Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Bioorg Med Chem Lett ; 46: 128163, 2021 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-34087433

RESUMO

Myostatin, a negative regulator of muscle mass is a promising target for the treatment of muscle atrophic diseases. The novel myostatin inhibitory peptide, DF-3 is derived from the N-terminal α-helical domain of follistatin, which is an endogenous inhibitor of myostatin and other TGF-ß family members. It has been suggested that the optimization of hydrophobic residues is important to enhance the myostatin inhibition. This study describes a structure-activity relationship study focused on hydrophobic residues of DF-3 and designed to obtain a more potent peptide. A methionine residue in DF-3, which is susceptible to oxidation, was successfully converted to homophenylalanine in DF-100, and a new derivative DF-100, with four amino acid substitutions in DF-3 shows twice the potent inhibitory ability as DF-3. This report provides a new platform of a 14-mer peptide muscle enhancer.


Assuntos
Folistatina/química , Miostatina/antagonistas & inibidores , Peptídeos/farmacologia , Relação Dose-Resposta a Droga , Humanos , Estrutura Molecular , Miostatina/metabolismo , Peptídeos/química , Relação Estrutura-Atividade
2.
Endocrinology ; 162(10)2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33539535

RESUMO

Diabetes is caused by insufficient insulin production from pancreatic beta cells or insufficient insulin action, leading to an inability to control blood glucose. While a wide range of treatments exist to alleviate the symptoms of diabetes, therapies addressing the root cause of diabetes through replacing lost beta cells with functional cells remain an object of active pursuit. We previously demonstrated that genetic deletion of Fstl3, a critical regulator of activin activity, enhanced beta cell number and glucose-responsive insulin production. These observations suggested the hypothesis that FSTL3 neutralization could be used to therapeutically enhance beta cell number and function in humans. To pursue this possibility, we developed an FSTL3-neutralizing antibody, FP-101, and characterized its ability to prevent or disrupt FSTL3 from complexing with activin or related ligands. This antibody was selective for FSTL3 relative to the closely related follistatin, thereby reducing the chance for off-target effects. In vitro assays with FP-101 and activin revealed that FP-101-mediated neutralization of FSTL3 can enhance both insulin secretion and glucose responsiveness to nonfunctional mouse and human islets under conditions that model diabetes. Thus, FSTL3 neutralization may provide a novel therapeutic strategy for treating diabetes through repairing dysfunctional beta cells.


Assuntos
Anticorpos Neutralizantes/química , Proteínas Relacionadas à Folistatina/genética , Folistatina/química , Deleção de Genes , Células Secretoras de Insulina/metabolismo , Ativinas/química , Animais , Glicemia/análise , Glucose/metabolismo , Células HEK293 , Humanos , Secreção de Insulina , Células Secretoras de Insulina/citologia , Ilhotas Pancreáticas/citologia , Ligantes , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos C57BL
3.
Bioorg Med Chem Lett ; 30(3): 126892, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31874826

RESUMO

Follistatin is well known as an inhibitor of transforming growth factor (TGF)-ß superfamily ligands including myostatin and activin A. Myostatin, a negative regulator of muscle growth, is a promising target with which to treat muscle atrophic diseases. Here, we focused on the N-terminal domain (ND) of follistatin (Fst) that interacts with the type I receptor binding site of myostatin. Through bioassay of synthetic ND-derived fragment peptides, we identified DF-3, a new myostatin inhibitory 14-mer peptide which effectively inhibits myostatin, but fails to inhibit activin A or TGF-ß1, in an in vitro luciferase reporter assay. Injected intramuscularly, DF-3 significantly increases skeletal muscle mass in mice and consequently, it can serve as a platform for development of muscle enhancement based on myostatin inhibition.


Assuntos
Folistatina/química , Miostatina/antagonistas & inibidores , Peptídeos/química , Ativinas/antagonistas & inibidores , Ativinas/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/crescimento & desenvolvimento , Miostatina/metabolismo , Peptídeos/metabolismo , Peptídeos/farmacologia , Ligação Proteica , Relação Estrutura-Atividade , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/metabolismo
4.
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
5.
Artigo em Inglês | MEDLINE | ID: mdl-29496578

RESUMO

Follistatin (FST) is a single-chain gonadal protein involving in various biological effects. FST plays important roles in not only ovary development but also body growth, whereas myostatin (MSTN) negatively regulates muscle growth. In this study, FST gene in bighead carp (HynFST) was cloned and characterized. A 5797 bp genomic sequence of HynFST, consisting six exons and five introns were cloned. The full-length cDNA of HynFST (2134 bp) has an open reading fragment encoding a polypeptide of 349 amino acids. Sequence comparison and phylogenetic analysis confirmed that FSTs are conserved throughout the vertebrates and HynFST belongs to FST-1 isoform. Nine single nucleotide polymorphisms (SNPs) of the HynFST were identified and three of them (g.2443 T > C, g.2852 T > C and g.5483A > G) were significantly associated with four growth-related traits. The average body weight of those fish with the combined genotype (CC CC GG) was 12.15-22.63% higher than that of triplotype (TT TT AA) in two bighead carp populations. HynFST was expressed in most of the development stages and various tissues with highest level in ovary. The co-expression results for FST and MSTN in brain and muscle of divergent weight groups showed that FST may inhibit MSTN expression, thus enhancing growth in bighead carp. Our results suggest that FST has significant genetic effects on the regulation of early growth in bighead carp. This study would facilitate the elucidation of multiple functions of FST gene in fish and exploration of the potentials as a gene marker in selective breeding programs for growth of bighead carp.


Assuntos
Cyprinidae/crescimento & desenvolvimento , Cyprinidae/genética , Proteínas de Peixes/genética , Folistatina/genética , Regulação da Expressão Gênica no Desenvolvimento , Sequência de Aminoácidos , Animais , Peso Corporal , Clonagem Molecular , Éxons/genética , Feminino , Proteínas de Peixes/química , Folistatina/química , Íntrons/genética , Masculino , Filogenia , Polimorfismo de Nucleotídeo Único , Razão de Masculinidade
6.
Exp Mol Med ; 49(9): e377, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28912572

RESUMO

Delivery of follistatin (FST) represents a promising strategy for both muscular dystrophies and diabetes, as FST is a robust antagonist of myostatin and activin, which are critical regulators of skeletal muscle and adipose tissues. FST is a multi-domain protein, and deciphering the function of different domains will facilitate novel designs for FST-based therapy. Our study aims to investigate the role of the N-terminal domain (ND) of FST in regulating muscle and fat mass in vivo. Different FST constructs were created and packaged into the adeno-associated viral vector (AAV). Overexpression of wild-type FST in normal mice greatly increased muscle mass while decreasing fat accumulation, whereas overexpression of an N terminus mutant or N terminus-deleted FST had no effect on muscle mass but moderately decreased fat mass. In contrast, FST-I-I containing the complete N terminus and double domain I without domain II and III had no effect on fat but increased skeletal muscle mass. The effects of different constructs on differentiated C2C12 myotubes were consistent with the in vivo finding. We hypothesized that ND was critical for myostatin blockade, mediating the increase in muscle mass, and was less pivotal for activin binding, which accounts for the decrease in the fat tissue. An in vitro TGF-beta1-responsive reporter assay revealed that FST-I-I and N terminus-mutated or -deleted FST showed differential responses to blockade of activin and myostatin. Our study provided direct in vivo evidence for a role of the ND of FST, shedding light on future potential molecular designs for FST-based gene therapy.


Assuntos
Tecido Adiposo/anatomia & histologia , Tecido Adiposo/metabolismo , Folistatina/metabolismo , Músculo Esquelético/anatomia & histologia , Músculo Esquelético/metabolismo , Domínios e Motivos de Interação entre Proteínas , Animais , Biomarcadores , Diferenciação Celular/genética , Linhagem Celular , Dependovirus/genética , Feminino , Imunofluorescência , Folistatina/química , Folistatina/genética , Expressão Gênica , Ordem dos Genes , Genes Reporter , Vetores Genéticos/genética , Humanos , Camundongos , Mutação , Mioblastos/citologia , Mioblastos/metabolismo , Tamanho do Órgão , Regiões Promotoras Genéticas , Domínios e Motivos de Interação entre Proteínas/genética , Transdução de Sinais
7.
Mol Reprod Dev ; 84(7): 626-637, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28500669

RESUMO

The hypothesis that, in contrast to other transforming growth factor-beta (TGFß) superfamily ligands, the dose-response curve of Anti-Müllerian hormone (AMH) is unmodulated was tested by examining whether known TGFB superfamily modulators affect AMH signaling, using a P19/BRE luciferase reporter assay. AMHC and AMHN,C activated the reporter with an EC50 of approximately 0.5 nM. Follistatins (FS) produced concentration-dependent increases in AMHC - and AMHN,C -initiated reporter activity, with FS288 being more potent than FS315; however, the maximum bioactivity of AMH was not altered by either follistatin. Thirteen other TGFß regulators (Chordin, Chordin-like 1, Chordin-like 2, Differential screening-selected gene aberrative in neuroblastoma [DAN], Decorin, Endoglin, Follistatin-like 1, Follistatin-like 3, Follistatin-like 4, Noggin, α2 macroglobulin, TGFß receptor 3, Von Willebrand factor C domain-containing 2) had little or no effect. Surface plasmon resonance analysis showed no significant association between FS288 and AMHC , suggesting that FS288 indirectly regulates AMH signaling. Activin A, a direct target of FS288, did not itself induce reporter activity in P19 cells, but did prevent the FS288-induced increase in AMH signaling. Hence, local concentrations of FS288 and Activin A may influence the response of some cell types to AMH.


Assuntos
Hormônio Antimülleriano/química , Folistatina/química , Transdução de Sinais , Ressonância de Plasmônio de Superfície , Animais , Hormônio Antimülleriano/genética , Linhagem Celular , Folistatina/genética , Folistatina/metabolismo , Humanos , Camundongos
8.
Cell Rep ; 16(8): 2077-2086, 2016 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-27524626

RESUMO

The DAN family, including Gremlin-1 and Gremlin-2 (Grem1 and Grem2), represents a large family of secreted BMP (bone morphogenetic protein) antagonists. However, how DAN proteins specifically inhibit BMP signaling has remained elusive. Here, we report the structure of Grem2 bound to GDF5 at 2.9-Å resolution. The structure reveals two Grem2 dimers binding perpendicularly to each GDF5 monomer, resembling an H-like structure. Comparison to the unbound Grem2 structure reveals a dynamic N terminus that undergoes significant transition upon complex formation, leading to simultaneous interaction with the type I and type II receptor motifs on GDF5. Binding studies show that DAN-family members can interact with BMP-type I receptor complexes, whereas Noggin outcompetes the type I receptor for ligand binding. Interestingly, Grem2-GDF5 forms a stable aggregate-like structure in vitro that is not clearly observed for other antagonists, including Noggin and Follistatin. These findings exemplify the structural and functional diversity across the various BMP antagonist families.


Assuntos
Proteína Morfogenética Óssea 2/química , Fator 5 de Diferenciação de Crescimento/química , Peptídeos e Proteínas de Sinalização Intercelular/química , Osteoblastos/metabolismo , Proteínas/química , Motivos de Aminoácidos , Animais , Sítios de Ligação , Ligação Competitiva , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular , Cristalografia por Raios X , Citocinas , Embrião não Mamífero , Folistatina/química , Folistatina/genética , Folistatina/metabolismo , Expressão Gênica , Fator 5 de Diferenciação de Crescimento/genética , Fator 5 de Diferenciação de Crescimento/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Modelos Moleculares , Osteoblastos/citologia , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Proteínas/genética , Proteínas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Xenopus laevis
9.
Hum Reprod Update ; 22(3): 342-57, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26884470

RESUMO

BACKGROUND: Activins are members of the pleiotrophic family of the transforming growth factor-beta (TGF-ß) superfamily of cytokines, initially isolated for their capacity to induce the release of FSH from pituitary extracts. Subsequent research has demonstrated that activins are involved in multiple biological functions including the control of inflammation, fibrosis, developmental biology and tumourigenesis. This review summarizes the current knowledge on the roles of activin in reproductive and developmental biology. It also discusses interesting advances in the field of modulating the bioactivity of activins as a therapeutic target, which would undoubtedly be beneficial for patients with reproductive pathology. METHODS: A comprehensive literature search was carried out using PUBMED and Google Scholar databases to identify studies in the English language which have contributed to the advancement of the field of activin biology, since its initial isolation in 1987 until July 2015. 'Activin', 'testis', 'ovary', 'embryonic development' and 'therapeutic targets' were used as the keywords in combination with other search phrases relevant to the topic of activin biology. RESULTS: Activins, which are dimers of inhibin ß subunits, act via a classical TGF-ß signalling pathway. The bioactivity of activin is regulated by two endogenous inhibitors, inhibin and follistatin. Activin is a major regulator of testicular and ovarian development. In the ovary, activin A promotes oocyte maturation and regulates granulosa cell steroidogenesis. It is also essential in endometrial repair following menstruation, decidualization and maintaining pregnancy. Dysregulation of the activin-follistatin-inhibin system leads to disorders of female reproduction and pregnancy, including polycystic ovary syndrome, ectopic pregnancy, miscarriage, fetal growth restriction, gestational diabetes, pre-eclampsia and pre-term birth. Moreover, a rise in serum activin A, accompanied by elevated FSH, is characteristic of female reproductive aging. In the male, activin A is an autocrine and paracrine modulator of germ cell development and Sertoli cell proliferation. Disruption of normal activin signalling is characteristic of many tumours affecting reproductive organs, including endometrial carcinoma, cervical cancer, testicular and ovarian cancer as well as prostate cancer. While activin A and B aid the progression of many tumours of the reproductive organs, activin C acts as a tumour suppressor. Activins are important in embryonic induction, morphogenesis of branched glandular organs, development of limbs and nervous system, craniofacial and dental development and morphogenesis of the Wolffian duct. CONCLUSIONS: The field of activin biology has advanced considerably since its initial discovery as an FSH stimulating agent. Now, activin is well known as a growth factor and cytokine that regulates many aspects of reproductive biology, developmental biology and also inflammation and immunological mechanisms. Current research provides evidence for novel roles of activins in maintaining the structure and function of reproductive and other organ systems. The fact that activin A is elevated both locally as well as systemically in major disorders of the reproductive system makes it an important biomarker. Given the established role of activin A as a pro-inflammatory and pro-fibrotic agent, studies of its involvement in disorders of reproduction resulting from these processes should be examined. Follistatin, as a key regulator of the biological actions of activin, should be evaluated as a therapeutic agent in conditions where activin A overexpression is established as a contributing factor.


Assuntos
Ativinas/fisiologia , Ovário/fisiologia , Reprodução/fisiologia , Testículo/fisiologia , Ativinas/química , Animais , Feminino , Folistatina/química , Folistatina/fisiologia , Glicoproteínas , Humanos , Subunidades beta de Inibinas , Inibinas/química , Inibinas/fisiologia , Masculino , Neoplasias Ovarianas , Pré-Eclâmpsia , Gravidez
10.
Drug Metab Dispos ; 43(12): 1882-90, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26354950

RESUMO

Follistatin 315 heparan sulfate-binding deficient mutant human IgG4 Fc fusion (FST-ΔHBS-Fc) is a follistatin (FST) based Fc fusion protein currently being developed as a novel therapy for several potential indications, including muscle wasting. Previous assessments of the pharmacokinetics and therapeutic activity of FST-ΔHBS-Fc have shown a close association of the exposure-response relationship. The current work builds upon these initial studies by investigating the glycosylation characteristics of FST-ΔHBS-Fc after recombinant expression and its impact on the pharmacokinetics in mice and Cynomolgus monkeys. The data presented indicate that FST-ΔHBS-Fc is heterogeneously glycosylated at the three putative sites in FST when recombinantly expressed in stably transfected Chinese hamster ovary cells. Such carbohydrate heterogeneity, especially with regards to sialic acid incorporation, directly results in sugar-dependent clearance in both mice and Cynomolgus monkeys. Examination of the pharmacokinetics of FST-ΔHBS-Fc molecules containing variable sialic acid content in asialoglycoprotein receptor 1 (ASPGR-1) knockout mice supports the receptor's role as part of the clearance mechanism of the molecules. Based on the evaluation of several variably sialylated lots of material in pharmacokinetic assessments, we define specifications for average sialic acid incorporation into FST-ΔHBS-Fc that result in limited sugar-mediated clearance. Taken together, these studies highlight the importance of establishing an early understanding of the glycosylation/pharmacokinetic relationships of FST-ΔHBS-Fc, which will provide a basis for future application toward optimal systemic drug delivery and dosing strategies.


Assuntos
Terapia Biológica/tendências , Folistatina/química , Folistatina/farmacocinética , Animais , Células CHO , Cricetinae , Cricetulus , Glicosilação , Células HEK293 , Humanos , Macaca fascicularis , Masculino , Camundongos , Camundongos Knockout , Camundongos SCID
11.
Appl Microbiol Biotechnol ; 98(24): 10041-51, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25411099

RESUMO

Follistatin (FST) binds to myostatin (MSTN), a potent negative regulator of skeletal muscle growth. Inhibition of MSTN activity by FST treatment has shown to enhance muscle growth as well as ameliorate symptoms of muscular dystrophy in animal models, illustrating the potential of FST as an agent to enhance muscle growth in animal agriculture or to treat muscle wasting conditions or disease in humans. Therefore, we designed a study to produce biologically active recombinant chicken FST315 (chFST315) in an Escherichia coli host. Since FST contains multiple intramolecular disulfide bonds, we expressed chFST315 protein in either a system that utilizes a periplasmic expression strategy, or a genetically modified E. coli system (SHuffle strain) that is capable of disulfide bond formation in the cytoplasm. Periplasmic expression of chFST315 using the pMAL-p5x vector system, which was designed to express maltose-binding protein (MBP) fusion protein, failed to produce a soluble recombinant protein. However, cytoplasmic expression of chFST315 using pMAL-c5x vector in SHuffle E. coli strain resulted in a soluble expression of the recombinant protein (MBP-chFST315). Combination of heparin and amylose resin affinity chromatography yielded about 6 mg/L purified MBP-chFST315. The purified MBP-chFST315 showed binding affinity to MSTN and activin in a pull-down assay, as well as inhibited MSTN and activin activity in an in vitro reporter gene assay. In conclusion, results of the study demonstrate that for the first time a recombinant, biologically active FST molecule can be produced in a soluble form in E. coli. The ability to produce FST in a cost-effective system is expected to allow us to investigate the potentials of FST as an agent to improve skeletal muscle growth of meat producing animals via suppression of MSTN.


Assuntos
Escherichia coli/genética , Escherichia coli/metabolismo , Folistatina/genética , Folistatina/metabolismo , Animais , Galinhas , Cromatografia de Afinidade , Citoplasma/metabolismo , Folistatina/química , Folistatina/isolamento & purificação , Dados de Sequência Molecular , Miostatina/antagonistas & inibidores , Miostatina/metabolismo , Periplasma/metabolismo , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Análise de Sequência de DNA , Solubilidade
12.
J Anim Physiol Anim Nutr (Berl) ; 98(3): 522-9, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23957442

RESUMO

Follistatin (FST) acts as a positive regulator of muscle development by inhibiting the activities and expression of myostatin. The recombinant duck FST protein was injected into hatching eggs and was also added to the medium of duck myoblast to study its role on duck embryonic muscle development and gene expressions. Duck embryo weight increased 3.49% (p > 0.05) in FST treatment group as compared with control group, but minor effects were found on leg or breast muscle weights of ducklings at 2 days post-hatching (p > 0.05). Relative expression of Pax7 was upregulated in both leg and breast muscle tissues (p < 0.05), while MyoD was only upregulated in leg muscle (p < 0.05), and Myf5 was only upregulated in breast muscle (p < 0.05). Relative expression of myostatin was downregulated in both muscle tissues researched (p < 0.05). In vitro studies also showed some maker genes relevant to protein synthesis and degradation, cells' proliferation and differentiation had significant changes in myoblasts after treated with FST. These results suggested that in ovo feeding of recombinant FST protein to duck hatching eggs had an effect on duck embryo development but have less roles on the duck embryonic muscle development.


Assuntos
Patos/embriologia , Patos/metabolismo , Folistatina/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Músculo Esquelético/embriologia , Proteínas Recombinantes , Animais , Folistatina/química , Músculo Esquelético/crescimento & desenvolvimento , Mioblastos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
13.
Vaccine ; 32(1): 11-8, 2013 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-24211168

RESUMO

Inhibin and follistatin are known to reduce fecundity by inhibiting the actions of activin and FSH. Thus, the immunoneutralization of these hormones is a rational proposal for augmenting reproductive performance. The present study describes a comprehensive computational methodology comprising of a consensus approach of several B- and Th-cell epitope prediction tools for the identification of epitopic regions within the structure of these hormones that can be incorporated into a poly-epitope fecundity vaccine. The proposed peptide (RGD-WSPAALRLLQRPPEEPA-KK-YSFPISSILE) should be effective in multiple animal species, generating good immunological memory.


Assuntos
Epitopos/imunologia , Fertilidade/imunologia , Gado/imunologia , Vacinas/imunologia , Sequência de Aminoácidos , Animais , Bovinos , Galinhas , Mapeamento de Epitopos/métodos , Epitopos/química , Epitopos de Linfócito B/química , Epitopos de Linfócito B/imunologia , Epitopos de Linfócito T/química , Epitopos de Linfócito T/imunologia , Folistatina/química , Folistatina/imunologia , Cavalos , Inibinas/química , Inibinas/imunologia , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/imunologia , Conformação Proteica , Ratos , Ovinos , Sus scrofa , Vacinas de Subunidades Antigênicas/química , Vacinas de Subunidades Antigênicas/imunologia
14.
Reprod Fertil Dev ; 25(3): 570-80, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23164397

RESUMO

Activin A and its inhibitors follistatin and inhibin play key roles in development and function of the male reproductive tract. Quantitative (q) polymerase chain reaction (PCR) was used to evaluate the expression of Inhba (the gene encoding activin A subunits), Inha and Inhbb (genes encoding the inhibin B subunits), as well as the genes for follistatin (Fst) and follistatin-like 3 (Fstl3) and the activin receptor subunits, in the male mouse reproductive tract. A qPCR assay that discriminated between the two follistatin variants of Fst288 (tissue-bound form) and Fst315 (circulating) was established. Activin A protein was measured by ELISA, whereas the inhibin α-subunit and total follistatin proteins were measured by radioimmunoassay (RIA). A screen of 22 tissues demonstrated tissue-specific regulation of the follistatin variants, with Fst288 highly expressed in the vas deferens and Fst315 most highly expressed in the skin. The expression of Fst288 and Fst315 and follistatin protein levels increased progressively from the testis through to the distal vas deferens. Inhba and the activin receptors were highly expressed in the epididymis, but activin A protein was elevated in both the epididymis and vas deferens. Inhibin α-subunit mRNA and protein and Inhbb expression were highest in the testis. These results indicate a role for activin A within the epididymis, but also that activin A bioactivity may be increasingly inhibited by follistatin distally along the male reproductive tract.


Assuntos
Ativinas/metabolismo , Epididimo/metabolismo , Folistatina/metabolismo , Regulação da Expressão Gênica , Inibinas/metabolismo , Ducto Deferente/metabolismo , Regiões 3' não Traduzidas , Receptores de Ativinas/genética , Receptores de Ativinas/metabolismo , Ativinas/genética , Animais , Sequência de Bases , Éxons , Folistatina/química , Folistatina/genética , Genitália Masculina/metabolismo , Inibinas/genética , Masculino , Camundongos , Camundongos Endogâmicos , Dados de Sequência Molecular , Especificidade de Órgãos , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo
15.
Biochemistry ; 51(34): 6797-803, 2012 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-22809401

RESUMO

Heparin and related heparan sulfate interact with a number of cytokines and growth factors, thereby playing an essential role in many physiological and pathophysiological processes by involving both signal transduction and the regulation of the tissue distribution of cytokines/growth factors. Follistatin (FS) is an autocrine protein with a heparin-binding motif that serves to regulate the cell proliferative activity of the paracrine hormone, and member of the TGF-ß family, activin A (ActA). Follistatin is currently under investigation as an antagonist of another TGF-ß family member, myostatin (Mstn), for the promotion of muscle growth in diseases associated with muscle atrophy. In this study, we employ surface plasmon resonance (SPR) spectroscopy to dissect the binding interactions between the heparin polysaccharide and both free follistatin (FS288) and its complexes (FS288-ActA and FS288-Mstn). FS288 complexes show much higher heparin binding affinity than FS288 alone. SPR solution competition studies using heparin oligosaccharides showed that the binding of FS288 and its complex to heparin is dependent on chain length. Full chain heparin or large oligosaccharides, having 18-20 sugar residues, show the highest binding activity for FS288 and the FS288-ActA complex, whereas smaller heparin molecules could interact with the FS288-Mstn complex. These interactions were also analyzed in normal physiological buffers and at different salt concentrations and pH values. Unbound follistatin was much more sensitive to all salt concentrations of >150 mM. The binding of heparin to the FS288-ActA complex was disrupted at 500 mM salt, whereas it was actually strengthened for the FS288-Mstn complex. At acidic pH values, binding of heparin to FS288 and the FS288-ActA complex was enhanced. While slightly acidic pH values (pH 6.2 and 5.2) enhanced the binding of the FS288-Mstn complex to heparin, at pH 4 heparin binding was inhibited. Overall, these studies demonstrate that binding of a specific ligand to FS288 differentially regulates its affinity and behavior for heparin molecules.


Assuntos
Folistatina/metabolismo , Heparina/metabolismo , Ressonância de Plasmônio de Superfície/métodos , Ativinas/química , Ativinas/metabolismo , Folistatina/química , Heparina/química , Humanos , Concentração de Íons de Hidrogênio , Cinética , Ligantes , Miostatina/química , Miostatina/metabolismo , Ligação Proteica
16.
Mol Endocrinol ; 26(7): 1167-78, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22593183

RESUMO

Follistatin (FST)-type proteins are important antagonists of some members of the large TGF-ß family of cytokines. These include myostatin, an important negative regulator of muscle growth, and the closely related activin A, which is involved in many physiological functions, including maintenance of a normal reproductive axis. FST-type proteins, including FST and FST-like 3 (FSTL3), differentially inhibit various TGF-ß family ligands by binding each ligand with two FST-type molecules. In this study, we sought to examine features that are important for ligand antagonism by FST-type proteins. Previous work has shown that a modified construct consisting of the FST N-terminal domain (ND) followed by two repeating follistatin domains (FSD), herein called FST ND-FSD1-FSD1, exhibits strong specificity for myostatin over activin A. Using cell-based assays, we show that FST ND-FSD1-FSD1 is unique in its specificity for myostatin as compared with similar constructs containing domains from FSTL3 and that the ND is critical to its activity. Furthermore, we demonstrate that FSD3 of FST provides affinity to ligand inhibition and confers resistance to perturbations in the ND and FSD2, likely through the interaction of FSD3 of one FST molecule with the ND of the other FST molecule. Additionally, our data suggest that this contact provides cooperativity to ligand antagonism. Cross-linking studies show that this interaction also potentiates formation of 1:2 ligand-FST complexes, whereas lack of FSD3 allows formation of 1:1 complexes. Altogether, these studies support that domain differences generate FST-type molecules that are each uniquely suited ligand antagonists.


Assuntos
Ativinas/química , Proteínas Relacionadas à Folistatina/química , Folistatina/química , Miostatina/química , Ativinas/antagonistas & inibidores , Animais , Linhagem Celular , Folistatina/metabolismo , Proteínas Relacionadas à Folistatina/genética , Proteínas Relacionadas à Folistatina/metabolismo , Células HEK293 , Humanos , Camundongos , Miostatina/antagonistas & inibidores , Ligação Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína
17.
Reprod Biomed Online ; 24(3): 364-74, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22285243

RESUMO

Follistatin, an inhibitor of activin A, has key regulatory roles in the female reproductive tract. Follistatin has two splice variants: FST288, largely associated with cell surfaces, and FST315, the predominant circulating form. The mechanism regulating uterine expression of these variants is unknown. Quantitative RT-PCR was used to measure expression of follistatin splice variants (Fst288, Fst315), the activin bA subunit (Inhba) and the inhibin a subunit (Inha) in uterine tissues during early pregnancy (days 1­4, preimplantation) and in response to exogenous 17b-oestradiol (single s.c. injection) and progesterone (three daily s.c. injections) in ovariectomized mice. Uterine Fst288, Fst315 and Inhba expression increased during early pregnancy, with greater increases in Fst315 relative to Fst288 suggesting differential regulation of these variants. Fst288, Fst315, Inhba and Inha all increased in response to progesterone treatment. Fst288, but not Fst315, mRNA decreased in response to 17b-oestradiol treatment, whereas Inhba increased. A comparison of the absolute concentrations of uterine follistatin mRNA using crossing thresholds indicated that both variants were more highly expressed in early pregnancy in contrast to the hormone treatment models. It is concluded that progesterone regulates uterine expression of both follistatin variants, as well as activin A, during early pregnancy in the mouse uterus


Assuntos
Folistatina/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Progesterona/farmacologia , Útero/efeitos dos fármacos , Animais , Estradiol/farmacologia , Feminino , Folistatina/química , Folistatina/genética , Subunidades beta de Inibinas/genética , Subunidades beta de Inibinas/metabolismo , Inibinas/genética , Inibinas/metabolismo , Camundongos , Gravidez , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Útero/metabolismo
18.
Am J Pathol ; 179(2): 915-30, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21689628

RESUMO

Recovery from skeletal muscle injury is often incomplete because of the formation of fibrosis and inadequate myofiber regeneration; therefore, injured muscle could benefit significantly from therapies that both stimulate muscle regeneration and inhibit fibrosis. To this end, we focused on blocking myostatin, a member of the transforming growth factor-ß superfamily and a negative regulator of muscle regeneration, with the myostatin antagonist follistatin. In vivo, follistatin-overexpressing transgenic mice underwent significantly greater myofiber regeneration and had less fibrosis formation compared with wild-type mice after skeletal muscle injury. Follistatin's mode of action is likely due to its ability to block myostatin and enhance neovacularization. Furthermore, muscle progenitor cells isolated from follistatin-overexpressing mice were significantly superior to muscle progenitors isolated from wild-type mice at regenerating dystrophin-positive myofibers when transplanted into the skeletal muscle of dystrophic mdx/severe combined immunodeficiency mice. In vitro, follistatin stimulated myoblasts to express MyoD, Myf5, and myogenin, which are myogenic transcription factors that promote myogenic differentiation. Moreover, follistatin's ability to enhance muscle differentiation is at least partially due to its ability to block myostatin, activin A, and transforming growth factor-ß1, all of which are negative regulators of muscle cell differentiation. The findings of this study suggest that follistatin is a promising agent for improving skeletal muscle healing after injury and muscle diseases, such as the muscular dystrophies.


Assuntos
Fibrose/patologia , Folistatina/química , Músculo Esquelético/metabolismo , Animais , Linhagem Celular , Transplante de Células , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência/métodos , Proteína MyoD/metabolismo , Fator Regulador Miogênico 5/metabolismo , Miostatina/metabolismo , Neovascularização Patológica , Regeneração , Fator de Crescimento Transformador beta/metabolismo
19.
J Biol Chem ; 285(5): 3114-25, 2010 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-19940118

RESUMO

Clustering or overexpression of the transmembrane form of the extracellular matrix proteoglycan agrin in neurons results in the formation of numerous highly motile filopodia-like processes extending from axons and dendrites. Here we show that similar processes can be induced by overexpression of transmembrane-agrin in several non-neuronal cell lines. Mapping of the process-inducing activity in neurons and non-neuronal cells demonstrates that the cytoplasmic part of transmembrane agrin is dispensable and that the extracellular region is necessary for process formation. Site-directed mutagenesis reveals an essential role for the loop between beta-sheets 3 and 4 within the Kazal subdomain of the seventh follistatin-like domain of TM-agrin. An aspartic acid residue within this loop is critical for process formation. The seventh follistatin-like domain could be functionally replaced by the first and sixth but not by the eighth follistatin-like domain, demonstrating a functional redundancy among some follistatin-like domains of agrin. Moreover, a critical distance of the seventh follistatin-like domain to the plasma membrane appears to be required for process formation. These results demonstrate that different regions within the agrin protein are responsible for synapse formation at the neuromuscular junction and for process formation in central nervous system neurons and suggest a role for agrin's follistatin-like domains in the developing central nervous system.


Assuntos
Agrina/química , Folistatina/química , Agrina/metabolismo , Animais , Células COS , Membrana Celular/metabolismo , Sistema Nervoso Central/metabolismo , Galinhas , Chlorocebus aethiops , Feminino , Humanos , Mutagênese Sítio-Dirigida , Neurônios/metabolismo , Células PC12 , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ratos
20.
EMBO J ; 28(17): 2662-76, 2009 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-19644449

RESUMO

Myostatin is a member of the transforming growth factor-beta (TGF-beta) family and a strong negative regulator of muscle growth. Here, we present the crystal structure of myostatin in complex with the antagonist follistatin 288 (Fst288). We find that the prehelix region of myostatin very closely resembles that of TGF-beta class members and that this region alone can be swapped into activin A to confer signalling through the non-canonical type I receptor Alk5. Furthermore, the N-terminal domain of Fst288 undergoes conformational rearrangements to bind myostatin and likely acts as a site of specificity for the antagonist. In addition, a unique continuous electropositive surface is created when myostatin binds Fst288, which significantly increases the affinity for heparin. This translates into stronger interactions with the cell surface and enhanced myostatin degradation in the presence of either Fst288 or Fst315. Overall, we have identified several characteristics unique to myostatin that will be paramount to the rational design of myostatin inhibitors that could be used in the treatment of muscle-wasting disorders.


Assuntos
Folistatina/química , Heparina/metabolismo , Miostatina/química , Ativinas , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células CHO , Cricetinae , Cricetulus , Heparina/química , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...