Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
2.
World J Stem Cells ; 15(7): 654-664, 2023 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-37545754

RESUMO

Acute pancreatitis (AP) often leads to a high incidence of cardiac injury, posing significant challenges in the treatment of severe AP and contributing to increased mortality rates. Mesenchymal stem cells (MSCs) release bioactive molecules that participate in various inflammatory diseases. Similarly, extracellular vesicles (EVs) secreted by MSCs have garnered extensive attention due to their comparable anti-inflammatory effects to MSCs and their potential to avoid risks associated with cell transplantation. Recently, the therapeutic potential of MSCs-EVs in various inflammatory diseases, including sepsis and AP, has gained increasing recognition. Although preclinical research on the utilization of MSCs-EVs in AP-induced cardiac injury is limited, several studies have demonstrated the positive effects of MSCs-EVs in regulating inflammation and immunity in sepsis-induced cardiac injury and cardiovascular diseases. Furthermore, clinical studies have been conducted on the therapeutic application of MSCs-EVs for some other diseases, wherein the contents of these EVs could be deliberately modified through prior modulation of MSCs. Consequently, we hypothesize that MSCs-EVs hold promise as a potential therapy for AP-induced cardiac injury. This paper aims to discuss this topic. However, additional research is essential to comprehensively elucidate the underlying mechanisms of MSCs-EVs in treating AP-induced cardiac injury, as well as to ascertain their safety and efficacy.

3.
Dis Markers ; 2022: 3117805, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36092959

RESUMO

Objective: This study was to evaluate the efficacy of antimicrobial step-down therapy versus conventional antimicrobial therapy in the treatment of patients with sepsis. Methods: Between September 2020 and September 2021, 65 patients with sepsis treated in the intensive care unit (ICU) of our hospital were recruited and assigned at a ratio of 1 : 1 to receive either conventional antimicrobial therapy (sulbactam plus cefoperazone) (control group) or antimicrobial step-down therapy (imipenem/cilastatin) (observation group). The results of drug sensitivity tests and clinical effects were evaluated comprehensively after 3-5 d of treatment, downgraded, and upgraded, or maintenance treatment was administered for 10 d. Outcome measures included clinical and laboratory indices and treatment efficacy. Results: Antimicrobial step-down therapy resulted in a significantly higher efficacy and lower levels of white blood cell (WBC) count and C-reactive protein (CRP) versus conventional antimicrobial therapy (P < 0.05). The patients given antimicrobial step-down therapy showed a significantly shorter duration of antimicrobial drug administration, temperature recovery, time of respiratory support, and ICU stays versus conventional antimicrobial therapy (P < 0.05). Conclusion: Antimicrobial step-down therapy contributes to the mitigation of inflammatory responses in patients with sepsis and shortens the duration of antimicrobial drug use and ICU stay versus conventional antimicrobial therapy. The reliability of the conclusions can be further increased if multicenter and large sample clinical observations can be conducted, which is the direction of endeavor for future clinical studies.


Assuntos
Anti-Infecciosos , Sepse , Antibacterianos/uso terapêutico , Anti-Infecciosos/uso terapêutico , Humanos , Unidades de Terapia Intensiva , Reprodutibilidade dos Testes , Sepse/tratamento farmacológico
4.
J Cell Mol Med ; 26(13): 3648-3658, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35678255

RESUMO

Myocardial injury is a frequently occurring complication of sepsis. This study aims to investigate the molecular mechanism of long noncoding RNA (lncRNA) small nucleolar RNA host gene 1 (SNHG1)-mediated DNA methyltransferase 1/B-cell lymphoma-2 (DNMT1/Bcl-2) axis in sepsis-induced myocardial injury. Mice and HL-1 cells were treated with lipopolysaccharide (LPS) to establish animal and cellular models simulating sepsis and inflammation. LncRNA SNHG1 was screened out as a differentially expressed lncRNA in sepsis samples through microarray profiling, and the upregulated expression of lncRNA SNHG1 was confirmed in myocardial tissues of LPS-induced septic mice and HL-1 cells. Further experiments suggested that silencing of lncRNA SNHG1 reduced the inflammation and apoptotic rate of LPS-induced HL-1 cells. LncRNA SNHG1 inhibited Bcl-2 expression by recruiting DNMT1 to Bcl-2 promoter region to cause methylation. Inhibition of Bcl-2 promoter methylation reduced the inflammation and apoptotic rate of LPS-induced HL-1 cells. In vivo experiments substantiated that lncRNA SNHG1 silencing alleviated sepsis-induced myocardial injury in mice. Taken together, lncRNA SNHG1 promotes LPS-induced myocardial injury in septic mice by downregulating Bcl-2 through DNMT1-mediated Bcl-2 methylation.


Assuntos
DNA (Citosina-5-)-Metiltransferase 1 , MicroRNAs , Proteínas Proto-Oncogênicas c-bcl-2 , RNA Longo não Codificante , Sepse , Animais , Apoptose/fisiologia , Proliferação de Células/fisiologia , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Longo não Codificante/metabolismo , Sepse/genética , Sepse/metabolismo
5.
Immunopharmacol Immunotoxicol ; 44(3): 306-315, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35238277

RESUMO

BACKGROUND: Severe acute pancreatitis (SAP) is a life-threatening disorder associated with multisystem organ failure. This study aimed to investigate the function of high mobility group box 1 (HMGB1) in SAP-induced myocardial injury. METHODS: A rat model with SAP was induced. The pathological changes in rat pancreatic and cardiac tissues were examined by HE staining. Cardiomyocyte apoptosis in rat cardiac tissues, and the serum levels of myocardial injury markers and pro-inflammatory cytokines were examined. Rat primary cardiomyocytes were treated with H2O2 for in vitro experiments. The regulatory molecules of HMGB1 were predicted by bioinformatics analysis. Altered expression of HMGB1, microRNA (miR)-340-5p and CCCTC-binding factor (CTCF) was introduced in rats or cells to investigate their roles in myocardial injury. RESULTS: CTCF and HMGB1 were highly expressed but miR-340-5p was poorly expressed in cardiac tissues of rats with SAP. HMGB1 silencing reduced toll-like receptor 4 (TLR4) expression to promote proliferation and reduce apoptosis of H2O2-treated cardiomyocytes. miR-340-5p targeted HMGB1 mRNA, while CTCF suppressed miR-340-5p transcription. CTCF upregulation or miR-340-5p downregulation blocked the effects of HMGB1 silencing on cardiomyocytes. In vivo, CTCF silencing alleviated injury in rat pancreatic and cardiac tissues and reduced the expression of creatine kinase-MB (CK-MB), lactic dehydrogenase, interleukin (IL)-1ß, IL-6 and tumor necrosis factor-α (TNF-α) in rat serum. But further overexpression of HMGB1 or inhibition of miR-340-5p aggravated the symptoms in rats. CONCLUSION: This study demonstrated that CTCF reduces transcription of miR-340-5p to promote HMGB1 expression, which activates TLR4 expression and promotes myocardial injury in rats with SAP.


Assuntos
Proteína HMGB1 , MicroRNAs , Pancreatite , Animais , Ratos , Doença Aguda , Apoptose/genética , Fator de Ligação a CCCTC/metabolismo , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Peróxido de Hidrogênio/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Pancreatite/induzido quimicamente , Pancreatite/genética , Pancreatite/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
6.
Exp Ther Med ; 23(2): 140, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35069821

RESUMO

Acute pancreatitis (AP) is a common gastrointestinal disease that affects 1 million individuals worldwide. Inflammation and apoptosis are considered to be important pathogenic mechanisms of AP, and high mobility group box 1 (HMGB1) has been shown to play a particularly important role in the etiology of this disease. MicroRNAs (miRs) are emerging as critical regulators of gene expression and, as such, they represent a promising area of therapeutic target identification and development for a variety of diseases, including AP. Using the online database query (microRNA.org), the current study identified a site in the 3' untranslated region of HMGB1 mRNA that was a viable target for miR-340-5p. The present study aimed to investigate the association between miR-340-5p and HMGB1 expression in pancreatic acinar cells following lipopolysaccharide (LPS) treatment by performing luciferase, western blotting and reverse transcription-quantitative PCR assays. The results suggest that miR-340-5p attenuates the induction of HMGB1 by LPS, thereby inhibiting inflammation and apoptosis via blunted activation of Toll-like receptor 4 and enhanced AKT signaling. Thus, the therapeutic application of miR-340-5p may be a useful strategy in AP via upregulation of HMGB1 and subsequent promotion of inflammation and apoptosis.

7.
Cell Cycle ; 21(2): 172-186, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34882521

RESUMO

Circular RNA hsa_circ_0073748 (circ_0073748) is upregulated in patients with acute pancreatitis (AP), a clinically common sudden inflammatory response. MicroRNA (miR)-132-3p is a stress-induced factor with high conservation between species. Herein, expression and role of circ_0073748 and miR-132-3p in caerulein-induced pancreatitis were studied. Expression levels of circ_0073748, miR-132-3p, TNF receptor associated factor 3 (TRAF3), Bcl-2 and Bcl-2-associated X protein (Bax) were examined by reverse transcription-quantitative PCR and Western blotting. Cell proliferation was measured by MTS and EdU assays. Flow cytometry and assay kits detected apoptosis, inflammatory, and oxidative responses. Western blotting detected nuclear factor (NF)-κB signaling pathway. Circ_0073748 was upregulated and miR-132-3p was downregulated in AP patients' plasma and human pancreatic ductal HPDE6-C7 cells with caerulein induction. Interfering circ_0073748 and reinforcing miR-132-3p improved cell viability, EdU incorporation, and superoxide dismutase (SOD) activity of caerulein-treated HPDE6-C7 cells but suppressed malonaldehyde (MDA), IL-6 and TNF-α levels and apoptosis rate. Moreover, TRAF3 downregulation was allied with circ_0073748 silencing and miR-132-3p overexpression in caerulein-induced HPDE6-C7 cells. Mechanically, circ_0073748 was identified as a sponge for miR-132-3p to modulate TRAF3 expression, thus establishing a competitive endogenous RNA (ceRNA) regulation model. Notably, circ_0073748 blockage could suppress expressions of phosphorylated P65 (p-P65) and p-IκB in caerulein-induced HPDE6-C7 cells by promoting miR-132-3p and inhibiting TRAF3. Silencing circ_0073748 and upregulating miR-132-3p could alleviate caerulein-induced HPDE6-C7 injury and inactivate canonical NF-κB signal by inhibiting TRAF3. Circ_0073748/miR-132-3p/TRAF3 ceRNA pathway might be one underlying mechanism and therapeutic target of caerulein-induced AP.


Assuntos
MicroRNAs , Pancreatite , Doença Aguda , Apoptose/genética , Ceruletídeo/metabolismo , Ceruletídeo/toxicidade , Humanos , MicroRNAs/metabolismo , NF-kappa B/metabolismo , Pancreatite/genética , Pancreatite/metabolismo , Fator 3 Associado a Receptor de TNF/genética , Fator 3 Associado a Receptor de TNF/metabolismo
8.
J Cell Mol Med ; 25(19): 9241-9254, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34448533

RESUMO

Acute pancreatitis (AP) is a serious condition carrying a mortality of 25-40%. Extracellular vesicles (EVs) have reported to exert potential functions in cell-to-cell communication in diseases such as pancreatitis. Thus, we aimed at investigating the mechanisms by which EV-encapsulated metastasis-associated lung adenocarcinoma transcript-1 (MALAT1) might mediate the M1 polarization of macrophages in AP. Expression patterns of MALAT1, microRNA-181a-5p (miR-181a-5p) and high-mobility group box 1 protein (HMGB1) in serum of AP patients were determined. EVs were isolated from serum and pancreatic cells. The binding affinity among miR-181a-5p, MALAT1 and HMGB1 was identified. AP cells were co-cultured with EVs from caerulein-treated MPC-83 cells to determine the levels of M1/2 polarization markers and TLR4, NF-κB and IKBa. Finally, AP mouse models were established to study the effects of EV-encapsulated MALAT1 on the M1 polarization of macrophages in AP in vivo. MALAT1 was transferred into MPC-83 cells via EVs, which promoted M1 polarization of macrophages in AP. MALAT1 competitively bound to miR-181a-5p, which targeted HMGB1. Moreover, MALAT1 activated the TLR4 signalling pathway by regulating HMGB1. EV-encapsulated MALAT1 competitively bound to miR-181a-5p to upregulate the levels of IL-6 and TNF-α by regulating HMGB1 via activation of the TLR4 signalling pathway, thereby inducing M1 polarization of macrophages in AP. In vivo experimental results also confirmed that MALAT1 shuttled by EVs promoted M1 polarization of macrophages in AP via the miR-181a-5p/HMGB1/TLR4 axis. Overall, EV-loaded MALAT1 facilitated M1 polarization of macrophages in AP via miR-181a-5p/HMGB1/TLR4, highlighting a potential target for treating AP.


Assuntos
Vesículas Extracelulares/metabolismo , Proteína HMGB1/genética , Macrófagos/imunologia , Macrófagos/metabolismo , MicroRNAs/genética , Pancreatite/etiologia , RNA Longo não Codificante/metabolismo , Adulto , Idoso , Animais , Apoptose/genética , Linhagem Celular Tumoral , Bases de Dados Genéticas , Modelos Animais de Doenças , Suscetibilidade a Doenças , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Inativação Gênica , Humanos , Ativação de Macrófagos , Masculino , Camundongos , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Pancreatite/metabolismo , Pancreatite/patologia , RNA Longo não Codificante/genética , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo
9.
Arch Med Res ; 52(6): 603-610, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33947580

RESUMO

BACKGROUND AND AIMS: The main cause of sepsis-induced Acute kidney injury (AKI) is acute infection after surgery and subsequent progression. However, the mechanism by which AKI is caused and developed from sepsis are not completely known. Herein, we determined the role of CCAAT/enhancer-binding protein ß (C/EBP ß) in sepsis-induced AKI METHODS: C/EBP ß expression was up or down-regulated in LPS-stimulated human renal tubular epithelial cells in vitro by recombinant adenoviruses or siRNA. Subsequent analyses included the test of TNF-α and IL-6 levels by ELISA, cell cycle assay by flow cytometry. RESULTS: C/EBP ß was aberrantly expressed in renal tubular epithelial HK-2 cells exposed to LPS. C/EBP ß overexpression significantly enhanced, but C/EBP ß silencing obviously decreased the production and secretion of inflammatory cytokines TNF-α and IL-6 induced by LPS stimulus in HK-2 cells. And the cell cycle arrest of HK-2 cells induced by LPS was also enhanced after C/EBP ß overexpression while attenuated after C/EBP ß silencing. Consistent pattern of changes in Cyclin D1 and p21 expression were observed in LPS-stimulated HK-2 cells after C/EBP ß silencing and C/EBP ß overexpression. Additionally, the increased p-NF-κB levels induced by LPS were found to be obviously decreased after C/EBP ß silencing in HK-2 cells. And the enhanced TNF-α and IL-6 secretion as well as cell cycle arrest by C/EBP ß overexpression were blocked by BAY11-7082 inhibitor of NF-κB pathway. CONCLUSIONS: C/EBP ß could mediate the LPS-induced aberrant inflammatory response and cell cycle arrest in tubular epithelial cells by NF-κB pathway.


Assuntos
Lipopolissacarídeos , NF-kappa B , Pontos de Checagem do Ciclo Celular , Células Epiteliais/metabolismo , Humanos , Lipopolissacarídeos/toxicidade , NF-kappa B/metabolismo , Transdução de Sinais
10.
Acta Biochim Biophys Sin (Shanghai) ; 53(6): 719-728, 2021 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-33856026

RESUMO

Acute kidney injury (AKI) is a complex syndrome with an abrupt decrease of kidney function, which is associated with high morbidity and mortality. Sepsis is the common cause of AKI. Mounting evidence has demonstrated that long non-coding RNAs (lncRNAs) play critical roles in the development and progression of sepsis-induced AKI. In this study, we aimed to illustrate the function and mechanism of lncRNA SNHG14 in lipopolysaccharide (LPS)-induced AKI. We found that SNHG14 was highly expressed in the plasma of sepsis patients with AKI. SNHG14 inhibited cell proliferation and autophagy and promoted cell apoptosis and inflammatory cytokine production in LPS-stimulated HK-2 cells. Functionally, SNHG14 acted as a competing endogenous RNA (ceRNA) to negatively regulate miR-495-3p expression in HK-2 cells. Furthermore, we identified that HIPK1 is a direct target of miR-495-3p in HK-2 cells. We also revealed that the SNHG14/miR-495-3p/HIPK1 interaction network regulated HK-2 cell proliferation, apoptosis, autophagy, and inflammatory cytokine production upon LPS stimulation. In addition, we demonstrated that the SNHG14/miR-495-3p/HIPK1 interaction network regulated the production of inflammatory cytokines (TNF-α, IL-6, and IL-1ß) via modulating NF-κB/p65 signaling in LPS-challenged HK-2 cells. In conclusion, our findings suggested a novel therapeutic axis of SNHG14/miR-495-3p/HIPK1 to treat sepsis-induced AKI.


Assuntos
Injúria Renal Aguda/sangue , Injúria Renal Aguda/induzido quimicamente , Células Epiteliais/metabolismo , Lipopolissacarídeos/efeitos adversos , MicroRNAs/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , RNA Longo não Codificante/sangue , Sepse/sangue , Transdução de Sinais/genética , Apoptose/genética , Autofagia/genética , Estudos de Casos e Controles , Linhagem Celular Transformada , Proliferação de Células/genética , Citocinas/biossíntese , Células Epiteliais/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Túbulos Renais/citologia , MicroRNAs/genética , Proteínas Serina-Treonina Quinases/genética , RNA Longo não Codificante/genética , Sepse/complicações , Transfecção
11.
Inflammation ; 44(4): 1565-1580, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33725236

RESUMO

The purpose of the present study is to investigate the role of CREB in cardiomyocytes proliferation in regulation of HDAC2-dependent TLR4/NF-κB pathway in severe acute pancreatitis (SAP)-induced myocardial injury. The SAP rat model was developed by injecting sodium touracholate into SD rats and then infected with lentivirus vectors expressing sh-CREB in the presence/absence of LPS. The pathological alterations of rat pancreatic and cardiac tissues were observed by HE staining. TUNEL assay was used to study apoptosis of cardiomyocytes. Next, the loss- and gain-function assay was conducted in LPS-induced myocardial injury cardiomyocytes to define the roles of CREB, HDAC2, and TLR4 in cardiomyocyte proliferation, apoptosis, inflammation, and myocardial injury in vitro. ChIP assay was used to study the enrichment of CREB bound to HDAC2 promoter. RT-qPCR and Western blot analysis were used to detect the expressions of related mRNA and proteins in the NF-κB pathway, respectively. CREB was found to be overexpressed in both SAP tissues and cells. CREB directly bound to the promoter of HDAC2 and activated its expression. Overexpressed CREB or HDAC2 inhibited proliferation and promoted apoptosis of cardiomyocytes. Suppression of CREB inhibited the HDAC2/TLR4/NF-κB cascade to promote proliferation and inhibit apoptosis of cardiomyocytes. The in vitro results were validated in vivo experiments. Coherently, suppression of CREB can inhibit HDAC2/TLR4/NF-κB cascade to promote cardiomyocyte proliferation, thus ameliorating SAP-induced myocardial injury.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/antagonistas & inibidores , Histona Desacetilase 2/antagonistas & inibidores , Miócitos Cardíacos/metabolismo , NF-kappa B/antagonistas & inibidores , Pancreatite/metabolismo , Receptor 4 Toll-Like/antagonistas & inibidores , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/biossíntese , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Inativação Gênica/fisiologia , Histona Desacetilase 2/biossíntese , Histona Desacetilase 2/genética , Masculino , Miócitos Cardíacos/patologia , NF-kappa B/biossíntese , NF-kappa B/genética , Pancreatite/genética , Pancreatite/prevenção & controle , Gravidade do Paciente , Ratos , Ratos Sprague-Dawley , Receptor 4 Toll-Like/biossíntese , Receptor 4 Toll-Like/genética
12.
Life Sci ; 272: 119189, 2021 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-33571516

RESUMO

AIMS: Acute pancreatitis (AP) is an inflammatory disease of the pancreas that may affect local tissues or remote organ systems, while severe acute pancreatitis (SAP) is a life-threatening disorder associated with multiple organ failure. In this investigation, we set about to determine whether microRNA-29a-3p (miR-29a-3p) carried by mesenchymal stem cell (MSCs)-derived extracellular vesicles (EVs) affects the myocardial injury during SAP. MAIN METHODS: EVs were isolated from MSCs of rat bone marrow by differential centrifugation. An SAP rat model was developed and treated with MSCs-EVs and/or alteration of miR-29a-3p and HMGB1 expression, followed by assessment of the rats' cardiac function and inflammation. Next, cardiomyocytes H9C2 were co-cultured with MSC-EVs and internalization of EVs was evaluated, followed by evaluation of whether EVs could transmit miR-29a-3p cargos into H9C2 cells and affect their biological functions. KEY FINDINGS: EVs derived from MSCs were observed to protect against SAP-induced myocardial injury. In SAP-induced rats, miR-29a-3p was under-expressed in myocardial tissues. In addition, we also confirmed that miR-29a-3p could be transferred into the H9C2 cardiomyocytes by MSC-derived EVs, which downregulated the expression of inflammatory markers and improve cardiac function to attenuate myocardial injury. Furthermore, miR-29a-3p inhibited the expression of HMGB1 to downregulate TLR4 expression and further inactivate the Akt signaling pathway. SIGNIFICANCE: These findings support the cardioprotective action of miR-29a-3p transmitted by MSCs-derived EVs in SAP-induced myocardial injury via downregulation of the HMGB1/TLR4/Akt axis, highlighting a promising target for the EV-based therapy for SAP.


Assuntos
MicroRNAs/genética , Isquemia Miocárdica/genética , Isquemia Miocárdica/prevenção & controle , Animais , Apoptose/genética , China , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Coração/fisiologia , Inflamação/metabolismo , Masculino , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Pancreatite/complicações , Pancreatite/genética , Pancreatite/metabolismo , Substâncias Protetoras/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/genética
13.
Cell Transplant ; 29: 963689720967672, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33172292

RESUMO

Myocardial infarction (MI) is one of the most serious cardiovascular diseases associated with myocardial ischemia/reperfusion (I/R) injury. Glaucocalyxin A (GLA) is a biologically active ent-kauranoid diterpenoid that has been found to ameliorate myocardial I/R injury in mice. However, the mechanism has not been fully investigated. In the present study, we aimed to investigate the effect of GLA on rat cardiomyocytes H9c2 cells exposed to hypoxia/reoxygenation (H/R). The results showed that GLA treatment improved cell viability of H/R-stimulated H9c2 cells. Administration with GLA suppressed the H/R-stimulated reactive oxygen species (ROS) production in H9c2 cells. GLA also elevated the activities of antioxidant enzymes, including superoxide dismutase and glutathione peroxidase in H/R-stimulated H9c2 cells. Moreover, GLA prevented H/R-stimulated cell apoptosis in H9c2 cells, as evidenced by increased bcl-2 expression, decreased bax expression, as well as reduced caspase-3 activity. Furthermore, GLA enhanced the activation of protein kinase B (Akt)/nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathway in H9c2 cells exposed to H/R. Additionally, treatment with LY294002 reserved the protective effects of GLA on H/R-stimulated oxidative injury in H9c2 cells. In conclusion, these findings suggested that GLA protected H9c2 cells from H/R-stimulated oxidative damage, which was mediated by the Akt/Nrf2/HO-1 signaling pathway. Thus, GLA might be a promising therapeutic agent for the prevention and treatment of myocardial I/R.


Assuntos
Diterpenos do Tipo Caurano/farmacologia , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Fator 2 Relacionado a NF-E2/metabolismo , Animais , Western Blotting , Hipóxia Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Heme Oxigenase (Desciclizante)/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
14.
Chem Biol Interact ; 316: 108922, 2020 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-31837296

RESUMO

Homeodomain interacting protein kinase-2 (HIPK2) has emerged as a crucial stress-responsive kinase that plays a critical role in regulating cell survival and apoptosis. However, whether HIPK2 participates in regulating cardiomyocyte survival during myocardial ischemia/reperfusion injury remains unclear. Here, we investigated the regulatory effect of HIPK2 on hypoxia/reoxygenation (H/R)-induced cardiomyocyte injury and its potential underlying molecular mechanism. We found that HIPK2 expression was induced in response to H/R exposure. HIPK2 depletion by small interfering RNA (siRNA)-mediated gene silencing significantly decreased the viability and exacerbated H/R-induced apoptosis and reactive oxygen species (ROS) production in cardiomyocytes. Comparatively, HIPK2 overexpression effectively rescued H/R-impaired viability and repressed H/R-induced apoptosis and ROS production in cardiomyocytes. HIPK2 overexpression significantly increased the nuclear expression of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and enhanced Nrf2-mediated transcriptional activity. Moreover, HIPK2 overexpression significantly increased the transcription of Nrf2/ARE target genes. Additionally, Nrf2 inhibition partially reversed the HIPK2-mediated protective effect. Overall, these results demonstrate that HIPK2 overexpression protects cardiomyocytes from H/R-induced injury by enhancing Nrf2/ARE antioxidant signaling, data that suggest HIPK2 is a potential target for cardioprotection.


Assuntos
Apoptose/efeitos dos fármacos , Hipóxia Celular , Estresse Oxidativo/efeitos dos fármacos , Oxigênio/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Elementos de Resposta Antioxidante/genética , Células Cultivadas , Camundongos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo
15.
J Cell Physiol ; 235(4): 3340-3349, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31566716

RESUMO

Non-small-cell lung cancer (NSCLC) is a kind of lung cancer with high incidence and poor outcomes all over the world. Studies have validated that the upregulation of long noncoding RNA LINC00657 is related to several cancers. Nevertheless, the underlying regulatory mechanism of LINC00657 in NSCLC has not been well elucidated. In the present study, quantitative reverse-transcription polymerase chain reaction (RT-qPCR) revealed that LINC00657 level was apparently elevated in NSCLC cells. Loss-of function assays demonstrated that LINC00657 silence retarded cell proliferation and migration in NSCLC cells. Moreover, the chromatin immunoprecipitation result identified the transcription factor SP1 could bind with LINC00657 promoter, and RT-qPCR proved SP1 positively regulated LINC00657 expression in NSCLC cells. In addition, the mechanistic investigations unveiled that LINC00657 was an endogenous sponge of miR-26b-5p and therefore boosted the expression of copper metabolism MURR1 domain-containing 8 (COMMD8), one of the targets of miR-26b-5p. Besides, miR-26b-5p could negatively regulate LINC00657 or COMMD8 in NSCLC cells. With the application of rescue assays, we uncovered that overexpression of COMMD8 partly mitigated the impairment of LINC00657 repression on NSCLC cell proliferation and migration. Together, our study illustrated that SP1-stimulated LINC00657 promoted NSCLC progression through targeting miR-26b-5p/COMMD8 axis, offering a novel potential therapeutic target for NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , MicroRNAs/genética , Proteínas/genética , RNA Longo não Codificante/genética , Fator de Transcrição Sp1/genética , Células A549 , Apoptose , Carcinoma Pulmonar de Células não Pequenas/patologia , Movimento Celular , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos
16.
Oncotarget ; 8(38): 64143-64156, 2017 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-28969058

RESUMO

Lung adenocarcinoma (LUAD) is the most common histological subtype of non-small cell lung cancer, but novel biomarkers for early diagnosis are lacking. Extensive effort has been exerted to identify miRNA biomarkers in LUAD. Unfortunately, high inter-lab variability and small sample sizes have produced inconsistent conclusions in this field. To resolve the above-mentioned limitations, we performed a comprehensive analysis based on LUAD miRNome profiling studies using the robust rank aggregation (RRA) method. Moreover, miRNA-gene interaction network, pathway enrichment analysis and Kaplan-Meier survival curves were used to investigate the clinical values and biological functions of the identified miRNAs. A total of six common differentially expressed miRNAs (DEMs) were identified in LUAD. An independent cohort further confirmed that four miRNAs (miR-21-5p, miR-210-3p, miR-182-5p and miR-183-5p) were up-regulated and two miRNAs (miR-126-3p and miR-218-5p) were down-regulated in LUAD tissues. Pathway enrichment analysis also suggested that the above-listed six DEMs may affect LUAD progression via the estrogen signaling pathway. Survival analysis based on the TCGA dataset revealed the potential prognostic values of six DEMs in patients with LUAD (P-value<0.01). In conclusion, we identified a panel of six miRNAs from LUAD using miRNome profiling studies. Our results provide evidence for the use of these six DEMs as novel diagnostic and prognostic biomarkers for LUAD patients.

17.
Am J Transl Res ; 8(4): 1730-7, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27186296

RESUMO

OBJECTIVE: The aim of this study is to estimate the role of JAK/STAT3 signaling pathway on apoptosis of lung adenocarcinoma induced by icotinib. METHODS: EGFR mutation was detected in lung adenocarcinoma cell line PC-9 by ARMS assay; The inhibitory rates of cell proliferation of PC-9 cells which were exposed to different concentrations of icotinib (0~100 µMol/L) for different time (24~72 h) respectively were evaluated by MTT assay; Apoptosis of PC-9 cells exposed to different concentrations of icotinib (0, 0.1, 1 and 10 µMol/L) for 48 h were evaluated by TUNEL assay; JAK2, STAT3, Bcl-2, Bax mRNA expressions were evaluated by Real-time PCR assay; The protein levels of P-STAT3 and IL-6 were evaluated by Western-blot assay. RESULTS: Human lung adenocarcinoma cell line PC-9 had an exon 19 deletion mutation in EGFR gene; Followed by treatment of icotinib, the proliferation of PC-9 cells were all inhibited significantly, especially in 48 and 72 h (P<0.01) in all concentrations; The inhibitory rates of cell proliferation in different treating time had statistical significance (P<0.01); Cell apoptosis in different concentrations were increased significantly (P<0.05); Along with the increasing concentrations, gene expression levels of JAK2, STAT3 and Bcl-2 decreased significantly (P<0.05), Bax increased significantly (P<0.05), JAK2/STAT3 ratios increased significantly (P<0.01), and Bcl-2/bax ratios decreased significantly (P<0.01); P-STAT3 and IL-6 protein levels were inhibited significantly in higher concentration. CONCLUSIONS: JAK/STAT3 signaling pathway participates in apoptosis of PC-9 cells induced by icotinib. The most likely mechanism is icotinib inhibited the gene expression levels of JAK2, STAT3 and Bcl-2, so with the P-STAT3 and IL-6 protein levels, and mediated gene Bax overexpression.

18.
Am J Transl Res ; 6(3): 267-80, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24936219

RESUMO

Lung adenocarcinoma, the most common type of lung cancer, has increased in recent years. Prognosis is still poor, and pathogenesis remains unclear. This study aimed to investigate the membrane protein profile differences between lung adenocarcinoma and normal tissue. Manual microdissection was used to isolate the target cells from tumor tissue and normal tissue. iTRAQ labeling combined with 2D-LC-MS/MS yielded a differential expression profile of membrane proteins. Bioinformatic analysis was performed using Gene Ontology, WEGO, PID, and KEGG. S100A14 protein was selectively verified by Western blotting. The relationship of S100A14 expression with clinicopathological features in lung cancer patients was evaluated using immunohistochemistry. As a result, 568 differential proteins were identified; 257 proteins were upregulated and 311 were downregulated. Of these proteins, 48% were found to be membrane bound or membrane associated. These proteins enable the physiological functions of binding, catalysis, molecular transduction, transport, and molecular structure. For these differential proteins, 35 pathways were significantly enriched through the Pathway Interaction Database, whereas 19 pathways were enriched via KEGG. The overexpression and cellular distribution of S100A14 in lung cancer were confirmed. We found that upregulation of S100A14 was associated with well or moderate differentiation. The iTRAQ-coupled 2D-LC-MS/MS technique is a potential method for comparing membrane protein profiles between tumor and normal tissue. Such analysis may also help in identifying novel biomarkers and the mechanisms underlying carcinogenesis.

19.
Int J Clin Exp Med ; 7(4): 1190-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24955208

RESUMO

In our previous study, the upregulation of adipophilin in lung adenocarcinoma were identified compared with normal lung tissues by quantitative proteomics. In this study, our aim was to verify the result from quantitative proteomics, further investigate the relationship between adipophilin expression and clinicopathologic factors of lung cancer patients. The expression levels of adipophilin were examined in 10 pairs of lung adenocarcinoma and normal lung tissues using western blotting and the expression and cellular distribution of adipophilin were determined by IHC in 62 formalin-fixed and paraffin embedded primary lung cancer specimens. Adipophilin expression was significantly higher in lung adenocarcinoma specimens than in normal tissues and lung squamous cell carcinomas (P<0.05). There were no significant difference of adipophilin expression between lung squamous cell carcinomas and normal lung tissues. The expression of adipophilin in lung cancer did not correlate with any clinicopathologic factors such as lymph node metastasis, patients' age, gender, tumor size, grade, and TNM stage. In Conclusion, Adipophilin was upregulated in lung adenocarcinoma, suggesting that adipophilin play an important role in tumorigenesis of lung adenocarcinoma and may serve as a potential marker for lung adenocarcinoma.

20.
Thorac Cancer ; 5(4): 349-53, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26767023

RESUMO

A 49-year-old man was diagnosed with small cell lung cancer in May 2005. Chemotherapy was started with 60 mg/day cisplatin iv drip (from days one to three), 2 mg topotecan (TP) hydrochloride iv drip (from days one to four), and traditional Chinese medicine (TCM) AiDi injection for anti-tumor. After four cycles, he underwent conformal radiotherapy with 56Gy/28 fractions in October 2005. In April 2006, a mass on the right supraclavicular area was found. Therefore, he underwent another course of radiotherapy. The fields included the right supraclavicular area and the radiation dose was 50Gy/25 fractions. After completion of chemoradiotherapy, the patient achieved complete remission. Subsequently, the patient received prophylactic cranial irradiation (PCI). Until April of 2012, he had been followed up regularly. Since the SCLC diagnosis, he had received TCM for seven years. In April 2012, the patient complained of coughing again. Subsequently, the patient was given five cycles of an etoposide carboplatin regimen. A computed tomography (CT) scan was performed for review, which showed no obvious change. The patient underwent a second-line chemotherapy irinotecan cisplatin three times. However, the symptoms and CT of this patient showed no significant improvement. We changed the chemotherapy regimen to TP (topotecan 1.2 mg iv drip, days one to five; carboplatin 100 mg iv drip, days one to five). After two TP regimens, the patient died in his sleep on 3 March 2013. In this case, the standardized sequential chemotherapy and radiotherapy treatment, PCI, TCM, and good compliance may have contributed to the patient's longer survival.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA