Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Cardiovasc Pharmacol ; (0)2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38113918

RESUMO

ABSTRACT: Studies have examined the therapeutic effect of levosimendan on cardiovascular diseases such as heart failure, perioperative cardiac surgery, and septic shock, but the specific mechanism in mice remains largely unknown. This study aimed to investigate the relaxation mechanism of levosimendan in the thoracic aorta smooth muscle of mice. Levosimendan-induced relaxation of isolated thoracic aortic rings that were precontracted with norepinephrine (NE) or KCl was recorded in an endothelium-independent manner. Vasodilatation by levosimendan was not associated with the production of the endothelial relaxation factors NO and PGI2. The voltage-dependent K+ channel (KV) blocker (4-aminopyridine) and selective KCa blocker (tetraethylammonium) had no effect on thoracic aortas treated with levosimendan, indicating that KV and KCa channels may not be involved in the levosimendan-induced relaxation mechanism. Although the inwardly rectifying K+ channel (Kir) blocker (barium chloride) and the KATP channel blocker (glibenclamide) significantly inhibited levosimendan-induced vasodilation in the isolated thoracic aorta, barium chloride had a much stronger inhibitory effect on levosimendan-induced vasodilation than glibenclamide, suggesting that levosimendan-induced vasodilation may be mediated by Kir channels. The vasodilation effect and expression of Kir 2.1 induced by levosimendan were further enhanced by the PKC inhibitor staurosporine. Extracellular calcium influx was inhibited by levosimendan without affecting intracellular Ca2+ levels in the isolated thoracic aorta. These results suggest that Kir channels play a more important role than KATP channels in regulating vascular tone in larger arteries and that the activity of the Kir channel is enhanced by the PKC pathway.

2.
Cell Biosci ; 12(1): 204, 2022 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-36539848

RESUMO

BACKGROUND: Sustained cardiac hypertrophy often develops maladaptive myocardial remodeling, and eventually progresses to heart failure and sudden death. Therefore, maladaptive hypertrophy is considered as a critical therapeutic target for many heart diseases. Mitophagy, a crucial mechanism in mitochondria quality control and cellular homeostasis, has been implicated in diverse cardiac disorders such as myocardial infarction, diabetic cardiomyopathy, cardiac hypertrophy and heart failure. However, what role mitophagy plays in heart diseases remains an enigma. PARKIN functions as an E3 ubiquitin protein ligase and mediates mitophagy cascades. It is still unclear whether PARKIN participates in the regulation of cardiac hypertrophy. RESULTS: PARKIN was downregulated in cardiomyocytes and hearts under hypertrophic stress. Enforced expression of PARKIN inhibited Ang II-induced cardiomyocyte hypertrophy. Compared to wide-type mice with Ang II-induced cardiac hypertrophy, Parkin transgenic mice subjected to Ang II administration showed attenuated cardiac hypertrophy and improved cardiac function. In addition, mitophagy machinery was impaired in response to Ang II, which was rescued by overexpression of PARKIN. PARKIN exerted the anti-hypertrophy effect through restoring mitophagy. In further exploring the underlying mechanisms, we found that PARKIN was transcriptionally activated by FOXO3a. FOXO3a promoted mitophagy and suppressed cardiac hypertrophy by targeting Parkin. CONCLUSIONS: The present study reveals a novel cardiac hypertrophy regulating model composed of FOXO3a, PARKIN and mitophagy program. Modulation of their levels may provide a new approach for preventing cardiac hypertrophy and heart failure.

3.
Cardiovasc Diagn Ther ; 12(3): 289-304, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35800356

RESUMO

Background: Myocardial infarction (MI) is a common cause of death. Thioredoxin-interacting protein (TXNIP) expression increases after MI, and it exerts a negative regulatory effect on cardiac function after MI. Our study aimed to investigate the specific regulatory mechanism of TXNIP on angiogenesis and cardiomyocyte apoptosis after MI. Methods: The TXNIP gene knock-in (TXNIP-KI) and knock-out (TXNIP-KO) mice were generated, respectively. Eight-week-old male TXNIP-KO, TXNIP-KI, and wild type (WT) mice were subjected to MI by permanent ligation of the left anterior descending artery. Cardiomyocyte apoptosis was detected by TUNEL assay on the 4th post-surgery day. The expressions of TXNIP, hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF), phosphorylated protein kinase B (p-AKT), p-AMP-activated protein kinase (p-AMPK), cleaved caspase-3, and caspase-3 were detected by Western blot. Quantitative real-time PCR was performed to detect the expression of TXNIP, HIF-1α, VEGF, prolyl hydroxylase (PHD) 1, and factor inhibiting HIF (FIH). In addition, the superoxide dismutase (SOD) activity and malondialdehyde (MDA) level in each group were also measured. On day 7 after MI, the hearts of sacrificed animals were analyzed by immunohistochemistry to assess CD31 expression and determine the density of angiogenesis. One month after treatment, the cardiac functional and structural changes were determined by echocardiography and the level of myocardial fibrosis was observed by Masson staining. Results: Compared with WT mice, TXNIP-KO mice had a significantly improved cardiac functional recovery after MI, and the proportion of myocardial fibrosis area was dramatically reduced, cardiomyocyte apoptosis was decreased, and angiogenesis was significantly increased; TXNIP-KI mice reversed in these changes. The expression of HIF-1α, p-AKT, and p-AMPK increased after MI in TXNIP-KO mice, and the mRNA expression of PHD 1 and FIH decreased. TXNIP-KI mice reversed in these changes. Conclusions: After MI, TXNIP down-regulated the level of HIF-1α and VEGF, reduced the number of angiogenesis, increased cardiomyocyte apoptosis, and ultimately led to a poor prognosis of ischemic myocardium. TXNIP was a protein with negative effects after MI and was expected to be a target for the prevention and treatment of MI.

4.
PLoS One ; 17(7): e0267331, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35802723

RESUMO

AIMS: The renin-angiotensin system (RAS) is over-activated and the serum angiotensin II (Ang II) level increased in obese patients, while their correlations were incompletely understood. This study aims to explore the role of Ang II in diet-induced obesity by focusing on adipose lipid anabolism and catabolism. METHODS: Rat model of AT1aR gene knockout were established to investigate the special role of Ang II on adipose lipid metabolism. Wild-type (WT) and AT1aR gene knockout (AT1aR-/-) SD rats were fed with normal diet or high-fat diet for 12 weeks. Adipose morphology and adipose lipid synthesis and lipolysis were examined. RESULTS: AT1aR deficiency activated lipolysis-related enzymes and increased the levels of NEFAs and glycerol released from adipose tissue in high-fat diet rats, while did not affect triglycerides synthesis. Besides, AT1aR knockout promoted energy expenditure and fatty acids oxidation in adipose tissue. cAMP levels and PKA phosphorylation in the adipose tissue were significantly increased in AT1aR-/- rats fed with high-fat. Activated PKA could promote adipose lipolysis and thus improved adipose histomorphology and insulin sensitivity in high-fat diet rats. CONCLUSIONS: AT1aR deficiency alleviated adipocyte hypertrophy in high-fat diet rats by promoting adipose lipolysis probably via cAMP/PKA pathway, and thereby delayed the onset of obesity and related metabolic diseases.


Assuntos
Dieta Hiperlipídica , Lipólise , Obesidade , Receptor Tipo 1 de Angiotensina , Tecido Adiposo/metabolismo , Angiotensina II/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Ácidos Graxos não Esterificados/metabolismo , Técnicas de Inativação de Genes , Obesidade/genética , Obesidade/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/genética
5.
Brain Res Bull ; 164: 146-156, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32858129

RESUMO

Accumulating evidence suggests that chronic metformin posttreatment offers potent neuroreparative effects against acute brain injury. However, in previous studies, metformin was not initially administered beyond 24 h postinjury, and the effects of delayed metformin treatment in traumatic brain injury (TBI) and other types of acute brain injury and the related mechanisms are unclear. To test this, male C57BL/6 mice received once daily metformin treatment (20, 50 or 100 mg/kg/d, i.p.) at day 1-14, day 1-2, day 1-10, day 3-10, day 5-12 or day 5-28 after cryogenic TBI (cTBI). The results showed that 100 mg/kg/d metformin administered at day 1-14 postinjury significantly promoted motor functional recovery in the beam walking and gait tests and reduced the infarct volume. Metformin (100 mg/kg/d) administered at day 1-10 or day 3-10 but not day 1-2 or day 5-12 after cTBI significantly improved motor functional outcomes at day 7 and 14, and reduced the infarct volume at day 14. Interestingly, the therapeutic time window was further expanded when the duration of metformin treatment starting at day 5 postinjury was extended to 2 weeks. Furthermore, compared with cTBI, the administration of metformin at day 3-10 or day 5-28 after cTBI significantly elevated the expression of phosphorylated adenosine monophosphate-activated protein kinase (AMPK) and growth associated protein 43 (an axonal regeneration marker) and the number of vascular branch points and decreased the area of glial scar and the number of amoeboid microglia in the peri-infarct area at day 14 or 28 postinjury. The above beneficial effects of metformin were blocked by the intracerebroventricular injection of the AMPK inhibitor compound C (40 µg/mouse/d). Our data provide the first evidence that metformin has a wide therapeutic time window for at least 5 days after cTBI, during which it can improve functional recovery by promoting tissue repair and inhibiting glial scar formation and microglial activation in a central AMPK-dependent manner.


Assuntos
Adenilato Quinase/metabolismo , Lesões Encefálicas Traumáticas/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Hipoglicemiantes/uso terapêutico , Metformina/uso terapêutico , Destreza Motora/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Recuperação de Função Fisiológica/efeitos dos fármacos , Animais , Encéfalo/metabolismo , Lesões Encefálicas Traumáticas/metabolismo , Modelos Animais de Doenças , Hipoglicemiantes/farmacologia , Masculino , Metformina/farmacologia , Camundongos , Fármacos Neuroprotetores/farmacologia , Fosforilação/efeitos dos fármacos
6.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 36(2): 119-123, 2020 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-32744003

RESUMO

Objective: To investigate whether the increased expression of thioredoxin interacting protein (TXNIP) in diabetes affects the senescence of islet ß cells. Methods: Six normal mice (db/m) and six diabetic mice (db/db) were randomly selected. Fasting blood glucose was measured by blood sugar meter, the expression levels of TXNIP protein, p16, p21 and Rb in pancreatic tissues were detected by Western blot, senescence-associated beta-galactosidase activity in pancreatic tissue was determined by immunochemical staining. INS-1 islet beta cells were randomly divided into 7 groups (n=6), and transfected with lentiviruses (30 µl) for 4 to 6 hours, then was screened with puromycin (PM, 3 µg/m) for 7 days to construct normal group, scramble ShRNA group (interference with airborne poison group), TXNIP-ShRNA-1 group (TXNIP silence group-1), TXNIP-ShRNA-2 group (TXNIP silence group 2), TXNIP-ShRNA-3 group (TXNIP silence group 3), Ad-GFP group (overexpression of the air virus group), Ad-TXNIP-GFP group (TXNIP overexpression group) stably transferred INS-1 islet beta cell line. TXNIP protein expression was detected by Western blot, aging-related beta-galactosidase activity was detected by immunochemical staining, the changes of expression of p16, p21 and Rb was determined by Western blot. Results: Compared with normal mice, the fasting blood glucose of db/db group was increased significantly (P<0. 01), the expression of TXNIP protein was increased significantly in pancreatic tissues(P<0. 05), positive staining rate of ß- galactosidase was increased significantly in pancreatic tissues, p16/p21/Rb protein expression levels were increased significantly (P<0. 05). Compared with Ad-GFP group, the positive staining rate of ß- galactosidase in Ad-TXNIP-GFP group was increased significantly, p16/p21/Rb protein expression levels were increased significantly (P<0. 01). Compared to the scramble ShRNA group, the positive staining rate of ß- galactosidase in TXNIP-ShRNA group was decreased, p16/p21/Rb protein expression levels were decreased significantly (P<0. 05). Conclusion: Diabetes can induce islet ß-cell senescence by up-regulating TXNIP expression.


Assuntos
Proteínas de Transporte/metabolismo , Senescência Celular , Diabetes Mellitus Experimental , Ilhotas Pancreáticas/citologia , Tiorredoxinas/metabolismo , Animais , Proteínas de Transporte/genética , Camundongos
7.
Sheng Li Xue Bao ; 70(2): 149-157, 2018 Apr 25.
Artigo em Chinês | MEDLINE | ID: mdl-29691579

RESUMO

This study investigated the effect of angiotensin II (Ang II) on apoptosis and thioredoxin-interacting protein (TXNIP) expression in INS-1 islet cells and the underlying mechanism. INS-1 cells cultured in vitro were treated with different concentration of Ang II for different time, and the viability was measured using cell counting kit-8 (CCK-8). After treatment with 1 × 10-6 mol/L Ang II for 24 h, flow cytometry and Western blot were used to measure the cell apoptosis, and Western blot was used to analyze the protein expression of TXNIP, carbohydrate response element-binding protein (ChREBP) and angiotensin II type 1 receptor (AT1R). Real-time PCR was used to detect TXNIP and ChREBP mRNA expression. IF/ICC was used to observe the TXNIP, ChREBP and AT1R expression. The results showed that Ang II reduced cell viability and induced the expression of TXNIP in a dose- and time-dependent manner (P < 0.05, n = 6) compared with the control group. Ang II induced apoptosis and up-regulated the expression of ChREBP and AT1R (P < 0.05, n = 6). AT1R inhibitor, telmisartan (TM), blocked Ang II-induced TXNIP and ChREBP overexpression (P < 0.05, n = 6) and inhibited Ang II-induced apoptosis. Taken together, Ang II increased ChREBP activation through AT1R, which subsequently increased TXNIP expression and promoted cell apoptosis. These findings suggest a therapeutic potential of targeting TXNIP in preventing Ang II-induced INS-1 cell apoptosis in diabetes.


Assuntos
Angiotensina II/farmacologia , Apoptose , Proteínas de Transporte/fisiologia , Células Secretoras de Insulina/fisiologia , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/fisiologia , Proteínas de Ciclo Celular , Linhagem Celular , Ratos , Receptor Tipo 1 de Angiotensina/fisiologia , Telmisartan/farmacologia , Regulação para Cima
8.
Sheng Li Xue Bao ; 70(2): 158-166, 2018 Apr 25.
Artigo em Chinês | MEDLINE | ID: mdl-29691580

RESUMO

Diabetes can cause a significant increase in the expression of thioredoxin (Trx)-interacting protein (TXNIP), which binds to Trx and inhibits its activity. The present study was aimed to investigate the effect of TXNIP on proliferation of rat INS-1 islet ß cells and the underlying mechanism. TXNIP overexpressing adenovirus vectors (Ad-TXNIP-GFP and Ad-TXNIPc247s-GFP) were constructed and used to infect INS-1 cells. Ad-TXNIPc247s-GFP vector carries a mutant C247S TXNIP gene, and its expression product (TXNIPc247s) cannot attach and inhibit Trx activity. The expression of TXNIP was detected by real-time PCR and Western blot. EdU and Ki67 methods were used to detect cell proliferation. Protein phosphorylation levels of ERK and AKT were detected by Western blot. The results showed that both TXNIP and TXNIPc247s protein overexpressions inhibited the proliferation of INS-1 cells, and the former's inhibitory effect was greater. Moreover, both of the two kinds of overexpressions inhibited the phosphorylation of ERK and AKT. These results suggest that TXNIP overexpression may inhibit the proliferation of INS-1 cells through Trx-dependent and non-Trx-dependent pathways, and the mechanism involves the inhibition of ERK and AKT phosphorylation.


Assuntos
Proteínas de Transporte/fisiologia , Vetores Genéticos , Células Secretoras de Insulina/citologia , Adenoviridae , Animais , Proteínas de Ciclo Celular , Linhagem Celular , Proliferação de Células , Diabetes Mellitus , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Oxirredução , Fosforilação , Proteínas Proto-Oncogênicas c-akt/fisiologia , Ratos
9.
Sheng Li Xue Bao ; 69(4): 445-451, 2017 Aug 25.
Artigo em Chinês | MEDLINE | ID: mdl-28825103

RESUMO

Thioredoxin (Trx) interacting protein (TXNIP) is a Trx-binding protein that inhibits the antioxidative function of Trx and is highly expressed in the serum and tissue samples from diabetes patients. This study was to explore whether TXNIP overexpression could cause INS-1 cell autophagy under normal glucose and lipid concentrations, and to analyze the role of autophagy in the apoptosis of INS-1 cells. The INS-1 cells cultured under normal conditions were divided into three groups: normal control, empty adenovirus vector (Ad-eGFP) and TXNIP overexpression (Ad-TXNIP-eGFP) groups. Forty-eight hours after transfection, the expression levels of TXNIP mRNA and protein were measured. Western blot was used to examine the protein expression levels of Beclin-1 and P62, as well as LC3-II/LC3-I ratio, which are associated with autophagy. IF/ICC was used to measure the autophagosome. In addition, the cleaved caspase-3/caspase-3 ratio, the apoptosis marker, was also measured, and the apoptotic rates were detected by flow cytometry (FCM). The results showed that the TXNIP mRNA and protein levels were significantly up-regulated in Ad-TXNIP-eGFP group, suggesting that TXNIP overexpression model was successfully established. In Ad-TXNIP-eGFP group, the protein levels of Beclin-1 and LC3-II/LC3-I ratio were increased, while the protein expression of P62 was decreased, compared with those in Ad-eGFP group. Red fluorescent intensity, representing autophagy level, was higher in Ad-TXNIP-eGFP group than that in Ad-eGFP group. These results suggested that TXNIP overexpression can significantly promote INS-1 cell autophagy. Meanwhile, cleaved caspase 3/caspase 3 ratio and the number of apoptotic cells were significantly increased in Ad-TXNIP-eGFP group. The inhibitor of autophagy, 3-MA, reduced TXNIP overexpression-induced apoptosis in INS-1 cells. Taken together, our data demonstrate that autophagy appears to be an important pathway in TXNIP overexpression-induced apoptosis in INS-1 cells.


Assuntos
Apoptose , Autofagia , Proteínas de Transporte/metabolismo , Ilhotas Pancreáticas/citologia , Proteínas de Transporte/genética , Caspase 3/metabolismo , Linhagem Celular , Humanos , Transfecção
10.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 23(6): 1586-90, 2015 Dec.
Artigo em Chinês | MEDLINE | ID: mdl-26708876

RESUMO

OBJECTIVE: To investigate the effects of indomethacin on proliferation and senescence of leukemia cells and to analyze whether non-steroidal anti-inflammatory drugs have any adjunctively therapeutic effects on leukemia. METHODS: Three kinds of leukemia cell lines (U266, K562 and U937) were treated with either indomethacin (final concentration 30 µmol/L) or solvent DMSO (control) for 7 days. Cell viability was determined by trypan blue, cell apoptosis and cell cycle were determined by flow cytometry, mRNA expression of hyperplastic suppression gene P21 and P27 was determined by RT-PCR, and cell senescence rate was assayed at day 0, 4, 7 after treatment. RESULTS: After treatment with indomethacin, the cell viability in U266 and U937 groups decreased significantly, while it was not changed in K562 group. Cell cycle in U266 and U937 groups was blocked at G(2)/M phase, but the blooking effect was not found in K562 cells. The cell apoptotic rate was enhanced in 3 treated groups (P < 0.01). The mRNA expression of P21 and P27 was significantly increased after indomethacin treatment in U266 and K562 cell lines except for the P27 mRNA expression of U937. The expression levels of P21 and P27 mRNA in U266 and K562 increased obviously. The cell senescence rate in K562 and U937 group also increased. CONCLUSION: The indomethacin possesses inhibitory effects on leukemia cells. However, its mechanisms are varied from different types of leukemia cells. It suggests that non-steroidal anti-inflammatory drugs can play a role in adjuvant therapy for leukemia, while its application would be adjusted according to different types of leukemia.


Assuntos
Proliferação de Células , Senescência Celular , Leucemia , Apoptose , Ciclo Celular , Sobrevivência Celular , Humanos , Indometacina
11.
Sheng Li Xue Bao ; 65(3): 309-18, 2013 Jun 25.
Artigo em Chinês | MEDLINE | ID: mdl-23788188

RESUMO

Adenovirus transfection technique was used in the current study to show if thioredoxin-interacting protein (TXNIP) overexpression can induce cell apoptosis and injury in H9C2 cardiomyocytes cultured in normal glucose condition. And the mechanisms were then investigated. Briefly, H9C2 cardiomyocytes in logarithmic growth phase were randomly divided into three groups: normal cultured group, empty adenovirus vector group (Ad-eGFP) and TXNIP overexpression group (Ad-TXNIP-eGFP). All cells were cultured in DMEM containing normal concentration of glucose (5 mmol/L) and lipid. 72 h after adenovirus transfection, cells and culture mediums were collected for further assay. The results showed that Ad-eGFP and Ad-TXNIP-eGFP adenovirus transfected H9C2 cells successfully, and the transfection efficiency reached the peak at 72 h. Compared with Ad-eGFP group, Ad-TXNIP-eGFP transfection significantly increased TXNIP mRNA (P < 0.05) and protein expression level (P < 0.01). TXNIP overexpression induced remarkable cell apoptosis and injury as evidenced by increased caspase-3 activity (P < 0.05), apoptotic rate (P < 0.01) and LDH activity (P < 0.01). To further analysis the mechanisms of TXNIP-induced cell apoptosis, we also determined Trx activity, Trx related free radical injury and p38 kinase activation, which are involved in free radical induced apoptosis. The results showed that, compared with those in Ad-eGFP group, Trx activity was significantly decreased (P < 0.01), while malondialdehyde (MDA), 3-nitrotyrosine contents and p38 kinase activity were significantly increased (P < 0.01) in TXNIP overexpression group. These results suggest that TXNIP overexpression alone can induce severe apoptosis and injury in H9C2 cardiomyocytes even they are cultured in normal glucose and lipid concentration conditions. The mechanism involved is that overexpressed TXNIP can bind and inhibit Trx, impairs its antioxidative and antiapoptotic function, and then increases free radical induced injury and p38 kinase dependent apoptosis.


Assuntos
Apoptose , Proteínas de Transporte/metabolismo , Miócitos Cardíacos/citologia , Adenoviridae/genética , Animais , Proteínas de Transporte/genética , Caspase 3/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular , Vetores Genéticos , Ratos , Tiorredoxinas/metabolismo
12.
Sheng Li Xue Bao ; 62(3): 261-8, 2010 Jun 25.
Artigo em Chinês | MEDLINE | ID: mdl-20571744

RESUMO

The aim of the present study is to investigate the change of thioredoxin (Trx) system in myocardial tissue of type 2 diabetic rats after myocardial injury and the underlying mechanism. Adult Sprague Dawley rats were randomly divided into two groups: normal control (NC) group and diabetes (DM) group. Rats in DM group were subjected to high-sugar, high-fat diet and streptozotocin (STZ) injection. Rats in NC group were only given normal diet and equal amount of citric acid buffer injection. At week 1, 2, 4, 12, 21 after STZ injection, plasma glucose concentration and the concentrations of insulin, creatine kinase MB (CK-MB), cardiac troponin I (cTnI) in serum were measured. Myocardial Trx and thioredoxin reductase (TR) activities, as well as caspase-3 activity, were determined by respective assay methods. Protein and mRNA levels of Trx, TR, Trx interacting protein (TXNIP) were determined by Western blot and real time PCR, respectively. The results showed that type 2 diabetic rat model was successfully established at week 1 after STZ injection, and myocardial injury was induced from week 2. Moreover, caspase-3 activity was significantly increased at week 4, 12 in diabetic rats. The activities of myocardial Trx and TR in diabetic rats was decreased from week 2, and continually aggravated as the disease developed. Compared with those in NC group, the mRNA levels of Trx1, Trx2, TR1, TR2 in DM group decreased at week 4, and then increased in week 12. In DM group, the protein levels of Trx1, Trx2, TR1 and TR2 increased significantly at week 12. The mRNA expressions of myocardial TXNIP in diabetic rats were significantly increased at week 4, 12, 24 and protein expression was increased at week 12. These results suggest diabetes can decrease myocardial Trx, TR activity, inducing myocardial cell apoptosis and heart injury. The inhibitory effect of diabetes is mainly associated with TXNIP up-regulation and Trx nitration.


Assuntos
Diabetes Mellitus Experimental/fisiopatologia , Miocárdio/patologia , Tiorredoxina Dissulfeto Redutase/metabolismo , Tiorredoxinas/metabolismo , Animais , Apoptose , Proteínas de Transporte/metabolismo , Caspase 3/metabolismo , Proteínas de Ciclo Celular , Creatina Quinase Forma MB/sangue , Dieta Hiperlipídica , Insulina/sangue , Ratos , Ratos Sprague-Dawley , Estreptozocina , Troponina I/sangue , Regulação para Cima
13.
J Pharmacol Exp Ther ; 328(3): 777-84, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19033557

RESUMO

Oxidative/nitrative stress caused by peroxynitrite, the reaction product of superoxide (O2(.-)) and nitric oxide (NO), is the primary cause of myocardial ischemia/reperfusion injury. The present study determined whether INO-4885 [5,10,15,20-tetra[N-(benzyl-4'-carboxylate)-2-pyridinium]-21H,23H-porphine iron(III) chloride], a new peroxynitrite decomposition catalyst, may provide cellular protection and protect heart from myocardial ischemia/reperfusion injury. Adult male mice were subjected to 30 min of ischemia and 3 or 24 h of reperfusion. Mice were randomized to receive vehicle, INO-4885 without catalytic moiety, or INO-4885 (3-300 microg/kg i.p.) 10 min before reperfusion. Infarct size, apoptosis, nitrotyrosine content, NO/O2(.-) production, and inducible nitric-oxide synthase (iNOS)/NADPH oxidase expression were determined. INO-4885 treatment reduced ischemia/reperfusion-induced protein nitration and caspase 3 activation in a dose-dependent fashion in the range of 3 to 100 microg/kg. However, doses exceeding 100 microg/kg produced nonspecific effects and attenuated its protective ability. At the optimal dose (30 microg/kg), INO-4885 significantly reduced infarct size (p < 0.01), decreased apoptosis (p < 0.01), and reduced tissue nitrotyrosine content (p < 0.01). As expected, INO-4885 had no effect on ischemia/reperfusion-induced iNOS expression and NO overproduction. To our surprise, this compound significantly reduced superoxide production and partially blocked NADPH oxidase overexpression in the ischemic/reperfused cardiac tissue. Additional experiments demonstrated that INO-4885 provided better cardioprotection than N-(3-(aminomethyl)benzyl)acetamidine (1400W, a selective iNOS inhibitor), apocynin (an NADPH oxidase inhibitor), or Tiron (a cell-permeable superoxide scavenger). Taken together, our data demonstrated that INO-4885 is a cardioprotective molecule that attenuates myocardial reperfusion injury by facilitating peroxynitrite decomposition and inhibiting NADPH oxidase-derived O2(.-) production.


Assuntos
Coração/fisiopatologia , Metaloporfirinas/uso terapêutico , Infarto do Miocárdio/prevenção & controle , Ácido Peroxinitroso/uso terapêutico , Traumatismo por Reperfusão/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Coração/efeitos dos fármacos , Coração/fisiologia , Masculino , Camundongos , NADPH Oxidases/efeitos dos fármacos , NADPH Oxidases/genética , Óxido Nítrico Sintase Tipo II/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/genética , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia
14.
Sheng Li Xue Bao ; 58(1): 90-4, 2006 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-16489410

RESUMO

Using two-kidney one-clip renal hypertensive (2K1C group), stress-induced hypertensive (neural group), DOCA-salt treated hypertensive (DOCA group) and spontaneously hypertensive rats (SHR group), to investigate the change in AT(1A)-receptor autoantibodies (AT(1A)-AAs) during the development of the four types of hypertension. The biological activities of AT(1A)-AAs were examined. It was shown that the frequency of occurrence and titres of AT(1A)-AAs increased significantly during the development of hypertension. In the four hypertensive groups studied, the occurrence of AT(1A)-AAs was most prominent in SHR, 2K1C and neural groups. The biological effects of AT(1A)-AAs were shown to increase the beating frequency of cultured neonatal myocardial and vascular contractile tension. It is suggested that autoimmune mechanisms are involved the pathogenesis of different types of hypertension and the AT(1A)-AAs may be one of the mechanisms leading to cardiac hypertrophy.


Assuntos
Autoanticorpos/sangue , Hipertensão Renovascular/imunologia , Hipertensão/imunologia , Hipertensão/fisiopatologia , Receptor Tipo 1 de Angiotensina/imunologia , Animais , Desoxicorticosterona/administração & dosagem , Hipertensão/classificação , Hipertensão/etiologia , Hipertensão Renovascular/fisiopatologia , Masculino , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Ratos Wistar , Estresse Fisiológico/fisiologia
15.
Sheng Li Xue Bao ; 56(6): 730-4, 2004 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-15614423

RESUMO

The purpose of this study was to compare the vasodilating effects of angiotensin-(1-7) [Ang-(1-7)] on the different vessels and to clarify its mechanisms by using relaxing responses of preconstricted vascular rings. The results showed: (1) Ang-(1-7) dose-dependently induced vasorelaxation in all the vessels studied. However, there is apparent heterogeneity in the responsiveness of vessels from different origin. (2) The Ang-(1-7)-induced vasorelaxation was endothelium dependent and largely mediated by NO system. (3) The vasodilator action of Ang-(1-7) was not mediated by AT1 or AT2 receptor subtypes. It is suggested that the Ang-(1-7)-induced vasorelaxation is endothelium dependent by some other unclarified angiotensin receptor subtypes and is largely mediated by NO system.


Assuntos
Angiotensina I/farmacologia , Endotélio Vascular/efeitos dos fármacos , Óxido Nítrico/biossíntese , Fragmentos de Peptídeos/farmacologia , Vasodilatadores/farmacologia , Angiotensina I/fisiologia , Animais , Endotélio Vascular/metabolismo , Feminino , Masculino , Coelhos , Receptor Tipo 1 de Angiotensina/fisiologia
16.
Artigo em Chinês | MEDLINE | ID: mdl-21207854

RESUMO

AIM AND METHODS: The effects of losartan (after operation 2 week to 10 week, 5 mg/kg d ig) on generation of AT1R-AA in sera were observed during development of hypertension in rats. The renovascular hypertension (RVH) model was established by two-kidney one-clip method, a synthetic peptide corresponding to amino acid sequence 165-191 of the second extracellular loop of the angiotensin II-1 receptor (AT1R) was used as antigen, SA-ELISA were used to examine sera AT1R autoantibody (AT1R-AA). RESULTS: The frequencies and titres of AT1R-AA after operation one week rats were significantly increased (P < 0.05). The treatment with losartan not only inhibited structural and functional changes, but also the frequencies and titres of AT1R-AA was significantly lower (P < 0.05) than RVH group. CONCLUSION: It is suggested that the losartan significantly inhibits generation of the AT1R-AA.


Assuntos
Autoanticorpos/biossíntese , Hipertensão Renovascular/sangue , Losartan/farmacologia , Receptores de Angiotensina/imunologia , Animais , Autoanticorpos/sangue , Hipertensão Renovascular/imunologia , Masculino , Ratos , Ratos Wistar
17.
Sheng Li Xue Bao ; 54(4): 317-20, 2002 Aug 25.
Artigo em Chinês | MEDLINE | ID: mdl-12195280

RESUMO

The aim of this study was to observe the change in angiotensin II receptor subtype 1 (AT(1)) autoantibody during the development of renovascular hypertension (RVH). The Goldblatt renovascular hypertension model was established by the two-kidney one-clip method, and a synthetic peptide corresponding to amino acid sequence 165-191 of the second extracellular loop of the AT(1)-receptor was used as the antigen. Sera AT(1)-receptor autoantibody was detected by SA-ELISA. It was shown that two weeks after operation both the frequency of occurrence and the titre of autoantibodies to AT(1)-receptor were significantly increased as compared with the pre-treatment control. The increase in autoantibodies lasted several weeks and then decreased gradually to the pre-clipping level at 12 weeks. It is suggested that autoimmune mechanisms are involved in the pathogenesis of renovascular hypertension and the AT(1) autoantibodies may be one of the mechanisms leading to cardiac hypertrophy.


Assuntos
Autoanticorpos/sangue , Hipertensão Renovascular/imunologia , Receptor Tipo 1 de Angiotensina/metabolismo , Animais , Modelos Animais de Doenças , Hipertensão Renovascular/sangue , Rim/fisiopatologia , Ratos , Receptor Tipo 1 de Angiotensina/imunologia
18.
J Am Coll Cardiol ; 39(11): 1866-73, 2002 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-12039504

RESUMO

OBJECTIVES: We sought to investigate the mechanism responsible for the occurrence of anticardiac receptor autoantibodies. BACKGROUND: Increasing evidence suggests the involvement of autoimmune mechanisms in the pathogenesis of a number of cardiovascular diseases. Among them, the biologic, functional and pathogenic properties of anticardiac receptor antibodies have been extensively investigated. However, the mechanism responsible for the occurrence of anticardiac receptor autoantibodies remains poorly understood. METHODS: Two rat models (aortic banding [AB] and adriamycin [ADR] groups) were constructed. Determination of cardiac function and morphology and T-lymphocyte subtypes, enzyme-linked immunosorbent assay and cardiomyocyte cultures were performed. RESULTS: It was shown, in the AB and ADR groups, that the frequency and titer of autoantibodies to beta(1) and muscarinic type 2 receptors were increased when myocardial remodeling occurred, as evidenced by significant cardiac morphologic changes, deposition of collagen and obvious functional impairment. This suggests that cardiac remodeling itself, in two disparate models of heart failure and cardiomyopathy, was able to trigger the genesis of anticardiac receptor autoantibodies. These autoantibodies have biologic effects similar to those seen in human autoantibodies. They have also shown a characteristic self-growth, as well as a time-course decline, suggesting that a negative finding of anticardiac receptor autoantibodies in sera of patients with heart disease does not necessarily imply that there is no autoimmune reaction involved in the pathogenesis. CONCLUSIONS: Our results demonstrated that myocardial damage was able to trigger the occurrence of an autoimmune reaction, resulting in the genesis of anticardiac receptor autoantibodies with properties similar to those seen in patients with idiopathic dilated cardiomyopathy.


Assuntos
Autoanticorpos/sangue , Cardiomiopatia Dilatada/imunologia , Receptores Adrenérgicos beta 1/imunologia , Receptores Muscarínicos/imunologia , Remodelação Ventricular/imunologia , Animais , Autoimunidade , Peso Corporal , Relação CD4-CD8 , Cardiomiopatia Dilatada/patologia , Modelos Animais de Doenças , Feminino , Coração/anatomia & histologia , Técnicas In Vitro , Masculino , Miocárdio/citologia , Miocárdio/imunologia , Tamanho do Órgão , Ratos , Ratos Wistar , Receptores Adrenérgicos beta 1/metabolismo , Receptores Muscarínicos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA