Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 193
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 54(3): 454-467.e6, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33561388

RESUMEN

Heparin, a mammalian polysaccharide, is a widely used anticoagulant medicine to treat thrombotic disorders. It is also known to improve outcomes in sepsis, a leading cause of mortality resulted from infection-induced immune dysfunction. Whereas it is relatively clear how heparin exerts its anticoagulant effect, the immunomodulatory mechanisms enabled by heparin remain enigmatic. Here, we show that heparin prevented caspase-11-dependent immune responses and lethality in sepsis independent of its anticoagulant properties. Heparin or a chemically modified form of heparin without anticoagulant function inhibited the alarmin HMGB1-lipopolysaccharide (LPS) interaction and prevented the macrophage glycocalyx degradation by heparanase. These events blocked the cytosolic delivery of LPS in macrophages and the activation of caspase-11, a cytosolic LPS receptor that mediates lethality in sepsis. Survival was higher in septic patients treated with heparin than those without heparin treatment. The identification of this previously unrecognized heparin function establishes a link between innate immune responses and coagulation.


Asunto(s)
Anticoagulantes/uso terapéutico , Caspasas/metabolismo , Heparina/uso terapéutico , Macrófagos/inmunología , Sepsis/tratamiento farmacológico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Caspasas/genética , Línea Celular , Femenino , Glucuronidasa/genética , Glucuronidasa/metabolismo , Glicocálix/metabolismo , Proteína HMGB1/metabolismo , Humanos , Inmunomodulación , Lipopolisacáridos/metabolismo , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Sepsis/mortalidad , Análisis de Supervivencia , Adulto Joven
2.
Immunity ; 51(6): 983-996.e6, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31836429

RESUMEN

Excessive activation of the coagulation system leads to life-threatening disseminated intravascular coagulation (DIC). Here, we examined the mechanisms underlying the activation of coagulation by lipopolysaccharide (LPS), the major cell-wall component of Gram-negative bacteria. We found that caspase-11, a cytosolic LPS receptor, activated the coagulation cascade. Caspase-11 enhanced the activation of tissue factor (TF), an initiator of coagulation, through triggering the formation of gasdermin D (GSDMD) pores and subsequent phosphatidylserine exposure, in a manner independent of cell death. GSDMD pores mediated calcium influx, which induced phosphatidylserine exposure through transmembrane protein 16F, a calcium-dependent phospholipid scramblase. Deletion of Casp11, ablation of Gsdmd, or neutralization of phosphatidylserine or TF prevented LPS-induced DIC. In septic patients, plasma concentrations of interleukin (IL)-1α and IL-1ß, biomarkers of GSDMD activation, correlated with phosphatidylserine exposure in peripheral leukocytes and DIC scores. Our findings mechanistically link immune recognition of LPS to coagulation, with implications for the treatment of DIC.


Asunto(s)
Caspasas Iniciadoras/metabolismo , Coagulación Intravascular Diseminada/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipopolisacáridos/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Fosfatidilserinas/metabolismo , Tromboplastina/metabolismo , Animales , Coagulación Sanguínea/fisiología , Caspasas Iniciadoras/genética , Línea Celular Tumoral , Endotoxemia/patología , Activación Enzimática , Células HT29 , Células HeLa , Humanos , Interleucina-1alfa/sangre , Interleucina-1beta/sangre , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Unión a Fosfato/genética , Piroptosis/fisiología , Transducción de Señal/fisiología
3.
Immunity ; 49(4): 740-753.e7, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30314759

RESUMEN

Caspase-11, a cytosolic endotoxin (lipopolysaccharide: LPS) receptor, mediates pyroptosis, a lytic form of cell death. Caspase-11-dependent pyroptosis mediates lethality in endotoxemia, but it is unclear how LPS is delivered into the cytosol for the activation of caspase-11. Here we discovered that hepatocyte-released high mobility group box 1 (HMGB1) was required for caspase-11-dependent pyroptosis and lethality in endotoxemia and bacterial sepsis. Mechanistically, hepatocyte-released HMGB1 bound LPS and targeted its internalization into the lysosomes of macrophages and endothelial cells via the receptor for advanced glycation end-products (RAGE). Subsequently, HMGB1 permeabilized the phospholipid bilayer in the acidic environment of lysosomes. This resulted in LPS leakage into the cytosol and caspase-11 activation. Depletion of hepatocyte HMGB1, inhibition of hepatocyte HMGB1 release, neutralizing extracellular HMGB1, or RAGE deficiency prevented caspase-11-dependent pyroptosis and death in endotoxemia and bacterial sepsis. These findings indicate that HMGB1 interacts with LPS to mediate caspase-11-dependent pyroptosis in lethal sepsis.


Asunto(s)
Caspasas/inmunología , Endotoxinas/inmunología , Proteína HMGB1/inmunología , Piroptosis/inmunología , Sepsis/inmunología , Animales , Caspasas/genética , Caspasas/metabolismo , Células Cultivadas , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Endotoxinas/metabolismo , Células HEK293 , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Humanos , Lipopolisacáridos/inmunología , Lipopolisacáridos/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor para Productos Finales de Glicación Avanzada/inmunología , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Sepsis/genética , Sepsis/metabolismo , Células THP-1
4.
Mediators Inflamm ; 2023: 2252255, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36741074

RESUMEN

Background: Sepsis, which could cause a systemic inflammatory response, is a life-threatening disease with a high morbidity and mortality rate. There is evidence that brain injury may be related to severe systemic infection induced by sepsis. The brain injury caused by sepsis could increase the risk of mortality in septic patients, which seriously affects the septic patient's prognosis of survival. Although there remains a focus on sepsis research, clinical measures to prevent and treat brain injury in sepsis are not yet available, and the high mortality rate is still a big health burden. Therefore, it is necessary to investigate the new molecules or regulated pathways that can effectively inhibit the progress of sepsis. Objective: NLR family pyrin domain-containing 3 (NLRP3) increased in the procession of sepsis and functioned as the key regulator of pyroptosis. Heat shock factor 1 (HSF1) can protect organs from multiorgan dysfunction syndrome induced by lipopolysaccharides in mice, and NLRP3 could be inhibited by HSF1 in many organs. However, whether HSF1 regulated NLRP3 in sepsis-induced brain injury, as well as the detailed mechanism of HSF1 in brain injury, remains unknown in the sepsis model. In this research, we try to explore the relationship between HSF1 and NLRP3 in a sepsis model and try to reveal the mechanism of HSF1 inhibiting the process of brain injury. Methods: In this study, we used wild-type mice and hsf1 -/- mice for in vivo research and PC12 cells for in vitro research. Real-time PCR and Western blot were used to analyze the expression of HSF1, NLRP3, cytokines, and pyrolytic proteins. EthD-III staining was chosen to detect the pyroptosis of the hippocampus and PC12 cells. Results: The results showed that HSF1 is negatively related to pyroptosis. The pyroptosis in cells of brain tissue was significantly increased in the hsf1 -/- mouse model compared to hsf1 +/+ mice. In PC12 cells, hsf1 siRNA can upregulate pyroptosis while HSF1-transfected plasmid could inhibit the pyroptosis. HSF1 could negatively regulate the NLRP3 pathway in PC12 cells, while hsf1 siRNA enhanced the pyroptosis in PC12 cells, which could be reversed by nlrp3 siRNA. Conclusion: These results imply that HSF1 could alleviate sepsis-induced brain injury by inhibiting pyroptosis through the NLRP3-dependent pathway in brain tissue and PC12 cells, suggesting HSF1 as a potential molecular target for treating brain injury in sepsis clinical studies.


Asunto(s)
Lesiones Encefálicas , Factores de Transcripción del Choque Térmico , Proteína con Dominio Pirina 3 de la Familia NLR , Sepsis , Animales , Ratones , Ratas , Factores de Transcripción del Choque Térmico/farmacología , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis , ARN Interferente Pequeño , Sepsis/metabolismo
5.
Blood ; 135(14): 1087-1100, 2020 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-32016282

RESUMEN

Bacterial infection not only stimulates innate immune responses but also activates coagulation cascades. Overactivation of the coagulation system in bacterial sepsis leads to disseminated intravascular coagulation (DIC), a life-threatening condition. However, the mechanisms by which bacterial infection activates the coagulation cascade are not fully understood. Here we show that type 1 interferons (IFNs), a widely expressed family of cytokines that orchestrate innate antiviral and antibacterial immunity, mediate bacterial infection-induced DIC by amplifying the release of high-mobility group box 1 (HMGB1) into the bloodstream. Inhibition of the expression of type 1 IFNs and disruption of their receptor IFN-α/ßR or downstream effector (eg, HMGB1) uniformly decreased gram-negative bacteria-induced DIC. Mechanistically, extracellular HMGB1 markedly increased the procoagulant activity of tissue factor by promoting the externalization of phosphatidylserine to the outer cell surface, where phosphatidylserine assembles a complex of cofactor-proteases of the coagulation cascades. These findings not only provide novel insights into the link between innate immune responses and coagulation, but they also open a new avenue for developing novel therapeutic strategies to prevent DIC in sepsis.


Asunto(s)
Coagulación Intravascular Diseminada/inmunología , Endotoxemia/inmunología , Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/inmunología , Interferón-alfa/inmunología , Interferón beta/inmunología , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Animales , Coagulación Sanguínea , Coagulación Intravascular Diseminada/sangre , Coagulación Intravascular Diseminada/etiología , Endotoxemia/sangre , Endotoxemia/complicaciones , Infecciones por Bacterias Gramnegativas/sangre , Infecciones por Bacterias Gramnegativas/complicaciones , Proteína HMGB1/sangre , Proteína HMGB1/inmunología , Humanos , Inmunidad Innata , Ratones Endogámicos C57BL
6.
J Mol Cell Cardiol ; 150: 65-76, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33098823

RESUMEN

Palmitic acid (PA)-induced myocardial injury is considered a critical contributor to the development of obesity and type 2 diabetes mellitus (T2DM)-related cardiomyopathy. However, the underlying mechanism has not been fully understood. Here, we demonstrated that PA induced the cell death of H9c2 cardiomyoblasts in a dose- and time-dependent manner, while different ferroptosis inhibitors significantly abrogated the cell death of H9c2 cardiomyoblasts and primary neonatal rat cardiomyocytes exposed to PA. Mechanistically, PA decreased the protein expression levels of both heat shock factor 1 (HSF1) and glutathione peroxidase 4 (GPX4) in a dose- and time-dependent manner, which were restored by different ferroptosis inhibitors. Overexpression of HSF1 not only alleviated PA-induced cell death and lipid peroxidation but also improved disturbed iron homeostasis by regulating the transcription of iron metabolism-related genes (e.g., Fth1, Tfrc, Slc40a1). Additionally, PA-blocked GPX4 protein expression was evidently restored by HSF1 overexpression. Inhibition of endoplasmic reticulum (ER) stress rather than autophagy contributed to HSF1-mediated GPX4 expression. Moreover, GPX4 overexpression protected against PA-induced ferroptosis, whereas knockdown of GPX4 reversed the anti-ferroptotic effect of HSF1. Consistent with the in vitro findings, PA-challenged Hsf1-/- mice exhibited more serious ferroptosis, increased Slc40a1 and Fth1 mRNA expression, decreased GPX4 and TFRC expression and enhanced ER stress in the heart compared with Hsf1+/+ mice. Altogether, HSF1 may function as a key defender against PA-induced ferroptosis in cardiomyocytes by maintaining cellular iron homeostasis and GPX4 expression.


Asunto(s)
Ferroptosis , Factores de Transcripción del Choque Térmico/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Ácido Palmítico/farmacología , Animales , Línea Celular , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Ferroptosis/efectos de los fármacos , Ferroptosis/genética , Regulación de la Expresión Génica/efectos de los fármacos , Factores de Transcripción del Choque Térmico/genética , Hierro/metabolismo , Ratones , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/ultraestructura , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Ratas Sprague-Dawley , Transcripción Genética/efectos de los fármacos
7.
J Cell Mol Med ; 25(2): 751-762, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33219625

RESUMEN

Vascular smooth muscle cells (VSMCs) play a significant role in atherosclerosis. As a multifunctional protein, nucleolin (NCL) is involved in many important physiological and pathological processes. In this study, we aimed to investigate the role of nucleolin in VSMCs proliferation and cell cycle. The expression of nucleolin increased in VSMCs of mice with aortas advanced plaques. With the left common carotid-artery ligation-injury model, immunofluorescence staining revealed that nucleolin and Ki67 expression increased in VSMCs in mice left carotid artery compared with right carotid artery after surgery. POVPC or ox-LDL up-regulated nucleolin mRNA and protein expression in a dose- and time-dependent manner in HAVSMCs. POVPC (5µg/ml) or ox-LDL (50µg/ml) promoted the proliferation of HAVSMCs. Nucleolin ablation relieved the pro-proliferation role of VSMCs. The cell cycle assay and cell ability results showing that POVPC or ox-LDL increased the proliferation, but nucleolin ablation inhibited the proliferation of HAVSMCs. And nucleolin ablation can prevent DNA replication at S phase and induce cell cycle arrest in S phase. The bioinformatics database predicts protein-protein interactions with nucleolin and aurora B. Nucleolin overexpression and ablation affected the expression of aurora B. These findings indicate for the first time that nucleolin actively involved the proliferation of VSMCs via aurora B.


Asunto(s)
Apolipoproteínas E/metabolismo , Aurora Quinasa B/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aurora Quinasa B/genética , Western Blotting , Ciclo Celular/fisiología , Proliferación Celular/genética , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Humanos , Lipoproteínas LDL/metabolismo , Masculino , Ratones , Miocitos del Músculo Liso/metabolismo , Fosfoproteínas/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Nucleolina
8.
J Mol Cell Cardiol ; 145: 1-13, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32470468

RESUMEN

Myocardial ischaemia is usually accompanied by inflammatory response which plays a critical role in the myocardial healing and scar formation, while persistent inflammatory response contributes greatly to the myocardial remodeling and consequent heart failure. Metformin (Met), a widely used hypoglycemic drug, has increasingly been shown to exert remarkable cardioprotective effect on ischaemic myocardial injury such as acute myocardial infarction (AMI). However, the underlying mechanisms are still far from being fully understood. In this study, a mouse model of AMI was established through ligating the left anterior descending coronary artery (LAD), 100 mg/kg Met was given immediately after operation once daily for 3 days. It was demonstrated that Met effectively improved the cardiac haemodynamics (LVSP, LVEDP, +dp/dt, -dp/dt), diminished the infarct size, alleviated the disarrangement of myocardial cells and reduced the infiltration of inflammatory cells (macrophages, neutrophils and lymphocytes) in the heart of AMI mice. Mechanistically, Met decreased the expression of NLRP3 and enhanced the accumulation of LC3 puncta in F4/80-positive macrophages in the heart of AMI mice. Single cell suspension of cardiac macrophages was prepared from AMI mice and exhibited increased NLRP3 mRNA and protein expression. In contrast, Met decreased the expression of NLRP3 and p62, whereas increased the ratio of LC3II/LC3I. Additionally, both conditioned medium from H9c2 cardiomyocytes exposed to hydrogen peroxide (H9c2-H2O2-CM) and combination of mtDNA and ATP (mtDNA-ATP) increased the expression of NLRP3 and cleaved caspase-1 (p10) as well as intracellular ROS production in RAW264.7 macrophages, which were abrogated by Met treatment. Strikingly, chloroquine (CQ), 3-methyladenine (3-MA) and knockdown of autophagy-related gene (Atg5) abrogated the inhibitory effects of Met on H9c2-H2O2-CM and mtDNA-ATP-induced NLRP3 expression, release of IL-1ß and IL-18 as well as ROS production in RAW264.7 macrophages. Collectively, these findings suggest that Met protects against ischaemic myocardial injury through alleviating autophagy-ROS-NLRP3 axis-mediated inflammatory response in macrophages.


Asunto(s)
Autofagia , Inflamación/patología , Macrófagos/patología , Metformina/uso terapéutico , Isquemia Miocárdica/patología , Isquemia Miocárdica/prevención & control , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Autofagia/efectos de los fármacos , ADN Mitocondrial/metabolismo , Femenino , Hemodinámica/efectos de los fármacos , Peróxido de Hidrógeno/toxicidad , Macrófagos/efectos de los fármacos , Macrófagos/ultraestructura , Masculino , Metformina/farmacología , Ratones , Ratones Endogámicos C57BL , Isquemia Miocárdica/fisiopatología , Miocardio/patología , Células RAW 264.7 , Ratas
9.
Basic Res Cardiol ; 115(3): 29, 2020 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-32248306

RESUMEN

Autophagy in cardiomyocyte is involved in myocardial ischemia/reperfusion (M-I/R) injury. Caspase recruitment domain-containing protein 9 (CARD9) plays a critical role in cardiovascular diseases (CVDs) such as hypertension and cardiac fibrosis. However, its role in autophagy following M-I/R injury is yet to be fully elucidated. Here, we found that CARD9 expression increased in M-I/R mouse hearts, and in H9c2 or neonatal rat ventricular myocytes (NRVMs) in response to hypoxia/reoxygenation (H/R) or H2O2. CARD9-/- mice exhibited a significant cardiac dysfunction following M-I/R injury (30 min of left ascending coronary (LAD) ischemia and 12 h of reperfusion) compared to wild-type (WT) mice. CARD9 deletion impaired autophagy during M-I/R in vivo and in vitro, evidenced by decrease of microtubule-associated protein 1 light chain 3 (LC3) lipidation and p62 accumulation. Conversely, CARD9 overexpression increased autophagic flux as indicated by enhanced expression of LC3 II/LC3 I and a reduction in p62. The protective effect of CARD9 on cardiomyocytes against H/R-induced oxidative stress was abolished by treatment with autophagy inhibitors, 3-methyladenine (3-MA) or Bafilomycin A1(BafA1). CARD9 interacted with RUN domain Beclin-1-interacting cysteine-rich-containing (Rubicon), a negative regulator of autophagy, and enhanced UV-irradiation-resistance-associated gene (UVRAG)-Beclin1-phosphatidylinositol 3-kinase catalytic subunit type 3 (PI3KC3) interaction and UVRAG-Vps16-mediated Rab7 activation to promote autophagosome formation, maturation, and endocytosis. Ablation of Rubicon by siRNA effectively prevented the detrimental effect of CARD9 knockdown on cardiomyocytes. These results suggest that CARD9 has protective effects on the myocardium against M-I/R injury by activating autophagy and restoring autophagic flux in vivo and in vitro.


Asunto(s)
Autofagia/fisiología , Proteínas Adaptadoras de Señalización CARD/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/patología , Ratas
10.
Arterioscler Thromb Vasc Biol ; 39(9): 1787-1801, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31315433

RESUMEN

OBJECTIVE: Macrophage foam cell formation is an important process in atherosclerotic plaque development. The small GTPase Rheb (Ras homolog enriched in brain 1) regulates endocytic trafficking that is critical for foam cell formation. However, it is unclear whether and how macrophage Rheb regulates atherogenesis, which are the focuses of the current study. Approach and Results: Immunofluorescence study confirmed the colocalization of Rheb in F4/80 and Mac-2 (galectin-3)-labeled lesional macrophages. Western blot and fluorescence-activated cell sorting analysis showed that Rheb expression was significantly increased in atherosclerotic lesions of atherosclerosis-prone (apoE-/- [apolipoprotein E deficient]) mice fed with Western diet. Increased Rheb expression was also observed in oxidized LDL (low-density lipoprotein)-treated macrophages. To investigate the in vivo role of macrophage Rheb, we established mature RhebmKO (macrophage-specific Rheb knockout) mice by crossing the Rheb floxed mice with F4/80-cre mice. Macrophage-specific knockout of Rheb in mice reduced Western diet-induced atherosclerotic lesion by 32%, accompanied with a decrease in macrophage content in plaque. Mechanistically, loss of Rheb in macrophages repressed oxidized LDL-induced lipid uptake, inflammation, and macrophage proliferation. On the contrary, lentivirus-mediated overexpression of Rheb in macrophages increased oxidized LDL-induced lipid uptake and inflammation, and the stimulatory effect of Rheb was suppressed by the mTOR (mammalian target of rapamycin) inhibitor rapamycin or the PKA (protein kinase A) activator forskolin. CONCLUSIONS: Macrophage Rheb plays important role in Western diet-induced atherosclerosis by promoting macrophage proliferation, inflammation, and lipid uptake. Inhibition of expression and function of Rheb in macrophages is beneficial to prevent diet-induced atherosclerosis.


Asunto(s)
Aterosclerosis/prevención & control , Inflamación/prevención & control , Metabolismo de los Lípidos , Macrófagos/fisiología , Proteína Homóloga de Ras Enriquecida en el Cerebro/fisiología , Animales , Proliferación Celular , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Lipoproteínas LDL/fisiología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Diana Mecanicista del Complejo 1 de la Rapamicina/fisiología , Ratones , Ratones Endogámicos C57BL , Proteína Homóloga de Ras Enriquecida en el Cerebro/deficiencia
11.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 45(2): 109-114, 2020 Feb 28.
Artículo en Inglés, Zh | MEDLINE | ID: mdl-32386033

RESUMEN

OBJECTIVES: To investigate effect of MIPU1 silence on proliferation, apoptosis, migration and invasion in U251 cells. METHODS: The shRNA recombinant plasmids targeting MIPU1 gene was transfected into U251 cells. Western blotting was used to identify the inhibitory efficiency at 72 h after transfection. The cell viability was measured by MTT colorimetric assay. Hoechest staining and caspase-3 activity were used to detect apoptosis. Then wound healing assay and transwell migration assay were applied to detect the migration and invasion of cells. RESULTS: The expression of MIPU1 protein was effectively knocked down in transfected cells (P<0.05). The cellular proliferation was obviously inhibited and apoptosis was increased in shRNA-transfected MIPU1 cells (all P<0.05). The migration and invasion ability of cells transfected with positive plasmid was lower than that in the control group (P<0.05). CONCLUSIONS: Down-regulation of MIPU1 can promote apoptosis while inhibit the proliferation, invasion, and migration of U251 cells.


Asunto(s)
Apoptosis , Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Humanos , Invasividad Neoplásica , Interferencia de ARN , ARN Interferente Pequeño , Transfección
12.
J Cell Physiol ; 234(4): 4044-4053, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30256395

RESUMEN

Nucleolin is a multifunctional phosphoprotein and is involved in protecting from myocardial ischemia/reperfusion (I/R) injury. The function of nucleolin is regulated by posttranslational modifications, including phosphorylation and glycosylation. To study whether phosphorylation of nucleolin (P-nucleolin) was involved in the protection from myocardial I/R injury. We investigated the expression pattern of P-nucleolin (Thr-76 and 84) in hearts subjected to I/R injury, or rat cardiac myoblast cells (H9C2) subjected to hydrogen peroxide (H 2 O 2 ). The results showed that the expression of P-nucleolin and the ratio of P-nucleolin/nucleolin were significantly increased both in vivo and in vitro. Mutant nucleolin was obtained by site directed mutagenesis in vitro: threonine at 76 and 84 was replaced by alanine, and we found that the protective effect of nucleolin on apoptosis induced by oxidative stress was dependent on its phosphorylation at 76 and 84 in H9C2 cells. Furthermore, the cardio-protective roles of P-nucleolin (Thr-76 and 84) in H9C2 cardiomyocytes, were attributable to the upregulation of microRNA (miR)-21. Further analysis found that P-nucleolin (Thr-76 and 84) could bind to miR-21, and P-nucleolin colocalized with argonaute 2 (Ago2) in cytoplasm and could interact with Ago2 in a RNA-independent manner under cell oxidative stress. The current study revealed that P-nucleolin (Thr-76 and 84) increased in I/R injury myocardium, P-nucleolin was indispensable to upregulate miR-21 and inhibited apoptosis induced by H 2 O 2 in H9C2 cardiomyocytes. These findings provided new insight into the molecular mechanisms of nucleolin in myocardial I/R injury and oxidative stress cells.


Asunto(s)
Apoptosis , MicroARNs/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Estrés Oxidativo , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteínas Argonautas/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Peróxido de Hidrógeno/toxicidad , Masculino , Ratones Endogámicos BALB C , MicroARNs/genética , Mutación , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Estrés Oxidativo/efectos de los fármacos , Fosfoproteínas/genética , Fosforilación , Proteínas de Unión al ARN/genética , Ratas , Transducción de Señal , Regulación hacia Arriba , Nucleolina
13.
J Cell Physiol ; 234(10): 17690-17703, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30793306

RESUMEN

Proangiogenesis is generally regarded as an effective approach for treating ischemic heart disease. Vascular endothelial growth factor (VEGF)-A is a strong and essential proangiogenic factor. Reactive oxygen species (ROS), endoplasmic reticulum (ER) stress, and autophagy are implicated in the process of angiogenesis. This study is designed to clarify the regulatory mechanisms underlying VEGF-A, ROS, ER stress, autophagy, and angiogenesis in acute myocardial infarction (AMI). A mouse model of AMI was successfully established by occluding the left anterior descending coronary artery. Compared with the sham-operated mice, the microvessel density, VEGF-A content, ROS production, expression of vascular endothelial cadherin, positive expression of 78 kDa glucose-regulated protein/binding immunoglobulin protein (GRP78/Bip), and LC3 puncta in CD31-positive endothelial cells of the ischemic myocardium were overtly elevated. Moreover, VEGF-A exposure predominantly increased the expression of beclin-1, autophagy-related gene (ATG) 4, ATG5, inositol-requiring enzyme-1 (IRE-1), GRP78/Bip, and LC3-II/LC3-I as well as ROS production in the human umbilical vein endothelial cells (HUVECs) in a dose and time-dependent manner. Both beclin-1 small interfering RNA and 3-methyladenine treatment predominantly mitigated VEGF-A-induced tube formation and migration of HUVECs, but they failed to elicit any notable effect on VEGF-A-increased expression of GRP78/Bip. Tauroursodeoxycholic acid not only obviously abolished VEGF-A-induced increase of IRE-1, GRP78/Bip, beclin-1 expression, and LC3-II/LC3-I, but also negated VEGF-A-induced tube formation and migration of HUVECs. Furthermore, N-acetyl- l-cysteine markedly abrogated VEGF-A-increased ROS production, IRE-1, GRP78/Bip, beclin-1 expression, and LC3-II/LC3-I in the HUVECs. Taken together, our data demonstrated that increased spontaneous production of VEGF-A may induce angiogenesis after AMI through initiating ROS-ER stress-autophagy axis in the vascular endothelial cells.


Asunto(s)
Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Neovascularización Fisiológica , Factor A de Crecimiento Endotelial Vascular/fisiología , Acetilcisteína/farmacología , Animales , Autofagia/efectos de los fármacos , Autofagia/fisiología , Beclina-1/antagonistas & inhibidores , Beclina-1/genética , Beclina-1/fisiología , Modelos Animales de Enfermedad , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Isquemia Miocárdica/patología , Isquemia Miocárdica/fisiopatología , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo
14.
Immunol Cell Biol ; 97(10): 902-915, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31472096

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are functionally immunosuppressive cells that are persistently increased in abundance and associated with adverse clinical outcomes in sepsis. Here, we investigated the therapeutic potential of an anaplastic lymphoma kinase inhibitor, LDK378, in cecal ligation and puncture (CLP)-induced polymicrobial sepsis and examined its effects on the recruitment of MDSCs. LDK378 significantly improved the survival of CLP-induced polymicrobial septic mice, which was paralleled by reduced organ injury, decreased release of inflammatory cytokines and decreased recruitment of MDSCs to the spleen. Importantly, LDK378 inhibited the migration of MDSCs to the spleen by blocking the CLP-mediated upregulation of CC chemokine receptor 2 (CCR2), a chemokine receptor critical for the recruitment of MDSCs. Mechanistically, LDK378 treatment blocked the CLP-induced CCR2 upregulation of MDSCs via partially inhibiting the phosphorylation of p38 and G-protein-coupled receptor kinase-2 (GRK2) in bone marrow MDSCs of septic mice. Furthermore, in vitro experiments also showed that lipopolysaccharide (LPS)-induced migration of MDSCs was similarly owing to the activation of GRK2 and upregulation of CCR2 by LPS, whereas the treatment with LDK378 partially blocked the LPS-induced phosphorylation of p38 and GRK2 and decreased the expression of CCR2 on the cell surface, therefore leading to the suppression of MDSC migration. Together, these findings unravel a novel function of LDK378 in the host response to infection and suggest that LDK378 could be a potential therapeutic agent for sepsis.


Asunto(s)
Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Células Supresoras de Origen Mieloide/metabolismo , Pirimidinas/farmacología , Receptores CCR2/metabolismo , Sepsis/metabolismo , Sepsis/patología , Bazo/patología , Sulfonas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Ciego/patología , Movimiento Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Terapia de Inmunosupresión , Inflamación/patología , Ligadura , Lipopolisacáridos , Masculino , Ratones Endogámicos BALB C , Modelos Biológicos , Células Supresoras de Origen Mieloide/efectos de los fármacos , Punciones , Sepsis/prevención & control , Transducción de Señal/efectos de los fármacos
15.
J Biochem Mol Toxicol ; 33(4): e22279, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30537341

RESUMEN

Platelet activation contributes to organs failure in inflammation and plays an important role in endotoxemia. Clopidogrel inhibits platelet aggregation and activation. However, the role of clopidogrel in modulating inflammatory progression of endotoxemia remains largely unexplored. Therefore, we investigated the role of clopidogrel on the activation of platelet and leukocytes in lipopolysaccharide (LPS)-induced inflammation in mice. Animals were treated with clopidogrel or vehicle before LPS induction. The expression of neutrophil-platelet aggregates and platelet activation and tissue factor was determined. Immunofluorescence was used to analyze platelet-leukocyte interactions and tissue factor (TF) expression on leukocytes. Clopidogrel pretreatment markedly decreased lung damage, inhibited platelet-neutrophil aggregates and TF expression. In addition, clopidogrel reduced thrombocytopenia and affected the number of circulating white blood cell in endotoxemia mice. Moreover, clopidogrel also reduced platelet shedding of CD40L and CD62P in endotoxemic mice. Taken together, clopidogrel played an important role through reducing platelet activation and inflammatory process in endotoxemia.


Asunto(s)
Plaquetas/efectos de los fármacos , Clopidogrel/farmacología , Endotoxemia/inducido químicamente , Inflamación/prevención & control , Lipopolisacáridos/toxicidad , Neutrófilos/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Animales , Plaquetas/citología , Plaquetas/metabolismo , Ligando de CD40/metabolismo , Inflamación/inducido químicamente , Interleucina-1beta/sangre , Interleucina-1beta/metabolismo , Ratones Endogámicos BALB C , Modelos Animales , Neutrófilos/citología , Neutrófilos/metabolismo , Selectina-P/metabolismo , Neumonía/inducido químicamente , Neumonía/prevención & control , Tromboplastina/metabolismo , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/metabolismo
16.
Mediators Inflamm ; 2019: 6453296, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30918470

RESUMEN

OBJECTIVES: To investigate the protective effect of ginsenoside Rg1 on relieving sepsis-induced lung inflammation and injury in vivo and in vitro. METHODS: Cultured human pulmonary epithelial cell line A549 was challenged with LPS to induce cell injury, and CLP mouse model was generated to mimic clinical condition of systemic sepsis. Rg1 was applied to cells or animals at indicated dosage. Apoptosis of cultured cells was quantified by flow cytometry, along with ELISA for inflammatory cytokines in supernatant. For septic mice, lung tissue pathology was examined, plus ELISA assay for serum cytokines. Western blotting was used to examine the activation of inflammatory pathways and ER stress marker proteins in both cells and mouse lung tissues. Reactive oxygen species (ROS) level was quantified by DCFDA kit. RESULTS: Ginsenoside Rg1 treatment remarkably suppressed apoptosis rate of LPS-induced A549 cells, relieved mouse lung tissue damage, and elevated survival rate. Rg1 treatment also rescued cells from LPS-induced intracellular ROS. In both A549 cells and mouse lung tissues, further study showed that Rg1 perfusion significantly suppressed the secretion of inflammatory cytokines including tumor necrosis factor- (TNF-) alpha and interleukin- (IL-) 6 and relieved cells from ER stress as supported by decreased expression of marker proteins via upregulating sirtuin 1 (SIRT1). CONCLUSION: Our results showed that ginsenoside Rg1 treatment effectively relieved sepsis-induced lung injury in vitro and in vivo, mainly via upregulating SIRT1 to relieve ER stress and inflammation. These findings provide new insights for unrevealing potential candidate for severe sepsis accompanied with lung injury.


Asunto(s)
Ginsenósidos/uso terapéutico , Inflamación/tratamiento farmacológico , Neumonía/tratamiento farmacológico , Sepsis/complicaciones , Sepsis/metabolismo , Sirtuina 1/metabolismo , Células A549 , Animales , Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Inflamación/metabolismo , Interleucina-6/metabolismo , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/metabolismo , Ratones , Neumonía/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
17.
J Cell Mol Med ; 22(5): 2692-2705, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29512890

RESUMEN

Hydroxysafflor Yellow A (HSYA), a most representative ingredient of Carthamus tinctorius L., had long been used in treating ischaemic cardiovascular diseases in China and exhibited prominently anticoagulant and pro-angiogenic activities, but the underlying mechanisms remained largely unknown. This study aimed to further elucidate the pro-angiogenic effect and mechanism of HSYA on ischaemic cardiac dysfunction. A C57 mouse model of acute myocardial infarction (AMI) was firstly established, and 25 mg/kg HSYA was intraperitoneally injected immediately after operation and given once, respectively, each morning and evening for 2 weeks. It was found that HSYA significantly improved ischaemia-induced cardiac haemodynamics, enhanced the survival rate, alleviated the myocardial injury and increased the expressions of CD31, vascular endothelial growth factor-A (VEGF-A) and nucleolin in the ischaemic myocardium. In addition, HSYA promoted the migration and tube formation of human umbilical vein endothelial cells (HUVECs), enhanced the expressions of nucleolin, VEGF-A and matrix metalloproteinase-9 (MMP-9) in a dose- and time-dependent manner. However, down-regulation of nucleolin expression sharply abrogated the effect mentioned above of HSYA. Further protein-RNA coimmunoprecipitation and immunoprecipitation-RT-PCR assay showed that nucleolin binded to VEGF-A and MMP-9 mRNA and overexpression of nucleolin up-regulated the mRNA expressions of VEGF-A and MMP-9 in the HUVECs through enhancing the stability of VEGF-A and MMP-9 mRNA. Furthermore, HSYA increased the mRNA expressions of VEGF-A and MMP-9 in the extract of antinucleolin antibody-precipitated protein from the heart of AMI mice. Our data revealed that nucleolin mediated the pro-angiogenic effect of HSYA through post-transcriptional regulation of VEGF-A and MMP-9 expression, which contributed to the protective effect of HSYA on ischaemic cardiac dysfunction.


Asunto(s)
Chalcona/análogos & derivados , Regulación de la Expresión Génica , Metaloproteinasa 9 de la Matriz/genética , Isquemia Miocárdica/tratamiento farmacológico , Isquemia Miocárdica/fisiopatología , Neovascularización Fisiológica , Fosfoproteínas/metabolismo , Quinonas/uso terapéutico , Proteínas de Unión al ARN/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Movimiento Celular , Chalcona/farmacología , Chalcona/uso terapéutico , Regulación de la Expresión Génica/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Isquemia Miocárdica/genética , Miocardio/patología , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Unión Proteica/efectos de los fármacos , Quinonas/farmacología , Estabilidad del ARN/efectos de los fármacos , Estabilidad del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Supervivencia , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Nucleolina
18.
J Cell Physiol ; 233(12): 9516-9525, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29968904

RESUMEN

Nucleolin is a multifunctional protein and participates in many important biological processes. Our previous study found that nucleolin protects the heart against myocardial ischemia-reperfusion injury. In this study, we aimed to investigate the role of nucleolin in doxorubicin (DOX)-induced cardiotoxicity. The expression pattern of nucleolin in hearts subjected to DOX injury was investigated, and we found that administration of DOX induced nucleolin expression significantly in vivo and in vitro. Gene transfection and RNA interference approaches were used in cardiomyocytes to investigate the function of nucleolin. Nucleolin overexpression protects cardiomyocytes against DOX-induced injury. Nucleolin-ablated cardiomyocytes become susceptible to the injury induced by DOX. The hearts of cardiac-myocyte-specific nucleolin transgenic mice are more resistant to DOX injury. Furthermore, nucleolin upregulates microRNA(miRNA)-21 expression in vivo and in vitro, and the miRNA-21 inhibitor negates the protective effect of nucleolin against injury induced by DOX. These results have demonstrated that nucleolin is involved in the regulation of DOX-induced cardiac injury and dysfunction via the regulation of miRNA-21 expression, and may be a novel therapeutic target for DOX-induced cardiotoxicity.


Asunto(s)
Cardiotoxicidad/genética , Cardiotoxicidad/prevención & control , Doxorrubicina/efectos adversos , MicroARNs/genética , Fosfoproteínas/metabolismo , Sustancias Protectoras/metabolismo , Proteínas de Unión al ARN/metabolismo , Regulación hacia Arriba , Animales , Cardiotoxicidad/patología , Muerte Celular/efectos de los fármacos , Masculino , Ratones Transgénicos , MicroARNs/metabolismo , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Especificidad de Órganos , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Nucleolina
19.
Mol Med ; 24(1): 66, 2018 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-30587103

RESUMEN

BACKGROUND: Caspase-11, a cytosolic receptor of bacterial endotoxin (lipopolysaccharide: LPS), mediates immune responses and lethality in endotoxemia and experimental sepsis. However, the upstream pathways that regulate caspase-11 activation in endotoxemia and sepsis are not fully understood. The aim of this study is to test whether TIR-domain-containing adapter-inducing interferon-ß (TRIF) signaling is critical for caspase-11-dependent immune responses and lethality in endotoxemia. METHODS: Mice of indicated genotypes were subjected to endotoxemia or cecum ligation and puncture (CLP) and monitored daily by signs of a moribund state for lethality. Serum interleukin (IL)-1α, IL-1ß, IL-6 and tumor necrosis factor (TNF) were measured by ELISA. Data were analyzed by using student's t-test or one-way ANOVA followed by post-hoc Bonferroni test. Survival data were analyzed by using the log-rank test. RESULTS: Blockade of type 1 interferon signaling or genetic deletion of TRIF or guanylate-binding proteins (GBPs) prevented caspase-11-dependent immune responses, organ injury and lethality in endotoxemia and experimental sepsis. In vitro, deletion of GBPs blocked cytosolic LPS-induced caspase-11 activation in mouse macrophages. CONCLUSIONS: These findings demonstrate that TRIF signaling is required for caspase-11-dependent immune responses and lethality in endotoxemia and sepsis, and provide novel mechanistic insights into how LPS induces caspase-11 activation during bacterial infection.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/inmunología , Caspasas/inmunología , Endotoxemia/inmunología , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Caspasas Iniciadoras , Endotoxemia/inducido químicamente , Femenino , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/inmunología , Interferón Tipo I/inmunología , Lipopolisacáridos , Macrófagos Peritoneales/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados
20.
Pancreatology ; 18(4): 370-378, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29550097

RESUMEN

OBJECTIVE: To perform a meta-analysis of all available studies on the effect of prophylactic somatostatin administration on prevention of post-endoscopic retrograde cholangiopancreatography (ERCP) pancreatitis (PEP) and post-ERCP hyperamylasemia (PEHA). METHODS: Electronic databases, including PubMed, EMBASE, the Cochrane library, and the Science Citation Index were searched to retrieve relevant trials. Randomized, placebo-controlled trials in adult patients that compared somatostatin versus placebo in prevention of PEP were included. Meta-analysis was performed using a random-effects model to assess the ratios of PEP, PEHA and post-ERCP abdominal pain. RESULTS: Total ratio of PEP of somatostatin group was significantly lower than that of placebo group. For the short-term injection or bolus injection there were no heterogeneity and no significance between the ratio of PEP of somatostatin group and placebo group. For the long-term injection subgroup there was heterogeneity, and the ratio of PEP of somatostatin group was significantly lower than that of placebo group. There was no significance between the ratio of PEP of somatostatin group and placebo group for the low-risk PEP subgroup, while the ratio of PEP of somatostatin group was significantly lower than that of placebo group for the high-risk PEP subgroup. The ratio of PEP of somatostatin group was significantly lower than that of placebo group for the long-term injection high-risk PEP subgroup. There was no significance between the ratio of PEHA of somatostatin group and placebo group for the short-term injection subgroup or bolus injection subgroup. The ratio of PEHA of somatostatin group was significantly lower than that of placebo group for the long-term injection subgroup. The total ratio of post-ERCP abdominal pain of somatostatin group was significantly lower than that of placebo group. The funnel plot of incidence of PEP and PEHA showed no asymmetry with a negative slope. CONCLUSION: Prophylactic use of long-term injection of somatostatin can significantly reduce the incidence of PEP, PEHA and post-ERCP abdominal pain for the high-risk PEP patients, while it is not necessary to be used for the low-risk PEP patients.


Asunto(s)
Colangiopancreatografia Retrógrada Endoscópica/efectos adversos , Hiperamilasemia/prevención & control , Pancreatitis/prevención & control , Somatostatina/uso terapéutico , Humanos , Hiperamilasemia/etiología , Pancreatitis/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA