Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cell Mol Life Sci ; 81(1): 228, 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38777955

RESUMO

Diabetic cardiomyopathy (DCM) is a prevalent complication of type 2 diabetes (T2D). 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) is a glycolysis regulator. However, the potential effects of PFKFB3 in the DCM remain unclear. In comparison to db/m mice, PFKFB3 levels decreased in the hearts of db/db mice. Cardiac-specific PFKFB3 overexpression inhibited myocardial oxidative stress and cardiomyocyte apoptosis, suppressed mitochondrial fragmentation, and partly restored mitochondrial function in db/db mice. Moreover, PFKFB3 overexpression stimulated glycolysis. Interestingly, based on the inhibition of glycolysis, PFKFB3 overexpression still suppressed oxidative stress and apoptosis of cardiomyocytes in vitro, which indicated that PFKFB3 overexpression could alleviate DCM independent of glycolysis. Using mass spectrometry combined with co-immunoprecipitation, we identified optic atrophy 1 (OPA1) interacting with PFKFB3. In db/db mice, the knockdown of OPA1 receded the effects of PFKFB3 overexpression in alleviating cardiac remodeling and dysfunction. Mechanistically, PFKFB3 stabilized OPA1 expression by promoting E3 ligase NEDD4L-mediated atypical K6-linked polyubiquitination and thus prevented the degradation of OPA1 by the proteasomal pathway. Our study indicates that PFKFB3/OPA1 could be potential therapeutic targets for DCM.


Assuntos
Cardiomiopatias Diabéticas , GTP Fosfo-Hidrolases , Miócitos Cardíacos , Fosfofrutoquinase-2 , Ubiquitinação , Fosfofrutoquinase-2/metabolismo , Fosfofrutoquinase-2/genética , Animais , Cardiomiopatias Diabéticas/metabolismo , Cardiomiopatias Diabéticas/patologia , Cardiomiopatias Diabéticas/genética , Camundongos , GTP Fosfo-Hidrolases/metabolismo , GTP Fosfo-Hidrolases/genética , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Masculino , Estresse Oxidativo , Apoptose/genética , Miocárdio/metabolismo , Miocárdio/patologia , Camundongos Endogâmicos C57BL , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/genética , Glicólise , Humanos , Estabilidade Proteica
2.
Prostaglandins Other Lipid Mediat ; 167: 106740, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37119935

RESUMO

Epoxyeicosatrienoic acids (EETs), which are synthesized from arachidonic acid by cytochrome P450 epoxygenases, function primarily as autocrine and paracrine effectors in the cardiovascular system. So far, most research has focused on the vasodilatory, anti-inflammatory, anti-apoptotic and mitogenic properties of EETs in the systemic circulation. However, whether EETs could suppress tissue factor (TF) expression and prevent thrombus formation remains unknown. Here we utilized in vivo and in vitro models to investigate the effects and underlying mechanisms of exogenously EETs on LPS induced TF expression and inferior vein cava ligation induced thrombosis. We observed that the thrombus formation rate and the size of the thrombus were greatly reduced in 11,12-EET treated mice,accompanied by decreased TF and inflammatory cytokines expression. Further in vitro studies showed that by enhancing p38 MAPK activation and subsequent tristetraprolin (TTP) phosphorylation, LPS strengthened the stability of TF mRNA and induced increased TF expression. However, by strengthening PI3K-dependent Akt phosphorylation, which acted as a negative regulator of p38-TTP signaling pathway,11,12-EET reduced LPS-induced TF expression in monocytes. In addition, 11,12-EET inhibited LPS-induced NF-κB nuclear translocation by activating the PI3K/Akt pathway. Further study indicated that the inhibitory effect of 11,12-EET on TF expression was mediated by antagonizing LPS-induced activation of thromboxane prostanoid receptor. In conclusion, our study demonstrated that 11,12-EET prevented thrombosis by reducing TF expression and targeting the CYP2J2 epoxygenase pathway may represent a novel approach to mitigate thrombosis related diseases.


Assuntos
Proteínas Proto-Oncogênicas c-akt , Trombose , Animais , Camundongos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Lipopolissacarídeos/farmacologia , Tromboplastina/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Transdução de Sinais , Citocromo P-450 CYP2J2 , Ácido 8,11,14-Eicosatrienoico/metabolismo , Trombose/tratamento farmacológico , Estabilidade de RNA
3.
Ecotoxicol Environ Saf ; 248: 114296, 2022 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-36399994

RESUMO

Elevated atmospheric carbon dioxide (eCO2) concentrations can alter the carbon:nitrogen ratio and palatability of host plants for herbivorous insects, but rhizobacteria likely mitigate the alteration and influence physiological adaptation of insects. In this study, we conducted transcriptomic analysis of maize (Zea mays) response to Azotobacter chroococcum (AC) inoculation under eCO2 conditions in contrast to ambient CO2 (aCO2), and studied the effects of plant-defense change of maize under eCO2 on the oriental armyworm, Mythimna separata. Results showed that there were 16, 14, 16 and 135 differentially expressed genes that were associated with plant-defense response in maize leaves between aCO2-CK and aCO2-AC, eCO2-CK and eCO2-AC, aCO2-CK and eCO2-CK, aCO2-AC and eCO2-AC, respectively. Moreover, A. chroococcum inoculation and eCO2 influenced plant hormone signal transduction of maize. Interestingly, A. chroococcum inoculation significantly decreased the contents of JA (jasmonic acid) and JA-Ile (isoleucine conjugate of JA) in leaves, but eCO2 markedly increased contents of JA-Ile, JA and SA (salicylic acid). Compared to aCO2, eCO2 significantly decreased activity of protective enzyme (catalase), and increased activities of digestive (lipase and protease), protective (peroxidase) and detoxifying enzymes (carboxylesterase, Mixed-functional oxidase and glutathione s-transferase), prolonged developmental time, and decreased survival rate and body weight of larvae (P < 0.05). A. chroococcum inoculation significantly increased the activity of protective enzyme (catalase), and decreased the activities of detoxifying enzymes (carboxylesterase, glutathione s-transferase and mixed-functional oxidase), thus increased the growth rate and body weight of larvae in comparison with no-inoculation of A. chroococcum (P < 0.05). The indices of M. separata were significantly correlated with the foliar contents of JA, JA-Ile and SA (|r| = 0.44-0.85, P < 0.05), indicating that A. chroococcum inoculation altered the physiological adaptation of M. separata under eCO2 by disturbing defense substances in maize. Our results in understanding effects of A. chroococcum inoculation on maize resistance to herbivorous insects will be valuable for agricultural pest control in the future at eCO2 conditions.


Assuntos
Dióxido de Carbono , Zea mays , Animais , Zea mays/genética , Catalase , Dióxido de Carbono/farmacologia , Spodoptera , Glutationa Transferase , Hidrolases de Éster Carboxílico
4.
Pharmacol Res ; 183: 106367, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35882293

RESUMO

Diabetic nephropathy (DN) is the most common cause of end-stage renal disease, and few treatment options that prevent the progressive loss of renal function are available. Studies have shown that dietary fiber intake improves kidney diseases and metabolism-related diseases, most likely through short-chain fatty acids (SCFAs). The present study aimed to examine the protective effects of inulin-type fructans (ITFs) on DN through 16 S rRNA gene sequencing, gas chromatographymass spectrometry (GCMS) analysis and fecal microbiota transplantation (FMT). The results showed that ITFs supplementation protected against kidney damage in db/db mice and regulated the composition of the gut microbiota. Antibiotic treatment and FMT experiments further demonstrated a key role of the gut microbiota in mediating the beneficial effects of ITFs. The ITFs treatment-induced changes in the gut microbiota led to an enrichment of SCFA-producing bacteria, especially the genera Akkermansia and Candidatus Saccharimonas, which increased the fecal and serum acetate concentrations. Subsequently, acetate supplementation improved glomerular damage and renal fibrosis by attenuating mitochondrial dysfunction and reducing toxic glucose metabolite levels. In conclusion, ITFs play a renoprotective role by modulating the gut microbiota and increasing acetate production. Furthermore, acetate mediates renal protection by regulating glucose metabolism, decreasing glycotoxic product levels and improving mitochondrial function.


Assuntos
Diabetes Mellitus , Nefropatias Diabéticas , Microbioma Gastrointestinal , Animais , Bactérias/metabolismo , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/prevenção & controle , Ácidos Graxos Voláteis/metabolismo , Frutanos/farmacologia , Frutanos/uso terapêutico , Inulina/metabolismo , Inulina/uso terapêutico , Camundongos
5.
Am J Physiol Heart Circ Physiol ; 321(2): H353-H368, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34142887

RESUMO

Arterial stiffness, a consequence of smoking, is an underlying risk factor of cardiovascular diseases. Epoxyeicosatrienoic acids (EETs), hydrolyzed by soluble epoxide hydrolase (sEH), have beneficial effects against vascular dysfunction. However, the role of sEH knockout in nicotine-induced arterial stiffness was not characterized. We hypothesized that sEH knockout could prevent nicotine-induced arterial stiffness. In the present study, Ephx2 (the gene encodes sEH enzyme) null (Ephx2-/-) mice and wild-type (WT) littermate mice were infused with or without nicotine and administered with or without nicotinamide [NAM, sirtuin-1 (SIRT1) inhibitor] simultaneously for 4 wk. Nicotine treatment increased sEH expression and activity in the aortas of WT mice. Nicotine infusion significantly induced vascular remodeling, arterial stiffness, and SIRT1 deactivation in WT mice, which was attenuated in Ephx2 knockout mice (Ephx2-/- mice) without NAM treatment. However, the arterial protective effects were gone in Ephx2-/- mice with NAM treatment. In vitro, 11,12-EET treatment attenuated nicotine-induced matrix metalloproteinase 2 (MMP2) upregulation via SIRT1-mediated yes-associated protein (YAP) deacetylation. In conclusion, sEH knockout attenuated nicotine-induced arterial stiffness and vascular remodeling via SIRT1-induced YAP deacetylation.NEW & NOTEWORTHY We presently show that sEH knockout repressed nicotine-induced arterial stiffness and extracellular matrix remodeling via SIRT1-induced YAP deacetylation, which highlights that sEH is a potential therapeutic target in smoking-induced arterial stiffness and vascular remodeling.


Assuntos
Aorta/efeitos dos fármacos , Epóxido Hidrolases/genética , Niacinamida/farmacologia , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Sirtuína 1/metabolismo , Rigidez Vascular/efeitos dos fármacos , Complexo Vitamínico B/farmacologia , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Ácido 8,11,14-Eicosatrienoico/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Aorta/metabolismo , Aorta/fisiopatologia , Metaloproteinase 2 da Matriz/efeitos dos fármacos , Metaloproteinase 2 da Matriz/genética , Camundongos , Camundongos Knockout , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/efeitos dos fármacos , Rigidez Vascular/genética , Vasodilatadores/farmacologia , Proteínas de Sinalização YAP
6.
J Biol Chem ; 296: 100667, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33864813

RESUMO

The epoxyeicosatrienoic acid (EET) exerts beneficial effects on insulin resistance and/or hypertension. EETs could be readily converted to less biological active diols by soluble epoxide hydrolase (sEH). However, whether sEH inhibition can ameliorate the comorbidities of insulin resistance and hypertension and the underlying mechanisms of this relationship are unclear. In this study, C57BL/6 mice were rendered hypertensive and insulin resistant through a high-fat and high-salt (HF-HS) diet. The sEH inhibitor, 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea (TPPU), was used to treat mice (1 mg/kg/day) for 8 weeks, followed by analysis of metabolic parameters. The expression of sEH and the sodium-glucose cotransporter 2 (SGLT2) was markedly upregulated in the kidneys of mice fed an HF-HS diet. We found that TPPU administration increased kidney EET levels, improved insulin resistance, and reduced hypertension. Furthermore, TPPU treatment prevented upregulation of SGLT2 and the associated increased urine volume and the excretion of urine glucose and urine sodium. Importantly, TPPU alleviated renal inflammation. In vitro, human renal proximal tubule epithelial cells (HK-2 cells) were used to further investigate the underlying mechanism. We observed that 14,15-EET or sEH knockdown or inhibition prevented the upregulation of SGLT2 upon treatment with palmitic acid or NaCl by inhibiting the inhibitory kappa B kinase α/ß/NF-κB signaling pathway. In conclusion, sEH inhibition by TPPU alleviated insulin resistance and hypertension induced by an HF-HS diet in mice. The increased urine excretion of glucose and sodium was mediated by decreased renal SGLT2 expression because of inactivation of the inhibitory kappa B kinase α/ß/NF-κB-induced inflammatory response.


Assuntos
Epóxido Hidrolases/antagonistas & inibidores , Regulação da Expressão Gênica , Hipertensão/prevenção & controle , Resistência à Insulina , Rim/metabolismo , Doenças Metabólicas/prevenção & controle , Transportador 2 de Glucose-Sódio/metabolismo , Animais , Regulação para Baixo , Hipertensão/etiologia , Hipertensão/metabolismo , Hipertensão/patologia , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Rim/efeitos dos fármacos , Rim/patologia , Masculino , Doenças Metabólicas/etiologia , Doenças Metabólicas/metabolismo , Doenças Metabólicas/patologia , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , Transportador 2 de Glucose-Sódio/genética
7.
Mol Cell Endocrinol ; 523: 111149, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33387601

RESUMO

Studies have shown that epoxyeicosatrienoic acids (EETs) can regulate glucose homeostasis, but the specific mechanisms need further exploration. The sodium-glucose co-transporter 2 (SGLT2) is highly expressed in diabetic kidneys, which further promotes renal reabsorption of glucose to respond to the hyperglycemic state of diabetes. Herein, whether EETs can be a latent inhibitor of SGLT2 to regulate glucose homeostasis in diabetic state needs to be elucidated. Our study demonstrated that EETs attenuated the glucose reabsorption via renal tubular epithelial cells in diabetic mice, which partly accounted for the beneficial effects of EETs on glucose homeostasis. Moreover, 14,15-EET suppressed SGLT2 expression in both diabetic kidney and renal tubular epithelial cells. Further, inhibition of NF-κB with BAY 11-7082 decreased insulin-induced SGLT2 expression while NF-κB overexpression reversed the above effects. In addition, 14,15-EET attenuated SGLT2 expression via inactivating NF-κB. Mechanistically, 14,15-EET attenuated NF-κB mediated SGLT2 transcription at the -1821/-1812 P65-binding site. These results showed that EETs ameliorated glucose homeostasis via preventing NF-κB-mediated transcription of SGLT2 in renal tubular epithelial cells, providing a unique therapeutic strategy for insulin resistance and diabetes.


Assuntos
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Células Epiteliais/metabolismo , Glucose/metabolismo , Homeostase , Túbulos Renais Proximais/citologia , NF-kappa B/metabolismo , Transportador 2 de Glucose-Sódio/genética , Transcrição Gênica , Ácido 8,11,14-Eicosatrienoico/farmacologia , Animais , Linhagem Celular , Diabetes Mellitus Experimental/patologia , Células Epiteliais/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Humanos , Insulina/farmacologia , Camundongos Endogâmicos C57BL , Compostos de Fenilureia/administração & dosagem , Piperidinas/administração & dosagem , Transportador 2 de Glucose-Sódio/metabolismo , Transcrição Gênica/efeitos dos fármacos
8.
Clin Cardiol ; 43(12): 1478-1493, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33026120

RESUMO

BACKGROUD: The association between underlying comorbidities and cardiac injury and the prognosis in coronavirus disease 2019 (COVID-19) patients was assessed in this study. HYPOTHESIS: The underlying comorbidities and cardiac injury may be associated with the prognosis in COVID-19 patients. METHODS: A systematic search was conducted in PubMed, EMBASE, Web of science, and The Cochrane library from December 2019 to July 2020. The odds ratio (OR) and 95% confidence intervals (95% CI) were used to estimate the probability of comorbidities and cardiac injury in COVID-19 patients with or without severe type, or in survivors vs nonsurvivors of COVID-19 patients. RESULTS: A total of 124 studies were included in this analysis. A higher risk for severity was observed in COVID-19 patients with comorbidities. The pooled result in patients with hypertension (OR 2.57, 95% CI: 2.12-3.11), diabetes (OR 2.54, 95% CI: 1.89-3.41), cardiovascular diseases (OR 3.86, 95% CI: 2.70-5.52), chronic obstractive pulmonary disease (OR 2.71, 95% CI: 1.98-3.70), chronic kidney disease (OR 2.20, 95% CI: 1.27-3.80), and cancer (OR 2.42, 95% CI: 1.81-3.22) respectively. All the comorbidities presented a higher risk of mortality. Moreover, the prevalence of acute cardiac injury is higher in severe group than in nonsevere group, and acute cardiac injury is associated with an increased risk for in-hospital mortality. CONCLUSION: Comorbidities and acute cardiac injury are closely associated with poor prognosis in COVID-19 patients. It is necessary to continuously monitor related clinical indicators of organs injury and concern comorbidities in COVID-19 patients.


Assuntos
COVID-19/mortalidade , Índice de Gravidade de Doença , COVID-19/fisiopatologia , Doenças Cardiovasculares/mortalidade , Causas de Morte , Comorbidade , Diabetes Mellitus/mortalidade , Feminino , Humanos , Hipertensão/mortalidade , Masculino , Neoplasias/mortalidade , Pandemias/estatística & dados numéricos , Prognóstico , Insuficiência Renal Crônica/mortalidade
10.
Mol Med Rep ; 22(3): 1859-1867, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32582979

RESUMO

Age-related alterations in the renin-angiotensin-aldosterone system (RAAS) have been reported in the cardiovascular system; however, the detailed mechanism of the RAAS component mineralocorticoid receptors (MR) has not been elucidated. The present study aimed to investigate the associations between MR and cardiac aging in rats, as well as the regulatory effects of oxidative stress and mitochondrial abnormalities in the aging process. MR expression in the hearts of male Sprague­Dawley rats aged 3 months (young rats) and 24 months (old rats) was evaluated in vivo. In addition, in vitro, H9C2 cells were treated with a specific MR antagonist, eplerenone, in order to investigate the molecular mechanism underlying the inhibition of myocyte aging process. The results demonstrated that MR expression was significantly higher in 24­month­old rat hearts compared with in 3­month­old rat hearts. These changes were accompanied by increased p53 expression, decreased peroxisome proliferator­activated receptor γ coactivator­1α expression, decreased mitochondrial renewal as assessed by electron microscopy, increased oxidative stress and decreased superoxide dismutase. In vitro, selective antagonism of MR partially blocked H2O2­induced myocardial aging as assessed by p16, p21 and p53 expression levels and excessive reactive oxygen species (ROS) accumulation. These results indicated that increased MR expression may drive age­related cardiac dysfunction via mitochondrial damage, increased ROS accumulation and an imbalanced redox state.


Assuntos
Envelhecimento/metabolismo , Eplerenona/farmacologia , Antagonistas de Receptores de Mineralocorticoides/farmacologia , Miócitos Cardíacos/citologia , Receptores de Mineralocorticoides/metabolismo , Animais , Linhagem Celular , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Peróxido de Hidrogênio/efeitos adversos , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima
11.
Biochem Pharmacol ; 177: 113951, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32251672

RESUMO

Dipeptidyl peptidase-4 (DPP4) is elevated in numerous cardiovascular pathological processes and DPP4 inhibition is associated with reduced inflammation and oxidative stress. The aim of this study was to examine the role of DPP4 in endothelial senescence. Sprague-Dawley rats (24 months) were orally administrated saxagliptin (10 mg·kg-1·d-1), a DPP4 inhibitor, for 12 weeks in drinking water. Body weight, heart rate, blood glucose, and blood pressure were measured and vascular histological experiments were performed. In vitro studies were performed using H2O2-induced senescent human umbilical vein endothelial cells. Both in vivo and in vitro studies confirmed the elevation of DPP4 in senescent vascular endothelium, and inhibition or knockdown of DPP4 ameliorated endothelial senescence. In addition, DPP4 inhibition or silencing reduced endothelial oxidative stress levels in aging vasculature and senescent endothelial cells. Moreover, DPP4 inhibition or knockdown normalized the expression and phosphorylation of AMP-activated protein kinase-α (AMPKα) and sirtuin 1 (SIRT1) expression. Furthermore, the beneficial effects of DPP4 inhibition or knockdown on endothelial cell senescence were at least partly dependent on SIRT1 and Nrf2 activation. In conclusion, our study demonstrated that DPP4 inhibition or silencing ameliorated endothelial senescence both in vivo and in vitro by regulating AMPK/SIRT1/Nrf2. DPP4 may be a new therapeutic target to combat endothelial senescence.


Assuntos
Adamantano/análogos & derivados , Senescência Celular/efeitos dos fármacos , Dipeptídeos/farmacologia , Inibidores da Dipeptidil Peptidase IV/farmacologia , Células Endoteliais/efeitos dos fármacos , Proteínas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/metabolismo , Adamantano/administração & dosagem , Adamantano/farmacologia , Animais , Células Cultivadas , Dipeptídeos/administração & dosagem , Dipeptidil Peptidase 4/genética , Dipeptidil Peptidase 4/metabolismo , Inibidores da Dipeptidil Peptidase IV/administração & dosagem , Células Endoteliais/metabolismo , Humanos , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Interferência de RNA , Ratos Sprague-Dawley , Sirtuína 1/metabolismo
12.
Shock ; 53(6): 761-771, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31318834

RESUMO

BACKGROUND: Sepsis is a life-threatening organ dysfunction initiated by a dysregulated response to infection, with imbalanced inflammation and immune homeostasis. Macrophages play a pivotal role in sepsis. N-[1-(1-oxopropyl)-4-piperidinyl]-N'-[4-(trifluoromethoxy)phenyl)-urea (TPPU) is an inhibitor of soluble epoxide hydrolase (sEH), which can rapidly hydrolyze epoxyeicosatrienoic acids (EETs) to the bio-inactive dihydroxyeicosatrienoic acids. TPPU was linked with the regulation of macrophages and inflammation. Here, we hypothesized that sEH inhibitor TPPU ameliorates cecal ligation and puncture (CLP)-induced sepsis by regulating macrophage functions. METHODS: A polymicrobial sepsis model induced by CLP was used in our study. C57BL/6 mice were divided into four groups: sham+ phosphate buffer saline (PBS), sham+TPPU, CLP+PBS, CLP+TPPU. Mice were observed 48 h after surgery to assess the survival rate. For other histological examinations, mice were sacrificed 6 h after surgery. Macrophage cell line RAW264.7 was used for in vitro studies. RESULTS: TPPU treatment, accompanied with increased EETs levels, markedly improved the survival of septic mice induced by CLP surgery, which was associated with alleviated organ damage and dysfunction triggered by systemic inflammatory response. Moreover, TPPU treatment significantly inhibited systemic inflammatory response via EETs-induced inactivation of mitogen-activated protein kinase signaling due to enhanced macrophage phagocytic ability and subsequently reduced bacterial proliferation and dissemination, and decreased inflammatory factors release. CONCLUSION: sEH inhibitor TPPU ameliorates cecal ligation and puncture-induced sepsis by regulating macrophage functions, including improved phagocytosis and reduced inflammatory response. Our data indicate that sEH inhibition has potential therapeutic effects on polymicrobial-induced sepsis.


Assuntos
Inibidores Enzimáticos/uso terapêutico , Epóxido Hidrolases/antagonistas & inibidores , Macrófagos/efeitos dos fármacos , Compostos de Fenilureia/uso terapêutico , Piperidinas/uso terapêutico , Sepse/tratamento farmacológico , Animais , Ceco/lesões , Modelos Animais de Doenças , Citometria de Fluxo , Inflamação/tratamento farmacológico , Inflamação/etiologia , Ligadura , Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fagocitose , Sepse/mortalidade
13.
Clin Cardiol ; 43(3): 235-241, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31777973

RESUMO

The optimal antiplatelet therapy after percutaneous coronary intervention (PCI) remains to be elucidated. Monotherapy with a P2Y12 inhibitor may be inferior to dual antiplatelet therapy in patients after PCI. PubMed, EMBASE (by Ovidsp), Web of Science, and The Cochrane Library were searched from database inception to 2 October 2019. The composite of cardiovascular outcomes, all-cause mortality, myocardial infarction (MI), stroke, stent thrombosis, and major bleeding were evaluated. Pooled outcomes were presented as relative risk (RR) and 95% confidence intervals (CIs). A total of four trials randomizing 29 089 participants were included. Compared with the dual antiplatelet therapy group (n = 14 559), the P2Y12 inhibitor monotherapy group (n = 14 530) significantly decreased the incidence of bleeding events (2.0% vs 3.1%; RR: 0.60; 95% CI: 0.43-0.84; P = .005). There were no significant differences in all-cause mortality (1.3% vs 1.5%; RR: 0.87; 95% CI, 0.71-1.06; P = .16), myocardial infarction (2.1% vs 1.9%; RR, 1.06; 95% CI, 0.90-1.25; P = .46), stroke (0.6% vs 0.5%; RR, 1.18; 95% CI, 0.67-2.07; P = .57), or stent thrombosis (0.5% vs 0.4%; RR, 1.14; 95% CI, 0.81-1.61; P = .44) between the two groups. P2Y12 inhibitor monotherapy did not show any significant difference in the adverse cardiac and cerebrovascular events, but markedly decreased the risk of bleeding among patients after PCI vs dual antiplatelet therapy. However, it still needs to be further confirmed due to limited data.


Assuntos
Plaquetas/efeitos dos fármacos , Doença da Artéria Coronariana/terapia , Intervenção Coronária Percutânea , Inibidores da Agregação Plaquetária/uso terapêutico , Antagonistas do Receptor Purinérgico P2Y/uso terapêutico , Receptores Purinérgicos P2Y12/efeitos dos fármacos , Idoso , Plaquetas/metabolismo , Doença da Artéria Coronariana/diagnóstico , Doença da Artéria Coronariana/mortalidade , Trombose Coronária/mortalidade , Trombose Coronária/prevenção & controle , Feminino , Hemorragia/induzido quimicamente , Humanos , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/mortalidade , Infarto do Miocárdio/prevenção & controle , Intervenção Coronária Percutânea/efeitos adversos , Intervenção Coronária Percutânea/instrumentação , Intervenção Coronária Percutânea/mortalidade , Inibidores da Agregação Plaquetária/efeitos adversos , Antagonistas do Receptor Purinérgico P2Y/efeitos adversos , Ensaios Clínicos Controlados Aleatórios como Assunto , Receptores Purinérgicos P2Y12/sangue , Medição de Risco , Fatores de Risco , Stents , Acidente Vascular Cerebral/mortalidade , Acidente Vascular Cerebral/prevenção & controle , Fatores de Tempo , Resultado do Tratamento
14.
FASEB J ; 33(7): 8436-8452, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30995868

RESUMO

Cardiac aging is characterized by myocardial hypertrophy, fibrosis, and diastolic dysfunction. Human kallikrein (hKLK1) protects against fibrosis in various pathogenic states. However, the effects of hKLK1 overexpression on cardiac aging-related fibrosis and the underlying mechanisms remain unknown. Moreover, the role of hKLK1 in regulating macrophage function leading to cardiac fibrosis has not been investigated. Thus, in this study, we determined the effects of hKLK1 on cardiac aging and explored the mechanisms through which hKLK1 regulated aging-related fibrosis. Echocardiographic measurements showed that aging caused significant alternations in cardiac morphology, hypertrophy, and fibrosis in rats, and hKLK1 overexpression protected against aging-induced cardiac dysfunction. Compared with wild-type hearts, the hKLK1 transgene decreased the expression of monocyte chemoattractant protein 1 and suppressed mitochondrial dysfunction and excess oxidative stress, leading to decreased recruitment and retention of alternatively activated (M2) macrophages and reduced secretion of profibrotic cytokines mediated by the TGF-ß1-Smad3 signaling pathway in hearts of aging rats. Furthermore, these cardioprotective effects of hKLK1 overexpression were associated with the Janus kinase-signal transducer and activator of transcription 3 signaling pathway. H2O2-induced senescence promoted the differentiation of RAW264.7 cells into M2-type cells induced by IL-4 treatment. Bradykinin treatment relieved the migratory capacity of macrophages induced by H2O2. Thus, hKLK1 overexpression reduced cardiac fibrosis and improved aging-related cardiac dysfunction through reduced shift of macrophages to M2 macrophages, indicating that hKLK1 may alleviate aging-related cardiac dysfunction.-Hu, D., Dong, R., Yang, Y., Chen, Z., Tang, Y., Fu, M., Wang, D. W., Xu, X., Tu, L. Human kallikrein overexpression alleviates cardiac aging by alternatively regulating macrophage polarization in aged rats.


Assuntos
Envelhecimento/metabolismo , Envelhecimento/fisiologia , Coração/fisiologia , Calicreínas/metabolismo , Macrófagos/metabolismo , Macrófagos/fisiologia , Animais , Linhagem Celular , Fibrose/metabolismo , Humanos , Ativação de Macrófagos/fisiologia , Masculino , Camundongos , Miocárdio/metabolismo , Estresse Oxidativo/fisiologia , Células RAW 264.7 , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Transdução de Sinais/fisiologia , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
15.
Aging Cell ; 17(2)2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29318723

RESUMO

Ample evidences demonstrate that cytochrome P450 epoxygenase-derived epoxyeicosatrienoic acids (EETs) exert diverse biological activities, which include potent vasodilatory, anti-inflammatory, and cardiovascular protective effects. In this study, we investigated the effects of endothelium-specific CYP2J2 overexpression on age-related insulin resistance and metabolic dysfunction. Endothelium-specific targeting of the human CYP epoxygenase, CYP2J2, transgenic mice (Tie2-CYP2J2-Tr mice) was utilized. The effects of endothelium-specific CYP2J2 overexpression on aging-associated obesity, inflammation, and peripheral insulin resistance were evaluated by assessing metabolic parameters in young (3 months old) and aged (16 months old) adult male Tie2-CYP2J2-Tr mice. Decreased insulin sensitivity and attenuated insulin signaling in aged skeletal muscle, adipose tissue, and liver were observed in aged adult male mice, and moreover, these effects were partly inhibited in 16-month-old CYP2J2-Tr mice. In addition, CYP2J2 overexpression-mediated insulin sensitization in aged mice was associated with the amelioration of inflammatory state. Notably, the aging-associated increases in fat mass and adipocyte size were only observed in 16-month-old wild-type mice, and CYP2J2 overexpression markedly prevented the increase in fat mass and adipocyte size in aged Tie2-CYP2J2-Tr mice, which was associated with increased energy expenditure and decreased lipogenic genes expression. Furthermore, these antiaging phenotypes of Tie2-CYP2J2-Tr mice were also associated with increased muscle blood flow, enhanced active-phase locomotor activity, and improved mitochondrial dysfunction in skeletal muscle. Collectively, our findings indicated that endothelium-specific CYP2J2 overexpression alleviated age-related insulin resistance and metabolic dysfunction, which highlighted CYP epoxygenase-EET system as a potential target for combating aging-related metabolic disorders.


Assuntos
Sistema Enzimático do Citocromo P-450/biossíntese , Inflamação/enzimologia , Resistência à Insulina , Tecido Adiposo/enzimologia , Tecido Adiposo/metabolismo , Fatores Etários , Animais , Citocromo P-450 CYP2J2 , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Eicosanoides/metabolismo , Humanos , Inflamação/genética , Insulina/metabolismo , Insulina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Esquelético/enzimologia , Músculo Esquelético/metabolismo
16.
Oncotarget ; 8(31): 50958-50971, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28881620

RESUMO

Integrity of endothelial barrier is a determinant of the prognosis in the acute lung injury caused by sepsis. The epoxyeicosatrienoic acids (EETs), metabolites of arachidonic acid, exhibit protective effects in various pathogenic states, however, whether EETs play a role in endothelial barrier enhancement and the involved mechanisms remain to be investigated. Here, we show that increased EETs level by endothelial specific cytochrome P450 epoxygenase 2J2 over-expression and soluble epoxide hydrolase (sEH) inhibitor TPPU reduced lipopolysaccharide-induced endothelial hyper-permeability in vivo, accompanied by improved survival of septic mice. In addition, sEH inhibitor AUDA and 11,12-EET also decreased endothelial hyper-permeability in the in-vitro study. Importantly, the relative mechanisms were associated with reduced GRP78-Src interaction and ROS production, and subsequently reduced RhoA/ROCK activation, and eventually decreased VE-cadherin and myosin light chain (MLC) phosphorylation. Thus CYP2J2-EETs is crucial for RhoA-dependent regulation of cytoskeletal architecture leading to reversible changes in vascular permeability, which may contribute to the development of new therapeutic approaches for pulmonary edema and other diseases caused by abnormal vascular permeability.

17.
Cell Physiol Biochem ; 42(2): 761-779, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28628912

RESUMO

BACKGROUND/AIMS: Sepsis is a common disease that continues to increase in prevalence worldwide, and diabetes mellitus may make the situation worse. This study was designed to determine the role of Liver Kinase B1 (LKB1)/adenosine monophosphate-activated protein kinase (AMPK) signaling pathway in diabetic mice complicated with systemic endotoxemia. METHODS: The effects of LKB1/AMPK signaling pathway activation on endotoxemia were investigated in streptozotocin induced diabetic mice (STZ-mice) and db/db diabetic mice. Primary peritoneal macrophages and human umbilical vein endothelial cells (HUVECs) monolayers were simultaneously stimulated by both high glucose and LPS and used as a model to investigate the potential molecular mechanisms in vitro. RESULTS: After treatment with LPS, high glucose or both LPS and high glucose, phosphor-AMPK expression was decreased, and moreover, AMPK activation by metformin treatment alleviated the decrease in phosphor-AMPK expression in HUVECs and macrophages as well as in lung tissue. Furthermore, both LPS and high glucose co-treatment decreased LKB1 and phosphor-AMPK expression via enhanced oxidative stress response, and importantly, LKB1 overexpression mediated by adenovirus inhibited the decrease in phosphor-AMPK expression in macrophages and HUVECs. AMPK activation by metformin administration improved the survival of STZ-induced diabetic mice and db/db diabetic mice, which was associated with reduced lung endothelial hyperpermeability and systemic inflammatory response. Furthermore, the permeability of HUVECs monolayers induced by both high glucose and LPS stimulation was also alleviated by AMPK activation, which was partly via suppression of VE-cadherin phosphorylation. CONCLUSION: These data demonstrated that LKB1/AMPK signaling pathway activation improved the survival of diabetic mice complicated with endotoxemia. Thus, LKB1/AMPK signaling pathway may serve as a potentially useful therapeutic target for severe infection in diabetic patients.


Assuntos
Proteínas Quinases Ativadas por AMP/genética , Diabetes Mellitus Experimental/genética , Endotoxemia/genética , Proteínas Serina-Treonina Quinases/genética , Animais , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/patologia , Progressão da Doença , Endotoxemia/patologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Hipoglicemiantes/administração & dosagem , Fígado/metabolismo , Fígado/patologia , Metformina/administração & dosagem , Camundongos , Camundongos Endogâmicos NOD/genética , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA