Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Hum Mol Genet ; 27(2): 359-372, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29202173

RESUMO

Congenital hypogonadotropic hypogonadism (CHH) is a rare genetic disease characterized by absent puberty and infertility due to GnRH deficiency, and is often associated with anosmia [Kallmann syndrome (KS)]. The genetic etiology of CHH is heterogeneous, and more than 30 genes have been implicated in approximately 50% of patients with CHH. We hypothesized that genes encoding axon-guidance proteins containing fibronectin type-III (FN3) domains (similar to ANOS1, the first gene associated with KS), are mutated in CHH. We performed whole-exome sequencing in a cohort of 133 CHH probands to test this hypothesis, and identified rare sequence variants (RSVs) in genes encoding for the FN3-domain encoding protein deleted in colorectal cancer (DCC) and its ligand Netrin-1 (NTN1). In vitro studies of these RSVs revealed altered intracellular signaling associated with defects in cell morphology, and confirmed five heterozygous DCC mutations in 6 probands-5 of which presented as KS. Two KS probands carry heterozygous mutations in both DCC and NTN1 consistent with oligogenic inheritance. Further, we show that Netrin-1 promotes migration in immortalized GnRH neurons (GN11 cells). This study implicates DCC and NTN1 mutations in the pathophysiology of CHH consistent with the role of these two genes in the ontogeny of GnRH neurons in mice.


Assuntos
Receptor DCC/genética , Hipogonadismo/genética , Netrina-1/genética , Adulto , Estudos de Coortes , Receptor DCC/metabolismo , Feminino , Domínio de Fibronectina Tipo III , Hormônio Liberador de Gonadotropina/deficiência , Humanos , Hipogonadismo/metabolismo , Hipogonadismo/patologia , Masculino , Mutação , Netrina-1/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Linhagem , Sequenciamento do Exoma
2.
EMBO Mol Med ; 9(10): 1379-1397, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28754744

RESUMO

Congenital hypogonadotropic hypogonadism (CHH) is a rare genetic form of isolated gonadotropin-releasing hormone (GnRH) deficiency caused by mutations in > 30 genes. Fibroblast growth factor receptor 1 (FGFR1) is the most frequently mutated gene in CHH and is implicated in GnRH neuron development and maintenance. We note that a CHH FGFR1 mutation (p.L342S) decreases signaling of the metabolic regulator FGF21 by impairing the association of FGFR1 with ß-Klotho (KLB), the obligate co-receptor for FGF21. We thus hypothesized that the metabolic FGF21/KLB/FGFR1 pathway is involved in CHH Genetic screening of 334 CHH patients identified seven heterozygous loss-of-function KLB mutations in 13 patients (4%). Most patients with KLB mutations (9/13) exhibited metabolic defects. In mice, lack of Klb led to delayed puberty, altered estrous cyclicity, and subfertility due to a hypothalamic defect associated with inability of GnRH neurons to release GnRH in response to FGF21. Peripheral FGF21 administration could indeed reach GnRH neurons through circumventricular organs in the hypothalamus. We conclude that FGF21/KLB/FGFR1 signaling plays an essential role in GnRH biology, potentially linking metabolism with reproduction.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Síndrome de Kallmann/genética , Proteínas de Membrana/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Animais , Células COS , Caenorhabditis elegans/genética , Chlorocebus aethiops , Estudos de Coortes , Feminino , Fatores de Crescimento de Fibroblastos/genética , Hormônio Liberador de Gonadotropina/genética , Células HEK293 , Humanos , Hipotálamo/metabolismo , Proteínas Klotho , Masculino , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Neurônios/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética
3.
Gene ; 516(1): 146-51, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23276709

RESUMO

FGFR1 mutations have been identified in both Kallmann syndrome and normosmic HH (nIHH). To date, few mutations in the FGFR1 gene have been structurally or functionally characterized in vitro to identify molecular mechanisms that contribute to the disease pathogenesis. We attempted to define the in vitro functionality of two FGFR1 mutants (R254W and R254Q), resulting from two different amino acid substitutions of the same residue, and to correlate the in vitro findings to the patient phenotypes. Two unrelated GnRH deficient probands were found to harbor mutations in FGFR1 (R254W and R254Q). Mutant signaling activity and expression levels were evaluated in vitro and compared to a wild type (WT) receptor. Signaling activity was determined by a FGF2/FGFR1 dependent transcription reporter assay. Receptor total expression levels were assessed by Western blot and cell surface expression was measured by a radiolabeled antibody binding assay. The R254W maximal receptor signaling capacity was reduced by 45% (p<0.01) while R254Q activity was not different from WT. However, both mutants displayed diminished total protein expression levels (40 and 30% reduction relative to WT, respectively), while protein maturation was unaffected. Accordingly, cell surface expression levels of the mutant receptors were also significantly reduced (35% p<0.01 and 15% p<0.05, respectively). The p.R254W and p.R254Q are both loss-of-function mutations as demonstrated by their reduced overall and cell surface expression levels suggesting a deleterious effect on receptor folding and stability. It appears that a tryptophan substitution at R254 is more disruptive to receptor structure than the more conserved glutamine substitution. No clear correlation between the severity of in vitro loss-of-function and phenotypic presentation could be assigned.


Assuntos
Hipogonadismo/genética , Mutação , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Adolescente , Animais , Células COS , Chlorocebus aethiops , Simulação por Computador , Regulação da Expressão Gênica , Genótipo , Glicosídeo Hidrolases/genética , Glicosídeo Hidrolases/metabolismo , Hormônio Liberador de Gonadotropina/genética , Humanos , Síndrome de Kallmann/genética , Masculino , Fenótipo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais
4.
J Clin Endocrinol Metab ; 97(4): E694-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22319038

RESUMO

CONTEXT: Kallmann syndrome (KS), combined pituitary hormone deficiency (CPHD), and septo-optic dysplasia (SOD) all result from development defects of the anterior midline in the human forebrain. OBJECTIVE: The objective of the study was to investigate whether KS, CPHD, and SOD have shared genetic origins. DESIGN AND PARTICIPANTS: A total of 103 patients with either CPHD (n = 35) or SOD (n = 68) were investigated for mutations in genes implicated in the etiology of KS (FGFR1, FGF8, PROKR2, PROK2, and KAL1). Consequences of identified FGFR1, FGF8, and PROKR2 mutations were investigated in vitro. RESULTS: Three patients with SOD had heterozygous mutations in FGFR1; these were either shown to alter receptor signaling (p.S450F, p.P483S) or predicted to affect splicing (c.336C>T, p.T112T). One patient had a synonymous change in FGF8 (c.216G>A, p.T72T) that was shown to affect splicing and ligand signaling activity. Four patients with CPHD/SOD were found to harbor heterozygous rare loss-of-function variants in PROKR2 (p.R85G, p.R85H, p.R268C). CONCLUSIONS: Mutations in FGFR1/FGF8/PROKR2 contributed to 7.8% of our patients with CPHD/SOD. These data suggest a significant genetic overlap between conditions affecting the development of anterior midline in the human forebrain.


Assuntos
Fator 8 de Crescimento de Fibroblasto/genética , Hipopituitarismo/genética , Síndrome de Kallmann/genética , Mutação , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Peptídeos/genética , Displasia Septo-Óptica/genética , Animais , Feminino , Fator 8 de Crescimento de Fibroblasto/metabolismo , Estudos de Associação Genética , Heterozigoto , Humanos , Hipopituitarismo/metabolismo , Síndrome de Kallmann/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Neuro-Hipófise/metabolismo , Neuro-Hipófise/patologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Displasia Septo-Óptica/metabolismo , Transdução de Sinais , Reino Unido , Estados Unidos
5.
Mol Cell Endocrinol ; 346(1-2): 44-50, 2011 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-21664414

RESUMO

Prokineticin, 1 (PROK1) and prokineticin 2 (PROK2), are two closely related proteins that were identified as the mammalian homologs of their two amphibian homologs, mamba intestinal toxin (MIT-1) and Bv8. MIT-1 was initially identified as a non-toxic constituent in the venom of the black mamba snake (Dendroaspis polylepis) (Joubert and Strydom, 1980) while Bv8 was identified in the skin secretion of the toad, Bombina variegate (Mollay et al., 1999). All three homologs stimulate gastrointestinal motility thus accounting for their family name "prokineticins" (Schweitz et al., 1990, 1999). However, since its initial description, both PROK1 and PROK2 have been found to regulate a dazzling array of biological functions throughout the body. In particular, PROK1 acts as a potent angiogenic mitogen on endocrine vascular epithelium, thus earning its other name, Endocrine gland-vascular endothelial factor (EG-VEGF) (LeCouter et al., 2002). In contrast, the PROK2 signaling pathway is a critical regulator of olfactory bulb morphogenesis and sexual maturation in mammals and this function is the focus of this review.


Assuntos
Hormônios Gastrointestinais/genética , Hormônios Gastrointestinais/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Reprodução/fisiologia , Animais , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Bulbo Olfatório/metabolismo , Bulbo Olfatório/fisiologia , Reprodução/genética , Maturidade Sexual/genética , Maturidade Sexual/fisiologia
6.
N Engl J Med ; 364(3): 215-25, 2011 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-21247312

RESUMO

BACKGROUND: Functional hypothalamic amenorrhea is a reversible form of gonadotropin-releasing hormone (GnRH) deficiency commonly triggered by stressors such as excessive exercise, nutritional deficits, or psychological distress. Women vary in their susceptibility to inhibition of the reproductive axis by such stressors, but it is unknown whether this variability reflects a genetic predisposition to hypothalamic amenorrhea. We hypothesized that mutations in genes involved in idiopathic hypogonadotropic hypogonadism, a congenital form of GnRH deficiency, are associated with hypothalamic amenorrhea. METHODS: We analyzed the coding sequence of genes associated with idiopathic hypogonadotropic hypogonadism in 55 women with hypothalamic amenorrhea and performed in vitro studies of the identified mutations. RESULTS: Six heterozygous mutations were identified in 7 of the 55 patients with hypothalamic amenorrhea: two variants in the fibroblast growth factor receptor 1 gene FGFR1 (G260E and R756H), two in the prokineticin receptor 2 gene PROKR2 (R85H and L173R), one in the GnRH receptor gene GNRHR (R262Q), and one in the Kallmann syndrome 1 sequence gene KAL1 (V371I). No mutations were found in a cohort of 422 controls with normal menstrual cycles. In vitro studies showed that FGFR1 G260E, FGFR1 R756H, and PROKR2 R85H are loss-of-function mutations, as has been previously shown for PROKR2 L173R and GNRHR R262Q. CONCLUSIONS: Rare variants in genes associated with idiopathic hypogonadotropic hypogonadism are found in women with hypothalamic amenorrhea, suggesting that these mutations may contribute to the variable susceptibility of women to the functional changes in GnRH secretion that characterize hypothalamic amenorrhea. Our observations provide evidence for the role of rare variants in common multifactorial disease. (Funded by the Eunice Kennedy Shriver National Institute of Child Health and Human Development and others; ClinicalTrials.gov number, NCT00494169.).


Assuntos
Amenorreia/genética , Hormônio Liberador de Gonadotropina/deficiência , Doenças Hipotalâmicas/genética , Mutação , Amenorreia/etiologia , Proteínas da Matriz Extracelular/genética , Feminino , Expressão Gênica , Predisposição Genética para Doença , Hormônio Liberador de Gonadotropina/genética , Humanos , Hipogonadismo/genética , Doenças Hipotalâmicas/complicações , Hormônio Luteinizante/metabolismo , Proteínas do Tecido Nervoso/genética , Precursores de Proteínas/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptores Acoplados a Proteínas G/genética , Receptores LHRH/genética , Receptores de Peptídeos/genética , Análise de Sequência de DNA
7.
J Clin Endocrinol Metab ; 94(11): 4380-90, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19820032

RESUMO

CONTEXT: FGFR1 mutations have been identified in about 10% of patients with Kallmann syndrome. Recently cases of idiopathic hypogonadotropic hypogonadism (IHH) with a normal sense of smell (nIHH) have been reported. AIMS: The objective of the study was to define the frequency of FGFR1 mutations in a large cohort of nIHH, delineate the spectrum of reproductive phenotypes, assess functionality of the FGFR1 mutant alleles in vitro, and investigate genotype-phenotype relationships. DESIGN: FGFR1 sequencing of 134 well-characterized nIHH patients (112 men and 22 women) and 270 healthy controls was performed. The impact of the identified mutations on FGFR1 function was assessed using structural prediction and in vitro studies. RESULTS: Nine nIHH subjects (five males and four females; 7%) harbor a heterozygous mutation in FGFR1 and exhibit a wide spectrum of pubertal development, ranging from absent puberty to reversal of IHH in both sexes. All mutations impair receptor function. The Y99C, Y228D, and I239T mutants impair the tertiary folding, resulting in incomplete glycosylation and reduced cell surface expression. The R250Q mutant reduces receptor affinity for FGF. The K618N, A671P, and Q680X mutants impair tyrosine kinase activity. However, the degree of functional impairment of the mutant receptors did not always correlate with the reproductive phenotype, and variable expressivity of the disease was noted within family members carrying the same FGFR1 mutation. These discrepancies were partially explained by additional mutations in known IHH loci. CONCLUSIONS: Loss-of-function mutations in FGFR1 underlie 7% of nIHH with different degrees of impairment in vitro. These mutations act in concert with other gene defects in several cases, consistent with oligogenicity.


Assuntos
Hipogonadismo/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Animais , Células COS , Chlorocebus aethiops , Estudos de Coortes , DNA/sangue , DNA/genética , DNA/isolamento & purificação , Estradiol/sangue , Feminino , Fator 2 de Crescimento de Fibroblastos/farmacologia , Genótipo , Humanos , Masculino , Mutação , Fenótipo , Fosfotirosina/metabolismo , Puberdade Tardia/genética , Valores de Referência , Testosterona/sangue
8.
J Clin Invest ; 118(8): 2822-31, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18596921

RESUMO

Idiopathic hypogonadotropic hypogonadism (IHH) with anosmia (Kallmann syndrome; KS) or with a normal sense of smell (normosmic IHH; nIHH) are heterogeneous genetic disorders associated with deficiency of gonadotropin-releasing hormone (GnRH). While loss-of-function mutations in FGF receptor 1 (FGFR1) cause human GnRH deficiency, to date no specific ligand for FGFR1 has been identified in GnRH neuron ontogeny. Using a candidate gene approach, we identified 6 missense mutations in FGF8 in IHH probands with variable olfactory phenotypes. These patients exhibited varied degrees of GnRH deficiency, including the rare adult-onset form of hypogonadotropic hypogonadism. Four mutations affected all 4 FGF8 splice isoforms (FGF8a, FGF8b, FGF8e, and FGF8f), while 2 mutations affected FGF8e and FGF8f isoforms only. The mutant FGF8b and FGF8f ligands exhibited decreased biological activity in vitro. Furthermore, mice homozygous for a hypomorphic Fgf8 allele lacked GnRH neurons in the hypothalamus, while heterozygous mice showed substantial decreases in the number of GnRH neurons and hypothalamic GnRH peptide concentration. In conclusion, we identified FGF8 as a gene implicated in GnRH deficiency in both humans and mice and demonstrated an exquisite sensitivity of GnRH neuron development to reductions in FGF8 signaling.


Assuntos
Fator 8 de Crescimento de Fibroblasto/metabolismo , Hormônio Liberador de Gonadotropina/deficiência , Transdução de Sinais , Adulto , Animais , Estudos de Casos e Controles , Estudos de Coortes , Feminino , Fator 8 de Crescimento de Fibroblasto/química , Fator 8 de Crescimento de Fibroblasto/genética , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Heterozigoto , Humanos , Hipogonadismo/genética , Hipogonadismo/fisiopatologia , Síndrome de Kallmann/genética , Síndrome de Kallmann/fisiopatologia , Masculino , Camundongos , Camundongos Transgênicos , Modelos Moleculares , Mutação , Neurônios/citologia , Neurônios/metabolismo , Transtornos do Olfato/genética , Linhagem
9.
J Clin Endocrinol Metab ; 93(9): 3551-9, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18559922

RESUMO

CONTEXT: Mice deficient in prokineticin 2(PROK2) and prokineticin receptor2 (PROKR2) exhibit variable olfactory bulb dysgenesis and GnRH neuronal migration defects reminiscent of human GnRH deficiency. OBJECTIVES: We aimed to screen a large cohort of patients with Kallmann syndrome (KS) and normosmic idiopathic hypogonadotropic hypogonadism (IHH) for mutations in PROK2/PROKR2, evaluate their prevalence, define the genotype/phenotype relationship, and assess the functionality of these mutant alleles in vitro. DESIGN: Sequencing of the PROK2 and PROKR2 genes was performed in 170 KS patients and 154 nIHH. Mutations were examined using early growth response 1-luciferase assays in HEK 293 cells and aequorin assays in Chinese hamster ovary cells. RESULTS: Four heterozygous and one homozygous PROK2 mutation (p.A24P, p.C34Y, p.I50M, p.R73C, and p.I55fsX1) were identified in five probands. Four probands had KS and one nIHH, and all had absent puberty. Each mutant peptide impaired receptor signaling in vitro except the I50M. There were 11 patients who carried a heterozygous PROKR2 mutation (p.R85C, p.Y113H, p.V115M, p.R164Q, p.L173R, p.W178S, p.S188L, p.R248Q, p.V331M, and p.R357W). Among them, six had KS, four nIHH, and one KS proband carried both a PROKR2 (p.V115M) and PROK2 (p.A24P) mutation. Reproductive phenotypes ranged from absent to partial puberty to complete reversal of GnRH deficiency after discontinuation of therapy. All mutant alleles appear to decrease intracellular calcium mobilization; seven exhibited decreased MAPK signaling, and six displayed decreased receptor expression. Nonreproductive phenotypes included fibrous dysplasia, sleep disorder, synkinesia, and epilepsy. Finally, considerable variability was evident in family members with the same mutation, including asymptomatic carriers. CONCLUSION: Loss-of-function mutations in PROK2 and PROKR2 underlie both KS and nIHH.


Assuntos
Hormônios Gastrointestinais/genética , Hormônio Liberador de Gonadotropina/deficiência , Hipogonadismo/genética , Mutação de Sentido Incorreto , Neuropeptídeos/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Peptídeos/genética , Adolescente , Equorina/genética , Animais , Células CHO , Cricetinae , Cricetulus , Análise Mutacional de DNA , Feminino , Frequência do Gene , Heterogeneidade Genética , Genótipo , Humanos , Síndrome de Kallmann/genética , Masculino , Modelos Biológicos , Linhagem , Transfecção
10.
Blood ; 111(10): 5195-204, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18326817

RESUMO

Hemojuvelin (HJV) is a coreceptor for bone morphogenetic protein (BMP) signaling that regulates hepcidin expression and iron metabolism. However, the precise combinations of BMP ligands and receptors used by HJV remain unknown. HJV has also been demonstrated to bind to neogenin, but it is not known whether this interaction has a role in regulating hepcidin expression. In the present study, we show that BMP-2, BMP-4, and BMP-6 are endogenous ligands for HJV in hepatoma-derived cell lines, and that all 3 of these ligands are expressed in human liver. We demonstrate in vitro that HJV selectively uses the BMP type II receptors ActRIIA and BMPRII, but not ActRIIB, and HJV enhances utilization of ActRIIA by BMP-2 and BMP-4. Interestingly, ActRIIA is the predominant BMP type II receptor expressed in human liver. While HJV can use all 3 BMP type I receptors (ALK2, ALK3, and ALK6) in vitro, only ALK2 and ALK3 are detected in human liver. Finally, we show that HJV-induced BMP signaling and hepcidin expression are not altered by neogenin overexpression or by inhibition of endogenous neogenin expression. Thus, HJV-mediated BMP signaling and hepcidin regulation occur via a distinct subset of BMP ligands and BMP receptors, independently of neogenin.


Assuntos
Peptídeos Catiônicos Antimicrobianos/genética , Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Regulação da Expressão Gênica , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/fisiologia , Receptores de Ativinas Tipo I/metabolismo , Receptores de Activinas Tipo II/metabolismo , Proteína Morfogenética Óssea 2 , Proteína Morfogenética Óssea 4 , Proteína Morfogenética Óssea 6 , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Carcinoma Hepatocelular/química , Carcinoma Hepatocelular/patologia , Proteínas Ligadas por GPI , Proteína da Hemocromatose , Hepcidinas , Humanos , Ligantes , Fígado/química , Fator de Crescimento Transformador beta
11.
J Clin Invest ; 117(7): 1933-9, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17607365

RESUMO

Systemic iron balance is regulated by hepcidin, a peptide hormone secreted by the liver. By decreasing cell surface expression of the iron exporter ferroportin, hepcidin decreases iron absorption from the intestine and iron release from reticuloendothelial stores. Hepcidin excess has been implicated in the pathogenesis of anemia of chronic disease, while hepcidin deficiency has a key role in the pathogenesis of the iron overload disorder hemochromatosis. We have recently shown that hemojuvelin is a coreceptor for bone morphogenetic protein (BMP) signaling and that BMP signaling positively regulates hepcidin expression in liver cells in vitro. Here we show that BMP-2 administration increases hepcidin expression and decreases serum iron levels in vivo. We also show that soluble hemojuvelin (HJV.Fc) selectively inhibits BMP induction of hepcidin expression in vitro and that administration of HJV.Fc decreases hepcidin expression, increases ferroportin expression, mobilizes splenic iron stores, and increases serum iron levels in vivo. These data support a role for modulators of the BMP signaling pathway in treating diseases of iron overload and anemia of chronic disease.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Ferro/metabolismo , Transdução de Sinais , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Proteínas Morfogenéticas Ósseas/classificação , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/farmacologia , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular , Regulação da Expressão Gênica , Hepcidinas , Humanos , Interleucina-6/farmacologia , Ligantes , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Sistema Fagocitário Mononuclear/metabolismo , Transdução de Sinais/efeitos dos fármacos , Solubilidade , Fator de Crescimento Transformador beta/classificação , Fator de Crescimento Transformador beta/farmacologia
12.
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
13.
Endocrinology ; 146(8): 3614-21, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15890774

RESUMO

Bone morphogenetic proteins (BMPs) play important roles in reproduction including primordial germ cell formation, follicular development, spermatogenesis, and FSH secretion. Dragon, a recently identified glycosylphosphatidylinositol-anchored member of the repulsive guidance molecule family, is also a BMP coreceptor. In the present study, we determined the tissue and cellular localization of Dragon in reproductive organs using immunohistochemistry and in situ hybridization. Among reproductive organs, Dragon was expressed in testis, epididymis, ovary, uterus, and pituitary. In the testis of early postnatal mice, Dragon was found in gonocytes and spermatogonia, whereas in immature testes, Dragon was only weakly expressed in spermatogonia. Interestingly, pregnant mare serum gonadotropin treatment of immature mice robustly induced Dragon production in spermatocytes. In adult testis, Dragon was found in spermatocytes and round spermatids. In the ovary, Dragon was detected exclusively within oocytes and primarily those within secondary follicles. In the pituitary, Dragon-expressing cells overlapped FSH-expressing cells. Dragon was also expressed in a number of cell lines originating from reproductive tissues including Ishikawa, Hela, LbetaT2, MCF-7, and JEG3 cells. Immunocytochemistry and gradient sucrose ultracentrifugation studies showed Dragon was localized in lipid rafts within the plasma membrane. In reproductive cell lines, Dragon expression enhanced signaling of exogenous BMP2 or BMP4. The present studies demonstrate that Dragon expression is dynamically regulated throughout the reproductive tract and that Dragon protein modulates BMP signaling in cells from reproductive tissues. The overlap between Dragon expression and the functional BMP signaling system suggests that Dragon may play a role in mammalian reproduction.


Assuntos
Proteínas do Tecido Nervoso/genética , Moléculas de Adesão de Célula Nervosa/genética , Animais , Epididimo/fisiologia , Feminino , Regulação da Expressão Gênica , Células HeLa , Humanos , Imuno-Histoquímica , Hibridização In Situ , Masculino , Microdomínios da Membrana/ultraestrutura , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Ovário/fisiologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espermatócitos/fisiologia , Útero/fisiologia
14.
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
15.
Mol Endocrinol ; 18(1): 228-40, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14563935

RESUMO

Follistatin (FS) is an important regulator of pituitary FSH secretion through its potent ability to bind and bioneutralize activin. It also represents a prototype for binding proteins that control bioavailability of other TGFbeta-related growth factors such as the bone morphogenetic proteins. The 288-residue FS molecule has a distinctive structure comprised principally of three 10-cysteine FS domains. These are preceded by an N-terminal segment shown by us previously to contain hydrophobic residues essential for activin binding. To establish the contribution of the FS domains themselves to FS's bioactivity, we prepared mutants with deleted or exchanged domains and intradomain point mutations. Mutants were expressed from mammalian (Chinese hamster ovary) cells and evaluated for activin binding and for biological activity in assays measuring differing aspects of FS bioactivity: activin-mediated transcriptional activity and suppression of FSH secretion in primary pituitary cell cultures. The N-terminal domain (residues 1-63) alone could not bind activin or suppress activin-mediated transcription, either alone or combined in solution with the FS domain region (residues 64-288). Deletion of FS domains 1 or 2 abolished activin binding and biological activity in both assays, whereas deletion of domain 3 was tolerated. Bioactivity was also reduced or eliminated after exchange of domains (FS 2/1/3 and FS 3/1/2) or doubling of domain 1 (FS 1/1/3) or domain 2 (FS 2/2/3). Several hydrophobic residues clustered within the C-terminal region of FS domains 1 and 2 are highly conserved among all FS domains. Mutation of any of these to Asp or Ala either reduced or eliminated FS bioactivity and disrupted distant epitopes for heparin binding (FS domain 1) or antibody recognition (FS domain 2), suggesting their role in maintaining the conformational integrity of the domain and possibly the FS molecule as a whole. These results are consistent with the importance of domain conformation as well as the overall order of the domains in FS function. A continuous sequence comprising the N-terminal domain and followed by FS domains 1 and 2 fulfills the minimum structural requirement for activin binding and FS bioactivity.


Assuntos
Folistatina/química , Folistatina/genética , Ativinas/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Divisão Celular , Folistatina/metabolismo , Folistatina/farmacologia , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação Puntual , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Fator de Crescimento Transformador beta/fisiologia
16.
Endocrinology ; 144(5): 1671-4, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12697670

RESUMO

Modulation of activin and other TGF beta superfamily signaling is the primary mechanism of action for both follistatin (FS) and FS-like 3 (FSTL-3). However, most studies of these ligands use activin A due to its wide availability. We have now tested the ability of FS288 and FSTL-3 to bind and neutralize activin B relative to activin A. Activin B bound to both FS and FSTL-3 at a potency approximately 10-fold lower than that of activin A. Moreover, whereas both activins had similar biological activity in 293 cell reporter assays, FS and FSTL-3 were approximately 3-fold more effective in neutralizing activin A relative to activin B. These results suggest that neutralization of activins A and B by FS and FSTL-3 are not identical, so that the relative activity of each activin in tissues where both are produced, such as in the ovary, could be quite different. In addition, biological systems that use primarily activin B, but which have been examined in vitro using activin A, may need to be reevaluated to determine the actual physiologic roles of FS or FSTL-3.


Assuntos
Ativinas/metabolismo , Proteínas Relacionadas à Folistatina/metabolismo , Folistatina/metabolismo , Subunidades beta de Inibinas/metabolismo , Linhagem Celular , Humanos
17.
Endocrinology ; 143(5): 1613-24, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11956142

RESUMO

Follistatin-related protein (FSRP) is a new addition to the expanding follistatin (FS)-related gene family whose members contain at least one conserved 10-cysteine follistatin domain. In contrast to other members of this family, FSRP and follistatin also share a common exon/intron domain structure, substantial primary sequence homology, and an ability to irreversibly bind activin. In this study, we further explored the hypothesis that FSRP is a functional as well as structural homologue of FS. N-terminal sequencing of recombinant FSRP revealed that signal peptide cleavage occurs within exon 1, a significant structural difference from FS, in which cleavage occurs at the exon/intron boundary. Solid-phase radioligand competition assays revealed both FS and FSRP to preferentially bind activin with the next closest TGF-beta superfamily member, bone-morphogenic protein-7, being at least 500-fold less potent. Consistent with their similar activin-binding affinities, FSRP and FS both prevented exogenous (endocrine or paracrine) activin from accessing its receptor and inducing gene transcription in bioassays. However, FS was at least 100-fold more potent than FSRP in inhibiting gene transcription and FSH release mediated by endogenously produced (autocrine) activin-A or activin-B in multiple cell systems. Finally, FSRP lacks the heparin-binding sequence found in FS, and we found that it was also unable to bind cell surface heparin sulfated proteoglycans. These findings suggest that structural differences between FSRP and FS may underlie their different neutralizating capabilities with respect to exogenous vs. endogenous activin. Taken together with our previous studies showing that activin binding is essential for FS's biological activity, the differential activities of FSRP and FS further indicate that activin binding is necessary but not sufficient to account for all of FS's actions.


Assuntos
Ativinas/antagonistas & inibidores , Ativinas/farmacologia , Glicoproteínas/farmacologia , Receptores de Ativinas/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Ligação Competitiva/efeitos dos fármacos , Células COS , Linhagem Celular , Folistatina , Proteínas Relacionadas à Folistatina , Heparina/metabolismo , Humanos , Ligantes , Luciferases/biossíntese , Luciferases/genética , Masculino , Camundongos , Dados de Sequência Molecular , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Neoplasias Hipofisárias/metabolismo , Ligação Proteica , Proteoglicanas/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA