of prohypertrophic stimulation. Genistein could not attenuate PE-induced cardiomyocyte hypertrophy even after pretreatment with sp61005 (a selective inhibitor of JNK1/2). CONCLUSION: Genistein attenuated pressure overload-induced cardiac hypertrophy in vivo and PE induced NRCM hypertrophy in vitro. The underlying mechanisms of genistein activity may be associated with blocking the JNK1/2 signaling pathways.


Assuntos
Cardiomegalia , Genisteína , Inibidores de Proteínas Quinases , Animais , Cardiomegalia/tratamento farmacológico , Genisteína/farmacologia , Glicogênio Sintase Quinase 3 beta , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Distribuição Aleatória
19.
Int J Mol Sci ; 20(22)2019 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-31717348

RESUMO

Auraptene is the most abundant coumarin derivative from plants. The pharmacological value of this compound has been well demonstrated, especially in the prevention of cancer and neurodegenerative diseases. Platelet activation is a major factor contributing to arterial thrombosis. Thus, this study evaluated the influence of auraptene in platelet aggregation and thrombotic formation. Auraptene inhibited platelet aggregation in human platelets stimulated with collagen only. However, auraptene was not effective in inhibiting platelet aggregation stimulated with thrombin, arachidonic acid, and U46619. Auraptene also repressed ATP release, [Ca2+]i mobilization, and P-selectin expression. Moreover, it markedly blocked PAC-1 binding to integrin αIIbß3. However, it had no influence on properties related to integrin αIIbß3-mediated outside-in signaling, such as the adhesion number, spreading area of platelets, and fibrin clot retraction. Auraptene inhibited the phosphorylation of Lyn-Fyn-Syk, phospholipase Cγ2 (PLCγ2), protein kinase C (PKC), Akt, and mitogen-activated protein kinases (MAPKs; extracellular-signal-regulated kinase (ERK1/2), and c-Jun N-terminal kinase (JNK1/2), but not p38 MAPK). Neither SQ22536, an adenylate cyclase inhibitor, nor ODQ, a guanylate cyclase inhibitor, reversed the auraptene-mediated inhibition of platelet aggregation. Auraptene reduced mortality caused by adenosine diphosphate (ADP)-induced pulmonary thromboembolism. In conclusion, this study provides definite evidence that auraptene signifies a potential therapeutic agent for preventing thromboembolic disorders.


Assuntos
Cumarínicos/uso terapêutico , Ativação Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Embolia Pulmonar/tratamento farmacológico , Embolia Pulmonar/mortalidade , Transdução de Sinais , Trifosfato de Adenosina/metabolismo , Animais , Plaquetas/efeitos dos fármacos , Cálcio/metabolismo , Cumarínicos/química , Cumarínicos/farmacologia , Humanos , Camundongos , Nucleotídeos Cíclicos/metabolismo , Selectina-P/metabolismo , Fosforilação/efeitos dos fármacos , Embolia Pulmonar/sangue , Transdução de Sinais/efeitos dos fármacos
20.
Molecules ; 24(5)2019 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-30823626

RESUMO

1-O-Hexyl-2,3,5-trimethylhydroquinone (HTHQ) has previously been found to have effective anti-oxidant and anti-lipid-peroxidative activity. We aimed to elucidate whether HTHQ can prevent dopaminergic neuronal cell death by investigating the effect on l-DOPA-induced cytotoxicity in PC12 cells. HTHQ protected from both l-DOPA-induced cell death and superoxide dismutase activity reduction. When assessing the effect of HTHQ on oxidative stress-related signaling pathways, HTHQ inhibited l-DOPA-induced phosphorylation of sustained extracellular signal-regulated kinases (ERK1/2), p38 mitogen-activated protein kinase (MAPK), and c-Jun N-terminal kinase (JNK1/2). HTHQ also normalized l-DOPA-reduced Bcl-2-associated death protein (Bad) phosphorylation and Bcl-2-associated X protein (Bax) expression, promoting cell survival. Taken together, HTHQ exhibits protective effects against l-DOPA-induced cell death through modulation of the ERK1/2-p38MAPK-JNK1/2-Bad-Bax signaling pathway in PC12 cells. These results suggest that HTHQ may show ameliorative effects against oxidative stress-induced dopaminergic neuronal cell death, although further studies in animal models of Parkinson's disease are required to confirm this.


Assuntos
Neurônios Dopaminérgicos/efeitos dos fármacos , Hidroquinonas/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Doença de Parkinson/tratamento farmacológico , Animais , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neurônios Dopaminérgicos/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Levodopa/efeitos adversos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células PC12 , Doença de Parkinson/genética , Doença de Parkinson/patologia , Ratos , Proteína X Associada a bcl-2/genética , Proteína de Morte Celular Associada a bcl/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA