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











Base de dados
Intervalo de ano de publicação
1.
Front Physiol ; 14: 1256852, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37621762

RESUMO

Cardiovascular disease is a leading cause of death worldwide. Loss of cardiomyocytes that occurs during many types of damage to the heart such as ischemic injury and stress caused by pressure overload, diminishes cardiac function due to their limited regenerative capacity and promotes remodeling, which further damages the heart. Cardiomyocyte death occurs through two primary mechanisms, necrosis and apoptosis. Apoptosis is a highly regulated form of cell death that can occur through intrinsic (mitochondrial) or extrinsic (receptor mediated) pathways. Extrinsic apoptosis occurs through a subset of Tumor Necrosis Receptor (TNF) family receptors termed "Death Receptors." While some ligands for death receptors have been extensively studied in the heart, such as TNF-α, others have been virtually unstudied. One poorly characterized cardiac TNF related ligand is TNF-Related Apoptosis Inducing Ligand (TRAIL). TRAIL binds to two apoptosis-inducing receptors, Death Receptor (DR) 4 and DR5. There are also three decoy TRAIL receptors, Decoy Receptor (DcR) 1, DcR2 and osteoprotegerin (OPG). While TRAIL has been extensively studied in the cancer field due to its ability to selectively induce apoptosis in transformed cell types, emerging clinical evidence points towards a role for TRAIL and its receptors in cardiac pathology. This article will highlight our current understanding of TRAIL and its receptors in normal and pathological conditions in the heart.

3.
Am J Physiol Heart Circ Physiol ; 321(4): H633-H649, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34415184

RESUMO

ß-Adrenergic receptors (ßARs) regulate normal and pathophysiological heart function through their impact on contractility. ßARs are also regulators of immune function where they play a unique role depending on the disease condition and immune cell type. Emerging evidence suggests an important role for the ß2AR subtype in regulating remodeling in the pathological heart; however, the importance of these responses has never been examined. In heart failure, catecholamines are elevated, leading to chronic ßAR activation and contributing to the detrimental effects in the heart. We hypothesized that immune cell ß2AR plays a critical role in the development of heart failure in response to chronic catecholamine elevations through their regulation of immune cell infiltration. To test this, chimeric mice were generated by performing bone marrow transplant (BMT) experiments using wild-type (WT) or ß2AR knockout (KO) donors. WT and ß2ARKO BMT mice were chronically administered the ßAR agonist isoproterenol. Immune cell recruitment to the heart was examined by histology and flow cytometry. Numerous changes in immune cell recruitment were observed with isoproterenol administration in WT BMT mice including proinflammatory myeloid populations and lymphocytes with macrophages made up the majority of immune cells in the heart and which were absent in ß2ARKO BMT animal. ß2ARKO BMT mice had decreased cardiomyocyte death, hypertrophy, and interstitial fibrosis following isoproterenol treatment, culminating in improved function. These findings demonstrate an important role for immune cell ß2AR expression in the heart's response to chronically elevated catecholamines.NEW & NOTEWORTHY Immune cell ß2-adrenergic receptors (ß2ARs) are important for proinflammatory macrophage infiltration to the heart in a chronic isoproterenol administration model of heart failure. Mice lacking immune cell ß2AR have decreased immune cell infiltration to their heart, primarily proinflammatory macrophage populations. This decrease culminated to decreased cardiac injury with lessened cardiomyocyte death, decreased interstitial fibrosis and hypertrophy, and improved function demonstrating that ß2AR regulation of immune responses plays an important role in the heart's response to persistent ßAR stimulation.


Assuntos
Insuficiência Cardíaca/metabolismo , Macrófagos/metabolismo , Miocárdio/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transferência Adotiva , Animais , Transplante de Medula Óssea , Morte Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Feminino , Fibrose , Insuficiência Cardíaca/imunologia , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Mediadores da Inflamação/metabolismo , Isoproterenol , Linfócitos/imunologia , Linfócitos/metabolismo , Ativação de Macrófagos , Macrófagos/imunologia , Macrófagos/transplante , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Miocárdica , Miocárdio/imunologia , Miocárdio/patologia , Fenótipo , Receptores Adrenérgicos beta 2/genética , Transdução de Sinais , Remodelação Ventricular
4.
Cell Signal ; 83: 109993, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33781845

RESUMO

The Reversion Inducing Cysteine Rich Protein With Kazal Motifs (RECK) is a glycosylphosphatidylinositol (GPI) anchored membrane-bound regulator of matrix metalloproteinases (MMPs). It is expressed throughout the body and plays a role in extracellular matrix (ECM) homeostasis and inflammation. In initial studies, RECK expression was found to be downregulated in various invasive cancers and associated with poor prognostic outcome. Restoring RECK, however, has been shown to reverse the metastatic phenotype. Downregulation of RECK expression is also reported in non-malignant diseases, such as periodontal disease, renal fibrosis, and myocardial fibrosis. As such, RECK induction has therapeutic potential in several chronic diseases. Mechanistically, RECK negatively regulates various matrixins involved in cell migration, proliferation, and adverse remodeling by targeting the expression and/or activation of multiple MMPs, A Disintegrin And Metalloproteinase Domain-Containing Proteins (ADAMs), and A Disintegrin And Metalloproteinase With Thrombospondin Motifs (ADAMTS). Outside of its role in remodeling, RECK has also been reported to exert anti-inflammatory effects. In cardiac diseases, for example, it has been shown to counteract several downstream effectors of Angiotensin II (Ang-II) that play a role in adverse cardiac and vascular remodeling, such as Interleukin-6 (IL-6)/IL-6 receptor (IL-6R)/glycoprotein 130 (IL-6 signal transducer) signaling and Epidermal Growth Factor Receptor (EGFR) transactivation. This review article focuses on the current understanding of the multifunctional effects of RECK and how its downregulation may contribute to adverse cardiovascular remodeling.


Assuntos
Movimento Celular , Regulação para Baixo , Proteínas Ligadas por GPI/biossíntese , Transdução de Sinais , Remodelação Vascular , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Proteínas Ligadas por GPI/genética , Humanos , Motivos Kazal
5.
Physiol Genomics ; 53(3): 99-115, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33491589

RESUMO

Heart failure (HF) patients with deteriorating right ventricular (RV) structure and function have a nearly twofold increased risk of death compared with those without. Despite the well-established clinical risk, few studies have examined the molecular signature associated with this HF condition. The purpose of this study was to integrate morphological, molecular, and functional data with the transcriptome data set in the RV of a preclinical model of cardiometabolic HF. Ossabaw swine were fed either normal diet without surgery (lean control, n = 5) or Western diet and aortic-banding (WD-AB; n = 4). Postmortem RV weight was increased and positively correlated with lung weight in the WD-AB group compared with CON. Total RNA-seq was performed and gene expression profiles were compared and analyzed using principal component analysis, weighted gene co-expression network analysis, module enrichment analysis, and ingenuity pathway analysis. Gene networks specifically associated with RV hypertrophic remodeling identified a hub gene in MAPK8 (or JNK1) that was associated with the selective induction of the extracellular matrix (ECM) component fibronectin. JNK1 and fibronectin protein were increased in the right coronary artery (RCA) of WD-AB animals and associated with a decrease in matrix metalloproteinase 14 protein, which specifically degrades fibronectin. RCA fibronectin content was correlated with increased vascular stiffness evident as a decreased elastin elastic modulus in WD-AB animals. In conclusion, this study establishes a molecular and transcriptome signature in the RV using Ossabaw swine with cardiometabolic HF. This signature was associated with altered ECM regulation and increased vascular stiffness in the RCA, with selective dysregulation of fibronectin.


Assuntos
Vasos Coronários/metabolismo , Perfilação da Expressão Gênica/métodos , Insuficiência Cardíaca/genética , Miocárdio/metabolismo , Transcriptoma , Remodelação Ventricular/genética , Animais , Dieta Ocidental , Feminino , Ontologia Genética , Redes Reguladoras de Genes , Insuficiência Cardíaca/metabolismo , Ventrículos do Coração/metabolismo , Humanos , RNA-Seq/métodos , Transdução de Sinais/genética , Suínos
6.
Int J Mol Sci ; 21(22)2020 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-33198112

RESUMO

Fibroblasts are an important resident cell population in the heart involved in maintaining homeostasis and structure during normal conditions. They are also crucial in disease states for sensing signals and initiating the appropriate repair responses to maintain the structural integrity of the heart. This sentinel role of cardiac fibroblasts occurs, in part, through their ability to secrete cytokines. ß-adrenergic receptors (ßAR) are also critical regulators of cardiac function in the normal and diseased state and a major therapeutic target clinically. ßAR are known to influence cytokine secretion in various cell types and they have been shown to be involved in cytokine production in the heart, but their role in regulating cytokine production in cardiac fibroblasts is not well understood. Thus, we hypothesized that ßAR activation on cardiac fibroblasts modulates cytokine production to influence fibroblast function. Using primary fibroblast cultures from neonatal rats and adult mice, increased interleukin (IL)-6 expression and secretion occurred following ß2AR activation. The use of pharmacological inhibitors and genetic manipulations showed that IL-6 elevations occurred through the Gαs-mediated activation of ERK1/2 and resulted in increased fibroblast proliferation. In vivo, a lack of ß2AR resulted in increased infarct size following myocardial infarction and impaired wound closure in a murine dermal wound healing assay. These findings identify an important role for ß2AR in regulating fibroblast proliferation through Gαs/ERK1/2-dependent alterations in IL-6 and may lead to the development of improved heart failure therapies through targeting fibrotic function of ß2AR.


Assuntos
Proliferação de Células/fisiologia , Fibroblastos/metabolismo , Coração/fisiologia , Interleucina-6/metabolismo , Miocárdio/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais/fisiologia , Animais , Secreções Corporais/metabolismo , Citocinas/metabolismo , Feminino , Fibroblastos/fisiologia , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Insuficiência Cardíaca/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/metabolismo , Ratos , Ratos Sprague-Dawley , Cicatrização/fisiologia
7.
Am J Physiol Heart Circ Physiol ; 318(5): H1162-H1175, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32216616

RESUMO

Nitric oxide (NO) and S-nitrosothiol (SNO) are considered cardio- and vasoprotective substances. We now understand that one mechanism in which NO/SNOs provide cardiovascular protection is through their direct inhibition of cardiac G protein-coupled receptor (GPCR) kinase 2 (GRK2) activity via S-nitrosylation of GRK2 at cysteine 340 (C340). This maintains GPCR homeostasis, including ß-adrenergic receptors, through curbing receptor GRK2-mediated desensitization. Previously, we have developed a knockin mouse (GRK2-C340S) where endogenous GRK2 is resistant to dynamic S-nitrosylation, which led to increased GRK2 desensitizing activity. This unchecked regulation of cardiac GRK2 activity resulted in significantly more myocardial damage after ischemic injury that was resistant to NO-mediated cardioprotection. Although young adult GRK2-C340S mice show no overt phenotype, we now report that as these mice age, they develop significant cardiovascular dysfunction due to the loss of SNO-mediated GRK2 regulation. This pathological phenotype is apparent as early as 12 mo of age and includes reduced cardiac function, increased cardiac perivascular fibrosis, and maladaptive cardiac hypertrophy, which are common maladies found in patients with cardiovascular disease (CVD). There are also vascular reactivity and aortic abnormalities present in these mice. Therefore, our data demonstrate that a chronic and global increase in GRK2 activity is sufficient to cause cardiovascular remodeling and dysfunction, likely due to GRK2's desensitizing effects in several tissues. Because GRK2 levels have been reported to be elevated in elderly CVD patients, GRK2-C340 mice can give insight into the aged-molecular landscape leading to CVD.NEW & NOTEWORTHY Research on G protein-coupled receptor kinase 2 (GRK2) in the setting of cardiovascular aging is largely unknown despite its strong established functions in cardiovascular physiology and pathophysiology. This study uses a mouse model of chronic GRK2 overactivity to further investigate the consequences of long-term GRK2 on cardiac function and structure. We report for the first time that chronic GRK2 overactivity was able to cause cardiac dysfunction and remodeling independent of surgical intervention, highlighting the importance of GRK activity in aged-related heart disease.


Assuntos
Envelhecimento/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Cardiopatias/etiologia , Coração/fisiologia , Miocárdio/metabolismo , Óxido Nítrico/metabolismo , Envelhecimento/metabolismo , Animais , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Coração/crescimento & desenvolvimento , Coração/fisiopatologia , Cardiopatias/metabolismo , Homeostase , Masculino , Camundongos , Mutação
8.
J Mol Cell Cardiol ; 136: 1-14, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31473246

RESUMO

Cardiomyocyte survival and death contributes to many cardiac diseases. A common mechanism of cardiomyocyte death is through apoptosis however, numerous death receptors (DR) have been virtually unstudied in the context of cardiovascular disease. Previous studies have identified TNF-related apoptosis inducing ligand (TRAIL) and its receptor, DR5, as being altered in a chronic catecholamine administration model of heart failure, and suggest a role of non-canonical signaling in cardiomyocytes. Furthermore, multiple clinical studies have identified TRAIL or DR5 as biomarkers in the prediction of severity and mortality following myocardial infarction and in heart failure development risk suggesting a role of DR5 signaling in the heart. While TRAIL/DR5 have been extensively studied as a potential cancer therapeutic due to their ability to selectively activate apoptosis in cancer cells, TRAIL and DR5 are highly expressed in the heart where their function is uncharacterized. However, many non-transformed cell types are resistant to TRAIL-induced apoptosis suggesting non-canonical functions in non-cancerous cell types. Our goal was to determine the role of DR5 in the heart with the hypothesis that DR5 does not induce cardiomyocyte apoptosis but initiates non-canonical signaling to promote cardiomyocyte growth and survival. Histological analysis of hearts from mice treated with a DR5 agonists showed increased hypertrophy with no differences in cardiomyocyte death, fibrosis or function. Mechanistic studies in the heart and isolated cardiomyocytes identified ERK1/2 activation with DR5 agonist treatment which contributed to hypertrophy. Furthermore, epidermal growth factor receptor (EGFR) was activated following DR5 agonist treatment through activation of MMP and HB-EGFR cleavage and specific inhibitors of MMP and EGFR prevented DR5-mediated ERK1/2 signaling and hypertrophy. Taken together, these studies identify a previously unidentified role for DR5 in the heart, which does not promote apoptosis but acts through non-canonical MMP-EGFR-ERK1/2 signaling mechanisms to contribute to cardiomyocyte hypertrophy.


Assuntos
Receptores ErbB/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Animais Recém-Nascidos , Cardiomegalia/metabolismo , Crescimento Celular , Sobrevivência Celular , Células Cultivadas , Receptores ErbB/genética , Fator de Transcrição GATA4/metabolismo , Regulação da Expressão Gênica , Hipertrofia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Metaloproteinases da Matriz/metabolismo , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Ftalimidas/farmacologia , Ratos Sprague-Dawley , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/agonistas , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Tiazolidinas/farmacologia
9.
JCI Insight ; 52019 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-30920389

RESUMO

Following injury, leukocytes are released from hematopoietic organs and migrate to the site of damage to regulate tissue inflammation and repair, however leukocytes lacking ß2-adrenergic receptor (ß2AR) expression have marked impairments in these processes. ß-blockade is a common strategy for the treatment of many cardiovascular etiologies, therefore the objective of our study was to assess the impact of prior ß-blocker treatment on baseline leukocyte parameters and their responsiveness to acute injury. In a temporal and ßAR isoform-dependent manner, chronic ß-blocker infusion increased splenic vascular cell adhesion molecule-1 (VCAM-1) expression and leukocyte accumulation (monocytes/macrophages, mast cells and neutrophils) and decreased chemokine receptor 2 (CCR2) expression, migration of bone marrow cells (BMC) and peripheral blood leukocytes (PBL), as well as infiltration into the heart following acute cardiac injury. Further, CCR2 expression and migratory responsiveness was significantly reduced in the PBL of patients receiving ß-blocker therapy compared to ß-blocker-naïve patients. These results highlight the ability of chronic ß-blocker treatment to alter baseline leukocyte characteristics that decrease their responsiveness to acute injury and suggest that prior ß-blockade may act to reduce the severity of innate immune responses.


Assuntos
Antagonistas Adrenérgicos beta/imunologia , Antagonistas Adrenérgicos beta/metabolismo , Leucócitos/imunologia , Leucócitos/fisiologia , Ferimentos e Lesões/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Medula Óssea , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Modelos Animais de Doenças , Feminino , Humanos , Imunidade Celular , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Isoformas de Proteínas , Receptores Adrenérgicos beta/metabolismo , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Receptores CCR2/metabolismo , Baço/metabolismo , Baço/patologia
10.
Arterioscler Thromb Vasc Biol ; 38(6): 1297-1308, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29674474

RESUMO

OBJECTIVE: To test the hypothesis that loss of IL-19 (interleukin-19) exacerbates atherosclerosis. APPROACH AND RESULTS: Il19-/- mice were crossed into Ldlr-/- (low-density lipoprotein receptor knock out) mice. Double knockout (dKO) mice had increased plaque burden in aortic arch and root compared with Ldlr-/- controls after 14 weeks of high-fat diet (HFD). dKO mice injected with 10 ng/g per day rmIL-19 had significantly less plaque compared with controls. qRT-PCR and Western blot analysis revealed dKO mice had increased systemic and intraplaque polarization of T cells and macrophages to proinflammatory Th1 and M1 phenotypes, and also significantly increased TNF (tumor necrosis factor)-α expression in spleen and aortic arch compared with Ldlr-/- controls. Bone marrow transplantation suggests that immune cells participate in IL-19 protection. Bone marrow-derived macrophages and vascular smooth muscle cells isolated from dKO mice had a significantly greater expression of inflammatory cytokine mRNA and protein compared with controls. Spleen and aortic arch from dKO mice had significantly increased expression of the mRNA stability protein HuR (human antigen R). Bone marrow-derived macrophage and vascular smooth muscle cell isolated from dKO mice also had greater HuR abundance. HuR stabilizes proinflammatory transcripts by binding AU-rich elements in the 3' untranslated region. Cytokine and HuR mRNA stability were increased in dKO bone marrow-derived macrophage and vascular smooth muscle cell, which was rescued by addition of IL-19 to these cells. IL-19-induced expression of miR133a, which targets and reduced HuR abundance; miR133a levels were lower in dKO mice compared with controls. CONCLUSIONS: These data indicate that IL-19 is an atheroprotective cytokine which decreases the abundance of HuR, leading to reduced inflammatory mRNA stability.


Assuntos
Aorta Torácica/metabolismo , Doenças da Aorta/metabolismo , Aterosclerose/metabolismo , Proteína Semelhante a ELAV 1/metabolismo , Deleção de Genes , Interleucina-10/deficiência , Estabilidade de RNA , RNA Mensageiro/metabolismo , Receptores de LDL/deficiência , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/patologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Doenças da Aorta/prevenção & controle , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/prevenção & controle , Células Cultivadas , Modelos Animais de Doenças , Progressão da Doença , Proteína Semelhante a ELAV 1/genética , Feminino , Predisposição Genética para Doença , Interleucina-10/administração & dosagem , Interleucina-10/genética , Interleucinas , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , MicroRNAs/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Fenótipo , Placa Aterosclerótica , Estabilidade de RNA/efeitos dos fármacos , RNA Mensageiro/genética , Receptores de LDL/genética , Fator de Necrose Tumoral alfa/metabolismo
11.
Circulation ; 136(10): 940-953, 2017 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-28667100

RESUMO

BACKGROUND: Activated fibroblasts (myofibroblasts) play a critical role in cardiac fibrosis; however, their origin in the diseased heart remains unclear, warranting further investigation. Recent studies suggest the contribution of bone marrow fibroblast progenitor cells (BM-FPCs) in pressure overload-induced cardiac fibrosis. We have previously shown that interleukin-10 (IL10) suppresses pressure overload-induced cardiac fibrosis; however, the role of IL10 in inhibition of BM-FPC-mediated cardiac fibrosis is not known. We hypothesized that IL10 inhibits pressure overload-induced homing of BM-FPCs to the heart and their transdifferentiation to myofibroblasts and thus attenuates cardiac fibrosis. METHODS: Pressure overload was induced in wild-type (WT) and IL10 knockout (IL10KO) mice by transverse aortic constriction. To determine the bone marrow origin, chimeric mice were created with enhanced green fluorescent protein WT mice marrow to the IL10KO mice. For mechanistic studies, FPCs were isolated from mouse bone marrow. RESULTS: Pressure overload enhanced BM-FPC mobilization and homing in IL10KO mice compared with WT mice. Furthermore, WT bone marrow (from enhanced green fluorescent protein mice) transplantation in bone marrow-depleted IL10KO mice (IL10KO chimeric mice) reduced transverse aortic constriction-induced BM-FPC mobilization compared with IL10KO mice. Green fluorescent protein costaining with α-smooth muscle actin or collagen 1α in left ventricular tissue sections of IL10KO chimeric mice suggests that myofibroblasts were derived from bone marrow after transverse aortic constriction. Finally, WT bone marrow transplantation in IL10KO mice inhibited transverse aortic constriction-induced cardiac fibrosis and improved heart function. At the molecular level, IL10 treatment significantly inhibited transforming growth factor-ß-induced transdifferentiation and fibrotic signaling in WT BM-FPCs in vitro. Furthermore, fibrosis-associated microRNA (miRNA) expression was highly upregulated in IL10KO-FPCs compared with WT-FPCs. Polymerase chain reaction-based selective miRNA analysis revealed that transforming growth factor-ß-induced enhanced expression of fibrosis-associated miRNAs (miRNA-21, -145, and -208) was significantly inhibited by IL10. Restoration of miRNA-21 levels suppressed the IL10 effects on transforming growth factor-ß-induced fibrotic signaling in BM-FPCs. CONCLUSIONS: Our findings suggest that IL10 inhibits BM-FPC homing and transdifferentiation to myofibroblasts in pressure-overloaded myocardium. Mechanistically, we show for the first time that IL10 suppresses Smad-miRNA-21-mediated activation of BM-FPCs and thus modulates cardiac fibrosis.


Assuntos
Ecocardiografia/métodos , Fibroblastos/metabolismo , Fibrose/metabolismo , Cardiopatias/complicações , Interleucina-10/genética , Interleucina-10/metabolismo , Miocárdio/metabolismo , Animais , Medula Óssea , Feminino , Fibroblastos/patologia , Humanos , Camundongos , Camundongos Transgênicos , Miocárdio/patologia , Transdução de Sinais
12.
J Cardiovasc Pharmacol ; 70(1): 3-9, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28059858

RESUMO

G protein-coupled receptors (GPCRs) remain primary therapeutic targets for numerous cardiovascular disorders, including heart failure (HF), because of their influence on cardiac remodeling in response to elevated neurohormone signaling. GPCR blockers have proven to be beneficial in the treatment of HF by reducing chronic G protein activation and cardiac remodeling, thereby extending the lifespan of patients with HF. Unfortunately, this effect does not persist indefinitely, thus next-generation therapeutics aim to selectively block harmful GPCR-mediated pathways while simultaneously promoting beneficial signaling. Transactivation of epidermal growth factor receptor (EGFR) has been shown to be mediated by an expanding repertoire of GPCRs in the heart, and promotes cardiomyocyte survival, thus may offer a new avenue of HF therapeutics. However, GPCR-dependent EGFR transactivation has also been shown to regulate cardiac hypertrophy and fibrosis by different GPCRs and through distinct molecular mechanisms. Here, we discuss the mechanisms and impact of GPCR-mediated EGFR transactivation in the heart, focusing on angiotensin II, urotensin II, and ß-adrenergic receptor systems, and highlight areas of research that will help us to determine whether this pathway can be engaged as future therapeutic strategy.


Assuntos
Receptores ErbB/genética , Receptores ErbB/metabolismo , Miócitos Cardíacos/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Ativação Transcricional/fisiologia , Animais , Humanos
13.
Proc Natl Acad Sci U S A ; 113(52): 15126-15131, 2016 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-27956622

RESUMO

Following cardiac injury, early immune cell responses are essential for initiating cardiac remodeling and tissue repair. We previously demonstrated the importance of ß2-adrenergic receptors (ß2ARs) in the regulation of immune cell localization following acute cardiac injury, with deficient leukocyte infiltration into the damaged heart. The purpose of this study was to investigate the mechanism by which immune cell-expressed ß2ARs regulate leukocyte recruitment to the heart following acute cardiac injury. Chemokine receptor 2 (CCR2) expression and responsiveness to C-C motif chemokine ligand 2 (CCL2)-mediated migration were abolished in ß2AR knockout (KO) bone marrow (BM), both of which were rescued by ß2AR reexpression. Chimeric mice lacking immune cell-specific CCR2 expression, as well as wild-type mice administered a CCR2 antagonist, recapitulated the loss of monocyte/macrophage and neutrophil recruitment to the heart following myocardial infarction (MI) observed in mice with immune cell-specific ß2AR deletion. Converse to ß2AR ablation, ß2AR stimulation increased CCR2 expression and migratory responsiveness to CCL2 in BM. Mechanistically, G protein-dependent ß2AR signaling was dispensable for these effects, whereas ß-arrestin2-biased ß2AR signaling was required for the regulation of CCR2 expression. Additionally, activator protein 1 (AP-1) was shown to be essential in mediating CCR2 expression in response to ß2AR stimulation in both murine BM and human monocytes. Finally, reconstitution of ß2ARKO BM with rescued expression of a ß-arrestin-biased ß2AR in vivo restored BM CCR2 expression as well as cardiac leukocyte infiltration following MI. These results demonstrate the critical role of ß-arrestin2/AP-1-dependent ß2AR signaling in the regulation of CCR2 expression and recruitment of leukocytes to the heart following injury.


Assuntos
Leucócitos/citologia , Miocárdio/patologia , Receptores Adrenérgicos beta/metabolismo , Receptores CCR2/metabolismo , Animais , Transplante de Medula Óssea , Movimento Celular , Quimiocinas/metabolismo , Vasos Coronários/patologia , Ecocardiografia , Insuficiência Cardíaca/patologia , Humanos , Antígenos Comuns de Leucócito/metabolismo , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/citologia , Infarto do Miocárdio/patologia , Isquemia Miocárdica/patologia , Miocárdio/metabolismo , Infiltração de Neutrófilos , Transdução de Sinais
14.
J Biol Chem ; 291(42): 21913-21924, 2016 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-27566547

RESUMO

GRK2, a G protein-coupled receptor kinase, plays a critical role in cardiac physiology. Adrenergic receptors are the primary target for GRK2 activity in the heart; phosphorylation by GRK2 leads to desensitization of these receptors. As such, levels of GRK2 activity in the heart directly correlate with cardiac contractile function. Furthermore, increased expression of GRK2 after cardiac insult exacerbates injury and speeds progression to heart failure. Despite the importance of this kinase in both the physiology and pathophysiology of the heart, relatively little is known about the role of GRK2 in skeletal muscle function and disease. In this study we generated a novel skeletal muscle-specific GRK2 knock-out (KO) mouse (MLC-Cre:GRK2fl/fl) to gain a better understanding of the role of GRK2 in skeletal muscle physiology. In isolated muscle mechanics testing, GRK2 ablation caused a significant decrease in the specific force of contraction of the fast-twitch extensor digitorum longus muscle yet had no effect on the slow-twitch soleus muscle. Despite these effects in isolated muscle, exercise capacity was not altered in MLC-Cre:GRK2fl/fl mice compared with wild-type controls. Skeletal muscle hypertrophy stimulated by clenbuterol, a ß2-adrenergic receptor (ß2AR) agonist, was significantly enhanced in MLC-Cre:GRK2fl/fl mice; mechanistically, this seems to be due to increased clenbuterol-stimulated pro-hypertrophic Akt signaling in the GRK2 KO skeletal muscle. In summary, our study provides the first insights into the role of GRK2 in skeletal muscle physiology and points to a role for GRK2 as a modulator of contractile properties in skeletal muscle as well as ß2AR-induced hypertrophy.


Assuntos
Clembuterol/efeitos adversos , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Contração Muscular/efeitos dos fármacos , Músculo Esquelético/enzimologia , Doenças Musculares/enzimologia , Transdução de Sinais/efeitos dos fármacos , Animais , Clembuterol/farmacocinética , Quinase 2 de Receptor Acoplado a Proteína G/genética , Hipertrofia/induzido quimicamente , Hipertrofia/enzimologia , Hipertrofia/genética , Hipertrofia/patologia , Camundongos , Camundongos Knockout , Contração Muscular/genética , Músculo Esquelético/patologia , Doenças Musculares/induzido quimicamente , Doenças Musculares/genética , Doenças Musculares/patologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais/genética
15.
Circulation ; 134(2): 153-67, 2016 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-27364164

RESUMO

BACKGROUND: Immune cell-mediated inflammation is an essential process for mounting a repair response after myocardial infarction (MI). The sympathetic nervous system is known to regulate immune system function through ß-adrenergic receptors (ßARs); however, their role in regulating immune cell responses to acute cardiac injury is unknown. METHODS: Wild-type (WT) mice were irradiated followed by isoform-specific ßAR knockout (ßARKO) or WT bone-marrow transplantation (BMT) and after full reconstitution underwent MI surgery. Survival was monitored over time, and alterations in immune cell infiltration after MI were examined through immunohistochemistry. Alterations in splenic function were identified through the investigation of altered adhesion receptor expression. RESULTS: ß2ARKO BMT mice displayed 100% mortality resulting from cardiac rupture within 12 days after MI compared with ≈20% mortality in WT BMT mice. ß2ARKO BMT mice displayed severely reduced post-MI cardiac infiltration of leukocytes with reciprocally enhanced splenic retention of the same immune cell populations. Splenic retention of the leukocytes was associated with an increase in vascular cell adhesion molecule-1 expression, which itself was regulated via ß-arrestin-dependent ß2AR signaling. Furthermore, vascular cell adhesion molecule-1 expression in both mouse and human macrophages was sensitive to ß2AR activity, and spleens from human tissue donors treated with ß-blocker showed enhanced vascular cell adhesion molecule-1 expression. The impairments in splenic retention and cardiac infiltration of leukocytes after MI were restored to WT levels via lentiviral-mediated re-expression of ß2AR in ß2ARKO bone marrow before transplantation, which also resulted in post-MI survival rates comparable to those in WT BMT mice. CONCLUSIONS: Immune cell-expressed ß2AR plays an essential role in regulating the early inflammatory repair response to acute myocardial injury by facilitating cardiac leukocyte infiltration.


Assuntos
Ruptura Cardíaca/etiologia , Leucócitos/metabolismo , Infarto do Miocárdio/complicações , Receptores Adrenérgicos beta 2/fisiologia , Idoso , Idoso de 80 Anos ou mais , Animais , Modelos Animais de Doenças , Feminino , Vetores Genéticos/uso terapêutico , Humanos , Macrófagos/metabolismo , Masculino , Metoprolol/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos , Quimera por Radiação , Receptores Adrenérgicos beta 2/deficiência , Receptores Adrenérgicos beta 2/genética , Proteínas Recombinantes de Fusão/metabolismo , Baço/metabolismo , Baço/patologia , Esplenectomia , Molécula 1 de Adesão de Célula Vascular/metabolismo
16.
Am J Physiol Heart Circ Physiol ; 308(4): H316-30, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25485901

RESUMO

Chronic stimulation of ß-adrenergic receptors (ßAR) can promote survival signaling via transactivation of epidermal growth factor receptor (EGFR) but ultimately alters cardiac structure and contractility over time, in part via enhanced cytokine signaling. We hypothesized that chronic catecholamine signaling will have a temporal impact on cardiac transcript expression in vivo, in particular cytokines, and that EGFR transactivation plays a role in this process. C57BL/6 mice underwent infusion with vehicle or isoproterenol (Iso)±gefitinib (Gef) for 1 or 2 wk. Cardiac contractility decreased following 2 wk of Iso treatment, while cardiac hypertrophy, fibrosis, and apoptosis were enhanced at both timepoints. Inclusion of Gef preserved contractility, blocked Iso-induced apoptosis, and prevented hypertrophy at the 2-wk timepoint, but caused fibrosis on its own. RNAseq analysis revealed hundreds of cardiac transcripts altered by Iso at each timepoint with subsequent RT-quantitative PCR validation confirming distinct temporal patterns of transcript regulation, including those involved in cardiac remodeling and survival signaling, as well as numerous cytokines. Although Gef infusion alone did not significantly alter cytokine expression, it abrogated the Iso-mediated changes in a majority of the ßAR-sensitive cytokines, including CCL2 and TNF-α. Additionally, the impact of ßAR-dependent EGFR transactivation on the acute regulation of cytokine transcript expression was assessed in isolated cardiomyocytes and in cardiac fibroblasts, where the majority of Iso-dependent, and EGFR-sensitive, changes in cytokines occurred. Overall, coincident with changes in cardiac structure and contractility, ßAR stimulation dynamically alters cardiac transcript expression over time, including numerous cytokines that are regulated via EGFR-dependent signaling.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Cardiomegalia/metabolismo , Quimiocina CCL2/metabolismo , Isoproterenol/farmacologia , Miócitos Cardíacos/metabolismo , Quinazolinas/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Apoptose , Cardiomegalia/fisiopatologia , Células Cultivadas , Quimiocina CCL2/genética , Receptores ErbB/antagonistas & inibidores , Fibrose/metabolismo , Fibrose/fisiopatologia , Gefitinibe , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Contração Miocárdica , Miócitos Cardíacos/efeitos dos fármacos , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Necrose Tumoral alfa/genética , Remodelação Ventricular
17.
Circulation ; 130(20): 1800-11, 2014 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-25205804

RESUMO

BACKGROUND: Enhanced arginine vasopressin levels are associated with increased mortality during end-stage human heart failure, and cardiac arginine vasopressin type 1A receptor (V1AR) expression becomes increased. Additionally, mice with cardiac-restricted V1AR overexpression develop cardiomyopathy and decreased ß-adrenergic receptor (ßAR) responsiveness. This led us to hypothesize that V1AR signaling regulates ßAR responsiveness and in doing so contributes to development of heart failure. METHODS AND RESULTS: Transaortic constriction resulted in decreased cardiac function and ßAR density and increased cardiac V1AR expression, effects reversed by a V1AR-selective antagonist. Molecularly, V1AR stimulation led to decreased ßAR ligand affinity, as well as ßAR-induced Ca(2+) mobilization and cAMP generation in isolated adult cardiomyocytes, effects recapitulated via ex vivo Langendorff analysis. V1AR-mediated regulation of ßAR responsiveness was demonstrated to occur in a previously unrecognized Gq protein-independent/G protein receptor kinase-dependent manner. CONCLUSIONS: This newly discovered relationship between cardiac V1AR and ßAR may be informative for the treatment of patients with acute decompensated heart failure and elevated arginine vasopressin.


Assuntos
Cardiomiopatia Hipertrófica/fisiopatologia , Contração Miocárdica/fisiologia , Receptores Adrenérgicos beta/fisiologia , Receptores de Vasopressinas/fisiologia , Sistemas do Segundo Mensageiro/fisiologia , Animais , Antagonistas dos Receptores de Hormônios Antidiuréticos/farmacologia , Arginina Vasopressina/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Cardiomiopatia Hipertrófica/complicações , Gatos , Linhagem Celular Tumoral , Colforsina/farmacologia , AMP Cíclico/biossíntese , Quinases de Receptores Acoplados a Proteína G/fisiologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Genes Reporter , Células HEK293 , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Indóis/farmacologia , Isoproterenol/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutagênese Sítio-Dirigida , Contração Miocárdica/efeitos dos fármacos , Pirrolidinas/farmacologia , Receptores de Vasopressinas/biossíntese , Receptores de Vasopressinas/genética , Proteínas Recombinantes de Fusão/metabolismo , Rolipram/farmacologia , Sistemas do Segundo Mensageiro/efeitos dos fármacos
18.
PLoS One ; 9(6): e99195, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24901703

RESUMO

ß-adrenergic receptor (ßAR)-mediated transactivation of epidermal growth factor receptor (EGFR) has been shown to promote cardioprotection in a mouse model of heart failure and we recently showed that this mechanism leads to enhanced cell survival in part via regulation of apoptotic transcript expression in isolated primary rat neonatal cardiomyocytes. Thus, we hypothesized that this process could regulate cardiac transcript expression in vivo. To comprehensively assess cardiac transcript alterations in response to acute ßAR-dependent EGFR transactivation, we performed whole transcriptome analysis of hearts from C57BL/6 mice given i.p. injections of the ßAR agonist isoproterenol in the presence or absence of the EGFR antagonist gefitinib for 1 hour. Total cardiac RNA from each treatment group underwent transcriptome analysis, revealing a substantial number of transcripts regulated by each treatment. Gefitinib alone significantly altered the expression of 405 transcripts, while isoproterenol either alone or in conjunction with gefitinib significantly altered 493 and 698 distinct transcripts, respectively. Further statistical analysis was performed, confirming 473 transcripts whose regulation by isoproterenol were significantly altered by gefitinib (isoproterenol-induced up/downregulation antagonized/promoted by gefinitib), including several known to be involved in the regulation of numerous processes including cell death and survival. Thus, ßAR-dependent regulation of cardiac transcript expression in vivo can be modulated by the EGFR antagonist gefitinib.


Assuntos
Regulação para Baixo/efeitos dos fármacos , Miocárdio/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Receptores Adrenérgicos beta/metabolismo , Regulação para Cima/efeitos dos fármacos , Agonistas Adrenérgicos beta/farmacologia , Animais , Feminino , Gefitinibe , Perfilação da Expressão Gênica , Redes Reguladoras de Genes/efeitos dos fármacos , Isoproterenol/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Adrenérgicos beta/química
19.
J Mol Cell Cardiol ; 72: 39-51, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24566221

RESUMO

ß-Adrenergic receptor (ßAR)-mediated transactivation of epidermal growth factor receptor (EGFR) has been shown to relay pro-survival effects via unknown mechanisms. We hypothesized that acute ßAR-mediated EGFR transactivation in the heart promotes differential subcellular activation of ERK1/2 and Akt, promoting cell survival through modulation of apoptosis. C57BL/6 mice underwent acute i.p. injection with isoproterenol (ISO)±AG 1478 (EGFR antagonist) to assess the impact of ßAR-mediated EGFR transactivation on the phosphorylation of ERK1/2 (P-ERK1/2) and Akt (P-Akt) in distinct cardiac subcellular fractions. Increased P-ERK1/2 and P-Akt were observed in cytosolic, plasma membrane and nuclear fractions following ISO stimulation. Whereas the P-ERK1/2 response was EGFR-sensitive in all fractions, the P-Akt response was EGFR-sensitive only in the plasma membrane and nucleus, results confirmed in primary rat neonatal cardiomyocytes (RNCM). ßAR-mediated EGFR-transactivation also decreased apoptosis in serum-depleted RNCM, as measured via TUNEL as well as caspase 3 activity/cleavage, which were sensitive to the inhibition of either ERK1/2 (PD184352) or Akt (LY-294002) signaling. Caspase 3 activity/cleavage was also sensitive to the inhibition of transcription, which, with an increase in nuclear P-ERK1/2 and P-Akt in response to ISO, suggested that ßAR-mediated EGFR transactivation may regulate apoptotic gene transcription. An Apoptosis PCR Array identified tnfsf10 (TRAIL) to be altered by ISO in an EGFR-sensitive manner, results confirmed via RT-PCR and ELISA measurement of both membrane-bound and soluble cardiomyocyte TRAIL levels. ßAR-mediated EGFR transactivation induces differential subcellular activation of ERK1/2 and Akt leading to increased cell survival through the modulation of caspase 3 activity and apoptotic gene expression in cardiomyocytes.


Assuntos
Receptores ErbB/genética , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Receptores Adrenérgicos beta/genética , Agonistas Adrenérgicos beta/farmacologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Apoptose/genética , Caspase 3/genética , Caspase 3/metabolismo , Gatos , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Regulação da Expressão Gênica , Isoproterenol/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Fosforilação , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinazolinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais , Tirfostinas/farmacologia
20.
J Pharmacol Exp Ther ; 338(2): 648-57, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21571945

RESUMO

Catecholamines released from the sympathetic nervous system in response to stress or injury affect expression of inflammatory cytokines generated by immune cells. α(1)-Adrenergic receptors (ARs) are expressed on innate immune cell populations, but their subtype expression patterns and signaling characteristics are not well characterized. Primary human monocytes, a human monocytic cell line, and monocyte-derived macrophage cells were used to measure expression of the proinflammatory mediator interleukin (IL)-1ß responding to lipopolysaccharide (LPS) in the presence or absence of α(1)-AR activation. Based on our previous findings, we hypothesized that α(1)-AR stimulation on innate immune cells positively regulates LPS-initiated IL-1ß production. IL-1ß production in response to LPS was synergistically higher for both monocytes and macrophages in the presence of the selective α(1)-AR agonist (R)-(-)-phenylephrine hydrochloride (PE). This synergistic IL-1ß response could be blocked with a selective α(1)-AR antagonist as well as inhibitors of protein kinase C (PKC). Radioligand binding studies characterized a homogenous α(1B)-AR subtype population on monocytes, which changed to a heterogeneous receptor subtype expression pattern when differentiated to macrophages. Furthermore, increased p38 mitogen-activated protein kinase (MAPK) activation was observed only with concurrent PE and LPS stimulation, peaking after 120 and 30 min in monocytes and macrophages, respectively. Blocking the PKC/p38 MAPK signaling pathway in both innate immune cell types inhibited the synergistic IL-1ß increase observed with concurrent PE and LPS treatments. This study characterizes α(1)-AR subtype expression on both human monocyte and macrophage cells and illustrates a mechanism by which increased IL-1ß production can be modulated by α(1)-AR input.


Assuntos
Macrófagos/metabolismo , Monócitos/metabolismo , Subunidades Proteicas/fisiologia , Receptores Adrenérgicos alfa 1/fisiologia , Receptor 4 Toll-Like/fisiologia , Regulação para Cima/imunologia , Adulto , Diferenciação Celular/imunologia , Linhagem Celular Transformada , Células Cultivadas , Humanos , Imunidade Inata , Mediadores da Inflamação/fisiologia , Interleucina-1beta/biossíntese , Interleucina-1beta/sangue , Lipopolissacarídeos/fisiologia , Macrófagos/imunologia , Macrófagos/patologia , Monócitos/imunologia , Monócitos/patologia , Subunidades Proteicas/biossíntese , Subunidades Proteicas/sangue , Receptores Adrenérgicos alfa 1/biossíntese , Receptores Adrenérgicos alfa 1/sangue , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/biossíntese , Receptor 4 Toll-Like/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA