Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
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.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
Muscle Nerve ; 39(3): 283-96, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19208403

RESUMO

In most cases, pharmacologic strategies to treat genetic muscle disorders and certain acquired disorders, such as sporadic inclusion body myositis, have produced modest clinical benefits. In these conditions, inhibition of the myostatin pathway represents an alternative strategy to improve functional outcomes. Preclinical data that support this approach clearly demonstrate the potential for blocking the myostatin pathway. Follistatin has emerged as a powerful antagonist of myostatin that can increase muscle mass and strength. Follistatin was first isolated from the ovary and is known to suppress follicle-stimulating hormone. This raises concerns for potential adverse effects on the hypothalamic-pituitary-gonadal axis and possible reproductive capabilities. In this review we demonstrate a strategy to bypass off-target effects using an alternatively spliced cDNA of follistatin (FS344) delivered by adeno-associated virus (AAV) to muscle. The transgene product is a peptide of 315 amino acids that is secreted from the muscle and circulates in the serum, thus avoiding cell-surface binding sites. Using this approach our translational studies show increased muscle size and strength in species ranging from mice to monkeys. Adverse effects are avoided, and no organ system pathology or change in reproductive capabilities has been seen. These findings provide the impetus to move toward gene therapy clinical trials with delivery of AAV-FS344 to increase size and function of muscle in patients with neuromuscular disease.


Assuntos
Folistatina/farmacologia , Folistatina/uso terapêutico , Terapia Genética/métodos , Doenças Musculares/terapia , Miostatina/antagonistas & inibidores , Processamento Alternativo , Animais , DNA Complementar/administração & dosagem , Dependovirus/genética , Folistatina/química , Folistatina/genética , Humanos , Camundongos , Camundongos Mutantes , Músculos/efeitos dos fármacos , Músculos/patologia , Músculos/fisiologia , Doenças Musculares/genética , Doenças Musculares/patologia , Miostatina/metabolismo
12.
J Biol Chem ; 282(52): 37378-88, 2007 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-17942404

RESUMO

The transmembrane protein with epidermal growth factor and two follistatin motifs 2 (TMEFF2) is expressed in prostate and brain and shed from the cell surface in a metalloproteinase-dependent fashion. Neither the sheddase(s) responsible for TMEFF2 shedding nor the physiological significance or activity of the soluble TMEFF2 ectodomain (TMEFF2-ECD) has been identified. In the present study we present new evidence that a disintegrin and metalloproteinase-17 (ADAM17) is responsible for phorbol 12-myristate 13-acetate-induced release of TMEFF2-ECD using small interfering RNA to ablate ADAM17 expression or by inhibiting enzymatic activity. A single well shedding assay monitoring the release of alkaline phosphatase-tagged TMEFF2-ECD into medium and the generation of 22- and 14-kDa C-terminal fragments in lysates were dependent on ADAM17 activity. A gamma-secretase inhibitor prevented the formation of a 10-kDa fragment in cell lysates, thus establishing TMEFF2 as a novel substrate for regulated intramembrane proteolysis. We assigned proliferation-inducing activity to TMEFF2. Inhibition of TMEFF2 shedding using synthetic metalloproteinase inhibitors or small interfering RNA targeting TMEFF2 expression yielded a statistically significant reduction of cell proliferation in the lymph node-derived prostate cancer cells (LNCaPs) and a human embryonic kidney (HEK293) cell line overexpressing TMEFF2. The TMEFF2-ECD was able to induce ERK1/2 phosphorylation in an epidermal growth factor receptor (or ErbB1)-dependent manner in HEK293 cells. Our data suggest that TMEFF2 contributes to cell proliferation in an ADAM17-dependent autocrine fashion in cells expressing this protein.


Assuntos
Proteínas ADAM/química , Membrana Celular/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Folistatina/química , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Ésteres de Forbol/farmacologia , Neoplasias da Próstata/metabolismo , Proteína ADAM17 , Motivos de Aminoácidos , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Masculino , Modelos Biológicos , Conformação Proteica , Estrutura Terciária de Proteína , RNA Interferente Pequeno/metabolismo
13.
Eur J Oral Sci ; 114 Suppl 1: 78-85; discussion 93-5, 379-80, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16674666

RESUMO

Genes encoding the major enamel matrix proteins and non-collagenous proteins of bone and dentin are members of the secretory calcium-binding phosphoprotein (SCPP) family, which originated from ancestral SPARC (secreted protein, acidic and rich in cysteine; BM-40/osteonectin). To better understand the role of SPARC in mineralizing systems, we isolated SPARC from developing pig teeth, deduced its primary structure from the cDNA sequence, and determined its quaternary structure by homology modelling with reference to human SPARC crystal structures. The guanidine/EDTA extract from porcine dentin was fractionated by anion-exchange and size-exclusion chromatography. Stains-all positive bands at 38 and 35 kDa gave the N-terminal sequences APQQEALPDETEV and DFEKNYNMYIFPV, which corresponded to the SPARC N terminus and an internal region of the protein. Porcine SPARC contains 300 amino acids, including the 17-amino acid signal peptide, and shares 96.2% amino acid sequence identity with human SPARC. Without post-translational modifications, the 283-amino acid secreted protein has a molecular mass of 32.3 kDa. The three-dimensional model revealed that porcine SPARC contains a single N-linked glycosylation at N113, seven intramolecular disulfide bridges, and assembles into dimers. SPARC is composed of three structural/functional domains: an acidic Ca2+-binding, a follistatin-like, and an extracellular calcium-binding domain.


Assuntos
Dentina/química , Osteonectina/isolamento & purificação , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteínas de Ligação ao Cálcio/química , Simulação por Computador , DNA Complementar/genética , Proteínas do Esmalte Dentário/genética , Proteínas do Esmalte Dentário/isolamento & purificação , Dissulfetos/química , Folistatina/química , Glicosilação , Humanos , Imageamento Tridimensional , Modelos Químicos , Modelos Moleculares , Dados de Sequência Molecular , Odontogênese/fisiologia , Sinais Direcionadores de Proteínas/genética , Estrutura Quaternária de Proteína , Homologia de Sequência de Aminoácidos , Suínos
15.
Endocrinology ; 147(7): 3586-97, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16627583

RESUMO

Follistatin (FST) and FST-like-3 (FSTL3) are activin-binding and neutralization proteins that also bind myostatin. Three FST isoforms have been described that differ in tissue distribution and cell-surface binding activity, suggesting that the FST isoforms and FSTL3 may have some nonoverlapping biological actions. We produced recombinant FST isoforms and FSTL3 and compared their biochemical and biological properties. Activin-binding affinities and kinetics were comparable between the isoforms and FSTL3, whereas cell-surface binding differed markedly (FST288 > FST303 > FST315 > FSTL3). Inhibition of endogenous activin bioactivity, whether the FST isoforms were administered endogenously or exogenously, correlated closely with surface binding activity, whereas neutralization of exogenous activin when FST and FSTL3 were also exogenous was consistent with their equivalent activin-binding affinities. This difference in activin inhibition was also evident in an in vitro bioassay because FST288 suppressed, whereas FST315 enhanced, activin-dependent TT cell proliferation. Moreover, when FSTL3, which does not associate with cell membranes, was expressed as a membrane-anchored protein, its endogenous activin inhibitory activity was dramatically increased. In competitive binding assays, myostatin was more potent than bone morphogenetic proteins (BMPs) 6 and 7, and BMPs 2 and 4 were inactive in binding to FST isoforms, whereas none of the BMPs tested competed with activin for binding to FSTL3. Neutralization of exogenous BMP or myostatin bioactivity correlated with the relative abilities of the isoforms to bind cell-surface proteoglycans. These results indicate that the differential biological actions among the FST isoforms and FSTL3 are primarily dependent on their relative cell-surface binding ability and ligand specificity.


Assuntos
Ativinas/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Relacionadas à Folistatina/metabolismo , Folistatina/química , Fator de Crescimento Transformador beta/metabolismo , Ativinas/química , Animais , Células COS , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Proliferação de Células , Chlorocebus aethiops , Folistatina/metabolismo , Humanos , Miostatina , Proteínas Recombinantes/química
16.
EMBO J ; 25(5): 1035-45, 2006 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-16482217

RESUMO

The secreted, multidomain protein follistatin binds activins with high affinity, inhibiting their receptor interaction. We have dissected follistatin's domain structure and shown that the minimal activin-inhibiting fragment of follistatin is comprised of the first and second Fs domains (Fs12). This protein can bind to activin dimer and form a stable complex containing two Fs12 molecules and one activin dimer. We have solved crystal structures of activin A alone and its complex with Fs12 fragment to 2 A resolution. The complex structure shows how Fs12 molecules wrap around the back of the 'wings' of activin, blocking the type II receptor-binding site on activin A. Arginine 192 in Fs2 is a key residue in this interaction, inserting itself in between activin's fingers. Complex formation imposes a novel orientation for the EGF- and Kazal-like subdomains in the Fs2 domain and activin A shows further variation from the canonical TGF-beta family fold. The structure provides a detailed description of the inhibitory mechanism and gives insights into interactions of follistatin with other TGF-beta family proteins.


Assuntos
Receptores de Activinas Tipo II/metabolismo , Ativinas/antagonistas & inibidores , Folistatina/química , Subunidades beta de Inibinas/antagonistas & inibidores , Estrutura Terciária de Proteína , Transdução de Sinais , Ativinas/genética , Ativinas/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Cristalografia por Raios X , Dimerização , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Embrião não Mamífero , Folistatina/genética , Folistatina/metabolismo , Humanos , Subunidades beta de Inibinas/genética , Subunidades beta de Inibinas/metabolismo , Ligantes , Modelos Moleculares , Dados de Sequência Molecular , Mutação/genética , Ligação Proteica , Ratos , Homologia de Sequência de Aminoácidos , Fator de Crescimento Transformador beta/metabolismo , Xenopus laevis
17.
Expert Opin Biol Ther ; 6(2): 147-54, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16436040

RESUMO

Skeletal muscle is the largest organ in the human body, and plays an important role in body movement and metabolism. Skeletal muscle mass is lost in genetic disorders such as muscular dystrophy, muscle wasting and ageing. Chemicals and proteins that restore muscle mass and function are potential drugs that can improve human health and could be used in the clinic. Myostatin is a muscle-specific member of the transforming growth factor (TGF)-beta superfamily that plays an essential role in the negative regulation of muscle growth. Inhibition of myostatin activity is a promising therapeutic method for restoring muscle mass and strength. Potential inhibitors of myostatin include follistatin domain-containing proteins, myostatin propeptide, myostatin antibodies and chemical compounds. These inhibitors could be beneficial for the development of clinical drugs for the treatment of muscular disorders. Bone morphogenetic protein (BMP) plays a significant role in the development of neuromuscular architecture and its proper functions. Modulation of BMP activity could be beneficial for muscle function in muscular disorders. This review will describe the current progress in therapy for muscular disorders, emphasising the importance of myostatin as a drug target.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Doenças Musculares/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Animais , Folistatina/química , Humanos , Modelos Biológicos , Músculo Esquelético/metabolismo , Músculos/metabolismo , Doenças Musculares/terapia , Distrofias Musculares/terapia , Miossarcoma/terapia , Miostatina , Estrutura Terciária de Proteína , Fator de Crescimento Transformador beta/química , Fator de Crescimento Transformador beta/metabolismo
18.
Dev Cell ; 9(4): 535-43, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16198295

RESUMO

TGF-beta ligands stimulate diverse cellular differentiation and growth responses by signaling through type I and II receptors. Ligand antagonists, such as follistatin, block signaling and are essential regulators of physiological responses. Here we report the structure of activin A, a TGF-beta ligand, bound to the high-affinity antagonist follistatin. Two follistatin molecules encircle activin, neutralizing the ligand by burying one-third of its residues and its receptor binding sites. Previous studies have suggested that type I receptor binding would not be blocked by follistatin, but the crystal structure reveals that the follistatin N-terminal domain has an unexpected fold that mimics a universal type I receptor motif and occupies this receptor binding site. The formation of follistatin:BMP:type I receptor complexes can be explained by the stoichiometric and geometric arrangement of the activin:follistatin complex. The mode of ligand binding by follistatin has important implications for its ability to neutralize homo- and heterodimeric ligands of this growth factor family.


Assuntos
Receptores de Ativinas Tipo I/metabolismo , Ativinas/química , Folistatina/química , Subunidades beta de Inibinas/química , Estrutura Terciária de Proteína , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Ativinas/genética , Ativinas/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Cricetinae , Cristalografia por Raios X , Folistatina/genética , Folistatina/metabolismo , Subunidades beta de Inibinas/genética , Subunidades beta de Inibinas/metabolismo , Ligantes , Modelos Moleculares , Dados de Sequência Molecular , Complexos Multiproteicos , Ligação Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Serina-Treonina Quinases , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptor do Fator de Crescimento Transformador beta Tipo II , Alinhamento de Sequência , Fator de Crescimento Transformador beta/metabolismo
19.
Reprod Biomed Online ; 10(6): 786-812, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15970011

RESUMO

The human reproductive process is regulated by complex mechanisms that involve many organs, including the brain, gonads and endocrine system. It has been more than 70 years since the name 'inhibin' was used to describe a substance produced in the gonads that negatively regulates pituitary secretion. Inhibin B controls FSH secretion via a negative feedback mechanism. It is a glycoprotein hormone secreted by the Sertoli cells of the testis and granulosa and theca cells of the ovary. Serum inhibin B concentrations are positively related to testicular volume and sperm counts. Current understanding of inhibin physiology and pathology in the human suggests that inhibin B may be of importance as a marker of Sertoli cell function in men with infertility and as a prognostic indicator in women undergoing ovulation induction therapy. Inhibin concentrations are elevated in patients with granulosa cell tumours and in post-menopausal women with mucinous ovarian cancers. Immunoreactivity against the inhibin alpha-subunit was identified in all cases of adrenal cortical adenoma and carcinoma, and levels are suppressed in the malignant prostate disease. This article discusses the structure, regulation and clinical use of inhibin and other related substances.


Assuntos
Infertilidade Masculina/fisiopatologia , Inibinas/sangue , Inibinas/fisiologia , Reprodução/fisiologia , Ativinas/química , Ativinas/metabolismo , Adulto , Fatores Etários , Cromossomos Humanos Y , Glândulas Exócrinas/metabolismo , Feminino , Fertilização in vitro/métodos , Folistatina/química , Folistatina/metabolismo , Humanos , Recém-Nascido , Inibinas/química , Masculino , Ciclo Menstrual/fisiologia , Neoplasias Ovarianas/sangue , Síndrome do Ovário Policístico/fisiopatologia , Gravidez , Deleção de Sequência , alfa-Macroglobulinas/química , alfa-Macroglobulinas/metabolismo
20.
J Clin Endocrinol Metab ; 89(10): 5067-75, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15472207

RESUMO

Follistatin (FST) is a monomeric activin-binding and neutralization protein that has at least three isoforms in human tissues and fluids. The full-length FS315 protein has an acidic 26-residue C-terminal tail that is not present in the shortest form, FS288, due to alternative splicing. An intermediate form, FS303, was identified in follicular fluid that is presumably derived by proteolytic processing of this tail domain. Interestingly, the biochemistry of each of these three isoforms is distinct, including their ability to bind to cell surface proteoglycans, an activity that ranks in the order FS288 > FS303 > FS315. This would suggest that the soluble, circulating FST isoform is likely to be FS315, a hypothesis supported by previous determinations that the serum and follicular fluid forms of FST are biochemically distinct. To test this hypothesis, we developed an immunoassay that is specific for full-length FS315. This assay was validated for use with human serum and follicular fluid samples and then used to examine FST in these fluid compartments. Our results indicate that FS315 is indeed the major circulating FST isoform but is undetectable in follicular fluid samples aspirated from normal women or women with polycystic ovary syndrome. These observations confirm the compartmentalization of FST isoforms according to their biochemical properties and biological actions so that the most soluble form is found in the circulation, whereas the forms that bind to cell surface proteoglycans are found in tissue compartments such as the ovarian follicle. They also confirm that the source of FST in human serum is not the ovarian follicle.


Assuntos
Líquido Folicular/metabolismo , Folistatina/sangue , Imunoensaio/métodos , Síndrome do Ovário Policístico/sangue , Anticorpos Monoclonais , Especificidade de Anticorpos , Calibragem , Linhagem Celular , Feminino , Folistatina/química , Folistatina/imunologia , Humanos , Imunoensaio/normas , Isomerismo , Reprodutibilidade dos Testes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA