Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Oncol ; 13: 1239124, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37681025

RESUMO

Objective: Uterine intravenous leiomyomatosis (IVL) is a rare and unique leiomyoma that is difficult to surgery due to its ability to extend into intra- and extra-uterine vasculature. And it is difficult to differentiate from uterine leiomyoma (LM) by conventional CT scanning, which results in a large number of missed diagnoses. This study aimed to evaluate the utility of a contrast-enhanced CT-based radiomic nomogram for preoperative differentiation of IVL and LM. Methods: 124 patients (37 IVL and 87 LM) were retrospectively enrolled in the study. Radiomic features were extracted from contrast-enhanced CT before surgery. Clinical, radiomic, and combined models were developed using LightGBM (Light Gradient Boosting Machine) algorithm to differentiate IVL and LM. The clinical and radiomic signatures were integrated into a nomogram. The diagnostic performance of the models was evaluated using the area under the curve (AUC) and decision curve analysis (DCA). Results: Clinical factors, such as symptoms, menopausal status, age, and selected imaging features, were found to have significant correlations with the differential diagnosis of IVL and LM. A total of 108 radiomic features were extracted from contrast-enhanced CT images and selected for analysis. 29 radiomics features were selected to establish the Rad-score. A clinical model was developed to discriminate IVL and LM (AUC=0.826). Radiomic models were used to effectively differentiate IVL and LM (AUC=0.980). This radiological nomogram combined the Rad-score with independent clinical factors showed better differentiation efficiency than the clinical model (AUC=0.985, p=0.046). Conclusion: This study provides evidence for the utility of a radiomic nomogram integrating clinical and radiomic signatures for differentiating IVL and LM with improved diagnostic accuracy. The nomogram may be useful in clinical decision-making and provide recommendations for clinical treatment.

2.
Nature ; 616(7956): 326-331, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36848932

RESUMO

cGAS is an evolutionarily conserved enzyme that has a pivotal role in immune defence against infection1-3. In vertebrate animals, cGAS is activated by DNA to produce cyclic GMP-AMP (cGAMP)4,5, which leads to the expression of antimicrobial genes6,7. In bacteria, cyclic dinucleotide (CDN)-based anti-phage signalling systems (CBASS) have been discovered8-11. These systems are composed of cGAS-like enzymes and various effector proteins that kill bacteria on phage infection, thereby stopping phage spread. Of the CBASS systems reported, approximately 39% contain Cap2 and Cap3, which encode proteins with homology to ubiquitin conjugating (E1/E2) and deconjugating enzymes, respectively8,12. Although these proteins are required to prevent infection of some bacteriophages8, the mechanism by which the enzymatic activities exert an anti-phage effect is unknown. Here we show that Cap2 forms a thioester bond with the C-terminal glycine of cGAS and promotes conjugation of cGAS to target proteins in a process that resembles ubiquitin conjugation. The covalent conjugation of cGAS increases the production of cGAMP. Using a genetic screen, we found that the phage protein Vs.4 antagonized cGAS signalling by binding tightly to cGAMP (dissociation constant of approximately 30 nM) and sequestering it. A crystal structure of Vs.4 bound to cGAMP showed that Vs.4 formed a hexamer that was bound to three molecules of cGAMP. These results reveal a ubiquitin-like conjugation mechanism that regulates cGAS activity in bacteria and illustrates an arms race between bacteria and viruses through controlling CDN levels.


Assuntos
Bactérias , Proteínas de Bactérias , Bacteriófagos , Nucleotidiltransferases , Ubiquitina , Animais , Bactérias/enzimologia , Bactérias/imunologia , Bactérias/metabolismo , Bactérias/virologia , Bacteriófagos/imunologia , Nucleotídeos Cíclicos/biossíntese , Nucleotídeos Cíclicos/metabolismo , Nucleotidiltransferases/química , Nucleotidiltransferases/metabolismo , Ubiquitina/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas Virais/metabolismo , Interações entre Hospedeiro e Microrganismos
3.
Front Cardiovasc Med ; 9: 913754, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36061559

RESUMO

Background: Trauma is a relatively uncommon etiology of carotid artery dissection. Trauma is both penetrative and trivial, which can lead to carotid artery dissection. In the current study, we present an unusual case in which carotid artery dissection was potentially triggered by the damaging thermal effect of 7D High-Intensity Macro- and Micro-Focused Ultrasound (7D HIFU), which has been proposed as a safe and effective non-surgical modality for skin rejuvenation. Case summary: A 41-year-old woman developed headache and clinical manifestations of cerebral infarction after 7D HIFU, aimed at removing neckline. Head and neck magnetic resonance angiography (MRA) and computed tomography angiogram (CTA) revealed severe stenosis and dissection of the left internal carotid artery. Neither the patient's history nor the physical examination showed any special indicators. After resection of the left carotid artery dissection, autologous great saphenous vein interposition grafting, and simple mastoidectomy, the patient underwent head and neck MRA, which revealed recanalization of the left internal carotid artery. Conclusion: Although mild or moderate complications of 7D HIFU, such as erythema, edema, transient dysesthesia, and motor nerve paresis, have been previously reported, a few previous literature studies documented severe complications of the cosmetic procedure. However, many recent studies pointed out the possibility of 7D HIFU damaging adjacent non-target tissues due to inadequate focal depth of HIFU treatment. Our case is the first to indicate that 7D HIFU could cause carotid artery dissection. We propose that better visualization systems and more rigorous operator training are needed to reduce the risk of the potential off-target damaging effect of 7D HIFU by reporting the case in which the damaging heat effect of 7D HIFU precipitated the carotid artery dissection HIFU.

4.
Cell Mol Life Sci ; 79(5): 269, 2022 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-35501612

RESUMO

Soluble receptor for advanced glycation end-products (sRAGE) was reported to inhibit cardiac apoptosis through the mitochondrial pathway during myocardial ischemia/reperfusion (I/R) injury. Meanwhile, the proapoptotic protein Bcl2 and adenovirus E1B 19-kDa-interacting protein 3 (Bnip3) was reported to mediate mitochondrial depolarization and be activated by the Forkhead box protein O3 (FoxO3a). Therefore, it is supposed that FoxO3a-Bnip3 pathway might be involved in the inhibiting effects of sRAGE on mitochondrial apoptosis during I/R. I/R surgery or glucose deprivation/reoxygenation was adopted to explore mitochondrial depolarization, apoptosis and related signaling pathways in mice hearts and cultured cardiomyocytes. The results showed that overexpression of sRAGE in cardiomyocytes dramatically improved cardiac function and reduced infarct areas in I/R treated mice. sRAGE inhibited mitochondrial depolarization and cardiac apoptosis during I/R, which correlated with reduced expression of Bnip3, Sirt2, phosphorylation of Akt and FoxO3a which translocated into nucleus in cultured cardiomyocytes. Either Sirt2 or FoxO3a silencing enhanced the inhibiting effects of sRAGE on mitochondrial depolarization induced by I/R in cultured cardiomyocytes. Meanwhile, overexpression or silencing of FoxO3a affected the inhibiting effects of sRAGE on Bnip3 and cleaved caspase-3 in cultured cardiomyocytes. Therefore, it is suggested that sRAGE inhibited I/R injuries via reducing mitochondrial apoptosis through the FoxO3a-Bnip3 pathway.


Assuntos
Traumatismo por Reperfusão Miocárdica , Animais , Apoptose , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Receptor para Produtos Finais de Glicação Avançada/genética , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Sirtuína 2/metabolismo , Sirtuína 2/farmacologia
5.
Apoptosis ; 27(5-6): 354-367, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35359221

RESUMO

Soluble receptor for advanced glycation end-product (sRAGE) was reported to protect myocardial ischemia/reperfusion (I/R) injuries via directly interacting with cardiomyocytes besides competing with RAGE for AGEs. However, the specific molecule for the interaction between sRAGE and cardiomyocytes are not clearly defined. Integrins which were reported to interact with RAGE on leukocytes were also expressed on myocardial cells, therefore it was supposed that sRAGE might interact with integrins on cardiomyocytes to protect hearts from ischemia/reperfusion injuries. The results showed that sRAGE increased the expression of integrinß3 but not integrinß1, ß2, ß4 or ß5 in cardiomyocytes during I/R injuries. Meanwhile, the suppressive effects of sRAGE on cardiac function, cardiac infraction size and apoptosis in mice were cancelled by inhibition of integrinß3 with cilengitide (CLG, 75 mg/kg). The results from cultured cardiomyocytes also proved that sRAGE attenuated myocardial apoptosis and autophagy through interacting with integrinß3 to activate Akt and STAT3 pathway during oxygen and glucose deprivation/reperfusion (OGD/R) treatment. Furthermore, the phosphorylation of STAT3 was significantly downregulated by the inhibition of Akt (LY294002, 10 µM) in OGD/R and sRAGE treated cardiomyocytes, which suggested that STAT3 pathway was induced by Akt in I/R and sRAGE treated cardiomyocytes. The present study contributes to the understanding of myocardial I/R pathogenesis and provided a novel integrinß3-dependent therapy strategy for sRAGE ameliorating I/R injuries.


Assuntos
Isquemia Miocárdica , Traumatismo por Reperfusão Miocárdica , Animais , Apoptose , Integrinas/genética , Isquemia , Camundongos , Isquemia Miocárdica/tratamento farmacológico , Isquemia Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor para Produtos Finais de Glicação Avançada/genética , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Reperfusão , Transdução de Sinais
6.
Front Cardiovasc Med ; 9: 851603, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35463776

RESUMO

Atherosclerotic disease has become the major cause of death worldwide. Smoking, as a widespread independent risk factor, further strengthens the health burden of atherosclerosis. Irisin is a cytokine that increases after physical activity and shows an atheroprotective effect, while its specific mechanism in the process of atherosclerosis is little known. The reversal effect of irisin on intimal thickening induced by smoking-mediated atherosclerosis was identified in Apoe -/- mice through the integrin αVß5 receptor. Endothelial cells treated with nicotine and irisin were further subjected to RNA-seq for further illustrating the potential mechanism of irisin in atherosclerosis, as well as the wound healing assays, CCK-8 assays, ß-gal staining and cell cycle determination to confirm phenotypic alterations. Endothelial differential expressed gene enrichment showed focal adhesion for migration and proliferation, as well as the P53 signaling pathway for cell senescence and cell cycle control. Irisin exerts antagonistic effects on nicotine-mediated migration and proliferation via the integrin αVß5/PI3K pathway. In addition, irisin inhibits nicotine-mediated endothelial senescence and cell cycle arrest in G0/G1 phase via P53/P21 pathway. This study further illustrates the molecular mechanism of irisin in atherosclerosis and stresses its potential as an anti-atherosclerotic therapy.

7.
Ann Transl Med ; 9(9): 805, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-34268418

RESUMO

BACKGROUND: Atherosclerosis is a chronic disease, with smoking being an independent risk factor. Irisin, a factor produced by myocytes, is expected to treat smoking-related arteriosclerosis, however its specific mechanism remains unclear. METHODS: Forty Apoe-/- mice with nicotine intervention were involved in this study. The atherosclerotic lesions, smooth muscle cell proliferation, and macrophage infiltration induced by nicotine, and the corresponding changes caused by the administration of irisin, were obtained. The integrin αVß5 inhibitor, cilengitide, was included to determine the cell entry pathway of irisin. Proteins and mRNA levels of phosphatidylinositol 3-kinase (PI3K) and downstreams were detected to clarify the specific molecular mechanism of irisin activity. RESULTS: H&E staining and Masson staining showed that nicotine could aggravate the intensity of atherosclerosis in mice, and Irisin could reverse the thickening of the vascular media induced by nicotine. Immunohistochemical staining of CD68 and α-SMA suggested that Irisin could inhibit nicotine-mediated macrophage infiltration and smooth muscle cell proliferation. The protective effect of Irisin was partially reduced after the administration of cilengitide, confirming that Irisin enters cells through multiple ways, including integrin αvß5. Nicotine was confirmed to activate the PI3K pathway to promote media thickening, while Irisin can inhibit the activation of the PI3K pathway, thus playing its anti-atherosclerosis role. Irisin was further observed to reverse nicotine-mediated P27 down-regulation. CONCLUSIONS: Irisin was found to inhibit nicotine-mediated medium thickening, smooth muscle cell proliferation, macrophage infiltration, and atherosclerosis progression via the integrin αVß5/PI3K/P27 pathway.

8.
Science ; 371(6535)2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33542149

RESUMO

The cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) synthase (cGAS) detects microbial and self-DNA in the cytosol to activate immune and inflammatory programs. cGAS also associates with chromatin, especially after nuclear envelope breakdown when cells enter mitosis. How cGAS is regulated during cell cycle transition is not clear. Here, we found direct biochemical evidence that cGAS activity was selectively suppressed during mitosis in human cell lines and uncovered two parallel mechanisms underlying this suppression. First, cGAS was hyperphosphorylated at the N terminus by mitotic kinases, including Aurora kinase B. The N terminus of cGAS was critical for sensing nuclear chromatin but not mitochondrial DNA. Chromatin sensing was blocked by hyperphosphorylation. Second, oligomerization of chromatin-bound cGAS, which is required for its activation, was prevented. Together, these mechanisms ensure that cGAS is inactive when associated with chromatin during mitosis, which may help to prevent autoimmune reaction.


Assuntos
Cromatina/metabolismo , Mitose , Nucleotidiltransferases/metabolismo , Aurora Quinase B/metabolismo , Ciclo Celular , Linhagem Celular , DNA/metabolismo , DNA Mitocondrial/metabolismo , Ativação Enzimática , Humanos , Nucleotídeos Cíclicos/metabolismo , Nucleotidiltransferases/química , Fosforilação , Multimerização Proteica
9.
Apoptosis ; 25(5-6): 341-353, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32333220

RESUMO

Soluble receptor for advanced glycation end-products (sRAGE), which exerts cardioprotective effect through inhibiting cardiomyocyte apoptosis and autophagy during ischemia/reperfusion (I/R) injury, is also known to enhance angiogenesis in post-ischemic reperfusion injury-critical limb ischemia (PIRI-CLI) mice. However, whether sRAGE protects the heart from myocardial I/R injury via promoting angiogenesis remains unclear. Myocardial model of I/R injury was conducted by left anterior descending (LAD) ligation for 30 min and reperfusion for 2 weeks in C57BL/6 mice. And I/R injury in cardiac microvascular endothelial cells (CMECs) was duplicated by oxygen and glucose deprivation. The results showed that I/R-induced cardiac dysfunction, inflammation and myocardial fibrosis were all reversed by sRAGE. CD31 immunohistochemistry staining showed that sRAGE increased the density of vessels after I/R injury. The results from cultured CMECs showed that sRAGE inhibited apoptosis and increased proliferation, migration, angiogenesis after exposure to I/R. These effects were dependent on signal transducer and activator of transcription 3 (STAT3) pathway. Together, the present study demonstrated that activation of STAT3 contributed to the protective effects of sRAGE on myocardial I/R injury via promoting angiogenesis.


Assuntos
Células Endoteliais/metabolismo , Produtos Finais de Glicação Avançada/genética , Isquemia Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/genética , Neovascularização Fisiológica , Receptor para Produtos Finais de Glicação Avançada/genética , Fator de Transcrição STAT3/genética , Animais , Apoptose/genética , Autofagia/genética , Débito Cardíaco/fisiologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Regulação da Expressão Gênica , Glucose/deficiência , Produtos Finais de Glicação Avançada/metabolismo , Frequência Cardíaca/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Oxigênio/farmacologia , Cultura Primária de Células , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Solubilidade , Volume Sistólico/fisiologia
10.
Int J Mol Med ; 43(6): 2507-2515, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30942429

RESUMO

The current study investigated the role of sRAGE in the production of IFN­Î³ in macrophages with I/R treatment. The number of macrophages in myocardial tissues treated with I/R with or without sRAGE was determined via immunohistochemical staining. Proliferative activity of macrophages was analyzed by a 5­BrdU incorporation assay. Differentiation of macrophages was detected via immunofluorescence staining of iNOS (M1 macrophage marker). IFN­Î³ production, due to sRAGE stimulation, in Raw 264.7 macrophages and the NF­κB signaling pathway were measured using western blotting. A ChIP assay was used to examine the interactions between NF­κB and the promoter of IFN­Î³. The results showed that the number of macrophages in I/R­treated myocardial tissues was increased following sRAGE infusion. Proliferation of macrophages was increased significantly in the presence of sRAGE; after I/R treatment, the cells preferred to differentiate into M1 macrophages. IFN­Î³ expression in Raw 264.7 macrophages was suppressed by an NF­κB inhibitor (Bay117082) but enhanced by sRAGE, with or without I/R treatment. Furthermore, sRAGE increased the phosphorylation of IκB, IKK and NF­κB, as well as the translocation of NF­κB into the nucleus of Raw 264.7 macrophages, with or without I/R treatment. ChIP results showed that sRAGE promoted NF­κB binding to the promoter of IFN­Î³ in Raw 264.7 macrophages. Therefore, the findings of the present study indicated that sRAGE protected the heart from I/R injuries, which might be mediated by promoting infiltration and the differentiation of macrophages into M1, which would then synthesize and secrete IFN­Î³ through activating the NF­κB signaling pathway.


Assuntos
Interferon gama/imunologia , Macrófagos/imunologia , Traumatismo por Reperfusão Miocárdica/imunologia , NF-kappa B/imunologia , Receptor para Produtos Finais de Glicação Avançada/imunologia , Animais , Proliferação de Células , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão Miocárdica/patologia , Células RAW 264.7 , Transdução de Sinais
11.
Nature ; 567(7747): 262-266, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30842662

RESUMO

Cyclic GMP-AMP (cGAMP) synthase (cGAS) detects infections or tissue damage by binding to microbial or self DNA in the cytoplasm1. Upon binding DNA, cGAS produces cGAMP that binds to and activates the adaptor protein STING, which then activates the kinases IKK and TBK1 to induce interferons and other cytokines2-6. Here we report that STING also activates autophagy through a mechanism that is independent of TBK1 activation and interferon induction. Upon binding cGAMP, STING translocates to the endoplasmic reticulum-Golgi intermediate compartment (ERGIC) and the Golgi in a process that is dependent on the COP-II complex and ARF GTPases. STING-containing ERGIC serves as a membrane source for LC3 lipidation, which is a key step in autophagosome biogenesis. cGAMP induced LC3 lipidation through a pathway that is dependent on WIPI2 and ATG5 but independent of the ULK and VPS34-beclin kinase complexes. Furthermore, we show that cGAMP-induced autophagy is important for the clearance of DNA and viruses in the cytosol. Interestingly, STING from the sea anemone Nematostella vectensis induces autophagy but not interferons in response to stimulation by cGAMP, which suggests that induction of autophagy is a primordial function of the cGAS-STING pathway.


Assuntos
Autofagia , Proteínas de Membrana/metabolismo , Nucleotidiltransferases/metabolismo , Transdução de Sinais , Animais , Autofagossomos/metabolismo , Proteína 5 Relacionada à Autofagia/deficiência , Proteína 5 Relacionada à Autofagia/genética , Proteína 5 Relacionada à Autofagia/metabolismo , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/deficiência , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/genética , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Proteína Beclina-1/deficiência , Proteína Beclina-1/genética , Proteína Beclina-1/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Citosol/virologia , Vírus de DNA/genética , Vírus de DNA/metabolismo , DNA Viral/metabolismo , Retículo Endoplasmático/metabolismo , Evolução Molecular , Complexo de Golgi/metabolismo , Células HEK293 , Humanos , Interferons/biossíntese , Interferons/imunologia , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Nucleotídeos Cíclicos/imunologia , Nucleotídeos Cíclicos/metabolismo , Proteínas de Ligação a Fosfato , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico , Anêmonas-do-Mar , Proteínas de Transporte Vesicular/metabolismo
12.
Free Radic Biol Med ; 130: 107-119, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30367996

RESUMO

The pathogenesis of myocardial ischemia/reperfusion (I/R) is poorly understood, but recent evidence suggests that autophagy plays crucial roles in I/R injuries. Soluble receptor for advanced glycation end-products (sRAGE) exerts protective effects during I/R by decreasing cardiac apoptosis, which is mediated via increasing the ubiquitin proteasome system (UPS) and signal transducer and activator of transcription 3 (STAT3). The present study examined the effects and mechanisms of sRAGE on I/R-triggered cardiac autophagy. I/R was performed in mice or primary neonatal cardiomyocytes with or without sRAGE administration or overexpression. Cardiac function and infarct size were detected in mouse hearts. Apoptosis, autophagy and autophagy-related signaling pathways were detected in mouse hearts and cardiomyocytes. The results demonstrated that sRAGE significantly improved cardiac function and reduced infarct size during I/R in mice. sRAGE inhibited I/R-induced apoptosis, which correlated with a reduction in autophagy-associated proteins, including ATG7, Beclin-1 and microtubule-associated protein 1 light chain 3 (LC3). sRAGE reduced autophagosome formation during I/R in vivo and in vitro. sRAGE significantly activated STAT3, but not mammalian target of rapamycin (mTOR), during I/R in vivo and in vitro, and suppression of STAT3 abolished the sRAGE inhibition of autophagy during I/R in vitro. Activation of autophagy using ATG7 overexpression with an adenovirus significantly abolished the sRAGE-induced reduction of cardiac apoptosis during I/R. These results suggest that sRAGE inhibits I/R injuries in the heart via a decrease in autophagy, a process that is dependent on STAT3 activation.


Assuntos
Produtos Finais de Glicação Avançada/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/fisiologia , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Autofagia , Proteína 7 Relacionada à Autofagia/genética , Proteína 7 Relacionada à Autofagia/metabolismo , Células Cultivadas , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais
13.
Lab Invest ; 99(3): 358-370, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30089851

RESUMO

The ubiquitin-proteasome system (UPS) is essential for protein degradation and plays critical roles in myocardial ischemia/reperfusion (MI/R) injuries. Previous studies have demonstrated that the soluble receptor for advanced glycation end-product (sRAGE) inhibited MI/R-induced apoptosis by upregulating proteasome subunits. However, the mechanism remains unknown. An MI/R model was established by left anterior descending (LAD) coronary artery ligation in mice. Recombinant sRAGE protein or saline was injected intramyocardially with or without neutralizing interferon-γ (IFN-γ) antibody injected intraperitoneally before ligation. In cardiomyocytes, ischemia was simulated with "ischemia buffer" and sRAGE was overexpressed by adenovirus. Adenovirus expressing the interference RNA of ß5i was used to knockdown ß5i in cardiomyocytes. IFN-γ was induced by sRAGE both in sham and MI/R mice. Blockade of IFN-γ using IFN-γ antibody abolished the rescue effects of sRAGE for cardiac dysfunction, infarct size and apoptosis provoked by MI/R. Blockade of IFN-γ reversed the upregulation of ß1i and ß5i expression induced by sRAGE during MI/R in heart, accompanied by decreasing chymotrypsin-like proteasome activity. In addition, IFN-γ antibody abolished the suppressing effect of sRAGE on MI/R-induced p38 and c-Jun N-terminal kinase (JNK) activation, as well as p53 expression, both in vivo and in vitro. However, knockdown of ß5i abolished the antiapoptosis effect of sRAGE during hypoxia/reoxygenation (H/R) in vitro, accompanied by decreased degradation of p53. Our data suggest a novel mechanism for sRAGE in preventing MI/R-induced apoptosis in heart: sRAGE inhibits MI/R-induced apoptosis in cardiomyocytes by degrading p53 by ß5i subunit that is increased via upregulation of IFN-γ.


Assuntos
Interferon gama/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Animais , Anticorpos Neutralizantes/administração & dosagem , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Interferon gama/antagonistas & inibidores , Interferon gama/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor para Produtos Finais de Glicação Avançada/antagonistas & inibidores , Receptor para Produtos Finais de Glicação Avançada/genética , Proteína Supressora de Tumor p53/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo
14.
Free Radic Biol Med ; 94: 17-26, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26878774

RESUMO

AIM: Apoptosis participated in the pathological process of myocardial ischemia/reperfusion (I/R) injury. Previous studies have reported that endogenous substance sRAGE protect against I/R injury through inhibiting myocardial apoptosis. But the mechanisms are currently unknown. Prior work has demonstrated that ubiquitin proteasome system (UPS) dysfunction is closely related to apoptosis. We explored the potential role of UPS in the effect of sRAGE inhibition on I/R-induced myocardial apoptosis. METHODS AND RESULTS: Adult male C57BL mice treated with sRAGE (100µg/day, i.p.) or saline were performed to ligate left anterior descending coronary artery (LAD) as an in vivo model. As an in vitro model, primary murine cardiomyocytes pretreated with sRAGE or sRAGE-containing adenovirus were simulated I/R by "ischemia buffer". The TUNEL and caspase-3 activity were assessed. Also the activity and expression of proteasome were detected. sRAGE decreased the number of TUNEL-positive cardiomyocytes and caspase-3 activity, however, the inhibition of sRAGE on I/R-induced apoptosis was abolished by proteasome inhibitor Bortezimb (BTZ). sRAGE inhibited the decreased proteasome activity, also the reduction in protein and gene levels of ß1i and ß5i following I/R. Suppression of STAT3 blocked the inhibition of sRAGE on apoptosis induced by I/R. The chromatin immunoprecipitation (CHIP) results confirmed that sRAGE promoted activating STAT3 binding to ß1i and ß5i promoter. CONCLUSIONS: Our data suggest that the inhibition of sRAGE on I/R-induced apoptosis is associated with activation and expression of proteasome, including improved proteasome activity and elevated ß1i and ß5i expression mediated by STAT3 activation. We predict that sRAGE is a novel intervention to target UPS activation for preventing and treating myocardial apoptosis.


Assuntos
Produtos Finais de Glicação Avançada/administração & dosagem , Isquemia Miocárdica/tratamento farmacológico , Complexo de Endopeptidases do Proteassoma/genética , Traumatismo por Reperfusão/tratamento farmacológico , Fator de Transcrição STAT3/genética , Animais , Apoptose/genética , Bortezomib/administração & dosagem , Caspase 3/genética , Produtos Finais de Glicação Avançada/genética , Humanos , Camundongos , Isquemia Miocárdica/genética , Isquemia Miocárdica/fisiopatologia , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/fisiopatologia , Ubiquitina/genética
15.
Heart Vessels ; 29(5): 679-89, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24141989

RESUMO

Angiogenesis plays an important role in myocardial infarction. Apelin and its natural receptor (angiotensin II receptor-like 1, AGTRL-1 or APLNR) induce sprouting of endothelial cells in an autocrine or paracrine manner. The aim of this study is to investigate whether apelin can improve the cardiac function after myocardial infarction by increasing angiogenesis in infarcted myocardium. Left ventricular end-diastolic pressure (LVEDP), left ventricular end systolic pressure (LVESP), left ventricular developed pressure (LVDP), maximal left ventricular pressure development (±LVdp/dtmax), infarct size, and angiogenesis were evaluated to analyze the cardioprotective effects of apelin on ischemic myocardium. Assays of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, 5-bromo-2'-deoxyuridine incorporation, wound healing, transwells, and tube formation were used to detect the effects of apelin on proliferation, migration, and chemotaxis of cardiac microvascular endothelial cells. Fluorescein isothiocyanate-labeled bovine serum albumin penetrating through monolayered cardiac microvascular endothelial cells was measured to evaluate the effects of apelin on permeability of microvascular endothelial cells. In vivo results showed that apelin increased ±LV dp/dtmax and LVESP values, decreased LVEDP values (all p < 0.05), and promoted angiogenesis in rat heart after ligation of the left anterior descending coronary artery. In vitro results showed that apelin dose-dependently enhanced proliferation, migration, chemotaxis, and tube formation, but not permeability of cardiac microvascular endothelial cells. Apelin also increased the expression of vascular endothelial growth factor receptors-2 (VEGFR2) and the endothelium-specific receptor tyrosine kinase (Tie-2) in cardiac microvascular endothelial cells. These results indicated that apelin played a protective role in myocardial infarction through promoting angiogenesis and decreasing permeability of microvascular endothelial cells via upregulating the expression of VEGFR2 and Tie-2 in cardiac microvascular endothelial cells.


Assuntos
Indutores da Angiogênese/farmacologia , Cardiotônicos/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Infarto do Miocárdio/tratamento farmacológico , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Permeabilidade Capilar/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Masculino , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , Miocárdio/patologia , Ratos Wistar , Receptor TIE-2/efeitos dos fármacos , Receptor TIE-2/metabolismo , Recuperação de Função Fisiológica , Fatores de Tempo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/efeitos dos fármacos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Função Ventricular Esquerda/efeitos dos fármacos , Pressão Ventricular/efeitos dos fármacos
16.
Science ; 341(6148): 903-6, 2013 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-23929945

RESUMO

Retroviruses, including HIV, can activate innate immune responses, but the host sensors for retroviruses are largely unknown. Here we show that HIV infection activates cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS) to produce cGAMP, which binds to and activates the adaptor protein STING to induce type I interferons and other cytokines. Inhibitors of HIV reverse transcriptase, but not integrase, abrogated interferon-ß induction by the virus, suggesting that the reverse-transcribed HIV DNA triggers the innate immune response. Knockout or knockdown of cGAS in mouse or human cell lines blocked cytokine induction by HIV, murine leukemia virus, and simian immunodeficiency virus. These results indicate that cGAS is an innate immune sensor of HIV and other retroviruses.


Assuntos
Infecções por HIV/imunologia , HIV/imunologia , Imunidade Inata , Nucleotidiltransferases/metabolismo , Animais , Linhagem Celular , Técnicas de Silenciamento de Genes , Células HEK293 , HIV/efeitos dos fármacos , HIV/enzimologia , Infecções por HIV/enzimologia , Infecções por HIV/virologia , Transcriptase Reversa do HIV/antagonistas & inibidores , Humanos , Interferon beta/biossíntese , Proteínas de Membrana/metabolismo , Camundongos , Nucleotidiltransferases/genética , Retroviridae/imunologia , Infecções por Retroviridae/enzimologia , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/virologia , Inibidores da Transcriptase Reversa/farmacologia
17.
Science ; 339(6121): 826-30, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23258412

RESUMO

Cytosolic DNA induces type I interferons and other cytokines that are important for antimicrobial defense but can also result in autoimmunity. This DNA signaling pathway requires the adaptor protein STING and the transcription factor IRF3, but the mechanism of DNA sensing is unclear. We found that mammalian cytosolic extracts synthesized cyclic guanosine monophosphate-adenosine monophosphate (cyclic GMP-AMP, or cGAMP) in vitro from adenosine triphosphate and guanosine triphosphate in the presence of DNA but not RNA. DNA transfection or DNA virus infection of mammalian cells also triggered cGAMP production. cGAMP bound to STING, leading to the activation of IRF3 and induction of interferon-ß. Thus, cGAMP functions as an endogenous second messenger in metazoans and triggers interferon production in response to cytosolic DNA.


Assuntos
AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Citosol/imunologia , DNA/imunologia , Imunidade Inata , Nucleotídeos Cíclicos/metabolismo , Sistemas do Segundo Mensageiro/imunologia , Animais , Extratos Celulares/química , Linhagem Celular , Células HEK293 , Herpesvirus Humano 1/imunologia , Humanos , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/biossíntese , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Interferência de RNA , Transfecção
18.
Science ; 339(6121): 786-91, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23258413

RESUMO

The presence of DNA in the cytoplasm of mammalian cells is a danger signal that triggers host immune responses such as the production of type I interferons. Cytosolic DNA induces interferons through the production of cyclic guanosine monophosphate-adenosine monophosphate (cyclic GMP-AMP, or cGAMP), which binds to and activates the adaptor protein STING. Through biochemical fractionation and quantitative mass spectrometry, we identified a cGAMP synthase (cGAS), which belongs to the nucleotidyltransferase family. Overexpression of cGAS activated the transcription factor IRF3 and induced interferon-ß in a STING-dependent manner. Knockdown of cGAS inhibited IRF3 activation and interferon-ß induction by DNA transfection or DNA virus infection. cGAS bound to DNA in the cytoplasm and catalyzed cGAMP synthesis. These results indicate that cGAS is a cytosolic DNA sensor that induces interferons by producing the second messenger cGAMP.


Assuntos
Citosol/imunologia , DNA/imunologia , Interferon Tipo I/biossíntese , Interferon beta/biossíntese , Nucleotidiltransferases/metabolismo , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , AMP Cíclico/biossíntese , GMP Cíclico/biossíntese , Citidina Trifosfato/metabolismo , Citosol/enzimologia , DNA/metabolismo , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Redes e Vias Metabólicas , Camundongos , Dados de Sequência Molecular , Nucleotidiltransferases/genética , Nucleotidiltransferases/isolamento & purificação
19.
Immunity ; 36(6): 959-73, 2012 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-22705106

RESUMO

RIG-I and MDA5 detect viral RNA in the cytoplasm and activate signaling cascades leading to the production of type-I interferons. RIG-I is activated through sequential binding of viral RNA and unanchored lysine-63 (K63) polyubiquitin chains, but how polyubiquitin activates RIG-I and whether MDA5 is activated through a similar mechanism remain unresolved. Here, we showed that the CARD domains of MDA5 bound to K63 polyubiquitin and that this binding was essential for MDA5 to activate the transcription factor IRF3. Mutations of conserved residues in MDA5 and RIG-I that disrupt their ubiquitin binding also abrogated their ability to activate IRF3. Polyubiquitin binding induced the formation of a large complex consisting of four RIG-I and four ubiquitin chains. This hetero-tetrameric complex was highly potent in activating the antiviral signaling cascades. These results suggest a unified mechanism of RIG-I and MDA5 activation and reveal a unique mechanism by which ubiquitin regulates cell signaling and immune response.


Assuntos
RNA Helicases DEAD-box/fisiologia , Vírus da Encefalomiocardite/fisiologia , Poliubiquitina/metabolismo , Animais , Sistema Livre de Células , Proteína DEAD-box 58 , RNA Helicases DEAD-box/química , RNA Helicases DEAD-box/genética , Vírus da Encefalomiocardite/genética , Fibroblastos/metabolismo , Fibroblastos/virologia , Células HEK293/metabolismo , Células HEK293/virologia , Humanos , Fator Regulador 3 de Interferon/metabolismo , Helicase IFIH1 Induzida por Interferon , Interferon beta/biossíntese , Interferon beta/genética , Camundongos , Complexos Multiproteicos , Mutagênese Sítio-Dirigida , Ligação Proteica , Mapeamento de Interação de Proteínas , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , RNA Viral/metabolismo , Receptores Imunológicos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/fisiologia , Transdução de Sinais/fisiologia , Relação Estrutura-Atividade , Ubiquitinação
20.
Mol Cell ; 44(4): 559-71, 2011 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-22099304

RESUMO

A20 is a potent anti-inflammatory protein that inhibits NF-κB, and A20 dysfunction is associated with autoimmunity and B cell lymphoma. A20 harbors a deubiquitination enzyme domain and can employ multiple mechanisms to antagonize ubiquitination upstream of NEMO, a regulatory subunit of the IκB kinase complex (IKK). However, direct evidence of IKK inhibition by A20 is lacking, and the inhibitory mechanism remains poorly understood. Here we show that A20 can directly impair IKK activation without deubiquitination or impairment of ubiquitination enzymes. We find that polyubiquitin binding by A20, which is largely dependent on A20's seventh zinc-finger motif (ZnF7), induces specific binding to NEMO. Remarkably, this ubiquitin-induced recruitment of A20 to NEMO is sufficient to block IKK phosphorylation by its upstream kinase TAK1. Our results suggest a noncatalytic mechanism of IKK inhibition by A20 and a means by which polyubiquitin chains can specify a signaling outcome.


Assuntos
Quinase I-kappa B/metabolismo , Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , NF-kappa B/metabolismo , Proteínas Nucleares , Transdução de Sinais/genética , Dedos de Zinco/genética , Autoimunidade/genética , Proteínas de Ligação a DNA , Ativação Enzimática/genética , Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Quinase I-kappa B/antagonistas & inibidores , Quinase I-kappa B/genética , Imunoprecipitação , Inflamação/genética , Interleucina-1beta/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Linfoma de Células B/genética , Linfoma de Células B/metabolismo , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilação , Poliubiquitina , Ligação Proteica , Estrutura Terciária de Proteína , Transdução de Sinais/efeitos dos fármacos , Proteína 3 Induzida por Fator de Necrose Tumoral alfa , Ubiquitinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA