Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Front Immunol ; 14: 1323670, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38143761

RESUMEN

Growth differentiation factor 11 (GDF11) is one of the important factors in the pathophysiological process of animals. It is widely expressed in many tissues and organs of animals, showing its wide biological activity and potential application value. Previous research has demonstrated that GDF11 has a therapeutic effect on various diseases, such as anti-myocardial aging and anti-tumor. This has not only sparked intense interest and enthusiasm among academics but also spurred some for-profit businesses to attempt to develop GDF11 as a medication for regenerative medicine or anti-aging application. Currently, Sotatercept, a GDF11 antibody drug, is in the marketing application stage, and HS-235 and rGDF11 are in the preclinical research stage. Therefore, we believe that figuring out which cells GDF11 acts on and its current problems should be an important issue in the scientific and commercial communities. Only through extensive, comprehensive research and discussion can we better understand the role and potential of GDF11, while avoiding unnecessary risks and misinformation. In this review, we aimed to summarize the role of GDF11 in different cells and its current controversies and challenges, providing an important reference for us to deeply understand the function of GDF11 and formulate more effective treatment strategies in the future.


Asunto(s)
Células , Factores de Diferenciación de Crecimiento , Humanos , Animales , Factores de Diferenciación de Crecimiento/metabolismo , Factores de Diferenciación de Crecimiento/uso terapéutico , Células/metabolismo , Biomarcadores , Neoplasias/terapia , Cardiomiopatías/terapia , Inflamación/terapia
2.
Cytokine Growth Factor Rev ; 71-72: 82-93, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37414617

RESUMEN

Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor-ß superfamily that has garnered significant attention due to its anti-cardiac aging properties. Many studies have revealed that GDF11 plays an indispensable role in the onset of cardiovascular diseases (CVDs). Consequently, it has emerged as a potential target and novel therapeutic agent for CVD treatment. However, currently, no literature reviews comprehensively summarize the research on GDF11 in the context of CVDs. Therefore, herein, we comprehensively described GDF11's structure, function, and signaling in various tissues. Furthermore, we focused on the latest findings concerning its involvement in CVD development and its potential for clinical translation as a CVD treatment. We aim to provide a theoretical basis for the prospects and future research directions of the GDF11 application regarding CVDs.


Asunto(s)
Enfermedades Cardiovasculares , Humanos , Enfermedades Cardiovasculares/tratamiento farmacológico , Factores de Diferenciación de Crecimiento/uso terapéutico , Envejecimiento , Factor de Crecimiento Transformador beta , Transducción de Señal , Proteínas Morfogenéticas Óseas/uso terapéutico
3.
Acta Pharmacol Sin ; 44(5): 999-1013, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36347996

RESUMEN

Non-healing diabetic wounds (DW) are a serious clinical problem that remained poorly understood. We recently found that topical application of growth differentiation factor 11 (GDF11) accelerated skin wound healing in both Type 1 DM (T1DM) and genetically engineered Type 2 diabetic db/db (T2DM) mice. In the present study, we elucidated the cellular and molecular mechanisms underlying the action of GDF11 on healing of small skin wound. Single round-shape full-thickness wound of 5-mm diameter with muscle and bone exposed was made on mouse dorsum using a sterile punch biopsy 7 days following the onset of DM. Recombinant human GDF11 (rGDF11, 50 ng/mL, 10 µL) was topically applied onto the wound area twice a day until epidermal closure (maximum 14 days). Digital images of wound were obtained once a day from D0 to D14 post-wounding. We showed that topical application of GDF11 accelerated the healing of full-thickness skin wounds in both type 1 and type 2 diabetic mice, even after GDF8 (a muscle growth factor) had been silenced. At the cellular level, GDF11 significantly facilitated neovascularization to enhance regeneration of skin tissues by stimulating mobilization, migration and homing of endothelial progenitor cells (EPCs) to the wounded area. At the molecular level, GDF11 greatly increased HIF-1ɑ expression to enhance the activities of VEGF and SDF-1ɑ, thereby neovascularization. We found that endogenous GDF11 level was robustly decreased in skin tissue of diabetic wounds. The specific antibody against GDF11 or silence of GDF11 by siRNA in healthy mice mimicked the non-healing property of diabetic wound. Thus, we demonstrate that GDF11 promotes diabetic wound healing via stimulating endothelial progenitor cells mobilization and neovascularization mediated by HIF-1ɑ-VEGF/SDF-1ɑ pathway. Our results support the potential of GDF11 as a therapeutic agent for non-healing DW.


Asunto(s)
Diabetes Mellitus Experimental , Células Progenitoras Endoteliales , Factores de Diferenciación de Crecimiento , Cicatrización de Heridas , Animales , Humanos , Ratones , Proteínas Morfogenéticas Óseas/metabolismo , Quimiocina CXCL12/efectos de los fármacos , Quimiocina CXCL12/metabolismo , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Células Progenitoras Endoteliales/metabolismo , Células Progenitoras Endoteliales/patología , Factores de Diferenciación de Crecimiento/uso terapéutico , Factores de Diferenciación de Crecimiento/metabolismo , Neovascularización Fisiológica , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapéutico , Subunidad alfa del Factor 1 Inducible por Hipoxia/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo
4.
Cancer Treat Res Commun ; 32: 100576, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35597155

RESUMEN

BACKGROUND: Despite advances in immunotherapy and targeted therapy, platinum-based chemotherapy remains crucial for many patients with advanced non-small cell lung cancer (NSCLC). Resistance to platinum chemotherapy is common, and predictive biomarkers are needed to tailor treatment to patients likely to respond. In vitro evidence implicates the transforming growth factor-ß (TGF-ß) superfamily ligands activin-A and growth differentiation factor 11 (GDF-11) in innate platinum resistance. We performed a validation study to assess their utility as predictive biomarkers of platinum chemotherapy response in advanced NSCLC. PATIENTS AND METHODS: Our study included 123 adult patients with advanced NSCLC without a driver mutation treated with platinum chemotherapy. 98 patients were from a retrospective cohort and 25 from a prospective cohort. We performed immunohistochemistry staining for Activin-A, GDF-11 and TGF-ß on tumour samples for each patient and analysed IHC expression with objective radiological response and overall survival. RESULTS: The overall median survival was 14.8 months. We performed statistical analysis around a cytoplasmic score of 8/18 for Activin-A and GDF-11 based on previously published work, and 110/30 for TGF-ß based on a calculated cutpoint for significance. No survival difference was detected between these groups for Activin-A (p=0.35), GDF-11 (p=0.57) or TGF-ß (p=0.34). There was no association between rates of progressive disease and high Activin-A expression (p=0.43), high GDF-11 expression (p=1.0) or high TGF-ß expression p=0.89). CONCLUSION: Within the confines of our study, Activin-A, GDF-11 and TGF-ß expression was not a predictor of objective radiological response to chemotherapy or overall survival.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Compuestos Organoplatinos , Activinas/metabolismo , Activinas/uso terapéutico , Adulto , Biomarcadores , Proteínas Morfogenéticas Óseas , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Factores de Diferenciación de Crecimiento/uso terapéutico , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Compuestos Organoplatinos/uso terapéutico , Platino (Metal)/uso terapéutico , Estudios Prospectivos , Estudios Retrospectivos , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/uso terapéutico , Factores de Crecimiento Transformadores/uso terapéutico
5.
Cell Death Dis ; 11(10): 917, 2020 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-33100331

RESUMEN

NLRP3 (Nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3) inflammasome-mediated cardiomyocytes pyroptosis plays a crucial part in progression of acute myocardial infarction (MI). GDF11 (Growth Differentiation Factor 11) has been reported to generate cytoprotective effects in phylogenesis and multiple diseases, but the mechanism that GDF11 contributes to cardioprotection of MI and cardiomyocytes pyroptosis remains poorly understood. In our study, we first determined that GDF11 was abnormally downregulated in the heart tissue of MI mice and hypoxic cardiomyocytes. Moreover, AAV9-GDF11 markedly alleviated heart function in MI mice. Meanwhile, GDF11 overexpression also decreased the pyroptosis of hypoxic cardiomyocytes. PROMO and JASPAR prediction software found that transcription factor HOXA3 was predicted as an important regulator of NLRP3, and was confirmed by ChIP assay. Further analysis identifying GDF11 promoted the Smad2/3 pathway resulted in HOXA3 overexpression. Taken together, our study implies that GDF11 prevents cardiomyocytes pyroptosis via HOXA3/NLRP3 signaling pathway in MI mice.


Asunto(s)
Proteínas Morfogenéticas Óseas/uso terapéutico , Cardiotónicos/uso terapéutico , Factores de Diferenciación de Crecimiento/uso terapéutico , Proteínas de Homeodominio/metabolismo , Infarto del Miocardio/genética , Animales , Proteínas Morfogenéticas Óseas/farmacología , Cardiotónicos/farmacología , Modelos Animales de Enfermedad , Factores de Diferenciación de Crecimiento/farmacología , Humanos , Masculino , Ratones , Piroptosis , Transfección , Regulación hacia Arriba
6.
Brain Res ; 1737: 146802, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32220534

RESUMEN

Growth differentiation factor 11 (GDF11), a member of the transforming growth factor-ß (TGF-ß) superfamily, regulates various biological processes in mammals. The effect of GDF11 in brain injury has not been fully elucidated. Our aim was to investigate the effects of GDF11 in cerebral ischemic injury. The expression level of GDF11 increased significantly in the peri-infarct cerebral cortex. Next, the effect of the intracerebroventricular injection of a GDF11 overexpression lentivirus or rGDF11 was investigated in middle cerebral artery occlusion (MCAO) rats. The preventative effects of the GDF11 overexpression virus on stroke were observed. The delivery of the lentivirus into rats before MCAO significantly reduced the infarct volume and the percentage of apoptotic cells and improved motor function in MCAO rats. Furthermore, it elevated the expression of p-Smad2/3 and promoted neurogenesis and angiogenesis in the ipsilateral SVZ during ischemic injury. More importantly, the therapeutic effects of rGDF11 on stroke were subsequently explored. The results in MCAO rats treated with rGDF11 were found similar to that in those treated with the GDF11 overexpression lentivirus. Together, these findings indicate that GDF11 has neuroprotective and neurorestorative effects in cerebral ischemic injury and provide new insights into the function and mechanism of GDF11 in stroke models.


Asunto(s)
Isquemia Encefálica/metabolismo , Factores de Diferenciación de Crecimiento/metabolismo , Animales , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/fisiopatología , Corteza Cerebral , Modelos Animales de Enfermedad , Factores de Diferenciación de Crecimiento/fisiología , Factores de Diferenciación de Crecimiento/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Masculino , Neovascularización Patológica/tratamiento farmacológico , Neurogénesis , Neuroprotección , Fármacos Neuroprotectores , Ratas , Accidente Cerebrovascular/tratamiento farmacológico
7.
Aging Cell ; 19(1): e13038, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31637864

RESUMEN

Aging is a negative regulator of general homeostasis, tissue function, and regeneration. Changes in organismal energy levels and physiology, through systemic manipulations such as calorie restriction and young blood infusion, can regenerate tissue activity and increase lifespan in aged mice. However, whether these two systemic manipulations could be linked has never been investigated. Here, we report that systemic GDF11 triggers a calorie restriction-like phenotype without affecting appetite or GDF15 levels in the blood, restores the insulin/IGF-1 signaling pathway, and stimulates adiponectin secretion from white adipose tissue by direct action on adipocytes, while repairing neurogenesis in the aged brain. These findings suggest that GDF11 has a pleiotropic effect on an organismal level and that it could be a linking mechanism of rejuvenation between heterochronic parabiosis and calorie restriction. As such, GDF11 could be considered as an important therapeutic candidate for age-related neurodegenerative and metabolic disorders.


Asunto(s)
Adiponectina/metabolismo , Proteínas Morfogenéticas Óseas/uso terapéutico , Restricción Calórica/métodos , Factores de Diferenciación de Crecimiento/uso terapéutico , Envejecimiento , Animales , Proteínas Morfogenéticas Óseas/farmacología , Factores de Diferenciación de Crecimiento/farmacología , Ratones , Fenotipo
8.
J Transl Med ; 17(1): 422, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31847906

RESUMEN

BACKGROUND: The growth differentiation factor 11 (GDF11) was shown to reverse age-related hypertrophy on cardiomyocytes and considered as anti-aging rejuvenation factor. The role of GDF11 in regulating metabolic homeostasis is unclear. In this study, we investigated the functions of GDF11 in regulating metabolic homeostasis and energy balance. METHODS: Using a hydrodynamic injection approach, plasmids carrying a mouse Gdf11 gene were delivered into mice and generated the sustained Gdf11 expression in the liver and its protein level in the blood. High fat diet (HFD)-induced obesity was employed to examine the impacts of Gdf11 gene transfer on HFD-induced adiposity, hyperglycemia, insulin resistance, and hepatic lipid accumulation. The impacts of GDF11 on metabolic homeostasis of obese and diabetic mice were examined using HFD-induced obese and STZ-induced diabetic models. RESULTS: Gdf11 gene transfer alleviates HFD-induced obesity, hyperglycemia, insulin resistance, and fatty liver development. In obese and STZ-induced diabetic mice, Gdf11 gene transfer restores glucose metabolism and improves insulin resistance. Mechanism study reveals that Gdf11 gene transfer increases the energy expenditure of mice, upregulates the expression of genes responsible for thermoregulation in brown adipose tissue, downregulates the expression of inflammatory genes in white adipose tissue and those involved in hepatic lipid and glucose metabolism. Overexpression of GDF11 also activates TGF-ß/Smad2, PI3K/AKT/FoxO1, and AMPK signaling pathways in white adipose tissue. CONCLUSIONS: These results demonstrate that GDF11 plays an important role in regulating metabolic homeostasis and energy balance and could be a target for pharmacological intervention to treat metabolic disease.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/uso terapéutico , Diabetes Mellitus Experimental/metabolismo , Dieta Alta en Grasa , Terapia Genética , Factores de Diferenciación de Crecimiento/genética , Factores de Diferenciación de Crecimiento/uso terapéutico , Homeostasis , Obesidad/prevención & control , Obesidad/terapia , Tejido Adiposo/patología , Animales , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/terapia , Metabolismo Energético/genética , Hígado Graso/complicaciones , Conducta Alimentaria , Regulación de la Expresión Génica , Intolerancia a la Glucosa/complicaciones , Hiperinsulinismo/complicaciones , Hipertrofia , Inflamación/complicaciones , Inflamación/genética , Resistencia a la Insulina , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Masculino , Ratones Obesos , Obesidad/complicaciones , Obesidad/genética , Consumo de Oxígeno/genética , Transducción de Señal , Estreptozocina , Aumento de Peso
9.
Brain Res Bull ; 153: 15-23, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31400495

RESUMEN

Recent animal studies on heterochronic parabiosis (a technique combining the blood circulation of two animals) have revealed that young blood has a powerful rejuvenating effect on brain aging. Circulating factors, especially growth differentiation factor 11 (GDF11) and C-C motif chemokine 11 (CCL11), may play a key role in this effect, which inspires hope for novel approaches to treating age-related cerebral diseases in humans, such as neurodegenerative and neurovascular diseases. Recently, attempts have begun to translate these astonishing and exciting findings from mice to humans and from bench to bedside. However, increasing reports have shown contradictory data, questioning the capacity of these circulating factors to reverse age-related brain dysfunction. In this review, we summarize the current research on the role of young blood, as well as the circulating factors GDF11 and CCL11, in the aging brain and age-related cerebral diseases. We highlight recent controversies, discuss related challenges and provide a future outlook.


Asunto(s)
Envejecimiento/metabolismo , Proteínas Morfogenéticas Óseas/uso terapéutico , Quimiocina CCL11/uso terapéutico , Factores de Diferenciación de Crecimiento/uso terapéutico , Factores de Edad , Envejecimiento/sangre , Envejecimiento/fisiología , Animales , Sangre , Proteínas Morfogenéticas Óseas/sangre , Quimiocina CCL11/sangre , Terapia Enzimática/métodos , Enzimas/sangre , Factores de Diferenciación de Crecimiento/sangre , Ratones , Enfermedades Neurodegenerativas/terapia , Parabiosis/métodos , Enfermedades Vasculares/terapia
10.
Basic Res Cardiol ; 114(3): 20, 2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30900023

RESUMEN

Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor beta 1 (TGF-ß1) superfamily that reverses age-related cardiac hypertrophy, improves muscle regeneration and angiogenesis, and maintains progenitor cells in injured tissue. Recently, targeted myocardial delivery of the GDF11 gene in aged mice was found to reduce heart failure and enhance the proliferation of cardiac progenitor cells after myocardial ischemia-reperfusion (I-R). No investigations have as yet explored the cardioprotective effect of exogenous recombinant GDF11 in acute I-R injury, despite the convenience of its clinical application. We sought to determine whether exogenous recombinant GDF11 protects against acute myocardial I-R injury and investigate the underlying mechanism in Sprague-Dawley rats. We found that GDF11 reduced arrhythmia severity and successfully attenuated myocardial infarction; GDF11 also increased cardiac function after I-R, enhanced HO-1 expression and decreased oxidative damage. GDF11 activated the canonical TGF-ß signaling pathway and inactivated the non-canonical pathways, ERK and JNK signaling pathways. Moreover, administration of GDF11 prior to reperfusion protected the heart from reperfusion damage. Notably, pretreatment with the activin-binding protein, follistatin (FST), inhibited the cardioprotective effects of GDF11 by blocking its activation of Smad2/3 signaling and its inactivation of detrimental TGF-ß signaling. Our data suggest that exogenous GDF11 has cardioprotective effects and may have morphologic and functional recovery in the early stage of myocardial I-R injury. GDF11 may be an innovative therapeutic approach for reducing myocardial I-R injury.


Asunto(s)
Factores de Diferenciación de Crecimiento/uso terapéutico , Corazón/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Daño por Reperfusión Miocárdica/prevención & control , Factor de Crecimiento Transformador beta/metabolismo , Animales , Apoptosis/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Proteína Forkhead Box O3/metabolismo , Factores de Diferenciación de Crecimiento/farmacología , Hemo Oxigenasa (Desciclizante)/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Estrés Oxidativo/efectos de los fármacos , Ratas Sprague-Dawley , Proteínas Smad Reguladas por Receptores/metabolismo
11.
Circ Res ; 123(11): 1220-1231, 2018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30571461

RESUMEN

RATIONALE: Possible beneficial effects of GDF11 (growth differentiation factor 11) on the normal, diseased, and aging heart have been reported, including reversing aging-induced hypertrophy. These effects have not been well validated. High levels of GDF11 have also been shown to cause cardiac and skeletal muscle wasting. These controversies could be resolved if dose-dependent effects of GDF11 were defined in normal and aged animals as well as in pressure overload-induced pathological hypertrophy. OBJECTIVE: To determine dose-dependent effects of GDF11 on normal hearts and those with pressure overload-induced cardiac hypertrophy. METHODS AND RESULTS: Twelve- to 13-week-old C57BL/6 mice underwent transverse aortic constriction (TAC) surgery. One-week post-TAC, these mice received rGDF11 (recombinant GDF11) at 1 of 3 doses: 0.5, 1.0, or 5.0 mg/kg for up to 14 days. Treatment with GDF11 increased plasma concentrations of GDF11 and p-SMAD2 in the heart. There were no significant differences in the peak pressure gradients across the aortic constriction between treatment groups at 1 week post-TAC. Two weeks of GDF11 treatment caused dose-dependent decreases in cardiac hypertrophy as measured by heart weight/tibia length ratio, myocyte cross-sectional area, and left ventricular mass. GDF11 improved cardiac pump function while preventing TAC-induced ventricular dilation and caused a dose-dependent decrease in interstitial fibrosis (in vivo), despite increasing markers of fibroblast activation and myofibroblast transdifferentiation (in vitro). Treatment with the highest dose (5.0 mg/kg) of GDF11 caused severe body weight loss, with significant decreases in both muscle and organ weights and death in both sham and TAC mice. CONCLUSIONS: Although GDF11 treatment can reduce pathological cardiac hypertrophy and associated fibrosis while improving cardiac pump function in pressure overload, high doses of GDF11 cause severe cachexia and death. Use of GDF11 as a therapy could have potentially devastating actions on the heart and other tissues.


Asunto(s)
Caquexia/etiología , Cardiomegalia/tratamiento farmacológico , Factores de Diferenciación de Crecimiento/uso terapéutico , Animales , Factores de Diferenciación de Crecimiento/administración & dosificación , Factores de Diferenciación de Crecimiento/efectos adversos , Factores de Diferenciación de Crecimiento/farmacología , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo
12.
Clin Oral Implants Res ; 29(10): 1050-1059, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30281171

RESUMEN

OBJECTIVES: The purpose of this study was to compare the effects of rhBMP2 with rhBMP9 on ridge augmentation following healing of extraction sockets in dogs. MATERIAL AND METHODS: Five male Beagle dogs, approximately 12 months of age, were used. The mesial roots of the four maxillary premolars were endodontically treated. The distal roots were extracted, and the buccal bony walls removed. All extraction sockets were filled with deproteinized bovine bone mineral (DBBM). A collagen membrane was soaked with 4 µg or 20 µg of rhBMP9, 20 µg of rhBMP2 or sterile saline and placed over the augmented sites. All animals were euthanized after 8 weeks of healing and investigated by micro-CT and histologic analysis. A one-way ANOVA with Tukey's HSD post hoc test was used to compare the differences between the four groups. RESULTS: New bone apposition in all defects was observed from the original bone. RhBMP samples showed an increase in bone formation in the buccal area and better integration of DBBM particles when compared to control sites. Both rhBMP9 defects showed higher values of bone (p = 0.024), bone marrow (p = 0.044), and total augmentation volume (p = 0.033) than the rhBMP2 (20 µg) or control sites. Highest bone area was found in rhBMP9 defects (p = 0.895). CONCLUSIONS: Within the limitations of the present study, rhBMP9 sites demonstrated higher bone-inducing potential in combination with DBBM than rhBMP2. While rhBMP9s failed to demonstrate a clear dose-response relationship to the outcomes, future studies are necessary to evaluate the appropriate dose and carrier systems.


Asunto(s)
Aumento de la Cresta Alveolar/métodos , Proteína Morfogenética Ósea 2/uso terapéutico , Factores de Diferenciación de Crecimiento/uso terapéutico , Animales , Regeneración Ósea/efectos de los fármacos , Sustitutos de Huesos/uso terapéutico , Perros , Factor 2 de Diferenciación de Crecimiento , Humanos , Masculino , Proteínas Recombinantes , Extracción Dental
13.
Am J Physiol Gastrointest Liver Physiol ; 315(6): G909-G920, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30188752

RESUMEN

Growth differentiation factor 11 (GDF11) has an anti-inflammatory effect in the mouse model of atherosclerosis and Alzheimer's disease, but how GDF11 regulates intestinal inflammation during ulcerative colitis (UC) is poorly defined. The Nod-like receptor family pyrin domain-1 containing 3 (NLRP3) inflammasome is closely associated with intestinal inflammation because of its ability to increase IL-1ß secretion. Our aim is to determine whether GDF11 has an effect on attenuating experimental colitis in mice. In this study, using a dextran sodium sulfate (DSS)-induced acute colitis mouse model, we reported that GDF11 treatment attenuated loss of body weight, the severity of the disease activity index, shortening of the colon, and histological changes in the colon. GDF11 remarkably suppressed IL-1ß secretion and NLRP3 inflammasome activation in colon samples and RAW 264.7 cells, such as the levels of NLRP3 and activated caspase-1. Furthermore, we found that GDF11 inhibited NLRP3 inflammasome activation by downregulating the Toll-like receptor 4/NF-κB p65 pathway and reactive oxygen species production via the typical Smad2/3 pathway. Thus, our research shows that GDF11 alleviates DSS-induced colitis by inhibiting NLRP3 inflammasome activation, providing some basis for its potential use in the treatment of UC. NEW & NOTEWORTHY Here, we identify a new role for growth differentiation factor 11 (GDF11), which ameliorates dextran sodium sulfate-induced acute colitis. Meanwhile, we discover a new phenomenon of GDF11 inhibiting IL-1ß secretion and Nod-like receptor family pyrin domain-1 containing 3 (NLRP3) inflammasome activation. These findings reveal that GDF11 is a new potential candidate for the treatment of ulcerative colitis patients with a hyperactive NLRP3 inflammasome.


Asunto(s)
Proteínas Morfogenéticas Óseas/uso terapéutico , Colitis Ulcerosa/tratamiento farmacológico , Factores de Diferenciación de Crecimiento/uso terapéutico , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Proteínas Morfogenéticas Óseas/farmacología , Células CHO , Caspasa 1/metabolismo , Colon/efectos de los fármacos , Colon/metabolismo , Cricetinae , Cricetulus , Femenino , Factores de Diferenciación de Crecimiento/farmacología , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo , Receptor Toll-Like 4/metabolismo , Factor de Transcripción ReIA/metabolismo
14.
Brain Res Bull ; 139: 38-47, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29432795

RESUMEN

The recent suggestion that growth differentiation factor 11 (GDF11) acts as a rejuvenation factor has remained controversial. However, in addition to its role in aging, the relationship between GDF11 and cerebral ischemia is still an important area that needs more investigation. Here we examined effects of GDF11 on angiogenesis and recovery of neurological function in a rat model of stroke. Exogenous recombinant GDF11 (rGDF11) at different doses were directly injected into the tail vein in rats subjected to cerebral ischemia/reperfusion (I/R). Neurobehavioral tests were performed, the proliferation of endothelial cells (ECs) and GDF11 downstream signal activin-like kinase 5 (ALK5) were assessed, and functional microvessels were measured. Results showed that rGDF11 at a dosage of 0.1 mg/kg/day could effectively activate cerebral angiogenesis in vivo. In addition, rGDF11 improved the modified neurological severity scores and the adhesive removal somatosensory test, promoted proliferation of ECs, induced ALK5 and increased vascular surface area and the number of vascular branch points in the peri-infarct cerebral cortex after cerebral I/R. These effects were suppressed by blocking ALK5. Our novel findings shed new light on the role of GDF11. Our results strongly suggest that GDF11 improves neurofunctional recovery from cerebral I/R injury and that this effect is mediated partly through its proangiogenic effect in the peri-infarct cerebral cortex, which is associated with ALK5. Thus, GDF11/ALK5 may represent new therapeutic targets for aiding recovery from stroke.


Asunto(s)
Factores de Diferenciación de Crecimiento/uso terapéutico , Infarto de la Arteria Cerebral Media/complicaciones , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/etiología , Trastornos Psicomotores/tratamiento farmacológico , Trastornos Psicomotores/etiología , Reperfusión , Análisis de Varianza , Animales , Antígenos CD/metabolismo , Infarto Encefálico/tratamiento farmacológico , Infarto Encefálico/etiología , Relación Dosis-Respuesta a Droga , Antígeno Ki-67/metabolismo , Masculino , Examen Neurológico , Ratas , Ratas Sprague-Dawley , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Transducción de Señal/efectos de los fármacos
15.
Clin Implant Dent Relat Res ; 19(4): 600-607, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28466608

RESUMEN

BACKGROUND: Recombinant human bone morphogenetic protein 9 (rhBMP9) has been considered the most osteoinductive growth factor of the BMP-family and has much translation potential for guided bone regeneration (GBR) procedures. PURPOSE: The aim of this study was to compare bone formation using rhBMP9 loaded with different carrier systems including deprotenized bovine bone mineral (BioOss, BO) or collagen barrier membranes (BioGide, BG) in a rabbit GBR model. MATERIALS AND METHODS: rhBMP9 was loaded either on BO; named BO/BMP9, or BG; named BG/BMP9 to investigate the better carrier system for rhBMP9. New bone formation was quantified in a rabbit calvarial defect model using four groups; (1) control (empty, n = 9), (2) BO + BG (n = 9), (3) BO/BMP9 + BG (n = 9; BMP9 loaded onto BO), and (4) BO + BG/BMP9 (n = 9; BMP9 loaded onto BG) by radiographically and histologically at 8 weeks post-surgery. RESULTS: Both BO/BMP9 + BG and BO + BG/BMP9 samples significantly promoted new bone formation when compared to BO + BG samples based on parameters including mineralized tissue volume by microCT analysis, as well as new bone height and new bone area by histomorphometry. Interestingly, BO + BG/BMP9 samples but not BO/BMP9 + BG achieved near perfect horizontal bone defect closure, while demonstrating new bone layers in the defect areas implanted with BG materials and bone formation around BO materials. CONCLUSION: Both BO and BG positively induced bone formation with rhBMP9 in an experimental rabbit GBR model when compared to BO + BG alone. This study revealed that BG-loaded with rhBMP9 promoted better wound closure when compared to BO-loaded with rhBMP9. GBR procedures with growth factors may thus benefit from loading rhBMP9 onto BG-collagen barrier membranes when compared to BO-bone grafting particles. Future large animal studies with different types of bone grafts and barrier membranes are needed to further investigate these trends.


Asunto(s)
Desarrollo Óseo/efectos de los fármacos , Factores de Diferenciación de Crecimiento/uso terapéutico , Regeneración Tisular Dirigida/métodos , Animales , Bovinos , Colágeno/metabolismo , Femenino , Factor 2 de Diferenciación de Crecimiento , Conejos , Proteínas Recombinantes/uso terapéutico , Cráneo/diagnóstico por imagen , Cráneo/efectos de los fármacos , Cráneo/crecimiento & desarrollo , Cráneo/cirugía , Microtomografía por Rayos X
16.
J Int Med Res ; 45(6): 1629-1635, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27565745

RESUMEN

The pathogenesis of myocardial ischaemia/reperfusion injury is multifactorial. Understanding the mechanisms of myocardial ischaemia/reperfusion will benefit patients with ischaemic heart disease. Growth differentiation factor 11 (GDF11), a member of the secreted transforming growth factor-ß superfamily, has been found to reverse age-related hypertrophy, revealing the important role of GDF11 in cardiovascular disease. However, the functions of GDF11 in myocardial ischaemia/reperfusion have not been elucidated yet. A number of signalling molecules are known to occur downstream of GDF11, including mothers against decapentaplegic homolog 3 (SMAD3) and forkhead box O3a (FOXO3a). A hypothesis is presented that GDF11 has protective effects in acute myocardial ischaemia/reperfusion injury through suppression of oxidative stress, prevention of calcium ion overload and promotion of the elimination of abnormal mitochondria via both canonical (SMAD3) and non-canonical (FOXO3a) pathways. Since circulating GDF11 may mainly derive from the spleen, the lack of a spleen may make the myocardium susceptible to damaging insults. Administration of GDF11 may be an efficacious therapy to protect against cardiovascular diseases in splenectomized patients.


Asunto(s)
Cardiotónicos/uso terapéutico , Factores de Diferenciación de Crecimiento/uso terapéutico , Modelos Biológicos , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/prevención & control , Animales , Humanos , Bazo/metabolismo
17.
J Craniomaxillofac Surg ; 45(1): 27-32, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27840120

RESUMEN

In the craniofacial bone field, fibrin sealants are used as coagulant and adhesive tools to stabilize grafts during surgery. Despite this, their exact role in osteogenesis is poorly characterized. In the present study, we aimed to characterize the osteogenic potential of TISSEEL fibrin sealant and used its technology to incorporate growth factors within its matrix. We focused on recombinant human bone morphogenetic protein (rhBMP)-9, which has previously been characterized as one of the strongest osteogenetic inducers in the BMP family. TISSEEL displayed an excellent ability to retain rhBMP9, which was gradually released over a 10-day period. Although TISSEEL decreased bone stromal ST2 cell attachment at 8 h, it displayed normal cell proliferation at 1, 3, and 5 days when compared to tissue culture plastic. Interestingly, TISSEEL had little influence on osteoblast differentiation; however its combination with rhBMP9 significantly increased ALP activity at 7 days, Alizarin Red staining at 14 days, and mRNA levels of osteoblast differentiation markers ALP, bone sialoprotein, and osteocalcin. In summary, although fibrin sealants were shown to have little influence on osteogenesis, their combination with bone-inducing growth factors such as rhBMP9 may serve as an attractive carrier/scaffold for future bone regenerative strategies. Future animal studies are now necessary.


Asunto(s)
Portadores de Fármacos/uso terapéutico , Adhesivo de Tejido de Fibrina/uso terapéutico , Factores de Diferenciación de Crecimiento/uso terapéutico , Osteogénesis/efectos de los fármacos , Ingeniería de Tejidos/métodos , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Portadores de Fármacos/administración & dosificación , Ensayo de Inmunoadsorción Enzimática , Factor 2 de Diferenciación de Crecimiento , Factores de Diferenciación de Crecimiento/administración & dosificación , Técnicas In Vitro , Ratones , Osteoblastos/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa
18.
Oncotarget ; 7(35): 55951-55956, 2016 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-27507054

RESUMEN

GDF11 has recently emerged as a powerful anti-aging candidate, found in young blood, capable of rejuvenating a number of aged tissues, such as heart, skeletal muscle and brain. However, recent reports have shown contradictory data questioning its capacity to reverse age-related tissue dysfunction. The availability of a mouse model of accelerated aging, which shares most of the features occurring in physiological aging, gives us an excellent opportunity to test in vivo therapies aimed at extending lifespan both in pathological and normal aging. On this basis, we wondered whether the proposed anti-aging functions of GDF11 would have an overall effect on longevity. We first confirmed the existence of a reduction in GDF11/8 levels in our mouse model of accelerated aging compared with wild-type littermates. However, we show herein that GDF11 daily administration does not extend lifespan of premature-aged mice.


Asunto(s)
Envejecimiento Prematuro/tratamiento farmacológico , Proteínas Morfogenéticas Óseas/uso terapéutico , Factores de Diferenciación de Crecimiento/uso terapéutico , Longevidad/fisiología , Miostatina/metabolismo , Rejuvenecimiento/fisiología , Envejecimiento Prematuro/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Modelos Animales de Enfermedad , Femenino , Factores de Diferenciación de Crecimiento/metabolismo , Humanos , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/uso terapéutico
19.
Biochim Biophys Acta ; 1862(7): 1237-46, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27085971

RESUMEN

Although BMP-9 has been reported to induce browning of white adipose tissues (WATs) and suppress high fat diet-induced obesity, detailed molecular mechanism needs to be further elucidated. We report here that administration of MB109, a recombinant derivative of human BMP-9, into obese mice enhanced gene expression of fibroblast growth factor 21 (FGF21), a metabolic regulator, and alleviates a spectrum of pathological symptoms due to high fat diet-induced obesity. In addition, periodical injection of MB109 (500µg/kg/week) reduced an amount of lipid droplets in the liver, serum levels of alanine aminotransferase (ALT), and total cholesterol. These results indicate that MB109 is also effective to treat obesity-mediated non-alcoholic fatty liver disease (NAFLD).


Asunto(s)
Fármacos Antiobesidad/uso terapéutico , Factores de Crecimiento de Fibroblastos/genética , Factores de Diferenciación de Crecimiento/uso terapéutico , Obesidad/tratamiento farmacológico , Regulación hacia Arriba/efectos de los fármacos , Alanina Transaminasa/sangre , Animales , Fármacos Antiobesidad/administración & dosificación , Colesterol/sangre , Dieta Alta en Grasa/efectos adversos , Factor 2 de Diferenciación de Crecimiento , Factores de Diferenciación de Crecimiento/administración & dosificación , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/sangre , Obesidad/etiología , Obesidad/genética , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico , Proteína Desacopladora 1/genética
20.
Circ Res ; 118(7): 1125-41; discussion 1142, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-27034275

RESUMEN

Growth differentiation factor 11 (GDF11) and myostatin (or GDF8) are closely related members of the transforming growth factor ß superfamily and are often perceived to serve similar or overlapping roles. Yet, despite commonalities in protein sequence, receptor utilization and signaling, accumulating evidence suggests that these 2 ligands can have distinct functions in many situations. GDF11 is essential for mammalian development and has been suggested to regulate aging of multiple tissues, whereas myostatin is a well-described negative regulator of postnatal skeletal and cardiac muscle mass and modulates metabolic processes. In this review, we discuss the biochemical regulation of GDF11 and myostatin and their functions in the heart, skeletal muscle, and brain. We also highlight recent clinical findings with respect to a potential role for GDF11 and/or myostatin in humans with heart disease. Finally, we address key outstanding questions related to GDF11 and myostatin dynamics and signaling during development, growth, and aging.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Factores de Diferenciación de Crecimiento/fisiología , Miostatina/fisiología , Adulto , Envejecimiento/fisiología , Secuencia de Aminoácidos , Animales , Proteínas Morfogenéticas Óseas/química , Proteínas Morfogenéticas Óseas/deficiencia , Encéfalo/crecimiento & desarrollo , Encéfalo/fisiología , Dimerización , Femenino , Folistatina/metabolismo , Proteínas Relacionadas con la Folistatina/metabolismo , Factores de Diferenciación de Crecimiento/química , Factores de Diferenciación de Crecimiento/deficiencia , Factores de Diferenciación de Crecimiento/uso terapéutico , Corazón/fisiología , Cardiopatías/metabolismo , Humanos , Masculino , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Músculos/fisiología , Miocardio/metabolismo , Miostatina/química , Miostatina/deficiencia , Especificidad de Órganos , Conformación Proteica , Estructura Terciaria de Proteína , Ratas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...