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










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 12: 647070, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679812

RESUMEN

The defective eradication of invading pathogens is a major cause of death in sepsis. As professional phagocytic cells, macrophages actively engulf/kill microorganisms and play essential roles in innate immune response against pathogens. Growth differentiation factor 3 (GDF3) was previously implicated as an important modulator of inflammatory response upon acute sterile injury. In this study, administration of recombinant GDF3 protein (rGDF3) either before or after CLP surgery remarkably improved mouse survival, along with significant reductions in bacterial load, plasma pro-inflammatory cytokine levels, and organ damage. Notably, our in vitro experiments revealed that rGDF3 treatment substantially promoted macrophage phagocytosis and intracellular killing of bacteria in a dose-dependent manner. Mechanistically, RNA-seq analysis results showed that CD5L, known to be regulated by liver X receptor α (LXRα), was the most significantly upregulated gene in rGDF3-treated macrophages. Furthermore, we observed that rGDF3 could promote LXRα nuclear translocation and thereby, augmented phagocytosis activity in macrophages, which was similar as LXRα agonist GW3965 did. By contrast, pre-treating macrophages with LXRα antagonist GSK2033 abolished beneficial effects of rGDF3 in macrophages. In addition, rGDF3 treatment failed to enhance bacteria uptake and killing in LXRα-knockout (KO) macrophages. Taken together, these results uncover that GDF3 may represent a novel mediator for controlling bacterial infection.


Asunto(s)
Factor 3 de Diferenciación de Crecimiento/farmacología , Receptores X del Hígado/inmunología , Macrófagos/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Proteínas Recombinantes/farmacología , Sepsis/prevención & control , Animales , Benzoatos/farmacología , Bencilaminas/farmacología , Células Cultivadas , Citocinas/inmunología , Citocinas/metabolismo , Expresión Génica/efectos de los fármacos , Expresión Génica/inmunología , Perfilación de la Expresión Génica/métodos , Factor 3 de Diferenciación de Crecimiento/administración & dosificación , Factor 3 de Diferenciación de Crecimiento/genética , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/microbiología , Receptores X del Hígado/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Fagocitosis/inmunología , Células RAW 264.7 , Proteínas Recombinantes/administración & dosificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sepsis/inmunología , Sepsis/microbiología
2.
Cardiovasc Res ; 117(3): 890-902, 2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-32170929

RESUMEN

AIMS: Cardiac dysfunction is a prevalent comorbidity of disrupted inflammatory homeostasis observed in conditions such as sepsis (acute) or obesity (chronic). Secreted and transmembrane protein 1a (Sectm1a) has previously been implicated to regulate inflammatory responses, yet its role in inflammation-associated cardiac dysfunction is virtually unknown. METHODS AND RESULTS: Using the CRISPR/Cas9 system, we generated a global Sectm1a-knockout (KO) mouse model and observed significantly increased mortality and cardiac injury after lipopolysaccharide (LPS) injection, when compared with wild-type (WT) control. Further analysis revealed significantly increased accumulation of inflammatory macrophages in hearts of LPS-treated KO mice. Accordingly, ablation of Sectm1a remarkably increased inflammatory cytokines levels both in vitro [from bone marrow-derived macrophages (BMDMs)] and in vivo (in serum and myocardium) after LPS challenge. RNA-sequencing results and bioinformatics analyses showed that the most significantly down-regulated genes in KO-BMDMs were modulated by LXRα, a nuclear receptor with robust anti-inflammatory activity in macrophages. Indeed, we identified that the nuclear translocation of LXRα was disrupted in KO-BMDMs when treated with GW3965 (LXR agonist), resulting in higher levels of inflammatory cytokines, compared to GW3965-treated WT-cells. Furthermore, using chronic inflammation model of high-fat diet (HFD) feeding, we observed that infiltration of inflammatory monocytes/macrophages into KO-hearts were greatly increased and accordingly, worsened cardiac function, compared to WT-HFD controls. CONCLUSION: This study defines Sectm1a as a new regulator of inflammatory-induced cardiac dysfunction through modulation of LXRα signalling in macrophages. Our data suggest that augmenting Sectm1a activity may be a potential therapeutic approach to resolve inflammation and associated cardiac dysfunction.


Asunto(s)
Cardiopatías/metabolismo , Inflamación/metabolismo , Receptores X del Hígado/metabolismo , Macrófagos/metabolismo , Proteínas de la Membrana/deficiencia , Función Ventricular Izquierda , Animales , Citocinas/genética , Citocinas/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Cardiopatías/etiología , Cardiopatías/genética , Cardiopatías/fisiopatología , Inflamación/etiología , Inflamación/genética , Inflamación/fisiopatología , Mediadores de Inflamación/metabolismo , Lipopolisacáridos , Receptores X del Hígado/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Células RAW 264.7 , Ratas Sprague-Dawley , Transducción de Señal
3.
Mol Ther ; 29(3): 1294-1311, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33279722

RESUMEN

Tissue-resident macrophages (TRMs) are sentinel cells for maintaining tissue homeostasis and organ function. In this study, we discovered that lipopolysaccharide (LPS) administration dramatically reduced TRM populations and suppressed their self-renewal capacities in multiple organs. Using loss- and gain-of-function approaches, we define Sectm1a as a novel regulator of TRM self-renewal. Specifically, at the earlier stage of endotoxemia, Sectm1a deficiency exaggerated acute inflammation-induced reduction of TRM numbers in multiple organs by suppressing their proliferation, which was associated with more infiltrations of inflammatory monocytes/neutrophils and more serious organ damage. By contrast, administration of recombinant Sectm1a enhanced TRM populations and improved animal survival upon endotoxin challenge. Mechanistically, we identified that Sectm1a-induced upregulation in the self-renewal capacity of TRM is dependent on GITR-activated T helper cell expansion and cytokine production. Meanwhile, we found that TRMs may play an important role in protecting local vascular integrity during endotoxemia. Our study demonstrates that Sectm1a contributes to stabling TRM populations through maintaining their self-renewal capacities, which benefits the host immune response to acute inflammation. Therefore, Sectm1a may serve as a new therapeutic agent for the treatment of inflammatory diseases.


Asunto(s)
Proteína Relacionada con TNFR Inducida por Glucocorticoide/metabolismo , Memoria Inmunológica/inmunología , Inflamación/complicaciones , Macrófagos/inmunología , Proteínas de la Membrana/metabolismo , Monocitos/inmunología , Insuficiencia Multiorgánica/prevención & control , Animales , Proteína Relacionada con TNFR Inducida por Glucocorticoide/genética , Homeostasis , Proteínas de la Membrana/genética , Ratones , Insuficiencia Multiorgánica/etiología , Linfocitos T Colaboradores-Inductores/inmunología
4.
Shock ; 55(1): 14-23, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-32590701

RESUMEN

ABSTRACT: Macrophage, as an integral component of the immune system and the first responder to local damage, is on the front line of defense against infection. Over the past century, the prevailing view of macrophage origin states that all macrophage populations resided in tissues are terminally differentiated and replenished by monocytes from bone-marrow progenitors. Nonetheless, this theory has been reformed by ground-breaking discoveries from the past decades. It is now believed that tissue-resident macrophages (TRMs) are originated from the embryonic precursors and seeded in tissue prenatally. They can replenish via self-renewal throughout the lifespan. Indeed, recent studies have demonstrated that tissue-resident macrophages should not be classified by the over-simplified macrophage polarization (M1/M2) dogma during inflammation. Moreover, multiple lines of evidence have indicated that tissue-resident macrophages play critical roles in maintaining tissue homeostasis and facilitating tissue repair through controlling infection and resolving inflammation. In this review, we summarize the properties of resident macrophages in the lung, spleen, and heart, and further highlight the impact of TRM populations on inflammation control and tissue repair. We also discuss the potential role of local proliferation in maintaining a physiologically stable TRM pool in response to acute inflammation.


Asunto(s)
Inflamación/etiología , Macrófagos/fisiología , Homeostasis/fisiología , Humanos , Inflamación/patología
5.
Cells ; 9(9)2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32839388

RESUMEN

Cardiac cells can adapt to pathological stress-induced energy crisis by shifting from fatty acid oxidation to glycolysis. However, the use of glucose-insulin-potassium (GIK) solution in patients undergoing cardiac surgery does not alleviate ischemia/reperfusion (I/R)-induced energy shortage. This indicates that insulin-mediated translocation of glucose transporter-4 (Glut-4) is impaired in ischemic hearts. Indeed, cardiac myocytes contain two intracellular populations of Glut-4: an insulin-dependent non-endosomal pool (also referred to as Glut-4 storage vesicles, GSVs) and an insulin-independent endosomal pool. Tumor susceptibility gene 101 (Tsg101) has been implicated in the endosomal recycling of membrane proteins. In this study, we aimed to examine whether Tsg101 regulated the sorting and re-distribution of Glut-4 to the sarcolemma membrane of cardiomyocytes under basal and ischemic conditions, using gain- and loss-of-function approaches. Forced overexpression of Tsg101 in mouse hearts and isolated cardiomyocytes could promote Glut-4 re-distribution to the sarcolemma, leading to enhanced glucose entry and adenosine triphosphate (ATP) generation in I/R hearts which in turn, attenuation of I/R-induced cardiac dysfunction. Conversely, knockdown of Tsg101 in cardiac myocytes exhibited opposite effects. Mechanistically, we identified that Tsg101 could interact and co-localize with Glut-4 in the sarcolemma membrane of cardiomyocytes. Our findings define Tsg101 as a novel regulator of cardiac Glut-4 trafficking, which may provide a new therapeutic strategy for the treatment of ischemic heart disease.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Miocitos Cardíacos/metabolismo , Sarcolema/metabolismo , Factores de Transcripción/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratas
6.
J Immunol ; 205(6): 1633-1643, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32769121

RESUMEN

The inability to effectively control invading bacteria or other pathogens is a major cause of multiple organ dysfunction and death in sepsis. As the first-line defense of the immune system, macrophages play a crucial role in the removal of pathogens during sepsis. In this study, we define secreted and transmembrane 1A (Sectm1a) as a novel ligand of glucocorticoid-induced TNFR (GITR) that greatly boosts macrophage phagocytosis and bactericidal capacity. Using a global Sectm1a knockout (KO) mouse model, we observed that Sectm1a deficiency significantly suppressed phagocytosis and bactericidal activity in both recruited macrophages and tissue-resident macrophages, which consequently aggravated bacterial burden in the blood and multiple organs and further increased systemic inflammation, leading to multiple organ injury and increased mortality during polymicrobial sepsis. By contrast, treatment of septic mice with recombinant Sectm1a protein (rSectm1a) not only promoted macrophage phagocytosis and bactericidal activity but also significantly improved survival outcome. Mechanistically, we identified that Sectm1a could bind to GITR in the surface of macrophages and thereby activate its downstream PI3K-Akt pathway. Accordingly, rSectm1a-mediated phagocytosis and bacterial killing were abolished in macrophages by either KO of GITR or pharmacological inhibition of the PI3K-Akt pathway. In addition, rSectm1a-induced therapeutic effects on sepsis injury were negated in GITR KO mice. Taken together, these results uncover that Sectm1a may represent a novel target for drug development to control bacterial dissemination during sepsis or other infectious diseases.


Asunto(s)
Proteína Relacionada con TNFR Inducida por Glucocorticoide/metabolismo , Macrófagos/fisiología , Proteínas de la Membrana/metabolismo , Insuficiencia Multiorgánica/inmunología , Sepsis/inmunología , Animales , Proteína Relacionada con TNFR Inducida por Glucocorticoide/genética , Humanos , Tolerancia Inmunológica , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Oncogénica v-akt/metabolismo , Fagocitosis , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal
7.
Redox Biol ; 32: 101453, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32057709

RESUMEN

Currently, most antioxidants do not show any favorable clinical outcomes in reducing myocardial ischemia-reperfusion (I/R) injury, suggesting an urgent need for exploring a new regulator of redox homeostasis in I/R hearts. Here, using heart-specific transgenic (TG) and knockdown (KD) mouse models, tumor susceptibility gene 101 (Tsg101) is defined as a novel cardiac-protector against I/R-triggered oxidative stress. RNA sequencing and bioinformatics data surprisingly reveal that most upregulated genes in Tsg101-TG hearts are transcribed by Nrf2. Accordingly, pharmacological inhibition of Nrf2 offsets Tsg101-elicited cardio-protection. Mechanistically, Tsg101 interacts with SQSTM1/p62 through its PRR domain, and promotes p62 aggregation, leading to recruitment of Keap1 for degradation by autophagosomes and release of Nrf2 to the nucleus. Furthermore, knockout of p62 abrogates Tsg101-induced cardio-protective effects during I/R. Hence, our findings uncover a previously unrecognized role of Tsg101 in the regulation of p62/Keap1/Nrf2 signaling cascades and provide a new strategy for the treatment of ischemic heart disease.


Asunto(s)
Autofagia , Factor 2 Relacionado con NF-E2 , Animales , Proteínas de Unión al ADN , Complejos de Clasificación Endosomal Requeridos para el Transporte , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Ratones , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Proteína Sequestosoma-1/metabolismo , Factores de Transcripción
8.
Cells ; 9(1)2020 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-31947892

RESUMEN

Macrophages are critical for regulation of inflammatory response during endotoxemia and septic shock. However, the mediators underlying their regulatory function remain obscure. Growth differentiation factor 3 (GDF3), a member of transforming growth factor beta (TGF-ß) superfamily, has been implicated in inflammatory response. Nonetheless, the role of GDF3 in macrophage-regulated endotoxemia/sepsis is unknown. Here, we show that serum GDF3 levels in septic patients are elevated and strongly correlate with severity of sepsis and 28-day mortality. Interestingly, macrophages treated with recombinant GDF3 protein (rGDF3) exhibit greatly reduced production of pro-inflammatory cytokines, comparing to controls upon endotoxin challenge. Moreover, acute administration of rGDF3 to endotoxin-treated mice suppresses macrophage infiltration to the heart, attenuates systemic and cardiac inflammation with less pro-inflammatory macrophages (M1) and more anti-inflammatory macrophages (M2), as well as prolongs mouse survival. Mechanistically, GDF3 is able to activate Smad2/Smad3 phosphorylation, and consequently inhibits the expression of nod-like receptor protein-3 (NLRP3) in macrophages. Accordingly, blockade of Smad2/Smad3 phosphorylation with SB431542 significantly offsets rGDF3-mediated anti-inflammatory effects. Taken together, this study uncovers that GDF3, as a novel sepsis-associated factor, may have a dual role in the pathophysiology of sepsis. Acute administration of rGDF3 into endotoxic shock mice could increase survival outcome and improve cardiac function through anti-inflammatory response by suppression of M1 macrophage phenotype. However, constitutive high levels of GDF3 in human sepsis patients are associated with lethality, suggesting that GDF3 may promote macrophage polarization toward M2 phenotype which could lead to immunosuppression.


Asunto(s)
Factor 3 de Diferenciación de Crecimiento/metabolismo , Corazón/fisiopatología , Inflamación/patología , Macrófagos/patología , Sepsis/prevención & control , Sepsis/fisiopatología , Adulto , Animales , Estudios de Casos y Controles , Polaridad Celular/efectos de los fármacos , Citocinas/biosíntesis , Endotoxinas , Factor 3 de Diferenciación de Crecimiento/sangre , Factor 3 de Diferenciación de Crecimiento/genética , Humanos , Inflamación/sangre , Ratones Endogámicos C57BL , Modelos Biológicos , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Sepsis/sangre , Proteínas Smad/metabolismo , Bazo/patología , Análisis de Supervivencia , Resultado del Tratamiento
9.
J Biol Chem ; 294(48): 18057-18068, 2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31619520

RESUMEN

Cardiac mitochondrial damage and subsequent inflammation are hallmarks of endotoxin-induced myocardial depression. Activation of the Parkin/PTEN-induced kinase 1 (PINK1) pathway has been shown to promote autophagy of damaged mitochondria (mitophagy) and to protect from endotoxin-induced cardiac dysfunction. Tumor susceptibility gene 101 (TSG101) is a key member of the endosomal recycling complexes required for transport, which may affect autophagic flux. In this study, we investigated whether TSG101 regulates mitophagy and influences the outcomes of endotoxin-induced myocardial dysfunction. TSG101 transgenic and knockdown mice underwent endotoxin/lipopolysaccharide treatment (10 µg/g) and were assessed for survival, cardiac function, systemic/local inflammation, and activity of mitophagy mediators in the heart. Upon endotoxin challenge and compared with WT mice, TSG101 transgenic mice exhibited increased survival, preserved cardiac contractile function, reduced inflammation, and enhanced mitophagy activation in the heart. By contrast, TSG101 knockdown mice displayed opposite phenotypes during endotoxemia. Mechanistically, both coimmunoprecipitation assays and coimmunofluorescence staining revealed that TSG101 directly binds to Parkin in the cytosol of myocytes and facilitates translocation of Parkin from the cytosol to the mitochondria. Our results indicate that TSG101 elevation could protect against endotoxin-triggered myocardial injury by promoting Parkin-induced mitophagy.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Cardiopatías/metabolismo , Lipopolisacáridos/toxicidad , Mitocondrias Cardíacas/metabolismo , Mitofagia/efectos de los fármacos , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Proteínas de Unión al ADN/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Cardiopatías/inducido químicamente , Cardiopatías/genética , Cardiopatías/patología , Masculino , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/genética , Mitocondrias Cardíacas/patología , Mitofagia/genética , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/genética , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Factores de Transcripción/genética , Ubiquitina-Proteína Ligasas/genética
10.
J Biol Chem ; 294(27): 10438-10448, 2019 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-31118273

RESUMEN

The initiation and development of diabetes are mainly ascribed to the loss of functional ß-cells. Therapies designed to regenerate ß-cells provide great potential for controlling glucose levels and thereby preventing the devastating complications associated with diabetes. This requires detailed knowledge of the molecular events and underlying mechanisms in this disorder. Here, we report that expression of microRNA-223 (miR-223) is up-regulated in islets from diabetic mice and humans, as well as in murine Min6 ß-cells exposed to tumor necrosis factor α (TNFα) or high glucose. Interestingly, miR-223 knockout (KO) mice exhibit impaired glucose tolerance and insulin resistance. Further analysis reveals that miR-223 deficiency dramatically suppresses ß-cell proliferation and insulin secretion. Mechanistically, using luciferase reporter gene assays, histological analysis, and immunoblotting, we demonstrate that miR-223 inhibits both forkhead box O1 (FOXO1) and SRY-box 6 (SOX6) signaling, a unique bipartite mechanism that modulates expression of several ß-cell markers (pancreatic and duodenal homeobox 1 (PDX1), NK6 homeobox 1 (NKX6.1), and urocortin 3 (UCN3)) and cell cycle-related genes (cyclin D1, cyclin E1, and cyclin-dependent kinase inhibitor P27 (P27)). Importantly, miR-223 overexpression in ß-cells could promote ß-cell proliferation and improve ß-cell function. Taken together, our results suggest that miR-223 is a critical factor for maintaining functional ß-cell mass and adaptation during metabolic stress.


Asunto(s)
Proteína Forkhead Box O1/metabolismo , MicroARNs/metabolismo , Factores de Transcripción SOXD/metabolismo , Regiones no Traducidas 3' , Animales , Línea Celular , Proliferación Celular , Ciclina D1/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Proteína Forkhead Box O1/química , Proteína Forkhead Box O1/genética , Prueba de Tolerancia a la Glucosa , Proteínas de Homeodominio/metabolismo , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Ratas , Factores de Transcripción SOXD/química , Factores de Transcripción SOXD/genética , Transducción de Señal , Transactivadores/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba/efectos de los fármacos
11.
FASEB J ; 33(6): 7451-7466, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30884248

RESUMEN

Development of physiologic cardiac hypertrophy has primarily been ascribed to the IGF-1 and its receptor, IGF-1 receptor (IGF-1R), and subsequent activation of the protein kinase B (Akt) pathway. However, regulation of endosome-mediated recycling and degradation of IGF-1R during physiologic hypertrophy has not been investigated. In a physiologic hypertrophy model of treadmill-exercised mice, we observed that levels of tumor susceptibility gene 101 (Tsg101), a key member of the endosomal sorting complex required for transport, were dramatically elevated in the heart compared with sedentary controls. To determine the role of Tsg101 on physiologic hypertrophy, we generated a transgenic (TG) mouse model with cardiac-specific overexpression of Tsg101. These TG mice exhibited a physiologic-like cardiac hypertrophy phenotype at 8 wk evidenced by: 1) the absence of cardiac fibrosis, 2) significant improvement of cardiac function, and 3) increased total and plasma membrane levels of IGF-1R and increased phosphorylation of Akt. Mechanistically, we identified that Tsg101 interacted with family-interacting protein 3 (FIP3) and IGF-1R, thereby stabilizing FIP3 and enhancing recycling of IGF-1R. In vitro, adenovirus-mediated overexpression of Tsg101 in neonatal rat cardiomyocytes resulted in cell hypertrophy, which was blocked by addition of monensin, an inhibitor of endosome-mediated recycling, and by small interfering RNA-mediated knockdown (KD) of FIP3. Furthermore, cardiac-specific KD of Tsg101 showed a significant reduction in levels of endosomal recycling compartment members (Rab11a and FIP3), IGF-1R, and Akt phosphorylation. Most interestingly, Tsg101-KD mice failed to develop cardiac hypertrophy after intense treadmill training. Taken together, our data identify Tsg101 as a novel positive regulator of physiologic cardiac hypertrophy through facilitating the FIP3-mediated endosomal recycling of IGF-1R.-Essandoh, K., Deng, S., Wang, X., Jiang, M., Mu, X., Peng, J., Li, Y., Peng, T., Wagner, K.-U., Rubinstein, J., Fan, G.-C. Tsg101 positively regulates physiologic-like cardiac hypertrophy through FIP3-mediated endosomal recycling of IGF-1R.


Asunto(s)
Cardiomegalia/fisiopatología , Proteínas de Unión al ADN/fisiología , Complejos de Clasificación Endosomal Requeridos para el Transporte/fisiología , Endosomas/metabolismo , Quinasa I-kappa B/fisiología , Receptor IGF Tipo 1/metabolismo , Factores de Transcripción/fisiología , Animales , Femenino , Perfilación de la Expresión Génica , Masculino , Ratones , Ratas
12.
Fish Shellfish Immunol ; 81: 21-28, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29981472

RESUMEN

Mucosal immune system is one of the most vital components in the innate immunity and constitutes the first line of host defense against bacterial infections, especially for the teleost, which live in the pathogen-rich aquatic environment. Cathepsins, a superfamily of hydrolytic enzymes produced and enclosed within lysosomes, play multiple roles at physiological and pathological states. In this regard, we sought here to identify Cathepsin A in turbot (SmCTSA), characterize its mucosal expression patterns following Vibrio anguillarum and Streptococcus iniae infections in mucosal tissues, and explore its binding ability with three microbial ligands for the first time. The SmCTSA was 2631 bp long containing a 1422 bp open reading frame (ORF) that encoded 473 amino acids. Phylogenetic analysis revealed that SmCTSA showed the closest relationship to half-smooth tongue sole (Cynoglossus semilaevis). In addition, SmCTSA was ubiquitously expressed in all examined healthy tissues, with high expression levels in head kidney (HK) and intestine, while the lowest expression level in blood. Moreover, SmCTSA was significantly differentially expressed at least two timepoints in each mucosal tissue, suggesting its potential important roles in innate immune responses of turbot. Finally, in vitro assays showed that recombinant SmCTSA bound Lipopolysaccharide (LPS) with high affinity, and lipoteichoic acid (LTA) and peptidoglycan (PGN) with relatively low affinity. This study provides valuable data for understanding the roles of ctsa in the host defense against bacterial infections.


Asunto(s)
Catepsina A/metabolismo , Enfermedades de los Peces/inmunología , Peces Planos/inmunología , Inmunidad Mucosa , Membrana Mucosa/inmunología , Animales , Sitios de Unión , Catepsina A/genética , Enfermedades de los Peces/microbiología , Expresión Génica , Regulación de la Expresión Génica , Inmunidad Innata , Ligandos , Lipopolisacáridos/metabolismo , Membrana Mucosa/microbiología , Filogenia , ARN Mensajero/metabolismo , Alimentos Marinos/microbiología , Infecciones Estreptocócicas/inmunología , Streptococcus iniae , Vibrio , Vibriosis/inmunología
13.
Cell Rep ; 23(12): 3607-3620, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29925002

RESUMEN

Exposure to cold temperature is well known to upregulate heat shock protein (Hsp) expression and recruit and/or activate brown adipose tissue and beige adipocytes in humans and animals. However, whether and how Hsps regulate adipocyte function for energy homeostatic responses is poorly understood. Here, we demonstrate a critical role of Hsp20 as a negative regulator of adipocyte function. Deletion of Hsp20 enhances non-shivering thermogenesis and suppresses inflammatory responses, leading to improvement of glucose and lipid metabolism under both chow diet and high-fat diet conditions. Mechanistically, Hsp20 controls adipocyte function by interacting with the subunit of the ubiquitin ligase complex, F-box only protein 4 (FBXO4), and regulating the ubiquitin-dependent degradation of peroxisome proliferation activated receptor gamma (PPARγ). Indeed, Hsp20 deficiency mimics and enhances the pharmacological effects of the PPARγ agonist rosiglitazone. Together, our findings suggest a role of Hsp20 in mediating adipocyte function by linking ß-adrenergic signaling to PPARγ activity.


Asunto(s)
Adipocitos/metabolismo , Proteínas F-Box/metabolismo , Proteínas del Choque Térmico HSP20/metabolismo , PPAR gamma/metabolismo , Ubiquitinación , Adipocitos/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Adiposidad/efectos de los fármacos , Animales , Frío , Metabolismo Energético/efectos de los fármacos , Glucosa/metabolismo , Proteínas del Choque Térmico HSP20/deficiencia , Proteínas del Choque Térmico HSP20/genética , Inflamación/patología , Resistencia a la Insulina , Metabolismo de los Lípidos/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/patología , Estabilidad Proteica/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Rosiglitazona/farmacología , Ubiquitinación/efectos de los fármacos
14.
Shock ; 49(4): 429-441, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28650928

RESUMEN

Septic shock increases vascular permeability, leading to multiple organ failure including cardiac dysfunction, a major contributor to septic death. Podosome, an actin-based dynamic membrane structure, plays critical roles in extracellular matrix degradation and angiogenesis. However, whether podosome contributes to endothelial barrier dysfunction during septic shock remains unknown. In this study, we found that the endothelial hyperpermeability, stimulated by phorbol 12-myristate 13-acetate and thrombin, was accompanied by increased formation of podosome clusters at the cell periphery, indicating a positive correlation between podosome clusters and endothelial leakage. Interestingly, we observed that circulating exosomes collected from septic mice were able to stimulate podosome cluster formation in cardiac endothelial cells, together with increased permeability in vitro/in vivo and cardiac dysfunction. Mechanistically, we identified that septic exosomes contained higher levels of reactive oxygen species (ROS) than normal ones, which were effectively transported to endothelial cells (ECs). Depletion of ROS in septic exosomes significantly reduced their capacity for promoting podosome cluster formation and thereby dampened vascular leakage. Finally, we elucidated that podosome cluster-induced endothelial hyperpermeability was associated with fragmentation/depletion of zonula occludens-1 (ZO-1) at the cell periphery. Our results demonstrate that septic exosomes were enriched with high amounts of ROS, which can be transported to ECs, leading to the generation of podosome clusters in target ECs and thereby, causing ZO-1 relocation, vascular leakage, and cardiac dysfunction.


Asunto(s)
Exosomas/metabolismo , Podosomas/metabolismo , Sepsis/metabolismo , Animales , Western Blotting , Permeabilidad Capilar/fisiología , Células Endoteliales/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Especies Reactivas de Oxígeno/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
15.
Sci Rep ; 6: 37535, 2016 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-27857228

RESUMEN

Dopamine is an important regulator of renal natriuresis and is critical for the adaptation of many animals to changing environmental salinity. However, the molecular mechanisms through which dopamine promotes this adaptation remain poorly understood. We studied the effects of dopamine on renal hypo-osmoregulation in the euryhaline fish Scatophagus argus (S. argus) during abrupt transfer from seawater (SW) to freshwater (FW). Following the transfer, serum dopamine concentration was decreased, and dopamine activated expression of the dopamine receptor 1 (designated SaDRD1) in the kidney, triggering the osmoregulatory signaling cascade. SaDRD1 protein is expressed in the renal proximal tubule cells in vivo, and is localized to the cell membrane of renal primary cells in vitro. Knockdown of SaDRD1 mRNA by siRNA significantly increased Na+/K+-ATPase (NKA) activity in cultured renal primary cells in vitro, suggesting that expression of SaDRD1 may oppose the activity of NKA. We demonstrate that exogenous dopamine enhances the response of NKA to hyposaline stress after transferring primary renal cells from isosmotic medium to hypoosmotic medium. Our results indicate that dopamine regulation via SaDRD1 ignited the renal dopaminergic system to balance the osmotic pressure through inhibiting NKA activity, providing a new perspective on the hyposaline adaptation of fish.


Asunto(s)
Adaptación Fisiológica/genética , Dopamina/metabolismo , Osmorregulación/genética , Receptores de Dopamina D1/metabolismo , Animales , Peces/genética , Peces/metabolismo , Agua Dulce , Branquias/fisiología , Presión Osmótica , Receptores de Dopamina D1/genética , Salinidad , Agua de Mar
16.
J Biol Chem ; 291(38): 20247-59, 2016 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-27502281

RESUMEN

Recent studies have shown that myocardial ischemia/reperfusion (I/R)-induced necrosis can be controlled by multiple genes. In this study, we observed that both strands (5p and 3p) of miR-223 were remarkably dysregulated in mouse hearts upon I/R. Precursor miR-223 (pre-miR-223) transgenic mouse hearts exhibited better recovery of contractile performance over reperfusion period and lesser degree of myocardial necrosis than wild type hearts upon ex vivo and in vivo myocardial ischemia. Conversely, pre-miR-223 knock-out (KO) mouse hearts displayed opposite effects. Furthermore, we found that the RIP1/RIP3/MLKL necroptotic pathway and inflammatory response were suppressed in transgenic hearts, whereas they were activated in pre-miR-223 KO hearts upon I/R compared with wild type controls. Accordingly, treatment of pre-miR-223 KO mice with necrostatin-1s, a potent necroptosis inhibitor, significantly decreased I/R-triggered cardiac necroptosis, infarction size, and dysfunction. Mechanistically, we identified two critical cell death receptors, TNFR1 and DR6, as direct targets of miR-223-5p, whereas miR-223-3p directly suppressed the expression of NLRP3 and IκB kinase α, two important mediators known to be involved in I/R-induced inflammation and cell necroptosis. Our findings indicate that miR-223-5p/-3p duplex works together and cooperatively inhibits I/R-induced cardiac necroptosis at multiple layers. Thus, pre-miR-223 may constitute a new therapeutic agent for the treatment of ischemic heart disease.


Asunto(s)
MicroARNs/biosíntesis , Daño por Reperfusión Miocárdica/metabolismo , Animales , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Imidazoles/farmacología , Indoles/farmacología , Ratones , Ratones Noqueados , MicroARNs/genética , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Necrosis , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/genética
17.
Mitochondrial DNA A DNA Mapp Seq Anal ; 27(4): 2691-2, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27158788

RESUMEN

The striped scat Selenotoca multifasciata is an ornamental and commercial fish in Asia. In the present study, we sequenced and annotated the complete mitochondrial genome of Selenotoca multifasciata. Its total length is 16,646 bp, and the mitochondrial genome is composed of 13 protein-coding genes, 2 rRNA genes, 22 tRNA genes and a non-coding control region. ND6 and eight tRNA genes were encoded on the light strand, whereas the remaining genes located on the heavy strand (H-strand). All the 16 mitochondrial genomes of the suborder Acanthuroidei available in GenBank were employed for phylogenetic analysis, and the result showed a close relationship between Selenotoca multifasciata and Scatophagus argus. This mitochondrial information may benefit relative ecological and phylogenetic studies.


Asunto(s)
Genoma Mitocondrial/genética , Perciformes/genética , Animales , Orden Génico/genética , Genes Mitocondriales/genética , Perciformes/clasificación , Filogenia , ARN Ribosómico/genética , ARN de Transferencia/genética , Análisis de Secuencia de ADN
18.
J Biol Chem ; 291(30): 15700-13, 2016 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-27226563

RESUMEN

MicroRNAs (miRNAs) have been extensively examined in pathological cardiac hypertrophy. However, few studies focused on profiling the miRNA alterations in physiological hypertrophic hearts. In this study we generated a transgenic mouse model with cardiac-specific overexpression of miR-223. Our results showed that elevation of miR-223 caused physiological cardiac hypertrophy with enhanced cardiac function but no fibrosis. Using the next generation RNA sequencing, we observed that most of dys-regulated genes (e.g. Atf3/5, Egr1/3, Sfrp2, Itgb1, Ndrg4, Akip1, Postn, Rxfp1, and Egln3) in miR-223-transgenic hearts were associated with cell growth, but they were not directly targeted by miR-223. Interestingly, these dys-regulated genes are known to regulate the Akt signaling pathway. We further identified that miR-223 directly interacted with 3'-UTRs of FBXW7 and Acvr2a, two negative regulators of the Akt signaling. However, we also validated that miR-223 directly inhibited the expression of IGF-1R and ß1-integrin, two positive regulators of the Akt signaling. Lastly, Western blotting did reveal that Akt was activated in miR-223-overexpressing hearts. Adenovirus-mediated overexpression of miR-223 in neonatal rat cardiomyocytes induced cell hypertrophy, which was blocked by the addition of MK2206, a specific inhibitor of Akt Taken together, these data represent the first piece of work showing that miR-223 tips the balance of promotion and inactivation of Akt signaling cascades toward activation of Akt, a key regulator of physiological cardiac hypertrophy. Thus, our study suggests that the ultimate phenotype outcome of a miRNA may be decided by the secondary net effects of the whole target network rather than by several primary direct targets in an organ/tissue.


Asunto(s)
Cardiomegalia/metabolismo , Regulación de la Expresión Génica , MicroARNs/biosíntesis , Transducción de Señal , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Adenoviridae , Animales , Cardiomegalia/genética , Cardiomegalia/patología , Modelos Animales de Enfermedad , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD , Ratones , Ratones Transgénicos , MicroARNs/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Transducción Genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
19.
Gene ; 561(1): 6-14, 2015 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-25550048

RESUMEN

In the present study, we cloned the full-length cDNAs of FOXL2, Cyp19a1a and Cyp19a1b and analyzed their expression patterns during gonadal development in spotted scat, Scatophagus argus. All three genes were expressed in ovaries and testes but showed sexual dimorphism. At early stages of gonadal development, the expression of FOXL2 in ovaries was higher than testes. FOXL2 expression deceased gradually as gonadal development continued, and reached the lowest level at the mature stage. Cyp19a1a and Cyp19a1b were expressed coordinately with FOXL2, except at the early vitellogenic stage in the ovary. The expression of FOXL2, Cyp19a1a and Cyp19a1b was mainly localized in granulosa cells of ovaries. In S. argus testes, strong expression of FOXL2 gene was observed in the interstitial cells including tubules and Leydig cells, while Cyp19a1a and Cyp19a1b were mainly expressed in Sertoli cells throughout gametogenesis. These results show that FOXL2 plays an essential role in sexual development, and imply that it may regulate Cyp19a1a and Cyp19a1b expression in S. argus.


Asunto(s)
Aromatasa/genética , Factores de Transcripción Forkhead/genética , Ovario/embriología , Perciformes/embriología , Testículo/embriología , Secuencia de Aminoácidos , Animales , Aromatasa/biosíntesis , Secuencia de Bases , Clonación Molecular , Femenino , Factores de Transcripción Forkhead/biosíntesis , Gametogénesis/fisiología , Regulación del Desarrollo de la Expresión Génica , Células de la Granulosa/metabolismo , Células Intersticiales del Testículo/metabolismo , Masculino , Túbulos Seminíferos/metabolismo , Análisis de Secuencia de ADN , Células de Sertoli/metabolismo , Desarrollo Sexual/genética , Desarrollo Sexual/fisiología
20.
Gen Comp Endocrinol ; 215: 25-35, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25304824

RESUMEN

Scatophagus argus, a euryhaline fish, is notable for its ability to tolerate a wide range of environmental salinities and especially for its tolerance to a rapid, marked reduction in salinity. Therefore, S. argus is a good model for studying the molecular mechanisms mediating abrupt hyperosmoregulation. The serum osmotic pressure decreased steeply within one hour after transferring S. argus from seawater (SW) to freshwater (FW) and remained at new balance throughout the duration of one week. To explain this phenomenon and understand the molecular responses to an abrupt hypoosmotic shock, hypoosmotic stress responsive genes were identified by constructing two suppression subtractive hybridization (SSH) cDNA libraries from the kidneys of S. argus that had been transferred from SW to FW. After trimming and blasting, 52 ESTs were picked out from the subtractive library. Among them, 11 genes were significantly up-regulated (p < 0.05). The kinetics studies of gene expression levels were conducted for 1 week after the transfer using quantitative real-time PCR. A significant variation in the expression of these genes occurred within 12h after the hypoosmotic shock, except for growth hormone (GH) and polyadenylate binding protein 1 (PBP1), which were significantly up-regulated 2 days post-transfer. Our results suggest different functional roles for these genes in response to hypoosmotic stress during the stress response phase (1 hpt-12 hpt) and stable phase (12 hpt-7 dpt). Furthermore, the plasma growth hormone level was detected to be significantly elevated at 1 hpt and 24 hpt following abrupt hypoosmotic shock. Meanwhile, several hematological parameters, hemoglobin (HGB), red blood cell (RBC) and mean cellular hemoglobin concentration (MCHC), were observed to be significantly increased at 12 hpt and 2 dpt compared with that of control group. Our results provide a solid basis from which to conduct future studies on the osmoregulatory mechanisms in the euryhaline fish.


Asunto(s)
Biomarcadores/metabolismo , Proteínas de Peces/genética , Regulación de la Expresión Génica , Riñón/metabolismo , Presión Osmótica , Perciformes/genética , Animales , Biblioteca de Genes , Hormona del Crecimiento/sangre , Perciformes/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Salinidad , Técnicas de Hibridación Sustractiva
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...