Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Life Sci Alliance ; 6(3)2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36631218

RESUMEN

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.


Asunto(s)
Desarrollo Óseo , Factores de Diferenciación de Crecimiento , Músculo Esquelético , Miostatina , Animales , Femenino , Ratones , Embarazo , Aminoácidos/química , Aminoácidos/genética , Desarrollo Óseo/genética , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Factores de Diferenciación de Crecimiento/genética , Factores de Diferenciación de Crecimiento/química , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Miostatina/genética , Miostatina/química , Factor de Crecimiento Transformador beta/metabolismo
2.
J Cardiovasc Aging ; 3(4)2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38235060

RESUMEN

Since the exogenous administration of GDF11, a TGF-ß superfamily member, was reported to have beneficial effects in some models of human disease, there have been many research studies in GDF11 biology. However, many studies have now confirmed that exogenous administration of GDF11 can improve physiology in disease models, including cardiac fibrosis, experimental stroke, and disordered metabolism. GDF11 is similar to GDF8 (also called Myostatin), differing only by 11 amino acids in their mature signaling domains. These two proteins are now known to be biochemically different both in vitro and in vivo. GDF11 is much more potent than GDF8 and induces more strongly SMAD2 phosphorylation in the myocardium compared to GDF8. GDF8 and GDF11 prodomain are only 52% identical and are cleaved by different Tolloid proteases to liberate the mature signaling domain from inhibition of the prodomain. Here, we review the state of GDF11 biology, highlighting both resolved and remaining controversies.

3.
Exp Biol Med (Maywood) ; 246(4): 467-482, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33197333

RESUMEN

Heparin and heparan sulfate (HS) are highly sulfated polysaccharides covalently bound to cell surface proteins, which directly interact with many extracellular proteins, including the transforming growth factor-ß (TGFß) family ligand antagonist, follistatin 288 (FS288). Follistatin neutralizes the TGFß ligands, myostatin and activin A, by forming a nearly irreversible non-signaling complex by surrounding the ligand and preventing interaction with TGFß receptors. The FS288-ligand complex has higher affinity than unbound FS288 for heparin/HS, which accelerates ligand internalization and lysosomal degradation; however, limited information is available for how FS288 interactions with heparin affect ligand binding. Using surface plasmon resonance (SPR) we show that preincubation of FS288 with heparin/HS significantly decreased the association kinetics for both myostatin and activin A with seemingly no effect on the dissociation rate. This observation is dependent on the heparin/HS chain length where small chain lengths less than degree of polymerization 10 (dp10) did not alter association rates but chain lengths >dp10 decreased association rates. In an attempt to understand the mechanism for this observation, we uncovered that heparin induced dimerization of follistatin. Consistent with our SPR results, we found that dimerization only occurs with heparin molecules >dp10. Small-angle X-ray scattering of the FS288 heparin complex supports that FS288 adopts a dimeric configuration that is similar to the FS288 dimer in the ligand-bound state. These results indicate that heparin mediates dimerization of FS288 in a chain-length-dependent manner that reduces the ligand association rate, but not the dissociation rate or antagonistic activity of FS288.


Asunto(s)
Folistatina/metabolismo , Heparina/farmacología , Multimerización de Proteína/efectos de los fármacos , Activinas , Animales , Células CHO , Cricetulus , Humanos , Concentración 50 Inhibidora , Ligandos , Modelos Moleculares , Miostatina , Dispersión del Ángulo Pequeño , Electricidad Estática , Porcinos , Difracción de Rayos X
4.
Sci Rep ; 10(1): 4561, 2020 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-32165710

RESUMEN

Insulin resistance is associated with aging in mice and humans. We have previously shown that administration of recombinant GDF11 (rGDF11) to aged mice alters aging phenotypes in the brain, skeletal muscle, and heart. While the closely related protein GDF8 has a role in metabolism, limited data are available on the potential metabolic effects of GDF11 or GDF8 in aging. To determine the metabolic effects of these two ligands, we administered rGDF11 or rGDF8 protein to young or aged mice fed a standard chow diet, short-term high-fat diet (HFD), or long-term HFD. Under nearly all of these diet conditions, administration of exogenous rGDF11 reduced body weight by 3-17% and significantly improved glucose tolerance in aged mice fed a chow (~30% vs. saline) or HF (~50% vs. saline) diet and young mice fed a HFD (~30%). On the other hand, exogenous rGDF8 showed signifcantly lesser effect or no effect at all on glucose tolerance compared to rGDF11, consistent with data demonstrating that GFD11 is a more potent signaling ligand than GDF8. Collectively, our results show that administration of exogenous rGDF11, but not rGDF8, can reduce diet-induced weight gain and improve metabolic homeostasis.


Asunto(s)
Envejecimiento/metabolismo , Peso Corporal/efectos de los fármacos , Proteínas Morfogenéticas Óseas/administración & dosificación , Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina , Miostatina/administración & dosificación , Envejecimiento/sangre , Envejecimiento/efectos de los fármacos , Animales , Proteínas Morfogenéticas Óseas/farmacología , Metabolismo Energético/efectos de los fármacos , Factores de Diferenciación de Crecimiento/administración & dosificación , Factores de Diferenciación de Crecimiento/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Miostatina/farmacología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos
5.
Sci Rep ; 9(1): 18613, 2019 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-31819086

RESUMEN

Recent advances in CRISPR/Cas gene editing technology have significantly expanded the possibilities and accelerated the pace of creating genetically engineered animal models. However, CRISPR/Cas-based strategies designed to precisely edit the genome can often yield unintended outcomes. Here, we report the use of zygotic CRISPR/Cas9 injections to generate a knock-in GFP reporter mouse at the Gdf11 locus. Phenotypic and genomic characterization of founder animals from these injections revealed a subset that contained the correct targeting event and exhibited GFP expression that, within the hematopoietic system, was restricted predominantly to lymphoid cells. Yet, in another subset of founder mice, we detected aberrant integration events at the target site that dramatically and inaccurately shifted hematopoietic GFP expression from the lymphoid to the myeloid lineage. Additionally, we recovered multiple Gdf11 deletion alleles that modified the C-terminus of the GDF11 protein. When bred to homozygosity, most of these alleles recapitulated skeletal phenotypes reported previously for Gdf11 knockout mice, suggesting that these represent null alleles. However, we also recovered one Gdf11 deletion allele that encodes a novel GDF11 variant protein ("GDF11-WE") predicted to contain two additional amino acids (tryptophan (W) and glutamic acid (E)) at the C-terminus of the mature ligand. Unlike the other Gdf11 deletion alleles recovered in this study, homozygosity for the Gdf11WE allele did not phenocopy Gdf11 knockout skeletal phenotypes. Further investigation using in vivo and in vitro approaches demonstrated that GDF11-WE retains substantial physiological function, indicating that GDF11 can tolerate at least some modifications of its C-terminus and providing unexpected insights into its biochemical activities. Altogether, our study confirms that one-step zygotic injections of CRISPR/Cas gene editing complexes provide a quick and powerful tool to generate gene-modified mouse models. Moreover, our findings underscore the critical importance of thorough characterization and validation of any modified alleles generated by CRISPR, as unintended on-target effects that fail to be detected by simple PCR screening can produce substantially altered phenotypic readouts.


Asunto(s)
Alelos , Proteínas Morfogenéticas Óseas/genética , Sistemas CRISPR-Cas , Eliminación de Gen , Edición Génica , Factores de Diferenciación de Crecimiento/genética , Animales , Femenino , Genes Reporteros , Ingeniería Genética , Genoma , Ácido Glutámico/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Hematopoyéticas/metabolismo , Homocigoto , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Células Mieloides/metabolismo , Fenotipo , Dominios Proteicos , Triptófano/metabolismo
6.
Am J Physiol Heart Circ Physiol ; 317(1): H201-H212, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31125255

RESUMEN

Administration of active growth differentiation factor 11 (GDF11) to aged mice can reduce cardiac hypertrophy, and low serum levels of GDF11 measured together with the related protein, myostatin (also known as GDF8), predict future morbidity and mortality in coronary heart patients. Using mice with a loxP-flanked ("floxed") allele of Gdf11 and Myh6-driven expression of Cre recombinase to delete Gdf11 in cardiomyocytes, we tested the hypothesis that cardiac-specific Gdf11 deficiency might lead to cardiac hypertrophy in young adulthood. We observed that targeted deletion of Gdf11 in cardiomyocytes does not cause cardiac hypertrophy but rather leads to left ventricular dilation when compared with control mice carrying only the Myh6-cre or Gdf11-floxed alleles, suggesting a possible etiology for dilated cardiomyopathy. However, the mechanism underlying this finding remains unclear because of multiple confounding effects associated with the selected model. First, whole heart Gdf11 expression did not decrease in Myh6-cre; Gdf11-floxed mice, possibly because of upregulation of Gdf11 in noncardiomyocytes in the heart. Second, we observed Cre-associated toxicity, with lower body weights and increased global fibrosis, in Cre-only control male mice compared with flox-only controls, making it challenging to infer which changes in Myh6-cre;Gdf11-floxed mice were the result of Cre toxicity versus deletion of Gdf11. Third, we observed differential expression of cre mRNA in Cre-only controls compared with the cardiomyocyte-specific knockout mice, also making comparison between these two groups difficult. Thus, targeted Gdf11 deletion in cardiomyocytes may lead to left ventricular dilation without hypertrophy, but alternative animal models are necessary to understand the mechanism for these findings. NEW & NOTEWORTHY We observed that targeted deletion of growth differentiation factor 11 in cardiomyocytes does not cause cardiac hypertrophy but rather leads to left ventricular dilation compared with control mice carrying only the Myh6-cre or growth differentiation factor 11-floxed alleles. However, the mechanism underlying this finding remains unclear because of multiple confounding effects associated with the selected mouse model.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Cardiomiopatía Dilatada/genética , Eliminación de Gen , Factores de Diferenciación de Crecimiento/genética , Integrasas/genética , Miocitos Cardíacos/metabolismo , Factores de Edad , Animales , Proteínas Morfogenéticas Óseas/deficiencia , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/fisiopatología , Progresión de la Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Predisposición Genética a la Enfermedad , Factores de Diferenciación de Crecimiento/deficiencia , Integrasas/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/patología , Cadenas Pesadas de Miosina/genética , Fenotipo , Función Ventricular Izquierda , Remodelación Ventricular
7.
J Biol Chem ; 294(16): 6333-6343, 2019 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-30814254

RESUMEN

Growth differentiation factor 8 (GDF8; also known as myostatin) and GDF11 are closely related members of the transforming growth factor ß (TGF-ß) family. GDF8 strongly and negatively regulates skeletal muscle growth, and GDF11 has been implicated in various age-related pathologies such as cardiac hypertrophy. GDF8 and GDF11 signaling activities are controlled by the extracellular protein antagonists follistatin; follistatin-like 3 (FSTL3); and WAP, follistatin/kazal, immunoglobulin, Kunitz, and netrin domain-containing (WFIKKN). All of these proteins contain a follistatin domain (FSD) important for ligand binding and antagonism. Here, we investigated the structure and function of the FSD from murine WFIKKN2 and compared it with the FSDs of follistatin and FSTL3. Using native gel shift and surface plasmon resonance analyses, we determined that the WFIKKN2 FSD can interact with both GDF8 and GDF11 and block their interactions with the type II receptor activin A receptor type 2B (ActRIIB). Further, we solved the crystal structure of the WFIKKN2 FSD to 1.39 Å resolution and identified surface-exposed residues that, when substituted with alanine, reduce antagonism of GDF8 in full-length WFIKKN2. Comparison of the WFIKKN2 FSD with those of follistatin and FSTL3 revealed differences in both the FSD structure and position of residues within the domain that are important for ligand antagonism. Taken together, our results indicate that both WFIKKN and follistatin utilize their FSDs to block the type II receptor but do so via different binding interactions.


Asunto(s)
Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Proteínas Portadoras/química , Factores de Diferenciación de Crecimiento/antagonistas & inhibidores , Miostatina/antagonistas & inhibidores , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/metabolismo , Animales , Proteínas Morfogenéticas Óseas/química , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Portadoras/metabolismo , Cristalografía por Rayos X , Proteínas Relacionadas con la Folistatina/química , Proteínas Relacionadas con la Folistatina/metabolismo , Factores de Diferenciación de Crecimiento/química , Factores de Diferenciación de Crecimiento/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Miostatina/química , Miostatina/metabolismo , Resonancia por Plasmón de Superficie
9.
Proc Natl Acad Sci U S A ; 115(5): E866-E875, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29348202

RESUMEN

Growth/differentiation factor 8 (GDF8), or myostatin, negatively regulates muscle mass. GDF8 is held in a latent state through interactions with its N-terminal prodomain, much like TGF-ß. Using a combination of small-angle X-ray scattering and mutagenesis, we characterized the interactions of GDF8 with its prodomain. Our results show that the prodomain:GDF8 complex can exist in a fully latent state and an activated or "triggered" state where the prodomain remains in complex with the mature domain. However, these states are not reversible, indicating the latent GDF8 is "spring-loaded." Structural analysis shows that the prodomain:GDF8 complex adopts an "open" configuration, distinct from the latency state of TGF-ß and more similar to the open state of Activin A and BMP9 (nonlatent complexes). We determined that GDF8 maintains similar features for latency, including the alpha-1 helix and fastener elements, and identified a series of mutations in the prodomain of GDF8 that alleviate latency, including I56E, which does not require activation by the protease Tolloid. In vivo, active GDF8 variants were potent negative regulators of muscle mass, compared with WT GDF8. Collectively, these results help characterize the latency and activation mechanisms of GDF8.


Asunto(s)
Miostatina/química , Activinas/química , Animales , Atrofia/patología , Diferenciación Celular , Dependovirus , Factor 2 de Diferenciación de Crecimiento , Factores de Diferenciación de Crecimiento/química , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Mutagénesis , Mutación , Miostatina/genética , Dominios Proteicos , Dispersión del Ángulo Pequeño , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
10.
Nat Struct Mol Biol ; 24(12): 1093-1099, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29131142

RESUMEN

Apolipoprotein (apo)A-I is an organizing scaffold protein that is critical to high-density lipoprotein (HDL) structure and metabolism, probably mediating many of its cardioprotective properties. However, HDL biogenesis is poorly understood, as lipid-free apoA-I has been notoriously resistant to high-resolution structural study. Published models from low-resolution techniques share certain features but vary considerably in shape and secondary structure. To tackle this central issue in lipoprotein biology, we assembled a team of structural biologists specializing in apolipoproteins and set out to build a consensus model of monomeric lipid-free human apoA-I. Combining novel and published cross-link constraints, small-angle X-ray scattering (SAXS), hydrogen-deuterium exchange (HDX) and crystallography data, we propose a time-averaged model consistent with much of the experimental data published over the last 40 years. The model provides a long-sought platform for understanding and testing details of HDL biogenesis, structure and function.


Asunto(s)
Apolipoproteína A-I/metabolismo , Lipoproteínas HDL/biosíntesis , Lipoproteínas HDL/metabolismo , Modelos Moleculares , Cardiotónicos/metabolismo , Simulación por Computador , Cristalografía por Rayos X , Humanos , Estructura Secundaria de Proteína
11.
BMC Biol ; 15(1): 19, 2017 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-28257634

RESUMEN

BACKGROUND: Growth/differentiation factor 8 (GDF8) and GDF11 are two highly similar members of the transforming growth factor ß (TGFß) family. While GDF8 has been recognized as a negative regulator of muscle growth and differentiation, there are conflicting studies on the function of GDF11 and whether GDF11 has beneficial effects on age-related dysfunction. To address whether GDF8 and GDF11 are functionally identical, we compared their signaling and structural properties. RESULTS: Here we show that, despite their high similarity, GDF11 is a more potent activator of SMAD2/3 and signals more effectively through the type I activin-like receptor kinase receptors ALK4/5/7 than GDF8. Resolution of the GDF11:FS288 complex, apo-GDF8, and apo-GDF11 crystal structures reveals unique properties of both ligands, specifically in the type I receptor binding site. Lastly, substitution of GDF11 residues into GDF8 confers enhanced activity to GDF8. CONCLUSIONS: These studies identify distinctive structural features of GDF11 that enhance its potency, relative to GDF8; however, the biological consequences of these differences remain to be determined.


Asunto(s)
Proteínas Morfogenéticas Óseas/química , Factores de Diferenciación de Crecimiento/química , Miostatina/química , Miostatina/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Proteínas Morfogenéticas Óseas/metabolismo , Células Cultivadas , Cristalografía por Rayos X , Folistatina/metabolismo , Genes Reporteros , Factores de Diferenciación de Crecimiento/antagonistas & inhibidores , Factores de Diferenciación de Crecimiento/metabolismo , Humanos , Inyecciones Intravenosas , Ligandos , Luciferasas/metabolismo , Ratones , Modelos Moleculares , Mioblastos/metabolismo , Miocardio/metabolismo , Miostatina/antagonistas & inhibidores , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Alineación de Secuencia , Transducción de Señal , Proteínas Smad/metabolismo , Homología Estructural de Proteína , Relación Estructura-Actividad
12.
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
13.
J Biol Chem ; 291(10): 5439-51, 2016 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-26755744

RESUMEN

Apolipoprotein (apo) A-I mediates many of the anti-atherogenic functions attributed to high density lipoprotein. Unfortunately, efforts toward a high resolution structure of full-length apoA-I have not been fruitful, although there have been successes with deletion mutants. Recently, a C-terminal truncation (apoA-I(Δ185-243)) was crystallized as a dimer. The structure showed two helical bundles connected by a long, curved pair of swapped helical domains. To compare this structure to that existing under solution conditions, we applied small angle x-ray scattering and isotope-assisted chemical cross-linking to apoA-I(Δ185-243) in its dimeric and monomeric forms. For the dimer, we found evidence for the shared domains and aspects of the N-terminal bundles, but not the molecular curvature seen in the crystal. We also found that the N-terminal bundles equilibrate between open and closed states. Interestingly, this movement is one of the transitions proposed during lipid binding. The monomer was consistent with a model in which the long shared helix doubles back onto the helical bundle. Combined with the crystal structure, these data offer an important starting point to understand the molecular details of high density lipoprotein biogenesis.


Asunto(s)
Apolipoproteína A-I/química , Metabolismo de los Lípidos , Simulación de Dinámica Molecular , Secuencia de Aminoácidos , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Sitios de Unión , Humanos , Datos de Secuencia Molecular , Unión Proteica , Multimerización de Proteína
14.
Circ Res ; 118(1): 29-37, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26489925

RESUMEN

RATIONALE: Growth differentiation factor 11 (GDF11) and GDF8 are members of the transforming growth factor-ß superfamily sharing 89% protein sequence homology. We have previously shown that circulating GDF11 levels decrease with age in mice. However, a recent study by Egerman et al reported that GDF11/8 levels increase with age in mouse serum. OBJECTIVE: Here, we clarify the direction of change of circulating GDF11/8 levels with age and investigate the effects of GDF11 administration on the murine heart. METHODS AND RESULTS: We validated our previous finding that circulating levels of GDF11/8 decline with age in mice, rats, horses, and sheep. Furthermore, we showed by Western analysis that the apparent age-dependent increase in GDF11 levels, as reported by Egerman et al, is attributable to cross-reactivity of the anti-GDF11 antibody with immunoglobulin, which is known to increase with age. GDF11 administration in mice rapidly activated SMAD2 and SMAD3 signaling in myocardium in vivo and decreased cardiac mass in both young (2-month-old) and old (22-month-old) mice in a dose-dependent manner after only 9 days. CONCLUSIONS: Our study confirms an age-dependent decline in serum GDF11/8 levels in multiple mammalian species and that exogenous GDF11 rapidly activates SMAD signaling and reduces cardiomyocyte size. Unraveling the molecular basis for the age-dependent decline in GDF11/8 could yield insight into age-dependent cardiac pathologies.


Asunto(s)
Envejecimiento/sangre , Proteínas Morfogenéticas Óseas/sangre , Factores de Diferenciación de Crecimiento/sangre , Miostatina/sangre , Animales , Biomarcadores/sangre , Caballos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Ovinos
15.
Endocrinology ; 157(1): 282-91, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26580671

RESUMEN

A potentially novel approach for treating obesity includes attenuating myostatin as this increases muscle mass and decreases fat mass. Notwithstanding, conflicting studies report that myostatin stimulates or inhibits adipogenesis and it is unknown whether reduced adiposity with myostatin attenuation results from changes in fat deposition or adipogenesis. We therefore quantified changes in the stem, transit amplifying and progenitor cell pool in white adipose tissue (WAT) and brown adipose tissue (BAT) using label-retaining wild-type and mstn(-/-) (Jekyll) mice. Muscle mass was larger in Jekyll mice, WAT and BAT mass was smaller and label induction was equal in all tissues from both wild-type and Jekyll mice. The number of label-retaining cells, however, dissipated quicker in WAT and BAT of Jekyll mice and was only 25% and 17%, respectively, of wild-type cell counts 1 month after induction. Adipose cell density was significantly higher in Jekyll mice and increased over time concomitant with label-retaining cell disappearance, which is consistent with enhanced expansion and differentiation of the stem, transit amplifying and progenitor pool. Stromal vascular cells from Jekyll WAT and BAT differentiated into mature adipocytes at a faster rate than wild-type cells and although Jekyll WAT cells also proliferated quicker in vitro, those from BAT did not. Differentiation marker expression in vitro, however, suggests that mstn(-/-) BAT preadipocytes are far more sensitive to the suppressive effects of myostatin. These results suggest that myostatin attenuation stimulates adipogenesis in vivo and that the reduced adiposity in mstn(-/-) animals results from nutrient partitioning away from fat and in support of muscle.


Asunto(s)
Adipogénesis , Tejido Adiposo Pardo/citología , Tejido Adiposo Blanco/citología , Miostatina/antagonistas & inhibidores , Células Madre/citología , Regulación hacia Arriba , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Adiposidad , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Proliferación Celular , Células Cultivadas , Regulación hacia Abajo , Metabolismo Energético , Femenino , Humanos , Cinética , Ratones , Ratones Noqueados , Desarrollo de Músculos , Miostatina/genética , Miostatina/metabolismo , Células Madre/metabolismo
16.
J Biol Chem ; 290(17): 10689-702, 2015 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-25733664

RESUMEN

Apolipoprotein (apo)A-IV is a lipid emulsifying protein linked to a range of protective roles in obesity, diabetes, and cardiovascular disease. It exists in several states in plasma including lipid-bound in HDL and chylomicrons and as monomeric and dimeric lipid-free/poor forms. Our recent x-ray crystal structure of the central domain of apoA-IV shows that it adopts an elongated helical structure that dimerizes via two long reciprocating helices. A striking feature is the alignment of conserved proline residues across the dimer interface. We speculated that this plays important roles in the structure of the lipid-free protein and its ability to bind lipid. Here we show that the systematic conversion of these prolines to alanine increased the thermodynamic stability of apoA-IV and its propensity to oligomerize. Despite the structural stabilization, we noted an increase in the ability to bind and reorganize lipids and to promote cholesterol efflux from cells. The novel properties of these mutants allowed us to isolate the first trimeric form of an exchangeable apolipoprotein and characterize it by small-angle x-ray scattering and chemical cross-linking. The results suggest that the reciprocating helix interaction is a common feature of all apoA-IV oligomers. We propose a model of how self-association of apoA-IV can result in spherical lipoprotein particles, a model that may have broader applications to other exchangeable apolipoprotein family members.


Asunto(s)
Apolipoproteínas A/química , Apolipoproteínas A/metabolismo , Alanina/química , Sustitución de Aminoácidos , Apolipoproteínas A/genética , Colesterol/metabolismo , Secuencia Conservada , Humanos , Liposomas/metabolismo , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Prolina/química , Unión Proteica , Multimerización de Proteína , Estabilidad Proteica , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Dispersión del Ángulo Pequeño , Termodinámica , Difracción de Rayos X
17.
J Biol Chem ; 290(12): 7506-16, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25657005

RESUMEN

Myostatin, a member of the TGF-ß family of ligands, is a strong negative regulator of muscle growth. As such, it is a prime therapeutic target for muscle wasting disorders. Similar to other TGF-ß family ligands, myostatin is neutralized by binding one of a number of structurally diverse antagonists. Included are the antagonists GASP-1 and GASP-2, which are unique in that they specifically antagonize myostatin. However, little is known from a structural standpoint describing the interactions of GASP antagonists with myostatin. Here, we present the First low resolution solution structure of myostatin-free and myostatin-bound states of GASP-1 and GASP-2. Our studies have revealed GASP-1, which is 100 times more potent than GASP-2, preferentially binds myostatin in an asymmetrical 1:1 complex, whereas GASP-2 binds in a symmetrical 2:1 complex. Additionally, C-terminal truncations of GASP-1 result in less potent myostatin inhibitors that form a 2:1 complex, suggesting that the C-terminal domains of GASP-1 are the primary mediators for asymmetric complex formation. Overall, this study provides a new perspective on TGF-ß antagonism, where closely related antagonists can utilize different ligand-binding strategies.


Asunto(s)
Proteínas Portadoras/metabolismo , Miostatina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Células CHO , Cromatografía en Gel , Cricetinae , Cricetulus , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Unión Proteica , Dispersión de Radiación , Resonancia por Plasmón de Superficie , Ultracentrifugación
18.
Expert Opin Ther Pat ; 25(5): 619-24, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25632990

RESUMEN

Muscular dystrophies (MD) are commonly characterized by progressive loss of muscle mass and function. It is hypothesized that therapeutic blockade of the TGF-ß ligand myostatin, a negative regulator of muscle mass, will stimulate muscle growth and restore muscle function. Although many anti-myostatin targets are currently being pursued in the clinical setting, the efficacies of the tested molecules have shown mixed results. The patent WO2014043344 describes a novel approach for myostatin inhibition using a modified fibronectin type III domain that could potentially be used to treat MD and other muscle-related pathologies.


Asunto(s)
Fibronectinas/metabolismo , Distrofias Musculares/tratamiento farmacológico , Miostatina/antagonistas & inhibidores , Animales , Progresión de la Enfermedad , Diseño de Fármacos , Humanos , Terapia Molecular Dirigida , Enfermedades Musculares/tratamiento farmacológico , Enfermedades Musculares/fisiopatología , Distrofias Musculares/fisiopatología , Patentes como Asunto
19.
J Biol Chem ; 289(9): 5596-608, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24425874

RESUMEN

Apolipoprotein (apo)A-IV plays important roles in dietary lipid and glucose metabolism, and knowledge of its structure is required to fully understand the molecular basis of these functions. However, typical of the entire class of exchangeable apolipoproteins, its dynamic nature and affinity for lipid has posed challenges to traditional high resolution structural approaches. We previously reported an x-ray crystal structure of a dimeric truncation mutant of apoA-IV, which showed a unique helix-swapping molecular interface. Unfortunately, the structures of the N and C termini that are important for lipid binding were not visualized. To build a more complete model, we used chemical cross-linking to derive distance constraints across the full-length protein. The approach was enhanced with stable isotope labeling to overcome ambiguities in determining molecular span of the cross-links given the remarkable similarities in the monomeric and dimeric apoA-IV structures. Using 51 distance constraints, we created a starting model for full-length monomeric apoA-IV and then subjected it to two modeling approaches: (i) molecular dynamics simulations and (ii) fitting to small angle x-ray scattering data. This resulted in the most detailed models yet for lipid-free monomeric or dimeric apoA-IV. Importantly, these models were of sufficient detail to direct the experimental identification of new functional residues that participate in a "clasp" mechanism to modulate apoA-IV lipid affinity. The isotope-assisted cross-linking approach should prove useful for further study of this family of apolipoproteins in both the lipid-free and -bound states.


Asunto(s)
Apolipoproteínas A/química , Simulación de Dinámica Molecular , Apolipoproteínas A/genética , Cristalografía por Rayos X , Humanos , Dispersión del Ángulo Pequeño , Difracción de Rayos X
20.
Endocrinology ; 155(3): 670-5, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24424069

RESUMEN

The immortal C2C12 cell line originates from dystrophic mouse thigh muscle and has been used to study the endocrine control of muscle cell growth, development, and function, including those actions regulated by myostatin. Previous studies suggest that high concentrations of recombinant myostatin generated in bacteria inhibit C2C12 proliferation and differentiation. Recombinant myostatin generated in eukaryotic systems similarly inhibits the proliferation of primary myosatellite cells, but consequently initiates, rather than inhibits, their differentiation and is bioactive at far lower concentrations. Our studies indicate that 2 different sources of recombinant myostatin made in eukaryotes stimulate, not inhibit, C2C12 proliferation. This effect occurred at different cell densities and serum concentrations and in the presence of IGF-I, a potent myoblast mitogen. This stimulatory effect was comparable to that obtained with TGFß1, a related factor that also inhibits primary myosatellite cell proliferation. Attenuating the myostatin/activin (ie, Acvr2b) and TGFß1 receptor signaling pathways with the Alk4/5 and Alk5 inhibitors, SB431542 and SB505142, respectively, similarly attenuated proliferation induced by serum, myostatin or TGFß1 and in a dose-dependent manner. In serum-free medium, both myostatin and TGFß1 stimulated Smad2 phosphorylation, but not that of Smad3, and a Smad3 inhibitor (SIS3) only inhibited proliferation in cells cultured in high serum. Thus, myostatin and TGFß1 stimulate C2C12 proliferation primarily via Smad2. These results together question the physiological relevance of the C2C12 model and previous studies using recombinant myostatin generated in bacteria. They also support the alternative use of primary myosatellite cells and recombinant myostatin generated in eukaryotes.


Asunto(s)
Proliferación Celular , Mioblastos/citología , Miostatina/fisiología , Animales , Línea Celular , Dimerización , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ligandos , Ratones , Estructura Terciaria de Proteína , Proteínas Recombinantes/metabolismo , Transducción de Señal , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...