Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 166
Filtrar
1.
J Cell Mol Med ; 28(7): e18140, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38494851

RESUMO

Growth differentiation factor 11 (GDF11), also known as bone morphogenetic protein 11 (BMP11), has been identified as a key player in various biological processes, including embryonic development, aging, metabolic disorders and cancers. GDF11 has also emerged as a critical component in liver development, injury and fibrosis. However, the effects of GDF11 on liver physiology and pathology have been a subject of debate among researchers due to conflicting reported outcomes. While some studies suggest that GDF11 has anti-aging properties, others have documented its senescence-inducing effects. Similarly, while GDF11 has been implicated in exacerbating liver injury, it has also been shown to have the potential to reduce liver fibrosis. In this narrative review, we present a comprehensive report of recent evidence elucidating the diverse roles of GDF11 in liver development, hepatic injury, regeneration and associated diseases such as non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), liver fibrosis and hepatocellular carcinoma. We also explore the therapeutic potential of GDF11 in managing various liver pathologies.


Assuntos
Neoplasias Hepáticas , Hepatopatia Gordurosa não Alcoólica , Humanos , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Fibrose , Cirrose Hepática/patologia , Fatores de Diferenciação de Crescimento/genética , Fatores de Diferenciação de Crescimento/metabolismo , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Neoplasias Hepáticas/patologia
2.
Life Sci ; 341: 122507, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38378101

RESUMO

Annually, the frequency of morbidity in depression has increased progressively in response to life stressors, and there is an increasing trend toward younger morbidity. The pathogenesis of depression is complicated and includes factors such as genetic inheritance and variations in physiological functions induced by various environmental factors. Currently, drug therapy has wide adaptability in clinical practice and plays an important role in the treatment of patients with mild depression. However, the therapeutic effects of most antidepressants are typically not significant and are associated with considerable adverse effects and addiction. Therefore, it is imperative to identify the deeper mechanisms of depression and search for alternative drug targets. Growth differentiation factor 11 (GDF11) is described as an anti-ageing molecule that belongs to a member of the transforming growth factor ß family. Additionally, the latest research findings suggested that GDF11 positively regulates neurogenesis and enhances neuronal activity, thereby attenuating depression-like behaviours. Although an increasing number of studies have focused on the multiple functions of GDF11 in skeletal dysplasia and carcinogenesis, its precise mechanism of action in depression remains unknown. Thus, in this review, we discuss the role of GDF11 and its mechanistic pathways in the pathogenesis of depression to develop novel therapies for depression.


Assuntos
Depressão , Fatores de Diferenciação de Crescimento , Humanos , Depressão/tratamento farmacológico , Fatores de Diferenciação de Crescimento/genética , Fatores de Diferenciação de Crescimento/metabolismo , Fatores de Diferenciação de Crescimento/farmacologia , Fator de Crescimento Transformador beta , Envelhecimento , Proteínas Morfogenéticas Ósseas
3.
Biochim Biophys Acta Mol Cell Res ; 1871(3): 119656, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38182060

RESUMO

Diabetic cardiomyopathy remains a formidable health challenge with a high mortality rate and no targeted treatments. Growth differentiation factor 11 (GDF11) has shown promising effects on cardiovascular diseases; however, its role and the underlying mechanism in regulating diabetic cardiomyopathy remain unclear. In this study, we developed mouse models of diabetic cardiomyopathy using leptin receptor-deficient (db/db) mice and streptozocin-induced C57BL/6 mice. The diabetic cardiomyopathy model mice exhibited apparent structural damage in cardiac tissues and a significant increase in the expression of apoptosis-related proteins. Notably, we observed a significant decreased expression of GDF11 in the myocardium of mice with diabetic cardiomyopathy. Moreover, GDF11 cardiac-specific knock-in mice (transgenic mice) exhibited improved cardiac function and reduced apoptosis. Moreover, exogenous administration of GDF11 mitigated high glucose-induced cardiomyocyte apoptosis. Mechanistically, we demonstrated that GDF11 alleviated high glucose-induced cardiomyocytes apoptosis by inhibiting the activation of the alkylation repair homolog 5 (ALKBH5)-forkhead box group O3a (FOXO3)-cerebellar degeneration-related protein 1 transcript (CDR1as)/Hippo signaling pathway. Consequently, this novel mechanism effectively counteracted myocardial cell apoptosis, providing valuable insights into potential therapeutic strategies for clinical diabetic cardiomyopathy.


Assuntos
Cardiomiopatias Diabéticas , Miócitos Cardíacos , Camundongos , Animais , Miócitos Cardíacos/metabolismo , Cardiomiopatias Diabéticas/induzido quimicamente , Cardiomiopatias Diabéticas/metabolismo , Via de Sinalização Hippo , Camundongos Endogâmicos C57BL , Fatores de Diferenciação de Crescimento/genética , Fatores de Diferenciação de Crescimento/metabolismo , Fatores de Diferenciação de Crescimento/farmacologia , Glucose/farmacologia , Glucose/metabolismo , Apoptose/genética
4.
Cell Mol Biol (Noisy-le-grand) ; 69(13): 250-255, 2023 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-38158658

RESUMO

Growth differentiation factor 11 (GDF11) is reported as a member of TGF-ß superfamily, which plays a key negative role in various tissue inflammation. However, the specific effect of GDF11 on infectious acute liver injury remains unknown. The current study is designed to certify the role of GDF11 both in LPS-induced RAW 264.7 cell line and rodent model of acute liver injury (ALI) and further investigate its molecular mechanism of inflammatory regulation. In vitro, LPS was used to stimulate the inflammatory activation of RAW 264.7 cells and then recombinant GDF11 (rGDF11) was used to treat the cells. In vivo, we injected LPS and rGDF11 in abdomen of mouse. The inflammatory indexes, GDF11 level, NLRP3 level, liver tissue injury, and liver function were examined using qRT-PCR, western blot, ELISA, IHC, IF and HE staining, respectively. Supplement of GDF11 protected the histology and function of liver tissue in LPS-induced ALI mice, in which the level of AST, ALT and TBiL associated with tissue damage were reduced after ALI. Moreover, increased GDF11 in RAW 264.7 cells and ALI mice reduced the expressions of COX-2, TNF-α, IL-1ß, and IL-6 via inhibiting NLRP3 inflammasome activation, suggesting the anti-inflammatory role of GDF11 in ALI. Besides, owing to the protective role of GDF11, the apoptotic degree in liver after LPS insult was attenuated, such as the reduced c-caspase-3 and annexin-V expressions. The results indicate that overexpression of GDF11 plays an antagonistic role in LPS-induced inflammatory response after ALI. Therefore, GDF11 may become a promising target for preventing infectious acute liver injury.


Assuntos
Doença Hepática Crônica Induzida por Substâncias e Drogas , Fatores de Diferenciação de Crescimento , Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Animais , Camundongos , Fatores de Diferenciação de Crescimento/genética , Inflamassomos/metabolismo , Inflamação , Lipopolissacarídeos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo
5.
Nat Commun ; 14(1): 7476, 2023 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-37978295

RESUMO

As a major neuron type in the brain, the excitatory neuron (EN) regulates the lifespan in C. elegans. How the EN acquires senescence, however, is unknown. Here, we show that growth differentiation factor 11 (GDF11) is predominantly expressed in the EN in the adult mouse, marmoset and human brain. In mice, selective knock-out of GDF11 in the post-mitotic EN shapes the brain ageing-related transcriptional profile, induces EN senescence and hyperexcitability, prunes their dendrites, impedes their synaptic input, impairs object recognition memory and shortens the lifespan, establishing a functional link between GDF11, brain ageing and cognition. In vitro GDF11 deletion causes cellular senescence in Neuro-2a cells. Mechanistically, GDF11 deletion induces neuronal senescence via Smad2-induced transcription of the pro-senescence factor p21. This work indicates that endogenous GDF11 acts as a brake on EN senescence and brain ageing.


Assuntos
Caenorhabditis elegans , Fatores de Diferenciação de Crescimento , Adulto , Camundongos , Humanos , Animais , Caenorhabditis elegans/metabolismo , Fatores de Diferenciação de Crescimento/genética , Fatores de Diferenciação de Crescimento/metabolismo , Envelhecimento/genética , Encéfalo/metabolismo , Neurônios/metabolismo , Proteínas Morfogenéticas Ósseas
6.
Peptides ; 170: 171112, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37918484

RESUMO

Growth differentiation factor-15 (GDF15) is a stress-activated cytokine that regulates cell growth and inflammatory and stress responses. We previously reported the role and regulation of GDF15 in pituitary corticotrophs. Dexamethasone increases Gdf15 gene expression levels and production. GDF15 suppresses adrenocorticotropic hormone synthesis in pituitary corticotrophs and subsequently mediates the negative feedback effect of glucocorticoids. Here, we analyzed corticotropin-releasing factor (Crf) promoter activity in hypothalamic 4B cells transfected with promoter-driven luciferase reporter constructs. The effects of time and GDF15 concentration on Crf mRNA levels were analyzed using quantitative real-time polymerase chain reaction. Glial cell-derived neurotrophic factor family receptor α-like (GFRAL) protein is expressed in 4B cells. GDF15 increased Crf promoter activity and Crf mRNA levels in 4B cells. The protein kinase A and C pathways also contributed to the GDF15-induced increase in Crf gene expression. GDF15 stimulates GFRAL, subsequently increasing the phosphorylation of AKT, an extracellular signal-related kinase, and the cAMP response element-binding protein. Therefore, GDF15-dependent pathways may be involved in regulating Crf expression under stressful conditions in hypothalamic cells.


Assuntos
Hormônio Liberador da Corticotropina , Fator 15 de Diferenciação de Crescimento , Hipotálamo , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , Fatores de Diferenciação de Crescimento/genética , Fatores de Diferenciação de Crescimento/metabolismo , Fatores de Diferenciação de Crescimento/farmacologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Regiões Promotoras Genéticas , Receptores de Hormônio Liberador da Corticotropina/genética , Receptores de Hormônio Liberador da Corticotropina/metabolismo , RNA Mensageiro/metabolismo , Animais , Ratos , Fator 15 de Diferenciação de Crescimento/metabolismo , Fator 15 de Diferenciação de Crescimento/farmacologia , Humanos
7.
J Cell Physiol ; 238(12): 2827-2840, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37801347

RESUMO

Growth differentiation factor 11 (GDF11) is a putative systemic rejuvenation factor. In this study, we characterized the mechanism by which GDF11 reversed aging of mesenchymal stem cells (MSCs). In culture, aged MSCs proliferate slower and are positive for senescence markers senescence-associated ß-galactosidase and P16ink4a . They have shortened telomeres, decreased GDF11 expression, and reduced osteogenic potential. GDF11 can block MSC aging in vitro and reverse age-dependent bone loss in vivo. The antiaging effect of GDF11 is via activation of the Smad2/3-PI3K-AKT-mTOR pathway. Unexpectedly, GDF11 also upregulated a DNA demethylase Tet2, which served as a key mediator for GDF11 to autoregulate itself via demethylation of the GDF11 promoter. Mutation of Tet2 facilitates MSC aging by blocking GDF11 expression. Mutagenesis of Tet2-regulated CpG sites also blocks GDF11 expression, leading to MSC aging. Together, a novel mutual regulatory relationship between GDF11 and an epigenetic factor Tet2 unveiled their antiaging roles.


Assuntos
Senescência Celular , Células-Tronco Mesenquimais , Senescência Celular/genética , Fatores de Diferenciação de Crescimento/genética , Fatores de Diferenciação de Crescimento/metabolismo , Células-Tronco Mesenquimais/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Humanos
8.
Cardiovasc Res ; 119(17): 2729-2742, 2023 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-37742057

RESUMO

AIMS: The heart rejuvenating effects of circulating growth differentiation factor 11 (GDF11), a transforming growth factor-ß superfamily member that shares 90% homology with myostatin (MSTN), remains controversial. Here, we aimed to probe the role of GDF11 in acute myocardial infarction (MI), a frequent cause of heart failure and premature death during ageing. METHODS AND RESULTS: In contrast to endogenous Mstn, myocardial Gdf11 declined during the course of ageing and was particularly reduced following ischaemia/reperfusion (I/R) injury, suggesting a therapeutic potential of GDF11 signalling in MI. Unexpectedly, boosting systemic Gdf11 by recombinant GDF11 delivery (0.1 mg/kg body weight over 30 days) prior to myocardial I/R augmented myocardial infarct size in C57BL/6 mice irrespective of their age, predominantly by accelerating pro-apoptotic signalling. While intrinsic cardioprotective signalling pathways remained unaffected by high circulating GDF11, targeted transcriptomics and immunomapping studies focusing on GDF11-associated downstream targets revealed attenuated Nkx2-5 expression confined to CD105-expressing cells, with pro-apoptotic activity, as assessed by caspase-3 levels, being particularly pronounced in adjacent cells, suggesting an indirect effect. By harnessing a highly specific and validated liquid chromatography-tandem mass spectrometry-based assay, we show that in prospectively recruited patients with MI circulating GDF11 but not MSTN levels incline with age. Moreover, GDF11 levels were particularly elevated in those at high risk for adverse outcomes following the acute event, with circulating GDF11 emerging as an independent predictor of myocardial infarct size, as estimated by standardized peak creatine kinase-MB levels. CONCLUSION: Our data challenge the initially reported heart rejuvenating effects of circulating GDF11 and suggest that high levels of systemic GDF11 exacerbate myocardial injury in mice and humans alike. Persistently high GDF11 levels during ageing may contribute to the age-dependent loss of cardioprotective mechanisms and thus poor outcomes of elderly patients following acute MI.


Assuntos
Fatores de Diferenciação de Crescimento , Traumatismos Cardíacos , Infarto do Miocárdio , Idoso , Animais , Humanos , Camundongos , Envelhecimento/metabolismo , Proteínas Morfogenéticas Ósseas , Fatores de Diferenciação de Crescimento/genética , Fatores de Diferenciação de Crescimento/metabolismo , Coração , Traumatismos Cardíacos/complicações , Traumatismos Cardíacos/metabolismo , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/complicações , Infarto do Miocárdio/metabolismo
9.
Nat Aging ; 3(2): 213-228, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-37118117

RESUMO

Cognitive decline and mood disorders increase in frequency with age. Many efforts are focused on the identification of molecules and pathways to treat these conditions. Here, we demonstrate that systemic administration of growth differentiation factor 11 (GDF11) in aged mice improves memory and alleviates senescence and depression-like symptoms in a neurogenesis-independent manner. Mechanistically, GDF11 acts directly on hippocampal neurons to enhance neuronal activity via stimulation of autophagy. Transcriptomic and biochemical analyses of these neurons reveal that GDF11 reduces the activity of mammalian target of rapamycin (mTOR), a master regulator of autophagy. Using a murine model of corticosterone-induced depression-like phenotype, we also show that GDF11 attenuates the depressive-like behavior of young mice. Analysis of sera from young adults with major depressive disorder (MDD) reveals reduced GDF11 levels. These findings identify mechanistic pathways related to GDF11 action in the brain and uncover an unknown role for GDF11 as an antidepressant candidate and biomarker.


Assuntos
Depressão , Transtorno Depressivo Maior , Camundongos , Animais , Depressão/tratamento farmacológico , Transtorno Depressivo Maior/tratamento farmacológico , Fatores de Diferenciação de Crescimento/genética , Fenótipo , Autofagia/genética , Mamíferos/metabolismo , Proteínas Morfogenéticas Ósseas/genética
10.
Kidney Blood Press Res ; 48(1): 209-219, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36780878

RESUMO

INTRODUCTION: Acute kidney injury (AKI) is a clinical emergency caused by the rapid decline of renal function caused by various etiologies. Growth differentiation factor 11 (GDF11) can promote renal tubular regeneration and improve kidney function in AKI, but the specific mechanism remains unclear. Herein, we investigated the effect and mechanisms of GDF11 in ameliorating AKI induced by ischemia-reperfusion (I/R). METHODS: An animal model of AKI was established by I/R method, and the changes of serum urea nitrogen and creatinine were measured to evaluate the AKI. Enzyme-linked immunosorbent assay (ELISA) was used to measure cytokines, malondialdehyde, superoxide dismutase, nitric oxide synthase, and arginase 1 levels. Flow cytometry was used to count the M1/M2 macrophages. IHC, WB, and q-PCR experiments were used to evaluate the expression of GDF11. RESULTS: The changes in serum levels of urea nitrogen and creatinine after I/R suggest that an animal model of AKI induced by I/R was successfully established. AKI caused by I/R significantly changed the M1/M2 macrophage polarization balance, with an increase in M2 being significantly higher than M1 as well as increased oxidative stress. Treatment with GDF11 after I/R significantly increased the differentiation of M2 cells and inhibited the differentiation of M1 macrophages, as well as decreased oxidative stress. CONCLUSION: GDF11 can promote the repair of AKI caused by I/R by regulating the balance of M1/M2 polarization in macrophages and oxidative stress.


Assuntos
Injúria Renal Aguda , Traumatismo por Reperfusão , Animais , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/metabolismo , Creatinina/metabolismo , Fatores de Diferenciação de Crescimento/genética , Fatores de Diferenciação de Crescimento/metabolismo , Isquemia/complicações , Rim/metabolismo , Macrófagos/metabolismo , Nitrogênio/metabolismo , Reperfusão/efeitos adversos , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/metabolismo , Ureia/metabolismo
11.
Elife ; 122023 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-36848184

RESUMO

Loss- and gain-of-function of MeCP2 causes Rett syndrome (RTT) and MECP2 duplication syndrome (MDS), respectively. MeCP2 binds methyl-cytosines to finely tune gene expression in the brain, but identifying genes robustly regulated by MeCP2 has been difficult. By integrating multiple transcriptomics datasets, we revealed that MeCP2 finely regulates growth differentiation factor 11 (Gdf11). Gdf11 is down-regulated in RTT mouse models and, conversely, up-regulated in MDS mouse models. Strikingly, genetically normalizing Gdf11 dosage levels improved several behavioral deficits in a mouse model of MDS. Next, we discovered that losing one copy of Gdf11 alone was sufficient to cause multiple neurobehavioral deficits in mice, most notably hyperactivity and decreased learning and memory. This decrease in learning and memory was not due to changes in proliferation or numbers of progenitor cells in the hippocampus. Lastly, loss of one copy of Gdf11 decreased survival in mice, corroborating its putative role in aging. Our data demonstrate that Gdf11 dosage is important for brain function.


Assuntos
Fenômenos Fisiológicos do Sistema Nervoso , Síndrome de Rett , Animais , Camundongos , Envelhecimento , Modelos Animais de Doenças , Fatores de Diferenciação de Crescimento/genética , Proteínas Morfogenéticas Ósseas/genética , Proteína 2 de Ligação a Metil-CpG/genética
12.
Life Sci Alliance ; 6(3)2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36631218

RESUMO

Growth differentiation factor 11 (GDF11) and GDF8 (MSTN) are closely related TGF-ß family proteins that interact with nearly identical signaling receptors and antagonists. However, GDF11 appears to activate SMAD2/3 more potently than GDF8 in vitro and in vivo. The ligands possess divergent structural properties, whereby substituting unique GDF11 amino acids into GDF8 enhanced the activity of the resulting chimeric GDF8. We investigated potentially distinct endogenous activities of GDF11 and GDF8 in vivo by genetically modifying their mature signaling domains. Full recoding of GDF8 to that of GDF11 yielded mice lacking GDF8, with GDF11 levels ∼50-fold higher than normal, and exhibiting modestly decreased muscle mass, with no apparent negative impacts on health or survival. Substitution of two specific amino acids in the fingertip region of GDF11 with the corresponding GDF8 residues resulted in prenatal axial skeletal transformations, consistent with Gdf11-deficient mice, without apparent perturbation of skeletal or cardiac muscle development or homeostasis. These experiments uncover distinctive features between the GDF11 and GDF8 mature domains in vivo and identify a specific requirement for GDF11 in early-stage skeletal development.


Assuntos
Desenvolvimento Ósseo , Fatores de Diferenciação de Crescimento , Músculo Esquelético , Miostatina , Animais , Feminino , Camundongos , Gravidez , Aminoácidos/química , Aminoácidos/genética , Desenvolvimento Ósseo/genética , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Fatores de Diferenciação de Crescimento/genética , Fatores de Diferenciação de Crescimento/química , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/metabolismo , Miostatina/genética , Miostatina/química , Fator de Crescimento Transformador beta/metabolismo
13.
Neurol Res ; 45(1): 70-80, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36328251

RESUMO

Background Previous data have indicated the regulation of circular RNA (circRNA) toward cerebral ischemia. This study aims to reveal the effects of circNUFIP2 on cerebral ischemia and the underlying mechanism. Methods Oxygen-glucose deprivation (OGD) hippocampal neuron (HT22) cell model and middle cerebral artery occlusion (MCAO) mouse model were used for this study. The expression of circRNA nuclear FMR1 interacting protein 2 (circNUFIP2), microRNA-1224-5p (miR-1224-5p) and growth differentiation factor 11 (GDF11) was detected by quantitative real-time polymerase-chain reaction. Protein expression was checked by Western blotting. The binding relationships among circNUFIP2, miR-1224-5p and GDF11 were identified by dual-luciferase reporter assay, RNA pull-down assay, and RNA immunoprecipitation assay. Cell proliferation and apoptosis were investigated by 5-Ethynyl-29-deoxyuridine and flow cytometry analysis, respectively. Results CircNUFIP2 and GDF11 expression were decreased, but miR-1224-5p was increased in OGD-treated HT22 cells when compared with their expression in control groups. OGD treatment inhibited HT22 cell proliferation but induced cell apoptosis and oxidative stress; however, these effects were attenuated after circNUFIP2 overexpression. Also, circNUFIP2 upregulation assuaged the cerebral infarction of MCAO mice. Besides, circNUFIP2 bound to miR-1224-5p and mediated OGD-induced HT22 cell damage through miR-1224-5p. Meanwhile, knockdown of GDF11, a target gene of miR-1224-5p, relieved miR-1224-5p depletion-caused effects in OGD-treated HT22 cells. Furthermore, circNUFIP2 regulated GDF11 expression by interacting with miR-1224-5p. Conclusion CircNUFIP2 overexpression protected neuron cells against cerebral ischemia-induced damage, at least in part, by the miR-1224-5p/GDF11 pathway, providing a possible target for the therapy of cerebral ischemic stroke.


Assuntos
Isquemia Encefálica , MicroRNAs , Traumatismo por Reperfusão , Acidente Vascular Cerebral , Camundongos , Animais , Oxigênio , Glucose/metabolismo , RNA Circular/metabolismo , RNA Circular/farmacologia , MicroRNAs/genética , MicroRNAs/metabolismo , Isquemia Encefálica/metabolismo , Acidente Vascular Cerebral/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Estresse Oxidativo , Apoptose , Hipocampo/metabolismo , Neurônios/metabolismo , Traumatismo por Reperfusão/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Morfogenéticas Ósseas/farmacologia , Fatores de Diferenciação de Crescimento/genética , Fatores de Diferenciação de Crescimento/metabolismo , Fatores de Diferenciação de Crescimento/farmacologia
14.
Am J Physiol Cell Physiol ; 323(5): C1402-C1409, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36094432

RESUMO

Skeletal muscle mass is negatively regulated by several TGF-ß superfamily members. Myostatin (MSTN) is the most prominent negative regulator of muscle mass. Recent studies show that in addition to MSTN, GDF11, which shares a high sequence identity with MSTN, induces muscle atrophy in vitro and in vivo at supraphysiological levels, whereas controversy regarding its roles exists. Furthermore, higher circulating GDF11 levels associate with frailty in humans. On the other hand, little is known about the effect of pathophysiological levels of GDF11 on muscle atrophy. Here we seek to determine whether pathophysiological levels of GDF11 are sufficient to activate Smad2/Smad3 signaling and induce muscle atrophy using human iPSC-derived myocytes (hiPSC myocytes). We first show that incubating hiPSC myocytes with pathophysiological concentrations of GDF11 significantly reduces myocyte diameters. We next demonstrate that pathophysiological levels of GDF11 are sufficient to activate Smad2/3 signaling. Finally, we show that pathophysiological levels of GDF11 are capable of inducing the expression of Atrogin-1, an atrophy-promoting E3 ubiquitin ligase and that FOXO1 blockage reverses the GDF11-induced Atrogin-1 expression and atrophic phenotype. Collectively, our results suggest that GDF11 induces skeletal muscle atrophy at the pathophysiological levels through the GDF11-FOXO1 axis.


Assuntos
Células-Tronco Pluripotentes Induzidas , Miostatina , Humanos , Miostatina/genética , Miostatina/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Fatores de Diferenciação de Crescimento/genética , Fatores de Diferenciação de Crescimento/metabolismo , Fatores de Diferenciação de Crescimento/farmacologia , Atrofia Muscular/patologia , Músculo Esquelético/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Células Musculares/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteína Smad3/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Proteína Smad2/genética
15.
Biochem Biophys Res Commun ; 623: 104-110, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-35921703

RESUMO

Protecting dopaminergic neurons is a key approach in the prevention of Parkinson's disease (PD). Transient receptor potential vanilloid 1 (TRPV1) is a nonselective cation channel that is widely distributed in the mammalian nervous system. In this study, we designed experiments to investigate the effect and mechanisms of TRPV1 against DA neurons damage of PD. Our results showed that trpv1-deficient mice showed a significant loss of TH + neurons than PD mice after MPTP intraperitoneal injection, in addition, a significant decline in motor function was observed in trpv1-deficient mice versus the MPTP model. In addition, our study indicated that GDF11 overexpression inhibited MPP + - induced oxidative stress, cell senescence, and apoptosis in neurons. Results also showed that TRPV1 prevented the down-regulation of GDF11 expression in PD model, gdf11 knockdown blocks the effects of TRPV1 on the antioxidant, antiaging, and antiapoptotic activities of dopaminergic neurons. Consequently, our findings indicate that TRPV1 protects dopaminergic neurons from injury by promoting GDF11 expression in PD model.


Assuntos
Fármacos Neuroprotetores , Doença de Parkinson , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Modelos Animais de Doenças , Neurônios Dopaminérgicos/metabolismo , Fatores de Diferenciação de Crescimento/genética , Mamíferos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacologia , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo
16.
Skelet Muscle ; 12(1): 10, 2022 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-35524286

RESUMO

Skeletal muscle wasting is a clinically remarkable phenotypic feature of pulmonary arterial hypertension (PAH) that increases the risk of mortality. Growth differentiation factor 11 (GDF11), centrally involved in PAH pathogenesis, has an inhibitory effect on skeletal muscle growth in other conditions. However, whether GDF11 is involved in the pathogenesis of skeletal muscle wasting in PAH remains unknown. We showed that serum GDF11 levels in patients were increased following PAH. Skeletal muscle wasting in the MCT-treated PAH model is accompanied by an increase in circulating GDF11 levels and local catabolic markers (Fbx32, Trim63, Foxo1, and protease activity). In vitro GDF11 activated phosphorylation of STAT3. Antagonizing STAT3, with Stattic, in vitro and in vivo, could partially reverse proteolytic pathways including STAT3/socs3 and iNOS/NO in GDF11-meditated muscle wasting. Our findings demonstrate that GDF11 contributes to muscle wasting and the inhibition of its downstream molecule STAT3 shows promise as a therapeutic intervention by which muscle atrophy may be directly prevented in PAH.


Assuntos
Fatores de Diferenciação de Crescimento , Atrofia Muscular , Hipertensão Arterial Pulmonar , Fator de Transcrição STAT3 , Proteínas Morfogenéticas Ósseas/metabolismo , Fatores de Diferenciação de Crescimento/genética , Fatores de Diferenciação de Crescimento/metabolismo , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/metabolismo , Fator de Transcrição STAT3/metabolismo
17.
Int J Mol Sci ; 23(9)2022 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-35563408

RESUMO

Antisense oligonucleotides (ASOs) are agents that modulate gene function. ASO-mediated out-of-frame exon skipping has been employed to suppress gene function. Myostatin, encoded by the MSTN gene, is a potent negative regulator of skeletal muscle growth. ASOs that induce skipping of out-of-frame exon 2 of the MSTN gene have been studied for their use in increasing muscle mass. However, no ASOs are currently available for clinical use. We hypothesized that ASOs against the splicing enhancer sequence within exon 1 of the MSTN gene would inhibit maturation of pre-mRNA, thereby suppressing gene function. To explore this hypothesis, ASOs against sequences of exon 1 of the MSTN gene were screened for their ability to reduce mature MSTN mRNA levels. One screened ASO, named KMM001, decreased MSTN mRNA levels in a dose-dependent manner and reciprocally increased MSTN pre-mRNA levels. Accordingly, KMM001 decreased myostatin protein levels. KMM001 inhibited SMAD-mediated myostatin signaling in rhabdomyosarcoma cells. Remarkably, it did not decrease GDF11 mRNA levels, indicating myostatin-specific inhibition. As expected, KMM001 enhanced the proliferation of human myoblasts. We conclude that KMM001 is a novel myostatin inhibitor that inhibits pre-mRNA maturation. KMM001 has great promise for clinical applications and should be examined for its ability to treat various muscle-wasting conditions.


Assuntos
Miostatina , Oligonucleotídeos Antissenso , Proteínas Morfogenéticas Ósseas/metabolismo , Elementos Facilitadores Genéticos , Éxons , Fatores de Diferenciação de Crescimento/genética , Humanos , Músculo Esquelético/metabolismo , Miostatina/antagonistas & inibidores , Miostatina/genética , Miostatina/metabolismo , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Precursores de RNA/genética , Precursores de RNA/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
18.
Placenta ; 121: 145-154, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35339026

RESUMO

INTRODUCTION: Preeclampsia (PE) is one of the main causes of maternal, fetal, and neonatal mortality. So far, the underlying mechanism of this pregnancy-specific syndrome remains unelucidated. The expression of Follistatin (FST) decreased in maternal serum (especially early onset severe PE) and placental trophoblasts of PE patients. However, whether FST-deficiency in preeclamptic placentas alters trophoblast function remains to be determined. METHODS: Trophoblast cell lines were cultured in vitro and LV3 short hairpin RNA (shRNA) was used to silence FST. Growth and differentiation factor 11 (GDF11) expression level in placentas and serum were detected by immunohistochemistry and enzyme-linked immune-sorbent assay, respectively. To verify the effect of reduced FST expression on trophoblasts, microRNA-24-3p, which was predicted to target the 3'-untranslated region (3'-UTR) of FST, was screened out, and miR-24-3p mimic, inhibitor was used to regulate FST expression in trophoblasts. RESULTS: Downregulation of FST significantly enhanced the apoptosis and impaired migration and invasion of trophoblast. Reduced FST caused the upregulation of GDF11 in trophoblasts. Interestingly, GDF11 reduced in preeclamptic placental microvascular endothelial cells. Dysregulation of FST-GDF11-Smad2/3 signaling pathway, leading to increased apoptosis of trophoblast. Expression levels of miR-24-3p, was significantly elevated in preeclamptic placentas. Trophoblast cells transfected with miR-24-3p mimics displayed impaired migration and invasion and increased apoptosis. Treated by miR-24-3p inhibitor, trophoblast cells exhibited rescued function. DISCUSSION: FST-deficiency impaired trophoblast function by upregulating GDF11 levels in trophoblasts. The regulation of FST-GDF11-Smad2/3 axis by microRNAs mimic or inhibitor may be critical to trophoblast function regulation and helps to deepen our understanding of the molecular mechanism of PE.


Assuntos
MicroRNAs , Pré-Eclâmpsia , Regiões 3' não Traduzidas , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Morfogenéticas Ósseas/farmacologia , Movimento Celular , Proliferação de Células/genética , Células Endoteliais/metabolismo , Feminino , Folistatina/genética , Folistatina/metabolismo , Folistatina/farmacologia , Fatores de Diferenciação de Crescimento/genética , Fatores de Diferenciação de Crescimento/metabolismo , Fatores de Diferenciação de Crescimento/farmacologia , Humanos , Recém-Nascido , MicroRNAs/metabolismo , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , Gravidez , Proteína Smad2/metabolismo , Trofoblastos/metabolismo
19.
Skelet Muscle ; 12(1): 7, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35287700

RESUMO

BACKGROUND: Myostatin (MSTN) is a transforming growth factor-ß superfamily member that acts as a major regulator of skeletal muscle mass. GDF-11, which is highly related to MSTN, plays multiple roles during embryonic development, including regulating development of the axial skeleton, kidneys, nervous system, and pancreas. As MSTN and GDF-11 share a high degree of amino acid sequence identity, behave virtually identically in cell culture assays, and utilize similar regulatory and signaling components, a critical question is whether their distinct biological functions result from inherent differences in their abilities to interact with specific regulatory and signaling components or whether their distinct biological functions mainly reflect their differing temporal and spatial patterns of expression. METHODS: We generated and characterized mice in which we precisely replaced in the germline the portion of the Mstn gene encoding the mature C-terminal peptide with the corresponding region of Gdf11. RESULTS: In mice homozygous for the knock-in allele, all of the circulating MSTN protein was replaced with GDF-11, resulting in ~ 30-40-fold increased levels of circulating GDF-11. Male mice homozygous for the knock-in allele had slightly decreased muscle weights, slightly increased weight gain in response to a high-fat diet, slightly increased plasma cholesterol and HDL levels, and significantly decreased bone density and bone mass, whereas female mice were mostly unaffected. CONCLUSIONS: GDF-11 appears to be capable of nearly completely functionally replacing MSTN in the control of muscle mass. The developmental and physiological consequences of replacing MSTN with GDF-11 are strikingly limited.


Assuntos
Músculo Esquelético , Miostatina , Sequência de Aminoácidos , Animais , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Feminino , Células Germinativas/metabolismo , Fatores de Diferenciação de Crescimento/genética , Fatores de Diferenciação de Crescimento/metabolismo , Masculino , Camundongos , Músculo Esquelético/metabolismo , Miostatina/genética , Miostatina/metabolismo , Transdução de Sinais
20.
Nat Commun ; 13(1): 243, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35017475

RESUMO

The vertebral column of individual mammalian species often exhibits remarkable robustness in the number and identity of vertebral elements that form (known as axial formulae). The genetic mechanism(s) underlying this constraint however remain ill-defined. Here, we reveal the interplay of three regulatory pathways (Gdf11, miR-196 and Retinoic acid) is essential in constraining total vertebral number and regional axial identity in the mouse, from cervical through to tail vertebrae. All three pathways have differing control over Hox cluster expression, with heterochronic and quantitative changes found to parallel changes in axial identity. However, our work reveals an additional role for Hox genes in supporting axial elongation within the tail region, providing important support for an emerging view that mammalian Hox function is not limited to imparting positional identity as the mammalian body plan is laid down. More broadly, this work provides a molecular framework to interrogate mechanisms of evolutionary change and congenital anomalies of the vertebral column.


Assuntos
Padronização Corporal/fisiologia , Proteínas Morfogenéticas Ósseas/metabolismo , Fatores de Diferenciação de Crescimento/metabolismo , MicroRNAs/metabolismo , Coluna Vertebral/metabolismo , Tretinoína/metabolismo , Animais , Evolução Biológica , Padronização Corporal/genética , Proteínas Morfogenéticas Ósseas/genética , Genes Homeobox , Fatores de Diferenciação de Crescimento/genética , Proteínas de Homeodomínio , Mamíferos , Camundongos , MicroRNAs/genética , Cauda/metabolismo , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...