Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Cell Mol Med ; 28(14): e18558, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39048917

RESUMO

Myocardial ischemia-reperfusion injury (MIRI) represents a critical pathology in acute myocardial infarction (AMI), which is characterized by high mortality and morbidity. Cardiac microvascular dysfunction contributes to MIRI, potentially culminating in heart failure (HF). Pigment epithelium-derived factor (PEDF), which belongs to the non-inhibitory serpin family, exhibits several physiological effects, including anti-angiogenesis, anti-inflammatory and antioxidant properties. Our study aims to explore the impact of PEDF and its functional peptide 34-mer on both cardiac microvascular perfusion in MIRI rats and human cardiac microvascular endothelial cells (HCMECs) injury under hypoxia reoxygenation (HR). It has been shown that MIRI is accompanied by ferroptosis in HCMECs. Furthermore, we investigated the effect of PEDF and its 34-mer, particularly regarding the Nrf2/HO-1 signalling pathway. Our results demonstrated that PEDF 34-mer significantly ameliorated cardiac microvascular dysfunction following MIRI. Additionally, they exhibited a notable suppression of ferroptosis in HCMECs, and these effects were mediated through activation of Nrf2/HO-1 signalling. These findings highlight the therapeutic potential of PEDF and 34-mer in alleviating microvascular dysfunction and MIRI. By enhancing cardiac microvascular perfusion and mitigating endothelial ferroptosis, PEDF and its derivative peptide represent promising candidates for the treatment of AMI.


Assuntos
Células Endoteliais , Proteínas do Olho , Ferroptose , Traumatismo por Reperfusão Miocárdica , Fator 2 Relacionado a NF-E2 , Fatores de Crescimento Neural , Serpinas , Transdução de Sinais , Serpinas/farmacologia , Serpinas/metabolismo , Fatores de Crescimento Neural/farmacologia , Fatores de Crescimento Neural/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Animais , Ferroptose/efeitos dos fármacos , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/patologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , Proteínas do Olho/metabolismo , Proteínas do Olho/farmacologia , Transdução de Sinais/efeitos dos fármacos , Ratos , Heme Oxigenase-1/metabolismo , Masculino , Ratos Sprague-Dawley , Microvasos/efeitos dos fármacos , Microvasos/metabolismo , Microvasos/patologia , Peptídeos/farmacologia
2.
Int J Mol Sci ; 24(3)2023 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-36769107

RESUMO

Maintaining the integrity and protecting the stability of tight junctions in endothelial cells is a potential therapeutic strategy against myocardial ischaemia. Laminin receptors (67LR) are highly expressed on endothelial cell membranes and are associated with endothelial barrier function. Herein, we sought to demonstrate the direct effects of pigment epithelial-derived factor (PEDF) on tight junctions between endothelial cells via 67LR during acute myocardial infarction (AMI) and elucidate its underlying mechanisms. We detected that PEDF directly increased the level of the tight junction protein zonula occludens protein 1 (ZO-1) after overexpression in vitro and in vivo using Western blotting. Evans Blue/TTC staining showed that PEDF significantly reduced the size of the infarcted myocardium. Immunofluorescence and the transwell cellular experiments suggested that PEDF significantly upregulated PI3K-AKT permeability and the distribution of ZO-1 between endothelial cells under OGD conditions. Interestingly, PEDF significantly upregulated the phosphorylation levels of PI3K-AKT-mTOR under oxygen and glucose deprivation conditions but had no significant effects on the total protein expression. The protective effect of PEDF on ZO-1 was significantly inhibited following the inhibition of PI3K-AKT-mTOR. The activation of phosphorylation of PI3K-AKT-mTOR by PEDF was blocked after silencing 67LR, as were the protective effects of PEDF on ZO-1. Therefore, we have reason to believe that PEDF increased ZO-1 expression through the 67LR-dependent PI3K-AKT-mTOR signaling pathway, thus maintaining tight junction stability and protecting cardiac function.


Assuntos
Infarto do Miocárdio , Proteínas Proto-Oncogênicas c-akt , Humanos , Células Endoteliais/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Junções Íntimas/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo , Receptores de Laminina/metabolismo
3.
Respir Res ; 23(1): 100, 2022 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-35459189

RESUMO

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a highly heterogeneous and fatal lung disease. In addition to dense fibrous tissue, abnormal angiogenesis is also an important feature of IPF. Pigment epithelium-derived factor (PEDF) is an angiogenesis inhibitor and a potential anti-fibrous factor. The purpose of this experiment is to observe the effect of PEDF on bleomycin (BLM)-induced pulmonary fibrosis in rats. METHODS: In vivo, pathological examination and detection of related factors were performed on pulmonary fibrosis induced by BLM in rats, and the temporal and spatial distribution of PEDF was investigated. Furthermore, lung gene delivery (PEDF-adeno-associated virus) was performed to investigate the effect of PEDF on pulmonary fibrosis. In vitro, lentiviral vectors were used to construct PEDF over-expression or knock out primary rat lung (PRL) fibroblasts. The effect of PEDF on fibroblast activation under TGF-ß1 stimulation was evaluated, and the activation of TGF-ß1/smad pathway and PPAR-γ expression (in the presence or absence of PPAR-γ inhibitors) were analyzed. RESULTS: In vivo results showed that PEDF expression decreased during the inflammatory phase and increased during the fibrotic phase. PEDF could inhibit the progression of pulmonary fibrosis in rats. In vitro results showed that PEDF could effectively inhibit TGF-ß1-stimulated fibroblast activation and reduce the production of α-SMA and collagen-I. PEDF could inhibit the TGF-ß1/smad pathway by up-regulating the activity of PPAR-γ. CONCLUSIONS: PEDF can act as an anti-fibrotic factor, inhibit fibroblast activation by upregulating PPAR-γ activity and reduce BLM-induced pulmonary fibrosis in rats.


Assuntos
Bleomicina , Fibrose Pulmonar Idiopática , Animais , Bleomicina/toxicidade , Proteínas do Olho , Fibroblastos/metabolismo , Fibrose , Fibrose Pulmonar Idiopática/metabolismo , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural , Receptores Ativados por Proliferador de Peroxissomo/efeitos adversos , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Ratos , Serpinas , Fator de Crescimento Transformador beta1/farmacologia
4.
Angiogenesis ; 24(4): 809-821, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34028626

RESUMO

The loss of normal alveolar capillary and deregulated angiogenesis occurs simultaneously in idiopathic pulmonary fibrosis (IPF); however the contributions of specific endothelial subpopulations in the development of pulmonary fibrosis are poorly understood. Herein, we perform single-cell RNA sequencing to characterize the heterogeneity of endothelial cells (ECs) in bleomycin (BLM)-induced lung fibrosis in rats. One subpopulation, characterized by the expression of Nos3 and Cav1, is mostly distributed in non-fibrotic lungs and also highly expresses genes related to the "response to mechanical stimulus" and "lung/heart morphogenesis" processes. Another subpopulation of ECs expanded in BLM-treated lungs, characterized by Cxcl12, is observed to be closely related to the pro-fibrotic process in the transcriptome data, such as "regulation of angiogenesis," "collagen binding," and "chemokine activity," and spatially localized to BLM-induced neovascularization. Using CellPhoneDB software, we generated a complex cell-cell interaction network, which predicts the potential roles of EC subpopulations in recruiting monocytes, inducing the proliferation of fibroblasts and promoting the production and remolding of the extracellular matrix (ECM). Taken together, our data demonstrate the high degree of heterogeneity of ECs in fibrotic lung and it is proposed that the interaction between ECs, macrophages, and stromal cells contributes to pathologic IPF.


Assuntos
Bleomicina , Fibrose Pulmonar Idiopática , Animais , Bleomicina/toxicidade , Células Endoteliais , Fibroblastos , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/genética , Pulmão , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Análise de Sequência de RNA
5.
J Cell Mol Med ; 22(11): 5732-5742, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30230261

RESUMO

Both decreased autophagy positive regulator AMP activated protein kinase (AMPK) level and promoted mitophagy are observed in oxygen-glucose deprivation (OGD) cardiomyocytes treated with pigment epithelium-derived factor (PEDF). This contradictory phenomenon and its underlying mechanisms have not been thoroughly elucidated. Our previous study reveals that PEDF increases the protein kinase Cα (PKCα) and phospho-PKCα (p-PKCα) contents to promote mitophagy. Thus, we investigated the association between PKCα and mitophagy. Here we identify an interaction between PKCα and Unc-51-like kinase 1 (ULK1), essential component of mitophagy. Further analyses show this is a direct interaction within a domain of ULK1 that termed the serine/threonine-rich domain (S/T domain). Notably, a deletion mutant ULK1 that lacks the binding domain is defective in mediating PEDF-induced mitophagy. Furthermore, we demonstrate that ULK1 is phosphorylated at Ser317/555/777 and Raptor is also phosphorylated by phospho-PKCα. Phospho-ULK1 (p-ULK1) at these sites are all essential for PEDF-induced mitophagy and reduce the release of mitochondrial ROS and DNA. This study therefore identifies a previously uncharacterized interaction between the ULK1 and PKCα that can replace the AMPK-dependent mitophagy processes.


Assuntos
Proteína Homóloga à Proteína-1 Relacionada à Autofagia/genética , Proteínas do Olho/genética , Isquemia Miocárdica/genética , Fatores de Crescimento Neural/genética , Proteína Quinase C-alfa/genética , Serpinas/genética , Proteínas Quinases Ativadas por AMP/genética , Animais , Autofagia/genética , Ventrículos do Coração/citologia , Ventrículos do Coração/patologia , Humanos , Mitocôndrias/genética , Mitofagia/genética , Isquemia Miocárdica/patologia , Isquemia Miocárdica/prevenção & controle , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fosforilação , Cultura Primária de Células , Ratos , Proteína Regulatória Associada a mTOR/genética
6.
Zhongguo Fei Ai Za Zhi ; 25(2): 71-77, 2022 Feb 20.
Artigo em Zh | MEDLINE | ID: mdl-35224959

RESUMO

BACKGROUND: Immunoneoadjuvant therapy opens a new prospect for local advanced lung cancer. The aim of our study was to explore the safety and feasibility of robotic-assisted bronchial sleeve resection in patients with locally advanced non-small cell lung cancer (NSCLC) after neoadjuvant chemoimmunotherapy. METHODS: Data of 13 patients with locally advanced NSCLC that underwent bronchial sleeve resection after neoadjuvant chemoimmunotherapy during August 2020 and February 2021 were retrospectively included. According to the surgical methods, patients were divided into thoracotomy bronchial sleeve resection (TBSR) group and robot-assisted bronchial sleeve resection (RABSR) group. Oncology, intraoperative, and postoperative data in the two groups were compared. RESULTS: The two groups of patients operated smoothly, the postoperative pathology confirmed that all the tumor lesions achieved R0 resection, and RABSR group no patient was transferred to thoracotomy during surgery. Partial remission (PR) rate and major pathological remissions (MPR) rate of patients in the TBSR group were 71.43% and 42.86%, respectively. Complete pathological response (pCR) was 28.57%. They were 66.67%, 50.00% and 33.33% in RABSR group, respectively. There were no significant differences in operative duration, number of lymph nodes dissected, intraoperative blood loss, postoperative drainage time and postoperative hospital stay between the two groups, but the bronchial anastomosis time of RABSR group was relatively short. Both groups of patients had a good prognosis. Successfully discharged from the hospital and post-operative 90-d mortality rate was 0. CONCLUSIONS: In patients with locally advanced central NSCLC after neoadjuvant chemoimmunotherapy can achieve the tumor reduction, tumor stage decline and increase the R0 resection rate, bronchial sleeve resection is safe and feasible. Under the premise of following the two principles of surgical safety and realizing the tumor R0 resection, robot-assisted bronchial sleeve resection can be preferred.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Robótica , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/cirurgia , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/cirurgia , Terapia Neoadjuvante , Pneumonectomia/métodos , Estudos Retrospectivos , Toracotomia , Resultado do Tratamento
7.
Int J Biol Sci ; 18(4): 1521-1538, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35280692

RESUMO

The occurrence and development of acute lung injury (ALI) involve a variety of pathological factors and complex mechanisms. How pulmonary cells communicate with each other and subsequently trigger an inflammatory cascade remains elusive. Extracellular vesicles (EVs) are a critical class of membrane-bound structures that have been widely investigated for their roles in pathophysiological processes, especially in immune responses and tumor progression. Most of the current knowledge of the functions of EVs is related to functions derived from viable cells (e.g., microvesicles and exosomes) or apoptotic cells (e.g., apoptotic bodies); however, there is limited understanding of the rapidly progressing inflammatory response in ALI. Herein, a comprehensive analysis of micron-sized EVs revealed a mass production of 1-5 µm pyroptotic bodies (PyrBDs) release in the early phase of ALI induced by lipopolysaccharide (LPS). Alveolar macrophages were the main source of PyrBDs in the early phase of ALI, and the formation and release of PyrBDs were dependent on caspase-1. Furthermore, PyrBDs promoted the activation of epithelial cells, induced vascular leakage and recruited neutrophils through delivery of damage-associated molecular patterns (DAMPs). Collectively, these findings suggest that PyrBDs are mainly released by macrophages in a caspase-1-dependent manner and serve as mediators of LPS-induced ALI.


Assuntos
Lesão Pulmonar Aguda , Vesículas Extracelulares , Lesão Pulmonar Aguda/induzido quimicamente , Caspase 1 , Humanos , Inflamação , Lipopolissacarídeos/toxicidade , Pulmão , Macrófagos Alveolares/patologia
8.
J Thorac Cardiovasc Surg ; 161(6): e417-e434, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32359908

RESUMO

OBJECTIVE: To select a suitable combination of classic angiogenic and vascular stabilization factors to improve the proliferation and maturity of neovascularization of lung tissue in a rat pulmonary arterial hypertension (PAH) model. METHODS: PAH rat model was established by intraperitoneal injection of monocrotaline. Proangiogenic factors hepatocyte growth factor (HGF) and vascular endothelial growth factor (VEGF), as well as vascular stabilization factors angiopoietin-1 (Ang-1), platelet-derived growth factor, and transforming growth factor-beta were transfected by pairs into the lung tissue of rats with PAH through lentivirus. Four weeks later, pulmonary artery angiography and hemodynamic parameters were determined to testify the remission of PAH. Immunofluorescence staining and Western blot were performed to investigate the structure and function of neovascularization. RESULTS: The pulmonary artery pressure and weight index of the right ventricle in HGF+Ang-1 and VEGF+Ang-1 groups were significantly decreased compared with vehicle group. The contrast medium filling time and right pulmonary artery root diameter were also significantly decreased. In addition, the maturity and perfusion of neovascularization in HGF+Ang-1 and VEGF+Ang-1 groups were promoted compared to vehicle group, and vascular leakage was reduced. Finally, the adherens junction integrity of vascular endothelial cells in HGF+Ang-1 and VEGF+Ang-1 combinations was upregulated compared with other combinations. CONCLUSIONS: HGF+Ang-1 transfection and VEGF+Ang-1 transfection alleviate PAH by promoting maturation and stability of new blood vessels, which may be potential candidates for PAH treatment.


Assuntos
Hipertensão Pulmonar/fisiopatologia , Pulmão , Neovascularização Fisiológica/efeitos dos fármacos , Artéria Pulmonar/fisiopatologia , Animais , Fator de Crescimento de Hepatócito/farmacologia , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Masculino , Monocrotalina , Ratos , Ratos Sprague-Dawley , Fator A de Crescimento do Endotélio Vascular/farmacologia
9.
Thorac Cancer ; 12(20): 2796-2802, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34463034

RESUMO

BACKGROUND: There are many studies on neoadjuvant immunotherapy for locally advanced non-small cell lung cancer (NSCLC) patients. Expert consensus recommends neoadjuvant immunotherapy for patients with resectable stage IB-IIIA NSCLC. However, there are few clinical studies or cases to verify this. METHODS: Data were collected from all NSCLC patients who underwent surgical resection after neoadjuvant chemoimmunotherapy admitted to the Affiliated Hospital of Xuzhou Medical University and Xuzhou Central Hospital between September 2020 and April 2021. Data collected included patient information, relevant examination results, intraoperative parameters, postoperative complications, pathological changes, and 90-day mortality. RESULTS: In total, 25 patients achieved R0 resection. Eleven (44%) patients completed surgery by thoracotomy, and three (12%) procedures were changed from minimally invasive procedures due to dense adhesions of hilar lymph nodes, which rendered it difficult to dissect the blood vessels. Thirteen (52%) patients achieved a major pathological response (MPR) with eight (32%) of these patients having a pathological complete response (pCR). Twenty-two (88%) patients showed radiological regression, and three (12%) patients had stable disease. The median drainage time was 8.50 (3-27) days. Thirteen (52%) postoperative complications were observed, but none were above grade 3. CONCLUSIONS: In this study, neoadjuvant chemoimmunotherapy was found to reduce tumor volume, cause pathological downstaging, and raise the surgical resection rate of patients with locally advanced NSCLC, and achieve a 100% R0 resection rate. There was an acceptable rate of postoperative complications. Thus, neoadjuvant chemoimmunotherapy is safe and practical.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/cirurgia , Imunoterapia/métodos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/cirurgia , Terapia Neoadjuvante/métodos , Idoso , Idoso de 80 Anos ou mais , Terapia Combinada , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Complicações Pós-Operatórias
10.
Int J Cardiol ; 283: 136-143, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30819588

RESUMO

BACKGROUND: Targeted increase in glucose uptake of ischemic myocardium is a potential therapeutic strategy for myocardial ischemia. PEDF presents a profound moderating effect on glucose metabolism of cells, but its role is still controversial. Here, we try to demonstrate the direct effect of PEDF on glucose uptake in ischemic myocyte and to elucidate its underlying mechanism. METHODS AND RESULTS: Lentivirus vectors carrying PEDF gene were delivered into the myocardium to locally overexpress PEDF in a myocardial ischemia/reperfusion rat model. PET imaging showed that PEDF local overexpression increased [18F]-FDG uptake of ischemic myocardium. In vitro, PEDF directly increased the glucose uptake in hypoxic cardiomyocytes. The expression of glucose transporter 4 (GLUT4) on plasma membrane of hypoxic cardiomyocytes was significantly upregulated by PEDF, but its total amount was not changed. The increased glucose uptake and cardioprotective effects induced by PEDF were blocked by the GLUT4 inhibitor indinavir. PEDF-mediated GLUT4 translocation and glucose uptake increase in hypoxic cardiomyocytes were prevented by phosphatidyl-inositol-3 kinase (PI3K) inhibitor or AKT inhibitor. The PEDF-mediated glucose uptake was also diminished when PEDF receptor (PEDFR) was downregulated or potent phospholipase A2 enzymatic activity was inhibited. CONCLUSIONS: PEDF can increase glucose uptake in ischemic myocardium through a PEDFR-dependent mechanism, involving PI3K/AKT signaling and GLUT4 translocation.


Assuntos
Proteínas do Olho/genética , Regulação da Expressão Gênica , Glucose/metabolismo , Isquemia Miocárdica/genética , Miocárdio/metabolismo , Fatores de Crescimento Neural/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serpinas/genética , Animais , Transporte Biológico , Western Blotting , DNA/genética , Modelos Animais de Doenças , Proteínas do Olho/biossíntese , Isquemia Miocárdica/diagnóstico , Isquemia Miocárdica/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fatores de Crescimento Neural/biossíntese , Tomografia por Emissão de Pósitrons/métodos , Ratos , Ratos Sprague-Dawley , Serpinas/biossíntese , Transdução de Sinais
11.
Hum Gene Ther ; 30(6): 762-776, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30734585

RESUMO

The failure to maintain the viability of ischemic myocardium is one of the mechanisms that causes ischemic heart dysfunction after revascularization. Hibernating myocardium is considered to be able to maintain long-term viability during chronic hypoperfusion. Pigment epithelium-derived factor (PEDF) decreases the contractility of hypoxic cardiomyocytes and protects cardiomyocytes against ischemic injury, which is strikingly similar to the pathophysiologic characteristics of hibernating myocardium. It was therefore postulated that PEDF may induce acute ischemic myocardium into a "hibernating-like" state to maintain its viability. Adult Sprague-Dawley rat models of acute myocardial infarction were surgically established. Lentiviral vectors carrying the PEDF gene (PEDF-LVs) were delivered into myocardium with infarction to overexpress PEDF locally. It was found that PEDF local overexpression significantly reduced myocardial infarct size and cardiomyocytes necrosis but did not improve cardiac function at rest. The contractile reserve assessed by low-dose dobutamine stress echocardiography and "perfusion-metabolism mismatch" assessed by positron emission tomography, which are the characteristics of viable myocardium in hibernation, were observed in the PEDF overexpressed ischemic heart. Ultrastructural changes observed by electron microscopy and glycogen deposition explored by Periodic acid-Schiff staining were similar to the histological characteristics of hibernating myocardium. Moreover, PEDF overexpression protected the cardiomyocytes against anoxic injury and retained their functional recovery potential after reoxygenation in vitro. PEDF local overexpression may induce acute ischemic myocardium into a "hibernating-like" state and maintain its viability. This novel effect of PEDF presents an important clinical approach to enhance functional recovery after revascularization therapy in acute myocardial infarction.


Assuntos
Proteínas do Olho/genética , Expressão Gênica , Vetores Genéticos/genética , Lentivirus/genética , Isquemia Miocárdica/genética , Fatores de Crescimento Neural/genética , Serpinas/genética , Animais , Biomarcadores , Modelos Animais de Doenças , Genes Reporter , Terapia Genética , Glicogênio/metabolismo , Testes de Função Cardíaca , Hibernação/genética , Masculino , Contração Miocárdica/genética , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Isquemia Miocárdica/diagnóstico , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/terapia , Miocárdio/metabolismo , Miocárdio/patologia , Miocárdio/ultraestrutura , Necrose/genética , Necrose/metabolismo , Tomografia por Emissão de Pósitrons , Ratos , Ratos Sprague-Dawley
12.
J Am Heart Assoc ; 8(5): e011220, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30819021

RESUMO

Background We occasionally noticed that native collateral blood flow showed a recessive trend in the early stages of acute myocardial infarction in rats, which greatly interferes with the accurate assessment of native collateral circulation levels. Here, we sought to recognize the coronary collateral circulation system in depth, especially the microcirculation part, on this basis. Methods and Results In this study, we detected native collateral flow with positron emission tomography perfusion imaging in rats and found that the native flow is relatively abundant when it is initially recruited. However, this flow is extremely unstable in the early stage of acute myocardial infarction and quickly fails. We used tracers to mark the collateral in an ischemic area and a massive preformed collateral network was labeled. The ultrastructures of these collateral microvessels are flawed, which contributes to extensive leakage and consequent interstitial edema in the ischemic region. Conclusions An unrecognized short-lived native coronary collateral microcirculation reserve is widely distributed in rat hearts. Recession of collateral blood flow transported by coronary collateral microcirculation reserve contributes to instability of native collateral blood flow in the early stage of acute myocardial infarction. The immature structure determines that these microvessels are short-lived and provide conditions for the development of early interstitial edema in acute myocardial infarction.


Assuntos
Circulação Colateral , Circulação Coronária , Vasos Coronários/fisiopatologia , Microcirculação , Microvasos/fisiopatologia , Infarto do Miocárdio/fisiopatologia , Animais , Permeabilidade Capilar , Células Cultivadas , Vasos Coronários/diagnóstico por imagem , Vasos Coronários/ultraestrutura , Modelos Animais de Doenças , Edema Cardíaco/diagnóstico por imagem , Edema Cardíaco/patologia , Edema Cardíaco/fisiopatologia , Masculino , Microvasos/diagnóstico por imagem , Microvasos/ultraestrutura , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/patologia , Imagem de Perfusão do Miocárdio , Tomografia por Emissão de Pósitrons , Ratos Sprague-Dawley , Fatores de Tempo
13.
Int J Mol Med ; 43(5): 1979-1990, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30864707

RESUMO

Myocardial edema is divided into cellular edema and interstitial edema; however, the dynamic change of cardiomyocyte edema has not been described in detail. Pigment epithelium­derived factor (PEDF) is known for its protective effects on ischemic cardiomyocytes; however, the association between PEDF and cardiomyocyte edema remains to be fully elucidated. In the present study, rat neonatal left ventricular cardiomyocytes were isolated and treated with oxygen­glucose deprivation (OGD) and recovery. During OGD and recovery, the cardiomyocytes exhibited significant edema following 30 min of OGD (OGD 30 min) and OGD 30 min with recovery for 6 h. PEDF significantly decreased the lactate content and extracellular acidification rate of the OGD­treated cardiomyocytes, thereby reducing cellular osmotic gradients and preventing the occurrence of cell edema. In addition, the glycolytic agonist, fructose­1, 6­diphosphate, eliminated the effect of PEDF on inhibiting edema in the OGD­treated cardiomyocytes. Furthermore, PEDF reduced the protein and mRNA expression of aquaporin 1 (AQP1), and thus downregulated cardiomyocyte edema during the OGD/recovery period. The addition of AQP1 agonist, arginine vasopressin, inhibited the inhibitory effect of PEDF on cardiomyocyte edema during OGD/recovery. In conclusion, the present study revealed a novel mechanism for the regulation of cardiomyocyte edema by PEDF involving lactate levels and the expression of AQP1 during OGD/recovery. The reduction of lactate content during OGD was associated with a decrease in the protein level of AQP1 during OGD/recovery; therefore, PEDF decreased cardiomyocyte edema and cellular apoptosis, prolonging the viability of the cells.


Assuntos
Aquaporina 1/metabolismo , Edema/patologia , Proteínas do Olho/farmacologia , Glucose/deficiência , Ácido Láctico/metabolismo , Miócitos Cardíacos/patologia , Fatores de Crescimento Neural/farmacologia , Oxigênio/metabolismo , Serpinas/farmacologia , Animais , Cardiotônicos/farmacologia , Glicólise/efeitos dos fármacos , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Ratos Sprague-Dawley , Sódio/metabolismo
14.
J Am Heart Assoc ; 8(22): e013323, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31718448

RESUMO

Background We previously found that the structural defects of the coronary collateral microcirculation reserve (CCMR) prevent these preformed collateral vessels from continuously delivering the native collateral blood and supporting the ischemic myocardium in rats. Here, we tested whether these native collaterals can be remodeled by artificially increasing pigment epithelium-derived factor (PEDF) expression and demonstrated the mechanism for this stimulation. Methods and Results We performed intramyocardial gene delivery (PEDF-lentivirus, 2×107 TU) along the left anterior descending coronary artery to artificially increase the expression of PEDF in the tissue of the region for 2 weeks. By blocking the left anterior descending coronary artery, we examined the effects of PEDF on native collateral blood flow and CCMR. The results of positron emission tomography perfusion imaging showed that PEDF increased the native collateral blood flow and significantly inhibited its decline during acute myocardial infarction. In addition, the number of CCMR vessels decreased and the size increased. Similar results were obtained from in vitro experiments. We tested whether PEDF induces CCMR remodeling in a fluid shear stress-like manner by detecting proteins and signaling pathways that are closely related to fluid shear stress. The nitric oxide pathway and the Notch-1 pathway participated in the process of CCMR remodeling induced by PEDF. Conclusions PEDF treatment activates the nitric oxide pathway, and the Notch-1 pathway enabled CCMR remodeling. Increasing the native collateral blood flow can promote the ventricular remodeling process and improve prognosis after acute myocardial infarction.


Assuntos
Aterosclerose/genética , Circulação Colateral/genética , Vasos Coronários/fisiopatologia , Proteínas do Olho/genética , Infarto do Miocárdio/fisiopatologia , Fatores de Crescimento Neural/genética , Serpinas/genética , Remodelação Vascular/genética , Remodelação Ventricular/genética , Animais , Circulação Colateral/fisiologia , Células Endoteliais/efeitos dos fármacos , Proteínas do Olho/farmacologia , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Lentivirus , Microcirculação/genética , Microcirculação/fisiologia , Isquemia Miocárdica/fisiopatologia , Reperfusão Miocárdica , Miocárdio , Fatores de Crescimento Neural/farmacologia , Óxido Nítrico/metabolismo , Tomografia por Emissão de Pósitrons , Ratos , Receptor Notch1/metabolismo , Serpinas/farmacologia , Estresse Mecânico
15.
Int J Mol Med ; 41(6): 3243-3252, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29532859

RESUMO

The prevention and management of myocardial ischemia/reperfusion (MI/R) injury is an essential part of coronary heart disease surgery and is becoming a major clinical problem in the treatment of ischemic heart disease. Previous studies by our group have demonstrated that pigment epithelium­derived factor (PEDF) improves cardiac function in rats with acute myocardial infarction and reduces hypoxia­induced cell injury. However, the protective function and mechanisms underlying the effect of PEDF in MI/R injury remain to be fully understood. In the present study, the positive effect of PEDF in MI/R injury was confirmed by construction of the adult Sprague­Dawley rat MI/R model. PEDF reduced myocardial infarct size and downregulated cardiomyocyte apoptosis in the I/R myocardium in this model. In addition, PEDF improved cardiac function and increased cardiac functional reserve in rats subjected to MI/R Injury. To further study the protective effect of PEDF and the underlying mechanisms in MI/R injury, a H9c2 cardiomyocyte hypoxia/reoxygenation (H/R) model was constructed. PEDF was confirmed to decrease H/R­induced apoptosis in H9c2 cells, and this anti­apoptotic function was abolished by pigment epithelium­derived factor­receptor (PEDF R) small interfering (si)RNA. Furthermore, administration of PEDF decreased the levels of reactive oxygen species (ROS) and malondialdehyde (MDA) in H/R H9c2 cells. Compared with the H/R group, PEDF decreased mitochondrial ROS, increased the mitochondrial DNA copy number, reduced xanthine oxidase and NADPH oxidase activity, as well as RAC family small GTPase 1 protein expression. However, these effects of PEDF were markedly attenuated by PEDF­R siRNA. To the best of our knowledge, the present study is the first to identify the protective effect of PEDF in MI/R injury, and confirm that the antioxidative effect PEDF occurred via inhibition of ROS generation via PEDF­R under MI/R conditions.


Assuntos
Proteínas do Olho/uso terapêutico , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio/metabolismo , Fatores de Crescimento Neural/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Serpinas/uso terapêutico , Animais , Western Blotting , Linhagem Celular , Citometria de Fluxo , Marcação In Situ das Extremidades Cortadas , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
16.
Int J Cardiol ; 257: 262-271, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29361350

RESUMO

AIMS: The activated AMP activated protein kinase (AMPK) serves as a transient protective cardiovascular kinase via preserving adenosine triphosphate (ATP) production under ischemic conditions. However, recent studies reveal that inhibition of AMPK in stroke is neuroprotection. Pigment epithelium derived factor (PEDF) is also known for the protection of ischemic cardiomyocytes. However, the relationship between PEDF and AMPK in cardiomyocytes is poorly understood. METHODS AND RESULTS: Rat neonatal and adult left ventricular cardiomyocytes were isolated and subjected to oxygen-glucose deprivation (OGD). During OGD, PEDF significantly reduced AMPKα levels to decrease ATP production and reduced ATP expenditure both in neonatal and adult cardiomyocytes, which increased energy reserves and cell viability. Importantly, pharmacological AMPK inhibitor reduced ATP production but failed to decrease ATP expenditure, thus leading cells into death. Furthermore, AMPKα was degraded by a ubiquitin-dependent proteasomal degradation pathway, which is associated with a PEDF/PEDFR/peroxisome proliferator activated receptor γ (PPARγ) axis. Inhibition of PPARγ or proteasome disrupted the interaction of AMPKα and PPARγ, which abolished AMPKα degradation. Importantly, the decrease of AMPKα and ATP level was normalized after recovery of oxygen and glucose. CONCLUSIONS: We demonstrate a novel mechanism for regulation of cardiac ATP production by PEDF involving AMPKα and PPARγ. PEDF promotes proteasomal degradation of AMPK and, subsequently, reduces ATP production. The reduction of ATP production associated with the decrease of ATP expenditure completed by PEDF increase energy reserves and reduces cell energy failure, prolonging the cell activity during OGD.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Trifosfato de Adenosina/metabolismo , Proteínas do Olho/metabolismo , Glucose/deficiência , Miócitos Cardíacos/metabolismo , Fatores de Crescimento Neural/metabolismo , Oxigênio/metabolismo , Serpinas/metabolismo , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Trifosfato de Adenosina/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Proteínas do Olho/farmacologia , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Fatores de Crescimento Neural/farmacologia , Ratos , Ratos Sprague-Dawley , Serpinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA