Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
J Pharmacol Exp Ther ; 390(2): 240-249, 2024 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-38902033

RESUMO

Sympathetic hyperinnervation is the leading cause of fatal ventricular arrhythmia (VA) after myocardial infarction (MI). Cardiac mast cells cause arrhythmias directly through degranulation. However, the role and mechanism of mast cell degranulation in sympathetic remodeling remain unknown. We investigated the role of oxytocin (OT) in stabilizing cardiac mast cells and improving sympathetic innervation in rats. MI was induced by coronary artery ligation. Western blotting, immunofluorescence, and toluidine staining of mast cells were performed to determine the expression and location of target protein. Mast cells accumulated significantly in peri-infarcted tissues and were present in a degranulated state. They expressed OT receptor (OTR), and OT infusion reduced the number of degranulated cardiac mast cells post-MI. Sympathetic hyperinnervation was attenuated as assessed by immunofluorescence for tyrosine hydroxylase (TH). Seven days post-MI, the arrhythmia score of programmed electrical stimulation was higher in vehicle-treated rats with MI than in rats treated with OT. An in vitro study showed that OT stabilized mast cells via the phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) signaling pathway. Further in vivo studies on OTR-deficient mice showed worsening mast cell degranulation and worsening sympathetic innervation. OT pretreatment inhibited cardiac mast cell degranulation post-MI and prevented sympathetic hyperinnervation, along with mast cell stabilization via the PI3K/Akt pathway. SIGNIFICANCE STATEMENT: This is the first study to elucidate the role and mechanism of oxytocin (OT) in inflammatory-sympathetic communication mediated sympathetic hyperinnervation after myocardial infarction (MI), providing new approaches to prevent fatal arrhythmias.


Assuntos
Degranulação Celular , Mastócitos , Infarto do Miocárdio , Ocitocina , Ratos Sprague-Dawley , Receptores de Ocitocina , Sistema Nervoso Simpático , Animais , Ocitocina/farmacologia , Ocitocina/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/metabolismo , Mastócitos/efeitos dos fármacos , Mastócitos/metabolismo , Ratos , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/metabolismo , Masculino , Degranulação Celular/efeitos dos fármacos , Receptores de Ocitocina/metabolismo , Receptores de Ocitocina/antagonistas & inibidores , Camundongos , Transdução de Sinais/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/etiologia
2.
J Biochem Mol Toxicol ; 38(8): e23780, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39056188

RESUMO

Sodium and potassium channels, especially Nav1.5 and Kir2.1, play key roles in the formation of action potentials in cardiomyocytes. These channels interact with, and are regulated by, synapse-associated protein 97 (SAP97). However, the regulatory role of SAP97 in myocyte remains incompletely understood. Here, we investigate the function of SAP97 phosphorylation in the regulation of Nav1.5 and Kir2.1 channel complexes and the upstream regulation of SAP97. We found that SAP97 is phosphorylated by casein kinase II (CK2) in vitro. In addition, transfection of casein kinase 2 interacting protein-1 (CKIP-1) into cardiomyocytes to drive CK2 from the nucleus to the cytoplasm, increased SAP97 phosphorylation and Nav1.5 and Kir2.1 current activity. These findings demonstrated that CKIP-1 modulates the subcellular translocation of CK2, which regulates Nav1.5 and Kir2.1 channel complex formation and activity in cardiomyocytes.


Assuntos
Caseína Quinase II , Miócitos Cardíacos , Canal de Sódio Disparado por Voltagem NAV1.5 , Canais de Potássio Corretores do Fluxo de Internalização , Miócitos Cardíacos/metabolismo , Caseína Quinase II/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Animais , Ratos , Fosforilação , Transporte Proteico , Humanos , Proteínas de Transporte/metabolismo , Ratos Sprague-Dawley
3.
Biotechnol Appl Biochem ; 71(1): 110-122, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37904285

RESUMO

We investigated if poly-lactic acid (PLA) nanopillar array can trigger the differentiation of human epicardial (ADSCs) (heADSCs) into cardiomyocyte-like cells and explored the effects of these cardiomyocyte-like cells on myocardial infarction (MI) in vivo. PLA nanopillar array (200 nm diameter) and plain PLA film (PLA planar) induced heADSCs were marked with carboxyfluorescein. After 7 days, the expressions of myocardiocyte-specific genes were significantly enhanced in cells seeded on PLA nanopillar array compared with that on PLA planar, especially CACNA1C, KCNH2, and MYL2 genes (p < 0.05). However, the expressions of cardiac troponin T (cTNT), KCNQ1, and KCNA5 were lower than those in PLA planar-induced heADSCs (p < 0.05), whereas GATA4 tended to increase with time. The cells with positively stained α-actinin and cTNT were elevated in heADSCs induced by PLA nanopillar array compared with those induced by PLA planar only (p < 0.05). In vivo experiments showed that cardiac function was improved after injecting PLA-nanopillar array-induced heADSCs into the ischemic heart (p < 0.05, compared with PLA planar + MI group). Furthermore, tyrosine hydroxylase density was significantly lower (p < 0.05). PLA nanopillar array directly drives the differentiation of heADSCs into cardiomyocyte-like cells, and the induced heADSCs exhibit a protective effect on ischemic myocardium by improving cardiac function in MI rats.


Assuntos
Infarto do Miocárdio , Miócitos Cardíacos , Humanos , Ratos , Animais , Poliésteres/metabolismo , Células-Tronco , Ácido Láctico/metabolismo
4.
J Mol Cell Cardiol ; 170: 87-99, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35717715

RESUMO

OBJECTIVE: Sudden cardiac death caused by ventricular arrhythmias (VAs) is the main cause of high mortality in patients with myocardial infarction (MI). Sympathetic neural remodeling caused by inflammation after MI is closely associated with the occurrence of VAs. METTL3, the earliest identified m6A methyltransferase, is critical in mediating inflammatory responses. Our aim was to investigate whether the m6A methyltransferase METTL3 was involved in sympathetic remodeling post-MI and its specific mechanism. METHODS AND RESULTS: A rat MI model was established via left coronary artery ligation. The expression of METTL3, TRAF6, NOX2, and NF-κB increased at 3 days and remained elevated at 7 days after MI, as determined via Western blotting. METTL3 was primarily present in macrophages, as determined via immunofluorescence. Intramyocardial injection of lentivirus carrying METTL3-shRNA inhibited METTL3 expression in vivo. Methylated immunoprecipitation-qPCR determined the METTL3 knockdown inhibited the m6A level of TRAF6 mRNA 3'-UTR. The co-immunoprecipitation experiment proved that METTL3 combines with TRAF6. Western blotting showed that silencing METTL3 inhibited TRAF6 level, NF-κB activation, and ROS production; decreased cytokine release (TNF-α and IL-1ß); and downregulated nerve growth factor expression. Finally, METTL3 knockdown reduced sympathetic remodeling after MI, as determined via immunofluorescence assays of tyrosine hydroxylase and growth-associated protein 43. Programmed electrical stimulation, renal sympathetic nerve activity recording, and haemodynamic measurements showed that METTL3 inhibition decreased sympathetic activity and improved cardiac function. CONCLUSIONS: Downregulation of METTL3 expression attenuated the excessive sympathetic neural remodeling induced by MI, further reducing the incidence of VAs and improving cardiac function. This was partly associated with the inhibition of the TRAF6/NF-κB pathway and ROS production.


Assuntos
Infarto do Miocárdio , NF-kappa B , Animais , Ratos , Arritmias Cardíacas , Metiltransferases/metabolismo , Infarto do Miocárdio/metabolismo , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Remodelação Ventricular
5.
J Cell Mol Med ; 26(10): 2959-2971, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35393774

RESUMO

Sympathetic activation after myocardial infarction (MI) leads to ventricular arrhythmias (VAs), which can result in sudden cardiac death (SCD). The toll-like receptor 4 (TLR4)/myeloid differentiation primary response 88 (MyD88)/nuclear factor-kappa B (NF-kB) axis within the hypothalamic paraventricular nucleus (PVN), a cardiac-neural sympathetic nerve centre, plays an important role in causing VAs. An MI rat model and a PVN-TLR4 knockdown model were constructed. The levels of protein were detected by Western blotting and immunofluorescence, and localizations were visualized by multiple immunofluorescence staining. Central and peripheral sympathetic activation was visualized by immunohistochemistry for c-fos protein, renal sympathetic nerve activity (RSNA) measurement, heart rate variability (HRV) analysis and norepinephrine (NE) level detection in serum and myocardial tissue measured by ELISA. The arrhythmia scores were measured by programmed electrical stimulation (PES), and cardiac function was detected by the pressure-volume loop (P-V loop). The levels of TLR4 and MyD88 and the nuclear translocation of NF-kB within the PVN were increased after MI, while sympathetic activation and arrhythmia scores were increased and cardiac function was decreased. However, inhibition of TLR4 significantly reversed these conditions. PVN-mediated sympathetic activation via the TLR4/MyD88/NF-kB axis ultimately leads to the development of VAs after MI.


Assuntos
Infarto do Miocárdio , Núcleo Hipotalâmico Paraventricular , Animais , Arritmias Cardíacas/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Infarto do Miocárdio/metabolismo , NF-kappa B/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
6.
J Cell Mol Med ; 26(4): 1264-1280, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35040253

RESUMO

Ventricular arrhythmias (VAs) triggers by sympathetic nerve hyperactivity contribute to sudden cardiac death in myocardial infarction (MI) patients. Microglia-mediated inflammation in the paraventricular nucleus (PVN) is involved in sympathetic hyperactivity after MI. N6-methyladenosine (m6 A), the most prevalent mRNA and epigenetic modification, is critical for mediating cell inflammation. We aimed to explore whether METTL3-mediated m6 A modification is involved in microglia-mediated sympathetic hyperactivity after MI in the PVN. MI model was established by left coronary artery ligation. METTL3-mediated m6 A modification was markedly increased in the PVN at 3 days after MI, and METTL3 was primarily located in microglia by immunofluorescence. RNA-seq, MeRIP-seq, MeRIP-qPCR, immunohistochemistry, ELISA, heart rate variability measurements, renal sympathetic nerve activity recording and programmed electrical stimulation confirmed that the elevated toll-like receptor 4 (TLR4) expression by m6 A modification on TLR4 mRNA 3'-UTR region combined with activated NF-κB signalling led to the overwhelming production of pro-inflammatory cytokines IL-1ß and TNF-α in the PVN, thus inducing the sympathetic hyperactivity and increasing the incidence of VAs post-MI. Targeting METTL3 attenuated the inflammatory response and sympathetic hyperactivity and reduced the incidence of VAs post-MI.


Assuntos
Metiltransferases , Infarto do Miocárdio , Animais , Coração , Humanos , Metilação , Metiltransferases/metabolismo , Infarto do Miocárdio/complicações , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos , Sistema Nervoso Simpático/metabolismo
7.
Heart Surg Forum ; 25(5): E698-E708, 2022 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-36317904

RESUMO

OBJECTIVE: To evaluate whether M2 macrophage-derived exosomes protect against MI/R injury and reveal the protective mechanism of exosomes [Kourembanas 2015]. METHODS: I/R model injury was induced by temporary left anterior descending coronary artery occlusion in Sprague-Dawley (SD) rats, macrophages isolated from bone marrow-derived macrophages (BMDMs) were induced to M2 polarization, and H9C2 cells subjected to hypoxia/reperfusion (H/R) were used to establish an in vitro model. I/R-induced rats and H/R-induced H9C2 cells were treated with M2-exos in vivo and in vitro, respectively. Masson staining was performed to observe myocardial fibrosis in rats. Immunohistochemical (IHC) staining of myocardial tissues showed the expression of NLRP3 inflammasome activation and pyrolysis. Exosomes derived from IL-4-treated macrophages (M2-exos) were detected by transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA) and western bolt. Western bolt was performed to determine the protein level, including NLRP3, pro-caspase-1, cleaved caspase-1, pro-IL-1ß, cleaved IL-1ß, gasdermin D (GSDMD), and N-terminus of gasdermin D (GSDMD-N). RESULTS: Activity of NLRP3 inflammasome and existence of pyroptosis in the rats subjected to MI/R were significantly higher than those in the control (P < 0.05). Moreover, we confirmed the accumulation of ROS during I/R injury in cardiomyocytes. M2-exos protected against I/R injury and reduced activity of NLRP3 inflammasome and existence of pyroptosis, accompanied with attenuating oxidative stress. In vitro studies showed similar effects, H9c2 cells co-cultured with M2-exos could attenuated H/R-induced cell injury, while M2-exos suppressed the expression of NLRP3 inflammasome and pyroptosis (P < 0.05).


Assuntos
Exossomos , Traumatismo por Reperfusão Miocárdica , Ratos , Animais , Piroptose , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Inflamassomos/metabolismo , Inflamassomos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Espécies Reativas de Oxigênio/farmacologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Exossomos/metabolismo , Ratos Sprague-Dawley , Reperfusão , Macrófagos/metabolismo
8.
Turk J Med Sci ; 52(5): 1468-1477, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36422496

RESUMO

BACKGROUND: Hypoxia-induced pulmonary arterial hypertension (PAH) is characterized by prostacyclin (PGI2 ) disorder, which manifests in the same manner as in monocrotaline (MCT)-induced PAH. Endogenous PGI2 inhibitor coupling factor 6 (CF6) is involved in MCT-induced PAH. This study aimed to explore the presence or absence of a correlation between hypoxia-induced PAH and CF6. METHODS: This study was conducted between January 2019 and June 2020. A total of 135 male Wistar rats (aged 8 weeks and weighing 200-250 g) were randomly divided into five groups: (A) control, (B) 1 week of hypoxia, (C) 2 weeks of hypoxia, (D) 3 weeks of hypoxia, and (E) 4 weeks of hypoxia. CF6 expression in both lung tissue and blood samples from the lung vasculature and tail vein was measured by western blotting, immunohistochemistry, reverse transcription polymerase chain reaction, and enzyme-linked immunosorbent assay. RESULTS: Hemodynamic and morphological changes in hypoxia-induced rats indicated PAH development. The results showed the presence of a correlation between the mRNA and protein levels of CF6 in lung tissue, activity of mitochondrial ATP synthase, and hypoxia time, and there was a significant increment in the group exposed to hypoxia for 4 weeks compared to the control group. The decrement expression of ATPase inhibitory factor 1 (IF 1) mRNA was consistent with the outcomes of ATP synthase activity in lung tissue in the 4 weeks of hypoxia group compared with the control group. However, the levels of CF6 and ATP synthase activity did not differ between blood samples from the lung vasculature and tail vein. DISCUSSION: : In hypoxia-induced PAH, CF6 showed downregulated expression in lung tissue, but not in pulmonary vasculature and circulation. Therefore, we speculated that CF6 and ATP synthase may play important roles in hypoxia-induced PAH.


Assuntos
Hipertensão Pulmonar , Hipertensão Arterial Pulmonar , Ratos , Masculino , Animais , ATPases Mitocondriais Próton-Translocadoras/efeitos adversos , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Ratos Wistar , Monocrotalina/efeitos adversos , Hipóxia/complicações , Hipóxia/metabolismo , RNA Mensageiro , Trifosfato de Adenosina/efeitos adversos
9.
J Cell Mol Med ; 23(1): 112-125, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30353660

RESUMO

Malignant ventricular arrhythmias (VAs) following myocardial infarction (MI) is a lethal complication resulting from sympathetic nerve hyperactivity. Numerous evidence have shown that inflammation within the paraventricular nucleus (PVN) participates in sympathetic hyperactivity. Our aim was to explore the role of Macrophage-inducible C-type lectin (Mincle) within the PVN in augmenting sympathetic activity following MI,and whether NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome/IL-1ß axis is involved in this activity. MI was induced by coronary artery ligation. Mincle expression localized in microglia within the PVN was markedly increased at 24 hours post-MI together with sympathetic hyperactivity, as indicated by measurement of the renal sympathetic nerve activity (RSNA) and norepinephrine (NE) concentration. Mincle-specific siRNA was administrated locally to the PVN, which consequently decreased microglial activation and sympathetic nerve activity. The MI rats exhibited a higher arrhythmia score after programmed electric stimulation than that treated with Mincle siRNA, suggesting that the inhibition of Mincle attenuated foetal ventricular arrhythmias post-MI. The underlying mechanism of Mincle in sympathetic hyperactivity was investigated in lipopolysaccharide (LPS)-primed naïve rats. Recombinant Sin3A-associated protein 130kD (rSAP130), an endogenous ligand for Mincle, induced high levels of NLRP3 and mature IL-1ß protein. PVN-targeted injection of NLRP3 siRNA or IL-1ß antagonist gevokizumab attenuated sympathetic hyperactivity. Together, the data indicated that the knockdown of Mincle in microglia within the PVN prevents VAs by attenuating sympathetic hyperactivity and ventricular susceptibility, in part by inhibiting its downstream NLRP3/IL-1ß axis following MI. Therapeutic interventions targeting Mincle signalling pathway could constitute a novel approach for preventing infarction injury.


Assuntos
Lectinas Tipo C/metabolismo , Microglia/metabolismo , Infarto do Miocárdio/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores Imunológicos/metabolismo , Sistema Nervoso Simpático/metabolismo , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Coração/fisiopatologia , Interleucina-1beta/antagonistas & inibidores , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lectinas Tipo C/genética , Masculino , Infarto do Miocárdio/genética , Infarto do Miocárdio/fisiopatologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Norepinefrina/metabolismo , Interferência de RNA , Ratos Sprague-Dawley , Receptores Imunológicos/genética , Sistema Nervoso Simpático/fisiopatologia
10.
J Cell Mol Med ; 21(11): 2695-2710, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28470940

RESUMO

Mounting evidence supports the hypothesis that inflammation modulates sympathetic sprouting after myocardial infarction (MI). The myeloid P2X7 signal has been shown to activate the nucleotide-binding and oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, a master regulator of inflammation. We investigated whether P2X7 signal participated in the pathogenesis of sympathetic reinnervation after MI, and whether NLRP3/interleukin-1ß (IL-1ß) axis is involved in the process. We explored the relationship between P2X7 receptor (P2X7 R) and IL-1ß in the heart tissue of lipopolysaccharide (LPS)-primed naive rats. 3'-O-(4-benzoyl) benzoyl adenosine 5'-triphosphate (BzATP), a P2X7 R agonist, induced caspase-1 activation and mature IL-1ß release, which was further neutralized by a NLRP3 inhibitor (16673-34-0). MI was induced by coronary artery ligation. Following infarction, a marked increase in P2X7 R was localized within infiltrated macrophages and observed in parallel with an up-regulation of NLRP3 inflammasome levels and the release of IL-1ß in the left ventricle. The administration of A-740003 (a P2X7 R antagonist) significantly prevented the NLRP3/IL-1ß increase. A-740003 and/or Anakinra (an IL-1 receptor antagonist) significantly reduced macrophage infiltration as well as macrophage-based IL-1ß and NGF (nerve growth factor) production and eventually blunted sympathetic hyperinnervation, as assessed by the immunofluorescence of tyrosine hydroxylase (TH) and growth-associated protein 43 (GAP 43). Moreover, the use of Anakinra partly attenuated sympathetic sprouting. This indicated that the effect of P2X7 on neural remodelling was mediated at least partially by IL-1ß. The arrhythmia score of programmed electric stimulation was in accordance with the degree of sympathetic hyperinnervation. In vitro studies showed that BzATP up-regulated secretion of nerve growth factor (NGF) in M1 macrophages via IL-1ß. Together, these data indicate that P2X7 R contributes to neural and cardiac remodelling, at least partly mediated by NLRP3/IL-1ß axis. Therapeutic interventions targeting P2X7 signal may be a novel approach to ameliorate arrhythmia following MI.


Assuntos
Interleucina-1beta/genética , Infarto do Miocárdio/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Receptores Purinérgicos P2X7/genética , Sistema Nervoso Simpático/efeitos dos fármacos , Acetamidas/farmacologia , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Animais , Caspases/genética , Caspases/metabolismo , Vasos Coronários/metabolismo , Vasos Coronários/patologia , Modelos Animais de Doenças , Estimulação Elétrica , Proteína GAP-43/genética , Proteína GAP-43/metabolismo , Regulação da Expressão Gênica , Inflamassomos/efeitos dos fármacos , Inflamassomos/metabolismo , Inflamação , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Interleucina-1beta/metabolismo , Ligadura , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Fator de Crescimento Neural/genética , Fator de Crescimento Neural/metabolismo , Agonistas do Receptor Purinérgico P2X/farmacologia , Antagonistas do Receptor Purinérgico P2X/farmacologia , Quinolinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2X7/metabolismo , Transdução de Sinais , Sistema Nervoso Simpático/metabolismo
11.
Respir Res ; 18(1): 127, 2017 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-28646872

RESUMO

BACKGROUND: Pulmonary arterial hypertension (PAH) is a devastating disease that lacks sufficient treatment. Studies have shown that the Nod-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome contributes to PAH pathogenesis, but the role of the upstream molecular P2X7 receptor (P2X7R) has remained unexplored. We investigated the role of P2X7R in the pathogenesis of PAH. METHODS AND RESULTS: PH was induced by a single subcutaneous injection of monocrotaline (MCT) (60 mg/kg) on left pneumonectomised Sprague-Dawley rats, as validated by significant increases in pulmonary artery pressure and vessel wall thickness. Marked P2X7R was detected by predominant PA immunostaining in lungs from PH rats. Western blot revealed a significant increase in the protein levels of P2X7R as well as NLRP3 and caspase-1 in the diseased lung tissue compared with normal tissue. The rats received A-740003 (a selective P2X7 receptor antagonist, 30 mg/kg) daily starting from 1 week before or 2 weeks after MCT injection. Consequently, A-740003 reversed the NLRP3 inflammasome upregulation, significantly decreased the mean right ventricular (RV) pressure and RV hypertrophy, and reversed pulmonary arterial remodelling 4 weeks after MCT injection, as both a pretreatment and rescue intervention. Notably, A-740003 significantly reduced macrophage and pro-inflammatory cytokine levels, as measured via bronchoalveolar lavage. The recruitment of macrophages as well as collagen fibre deposition in the perivascular areas were also reduced, as confirmed by histological staining. CONCLUSIONS: P2X7R contributes to the pathogenesis of PH, probably in association with activation of the NLRP3 inflammasome. Blockade of P2X7R might be applied as a novel therapeutic approach for the treatment of PAH.


Assuntos
Progressão da Doença , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Receptores Purinérgicos P2X7/fisiologia , Animais , Relação Dose-Resposta a Droga , Masculino , Antagonistas do Receptor Purinérgico P2X/farmacologia , Ratos , Ratos Sprague-Dawley
12.
Am J Physiol Cell Physiol ; 310(1): C41-53, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26491050

RESUMO

Inflammation-dominated sympathetic sprouting adjacent to the necrotic region following myocardial infarction (MI) has been implicated in the etiology of arrhythmias resulting in sudden cardiac death; however, the mechanisms responsible remain to be elucidated. Although being a key immune mediator, the role of Notch has yet to be explored. We investigated whether Notch regulates macrophage responses to inflammation and affects cardiac sympathetic reinnervation in rats undergoing MI. MI was induced by coronary artery ligation. A high level of Notch intracellular domain was observed in the macrophages that infiltrated the infarct area at 3 days post-MI. The administration of the Notch inhibitor N-N-(3,5-difluorophenacetyl-L-alanyl)-S-phenylglycine-t-butyl ester (DAPT) (intravenously 30 min before MI and then daily until death) decreased the number of macrophages and significantly increased the M2 macrophage activation profile in the early stages and attenuated the expression of nerve growth factor (NGF). Eventually, NGF-induced sympathetic hyperinnervation was blunted, as assessed by the immunofluorescence of tyrosine hydroxylase. At 7 days post-MI, the arrhythmia score of programmed electric stimulation in the vehicle-treated infarcted rats was higher than that in rats treated with DAPT. Further deterioration in cardiac function and decreases in the plasma levels of TNF-α and IL-1ß were also detected. In vitro studies revealed that LPS/IFN-γ upregulated the surface expression of NGF in M1 macrophages in a Notch-dependent manner. We concluded that Notch inhibition during the acute inflammatory response phase is associated with the downregulation of NGF, probably through a macrophage-dependent pathway, thus preventing the process of sympathetic hyperinnervation.


Assuntos
Anti-Inflamatórios/farmacologia , Dipeptídeos/farmacologia , Coração/inervação , Macrófagos/efeitos dos fármacos , Infarto do Miocárdio/tratamento farmacológico , Miocárdio/metabolismo , Receptor Notch1/antagonistas & inibidores , Sistema Nervoso Simpático/efeitos dos fármacos , Animais , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatologia , Arritmias Cardíacas/prevenção & controle , Estimulação Cardíaca Artificial , Células Cultivadas , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Masculino , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/fisiopatologia , Fator de Crescimento Neural/metabolismo , Fenótipo , Ratos Sprague-Dawley , Receptor Notch1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sistema Nervoso Simpático/metabolismo , Sistema Nervoso Simpático/fisiopatologia , Fator de Necrose Tumoral alfa/metabolismo
13.
Respir Res ; 17(1): 99, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27491388

RESUMO

BACKGROUND: Pulmonary arterial hypertension (PAH) is a progressive and life-threatening disease associated with high morbidity and mortality rates. However, the exact regulatory mechanism of PAH is unknown. Although coupling factor 6 (CF6) is known to function as a repressor, its role in PAH has not been explored. Here, we investigated the involvement of endogenous CF6 in the development of PAH. METHODS: PAH was induced with monocrotaline (MCT), as demonstrated by significant increases in pulmonary artery pressure and vessel wall thickness. The adeno-associated virus (AAV) carrying CF6 short hairpin RNA (shRNA) or control vector (2×10(10) gp) was intratracheally transfected into the lungs of rats 2 weeks before or after MCT injection. RESULTS: A 2-6-fold increase in CF6 was observed in the lungs and circulation of the MCT-injected rats as confirmed by qRT-PCR and ELISA. Immunohistochemistry analysis revealed a small quantity of CF6 localized to endothelial cells (ECs) under physiological conditions spread to surrounding tissues in a paracrine manner in PAH lungs. Notably, CF6 shRNA effectively inhibited CF6 expression, abolished lung macrophage infiltration, reversed endothelial dysfunction and vascular remodeling, and ameliorated the severity of pulmonary hypertension and right ventricular dysfunction at 4 weeks both as a pretreatment and rescue intervention. In addition, the circulating and lung levels of 6-keto-PGF1a, a stable metabolite of prostacyclin, were reversed by CF6 inhibition, suggesting that the effect of CF6 inhibition may partly be mediated through prostacyclin. CONCLUSIONS: CF6 contributes to the pathogenesis of PAH, probably in association with downregulation of prostacyclin. The blockage of CF6 might be applied as a novel therapeutic approach for PAH and PA remodeling.


Assuntos
Terapia Genética/métodos , Hipertensão Pulmonar/terapia , Pulmão/metabolismo , ATPases Mitocondriais Próton-Translocadoras/antagonistas & inibidores , ATPases Mitocondriais Próton-Translocadoras/genética , Fatores Acopladores da Fosforilação Oxidativa/antagonistas & inibidores , Fatores Acopladores da Fosforilação Oxidativa/genética , Interferência de RNA , 6-Cetoprostaglandina F1 alfa/metabolismo , Animais , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Hipertensão Pulmonar/induzido quimicamente , Injeções Espinhais , Pulmão/patologia , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Monocrotalina , Infiltração de Neutrófilos , Fatores Acopladores da Fosforilação Oxidativa/metabolismo , Artéria Pulmonar/patologia , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Remodelação Vascular , Disfunção Ventricular Direita/etiologia , Disfunção Ventricular Direita/genética , Disfunção Ventricular Direita/prevenção & controle
14.
J Cardiovasc Pharmacol ; 67(3): 252-9, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26566151

RESUMO

BACKGROUND: Myocardial infarction (MI) results in decreased inward-rectifier K⁺ current (IK1), which is mediated primarily by the Kir2.1 protein and is accompanied by upregulated T cells. Interferon γ (IFN-γ), secreted predominantly by Th1 cells, causes a decrease in IK1 in microglia. Whether Th1 cells can induce IK1/Kir2.1 remodeling following MI and whether valsartan can ameliorate this phenomenon remain unclear. METHODS: Rats experiencing MI received either valsartan or saline for 7 days. Th1-enriched lymphocytes and myocytes were cocultured with or without valsartan treatment. Th1 cells were monitored by flow cytometry. The protein levels of Kir2.1 were detected by Western blot analyses. IK1 was recorded through whole-cell patch clamping. The plasma levels of IFN-γ, interleukin 2, and tumor necrosis factor α were detected by enzyme-linked immunosorbent assay. RESULTS: Th1 cell number and cytokine expression levels were higher following MI, and the Kir2.1 protein level was decreased. In MI rats, valsartan reduced Th1 cell number and cytokine expression levels and increased the Kir2.1 expression and the IK1 current compared with the rats that received saline treatment; these results are consistent with the effect of valsartan in cocultured lymphocytes and myocytes. In vitro, IFN-γ overexpression suppressed the IK1 current, whereas interleukin 2 and tumor necrosis factor α had no significant effect on the current, establishing that Th1 cell regulation of IK1/Kir2.1 expression is mainly dependent on IFN-γ. CONCLUSIONS: Valsartan ameliorates IK1/Kir2.1 remodeling by downregulating the Th1 immune response following MI.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Infarto do Miocárdio/tratamento farmacológico , Miócitos Cardíacos/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização/efeitos dos fármacos , Células Th1/efeitos dos fármacos , Valsartana/farmacologia , Animais , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Regulação para Baixo , Hemodinâmica/efeitos dos fármacos , Interferon gama/sangue , Interleucina-2/sangue , Masculino , Potenciais da Membrana , Infarto do Miocárdio/sangue , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/metabolismo , Potássio/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Ratos Wistar , Células Th1/imunologia , Células Th1/metabolismo , Fatores de Tempo , Fator de Necrose Tumoral alfa/sangue
15.
BMC Cardiovasc Disord ; 16: 16, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26787044

RESUMO

BACKGROUND: To investigate the effects of semaphorin 3A (sema 3A) on cardiac autonomic regulation and subsequent ventricular arrhythmias (VAs) in post-infarcted hearts. METHOD AND RESULTS: In order to explore the functions of sema 3A in post-infarcted hearts, lentivirus-Sema 3A-shRNA and negative control vectors were delivered to the peri-infarcted myocardium rats respectively. Meanwhile, recombinant sema 3A and control (0.9% NaCl solution) were injected intravenously into infarcted rats to test the therapeutic potential of sema 3A. Results indicated that levels of sema 3A were higher in post-infarcted hearts compared with sham rats. However, sema 3A silencing leaded to sympathetic hyperinnervation, increased myocardial norepinephrine (NE) content and inducible VAs. Conversely, the intravenous administration of sema 3A to infarcted rats reduced sympathetic nerve sprouting, improved cardiac autonomic regulation, and decreased the incidence of inducible VAs. However, both infarct size and cardiac function were similar among infarcted hearts. CONCLUSIONS: The upregulation and administration of sema 3A exerted beneficial effects on infarction-induced cardiac autonomic disorders by increasing cardiac electrical stability and reducing VAs. Sema 3A might be a potential therapeutic agent for cardiac autonomic abnormalities induced arrhythmias.


Assuntos
Coração/efeitos dos fármacos , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , Semaforina-3A/genética , Sistema Nervoso Simpático/metabolismo , Taquicardia Ventricular/genética , Fibrilação Ventricular/genética , Animais , Sistema Nervoso Autônomo/efeitos dos fármacos , Sistema Nervoso Autônomo/metabolismo , Sistema Nervoso Autônomo/fisiopatologia , Western Blotting , Cromatografia Líquida de Alta Pressão , Eletrocardiografia , Técnicas de Silenciamento de Genes , Coração/inervação , Coração/fisiopatologia , Imuno-Histoquímica , Masculino , Infarto do Miocárdio/metabolismo , Norepinefrina/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Recombinantes/farmacologia , Semaforina-3A/farmacologia , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/fisiopatologia , Taquicardia Ventricular/fisiopatologia , Fibrilação Ventricular/fisiopatologia
16.
J Appl Toxicol ; 36(2): 257-65, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26011702

RESUMO

The kidney is the principal organ targeted by exposure to cadmium (Cd), a well-known toxic metal. Even at a low level, Cd damages glomerular filtration. However, little is known about the effects of Cd on the glomerular endothelium, which performs the filtration function and directly interacts with Cd in blood plasma. In this study, we cultured human renal glomerular endothelial cells (HRGECs) in the presence of serum with treatment of a short term (1 h) and low concentration (1 µm) of Cd, which mimics the pattern of glomerular endothelium exposure to Cd in vivo. We found that this short-term, low-dose Cd exposure does not induce cytotoxicity, but increases permeability in HRGECs monolayers and redistributes adherens junction proteins vascular endothelial-cadherin and ß-catenin. Though short-term, low-dose Cd exposure activates all three major mitogen activated protein kinases, only the inhibitor of p38 mitogen activated protein kinase partially prevents Cd-induced hyperpermeability in HRGECs. Our data indicate that the presence of Cd in blood circulation might directly disrupt the glomerular endothelial cell barrier and contribute to the development of clinical symptoms of glomerular diseases.


Assuntos
Cádmio/toxicidade , Células Cultivadas/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Taxa de Filtração Glomerular/efeitos dos fármacos , Rim/efeitos dos fármacos , Permeabilidade/efeitos dos fármacos , Cádmio/sangue , Crescimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Humanos
17.
Cardiovasc Drugs Ther ; 29(3): 209-18, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26095682

RESUMO

PURPOSE: Myocardial infarction (MI) results in an increased susceptibility to ventricular arrhythmias, due in part to decreased inward-rectifier K+ current (IK1), which is mediated primarily by the Kir2.1 protein. The use of renin-angiotensin-aldosterone system antagonists is associated with a reduced incidence of ventricular arrhythmias. Casein kinase 2 (CK2) binds and phosphorylates SP1, a transcription factor of KCNJ2 that encodes Kir2.1. Whether valsartan represses CK2 activation to ameliorate IK1 remodeling following MI remains unclear. METHODS: Wistar rats suffering from MI received either valsartan or saline for 7 days. The protein levels of CK2 and Kir2.1 were each detected via a Western blot analysis. The mRNA levels of CK2 and Kir2.1 were each examined via quantitative real-time PCR. RESULTS: CK2 expression was higher at the infarct border; and was accompanied by a depressed IK1/Kir2.1 protein level. Additionally, CK2 overexpression suppressed KCNJ2/Kir2.1 expression. By contrast, CK2 inhibition enhanced KCNJ2/Kir2.1 expression, establishing that CK2 regulates KCNJ2 expression. Among the rats suffering from MI, valsartan reduced CK2 expression and increased Kir2.1 expression compared with the rats that received saline treatment. In vitro, hypoxia increased CK2 expression and valsartan inhibited CK2 expression. The over-expression of CK2 in cells treated with valsartan abrogated its beneficial effect on KCNJ2/Kir2.1. CONCLUSIONS: AT1 receptor antagonist valsartan reduces CK2 activation, increases Kir2.1 expression and thereby ameliorates IK1 remodeling after MI in the rat model.


Assuntos
Caseína Quinase II/metabolismo , Infarto do Miocárdio/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/biossíntese , Regulação para Cima/efeitos dos fármacos , Valsartana/farmacologia , Animais , Masculino , Potenciais da Membrana/efeitos dos fármacos , Infarto do Miocárdio/enzimologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Cultura Primária de Células , Ratos
18.
Med Sci Monit ; 21: 3941-7, 2015 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-26675565

RESUMO

BACKGROUND This study aimed to decrease leukocytes counts by hydroxyurea (Hu) in an acute myocardial infarction (AMI) rat model and examine its effect on the inflammatory response of myocardial infarction and cardiac functions. MATERIAL AND METHODS AMI was successfully caused in 36 rats, and 12 control rats received sham operation. Rats in the AMI group were then randomly divided into Hu and vehicle group with 18 rats each. Rats in the Hu AMI group received Hu (200 mg/kg) intragastrically while vehicle AMI group received saline. Leukocytes counts, cardiac functions, myocardial tissue morphology, and levels of soluble intercellular adhesion molecule-1 (sICAM), P-selectin and platelet activating factor (PAF) were measured and compared among the three groups four weeks after AMI induction. RESULTS Leukocytes, neutrophils, and leukomonocyte counts in vehicle AMI rats were significantly higher than that of the normal control group (p<0.05). However, Hu treatment decreased their counts significantly (p<0.05). sICAM, P-selectin, and PAF level in vehicle AMI group were significantly higher than those of the normal group, and their level was also decreased by Hu treatment (p<0.05). Echocardiography analysis showed that Hu treatment increased left ventricular ejection fraction (LVEF) and left ventricular fractional shortening (LVFS) compared to that of vehicle AMI group (p<0.05). Histopathological examination showed that Hu significantly reduced the swelling of the heart muscle fiber in necrotic foci and the number of inflammatory cells infiltrated into myocardial interstitium compared to vehicle AMI group. CONCLUSIONS Decrease leukocytes counts by Hu significantly reduced inflammatory reaction and improved cardiac functions in AMI rats.


Assuntos
Testes de Função Cardíaca/efeitos dos fármacos , Hidroxiureia/farmacologia , Contagem de Leucócitos , Infarto do Miocárdio/fisiopatologia , Animais , Ecocardiografia , Eletrocardiografia , Masculino , Infarto do Miocárdio/diagnóstico por imagem , Ratos , Ratos Wistar
19.
Biochem Biophys Res Commun ; 450(1): 447-52, 2014 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-24909688

RESUMO

The vasculature of various organs is a targeted by the environmental toxin, cadmium (Cd). However, mechanisms leading to pathological conditions are poorly understood. In the present study, we examined the effect of cadmium chloride (CdCl2) on human umbilical vein endothelial cells (HUVECs). At 4 µM, CdCl2 induced a hyper-permeability defect in HUVECs, but not the inhibition of cell growth up to 24h. This effect of CdCl2 was dependent on the activation of the p38 mitogen-activated protein kinase (MAPK) pathway. The p38 MAPK inhibitor SB203850 suppressed the CdCl2-induced alteration in trans-endothelial electrical resistance in HUVEC monolayers, a model measurement of vascular endothelial barrier integrity. SB203850 also inhibited the Cd-induced membrane dissociation of vascular endothelial (VE) cadherin and ß-catenin, the important components of the adherens junctional complex. In addition, SB203850 reduces the Cd-induced expression and secretion of tumor necrosis factor α (TNF-α). Taken together, our findings suggest that Cd induces vascular hyper-permeability and disruption of endothelial barrier integrity through stimulation of p38 MAPK signaling.


Assuntos
Cádmio/toxicidade , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/fisiologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Células Endoteliais/patologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos
20.
Cardiology ; 127(3): 155-63, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24356397

RESUMO

OBJECTIVES: Diabetic cardiac autonomic neuropathy can lead to an increased incidence of ventricular arrhythmias (VAs). However, few data are available regarding the pathogenesis and therapy of the VAs accompanying diabetic cardiac autonomic neuropathy. We aimed to explore whether or not exogenous nerve growth factor (NGF) can reduce the sympathetic heterogeneity and the incidence of VAs in diabetes mellitus (DM). METHODS: Male Wistar rats were randomly divided into 3 groups: controls, rats with DM with saline infused into the left stellate ganglion (LSG), i.e. the DS group and rats with DM with NGF infused into the LSG, i.e. the DN group. After 28 weeks, all rats were subjected to electrophysiological experiments. Sympathetic innervations and NGF were studied by immunostaining, RT-PCR or Western blot analysis. RESULTS: The incidence of inducible VAs was significantly higher in the DS group than in the control group, but was markedly decreased in the DN group. In the DS group, the tyrosine hydroxylase (TH) and NGF expression were significantly lower than in the other groups, and significant proximal-distal heterogeneities existed regarding the TH and NGF expression in the left ventricle, but were markedly repaired in the DN group. CONCLUSIONS: NGF intervention in the LSG can reduce the heterogeneity of cardiac sympathetic innervations and the incidence of VAs in diabetic rats.


Assuntos
Antiarrítmicos/farmacologia , Arritmias Cardíacas/tratamento farmacológico , Diabetes Mellitus Experimental/tratamento farmacológico , Neuropatias Diabéticas/tratamento farmacológico , Fator de Crescimento Neural/farmacologia , Animais , Biomarcadores/metabolismo , Estimulação Elétrica , Masculino , RNA Mensageiro/metabolismo , Distribuição Aleatória , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA