Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
1.
Int Immunopharmacol ; 142(Pt B): 113092, 2024 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-39317051

RESUMEN

BACKGROUND: Growth differentiation factor 11 (GDF11) is considered to be a potential molecular target for treating pulpitis. However, whether GDF11 regulates osteogenic/odontogenic differentiation of dental pulp stem cells (DPSCs) to mediate pulpitis process remains unclear. METHODS: Lipopolysaccharide (LPS) was used to induce inflammation conditions in DPSCs. The levels of GDF11, sirtuin 3 (SIRT3), forkhead box O-3 (FOXO3), osteogenic/odontogenic differentiation-related markers were measured by quantitative real-time PCR (qRT-PCR) and western blot (WB). Immunofluorescence staining was used to measure mitophagy. Mitophagy-related proteins were analyzed by WB, and the levels of inflammation factors were examined using qRT-PCR, ELISA and immunohistochemistry. Alkaline phosphatase activity and alizarin red S intensity were evaluated to assess osteogenic differentiation. Acute pulp (AP) injury rat model was constructed to study the role of oe-GDF11 in vivo. RESULTS: GDF11 was downregulated in LPS-induced DPSCs, and LPS suppressed osteogenic/odontogenic differentiation and mitophagy. GDF11 overexpression promoted osteogenic/odontogenic differentiation in DPSCs through the activation of mitophagy. Furthermore, GDF11 upregulated SIRT3 to enhance FOXO3 expression by inhibiting its acetylation. GDF11 ameliorated LPS-induced inflammation and promoted osteogenic/odontogenic differentiation in DPSCs via enhancing SIRT3/FOXO3-mediated mitophagy. Besides, GDF11 overexpression suppressed inflammation and promoted dentin repair in AP rat models. CONCLUSION: GDF11 promoted SIRT3/FOXO3-mediated mitophagy to accelerate osteogenic/odontogenic differentiation in DPSCs, providing a novel target for pulpitis treatment.


Asunto(s)
Proteínas Morfogenéticas Óseas , Diferenciación Celular , Pulpa Dental , Proteína Forkhead Box O3 , Factores de Diferenciación de Crecimiento , Mitofagia , Osteogénesis , Células Madre , Pulpa Dental/citología , Pulpa Dental/metabolismo , Animales , Osteogénesis/efectos de los fármacos , Humanos , Proteína Forkhead Box O3/metabolismo , Proteína Forkhead Box O3/genética , Células Madre/metabolismo , Mitofagia/efectos de los fármacos , Ratas , Factores de Diferenciación de Crecimiento/metabolismo , Factores de Diferenciación de Crecimiento/genética , Proteínas Morfogenéticas Óseas/metabolismo , Células Cultivadas , Odontogénesis , Sirtuina 3/metabolismo , Sirtuina 3/genética , Ratas Sprague-Dawley , Masculino , Lipopolisacáridos , Dentina/metabolismo , Pulpitis/metabolismo , Pulpitis/patología
2.
Eur J Pharmacol ; 978: 176793, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-38960061

RESUMEN

In recent years, a common-used antidiabetic drug, liraglutide, was identified with extra effects on lipid metabolism. Its effects against excessive lipid deposition in bone marrow were gained much attention but not well established. Our aim in the present study is to explore the interaction of miRNAs-mRNAs altered by liraglutide administration during bone marrow adipogenesis in diabetes. To establish the diabetic animal model, rats were treated with high fat diet (HFD) and STZ injection. We then identified the lowering effect of liraglutide on lipids metabolism in the diabetes. During this process, high-throughput sequencing and bioinformatics analyses on miRNAs extracted from bone marrow mesenchymal stem cells (BMSCs) were conducted after liraglutide administration. We then identified five differentially expressed miRNAs (miRNA-150-5p, miRNA-129-5p, miRNA-201-3p, miRNA-201-5p, and miRNA-214-5p). The expressions of the DE miRNAs were verified as temporal specific expression patterns in Day 3 and in Day 7. Among them, miRNA-150-5p expression was more stable and consistent with the sequencing data. Of interest, miR-150-5p overexpression facilitated adipogenesis of BMSCs. But this promotion was alleviated by liraglutide. The predicted target gene of miR-150-5p, GDF11, was validated to be involved in liraglutide alleviated BMSCs' lipid accumulation in diabetes. In vitro, liraglutide increased the GDF11 expression, rescued its down-expression by siGDF11 and inhibit the adipogenesis of BMSCs cultured in high glucose medium. In vivo, liraglutide reversed the HFD-STZ induced excessive lipid droplets by up-regulation of GDF11 expression, which was discounted by agomiR-150-5p injection. Above all, liraglutide might alleviate bone marrow fat accumulation via inactivating miR-150-5p/GDF11 axis in diabetes.


Asunto(s)
Adipogénesis , Diabetes Mellitus Experimental , Liraglutida , Células Madre Mesenquimatosas , MicroARNs , Ratas Sprague-Dawley , Animales , MicroARNs/genética , MicroARNs/metabolismo , Liraglutida/farmacología , Liraglutida/uso terapéutico , Masculino , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Adipogénesis/efectos de los fármacos , Adipogénesis/genética , Ratas , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Factores de Diferenciación de Crecimiento/genética , Factores de Diferenciación de Crecimiento/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/genética , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Dieta Alta en Grasa/efectos adversos , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Regulación de la Expresión Génica/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo
3.
J Cell Mol Med ; 28(7): e18140, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38494851

RESUMEN

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.


Asunto(s)
Neoplasias Hepáticas , Enfermedad del Hígado Graso no Alcohólico , Humanos , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Fibrosis , Cirrosis Hepática/patología , Factores de Diferenciación de Crecimiento/genética , Factores de Diferenciación de Crecimiento/metabolismo , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Neoplasias Hepáticas/patología
4.
Life Sci ; 341: 122507, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38378101

RESUMEN

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.


Asunto(s)
Depresión , Factores de Diferenciación de Crecimiento , Humanos , Depresión/tratamiento farmacológico , Factores de Diferenciación de Crecimiento/genética , Factores de Diferenciación de Crecimiento/metabolismo , Factores de Diferenciación de Crecimiento/farmacología , Factor de Crecimiento Transformador beta , Envejecimiento , Proteínas Morfogenéticas Óseas
5.
J Cell Physiol ; 238(12): 2827-2840, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37801347

RESUMEN

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.


Asunto(s)
Senescencia Celular , Células Madre Mesenquimatosas , Senescencia Celular/genética , Factores de Diferenciación de Crecimiento/genética , Factores de Diferenciación de Crecimiento/metabolismo , Células Madre Mesenquimatosas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Humanos
6.
Kidney Blood Press Res ; 48(1): 209-219, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36780878

RESUMEN

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.


Asunto(s)
Lesión Renal Aguda , Daño por Reperfusión , Animales , Lesión Renal Aguda/etiología , Lesión Renal Aguda/metabolismo , Creatinina/metabolismo , Factores de Diferenciación de Crecimiento/genética , Factores de Diferenciación de Crecimiento/metabolismo , Isquemia/complicaciones , Riñón/metabolismo , Macrófagos/metabolismo , Nitrógeno/metabolismo , Reperfusión/efectos adversos , Daño por Reperfusión/complicaciones , Daño por Reperfusión/metabolismo , Urea/metabolismo
7.
Int J Mol Sci ; 23(9)2022 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-35563408

RESUMEN

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.


Asunto(s)
Miostatina , Oligonucleótidos Antisentido , Proteínas Morfogenéticas Óseas/metabolismo , Elementos de Facilitación Genéticos , Exones , Factores de Diferenciación de Crecimiento/genética , Humanos , Músculo Esquelético/metabolismo , Miostatina/antagonistas & inhibidores , Miostatina/genética , Miostatina/metabolismo , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Oligonucleótidos Antisentido/farmacología , Precursores del ARN/genética , Precursores del ARN/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
8.
Neoplasma ; 69(6): 1386-1395, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36591801

RESUMEN

Breast cancer (BC) is a prevalent neoplasm that occurs in women all over the world. Growth and differentiation factor 11 (GDF11) plays an essential role in cancer progression. This study focused on investigating the biological role and underlying mechanisms of GDF11 in BC. We detected the expression of GDF11 in 27 patients with BC and BC cell lines. Kaplan-Meier plotter was employed to analyze the relationship between GDF11 expression and overall survival (OS) of BC patients. The proliferative, migratory, invasive, and apoptotic abilities of T47D cells were examined. Correlation analysis of GDF11 with Smad ubiquitination regulatory factor 1 (SMURF1) was conducted. The association between GDF11 and the p53 pathway was analyzed by western blot and PFT-α (a p53 inhibitor)-mediated rescue assays. A brief analysis of the role of estrogen receptor alpha (ERα) signaling in BC progression was performed. The results showed that GDF11 was increased in BC tissues and cell lines, and the high expression of GDF11 was associated with the poor OS of BC patients. GDF11 knockdown inhibited the proliferation, migration, and invasion of T47D cells, but promoted cell apoptosis. Meanwhile, the GDF11 knockdown reduced the SMURF1 expression and invoked the p53 pathway activation. SMURF1 overexpression and PFT-α partially blocked the effects of GDF11 knockdown. In addition, GDF11 knockdown and SMURF1 silencing inhibited the activation of the ERα signaling pathway. In summary, GDF11 was involved in the progression of BC by regulating SMURF1-mediated p53 and ERα pathways, opening up a new way for BC treatment.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/genética , Línea Celular Tumoral , Transducción de Señal , Regulación Neoplásica de la Expresión Génica , Proliferación Celular , Proteínas Morfogenéticas Óseas/metabolismo , Factores de Diferenciación de Crecimiento/genética , Factores de Diferenciación de Crecimiento/metabolismo
9.
Arch Med Res ; 53(1): 44-50, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34266696

RESUMEN

BACKGROUND: Melanoma is the deadliest cutaneous malignant tumor with high risks. Though increasing evidence has widely referred to the involvement of long non-coding RNAs (lncRNAs) in the mechanism of tumor development, including melanoma, the functional roles of most lncRNAs in melanoma remain to be explored. In this study, we focus on disclosing the role of long intergenic non-protein coding RNA 1116 (LINC01116) in melanoma. METHODS: Firstly, we detected LINC01116 expression through RT-qPCR. Functional analysis and animal experiments were carried out to assess the role of LINC01116 in vivo and in vitro. Western blot analysis was employed for detection of important markers regarding epithelial mesenchymal transition (EMT). In addition, RNA pulls down, RIP and luciferase reporter assays were performed to probe into the regulatory mechanism of LINC01116. RESULTS: LINC01116 was significantly up regulated in melanoma cells. LINC01116 deficiency abrogated cell proliferation, migration, invasion and EMT in melanoma. Moreover, LINC01116 enhanced growth differentiation factor 11 (GDF11) and syndecan 3 (SDC3) expression through sponging microRNA-3612 (miR-3612). The oncogenic role of the LINC01116/miR-3612/GDF11/SDC3 axis in melanoma was finally demonstrated. CONCLUSION: Conclusively, LINC01116 sequestered miR-3612 and targeted GDF11 and SDC3 to contribute to the progression of melanoma.


Asunto(s)
Proteínas Morfogenéticas Óseas , Regulación Neoplásica de la Expresión Génica , Factores de Diferenciación de Crecimiento , Melanoma , MicroARNs , ARN Largo no Codificante , Sindecano-3 , Animales , Carcinogénesis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Factores de Diferenciación de Crecimiento/genética , Factores de Diferenciación de Crecimiento/metabolismo , Melanoma/genética , Melanoma/patología , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Sindecano-3/genética , Sindecano-3/metabolismo
10.
Stem Cell Res Ther ; 12(1): 456, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34384486

RESUMEN

BACKGROUND: The efficacy of stem cell therapy for ischemia repair has been limited by low cell retention rate. Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor-ß super family, which has multiple effects on development, physiology and diseases. The objective of the study is to investigate whether GDF11 could affect the efficacy of stem cell transplantation. METHODS: We explored the effects of GDF11 on proangiogenic activities of mesenchymal stem cells (MSCs) for angiogenic therapy in vitro and in vivo. RESULTS: Mouse bone marrow-derived MSCs were transduced with lentiviral vector to overexpress GDF11 (MSCGDF11). After exposed to hypoxia and serum deprivation for 48 h, MSCGDF11 were significantly better in viability than control MSCs (MSCvector). MSCGDF11 also had higher mobility and better angiogenic paracrine effects. The cytokine antibody array showed more angiogenic cytokines in the conditioned medium of MSCGDF11 than that of MSCvector, such as epidermal growth factor, platelet-derived growth factor-BB, placenta growth factor. When MSCs (1 × 106 cells in 50 µl) were injected into ischemic hindlimb of mice after femoral artery ligation, MSCGDF11 had higher retention rate in the muscle than control MSCs. Injection of MSCGDF11 resulted in better blood reperfusion and limb salvage than that of control MSCs after 14 days. Significantly more CD31+ endothelial cells and α-SMA + smooth muscle cells were detected in the ischemic muscles that received MSCGDF11. The effects of GDF11 were through activating TGF-ß receptor and PI3K/Akt signaling pathway. CONCLUSION: Our study demonstrated an essential role of GDF11 in promoting therapeutic functions of MSCs for ischemic diseases by enhancing MSC viability, mobility, and angiogenic paracrine functions.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Animales , Proteínas Morfogenéticas Óseas , Células Cultivadas , Células Endoteliales , Femenino , Factores de Diferenciación de Crecimiento/genética , Isquemia/terapia , Ratones , Neovascularización Fisiológica , Fosfatidilinositol 3-Quinasas , Embarazo
11.
Sci Rep ; 11(1): 16767, 2021 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-34408239

RESUMEN

Regulatory T cells (Tregs) are the key cells regulating peripheral autoreactive T lymphocytes. Tregs exert their function by suppressing effector T cells. Tregs have been shown to play essential roles in the control of a variety of physiological and pathological immune responses. However, Tregs are unstable and can lose the expression of FOXP3 and suppressive functions as a consequence of outer stimuli. Available literature suggests that secreted proteins regulate Treg functional states, such as differentiation, proliferation and suppressive function. Identification of secreted proteins that affect Treg cell function are highly interesting for both therapeutic and diagnostic purposes in either hyperactive or immunosuppressed populations. Here, we report a phenotypic screening of a human secretome library in human Treg cells utilising a high throughput flow cytometry technology. Screening a library of 575 secreted proteins allowed us to identify proteins stabilising or destabilising the Treg phenotype as suggested by changes in expression of Treg marker proteins FOXP3 and/or CTLA4. Four proteins including GDF-7, IL-10, PAP and IFNα-7 were identified as positive regulators that increased FOXP3 and/or CTLA4 expression. PAP is a phosphatase. A catalytic-dead version of the protein did not induce an increase in FOXP3 expression. Ten interferon proteins were identified as negative regulators that reduced the expression of both CTLA4 and FOXP3, without affecting cell viability. A transcriptomics analysis supported the differential effect on Tregs of IFNα-7 versus other IFNα proteins, indicating differences in JAK/STAT signaling. A conformational model experiment confirmed a tenfold reduction in IFNAR-mediated ISG transcription for IFNα-7 compared to IFNα-10. This further strengthened the theory of a shift in downstream messaging upon external stimulation. As a summary, we have identified four positive regulators of FOXP3 and/or CTLA4 expression. Further exploration of these Treg modulators and their method of action has the potential to aid the discovery of novel therapies for both autoimmune and infectious diseases as well as for cancer.


Asunto(s)
Proteínas Morfogenéticas Óseas/inmunología , Factores de Diferenciación de Crecimiento/inmunología , Factores Inmunológicos/inmunología , Interferón-alfa/inmunología , Proteínas Asociadas a Pancreatitis/inmunología , Linfocitos T Reguladores/inmunología , Proteínas Morfogenéticas Óseas/genética , Factores de Diferenciación de Crecimiento/genética , Humanos , Factores Inmunológicos/genética , Interferón-alfa/genética , Proteínas Asociadas a Pancreatitis/genética
12.
Cell Death Dis ; 12(7): 665, 2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-34215721

RESUMEN

It has been reported that growth differentiation factor 11 (GDF11) protects against myocardial ischemia/reperfusion (IR) injury, but the underlying mechanisms have not been fully clarified. Considering that GDF11 plays a role in the aging/rejuvenation process and that aging is associated with telomere shortening and cardiac dysfunction, we hypothesized that GDF11 might protect against IR injury by activating telomerase. Human plasma GDF11 levels were significantly lower in acute coronary syndrome patients than in chronic coronary syndrome patients. IR mice with myocardial overexpression GDF11 (oe-GDF11) exhibited a significantly smaller myocardial infarct size, less cardiac remodeling and dysfunction, fewer apoptotic cardiomyocytes, higher telomerase activity, longer telomeres, and higher ATP generation than IR mice treated with an adenovirus carrying a negative control plasmid. Furthermore, mitochondrial biogenesis-related proteins and some antiapoptotic proteins were significantly upregulated by oe-GDF11. These cardioprotective effects of oe-GDF11 were significantly antagonized by BIBR1532, a specific telomerase inhibitor. Similar effects of oe-GDF11 on apoptosis and mitochondrial energy biogenesis were observed in cultured neonatal rat cardiomyocytes, whereas GDF11 silencing elicited the opposite effects to oe-GDF11 in mice. We concluded that telomerase activation by GDF11 contributes to the alleviation of myocardial IR injury through enhancing mitochondrial biogenesis and suppressing cardiomyocyte apoptosis.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Factores de Diferenciación de Crecimiento/metabolismo , Mitocondrias Cardíacas/enzimología , Infarto del Miocardio/enzimología , Daño por Reperfusión Miocárdica/enzimología , Miocitos Cardíacos/enzimología , Biogénesis de Organelos , Telomerasa/metabolismo , Aminobenzoatos/farmacología , Animales , Apoptosis , Proteínas Morfogenéticas Óseas/genética , Estudios de Casos y Controles , Células Cultivadas , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Factores de Diferenciación de Crecimiento/genética , Humanos , Masculino , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/genética , Mitocondrias Cardíacas/patología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Naftalenos/farmacología , Ratas , Transducción de Señal , Telomerasa/antagonistas & inhibidores
13.
Biomed Res Int ; 2021: 5538535, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33855071

RESUMEN

Tumor necrosis factor alpha (TNF-α) plays a critical role in the progression of inflammation and affects the cells of the synovial membrane. Another key factor in the progression of rheumatoid inflammation is interleukin-6 (IL-6). Both TNF-α and IL-6 promote the proliferation of synovial membrane cells thus stimulating the production of matrix metalloproteinases and other cytotoxins and leading towards bone erosion and destruction of the cartilage. Growth differentiation factor-11 (GDF11) and growth differentiation factor-8 (GDF8) which is also known as myostatin are members of the transforming growth factor-ß family and could be used as antagonists to inflammatory responses which are associated with rheumatoid arthritis. In the current study, to elucidate the evolutionary relationships of GDF11 with its homologs from other closely related organisms, a comprehensive phylogenetic analysis was performed. From the phylogram, it was revealed that the clade of Primates that belong to superorder Euarchontoglires showed close evolutionary relationships with order Cetartiodactyla of the Laurasiatheria superorder. Fifty tetrapeptides were devised from conserved regions of GDF11 which served as ligands in protein-ligand docking against TNF-α and IL-6 followed by drug scanning and ADMET profiling of best selected ligands. The peptides SAGP showed strong interactions with IL-6, and peptides AFDP and AGPC showed strong interactions with TNF-α, and all three peptides fulfilled all the pharmacokinetic parameters which are important for bioavailability. The potential of GDF8 as an antagonist to TNF-α and IL-6 was also explored using a protein-protein docking approach. The binding patterns of GDF8 with TNF-α and IL-6 showed that GDF8 could be used as a potential inhibitor of TNF-α and IL-6 to treat rheumatoid arthritis.


Asunto(s)
Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Simulación por Computador , Factores de Diferenciación de Crecimiento/metabolismo , Inflamación/patología , Interleucina-6/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Secuencia de Aminoácidos , Disponibilidad Biológica , Proteínas Morfogenéticas Óseas/genética , Factores de Diferenciación de Crecimiento/genética , Humanos , Interleucina-6/metabolismo , Ligandos , Simulación del Acoplamiento Molecular , Péptidos/química , Péptidos/metabolismo , Péptidos/farmacocinética , Filogenia , Factor de Necrosis Tumoral alfa/metabolismo
14.
J Cell Mol Med ; 25(10): 4623-4636, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33764670

RESUMEN

Thoracic aortic dissection (TAD) is an aortic disease associated with dysregulated extracellular matrix composition and de-differentiation of vascular smooth muscle cells (SMCs). Growth Differentiation Factor 11 (GDF11) is a member of transforming growth factor ß (TGF-ß) superfamily associated with cardiovascular diseases. The present study attempted to investigate the expression of GDF11 in TAD and its effects on aortic SMC phenotype transition. GDF11 level was found lower in the ascending thoracic aortas of TAD patients than healthy aortas. The mouse model of TAD was established by ß-aminopropionitrile monofumarate (BAPN) combined with angiotensin II (Ang II). The expression of GDF11 was also decreased in thoracic aortic tissues accompanied with increased inflammation, arteriectasis and elastin degradation in TAD mice. Administration of GDF11 mitigated these aortic lesions and improved the survival rate of mice. Exogenous GDF11 and adeno-associated virus type 2 (AAV-2)-mediated GDF11 overexpression increased the expression of contractile proteins including ACTA2, SM22α and myosin heavy chain 11 (MYH11) and decreased synthetic markers including osteopontin and fibronectin 1 (FN1), indicating that GDF11 might inhibit SMC phenotype transition and maintain its contractile state. Moreover, GDF11 inhibited the production of matrix metalloproteinase (MMP)-2, 3, 9 in aortic SMCs. The canonical TGF-ß (Smad2/3) signalling was enhanced by GDF11, while its inhibition suppressed the inhibitory effects of GDF11 on SMC de-differentiation and MMP production in vitro. Therefore, we demonstrate that GDF11 may contribute to TAD alleviation via inhibiting inflammation and MMP activity, and promoting the transition of aortic SMCs towards a contractile phenotype, which provides a therapeutic target for TAD.


Asunto(s)
Aorta Torácica/cirugía , Disección Aórtica/prevención & control , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Factores de Diferenciación de Crecimiento/metabolismo , Contracción Muscular , Miocitos del Músculo Liso/fisiología , Disección Aórtica/etiología , Disección Aórtica/metabolismo , Disección Aórtica/patología , Animales , Proteínas Morfogenéticas Óseas/genética , Estudios de Casos y Controles , Proliferación Celular , Femenino , Factores de Diferenciación de Crecimiento/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Miocitos del Músculo Liso/citología
15.
Mol Ther ; 29(4): 1639-1657, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33388417

RESUMEN

Glaucoma is the leading cause of irreversible vision loss, affecting more than 70 million individuals worldwide. Circulatory disturbances of aqueous humor (AH) have long been central pathological contributors to glaucomatous lesions. Thus, targeting the AH outflow is a promising approach to treat glaucoma. However, the epigenetic mechanisms initiating AH outflow disorders and the targeted treatments remain to be developed. Studying glaucoma patients, we identified GDF7 (growth differentiation factor 7) hypomethylation as a crucial event in the onset of AH outflow disorders. Regarding the underlying mechanism, the hypomethylated GDF7 promoter was responsible for the increased GDF7 production and secretion in primary open-angle glaucoma (POAG). Excessive GDF7 protein promoted trabecular meshwork (TM) fibrosis through bone morphogenetic protein receptor type 2 (BMPR2)/Smad signaling and upregulated pro-fibrotic genes, α-smooth muscle actin (α-SMA) and fibronectin (FN). GDF7 protein expression formed a positive feedback loop in glaucomatous TM (GTM). This positive feedback loop was dependent on the activated TET (ten-eleven translocation) enzyme, which kept the GDF7 promoter region hypomethylated. The phenotypic transition in TM fortified the AH outflow resistance, thus elevating the intraocular pressure (IOP) and attenuating the nerve fiber layer. This methylation-dependent mechanism is also confirmed by a machine-learning model in silico with a specificity of 84.38% and a sensitivity of 89.38%. In rhesus monkeys, we developed GDF7 neutralization therapy to inhibit TM fibrosis and consequent AH outflow resistance that contributes to glaucoma. The neutralization therapy achieved high-efficiency control of the IOP (from 21.3 ± 0.3 to 17.6 ± 0.2 mmHg), a three-fold improvement in the outflow facility (from 0.1 to 0.3 µL/min · mmHg), and protection of nerve fibers. This study provides new insights into the epigenetic mechanism of glaucoma and proposes an innovative GDF7 neutralization therapy as a promising intervention.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Proteínas Morfogenéticas Óseas/genética , Fibrosis/terapia , Glaucoma de Ángulo Abierto/terapia , Factores de Diferenciación de Crecimiento/genética , Actinas/genética , Animales , Humor Acuoso/metabolismo , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Metilación de ADN/genética , Modelos Animales de Enfermedad , Fibrosis/genética , Fibrosis/patología , Glaucoma de Ángulo Abierto/genética , Glaucoma de Ángulo Abierto/patología , Factores de Diferenciación de Crecimiento/antagonistas & inhibidores , Humanos , Macaca mulatta/genética , Oxigenasas de Función Mixta/genética , Proteínas Proto-Oncogénicas/genética , Transducción de Señal/genética , Proteínas Smad/genética , Malla Trabecular/metabolismo , Malla Trabecular/patología
16.
Histol Histopathol ; 35(12): 1473-1481, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33146403

RESUMEN

Ulcerative colitis (UC) is a risk factor for carcinogenesis of colorectal cancer, which is associated with disruption of the epithelial barrier and disorder of the inflammatory response. It has been reported that the expression of microRNA (miR)-215 is upregulated in patients with long-term UC. The present study aimed to investigate the effects of miR-215 on lipopolysaccharide (LPS)-induced inflammatory injury in CCD-18Co cells, as well as to identify the underlying possible molecular mechanisms. CCD-18Co cells were treated with 1 µg/ml LPS to induce inflammatory injury. Reverse transcription-quantitative PCR was performed to determine the expression of miR-215 in LPS-treated CCD-18Co cells. Moreover, a dual luciferase reporter system assay was used to evaluate the interaction of miR-215 and growth differentiation factor 11 (GDF11) in CCD-18Co cells. The expression of miR-215 was significantly upregulated in LPS-treated CCD-18Co cells. Knockdown of miR-215 significantly alleviated the inflammatory response and oxidative stress in LPS-treated CCD-18Co cells. In addition, GDF11 was identified as a direct binding target of miR-215 in CCD-18Co cells. Knockdown of miR-215 significantly increased the expression of GDF11, but decreased the expression levels of Toll-like receptor (TLR)4, phosphorylated (p)-p65, iNOS, p-p38 and p-JNK in LPS-treated CCD-18Co cells. Collectively, the present findings indicated that knockdown of miR-215 alleviated oxidative stress and inflammatory response in LPS-treated CCD-18Co cells by upregulating GDF11 expression and inactivating the TLR4/NF-κB and JNK/p38 signaling pathways.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Colitis Ulcerosa/metabolismo , Colon/metabolismo , Factores de Diferenciación de Crecimiento/metabolismo , Mucosa Intestinal/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , MicroARNs/metabolismo , FN-kappa B/metabolismo , Receptor Toll-Like 4/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Antagomirs/farmacología , Proteínas Morfogenéticas Óseas/genética , Línea Celular , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/genética , Colitis Ulcerosa/prevención & control , Colon/efectos de los fármacos , Colon/patología , Regulación hacia Abajo , Factores de Diferenciación de Crecimiento/genética , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Lipopolisacáridos/toxicidad , MicroARNs/genética , Estrés Oxidativo , Fosforilación , Transducción de Señal
17.
Int J Mol Sci ; 21(15)2020 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-32759740

RESUMEN

Congenital dyserythropoietic anemia type II (CDA II) is a hypo-productive anemia defined by ineffective erythropoiesis through maturation arrest of erythroid precursors. CDA II is an autosomal recessive disorder due to loss-of-function mutations in SEC23B. Currently, management of patients with CDA II is based on transfusions, splenectomy, or hematopoietic stem-cell transplantation. Several studies have highlighted benefits of ACE-011 (sotatercept) treatment of ineffective erythropoiesis, which acts as a ligand trap against growth differentiation factor (GDF)11. Herein, we show that GDF11 levels are increased in CDA II, which suggests sotatercept as a targeted therapy for treatment of these patients. Treatment of stable clones of SEC23B-silenced erythroleukemia K562 cells with the iron-containing porphyrin hemin plus GDF11 increased expression of pSMAD2 and reduced nuclear localization of the transcription factor GATA1, with subsequent reduced gene expression of erythroid differentiation markers. We demonstrate that treatment of these SEC23B-silenced K562 cells with RAP-011, a "murinized" ortholog of sotatercept, rescues the disease phenotype by restoring gene expression of erythroid markers through inhibition of the phosphorylated SMAD2 pathway. Our data also demonstrate the effect of RAP-011 treatment in reducing the expression of erythroferrone in vitro, thus suggesting a possible beneficial role of the use of sotatercept in the management of iron overload in patients with CDA II.


Asunto(s)
Anemia Diseritropoyética Congénita/tratamiento farmacológico , Proteínas Morfogenéticas Óseas/genética , Factor de Transcripción GATA1/genética , Factores de Diferenciación de Crecimiento/genética , Proteínas Recombinantes de Fusión/farmacología , Proteínas de Transporte Vesicular/genética , Anemia Diseritropoyética Congénita/genética , Anemia Diseritropoyética Congénita/patología , Células Precursoras Eritroides/metabolismo , Eritropoyesis/genética , Femenino , Humanos , Células K562 , Mutación/genética , Fenotipo , Proteínas Recombinantes de Fusión/genética , Proteína Smad2/genética , Proteína smad3/genética
18.
Histol Histopathol ; 35(10): 1125-1133, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32720699

RESUMEN

BACKGROUND: Chronic kidney disease (CKD) has become a major public health issue, which can lead to renal fibrosis regardless of the initial injury. It has been previously reported that miRNA-1228-3p was correlate with the progression of kidney fibrosis. However, the mechanism by which miRNA-1228-3p regulates renal fibrosis remains unclear. METHODS: Renal tubular epithelial cells (HK-2) were treated with TGF-ß1 (10 ng/ml) in an in vitro model of renal fibrosis. Gene and protein expressions in HK-2 cells were measured by Western-blot and RT-qPCR, respectively. The relation between miRNA-1228-3p and its target gene was investigated by dual luciferase report analysis. RESULTS: Upregulation of miRNA-1228-3p significantly inhibited TGF-ß1-induced fibrosis of HK-2 cells in vitro by targeting GDF11. In addition, miRNA-1228-3p exhibited anti-fibrosis effect through inhibition of the smad2/smad4 signaling pathway. CONCLUSION: Upregulation of miRNA-1228-3p markedly inhibited the progression of renal fibrosis in vitro, indicating that miRNA-1228-3p may serve as a potential novel target for the treatment of renal fibrosis.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Túbulos Renales Proximales/efectos de los fármacos , MicroARNs/metabolismo , Factor de Crecimiento Transformador beta1/toxicidad , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Línea Celular , Células Epiteliales/metabolismo , Células Epiteliales/patología , Fibrosis , Factores de Diferenciación de Crecimiento/genética , Factores de Diferenciación de Crecimiento/metabolismo , Humanos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , MicroARNs/genética , Transducción de Señal , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Regulación hacia Arriba
19.
J Cell Mol Med ; 24(15): 8703-8717, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32588524

RESUMEN

Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor-ß super family. It has multiple effects on development, physiology and diseases. However, the role of GDF11 in the development of mesenchymal stem cells (MSCs) is not clear. To explore the effects of GDF11 on the differentiation and pro-angiogenic activities of MSCs, mouse bone marrow-derived MSCs were engineered to overexpress GDF11 (MSCGDF11 ) and their capacity for differentiation and paracrine actions were examined both in vitro and in vivo. Expression of endothelial markers CD31 and VEGFR2 at the levels of both mRNA and protein was significantly higher in MSCGDF11 than control MSCs (MSCVector ) during differentiation. More tube formation was observed in MSCGDF11 as compared with controls. In an in vivo angiogenesis assay with Matrigel plug, MSCGDF11 showed more differentiation into CD31+ endothelial-like cells and better pro-angiogenic activity as compared with MSCVector . Mechanistically, the enhanced differentiation by GDF11 involved activation of extracellular-signal-related kinase (ERK) and eukaryotic translation initiation factor 4E (EIF4E). Inhibition of either TGF-ß receptor or ERK diminished the effect of GDF11 on MSC differentiation. In summary, our study unveils the function of GDF11 in the pro-angiogenic activities of MSCs by enhancing endothelial differentiation via the TGFß-R/ERK/EIF4E pathway.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Diferenciación Celular/genética , Células Endoteliales/metabolismo , Factores de Diferenciación de Crecimiento/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Neovascularización Fisiológica/genética , Animales , Apoptosis/genética , Proteínas Morfogenéticas Óseas/metabolismo , Movimiento Celular , Células Cultivadas , Células Endoteliales/citología , Expresión Génica , Factores de Diferenciación de Crecimiento/metabolismo , Hipoxia , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
Rejuvenation Res ; 23(4): 324-332, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31805818

RESUMEN

Telomere length (TL), sirtuin (SIRT) 1, growth differentiation factor (GDF) 11, as well as inflammaging have been related to age-related diseases. In healthy subjects, we aimed to investigate whether leukocyte TL (LTL) associated with family history of coronary heart disease (CHD), age, sex, and lifestyle, and further potential covariations between LTL, GDF11, SIRT1 and selected proinflammatory markers. In 118 healthy subjects (18-81 years, 58% females), whole blood was collected for DNA and RNA isolation and polymerase chain reaction relative quantification of LTLs and gene-expression of SIRT1, GDF11, interleukin (IL)-18, and interferon (IFN)Æ´, respectively, and serum SIRT1 and IL-18 analyses. Shorter LTLs were associated with a seven-fold higher frequency of hereditary CHD in subjects with LTLs in quartile (Q)1 compared with Q2-4 (odds ratio = 7.5, 95% confidence interval: 2.5-21.6, p < 0.001, adjusted). We also observed that LTLs in Q4 compared with Q1-3 associated with higher leukocyte expression of SIRT1 and GDF11 (p = 0.052 and p = 0.058), lower IFNÆ´ expression (p = 0.009), and lower circulating IL-18 levels (p = 0.027). SIRT1 and GDF11 expression were strongly intercorrelated (Spearman's rho = 0.85, p < 0.001). Overall, smoking, snus, and alcohol consumption were not associated with LTLs. The observed shorter LTLs in association with elevated expression of SIRT1 and GDF11 and dampened inflammation in hereditary CHD subjects, suggest impending risk of disease. More research are warranted to shed light on early lifestyle interventions targeting these mechanisms, to promote healthier aging in individuals with hereditary burden. Graphical Abstract [Figure: see text].


Asunto(s)
Envejecimiento , Proteínas Morfogenéticas Óseas/metabolismo , Enfermedad Coronaria/fisiopatología , Factores de Diferenciación de Crecimiento/metabolismo , Leucocitos/metabolismo , Sirtuina 1/metabolismo , Homeostasis del Telómero , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Proteínas Morfogenéticas Óseas/genética , Familia , Femenino , Factores de Diferenciación de Crecimiento/genética , Voluntarios Sanos , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Persona de Mediana Edad , Sirtuina 1/genética , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA