Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
J Biol Chem ; 300(1): 105452, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37949218

RESUMO

Hepcidin, a peptide hormone that negatively regulates iron metabolism, is expressed by bone morphogenetic protein (BMP) signaling. Erythroferrone (ERFE) is an extracellular protein that binds and inhibits BMP ligands, thus positively regulating iron import by indirectly suppressing hepcidin. This allows for rapid erythrocyte regeneration after blood loss. ERFE belongs to the C1Q/TNF-related protein family and is suggested to adopt multiple oligomeric forms: a trimer, a hexamer, and a high molecular weight species. The molecular basis for how ERFE binds BMP ligands and how the different oligomeric states impact BMP inhibition are poorly understood. In this study, we demonstrated that ERFE activity is dependent on the presence of stable dimeric or trimeric ERFE and that larger species are dispensable for BMP inhibition. Additionally, we used an in silico approach to identify a helix, termed the ligand-binding domain, that was predicted to bind BMPs and occlude the type I receptor pocket. We provide evidence that the ligand-binding domain is crucial for activity through luciferase assays and surface plasmon resonance analysis. Our findings provide new insight into how ERFE oligomerization impacts BMP inhibition, while identifying critical molecular features of ERFE essential for binding BMP ligands.


Assuntos
Proteínas Morfogenéticas Ósseas , Hormônios Peptídicos , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Proteínas Morfogenéticas Ósseas/metabolismo , Ligantes , Transdução de Sinais/efeitos dos fármacos , Linhagem Celular , Hormônios Peptídicos/genética , Hormônios Peptídicos/isolamento & purificação , Hormônios Peptídicos/farmacologia , Multimerização Proteica/genética , Mutação , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Domínios Proteicos , Humanos
2.
FASEB J ; 38(1): e23377, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38133902

RESUMO

The roles of anti-Müllerian hormone (AMH) continue to expand, from its discovery as a critical factor in sex determination, through its identification as a regulator of ovarian folliculogenesis, its use in fertility clinics as a measure of ovarian reserve, and its emerging role in hypothalamic-pituitary function. In light of these actions, AMH is considered an attractive therapeutic target to address diverse reproductive needs, including fertility preservation. Here, we set out to characterize the molecular mechanisms that govern AMH synthesis and activity. First, we enhanced the processing of the AMH precursor to >90% by introducing more efficient proprotein convertase cleavage sites (RKKR or ISSRKKRSVSS [SCUT]). Importantly, enhanced processing corresponded with a dramatic increase in secreted AMH activity. Next, based on species differences across the AMH type II receptor-binding interface, we generated a series of human AMH variants and assessed bioactivity. AMHSCUT potency (EC50 4 ng/mL) was increased 5- or 10-fold by incorporating Gln484 Met/Leu535 Thr (EC50 0.8 ng/mL) or Gln484 Met/Gly533 Ser (EC50 0.4 ng/mL) mutations, respectively. Furthermore, the Gln484 Met/Leu535 Thr double mutant displayed enhanced efficacy, relative to AMHSCUT . Finally, we identified residues within the wrist pre-helix of AMH (Trp494 , Gln496 , Ser497 , and Asp498 ) that likely mediate type I receptor binding. Mutagenesis of these residues generated gain- (Trp494 Phe or Gln496 Leu) or loss- (Ser497 Ala) of function AMH variants. Surprisingly, combining activating type I and type II receptor mutations only led to modest additive increases in AMH potency/efficacy. Our study is the first to characterize AMH residues involved in type I receptor binding and suggests a step-wise receptor-complex assembly mechanism, in which enhancement in the affinity of the ligand for either receptor can increase AMH activity beyond the natural level.


Assuntos
Hormônio Antimülleriano , Hormônios Peptídicos , Feminino , Humanos , Hormônio Antimülleriano/genética , Ovário , Sequência de Aminoácidos , Fragmentos de Peptídeos
3.
Biochem J ; 481(7): 547-564, 2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38533769

RESUMO

Activins are one of the three distinct subclasses within the greater Transforming growth factor ß (TGFß) superfamily. First discovered for their critical roles in reproductive biology, activins have since been shown to alter cellular differentiation and proliferation. At present, members of the activin subclass include activin A (ActA), ActB, ActC, ActE, and the more distant members myostatin and GDF11. While the biological roles and signaling mechanisms of most activins class members have been well-studied, the signaling potential of ActE has remained largely unknown. Here, we characterized the signaling capacity of homodimeric ActE. Molecular modeling of the ligand:receptor complexes showed that ActC and ActE shared high similarity in both the type I and type II receptor binding epitopes. ActE signaled specifically through ALK7, utilized the canonical activin type II receptors, ActRIIA and ActRIIB, and was resistant to the extracellular antagonists follistatin and WFIKKN. In mature murine adipocytes, ActE invoked a SMAD2/3 response via ALK7, like ActC. Collectively, our results establish ActE as a specific signaling ligand which activates the type I receptor, ALK7.


Assuntos
Proteínas de Transporte , Fator de Crescimento Transformador beta , Camundongos , Animais , Fator de Crescimento Transformador beta/metabolismo , Ligantes , Receptores de Ativinas/genética , Receptores de Ativinas/metabolismo , Ativinas/metabolismo
4.
Proc Natl Acad Sci U S A ; 118(26)2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34155118

RESUMO

Anti-Müllerian hormone (AMH), or Müllerian-inhibiting substance, is a protein hormone that promotes Müllerian duct regression during male fetal sexual differentiation and regulation of folliculogenesis in women. AMH is a member of the transforming growth factor beta (TGF-ß) family, which has evolved to signal through its own dedicated type II receptor, AMH receptor type II (AMHR2). Structures of other TGF-ß family members have revealed how ligands infer specificity for their cognate receptors; however, it is unknown how AMH binds AMHR2 at the molecular level. Therefore, in this study, we solved the X-ray crystal structure of AMH bound to the extracellular domain of AMHR2 to a resolution of 2.6Å. The structure reveals that while AMH binds AMHR2 in a similar location to Activin and BMP ligand binding to their type II receptors, differences in both AMH and AMHR2 account for a highly specific interaction. Furthermore, using an AMH responsive cell-based luciferase assay, we show that a conformation in finger 1 of AMHR2 and a salt bridge formed by K534 on AMH and D81/E84 of AMHR2 are key to the AMH/AMHR2 interaction. Overall, our study highlights how AMH engages AMHR2 using a modified paradigm of receptor binding facilitated by modifications to the three-finger toxin fold of AMHR2. Furthermore, understanding these elements contributing to the specificity of binding will help in the design of agonists or antagonists or the selection of antibody therapies.


Assuntos
Hormônio Antimülleriano/química , Hormônio Antimülleriano/metabolismo , Receptores de Peptídeos/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Ativinas/química , Sequência de Aminoácidos , Proteínas Morfogenéticas Ósseas/química , Cristalografia por Raios X , Modelos Moleculares , Receptores de Peptídeos/química , Receptores de Fatores de Crescimento Transformadores beta/química , Homologia Estrutural de Proteína
5.
BMC Biol ; 21(1): 16, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36726183

RESUMO

BACKGROUND: Proteins of the TGFß family, which are largely studied as homodimers, are also known to form heterodimers with biological activity distinct from their component homodimers. For instance, heterodimers of bone morphogenetic proteins, including BMP2/BMP7, BMP2/BMP6, and BMP9/BMP10, among others, have illustrated the importance of these heterodimeric proteins within the context of TGFß signaling. RESULTS: In this study, we have determined that mature GDF5 can be combined with mature BMP2 or BMP4 to form BMP2/GDF5 and BMP4/GDF5 heterodimer. Intriguingly, this combination of a BMP2 or BMP4 monomer, which exhibit high affinity to heparan sulfate characteristic to the BMP class, with a GDF5 monomer with low heparan sulfate affinity produces a heterodimer with an intermediate affinity. Using heparin affinity chromatography to purify the heterodimeric proteins, we then determined that both the BMP2/GDF5 and BMP4/GDF5 heterodimers consistently signaled potently across an array of cellular and in vivo systems, while the activities of their homodimeric counterparts were more context dependent. These differences were likely driven by an increase in the combined affinities for the type 1 receptors, Alk3 and Alk6. Furthermore, the X-ray crystal structure of BMP2/GDF5 heterodimer was determined, highlighting the formation of two asymmetric type 1 receptor binding sites that are both unique relative to the homodimers. CONCLUSIONS: Ultimately, this method of heterodimer production yielded a signaling molecule with unique properties relative to the homodimeric ligands, including high affinity to multiple type 1 and moderate heparan binding affinity.


Assuntos
Proteína Morfogenética Óssea 2 , Proteínas Morfogenéticas Ósseas , Proteínas Morfogenéticas Ósseas/metabolismo , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Ligação Proteica , Proteínas de Transporte/metabolismo , Heparitina Sulfato
6.
EMBO J ; 37(3): 367-383, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29330193

RESUMO

Myostatin, a key regulator of muscle mass in vertebrates, is biosynthesised as a latent precursor in muscle and is activated by sequential proteolysis of the pro-domain. To investigate the molecular mechanism by which pro-myostatin remains latent, we have determined the structure of unprocessed pro-myostatin and analysed the properties of the protein in its different forms. Crystal structures and SAXS analyses show that pro-myostatin adopts an open, V-shaped structure with a domain-swapped arrangement. The pro-mature complex, after cleavage of the furin site, has significantly reduced activity compared with the mature growth factor and persists as a stable complex that is resistant to the natural antagonist follistatin. The latency appears to be conferred by a number of distinct features that collectively stabilise the interaction of the pro-domains with the mature growth factor, enabling a regulated stepwise activation process, distinct from the prototypical pro-TGF-ß1. These results provide a basis for understanding the effect of missense mutations in pro-myostatin and pave the way for the design of novel myostatin inhibitors.


Assuntos
Músculo Esquelético/metabolismo , Miostatina/metabolismo , Precursores de Proteínas/metabolismo , Linhagem Celular , Cristalografia por Raios X , Ativação Enzimática/fisiologia , Folistatina/farmacologia , Células HEK293 , Humanos , Miostatina/antagonistas & inibidores , Polimorfismo Genético , Estrutura Secundária de Proteína , Proteólise , Fator de Crescimento Transformador beta/metabolismo
7.
Biochem J ; 478(9): 1733-1747, 2021 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-33876824

RESUMO

Growth differentiation factor 8 (GDF8), a.k.a. myostatin, is a member of the larger TGFß superfamily of signaling ligands. GDF8 has been well characterized as a negative regulator of muscle mass. After synthesis, GDF8 is held latent by a noncovalent complex between the N-terminal prodomain and the signaling ligand. Activation of latent GDF8 requires proteolytic cleavage of the prodomain at residue D99 by a member of the tolloid family of metalloproteases. While tolloid proteases cleave multiple substrates, they lack a conserved consensus sequence. Here, we investigate the tolloid cleavage site of the GDF8 prodomain to determine what residues contribute to tolloid recognition and subsequent proteolysis. Using sequential alanine mutations, we identified several residues adjacent to the scissile bond, including Y94, that when mutated, abolish tolloid-mediated activation of latent GDF8. Using the astacin domain of Tll1 (Tolloid Like 1) we determined that prodomain mutants were more resistant to proteolysis. Purified latent complexes harboring the prodomain mutations, D92A and Y94A, impeded activation by tolloid but could be fully activated under acidic conditions. Finally, we show that co-expression of GDF8 WT with prodomain mutants that were tolloid resistant, suppressed GDF8 activity. Taken together our data demonstrate that residues towards the N-terminus of the scissile bond are important for tolloid-mediated activation of GDF8 and that the tolloid-resistant version of the GDF8 prodomain can function dominant negative to WT GDF8.


Assuntos
Alanina/metabolismo , Ácido Aspártico/metabolismo , Miostatina/genética , Metaloproteases Semelhantes a Toloide/genética , Tirosina/metabolismo , Alanina/genética , Sequência de Aminoácidos , Ácido Aspártico/genética , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Células HEK293 , Humanos , Luciferases/genética , Luciferases/metabolismo , Mutação , Miostatina/química , Miostatina/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteólise , Transdução de Sinais , Metaloproteases Semelhantes a Toloide/química , Metaloproteases Semelhantes a Toloide/metabolismo , Tirosina/genética
8.
Proc Natl Acad Sci U S A ; 116(31): 15505-15513, 2019 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-31315975

RESUMO

TGFß family ligands, which include the TGFßs, BMPs, and activins, signal by forming a ternary complex with type I and type II receptors. For TGFßs and BMPs, structures of ternary complexes have revealed differences in receptor assembly. However, structural information for how activins assemble a ternary receptor complex is lacking. We report the structure of an activin class member, GDF11, in complex with the type II receptor ActRIIB and the type I receptor Alk5. The structure reveals that receptor positioning is similar to the BMP class, with no interreceptor contacts; however, the type I receptor interactions are shifted toward the ligand fingertips and away from the dimer interface. Mutational analysis shows that ligand type I specificity is derived from differences in the fingertips of the ligands that interact with an extended loop specific to Alk4 and Alk5. The study also reveals differences for how TGFß and GDF11 bind to the same type I receptor, Alk5. For GDF11, additional contacts at the fingertip region substitute for the interreceptor interactions that are seen for TGFß, indicating that Alk5 binding to GDF11 is more dependent on direct contacts. In support, we show that a single residue of Alk5 (Phe84), when mutated, abolishes GDF11 signaling, but has little impact on TGFß signaling. The structure of GDF11/ActRIIB/Alk5 shows that, across the TGFß family, different mechanisms regulate type I receptor binding and specificity, providing a molecular explanation for how the activin class accommodates low-affinity type I interactions without the requirement of cooperative receptor interactions.


Assuntos
Ativinas/química , Ativinas/metabolismo , Complexos Multiproteicos/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/química , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Humanos , Camundongos , Modelos Moleculares , Complexos Multiproteicos/química , Ratos , Fator de Crescimento Transformador beta/metabolismo
9.
Biochem J ; 477(17): 3167-3182, 2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32779697

RESUMO

The DAN (differential screening-selected gene aberrative in neuroblastoma) family are a group of secreted extracellular proteins which typically bind to and antagonize BMP (bone morphogenetic protein) ligands. Previous studies have revealed discrepancies between the oligomerization state of certain DAN family members, with SOST (a poor antagonist of BMP signaling) forming a monomer while Grem1, Grem2, and NBL1 (more potent BMP antagonists) form non-disulfide linked dimers. The protein SOSTDC1 (Sclerostin domain containing protein 1) is sequentially similar to SOST, but has been shown to be a better BMP inhibitor. In order to determine the oligomerization state of SOSTDC1 and determine what effect dimerization might have on the mechanism of DAN family antagonism of BMP signaling, we isolated the SOSTDC1 protein and, using a battery of biophysical, biochemical, and structural techniques, showed that SOSTDC1 forms a highly stable non-covalent dimer. Additionally, this SOSTDC1 dimer was shown, using an in vitro cell based assay system, to be an inhibitor of multiple BMP signaling growth factors, including GDF5, while monomeric SOST was a very poor antagonist. These results demonstrate that SOSTDC1 is distinct from paralogue SOST in terms of both oligomerization and strength of BMP inhibition.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Aviárias/química , Multimerização Proteica , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Linhagem Celular , Galinhas , Humanos , Transdução de Sinais
10.
Proc Natl Acad Sci U S A ; 115(5): E866-E875, 2018 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-29348202

RESUMO

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.


Assuntos
Miostatina/química , Ativinas/química , Animais , Atrofia/patologia , Diferenciação Celular , Dependovirus , Fator 2 de Diferenciação de Crescimento , Fatores de Diferenciação de Crescimento/química , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese , Mutação , Miostatina/genética , Domínios Proteicos , Espalhamento a Baixo Ângulo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
11.
J Lipid Res ; 61(2): 244-251, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31831525

RESUMO

APOA5 is a low-abundance exchangeable apolipoprotein that plays critical roles in human triglyceride (TG) metabolism. Indeed, aberrations in the plasma concentration or structure of APOA5 are linked to hypertriglyceridemia, hyperchylomicronemia, myocardial infarction risk, obesity, and coronary artery disease. While it has been successfully produced at low yield in bacteria, the resulting protein had limitations for structure-function studies due to its low solubility under physiological buffer conditions. We hypothesized that the yield and solubility of recombinant APOA5 could be increased by: i) engineering a fusion protein construct in a codon optimized expression vector, ii) optimizing an efficient refolding protocol, and iii) screening buffer systems at physiological pH. The result was a high-yield (25 mg/l) bacterial expression system that produces lipid-free APOA5 soluble at concentrations of up to 10 mg/ml at a pH of 7.8 in bicarbonate buffers. Physical characterization of lipid-free APOA5 indicated that it exists as an array of multimers in solution, and far UV circular dichroism analyses show differences in total α-helicity between acidic and neutral pH buffering conditions. The protein was functional in that it bound and emulsified multilamellar dimyristoyl-phosphatidylcholine vesicles and could inhibit postprandial plasma TG accumulation when injected into C57BL/6J mice orally gavaged with Intralipid.


Assuntos
Apolipoproteína A-V/metabolismo , Animais , Apolipoproteína A-V/biossíntese , Apolipoproteína A-V/isolamento & purificação , Escherichia coli/citologia , Escherichia coli/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/metabolismo
12.
J Biol Chem ; 294(16): 6333-6343, 2019 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-30814254

RESUMO

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.


Assuntos
Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Proteínas de Transporte/química , Fatores de Diferenciação de Crescimento/antagonistas & inibidores , Miostatina/antagonistas & inibidores , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/química , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Transporte/metabolismo , Cristalografia por Raios X , Proteínas Relacionadas à Folistatina/química , Proteínas Relacionadas à Folistatina/metabolismo , Fatores de Diferenciação de Crescimento/química , Fatores de Diferenciação de Crescimento/metabolismo , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Miostatina/química , Miostatina/metabolismo , Ressonância de Plasmônio de Superfície
13.
Hum Mutat ; 40(10): 1813-1825, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31215115

RESUMO

Cleft lip with or without cleft palate (CL/P) is generally viewed as a complex trait with multiple genetic and environmental contributions. In 70% of cases, CL/P presents as an isolated feature and/or deemed nonsyndromic. In the remaining 30%, CL/P is associated with multisystem phenotypes or clinically recognizable syndromes, many with a monogenic basis. Here we report the identification, via exome sequencing, of likely pathogenic variants in two genes that encode interacting proteins previously only linked to orofacial clefting in mouse models. A variant in GDF11 (encoding growth differentiation factor 11), predicting a p.(Arg298Gln) substitution at the Furin protease cleavage site, was identified in one family that segregated with CL/P and both rib and vertebral hypersegmentation, mirroring that seen in Gdf11 knockout mice. In the second family in which CL/P was the only phenotype, a mutation in FST (encoding the GDF11 antagonist, Follistatin) was identified that is predicted to result in a p.(Cys56Tyr) substitution in the region that binds GDF11. Functional assays demonstrated a significant impact of the specific mutated amino acids on FST and GDF11 function and, together with embryonic expression data, provide strong evidence for the importance of GDF11 and Follistatin in the regulation of human orofacial development.


Assuntos
Proteínas Morfogenéticas Ósseas/genética , Fenda Labial/diagnóstico , Fenda Labial/genética , Folistatina/metabolismo , Estudos de Associação Genética , Predisposição Genética para Doença , Fatores de Diferenciação de Crescimento/genética , Mutação , Alelos , Substituição de Aminoácidos , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Linhagem Celular , Biologia Computacional/métodos , Folistatina/química , Estudos de Associação Genética/métodos , Genômica/métodos , Fatores de Diferenciação de Crescimento/antagonistas & inibidores , Humanos , Modelos Moleculares , Linhagem , Conformação Proteica , Sequenciamento do Exoma
14.
Am J Physiol Endocrinol Metab ; 316(6): E1036-E1049, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30888862

RESUMO

Circulating myostatin-attenuating agents are being developed to treat muscle-wasting disease despite their potential to produce serious off-target effects, as myostatin/activin receptors are widely distributed among many nonmuscle tissues. Our studies suggest that the myokine not only inhibits striated muscle growth but also regulates pituitary development and growth hormone (GH) action in the liver. Using a novel myostatin-null label-retaining model (Jekyll mice), we determined that the heterogeneous pool of pituitary stem, transit-amplifying, and progenitor cells in Jekyll mice depletes more rapidly after birth than the pool in wild-type mice. This correlated with increased levels of GH, prolactin, and the cells that secrete these hormones, somatotropes and lactotropes, respectively, in Jekyll pituitaries. Recombinant myostatin also stimulated GH release and gene expression in pituitary cell cultures although inhibiting prolactin release. In primary hepatocytes, recombinant myostatin blocked GH-stimulated expression of two key mediators of growth, insulin-like growth factor (IGF)1 and the acid labile subunit and increased expression of an inhibitor, IGF-binding protein-1. The significance of these findings was demonstrated by smaller muscle fiber size in a model lacking myostatin and liver IGF1 expression (LID-o-Mighty mice) compared with that in myostatin-null (Mighty) mice. These data together suggest that myostatin may regulate pituitary development and function and that its inhibitory actions in muscle may be partly mediated by attenuating GH action in the liver. They also suggest that circulating pharmacological inhibitors of myostatin could produce unintended consequences in these and possibly other tissues.


Assuntos
Hormônio do Crescimento/metabolismo , Hepatócitos/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Lactotrofos/metabolismo , Miostatina/genética , Hipófise/crescimento & desenvolvimento , Prolactina/metabolismo , Somatotrofos/metabolismo , Animais , Caquexia , Proteínas de Transporte/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Desenvolvimento de Medicamentos , Glicoproteínas/efeitos dos fármacos , Glicoproteínas/metabolismo , Hormônio do Crescimento/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/efeitos dos fármacos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Fator de Crescimento Insulin-Like I/efeitos dos fármacos , Lactotrofos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Knockout , Modelos Animais , Miostatina/farmacologia , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Cultura Primária de Células , Prolactina/efeitos dos fármacos , Proteínas Recombinantes , Somatotrofos/efeitos dos fármacos , Células-Tronco
15.
J Lipid Res ; 59(7): 1244-1255, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29773713

RESUMO

APOA1 is the most abundant protein in HDL. It modulates interactions that affect HDL's cardioprotective functions, in part via its activation of the enzyme, LCAT. On nascent discoidal HDL, APOA1 comprises 10 α-helical repeats arranged in an anti-parallel stacked-ring structure that encapsulates a lipid bilayer. Previous chemical cross-linking studies suggested that these APOA1 rings can adopt at least two different orientations, or registries, with respect to each other; however, the functional impact of these structural changes is unknown. Here, we placed cysteine residues at locations predicted to form disulfide bonds in each orientation and then measured APOA1's ability to adopt the two registries during HDL particle formation. We found that most APOA1 oriented with the fifth helix of one molecule across from fifth helix of the other (5/5 helical registry), but a fraction adopted a 5/2 registry. Engineered HDLs that were locked in 5/5 or 5/2 registries by disulfide bonds equally promoted cholesterol efflux from macrophages, indicating functional particles. However, unlike the 5/5 registry or the WT, the 5/2 registry impaired LCAT cholesteryl esterification activity (P < 0.001), despite LCAT binding equally to all particles. Chemical cross-linking studies suggest that full LCAT activity requires a hybrid epitope composed of helices 5-7 on one APOA1 molecule and helices 3-4 on the other. Thus, APOA1 may use a reciprocating thumbwheel-like mechanism to activate HDL-remodeling proteins.


Assuntos
Apolipoproteína A-I/metabolismo , HDL-Colesterol/metabolismo , Fosfatidilcolina-Esterol O-Aciltransferase/metabolismo , Apolipoproteína A-I/genética , Ativação Enzimática , Humanos , Mutação
16.
Circ Res ; 119(3): 434-49, 2016 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-27283840

RESUMO

RATIONALE: We have recently shown that the bone morphogenetic protein (BMP) antagonist Gremlin 2 (Grem2) is required for early cardiac development and cardiomyocyte differentiation. Our initial studies discovered that Grem2 is strongly induced in the adult heart after experimental myocardial infarction (MI). However, the function of Grem2 and BMP-signaling inhibitors after cardiac injury is currently unknown. OBJECTIVE: To investigate the role of Grem2 during cardiac repair and assess its potential to improve ventricular function after injury. METHODS AND RESULTS: Our data show that Grem2 is transiently induced after MI in peri-infarct area cardiomyocytes during the inflammatory phase of cardiac tissue repair. By engineering loss- (Grem2(-/-)) and gain- (TG(Grem2)) of-Grem2-function mice, we discovered that Grem2 controls the magnitude of the inflammatory response and limits infiltration of inflammatory cells in peri-infarct ventricular tissue, improving cardiac function. Excessive inflammation in Grem2(-/-) mice after MI was because of overactivation of canonical BMP signaling, as proven by the rescue of the inflammatory phenotype through administration of the canonical BMP inhibitor, DMH1. Furthermore, intraperitoneal administration of Grem2 protein in wild-type mice was sufficient to reduce inflammation after MI. Cellular analyses showed that BMP2 acts with TNFα to induce expression of proinflammatory proteins in endothelial cells and promote adhesion of leukocytes, whereas Grem2 specifically inhibits the BMP2 effect. CONCLUSIONS: Our results indicate that Grem2 provides a molecular barrier that controls the magnitude and extent of inflammatory cell infiltration by suppressing canonical BMP signaling, thereby providing a novel mechanism for limiting the adverse effects of excessive inflammation after MI.


Assuntos
Proteína Morfogenética Óssea 2/antagonistas & inibidores , Proteína Morfogenética Óssea 2/metabolismo , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/prevenção & controle , Proteínas/metabolismo , Animais , Células Cultivadas , Citocinas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Feminino , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Pirazóis/farmacologia , Pirazóis/uso terapêutico , Quinolinas/farmacologia , Quinolinas/uso terapêutico
17.
Circ Res ; 118(1): 29-37, 2016 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-26489925

RESUMO

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.


Assuntos
Envelhecimento/sangue , Proteínas Morfogenéticas Ósseas/sangue , Fatores de Diferenciação de Crescimento/sangue , Miostatina/sangue , Animais , Biomarcadores/sangue , Cavalos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos , Ovinos
18.
Circ Res ; 118(7): 1125-41; discussion 1142, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-27034275

RESUMO

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.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Fatores de Diferenciação de Crescimento/fisiologia , Miostatina/fisiologia , Adulto , Envelhecimento/fisiologia , Sequência de Aminoácidos , Animais , Proteínas Morfogenéticas Ósseas/química , Proteínas Morfogenéticas Ósseas/deficiência , Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiologia , Dimerização , Feminino , Folistatina/metabolismo , Proteínas Relacionadas à Folistatina/metabolismo , Fatores de Diferenciação de Crescimento/química , Fatores de Diferenciação de Crescimento/deficiência , Fatores de Diferenciação de Crescimento/uso terapêutico , Coração/fisiologia , Cardiopatias/metabolismo , Humanos , Masculino , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Músculos/fisiologia , Miocárdio/metabolismo , Miostatina/química , Miostatina/deficiência , Especificidade de Órgãos , Conformação Proteica , Estrutura Terciária de Proteína , Ratos , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Relação Estrutura-Atividade
19.
Biochem J ; 474(7): 1093-1107, 2017 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-28104757

RESUMO

Bone morphogenetic proteins (BMPs) are regulated by extracellular antagonists of the DAN (differential screening-selected gene aberrative in neuroblastoma) family. Similar to the BMP ligands, certain DAN family members have been shown to interact with heparin and heparan sulfate (HS). Structural studies of DAN family members Gremlin-1 and Gremlin-2 (Grem2) have revealed a dimeric growth factor-like fold where a series of lysine residues cluster along one face of the protein. In the present study, we used mutagenesis, heparin-binding measurements, and cell surface-binding analysis to identify lysine residues that are important for heparin/HS binding in Grem2. We determined that residues involved in heparin/HS binding, while not necessary for BMP antagonism, merge with the heparin/HS-binding epitope of BMP2. Furthermore, the Grem2-BMP2 complex has higher affinity for heparin than the individual proteins and this affinity is not abrogated when the heparin/HS-binding epitope of Grem2 is attenuated. Overall, the present study shows that the Grem2 heparin/HS and BMP-binding epitopes are unique and independent, where, interestingly, the Grem2-BMP2 complex exhibits a significant increase in binding affinity toward heparin moieties that appear to be partially independent of the Grem2 heparin/HS-binding epitope.


Assuntos
Proteína Morfogenética Óssea 2/química , Epitopos/química , Heparina/química , Heparitina Sulfato/química , Peptídeos e Proteínas de Sinalização Intercelular/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Células CHO , Clonagem Molecular , Cricetulus , Citocinas , Epitopos/genética , Epitopos/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Heparina/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mutação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade
20.
BMC Biol ; 15(1): 19, 2017 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-28257634

RESUMO

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.


Assuntos
Proteínas Morfogenéticas Ósseas/química , Fatores de Diferenciação de Crescimento/química , Miostatina/química , Miostatina/metabolismo , Sequência de Aminoácidos , Animais , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Proteínas Morfogenéticas Ósseas/metabolismo , Células Cultivadas , Cristalografia por Raios X , Folistatina/metabolismo , Genes Reporter , Fatores de Diferenciação de Crescimento/antagonistas & inibidores , Fatores de Diferenciação de Crescimento/metabolismo , Humanos , Injeções Intravenosas , Ligantes , Luciferases/metabolismo , Camundongos , Modelos Moleculares , Mioblastos/metabolismo , Miocárdio/metabolismo , Miostatina/antagonistas & inibidores , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Alinhamento de Sequência , Transdução de Sinais , Proteínas Smad/metabolismo , Homologia Estrutural de Proteína , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA