Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Heart Fail Rev ; 29(1): 95-111, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37768435

RESUMO

This review article offers a detailed examination of metabolic adaptations in pressure overload hypertrophic hearts, a condition that plays a pivotal role in the progression of heart failure with preserved ejection fraction (HFpEF) to heart failure with reduced ejection fraction (HFrEF). The paper delves into the complex interplay between various metabolic pathways, including glucose metabolism, fatty acid metabolism, branched-chain amino acid metabolism, and ketone body metabolism. In-depth insights into the shifts in substrate utilization, the role of different transporter proteins, and the potential impact of hypoxia-induced injuries are discussed. Furthermore, potential therapeutic targets and strategies that could minimize myocardial injury and promote cardiac recovery in the context of pressure overload hypertrophy (POH) are examined. This work aims to contribute to a better understanding of metabolic adaptations in POH, highlighting the need for further research on potential therapeutic applications.


Assuntos
Insuficiência Cardíaca , Humanos , Insuficiência Cardíaca/metabolismo , Metabolismo Energético , Volume Sistólico , Cardiomegalia/metabolismo
3.
Cell Biol Int ; 47(5): 943-953, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36934412

RESUMO

The activity of phosphatase and tensin homolog (PTEN) can be inhibited by miR-17-3p, which results in attenuating myocardial ischemia/reperfusion injury (IRI), however, the mechanism behind this phenomenon is still elusive. Suppression of PTEN leads to augmented protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling strength and constrained autophagy activation, which might be the one mechanism for the ameliorated myocardial IRI. Thus, we tested the hypothesis that miR-17-3p attenuated hypoxia/reoxygenation (H/R)-mediated damage in cardiomyocytes by downregulating excessive autophagy via the PTEN-Akt-mTOR axis. The expression of miR-17-3p was remarkably increased after H/R treatment (6-h hypoxia followed by 6-h reoxygenation; H6/R6), which was concomitant with the increase of the release of lactic acid dehydrogenase (cell injury marker) and the enhancement LC3II/I ratio (autophagy markers) in H9C2 cardiomyocytes. Ectoexpression of miR-17-3p agomir led to remarkable augmentation of miR-17-3p expression and evidently attenuated H/R-mediated cell damage and excessive autophagy. Furthermore, an increase in miR-17-3p expression elicited constrained phosphorylation of PTEN (Ser380 ) while enhanced the phosphorylation of Akt (Thr308 , Ser473 ) and mTOR (Ser536 ) after H/R stimulation. In addition, pretreatment with LY-294002 (an Akt selective inhibitor) and rapamycin (an mTOR selective inhibitor) significantly abrogated the protective function of miR-17-3p on H/R-mediated cell damage and autophagy in H9C2 cardiomyocytes. Taken together, these observations indicated that the enhancement of the PTEN/Akt/mTOR axis and the consequent suppression of autophagy overactivation might represent an underlying mechanism by which miR-17-3p attenuated H/R-mediated damage in H9C2 cells.


Assuntos
MicroRNAs , Proteínas Proto-Oncogênicas c-akt , Animais , Proteínas Proto-Oncogênicas c-akt/metabolismo , Miócitos Cardíacos/metabolismo , Linhagem Celular , MicroRNAs/metabolismo , Apoptose , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Mamíferos/metabolismo , Hipóxia/metabolismo , Autofagia
4.
BMC Immunol ; 23(1): 58, 2022 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-36434505

RESUMO

Propofol is widely used in clinical anesthesia due to its advantages of rapid onset and less adverse reactions. This study focused on the role of propofol in the balance of Th17/Treg in elderly patients with lung cancer during perioperative period. Patients undergoing lung cancer surgery were anesthetized by propofol or sevoflurane. Veinal blood was collected at different time points to evaluate the changes of Th17/Treg cell. Propofol better maintained the balance of Th17/Treg in vivo. The peripheral blood of patients with lung cancer was collected in vitro before surgery. Cluster of differentiation (CD)4+ T cells were obtained and then treated with propofol at different concentrations and γ-aminobutyric acid A (GABAA) receptor antagonists. Propofol affected Th17/Treg cell balance by increasing Th17 cells, decreasing Treg cells, thus elevating Th17/Treg ratio, and inhibited invasion and migration of lung cancer cells through GABAA receptor, which was counteracted by GABAA receptor inhibitors. Subsequently, tumor in situ model of lung cancer in aged mice was established. Propofol anesthetized mice had lower change of Th17/Treg ratio, higher survival rate and less metastasis. In brief, propofol regulated balance of Th17/Treg in elderly patients undergoing lung cancer surgery through GABAA receptor. Additionally, propofol could inhibit metastasis of lung cancer.


Assuntos
Neoplasias Pulmonares , Propofol , Animais , Camundongos , Células Th17 , Linfócitos T Reguladores , Propofol/farmacologia , Receptores de GABA-A , Receptores de GABA , Neoplasias Pulmonares/cirurgia
5.
Int J Mol Med ; 48(1)2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34036377

RESUMO

Propofol is a commonly used intravenous anesthetic. The aim of the study was to examine the mechanism of propofol in traumatic brain injury (TBI) by regulating interleukin (IL)­17 activity and maintaining the Th17/Treg balance. A rat model with moderate TBI was established using the weight­drop method. Rats with TBI were regularly injected with propofol and their brain injuries were monitored. The peripheral blood of rats was collected to measure the Th17/Treg ratio. MicroRNA (miR)­145­3p expression was detected in the brain tissues of rats and antagomiR­145­3p was injected into the lateral ventricles of their brains to verify the effect of miR­145­3p on brain injury. The downstream target of miR­145­3p was predicted. The targeting relationship between miR­145­3p and nuclear factor of activated T cells c2 (NFATc2) was confirmed. NFATC2 expression and phosphorylation of NF­κB pathway­related proteins were measured. Propofol alleviated brain injury in rats with TBI and maintained the Th17/Treg balance. Propofol upregulated miR­145­3p expression in rat brains, while the inhibition of miR­145­3p reversed the effect of propofol on brain injury. A binding relationship was observed between miR­145­3p and NFATc2. Furthermore, propofol decreased the phosphorylation of p65 and IκBα, and inhibited activation of the NF­κB pathway in the brains of rats with TBI. In conclusion, propofol maintained Th17/Treg balance and reduced inflammation in the rats with TBI via the miR­145­3p/NFATc2/NF­κB axis.


Assuntos
Lesões Encefálicas Traumáticas , MicroRNAs/imunologia , NF-kappa B/imunologia , Fatores de Transcrição NFATC/imunologia , Propofol/farmacologia , Transdução de Sinais/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Animais , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/imunologia , Inflamação/tratamento farmacológico , Inflamação/imunologia , Masculino , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/imunologia
6.
Acta Diabetol ; 58(9): 1251-1267, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33907874

RESUMO

AIMS: Diabetic cardiomyopathy (DCM) is a specific myocardial alteration in patients with diabetics. LncRNA KCNQ1OT1 has been previously demonstrated to be involved in various diabetic complications. Our aims are to further investigate the underlying regulatory mechanisms/pathways of KCNQ1OT1 in DCM. METHODS: In vitro and in vivo models of DCM were established in high glucose (HG)-treated human cardiomyocytes and in streptozotocin (STZ)-induced diabetic mice, respectively. Gene and protein expressions were examined by qPCR, western blotting and ELISA. Cell proliferation and apoptosis were determined by CCK8 assay, flow cytometry and TUNEL staining. The association between KCNQ1OT1 and miR-181a-5p, miR-181a-5p and PDCD4 was predicted using bioinformatics methods and subsequently confirmed by dual luciferase reporter and RNA immunoprecipitation assays. Mouse cardiac tissues were collected and analysed using HE staining, Masson's staining and immunohistochemical analysis. RESULTS: KCNQ1OT1 and PDCD4 were upregulated in HG-treated human cardiomyocytes, while miR-181a-5p was downregulated. In addition, KCNQ1OT1 could negatively regulate miR-181a-5p expression; meanwhile, miR-181a-5p also negatively regulated PDCD4 expression. KCNQ1OT1 silencing suppressed the expression of inflammatory cytokines and cell apoptosis in vitro, whereas inhibition of miR-181a-5p abrogated those effects of KCNQ1OT1 knockdown. Moreover, overexpressed PDCD4 abolished the inhibition on inflammation and apoptosis caused by miR-181a-5p overexpression. Finally, KCNQ1OT1 knockdown reduced the expression of PDCD4 via regulating miR-181a-5p and inhibited myocardial inflammation and cardiomyocyte apoptosis in the in vivo DCM model. CONCLUSIONS: Our findings suggest that KCNQ1OT1 and its target gene miR-181a-5p regulate myocardial inflammation and cardiomyocyte apoptosis by modulating PDCD4 in DCM.


Assuntos
Diabetes Mellitus Experimental , Cardiomiopatias Diabéticas , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Animais , Apoptose/genética , Proteínas Reguladoras de Apoptose/genética , Diabetes Mellitus Experimental/genética , Cardiomiopatias Diabéticas/genética , Humanos , Camundongos , MicroRNAs/genética , Miócitos Cardíacos , RNA Longo não Codificante , Proteínas de Ligação a RNA/genética
7.
Theranostics ; 11(10): 4710-4727, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33754023

RESUMO

Background: Telomere shortening and dysfunction may cause metabolic disorders, tissue damage and age-dependent pathologies. However, little is known about the association of telomere-associated protein Rap1 with mitochondrial energy metabolism and cardiac aging. Methods: Echocardiography was performed to detect cardiac structure and function in Rap1+/+ and Rap1-/- mice at different ages (3 months, 12 months and 20 months). Telomere length, DNA damage, cardiac senescence and cardiomyocyte size were analyzed using the real-time PCR, Western blotting, senescence associated ß-galactosidase assay and wheat germ agglutinin staining, respectively. Western blotting was also used to determine the level of cardiac fatty acid metabolism related key enzymes in mouse and human myocardium. Chromatin immunoprecipitation assay was used to verify the direct link between p53 and PPARα. The p53 inhibitor, Pifithrin-α and PPARα activator WY14643 were utilized to identify the effects of Rap1/p53/PPARα signaling pathway. Results: Telomere was shortened concomitant with extensive DNA damage in aged Rap1-/- mouse hearts, evidenced by reduced T/S ratios and increased nuclear γH2AX. Meanwhile, the aging-associated phenotypes were pronounced as reflected by altered mitochondrial ultrastructure, enhanced senescence, cardiac hypertrophy and dysfunction. Mechanistically, acetylated p53 and nuclear p53 was enhanced in the Rap1-/- mouse hearts, concomitant with reduced PPARα. Importantly, p53 directly binds to the promoter of PPARα in mouse hearts and suppresses the transcription of PPARα. In addition, aged Rap1-/- mice exhibited reduced cardiac fatty acid metabolism. Pifithrin-α alleviated cardiac aging and enhanced fatty acid metabolism in the aged Rap1-/- mice. Activating PPARα with WY14643 in primarily cultured Rap1-/- cardiomyocytes restored maximal oxygen consumption rates. Reduced Rap1 expression and impaired p53/PPARα signaling also presented in aged human myocardium. Conclusion: In summary, Rap1 may link telomere biology to fatty acid metabolism and aging-related cardiac pathologies via modulating the p53/PPARα signaling pathway, which could represent a therapeutic target in preventing/attenuating cardiac aging.


Assuntos
Envelhecimento/genética , Cardiomegalia/genética , Senescência Celular/genética , Miócitos Cardíacos/metabolismo , PPAR alfa/genética , Proteínas de Ligação a Telômeros/genética , Proteína Supressora de Tumor p53/genética , Animais , Benzotiazóis/farmacologia , Cardiomegalia/diagnóstico por imagem , Cardiomegalia/fisiopatologia , Dano ao DNA , Ecocardiografia , Ácidos Graxos/metabolismo , Cardiopatias/diagnóstico por imagem , Cardiopatias/genética , Cardiopatias/fisiopatologia , Histonas/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/ultraestrutura , Teste de Campo Aberto , PPAR alfa/metabolismo , Proliferadores de Peroxissomos/farmacologia , Pirimidinas/farmacologia , Complexo Shelterina , Transdução de Sinais , Telômero/metabolismo , Homeostase do Telômero , Proteínas de Ligação a Telômeros/metabolismo , Tolueno/análogos & derivados , Tolueno/farmacologia , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo
8.
Oxid Med Cell Longev ; 2020: 6454281, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33082912

RESUMO

BACKGROUND: Cerebral ischemic stroke is a refractory disease which seriously endangers human health. Remote ischemic perconditioning (RiPerC) by which the sublethal ischemic stimulus is administered during the ischemic event is beneficial after an acute stroke. However, the regulatory mechanism of RiPerC that relieves cerebral ischemic injury is still not completely clear. METHODS: In the present study, we investigated the regulatory mechanism of RiPerC in a rat model of ischemia induced by the middle cerebral artery occlusion (MCAO). Forty-eight adult male Sprague-Dawley (SD) rats were injected intracerebroventricularly with miR-98 agomir, miR-98 antagomir, or their negative controls (agomir-NC, antagomir-NC) 2 h before MCAO or MCAO+RiPerC followed by animal behavior tests and infraction volume measurement at 24 h after MCAO. The expression of miR-98, PIK3IP1, and tight junction proteins in rat hippocampus and cerebral cortex tissues was detected by quantitative polymerase chain reaction (qPCR) and Western blot (WB). Enzyme-linked immunosorbent assay (ELISA) was used to assess the IL-1ß, IL-6, and TNF-α levels in the rat serum. RESULTS: The results showed that in MCAO group, the expression of PIK3IP1 was upregulated, but decreased after RiPerC treatment. Then, we found that PIK3IP1 was a potential target of miR-98. Treatment with miR-98 agomir decreased the infraction volume, reduced brain edema, and improved neurological functions compared to control rats. But treating with miR-98 antagomir in RiPerC group, the protective effect on cerebral ischemia injury was canceled. CONCLUSION: Our finding indicated that RiPerC inhibited the MCAO-induced expression of PIK3IP1 through upregulated miR-98, thereby reducing the apoptosis induced by PIK3IP1 through the PI3K/AKT signaling pathway, thus reducing the cerebral ischemia-reperfusion injury.


Assuntos
Isquemia Encefálica/patologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Precondicionamento Isquêmico , MicroRNAs/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Regiões 3' não Traduzidas , Animais , Antagomirs/metabolismo , Antagomirs/uso terapêutico , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/etiologia , Córtex Cerebral/metabolismo , Modelos Animais de Doenças , Células HEK293 , Hipocampo/metabolismo , Humanos , Infarto da Artéria Cerebral Média/complicações , Interleucina-1beta/análise , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
9.
Nan Fang Yi Ke Da Xue Xue Bao ; 40(9): 1313-1318, 2020 Sep 30.
Artigo em Chinês | MEDLINE | ID: mdl-32990232

RESUMO

OBJECTIVE: To explore the effect of pretreatment of neuroblastoma cells with hot water extract of Korean ginseng on MNNG-induced parthanatos and its mechanism. METHODS: Neuroblastoma SH-SY5Y cells were pretreated with 1 mg/L hot water extract of Korean ginseng before induction with 250 µmol/L MNNG for 1 h or 4 h. CCK-8 and cell flow cytometry were used to detect cell survival rate. Western blotting was used to detect the changes in poly(ADP-ribose) (PAR) expression in the treated cells. Immunofluorescence assay was used to detect nuclear distribution of apoptosis-inducing factor (AIF), and flow cytometry was used to detect the level of reactive oxygen species (ROS) in the cells. RESULTS: Compared with the blank control cells, MNNG-treated SH-SY5Y cells showed significantly decreased survival rate as the concentration of MNNG and the stimulation time increased (P < 0.05). Stimulation with MNNG also resulted in significantly increased expression of PAR protein in the cells (P < 0.05). Pretreatment of the cells with hot water extract of Korean ginseng obviously inhibited MNNG-induced cell death and significantly reduced AIF expression and nucleation in the cells (P < 0.05). MNNG stimulation significantly increased ROS level in the cells, which was decreased significantly by pretreatment of the cells with the extract (P < 0.05). CONCLUSIONS: Pretreatment with hot water extract of Korean ginseng reduces MNNG-induced parthanatos and ROS production in SH-SY5Y cells.


Assuntos
Neuroblastoma , Panax , Fator de Indução de Apoptose/metabolismo , Humanos , Panax/metabolismo , Parthanatos , República da Coreia
10.
Oxid Med Cell Longev ; 2020: 7913418, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32774684

RESUMO

BACKGROUND: Constitutive nuclear factor kappa B (NFκB) activation has been shown to exacerbate during myocardial ischemia/reperfusion (I/R) injury. We recently showed that miR-181c-5p exacerbated cardiomyocytes injury and apoptosis by directly targeting the 3'-untranslated region of protein tyrosine phosphatase nonreceptor type 4 (PTPN4). However, whether miR-181c-5p mediates cardiac I/R injury through NFκB-mediated inflammation is unknown. Thus, the present study aimed to investigate the role of miR-181c-5p during myocardial I/R injury and explore its mechanism in relation to inflammation in H9C2 cardiomyocytes. METHODS AND RESULTS: In hypoxia/reoxygenation (H/R, 6 h hypoxia followed by 6 h reoxygenation)-stimulated H9C2 cardiomyocytes or postischemic myocardium of rat, the expression of miR-181c-5p was significantly upregulated, which was concomitant increased NFκB activity when compared to the nonhypoxic or nonischemic control groups. This is indicative that miR-181c-5p may be involved in NFκB-mediated inflammation during myocardial I/R injury. To investigate the potential role of miR-181c-5p in H/R-induced cell inflammation and injury, H9C2 cardiomyocytes were transfected with the miR-181c-5p agomir. Overexpression of miR-181c-5p significantly aggravated H/R-induced cell injury (increased lactate dehydrogenase (LDH) level) and exacerbated NFκB-mediated inflammation (greater phosphorylation and degradation of IκBα, phosphorylation of p65, and increased levels of proinflammatory cytokines tumor necrosis factor α (TNFα), interleukin (IL)-6, and IL-1ß). In contrast, inhibition of miR-181c-5p by its antagomir transfection in vitro had the opposite effect. Furthermore, overexpression of miR-181c-5p significantly enhanced lipopolysaccharide-induced NFκB signalling. Additionally, knockdown of PTPN4, the direct target of miR-181c-5p, significantly aggravated H/R-induced phosphorylation and degradation of IκBα, phosphorylation of p65, and the levels of proinflammatory cytokines. PTPN4 knockdown also cancelled miR-181c-5p antagomir mediated anti-inflammatory effects in H9C2 cardiomyocytes during H/R injury. CONCLUSIONS: It is concluded that miR-181c-5p may exacerbate myocardial I/R injury and NFκB-mediated inflammation via PTPN4, and that targeting miR-181c-5p/PTPN4/NFκB signalling may represent a novel strategy to combat myocardial I/R injury.


Assuntos
Hipóxia Celular/fisiologia , Inflamação/metabolismo , MicroRNAs/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Animais , Regulação para Baixo , Humanos , Masculino , Ratos , Ratos Sprague-Dawley
11.
Biosci Trends ; 14(3): 222-226, 2020 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-32321903

RESUMO

The new coronavirus (COVID-19) has been characterized as a world pandemic by WHO since March 11, 2020. Although it is likely that COVID-19 transmission is primarily via droplets and close contact, airborne transmission and fecal-oral route remains a possibility. The medical staff working in the operating room, such as anesthesiologists, surgeons and nurses, are at high risk of exposure to virus due to closely contacting patients. The perioperative management is under great challenge while performing surgeries for patients suffering COVID-19, including emergency cesarean section, which is one of the most common surgeries under such circumstances. How to prevent medical staff from cross-infection is an issue of great concern. In this article, we give a practice of anesthesia scenario design for emergency cesarean section in a supposed standard patient suffering COVID-19, aimed to optimize the work flow and implement the protective details through simulation of a real operation scenario, which may be useful for training and clinical practice of anesthesia management for patients suffering COVID-19 or other fulminating infectious diseases.


Assuntos
Anestesia , Cesárea , Infecções por Coronavirus , Controle de Infecções/métodos , Pandemias , Pneumonia Viral , Complicações Infecciosas na Gravidez , Betacoronavirus , COVID-19 , Tratamento de Emergência , Feminino , Humanos , Gravidez , SARS-CoV-2
12.
FASEB J ; 34(3): 4482-4496, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32020680

RESUMO

Ischemic heart disease is a leading cause of morbidity and mortality. Repressor activator protein 1 (Rap1), an established telomere-associated protein, is a novel modulator of hypoxia-induced apoptosis. This study aimed to explore the potential direct role of Rap1 in myocardial ischemia/reperfusion injury (I/RI) and to determine the underlying molecular mechanism. In a mouse model of myocardial I/RI (30-min of left descending coronary artery ligation followed by 2-h reperfusion), Rap1 deficiency significantly reduced myocardial infarct size (IS) and improved cardiac systolic/diastolic function. This was associated with a reduction in apoptosis in the post-ischemic myocardium. In H9C2 and primary cardiomyocytes, Rap1 knockdown or knockout significantly suppressed hypoxia/reoxygenation (H/R)-induced cell injury and apoptosis through increasing the phosphorylation/activation of STAT3 at site Ser727 and translocation of STAT3 to the nucleus. We surmise this since Stattic (selective STAT3 inhibitor) pretreatment canceled the abovementioned protective effect. Furthermore, co-immunoprecipitation assay revealed a direct interaction between Rap1 and STAT3, but not JAK2, suggesting that the association of Rap1 with STAT3 may contribute to the reduced activity of STAT3 (Ser727 ) upon H/R stimulation. In conclusion, Rap1 deficiency protects the heart from ischemic damage through STAT3-dependent reduction of cardiomyocyte apoptosis, which may yield viable target for pharmacological intervention in ischemic heart disease.


Assuntos
Sobrevivência Celular/fisiologia , L-Lactato Desidrogenase/metabolismo , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/terapia , Fator de Transcrição STAT3/metabolismo , Proteínas rap1 de Ligação ao GTP/metabolismo , Animais , Apoptose/genética , Apoptose/fisiologia , Western Blotting , Linhagem Celular , Sobrevivência Celular/genética , Ecocardiografia , Humanos , Imunoprecipitação , Marcação In Situ das Extremidades Cortadas , L-Lactato Desidrogenase/genética , Masculino , Camundongos Endogâmicos C57BL , Isquemia Miocárdica/genética , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Fator de Transcrição STAT3/genética , Proteínas rap1 de Ligação ao GTP/genética
13.
J Diabetes Investig ; 11(5): 1126-1136, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32064785

RESUMO

AIMS/INTRODUCTION: Diabetic hearts are more vulnerable to ischemia-reperfusion injury (I/RI). The activation of nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome can mediate the inflammatory process, and hence might contribute to myocardial I/RI. Activation of autophagy can eliminate excess reactive oxygen species and alleviate myocardial I/RI in diabetes. The present study aimed to investigate whether the activation of autophagy can alleviate diabetic myocardial I/RI through inhibition of NLRP3 inflammasome activation. MATERIALS AND METHODS: A dose of 65 mg/kg streptozotocin was given by tail vein injection to establish a type 1 diabetes model in the rats. The left anterior descending coronary artery was ligated for 30 min followed by reperfusion for 2 h to establish a myocardial I/RI model. H9C2 cardiomyocytes were exposed to high glucose (33 mmol/L) and subjected to hypoxia-reoxygenation (6 h hypoxia followed by 4 h reoxygenation). RESULTS: The diabetic rats showed significant inhibition of cardiac autophagy (decreased LC3-II/I and increased p62) that was concomitant with increased activation of NLRP3 inflammasome (increased NLRP3, apoptosis-related spots protein cleaved caspase-1, interleukin-18, interleukin-1ß) and more severe myocardial I/RI (elevated creatine kinase myocardial band, lactate dehydrogenase and larger infarct size). However, administration of rapamycin, an inhibitor of the autophagy, to activate autophagy resulted in the inhibition of NLRP3 inflammasome, and finally alleviated myocardial I/RI. In vitro, high glucose inhibited autophagy, while activating NLRP3 inflammasome in H9C2 cardiomyocytes and aggravating hypoxia-reoxygenation injury, but rapamycin reversed these adverse effects of high glucose. CONCLUSION: Activation of autophagy can suppress the formation of NLRP3 inflammasome, which in turn attenuates myocardial ischemia-reperfusion injury in diabetic rats.


Assuntos
Autofagia , Diabetes Mellitus Experimental/complicações , Inflamassomos/metabolismo , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miócitos Cardíacos/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Animais , Apoptose , Inflamassomos/genética , Masculino , Traumatismo por Reperfusão Miocárdica/etiologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
14.
Front Mol Neurosci ; 13: 598811, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33384583

RESUMO

Chronic stress exposure increases the risk of developing various neuropsychiatric illnesses. The ventral hippocampus (vHPC) is central to affective and cognitive processing and displays a high density of acetylcholine (ACh) muscarinic receptors (mAChRs). However, the precise role of vHPC mAChRs in anxiety remains to be fully investigated. In this study, we found that chronic restraint stress (CRS) induced social avoidance and anxiety-like behaviors in mice and increased mAChR expression in the vHPC. CRS increased the vHPC ACh release in behaving mice. Moreover, CRS altered the synaptic activities and enhanced neuronal activity of the vHPC neurons. Using pharmacological and viral approaches, we showed that infusing the antagonist of mAChRs or decreasing their expression in the vHPC attenuated the anxiety-like behavior and rescued the social avoidance behaviors in mice probably due to suppression of vHPC neuronal activity and its excitatory synaptic transmission. Our results suggest that the changes of neuronal activity and synaptic transmission in the vHPC mediated by mAChRs may play an important role in stress-induced anxiety-like behavior, providing new insights into the pathological mechanism and potential pharmacological target for anxiety disorders.

15.
Nan Fang Yi Ke Da Xue Xue Bao ; 39(7): 850-854, 2019 Jul 30.
Artigo em Chinês | MEDLINE | ID: mdl-31340920

RESUMO

OBJECTIVE: To investigate the effect of ulinastatin pretreatment on isoflurane-induced mitochondria-dependent neuronal apoptosis in the hippocampus of rats. METHODS: Thirty-six male SD rats were randomly assigned into control group, isoflurane group and ulinastatin group. In the latter two groups, the rats were subjected to acute exposure to 0.75% isoflurane for 6 h and pretreated with 50 000 U/kg of ulinastatin before isoflurane exposure, respectively. After the treatments, apoptosis of the hippocampal neurons was detected using TUNEL assay, and the mitochondrial membrane potential (△ ψm) was measured using JC-1 mitochondrial membrane potential kit; cytochrome C release and caspase-3 activity were examined with Western blotting, and intracellular reactive oxygen species (ROS) was detected using the fluorescent probe H2DCFDA. RESULTS: Compared with those in the control group, the rats with acute exposure to isoflurane showed markedly increased TUNEL-positive cells in the hippocampus (P < 0.05), which were obviously reduced by ulinastatin pretreatment (P < 0.05). The △ψm of the hippocampal neurons was significantly reduced after isoflurane exposure (P < 0.05), and was partly recovered by ulinastatin pretreatment (P < 0.05). Acute exposure to isoflurane resulted in obviously increased cellular ROS, cytochrome C release and caspase-3 activity in the hippocampal neurons (P < 0.05), and these changes were significantly inhibited by ulinastatin pretreatment (P < 0.05). CONCLUSIONS: Ulinastatin pretreatment provides neuroprotection against isoflurane-induced apoptosis of the hippocampal neurons in rats possibly by inhibiting mitochondria-dependent apoptosis pathway.


Assuntos
Apoptose , Animais , Glicoproteínas , Hipocampo , Isoflurano , Masculino , Ratos , Ratos Sprague-Dawley
16.
Oxid Med Cell Longev ; 2019: 1957920, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31178952

RESUMO

BACKGROUND: Activation of cell apoptosis is a major form of cell death during myocardial ischemia/reperfusion injury (I/RI). Therefore, examining ways to control cell apoptosis has important clinical significance for improving postischemic recovery. Clinical evidence demonstrated that miR-181c-5p was significantly upregulated in the early phase of myocardial infarction. However, whether or not miR-181c-5p mediates cardiac I/RI through cell apoptosis pathway is unknown. Thus, the present study is aimed at investigating the role and the possible mechanism of miR-181c-5p in apoptosis during I/R injury by using H9C2 cardiomyocytes. METHODS AND RESULTS: The rat origin H9C2 cardiomyocytes were subjected to hypoxia/reoxygenation (H/R, 6 hours hypoxia followed by 6 hours reoxygenation) to induce cell injury. The results showed that H/R significantly increased the expression of miR-181c-5p but not miR-181c-3p in H9C2 cells. In line with this, in an in vivo rat cardiac I/RI model, miR-181c-5p expression was also significantly increased. The overexpression of miR-181c-5p by its agomir transfection significantly aggravated H/R-induced cell injury (increased lactate dehydrogenase level and reduced cell viability) and exacerbated H/R-induced cell apoptosis (greater cleaved caspases 3 expression, Bax/Bcl-2 and more TUNEL-positive cells). In contrast, inhibition of miR-181c-5p in vitro had the opposite effect. By using computational prediction algorithms, protein tyrosine phosphatase nonreceptor type 4 (PTPN4) was predicted as a potential target gene of miR-181c-5p and was verified by the luciferase reporter assay. The overexpression of miR-181c-5p significantly attenuated the mRNA and protein expression of PTPN4 in H9C2 cardiomyocytes. Moreover, knockdown of PTPN4 significantly aggravated H/R-induced enhancement of LDH level, cleaved caspase 3 expression, and apoptotic cell death, which mimicked the proapoptotic effects of miR-181c-5p in H9C2 cardiomyocytes. CONCLUSIONS: These findings suggested that miR-181c-5p exacerbates H/R-induced cardiomyocyte injury and apoptosis via targeting PTPN4 and that miR-181c-5p/PTPN4 signaling may yield novel strategies to combat myocardial I/R injury.


Assuntos
Hipóxia Celular/fisiologia , MicroRNAs/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 4/metabolismo , Animais , Apoptose/fisiologia , Masculino , MicroRNAs/genética , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/patologia , Proteína Tirosina Fosfatase não Receptora Tipo 4/genética , Ratos , Ratos Sprague-Dawley , Transfecção
17.
Trials ; 20(1): 789, 2019 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-31888744

RESUMO

BACKGROUND: Cardiac surgery involving cardiopulmonary bypass (CPB) is known to be associated with a transient postoperative immunosuppression. When severe and persistent, this immune dysfunction predisposes patients to infectious complications, which contributes to a prolonged stay in the intensive care unit (ICU), and even mortality. Effective prevention and treatment methods are still lacking. Recent studies revealed that acupuncture-related techniques, such as electroacupuncture and transcutaneous electrical acupoint stimulation (TEAS), are able to produce effective cardioprotection and immunomodulation in adult and pediatric patients undergoing cardiac surgery with CPB, which leads to enhanced recovery. However, whether perioperative application of TEAS, a non-invasive technique, is able to improve immunosuppression of the patients with post-cardiosurgical conditions is unknown. Thus, as a preliminary study, the main objective is to evaluate the effects of TEAS on the postoperative expression of monocytic human leukocyte antigen (-D related) (mHLA-DR), a standardized "global" biomarker of injury or sepsis-associated immunosuppression, in patients receiving on-pump coronary artery bypass grafting (CABG). METHODS: This study is a single-center clinical trial. The 88 patients scheduled to receive CABG under CPB will be randomized into two groups: the group receiving TEAS, and the group receiving transcutaneous acupoint pseudo-electric stimulation (Sham TEAS). Expression of mHLA-DR serves as a primary endpoint, and other laboratory parameters (e.g., interleukin [IL]-6, IL-10) and clinical outcomes (e.g., postoperative infectious complications, ICU stay time, and mortality) as the secondary endpoints. In addition, immune indicators, such as high mobility group box 1 protein and regulatory T cells will also be measured. DISCUSSION: The current study is a preliminary monocentric clinical trial with a non-clinical primary endpoint, expression of mHLA-DR, aiming at determining whether perioperative application of TEAS has a potential to reverse CABG-associated immunosuppression. Although the immediate clinical impact of this study is limited, its results would inform further large-sample clinical trials using relevant patient-centered clinical outcomes as primary endpoints. TRIAL REGISTRATION: ClinicalTrials.gov, NCT02933996. Registered on 13 October 2016.


Assuntos
Pontos de Acupuntura , Ponte Cardiopulmonar , Ponte de Artéria Coronária , Antígenos HLA-DR/metabolismo , Monócitos/metabolismo , Período Perioperatório , Estimulação Elétrica Nervosa Transcutânea/métodos , Adolescente , Adulto , Idoso , Biomarcadores/metabolismo , Método Duplo-Cego , Feminino , Humanos , Unidades de Terapia Intensiva , Tempo de Internação , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento , Adulto Jovem
18.
Mol Pain ; 13: 1744806917701445, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28326940

RESUMO

Background: Mesenchymal stem cell (MSC) has been one of the potential tools in neuropathic pain therapy; however, the augmented efficacy may be expected when they are modified with human proenkephalin (hPPE) gene. In the current study, the antinociceptive effect of human bone marrow stem cells (hBMSCs) engineered with hPPE gene (hPPE-hBMSCs) on sciatic nerve chronic constriction injury (CCI)-induced neuropathic pain in rats was investigated. Methods: Primary-cultured hBMSCs were passaged and modified with hPPE, and the cell suspensions (6 × 106) were then intrathecally injected into a rat model of CCI. Paw mechanical withdrawal threshold and paw withdrawal thermal latency were measured before and after CCI surgery. The effects of hPPE gene transfer on hBMSCs bioactivity were analyzed in vitro and in vivo. Results: No changes were observed in the surface phenotypes and differentiation of hBMSCs after gene transfer. The hPPE-hBMSC group showed improved paw mechanical withdrawal threshold and paw thermal withdrawal latency values on the ipsilateral side of rats with CCI from day 9 post-surgery, and the analgesic effect was reversed by naloxone. Leucine-enkephalin (L-EK) secretion was augmented in the hPPE-engineered hBMSC group. Conclusions: The intrathecal administration of BMSCs modified with hPPE gene can effectively relieve pain caused by chronic constriction injury in rats and might be a potentially therapeutic tool for neuropathic pain in humans.


Assuntos
Encefalinas/genética , Células-Tronco Mesenquimais/citologia , Neuralgia/terapia , Dor Nociceptiva/terapia , Precursores de Proteínas/genética , Animais , Humanos , Injeções Espinhais/métodos , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Neuralgia/metabolismo , Limiar da Dor/efeitos dos fármacos , Ratos Sprague-Dawley , Medula Espinal/metabolismo , Medula Espinal/cirurgia
19.
Pain Res Manag ; 2017: 7346103, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28286408

RESUMO

Background. This study aimed to investigate the use of human bone marrow mesenchymal stem cells (hBMSCs) genetically engineered with the human proenkephalin (hPPE) gene to treat bone cancer pain (BCP) in a rat model. Methods. Primary cultured hBMSCs were passaged and modified with hPPE, and the cell suspensions (6 × 106) were then intrathecally injected into a rat model of BCP. Paw mechanical withdrawal threshold (PMWT) was measured before and after BCP. The effects of hPPE gene transfer on hBMSC bioactivity were analyzed in vitro and in vivo. Results. No changes were observed in the surface phenotypes and differentiation of hBMSCs after gene transfer. The hPPE-hBMSC group showed improved PMWT values on the ipsilateral side of rats with BCP from day 12 postoperatively, and the analgesic effect was reversed by naloxone. The levels of proinflammatory cytokines such as IL-1ß and IL-6 were ameliorated, and leucine-enkephalin (L-EK) secretion was augmented, in the hPPE-engineered hBMSC group. Conclusion. The intrathecal administration of BMSCs modified with the hPPE gene can effectively relieve pain caused by bone cancer in rats and might be a potentially therapeutic tool for cancer-related pain in humans.


Assuntos
Analgésicos/uso terapêutico , Células da Medula Óssea/metabolismo , Dor do Câncer/cirurgia , Encefalinas , Precursores de Proteínas , Transplante de Células-Tronco/métodos , Animais , Neoplasias Ósseas/complicações , Dor do Câncer/etiologia , Diferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Encefalinas/genética , Encefalinas/metabolismo , Encefalinas/uso terapêutico , Feminino , Terapia Genética , Vetores Genéticos , Humanos , Hiperalgesia/diagnóstico , Hiperalgesia/tratamento farmacológico , Injeções Espinhais , Naloxona/farmacologia , Limiar da Dor/fisiologia , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Precursores de Proteínas/uso terapêutico , Ratos , Ratos Sprague-Dawley
20.
Brain Res ; 1554: 29-35, 2014 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-24480471

RESUMO

Bone cancer pain is a major clinical problem and remains difficult to treat. ATP-sensitive potassium (KATP) channels may be involved in regulating nociceptive transmission at the spinal cord level. We determined the role of spinal KATP channels in the control of mechanical hypersensitivity in a rat model of bone cancer pain. The rat model of bone cancer pain was induced by implanting rat mammary gland carcinoma cells (Walker256) into the tibias. KATP modulators (pinacidil and glibenclamide) or the specific Kir6.2-siRNA were injected via an intrathecal catheter. The mechanical withdrawal threshold of rats was tested using von Frey filaments. The Kir6.2 mRNA and protein levels were measured by quantitative PCR and western blots, respectively. Intrathecal injection of pinacidil, a KATP channel opener, significantly increased the tactile withdrawal threshold of cancer cell-injected rats in a dose-dependent manner. In contrast, intrathecal delivery of glibenclamide, a KATP channel blocker, or the specific Kir6.2-siRNA significantly reduced the tactile withdrawal threshold of cancer cell-injected rats. The mRNA and protein levels of Kir6.2 in the spinal cord of cancer cell-injected rats were significantly lower than those in control rats. Our findings suggest that the KATP channel expression level in the spinal cord is reduced in bone cancer pain. Activation of KATP channels at the spinal level reduces pain hypersensitivity associated with bone cancer pain.


Assuntos
Neoplasias Ósseas/fisiopatologia , Nociceptividade/fisiologia , Dor/fisiopatologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Glibureto/farmacologia , Hiperalgesia/tratamento farmacológico , Hiperalgesia/fisiopatologia , Moduladores de Transporte de Membrana/farmacologia , Neoplasias Experimentais , Dor/metabolismo , Limiar da Dor/efeitos dos fármacos , Pinacidil/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Wistar , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Tato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA