Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Nat Metab ; 6(4): 708-723, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38499763

RESUMO

Cachexia affects 50-80% of patients with cancer and accounts for 20% of cancer-related death, but the underlying mechanism driving cachexia remains elusive. Here we show that circulating lactate levels positively correlate with the degree of body weight loss in male and female patients suffering from cancer cachexia, as well as in clinically relevant mouse models. Lactate infusion per se is sufficient to trigger a cachectic phenotype in tumour-free mice in a dose-dependent manner. Furthermore, we demonstrate that adipose-specific G-protein-coupled receptor (GPR)81 ablation, similarly to global GPR81 deficiency, ameliorates lactate-induced or tumour-induced adipose and muscle wasting in male mice, revealing adipose GPR81 as the major mediator of the catabolic effects of lactate. Mechanistically, lactate/GPR81-induced cachexia occurs independently of the well-established protein kinase A catabolic pathway, but it is mediated by a signalling cascade sequentially activating Gi-Gßγ-RhoA/ROCK1-p38. These findings highlight the therapeutic potential of targeting GPR81 for the treatment of this life-threatening complication of cancer.


Assuntos
Caquexia , Ácido Láctico , Neoplasias , Receptores Acoplados a Proteínas G , Caquexia/metabolismo , Caquexia/etiologia , Animais , Receptores Acoplados a Proteínas G/metabolismo , Camundongos , Humanos , Ácido Láctico/metabolismo , Masculino , Feminino , Neoplasias/metabolismo , Neoplasias/complicações , Transdução de Sinais
2.
Signal Transduct Target Ther ; 8(1): 263, 2023 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-37414783

RESUMO

Due to the essential role of cyclin D1 in regulating transition from G1 to S phase in cell cycle, aberrant cyclin D1 expression is a major oncogenic event in many types of cancers. In particular, the dysregulation of ubiquitination-dependent degradation of cyclin D1 contributes to not only the pathogenesis of malignancies but also the refractory to cancer treatment regiments with CDK4/6 inhibitors. Here we show that in colorectal and gastric cancer patients, MG53 is downregulated in more than 80% of tumors compared to the normal gastrointestinal tissues from the same patient, and the reduced MG53 expression is correlated with increased cyclin D1 abundance and inferior survival. Mechanistically, MG53 catalyzes the K48-linked ubiquitination and subsequent degradation of cyclin D1. Thus, increased expression of MG53 leads to cell cycle arrest at G1, and thereby markedly suppresses cancer cell proliferation in vitro as well as tumor growth in mice with xenograft tumors or AOM/DSS induced-colorectal cancer. Consistently, MG53 deficiency results in accumulation of cyclin D1 protein and accelerates cancer cell growth both in culture and in animal models. These findings define MG53 as a tumor suppressor via facilitating cyclin D1 degradation, highlighting the therapeutic potential of targeting MG53 in treating cancers with dysregulated cyclin D1 turnover.


Assuntos
Neoplasias Gástricas , Ubiquitina-Proteína Ligases , Humanos , Animais , Camundongos , Ubiquitina-Proteína Ligases/metabolismo , Ciclina D1/genética , Ciclina D1/metabolismo , Proliferação de Células , Pontos de Checagem do Ciclo Celular , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , Proteínas de Membrana
3.
Circ Res ; 131(12): 962-976, 2022 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-36337049

RESUMO

BACKGROUND: As an integral component of cell membrane repair machinery, MG53 (mitsugumin 53) is important for cardioprotection induced by ischemia preconditioning and postconditioning. However, it also impairs insulin signaling via its E3 ligase activity-mediated ubiquitination-dependent degradation of IR (insulin receptor) and IRS1 (insulin receptor substrate 1) and its myokine function-induced allosteric blockage of IR. Here, we sought to develop MG53 into a cardioprotection therapy by separating its detrimental metabolic effects from beneficial actions. METHODS: Using immunoprecipitation-mass spectrometry, site-specific mutation, in vitro kinase assay, and in vivo animal studies, we investigated the role of MG53 phosphorylation at serine 255 (S255). In particular, utilizing recombinant proteins and gene knock-in approaches, we evaluated the potential therapeutic effect of MG53-S255A mutant in treating cardiac ischemia/reperfusion injury in diabetic mice. RESULTS: We identified S255 phosphorylation as a prerequisite for MG53 E3 ligase activity. Furthermore, MG53S255 phosphorylation was mediated by GSK3ß (glycogen synthase kinase 3 beta) and markedly elevated in the animal models with metabolic disorders. Thus, IR-IRS1-GSK3ß-MG53 formed a vicious cycle in the pathogenesis of metabolic disorders where aberrant insulin signaling led to hyper-activation of GSK3ß, which in turn, phosphorylated MG53 and enhanced its E3 ligase activity, and further impaired insulin sensitivity. Importantly, S255A mutant eliminated the E3 ligase activity while retained cell protective function of MG53. Consequently, the S255A mutant, but not the wild type MG53, protected the heart against ischemia/reperfusion injury in db/db mice with advanced diabetes, although both elicited cardioprotection in normal mice. Moreover, in S255A knock-in mice, S255A mutant also mitigated ischemia/reperfusion-induced myocardial damage in the diabetic setting. CONCLUSIONS: S255 phosphorylation is a biased regulation of MG53 E3 ligase activity. The MG53-S255A mutant provides a promising approach for the treatment of acute myocardial injury, especially in patients with metabolic disorders.


Assuntos
Diabetes Mellitus Experimental , Traumatismo por Reperfusão , Camundongos , Animais , Fosforilação , Proteínas de Transporte/metabolismo , Serina/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Diabetes Mellitus Experimental/complicações , Proteínas de Membrana/metabolismo , Insulina/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Isquemia
4.
Biochem J ; 479(17): 1909-1916, 2022 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-36053137

RESUMO

MG53 is a tripartite motif (TRIM) family E3 ligase and plays important biological functions. Here we present the cryo-EM structure of human MG53, showing that MG53 is a homodimer consisting of a 'body' and two 'wings'. Intermolecular interactions are mainly distributed in the 'body' which is relatively stable, while two 'wings' are more dynamic. The overall architecture of MG53 is distinct from those of TRIM20 and TRIM25, illustrating the broad structural diversity of this protein family.


Assuntos
Proteínas de Transporte , Proteínas de Membrana , Proteínas com Motivo Tripartido/química , Proteínas de Transporte/metabolismo , Microscopia Crioeletrônica , Humanos , Proteínas de Membrana/metabolismo , Multimerização Proteica , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
5.
Circulation ; 145(15): 1154-1168, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35317609

RESUMO

BACKGROUND: Cardiac ischemia/reperfusion (I/R) injury has emerged as an important therapeutic target for ischemic heart disease, the leading cause of morbidity and mortality worldwide. At present, there is no effective therapy for reducing cardiac I/R injury. CaMKII (Ca2+/calmodulin-dependent kinase II) plays a pivotal role in the pathogenesis of severe heart conditions, including I/R injury. Pharmacological inhibition of CaMKII is an important strategy in the protection against myocardial damage and cardiac diseases. To date, there is no drug targeting CaMKII for the clinical therapy of heart disease. Furthermore, at present, there is no selective inhibitor of CaMKII-δ, the major CaMKII isoform in the heart. METHODS: A small-molecule kinase inhibitor library and a high-throughput screening system for the kinase activity assay of CaMKII-δ9 (the most abundant CaMKII-δ splice variant in human heart) were used to screen for CaMKII-δ inhibitors. Using cultured neonatal rat ventricular myocytes, human embryonic stem cell-derived cardiomyocytes, and in vivo mouse models, in conjunction with myocardial injury induced by I/R (or hypoxia/reoxygenation) and CaMKII-δ9 overexpression, we sought to investigate the protection of hesperadin against cardiomyocyte death and cardiac diseases. BALB/c nude mice with xenografted tumors of human cancer cells were used to evaluate the in vivo antitumor effect of hesperadin. RESULTS: Based on the small-molecule kinase inhibitor library and screening system, we found that hesperadin, an Aurora B kinase inhibitor with antitumor activity in vitro, directly bound to CaMKII-δ and specifically blocked its activation in an ATP-competitive manner. Hesperadin functionally ameliorated both I/R- and overexpressed CaMKII-δ9-induced cardiomyocyte death, myocardial damage, and heart failure in both rodents and human embryonic stem cell-derived cardiomyocytes. In addition, in an in vivo BALB/c nude mouse model with xenografted tumors of human cancer cells, hesperadin delayed tumor growth without inducing cardiomyocyte death or cardiac injury. CONCLUSIONS: Here, we identified hesperadin as a specific small-molecule inhibitor of CaMKII-δ with dual functions of cardioprotective and antitumor effects. These findings not only suggest that hesperadin is a promising leading compound for clinical therapy of cardiac I/R injury and heart failure, but also provide a strategy for the joint therapy of cancer and cardiovascular disease caused by anticancer treatment.


Assuntos
Insuficiência Cardíaca , Traumatismo por Reperfusão Miocárdica , Neoplasias , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Insuficiência Cardíaca/patologia , Humanos , Indóis , Isquemia/metabolismo , Camundongos , Camundongos Nus , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/metabolismo , Neoplasias/patologia , Ratos , Sulfonamidas
6.
Circ Res ; 130(6): 887-903, 2022 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-35152717

RESUMO

BACKGROUND: CaMKII (Ca2+/calmodulin-dependent kinase II) plays a central role in cardiac ischemia/reperfusion (I/R) injury-an important therapeutic target for ischemic heart disease. In the heart, CaMKII-δ is the predominant isoform and further alternatively spliced into 11 variants. In humans, CaMKII-δ9 and CaMKII-δ3, the major cardiac splice variants, inversely regulate cardiomyocyte viability with the former pro-death and the latter pro-survival. However, it is unknown whether specific inhibition of the detrimental CaMKII-δ9 prevents cardiac I/R injury and, if so, what is the underlying mechanism. Here, we aim to investigate the cardioprotective effect of specific CaMKII-δ9 inhibition against myocardial I/R damage and determine the underlying mechanisms. METHODS: The role and mechanism of CaMKII-δ9 in cardiac I/R injury were investigated in mice in vivo, neonatal rat ventricular myocytes, and human embryonic stem cell-derived cardiomyocytes. RESULTS: We demonstrate that CaMKII-δ9 inhibition with knockdown or knockout of its feature exon, exon 16, protects the heart against I/R-elicited injury and subsequent heart failure. I/R-induced cardiac inflammation was also ameliorated by CaMKII-δ9 inhibition, and compared with the previously well-studied CaMKII-δ2, CaMKII-δ9 overexpression caused more profound cardiac inflammation. Mechanistically, in addition to IKKß (inhibitor of NF-κB [nuclear factor-κB] kinase subunit ß), CaMKII-δ9, but not δ2, directly interacted with IκBα (NF-κB inhibitor α) with its feature exon 13-16-17 combination and increased IκBα phosphorylation and consequently elicited more pronounced activation of NF-κB signaling and inflammatory response. Furthermore, the essential role of CaMKII-δ9 in myocardial inflammation and damage was confirmed in human cardiomyocytes. CONCLUSIONS: We not only identified CaMKII-δ9-IKK/IκB-NF-κB signaling as a new regulator of human cardiomyocyte inflammation but also demonstrated that specifically targeting CaMKII-δ9, the most abundant CaMKII-δ splice variant in human heart, markedly suppresses I/R-induced cardiac NF-κB activation, inflammation, and injury and subsequently ameliorates myocardial remodeling and heart failure, providing a novel therapeutic strategy for various ischemic heart diseases.


Assuntos
Insuficiência Cardíaca , Traumatismo por Reperfusão Miocárdica , Miocardite , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Inflamação/genética , Inflamação/prevenção & controle , Isquemia , Camundongos , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miócitos Cardíacos , Inibidor de NF-kappaB alfa , NF-kappa B , Ratos
7.
Diabetes ; 71(2): 298-314, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34844991

RESUMO

Cardiometabolic diseases, including diabetes and its cardiovascular complications, are the global leading causes of death, highlighting a major unmet medical need. Over the past decade, mitsugumin 53 (MG53), also called TRIM72, has emerged as a powerful agent for myocardial membrane repair and cardioprotection, but its therapeutic value is complicated by its E3 ligase activity, which mediates metabolic disorders. Here, we show that an E3 ligase-dead mutant, MG53-C14A, retains its cardioprotective function without causing metabolic adverse effects. When administered in normal animals, both the recombinant human wild-type MG53 protein (rhMG53-WT) and its E3 ligase-dead mutant (rhMG53-C14A) protected the heart equally from myocardial infarction and ischemia/reperfusion (I/R) injury. However, in diabetic db/db mice, rhMG53-WT treatment markedly aggravated hyperglycemia, cardiac I/R injury, and mortality, whereas acute and chronic treatment with rhMG53-C14A still effectively ameliorated I/R-induced myocardial injury and mortality or diabetic cardiomyopathy, respectively, without metabolic adverse effects. Furthermore, knock-in of MG53-C14A protected the mice from high-fat diet-induced metabolic disorders and cardiac damage. Thus, the E3 ligase-dead mutant MG53-C14A not only protects the heart from acute myocardial injury but also counteracts metabolic stress, providing a potentially important therapy for the treatment of acute myocardial injury in metabolic disorders, including diabetes and obesity.


Assuntos
Proteínas de Membrana/genética , Síndrome Metabólica/genética , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Animais , Células Cultivadas , Citoproteção/genética , Cardiomiopatias Diabéticas/complicações , Cardiomiopatias Diabéticas/genética , Cardiomiopatias Diabéticas/metabolismo , Cardiomiopatias Diabéticas/fisiopatologia , Dieta Hiperlipídica , Feminino , Coração/fisiopatologia , Humanos , Masculino , Síndrome Metabólica/metabolismo , Síndrome Metabólica/patologia , Síndrome Metabólica/fisiopatologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Traumatismo por Reperfusão Miocárdica/etiologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Transdução de Sinais/genética
8.
Mol Biol Evol ; 38(7): 2930-2945, 2021 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-33744959

RESUMO

Cis-regulatory elements play important roles in tissue-specific gene expression and in the evolution of various phenotypes, and mutations in promoters and enhancers may be responsible for adaptations of species to environments. TRIM72 is a highly conserved protein that is involved in energy metabolism. Its expression in the heart varies considerably in primates, with high levels of expression in Old World monkeys and near absence in hominids. Here, we combine phylogenetic hypothesis testing and experimentation to demonstrate that mutations in promoter are responsible for the differences among primate species in the heart-specific expression of TRIM72. Maximum likelihood estimates of lineage-specific substitution rates under local-clock models show that relative to the evolutionary rate of introns, the rate of promoter was accelerated by 78% in the common ancestor of Old World monkeys, suggesting a role for positive selection in the evolution of the TRIM72 promoter, possibly driven by selective pressure due to changes in cardiac physiology after species divergence. We demonstrate that mutations in the TRIM72 promoter account for the differential myocardial TRIM72 expression of the human and the rhesus macaque. Furthermore, changes in TRIM72 expression alter the expression of genes involved in oxidative phosphorylation, which in turn affects mitochondrial respiration and cardiac energy capacity. On a broader timescale, phylogenetic regression analyses of data from 29 mammalian species show that mammals with high cardiac expression of TRIM72 have high heart rate, suggesting that the expression changes of TRIM72 may be related to differences in the heart physiology of those species.


Assuntos
Evolução Biológica , Miocárdio/metabolismo , Primatas/genética , Regiões Promotoras Genéticas/genética , Proteínas com Motivo Tripartido/genética , Animais , Metabolismo Basal , Regulação da Expressão Gênica/genética , Frequência Cardíaca , Humanos , Mutação , Fosforilação Oxidativa , Primatas/metabolismo , Proteínas com Motivo Tripartido/metabolismo
9.
Mol Cell ; 81(3): 629-637.e5, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33400924

RESUMO

As a master regulator of metabolism, AMP-activated protein kinase (AMPK) is activated upon energy and glucose shortage but suppressed upon overnutrition. Exaggerated negative regulation of AMPK signaling by nutrient overload plays a crucial role in metabolic diseases. However, the mechanism underlying the negative regulation is poorly understood. Here, we demonstrate that high glucose represses AMPK signaling via MG53 (also called TRIM72) E3-ubiquitin-ligase-mediated AMPKα degradation and deactivation. Specifically, high-glucose-stimulated reactive oxygen species (ROS) signals AKT to phosphorylate AMPKα at S485/491, which facilitates the recruitment of MG53 and the subsequent ubiquitination and degradation of AMPKα. In addition, high glucose deactivates AMPK by ROS-dependent suppression of phosphorylation of AMPKα at T172. These findings not only delineate the mechanism underlying the impairment of AMPK signaling in overnutrition-related diseases but also highlight the significance of keeping the yin-yang balance of AMPK signaling in the maintenance of metabolic homeostasis.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Diabetes Mellitus/enzimologia , Glucose/farmacologia , Proteínas de Membrana/metabolismo , Músculo Esquelético/efeitos dos fármacos , Obesidade/enzimologia , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP/genética , Animais , Glicemia/metabolismo , Diabetes Mellitus/sangue , Diabetes Mellitus/genética , Modelos Animais de Doenças , Células HEK293 , Humanos , Macaca mulatta , Masculino , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Músculo Esquelético/enzimologia , Obesidade/sangue , Obesidade/genética , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteólise , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Ubiquitinação
10.
Circulation ; 142(11): 1077-1091, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32677469

RESUMO

BACKGROUND: Ischemic heart disease is the leading cause of morbidity and mortality worldwide. Ischemic preconditioning (IPC) is the most powerful intrinsic protection against cardiac ischemia/reperfusion injury. Previous studies have shown that a multifunctional TRIM family protein, MG53 (mitsugumin 53; also called TRIM72), not only plays an essential role in IPC-mediated cardioprotection against ischemia/reperfusion injury but also ameliorates mechanical damage. In addition to its intracellular actions, as a myokine/cardiokine, MG53 can be secreted from the heart and skeletal muscle in response to metabolic stress. However, it is unknown whether IPC-mediated cardioprotection is causally related to MG53 secretion and, if so, what the underlying mechanism is. METHODS: Using proteomic analysis in conjunction with genetic and pharmacological approaches, we examined MG53 secretion in response to IPC and explored the underlying mechanism using rodents in in vivo, isolated perfused hearts, and cultured neonatal rat ventricular cardiomyocytes. Moreover, using recombinant MG53 proteins, we investigated the potential biological function of secreted MG53 in the context of IPC and ischemia/reperfusion injury. RESULTS: We found that IPC triggered robust MG53 secretion in rodents in vivo, perfused hearts, and cultured cardiac myocytes without causing cell membrane leakage. Mechanistically, IPC promoted MG53 secretion through H2O2-evoked activation of protein kinase-C-δ. Specifically, IPC-induced myocardial MG53 secretion was mediated by H2O2-triggered phosphorylation of protein kinase-C-δ at Y311, which is necessary and sufficient to facilitate MG53 secretion. Functionally, systemic delivery of recombinant MG53 proteins to mimic elevated circulating MG53 not only restored IPC function in MG53-deficient mice but also protected rodent hearts from ischemia/reperfusion injury even in the absence of IPC. Moreover, oxidative stress by H2O2 augmented MG53 secretion, and MG53 knockdown exacerbated H2O2-induced cell injury in human embryonic stem cell-derived cardiomyocytes, despite relatively low basal expression of MG53 in human heart. CONCLUSIONS: We conclude that IPC and oxidative stress can trigger MG53 secretion from the heart via an H2O2-protein kinase-C-δ-dependent mechanism and that extracellular MG53 can participate in IPC protection against cardiac ischemia/reperfusion injury.


Assuntos
Peróxido de Hidrogênio/farmacologia , Precondicionamento Isquêmico , Proteínas de Membrana/metabolismo , Traumatismo por Reperfusão Miocárdica , Proteína Quinase C-delta/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Proteína Quinase C-delta/genética
12.
Circulation ; 139(7): 901-914, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30586741

RESUMO

BACKGROUND: Mitsugumin 53 (MG53 or TRIM72), a striated muscle-specific E3 ligase, promotes ubiquitin-dependent degradation of the insulin receptor and insulin receptor substrate-1 and subsequently induces insulin resistance, resulting in metabolic syndrome and type 2 diabetes mellitus (T2DM). However, it is unknown how MG53 from muscle regulates systemic insulin response and energy metabolism. Increasing evidence demonstrates that muscle secretes proteins as myokines or cardiokines that regulate systemic metabolic processes. We hypothesize that MG53 may act as a myokine/cardiokine, contributing to interorgan regulation of insulin sensitivity and metabolic homeostasis. METHODS: Using perfused rodent hearts or skeletal muscle, we investigated whether high glucose, high insulin, or their combination (conditions mimicking metabolic syndrome or T2DM) alters MG53 protein concentration in the perfusate. We also measured serum MG53 levels in rodents and humans in the presence or absence of metabolic diseases, particularly T2DM. The effects of circulating MG53 on multiorgan insulin response were evaluated by systemic delivery of recombinant MG53 protein to mice. Furthermore, the potential involvement of circulating MG53 in the pathogenesis of T2DM was assessed by neutralizing blood MG53 with monoclonal antibodies in diabetic db/db mice. Finally, to delineate the mechanism underlying the action of extracellular MG53 on insulin signaling, we analyzed the potential interaction of MG53 with extracellular domain of insulin receptor using coimmunoprecipitation and surface plasmon resonance assays. RESULTS: Here, we demonstrate that MG53 is a glucose-sensitive myokine/cardiokine that governs the interorgan regulation of insulin sensitivity. First, high glucose or high insulin induces MG53 secretion from isolated rodent hearts and skeletal muscle. Second, hyperglycemia is accompanied by increased circulating MG53 in humans and rodents with diabetes mellitus. Third, systemic delivery of recombinant MG53 or cardiac-specific overexpression of MG53 causes systemic insulin resistance and metabolic syndrome in mice, whereas neutralizing circulating MG53 with monoclonal antibodies has therapeutic effects in T2DM db/db mice. Mechanistically, MG53 binds to the extracellular domain of the insulin receptor and acts as an allosteric blocker. CONCLUSIONS: Thus, MG53 has dual actions as a myokine/cardiokine and an E3 ligase, synergistically inhibiting the insulin signaling pathway. Targeting circulating MG53 opens a new therapeutic avenue for T2DM and its complications.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus/sangue , Metabolismo Energético , Resistência à Insulina , Proteínas de Membrana/metabolismo , Adulto , Animais , Anticorpos Monoclonais/farmacologia , Antígenos CD/metabolismo , Biomarcadores/sangue , Glicemia/efeitos dos fármacos , Estudos de Casos e Controles , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/enzimologia , Diabetes Mellitus/imunologia , Modelos Animais de Doenças , Metabolismo Energético/efeitos dos fármacos , Feminino , Células HEK293 , Homeostase , Humanos , Hipoglicemiantes/farmacologia , Masculino , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Musculares/metabolismo , Músculo Esquelético/enzimologia , Miocárdio/enzimologia , Ratos Sprague-Dawley , Ratos Zucker , Receptor de Insulina/metabolismo , Transdução de Sinais , Proteínas com Motivo Tripartido/metabolismo , Proteínas de Transporte Vesicular/metabolismo
13.
Cardiovasc Res ; 113(13): 1615-1626, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-29016703

RESUMO

AIMS: Ischemic heart disease is a leading cause of morbidity and mortality worldwide. Although timely restoration of coronary blood flow (reperfusion) is the most effective therapeutics of myocardial infarction, reperfusion causes further cardiac damage, i.e. ischemia-reperfusion (I/R) injury. ß-arrestins (Arrbs) have been traditionally defined as negative regulators of G protein-coupled receptor (GPCR) signalling, but recent studies have shown that they are essential for G protein-independent, GPCR-mediated biased signalling. Several ligands have been reported to be cardioprotective via Arrbs dependent pathway. However, it is unclear whether Arrbs exert receptor-independent physiological or pathological functions in the heart. Here, we sought to determine whether and how Arrbs play a role in regulating cardiomyocyte viability and myocardial remodelling following I/R injury. METHODS AND RESULTS: The expression of ß-arrestin 2 (Arrb2), but not ß-arrestin 1 (Arrb1), is upregulated in rat hearts subjected to I/R injury, or in cultured neonatal rat cardiomyocytes treated with hypoxia-reoxygenation (H/R) injury. Deficiency of Arrb2 in cultured neonatal rat cardiomyocytes alleviates H/R-induced cardiomyocyte death and Arrb2-/- mice are resistant to myocardial damage caused by I/R injury. In contrast, upregulation of Arrb2 triggers cardiomyocyte death and exaggerates I/R (or H/R)-induced detrimental effects. Mechanically, Arrb2 induces cardiomyocyte death by interacting with the p85 subunit of PI3K, and negatively regulating the formation of p85-PI3K/CaV3 survival complex, thus blocking activation of PI3K-Akt-GSK3ß cell survival signalling pathway. CONCLUSION: We define an upregulation of Arrb2 as a pathogenic factor in cardiac I/R injury, and also reveal a novel GPCR-independent mechanism of Arrb2-mediated cell death signalling in the heart.


Assuntos
Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestina 2/metabolismo , Animais , Caveolina 3/metabolismo , Morte Celular , Sobrevivência Celular , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Modelos Animais de Doenças , Predisposição Genética para Doença , Glicogênio Sintase Quinase 3 beta/metabolismo , Preparação de Coração Isolado , Masculino , Camundongos Knockout , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miócitos Cardíacos/patologia , Fenótipo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Transdução de Sinais , beta-Arrestina 1/genética , beta-Arrestina 1/metabolismo , beta-Arrestina 2/deficiência , beta-Arrestina 2/genética
14.
Appl Opt ; 56(28): 7915-7920, 2017 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-29047778

RESUMO

Foveated imaging systems have the ability to capture local high-resolution or high-magnification images with wide field of view (FOV); thus, they have great potential for applications in the field of monitoring and remote sensing of unmanned aerial vehicles. Hence, foveated optical systems are in strong demand. However, the existing foveated imaging systems either are equipped with expensive modulators or require fixing the local high resolution imaging field, which is not suitable for mass production or object tracking in industrial applications. We propose a low-cost dynamic real-time foveated imaging system for extensive use in the listed applications. Specifically, we place a microlens behind the first intermediary image plane to modulate the local focal length, constructing a local high magnification imaging channel. One two-axis translation stage drives the microlens to scan in the plane perpendicular to the optical axis, resulting in dynamic local high magnifying imaging. Furthermore, the peripheral imaging channel and the foveated imaging channel focus on the same detector, and the post image fusion is unnecessary; the system consists of only a common aspherical lens and thus is very inexpensive. The experimental system has a focal length of 25 mm, a full FOV of 30°, and an entrance pupil diameter of 5 mm, while the local high magnifying imaging channel has a focal length of 35 mm and FOV of 15°. Experiment results show that the low-cost dynamic real-time foveated imaging system performs very well.

15.
Mol Pharmacol ; 92(3): 211-218, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28432201

RESUMO

MG53 (also known as tripartite motif, TRIM72) is a cardiac and skeletal muscle-specific TRIM-family protein that exhibits multiple biologic functions. First, MG53 participates in plasma membrane repair of the heart, skeletal muscle, and, other tissues. Second, MG53 is essentially involved in the cardioprotection of cardiac ischemic, preconditioning, and postconditioning by activating the PI3K-Akt-GSK3ß and ERK1/2 survival signaling pathways. Moreover, systemic delivery of recombinant MG53 protein ameliorates the impact of a range of injury insults on the heart, skeletal muscle, lung, kidney, skin, and brain. It is noteworthy that chronic upregulation of MG53 induces insulin resistance and metabolic diseases, such as type 2 diabetes and its cardiovascular complications, by acting as an E3 ligase to mediate the degradation of insulin receptor and insulin receptor substrate-1. In addition, MG53 negatively regulates myogenesis. In summary, MG53 is a multifunctional protein involved in the vital physiologic and pathologic processes of multiple organs and is a promising therapeutic target for various human diseases. In this review, we comprehensively summarize current research progress on the biologic functions and therapeutic potential of MG53.


Assuntos
Proteínas de Transporte/fisiologia , Animais , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/química , Proteínas de Transporte/genética , Cardiomiopatias Diabéticas/etiologia , Humanos , Resistência à Insulina , Desenvolvimento Muscular , Músculo Esquelético/fisiologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Transcrição Gênica , Proteínas com Motivo Tripartido
16.
Sheng Li Xue Bao ; 68(4): 505-16, 2016 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-27546510

RESUMO

Mitsugumin 53 (MG53), also named Trim72, is a multi-functional TRIM-family protein, which is abundantly expressed in cardiac and skeletal muscle. It has been shown that MG53 not only plays important physiological roles but also acts as a crucial pathogenic factor of various diseases. First, MG53 preserves cardiac and skeletal muscle integrity via facilitating plasma membrane repair. Second, MG53 is essentially involved in cardiac ischemic preconditioning and postconditioning by activating PI3K-Akt-GSK3ß and ERK1/2 cell survival signaling pathways. Moreover, systemic delivery of recombinant MG53 is able to abolish mechanic or ischemia-reperfusion (I/R)-induced injury of multiple organs, including heart, skeletal muscle, lung, kidney and skin. Importantly, MG53 acts as an E3 ligase to mediate the degradation of insulin receptor and insulin receptor substrate-1, and subsequently induces insulin resistance and metabolic diseases such as type-2 diabetes and its cardiovascular complications. In addition, MG53 negatively regulates myogenesis. As a potential therapeutic target of human diseases, multiple facets of MG53 biological function and mechanisms of action should be taken into the consideration to maximize its beneficial effects and minimize potential side-effects. Here in this review, we intend to comprehensively summarize the current progresses on the biological functions of MG53, focusing on its clinical value as a therapeutic target.


Assuntos
Doenças Cardiovasculares , Diabetes Mellitus Tipo 2 , Resistência à Insulina , Proteínas de Transporte , Humanos , Transdução de Sinais , Proteínas com Motivo Tripartido
17.
JCI Insight ; 1(1): e84969, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26966719

RESUMO

Stimulation of ß1-adrenergic receptor (ß1AR), a GPCR, and the receptor for advanced glycation end-products (RAGE), a pattern recognition receptor (PRR), have been independently implicated in the pathogenesis of cardiomyopathy caused by various etiologies, including myocardial infarction, ischemia/reperfusion injury, and metabolic stress. Here, we show that the two distinctly different receptors, ß1AR and RAGE, are mutually dependent in mediating myocardial injury and the sequelae of cardiomyopathy. Deficiency or inhibition of RAGE blocks ß1AR- and RAGE-mediated myocardial cell death and maladaptive remodeling. Ablation or blockade of ß1AR fully abolishes RAGE-induced detrimental effects. Mechanistically, RAGE and ß1AR form a complex, which in turn activates Ca2+/calmodulin-dependent kinase II (CaMKII), resulting in loss of cardiomyocytes and myocardial remodeling. These results indicate that RAGE and ß1AR not only physically crosstalk at the receptor level, but also functionally converge at the common mediator, CaMKII, highlighting a combined inhibition of RAGE and ß1AR as a more effective therapy to treat diverse cardiovascular diseases, such as myocardial infarction, ischemia/reperfusion injury, and diabetic cardiovascular complications.

18.
Nat Med ; 22(2): 175-82, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26726877

RESUMO

Regulated necrosis (necroptosis) and apoptosis are crucially involved in severe cardiac pathological conditions, including myocardial infarction, ischemia-reperfusion injury and heart failure. Whereas apoptotic signaling is well defined, the mechanisms that underlie cardiomyocyte necroptosis remain elusive. Here we show that receptor-interacting protein 3 (RIP3) triggers myocardial necroptosis, in addition to apoptosis and inflammation, through activation of Ca(2+)-calmodulin-dependent protein kinase (CaMKII) rather than through the well-established RIP3 partners RIP1 and MLKL. In mice, RIP3 deficiency or CaMKII inhibition ameliorates myocardial necroptosis and heart failure induced by ischemia-reperfusion or by doxorubicin treatment. RIP3-induced activation of CaMKII, via phosphorylation or oxidation or both, triggers opening of the mitochondrial permeability transition pore and myocardial necroptosis. These findings identify CaMKII as a new RIP3 substrate and delineate a RIP3-CaMKII-mPTP myocardial necroptosis pathway, a promising target for the treatment of ischemia- and oxidative stress-induced myocardial damage and heart failure.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Insuficiência Cardíaca/genética , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Isquemia Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/genética , Miócitos Cardíacos/metabolismo , Necrose/genética , Estresse Oxidativo , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Animais , Antibióticos Antineoplásicos/toxicidade , Apoptose/genética , Western Blotting , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Sobrevivência Celular , Doxorrubicina/toxicidade , Insuficiência Cardíaca/induzido quimicamente , Insuficiência Cardíaca/patologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Poro de Transição de Permeabilidade Mitocondrial , Isquemia Miocárdica/complicações , Isquemia Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/patologia , Necrose/etiologia , Necrose/patologia , Fosforilação , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio , Reação em Cadeia da Polimerase em Tempo Real , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo
19.
J Mol Med (Berl) ; 93(10): 1107-18, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25894383

RESUMO

UNLABELLED: Proliferation and migration disorders of vascular smooth muscle cells (VSMCs) contribute to the pathogenesis of proliferative cardiovascular diseases. Although, over the past two decades, a large panel of drugs has been developed for targeting VSMC proliferation, cardiovascular disease remains the leading cause of death worldwide. Thus, there is a compelling need to identify novel signaling pathways and molecules controlling VSMC proliferation and migration, to provide not only mechanistic insights but also safe and effective therapies for the treatment of cardiovascular diseases. Our recent studies have demonstrated that p55γ, a regulatory subunit of phosphoinositide 3-kinase, functions as an endogenous brake on VSMC proliferation. Here, we demonstrate that the small peptide N24, the first 24 amino acids of the NH2 terminus of p55γ, is a functional mimetic which negatively regulates VSMC proliferation and migration. Specifically, luminal delivery of adenovirus expressing N24 or local administration of Tat transactivator protein (TAT)-tagged N24 by pluronic gel alleviates neointimal formation following balloon injury in rat carotid arteries. Enforced expression of N24 suppresses the proliferation and migration of VSMCs induced by serum- or platelet-derived growth factor-BB. Mechanistically, N24 induces cell cycle arrest via activating the p53-p21 signal pathway, without triggering cell death. N24 interacts with and stabilizes p53 by blocking its ubiquitin-dependent degradation, subsequently promotes p21 transcription, and arrests cell cycle progression. Indeed, knockdown of p21 or p53 abrogates the N24-mediated cell growth arrest. Thus, N24 is a p55γ mimetic inhibiting VSMC proliferation as well as migration, thereby conferring important therapeutic implications for anti-proliferative treatment. KEY MESSAGE: • N24 attenuates balloon injury-induced neointimal formation. • Overexpression of N24 inhibits cultured VSMC proliferation and migration. • Overexpression of N24 arrests the cell cycle at S phase. • N24 interacts with and stabilizes p53 resulting in growth suppression.


Assuntos
Miócitos de Músculo Liso/efeitos dos fármacos , Neointima/tratamento farmacológico , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/uso terapêutico , Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/farmacologia , Fosfatidilinositol 3-Quinases/uso terapêutico , Angioplastia com Balão/efeitos adversos , Animais , Aorta Torácica/citologia , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/patologia , Contagem de Células , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Produtos do Gene tat/farmacologia , Masculino , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/fisiologia , Neointima/patologia , Ratos Sprague-Dawley , Proteína Supressora de Tumor p53/metabolismo , Cicatrização/efeitos dos fármacos
20.
Cardiovasc Res ; 105(1): 75-85, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25388664

RESUMO

AIMS: Phosphatidylinositol 3 kinases (PI3Ks) play a pivotal role in vascular physiology and pathophysiology. We aimed to investigate the role of p55γ, a regulatory subunit of PI3Ks, in vascular smooth muscle cell (VSMC) proliferation and neointimal formation. METHODS AND RESULTS: We identified p55γ as an important factor that suppresses VSMC proliferation and injury-evoked neointimal formation. Western blot and mRNA analyses showed that p55γ expression declined in balloon-injured rat carotid arteries and in response to PDGF-BB and serum treatment in cultured VSMCs. Overexpression of p55γ inhibited, whereas short hairpin RNA knockdown of p55γ promoted PDGF-BB- and serum-induced VSMC proliferation. Importantly, in vivo adenoviral gene transfer of p55γ into carotid arteries attenuated, while knockdown of p55γ enhanced balloon injury-induced neointimal formation. Furthermore, p55γ sequentially up-regulated p53 and p21, resulting in cell-cycle arrest in S phase; small-interfering RNA knockdown of either p53 or p21 blocked p55γ-induced VSMC growth arrest. Mechanistically, p55γ interacted with and stabilized p53 protein by blocking mouse double minute 2 homologue-mediated p53 ubiquitination and degradation, subsequently activating its target gene p21. Concurrently, p55γ up-regulated Bcl-xl expression, resulting in non-apoptotic growth arrest effect. CONCLUSION: These findings mark p55γ as a novel upstream regulator of the p53-p21 signalling pathway that negatively regulates VSMC proliferation, suggesting that malfunction of p55γ may trigger vascular proliferative disorders.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/patologia , Neointima/enzimologia , Neointima/prevenção & controle , Animais , Lesões das Artérias Carótidas/enzimologia , Lesões das Artérias Carótidas/patologia , Pontos de Checagem do Ciclo Celular , Proliferação de Células , Células Cultivadas , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Masculino , Neointima/patologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima , Proteína bcl-X/antagonistas & inibidores , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...