Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(27): e2113749119, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35763574

RESUMO

Kisspeptin neurons in the mediobasal hypothalamus (MBH) are critical targets of ovarian estrogen feedback regulating mammalian fertility. To reveal molecular mechanisms underlying this signaling, we thoroughly characterized the estrogen-regulated transcriptome of kisspeptin cells from ovariectomized transgenic mice substituted with 17ß-estradiol or vehicle. MBH kisspeptin neurons were harvested using laser-capture microdissection, pooled, and subjected to RNA sequencing. Estrogen treatment significantly (p.adj. < 0.05) up-regulated 1,190 and down-regulated 1,139 transcripts, including transcription factors, neuropeptides, ribosomal and mitochondrial proteins, ion channels, transporters, receptors, and regulatory RNAs. Reduced expression of the excitatory serotonin receptor-4 transcript (Htr4) diminished kisspeptin neuron responsiveness to serotonergic stimulation. Many estrogen-regulated transcripts have been implicated in puberty/fertility disorders. Patients (n = 337) with congenital hypogonadotropic hypogonadism (CHH) showed enrichment of rare variants in putative CHH-candidate genes (e.g., LRP1B, CACNA1G, FNDC3A). Comprehensive characterization of the estrogen-dependent kisspeptin neuron transcriptome sheds light on the molecular mechanisms of ovary-brain communication and informs genetic research on human fertility disorders.


Assuntos
Núcleo Arqueado do Hipotálamo , Estrogênios , Fertilidade , Kisspeptinas , Neurônios , Ovário , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Estrogênios/metabolismo , Feminino , Fertilidade/genética , Perfilação da Expressão Gênica , Humanos , Hipogonadismo/congênito , Hipogonadismo/genética , Kisspeptinas/genética , Kisspeptinas/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Ovário/metabolismo
2.
Am J Hum Genet ; 106(1): 58-70, 2020 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-31883645

RESUMO

Congenital hypogonadotropic hypogonadism (CHH) is a rare genetic disorder characterized by infertility and the absence of puberty. Defects in GnRH neuron migration or altered GnRH secretion and/or action lead to a severe gonadotropin-releasing hormone (GnRH) deficiency. Given the close developmental association of GnRH neurons with the olfactory primary axons, CHH is often associated with anosmia or hyposmia, in which case it is defined as Kallmann syndrome (KS). The genetics of CHH are heterogeneous, and >40 genes are involved either alone or in combination. Several CHH-related genes controlling GnRH ontogeny encode proteins containing fibronectin-3 (FN3) domains, which are important for brain and neural development. Therefore, we hypothesized that defects in other FN3-superfamily genes would underlie CHH. Next-generation sequencing was performed for 240 CHH unrelated probands and filtered for rare, protein-truncating variants (PTVs) in FN3-superfamily genes. Compared to gnomAD controls the CHH cohort was statistically enriched for PTVs in neuron-derived neurotrophic factor (NDNF) (p = 1.40 × 10-6). Three heterozygous PTVs (p.Lys62∗, p.Tyr128Thrfs∗55, and p.Trp469∗, all absent from the gnomAD database) and an additional heterozygous missense mutation (p.Thr201Ser) were found in four KS probands. Notably, NDNF is expressed along the GnRH neuron migratory route in both mouse embryos and human fetuses and enhances GnRH neuron migration. Further, knock down of the zebrafish ortholog of NDNF resulted in altered GnRH migration. Finally, mice lacking Ndnf showed delayed GnRH neuron migration and altered olfactory axonal projections to the olfactory bulb; both results are consistent with a role of NDNF in GnRH neuron development. Altogether, our results highlight NDNF as a gene involved in the GnRH neuron migration implicated in KS.


Assuntos
Movimento Celular , Hipogonadismo/congênito , Hipogonadismo/genética , Mutação , Fatores de Crescimento Neural/genética , Neurônios/patologia , Adolescente , Animais , Estudos de Coortes , Feminino , Heterozigoto , Humanos , Hipogonadismo/patologia , Masculino , Camundongos , Camundongos Knockout , Fatores de Crescimento Neural/fisiologia , Neurônios/metabolismo , Linhagem , Peixe-Zebra
3.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 39(1): 35-38, 2022 Jan 10.
Artigo em Zh | MEDLINE | ID: mdl-34964963

RESUMO

OBJECTIVE: To investigate the clinical characteristics and genetic basis for a child with Keppen-Lubinsky syndrome (KPLBS). METHODS: Trio-whole exome sequencing (Trio-WES) was carried out for the proband and her parents. Candidate variant was verified by Sanger sequencing and bioinformatic analysis. RESULTS: The child has featured peculiar facies including large eyes, alar hypoplasia, microretrognathia, premature aging appearance in addition with growth delay and mental retardation. Trio-WES has identified that she has carried a de novo variant of the KCNJ6 gene, namely c.460G>C (p.Gly154Arg). The variant has not been recorded in the database. Prediction of protein structure indicated that the variant may affect the potassium ion selective filtration structure channel in the transmembrane region of KCNJ6 protein, which may result in up regulation of the function of the channel. CONCLUSION: The de novo c.460G>C (p.Gly154Arg) variant of the KCNJ6 gene probably underlay the KPLBS in this child. Above finding has enriched the genotypic and phenotype spectrum of this syndrome.


Assuntos
Deficiência Intelectual , Catarata , China , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Humanos , Hipogonadismo/congênito , Deficiência Intelectual/genética , Mutação , Sequenciamento do Exoma
4.
Hum Genet ; 140(1): 77-111, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32200437

RESUMO

A genetic basis of congenital isolated hypogonadotropic hypogonadism (CHH) can be defined in almost 50% of cases, albeit not necessarily the complete genetic basis. Next-generation sequencing (NGS) techniques have led to the discovery of a great number of loci, each of which has illuminated our understanding of human gonadotropin-releasing hormone (GnRH) neurons, either in respect of their embryonic development or their neuroendocrine regulation as the "pilot light" of human reproduction. However, because each new gene linked to CHH only seems to underpin another small percentage of total patient cases, we are still far from achieving a comprehensive understanding of the genetic basis of CHH. Patients have generally not benefited from advances in genetics in respect of novel therapies. In most cases, even genetic counselling is limited by issues of apparent variability in expressivity and penetrance that are likely underpinned by oligogenicity in respect of known and unknown genes. Robust genotype-phenotype relationships can generally only be established for individuals who are homozygous, hemizygous or compound heterozygotes for the same gene of variant alleles that are predicted to be deleterious. While certain genes are purely associated with normosmic CHH (nCHH) some purely with the anosmic form (Kallmann syndrome-KS), other genes can be associated with both nCHH and KS-sometimes even within the same kindred. Even though the anticipated genetic overlap between CHH and constitutional delay in growth and puberty (CDGP) has not materialised, previously unanticipated genetic relationships have emerged, comprising conditions of combined (or multiple) pituitary hormone deficiency (CPHD), hypothalamic amenorrhea (HA) and CHARGE syndrome. In this review, we report the current evidence in relation to phenotype and genetic peculiarities regarding 60 genes whose loss-of-function variants can disrupt the central regulation of reproduction at many levels: impairing GnRH neurons migration, differentiation or activation; disrupting neuroendocrine control of GnRH secretion; preventing GnRH neuron migration or function and/or gonadotropin secretion and action.


Assuntos
Hipogonadismo/congênito , Hipogonadismo/genética , Alelos , Animais , Estudos de Associação Genética/métodos , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Síndrome de Kallmann/genética , Mutação com Perda de Função/genética , Células Neuroendócrinas/metabolismo , Fenótipo
5.
Neuroendocrinology ; 111(12): 1176-1186, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33227799

RESUMO

BACKGROUND: Hypogonadotropic hypogonadism (HH) is hypogonadism due to either hypothalamic or pituitary dysfunction. While gonadotropin-releasing hormone (GnRH) can directly test pituitary function, no specific test of hypothalamic function exists. Kisspeptin-54 (KP54) is a neuropeptide that directly stimulates hypothalamic GnRH release and thus could be used to specifically interrogate hypothalamic function. Congenital HH (CHH) is typically due to variants in genes that control hypothalamic GnRH neuronal migration or function. Thus, we investigated whether KP54 could accurately identify hypothalamic dysfunction in men with CHH. METHODS: Men with CHH (n = 21) and healthy eugonadal men (n = 21) received an intravenous bolus of either GnRH (100 µg) or KP54 (6.4 nmol/kg), on 2 occasions, and were monitored for 6 h after administration of each neuropeptide. RESULTS: Maximal luteinizing hormone (LH) rise after KP54 was significantly greater in healthy men (12.5 iU/L) than in men with CHH (0.4 iU/L; p < 0.0001). KP54 more accurately differentiated CHH men from healthy men than GnRH (area under receiver operating characteristic curve KP54: 1.0, 95% CI 1.0-1.0; GnRH: 0.88, 95% CI 0.76-0.99). Indeed, all CHH men had an LH rise <2.0 iU/L following KP54, whereas all healthy men had an LH rise >4.0 iU/L. Anosmic men with CHH (i.e., Kallmann syndrome) had even lower LH rises after KP54 than did normosmic men with CHH (p = 0.017). Likewise, men identified to have pathogenic/likely pathogenic variants in CHH genes had even lower LH rises after KP54 than other men with CHH (p = 0.035). CONCLUSION: KP54 fully discriminated men with CHH from healthy men. Thus, KP54 could be used to specifically interrogate hypothalamic GnRH neuronal function in patients with CHH.


Assuntos
Hormônio Liberador de Gonadotropina/farmacologia , Hipogonadismo/sangue , Hipogonadismo/congênito , Hipogonadismo/diagnóstico , Kisspeptinas/farmacologia , Hormônio Luteinizante/sangue , Hormônio Luteinizante/efeitos dos fármacos , Adulto , Hormônio Liberador de Gonadotropina/administração & dosagem , Humanos , Síndrome de Kallmann/sangue , Síndrome de Kallmann/diagnóstico , Kisspeptinas/administração & dosagem , Masculino
6.
J Endocrinol Invest ; 44(11): 2445-2454, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33738751

RESUMO

PURPOSE: To investigate predictors of testicular response and non-reproductive outcomes (height, body proportions) after gonadotropin-induced puberty in congenital hypogonadotropic hypogonadism (CHH). DESIGN: A retrospective analysis of the puberty induction in CHH male patients, undergoing an off-label administration of combined gonadotropin (FSH and hCG). METHODS: Clinical and hormonal evaluations before and during gonadotropin stimulation in 19 CHH patients genotyped by Targeted Next Generation Sequencing for CHH genes; 16 patients underwent also semen analysis after gonadotropins. RESULTS: A lesser increase in testicular volume after 24 months of induction was significantly associated with: (I) cryptorchidism; (II) a positive genetic background; (III) a complete form of CHH. We found no significant correlation with the cumulative dose of hCG administered in 24 months. We found no association with the results of semen analyses, probably due to the low numerosity. Measures of body disproportion (eunuchoid habitus and difference between adult and target height: deltaSDSth), were significantly related to the: (I) age at the beginning of puberty induction; (II) duration of growth during the induction; (III) initial bone age. The duration of growth during induction was associated with previous testosterone priming and to partial forms of CHH. CONCLUSIONS: This study shows that a strong genetic background and cryptorchidism, as indicators of a complete GnRH deficiency since intrauterine life, are negative predictors of testicular response to gonadotropin stimulation in CHH. Body disproportion is associated with a delay in treatment and duration of growth during the induction, which is apparently inversely related to previous androgenization.


Assuntos
Estatura/efeitos dos fármacos , Gonadotropina Coriônica/uso terapêutico , Criptorquidismo , Hormônio Foliculoestimulante/uso terapêutico , Predisposição Genética para Doença , Hipogonadismo , Adulto , Criptorquidismo/diagnóstico , Criptorquidismo/etiologia , Relação Dose-Resposta a Droga , Disgenesia Gonadal/tratamento farmacológico , Disgenesia Gonadal/etiologia , Gonadotropinas/uso terapêutico , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Hipogonadismo/congênito , Hipogonadismo/genética , Hipogonadismo/terapia , Masculino , Puberdade/efeitos dos fármacos , Saúde Reprodutiva/estatística & dados numéricos , Análise do Sêmen/métodos , Análise do Sêmen/estatística & dados numéricos , Testículo , Tempo para o Tratamento/normas
7.
J Postgrad Med ; 67(2): 67-74, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33942770

RESUMO

Aims: To evaluate the prevalence of metabolic syndrome (MS) and whole-body composition in patients with congenital hypogonadism and investigate the effects of replacement therapy with testosterone undecanoate on MS, insulin resistance (IR), and whole-body composition in subset of patients. Methods: In a single arm prospective longitudinal intervention study, 33 patients with congenital hypogonadism, ages 20-39 years, were recruited and their parameters of MS, whole-body composition by DXA were compared with age and BMI matched healthy controls. In 21 patients, after 9 months we prospectively studied the effect (pre-post difference) of injection testosterone undecanoate (1,000 mg) replacement on MS, IR, and whole-body compositions. Results: The prevalence of MS was similar in patients and controls (27.3% vs. 9.1%, P = 0.05). Hypogonadism patients had higher prevalence of hypertension (33% vs. 3%, P < 0.01). Patients had decrease in lean body mass (P < 0.05) as compared to controls. After testosterone replacement, there was significant decrease in waist circumference (88.6 ± 13.1 cm vs. 83.9 ± 12.9 cm, P < 0.01), truncal fat (25.9 ± 7.3% vs. 24.0 ± 6.3%, P < 0.05), fasting C-peptide (2.1 ± 0.79 ng/ml vs. 0.68 ± 0.23 ng/ml, P < 0.01), serum proinsulin [1.43 (0.32-13.4) vs. 0.5 (0.5-3.2) pmol/l, P < 0.001] and a significant increase in lean body mass (46,906 ± 8,876 gm vs. 50,083 ± 7,590 gm, P < 0.001). Homeostasis model assessment of insulin resistance (HOMA-IR) (4.6 ± 1.7 vs. 0.5 ± 0.2, P < 0.001) and homeostatic model for assessment of insulin sensitivity (HOMA%S) [21 (12-65) vs. 206 (125-714), P < 0.001] were improved significantly following testosterone replacement. Conclusion: In this study, 36 weeks of testosterone replacement resulted in significant decrease in waist circumference, IR, truncal fat, total body fat and improvement in lean body mass, and insulin sensitivity.


Assuntos
Terapia de Reposição Hormonal , Hipogonadismo/tratamento farmacológico , Testosterona/uso terapêutico , Adulto , Índice de Massa Corporal , Seguimentos , Humanos , Hipogonadismo/congênito , Hipogonadismo/fisiopatologia , Resistência à Insulina , Estudos Longitudinais , Masculino , Síndrome Metabólica/epidemiologia , Pessoa de Meia-Idade , Estudos Prospectivos , Testosterona/farmacologia , Adulto Jovem
8.
Minerva Pediatr ; 72(6): 484-490, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32748610

RESUMO

The onset of puberty may be late - in the latter part of the predicted normal range or truly delayed - beyond this range. The latest age to start is usually regarded as 13 years in girls and 14 years in boys. There may also be a delayed completion of puberty, 16 years in girls and 17 years in boys. The initial approach requires a detailed history and clinical examination to exclude other medical or psychological problems. The presence or absence or pubertal signs should be documented. Investigations should be targeted at ruling out any medical causes and determining whether the delay is due to central gonadotropin deficiency (hypogonadotropic hypogonadism) or a gonadal disorder (hypergonadotropic hypogonadism). Physiological or constitutional delay of growth and puberty (CDGP) is more common in boys but is a diagnosis of exclusion. Current research suggests that CDGP and congenital hypogonadotropic hypogonadism have distinct genetic profiles which may aid in the differential diagnosis. Treatment may be given using low doses of sex steroids, testosterone or estradiol initially in a short course of 3-6 months but continuing in escalating doses mimicking the normal course of puberty, watching regularly for the spontaneous resumption of progress and gonadotropin secretion. In gonadotropin deficiency, sex hormone treatment needs to be continued until completion of pubertal development and growth. Counselling, reassurance and support are key elements in the management of adolescents with delayed puberty.


Assuntos
Hipogonadismo/complicações , Puberdade Tardia/etiologia , Adolescente , Adrenarca/fisiologia , Feminino , Gonadotropinas/deficiência , Transtornos do Crescimento/complicações , Humanos , Hipogonadismo/congênito , Hipogonadismo/genética , Masculino , Menarca/fisiologia , Puberdade/fisiologia , Puberdade Tardia/tratamento farmacológico , Fatores Sexuais
9.
Minerva Pediatr ; 72(4): 278-287, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32418410

RESUMO

Distinguishing between constitutional delay of growth and puberty (CDGP) and congenital hypogonadotropic hypogonadism (CHH) may be challenging. CDGP and CHH appear to belong to the same clinical spectrum (with low sex hormones and low LH and FSH), although one is classically transient and known as a self-limited form of delayed puberty (CDGP) while the other is permanent (CHH). Thus, the clinical history and the outcomes of these two conditions require different approaches, and an adequate and timely management for the patients is mandatory. Since the initial presentation of CDGP and CHH is almost identical and given the similarities of CDGP and partial forms of CHH (i.e. patients with partial and early interrupted pubertal development) the scientific community has been struggling to find some diagnostic tests able to allow an accurate differential diagnosis between these two conditions in delayed puberty. In this review we provide an up to date insight on the tests available, their meanings and accuracy, as well as some clues to effectively differentiate between constitutional pubertal delay and pathologic CHH.


Assuntos
Transtornos do Crescimento/diagnóstico , Hipogonadismo/diagnóstico , Puberdade Tardia/diagnóstico , Diagnóstico Diferencial , Feminino , Hormônio Foliculoestimulante/sangue , Humanos , Hipogonadismo/sangue , Hipogonadismo/congênito , Hipogonadismo/genética , Inibinas/sangue , Insulina/sangue , Kisspeptinas/sangue , Hormônio Luteinizante/sangue , Masculino , Proteínas , Puberdade Tardia/etiologia , Puberdade Tardia/genética , Receptores de Peptídeos/sangue , Receptores de Fatores de Crescimento Transformadores beta/sangue , Fatores Sexuais , Fatores de Tempo
10.
Hum Reprod ; 34(5): 863-871, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31067328

RESUMO

STUDY QUESTION: What is the peripubertal outcome of recombinant human FSH (r-hFSH) treatment during minipuberty in boys with congenital hypogonadotropic hypogonadism (CHH)? SUMMARY ANSWER: Sertoli-cell response to r-hFSH, given during the minipuberty of infancy, appears insufficient to maintain Sertoli cell function throughout childhood, as evaluated by inhibin B measurements. WHAT IS KNOWN ALREADY: Severe CHH in boys can be diagnosed during the minipuberty of infancy. Combined gonadotropin treatment at that age is suggested to improve testicular endocrine function and future fertility, yet long-term evidence is lacking. STUDY DESIGN, SIZE, DURATION: In this retrospective cohort study, we describe five CHH boys treated with r-hFSH in Helsinki University Hospital or Kuopio University Hospital between 2004 and 2018. Immediate follow-up data (0.1-1.4 months after cessation of the gonadotropin therapy) was available for four boys and long-term observations (at the age of 10.0-12.8 years) was available for three boys. As a retrospective control cohort, we provide inhibin B values of eight untreated CHH boys at the age of 12.7-17.8 years. PARTICIPANTS/MATERIALS, SETTING, METHODS: Four patients had combined pituitary hormone deficiency, and one had CHARGE syndrome due to a CHD7 mutation. The patients were treated at the age of 0.7-4.2 months with r-hFSH (3.4 IU/kg-7.5 IU/kg per week in 2 or 3 s.c. doses for 3-4.5 months) combined with T (25 mg i.m. monthly for three months for the treatment of micropenis). Inhibin B was chosen as the primary outcome measure. MAIN RESULTS AND THE ROLE OF CHANCE: During the r-hFSH + T treatment, inhibin B increased from 76 ± 18 ng/l to 176 ± 80 ng/l (P = 0.04) and penile length increased by 81 ± 50% (P = 0.04). Unexpectedly, two boys with robust inhibin B responses in infancy demonstrated low inhibin B values in peripuberty: declining from 290 ng/l (4 months) to 16 ng/l (12.4 years), and from 207 ng/l (6 months) to 21 ng/l (12.8 years). All boys underwent orchiopexy at 2.0 ± 0.7 years of age. Inhibin B values in long-term follow-up, available for the three boys, did not significantly differ from the untreated CHH controls. LIMITATIONS, REASONS FOR CAUTION: Limitations of this retrospective study are the small number and heterogeneity of the patients and their treatment schemes. WIDER IMPLICATIONS OF THE FINDINGS: We describe the first long-term follow-up data on CHH boys treated with r-hFSH and T as infants. The results from this small patient series suggest that the effects of infant r-hFSH treatment may be transient, and further longitudinal studies are required to determine the efficacy of this treatment approach to optimise the fertility potential in this patient population. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by the Finnish foundation for Pediatric Research, the Academy of Finland and the Emil Aaltonen Foundation. The authors have no competing interests. TRIAL REGISTRATION NUMBER: Non-applicable.


Assuntos
Gonadotropinas/deficiência , Hipogonadismo/tratamento farmacológico , Puberdade/efeitos dos fármacos , Células de Sertoli/efeitos dos fármacos , Adolescente , Criança , Pré-Escolar , Quimioterapia Combinada/métodos , Hormônio Foliculoestimulante Humano/administração & dosagem , Seguimentos , Gonadotropinas/sangue , Humanos , Hipogonadismo/sangue , Hipogonadismo/congênito , Hipogonadismo/diagnóstico , Lactente , Inibinas/sangue , Inibinas/metabolismo , Estudos Longitudinais , Masculino , Puberdade/sangue , Proteínas Recombinantes/administração & dosagem , Estudos Retrospectivos , Células de Sertoli/metabolismo , Índice de Gravidade de Doença , Testosterona/administração & dosagem , Resultado do Tratamento
11.
Hum Genomics ; 11(1): 1, 2017 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-28209183

RESUMO

BACKGROUND: Congenital hypogonadotrophic hypogonadism (CHH) and Kallmann syndrome (KS) are caused by disruption to the hypothalamic-pituitary-gonadal (H-P-G) axis. In particular, reduced production, secretion or action of gonadotrophin-releasing hormone (GnRH) is often responsible. Various genes, many of which play a role in the development and function of the GnRH neurons, have been implicated in these disorders. Clinically, CHH and KS are heterogeneous; however, in 46,XY patients, they can be characterised by under-virilisation phenotypes such as cryptorchidism and micropenis or delayed puberty. In rare cases, hypospadias may also be present. RESULTS: Here, we describe genetic mutational analysis of CHH genes in Indonesian 46,XY disorder of sex development patients with under-virilisation. We present 11 male patients with varying degrees of under-virilisation who have rare variants in known CHH genes. Interestingly, many of these patients had hypospadias. CONCLUSIONS: We postulate that variants in CHH genes, in particular PROKR2, PROK2, WDR11 and FGFR1 with CHD7, may contribute to under-virilisation phenotypes including hypospadias in Indonesia.


Assuntos
Hipogonadismo/congênito , Hipogonadismo/genética , Mutação , Adolescente , Criança , Pré-Escolar , Estudos de Coortes , DNA Helicases/genética , Proteínas de Ligação a DNA/genética , Hormônios Gastrointestinais/genética , Humanos , Hipogonadismo/patologia , Indonésia , Lactente , Masculino , Proteínas de Membrana/genética , Neuropeptídeos/genética , Proteínas Proto-Oncogênicas/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Peptídeos/genética
12.
Clin Endocrinol (Oxf) ; 86(3): 377-383, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27647266

RESUMO

OBJECTIVE: Men with congenital hypogonadotrophic hypogonadism (CHH) typically require lifelong hormonal therapy, and discontinuing treatment can have negative health consequences. Little is known about adherence to treatment or the psychosocial impact of CHH. DESIGN: A sequential, multiple methods approach was used. A quantitative online survey assessed adherence to treatment, depressive symptoms and illness perceptions. Subsequently, qualitative focus groups explored patient-reported factors for adherence. PATIENTS: Adult men with CHH on at least 1 year of treatment were recruited internationally. MEASUREMENTS: Adherence (Morisky medication adherence scale), depressive symptoms (Zung self-rating depression scale) and patient perception of CHH (revised illness perception questionnaire) were assessed in an online survey, and comparisons were made to reference groups. Patient focus group discussions were conducted and thematic analysis was employed to identify patient-reported factors for adherence. RESULTS: In total, 101 men on long-term treatment were included (mean age 37 ± 11 years). Forty three percent (43/101) exhibited low medication adherence and a significantly elevated prevalence of mild, moderate or severe depressive symptoms (27%, 17%, 20%, respectively, all P < 0·001 vs reference population). Patients reported negative illness perceptions and significant psychosocial consequences. Focus group discussions (n = 3, 26 total patients) identified patient-, health professional- and healthcare system-related barriers as targets for improving adherence. CONCLUSIONS: Congenital hypogonadotrophic hypogonadism men are challenged to adhere to long-term treatment. Poor adherence may contribute to adverse effects on bone, sexual and psychological health. The psychosocial morbidity of CHH is significant and appears to be underappreciated by healthcare providers.


Assuntos
Depressão/etiologia , Hipogonadismo/psicologia , Adesão à Medicação/psicologia , Adulto , Terapia de Reposição Hormonal , Humanos , Hipogonadismo/congênito , Hipogonadismo/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Psicologia , Inquéritos e Questionários
13.
Hum Reprod ; 32(3): 704-711, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28122887

RESUMO

STUDY QUESTION: What is the prevalence and functional consequence of ANOS1 (KAL1) mutations in a group of men with congenital hypogonadotropic hypogonadism (CHH)? SUMMARY ANSWER: Three of forty-two (7.1%) patients presented ANOS1 mutations, including a novel splice site mutation leading to exon skipping and a novel contiguous gene deletion associated with ichthyosis. WHAT IS KNOWN ALREADY: CHH is characterized by lack of pubertal development and infertility, due to deficient production, secretion or action of GnRH, and can be associated with anosmia/hyposmia (Kallmann syndrome, KS) or with a normal sense of smell (normosmic CHH). Mutations in the anosmin-1 (ANOS1) gene are responsible for the X-linked recessive form of KS. STUDY DESIGN, SIZE, DURATION: This cross-sectional study included 42 unrelated men with CHH (20 with KS and 22 with normosmic CHH). PARTICIPANTS/MATERIALS, SETTING, METHODS: Patients were screened for mutations in the ANOS1 gene by DNA sequencing. Identified mutations were further investigated by RT-PCR analysis and multiplex ligation-dependent probe amplification (MLPA) analysis. MAIN RESULTS AND THE ROLE OF CHANCE: Hemizygous mutations were identified in three (7.1%) KS cases: a novel splice acceptor site mutation (c.542-1G>C), leading to skipping of exon 5 in the ANOS1 transcript in a patient with self-reported normosmia (but hyposmic upon testing); a recurrent nonsense mutation (c.571C>T, p.Arg191*); and a novel 4.8 Mb deletion involving ANOS1 and eight other genes (VCX3B, VCX2, PNPLA4, VCX, STS, HDHD1, VCX3A and NLGN4X) in KS associated with ichthyosis. LIMITATIONS, REASONS FOR CAUTION: Objective olfactory testing was not performed in all cases of self-reported normosmia and this may have underestimated the olfactory deficits. WIDER IMPLICATIONS OF THE FINDINGS: This study further expands the spectrum of known genetic defects associated with CHH and suggests that patients with self-reported normal olfactory function should not be excluded from ANOS1 genetic testing. STUDY FUNDING/COMPETING INTEREST(S): This study was funded by the Portuguese Foundation for Science and Technology. The authors have no conflicts of interest. TRIAL REGISTRATION NUMBER: N/A.


Assuntos
Proteínas da Matriz Extracelular/genética , Hipogonadismo/genética , Síndrome de Kallmann/genética , Mutação , Proteínas do Tecido Nervoso/genética , Alelos , Estudos Transversais , Análise Mutacional de DNA , Éxons , Feminino , Frequência do Gene , Humanos , Hipogonadismo/congênito , Masculino , Linhagem
14.
Gynecol Endocrinol ; 33(9): 728-732, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28447505

RESUMO

In this retrospective multicenter cohort study, women with congenital hypogonadotrophic hypogonadism (CHH) (n = 57) who underwent intra-cytoplasmic sperm injection in-between 2010-2014 were compared to age-matched controls with tubal factor infertility (n = 114) to assess ovarian stimulation cycle and pregnancy outcomes. Live birth rates (LBRs) per started cycle were 31.6 and 24.6% in CHH and controls groups, respectively (p = 0.36). Comparable success rates were also confirmed with the logistic regression analysis (OR: 1.44, 95% CI: 0.78-2.67, p = 0.24). Of the 57 women with CHH, 19 were stimulated with the gonadotropin-releasing hormone (GnRH) antagonist protocol, 13 with the long-GnRH-agonist protocol. Pituitary suppression (PS) was not employed in the remaining 25 cases. Compared to women with PS, women without PS had significantly higher embryo implantation rates (21.6 versus 52.6%, p = 0.03). Although there was a trend favoring no PS, LBRs (25.0 versus 40.0%, p = 0.26) per cycle were short of statistical significance. LBRs per cycle (57.1 versus 31.2%, p = 0.11) and miscarriage rates (11.1 versus 16.7%, p = 0.75) were similar between CHH women who were given estrogen + progesterone and progesterone alone to support the luteal phase. In conclusion, the optimal stimulation protocol appears to be exogenous gonadotropin stimulation alone, without PS, and progesterone-only luteal phase support in CHH patients.


Assuntos
Coeficiente de Natalidade , Hipogonadismo/terapia , Infertilidade Feminina/terapia , Nascido Vivo , Injeções de Esperma Intracitoplásmicas , Adulto , Estudos de Coortes , Transferência Embrionária , Feminino , Fármacos para a Fertilidade Feminina/uso terapêutico , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/uso terapêutico , Humanos , Hipogonadismo/congênito , Infertilidade Feminina/congênito , Indução da Ovulação/métodos , Gravidez , Resultado do Tratamento , Adulto Jovem
15.
Toxicol Mech Methods ; 27(6): 451-457, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28413915

RESUMO

Patients with hypogonadism are at increased risk of cardiac and metabolic diseases. However, the pathogenesis of increased cardiometabolic risk in patients with hypogonadism is not clear. Oxidative stress plays an important role in the pathogenesis of cardiometabolic diseases. This study aimed to investigate possible differences in oxidative stress conditions between patients with hypogonadism and healthy controls. In this study, 38 male patients with congenital hypogonadotropic hypogonadism (CHH) (mean age: 21.7 ± 1.6 years) and 44 healthy male controls (mean age: 22.3 ± 1.4 years) with almost equal body mass index were enrolled. The demographic parameters, follicle-stimulating hormone (FSH), luteinizing hormone (LH), total and free testosterone, homeostatic model assessment of insulin resistance (HOMA-IR) and oxidative stress parameters, such as superoxide dismutase, catalase (CAT), glutathione peroxidase (GPx) and malondialdehyde (MDA), were compared between both groups. Compared to the healthy controls, triglycerides (p = .02), insulin levels, HOMA-IR values, CAT activities and MDA levels (p < .001 for all) were significantly higher and HDL cholesterol (p = .04), total and free testosterone, FSH, LH levels and GPx activity were significantly lower (p < .001 for all) in patients with CHH. There were significant correlations between total testosterone levels and CAT activity (r = -.33 p = .01), GPx activity (r = .36 p = .007) and MDA (r = -.47 p < .001) levels. The results of this study showed that young and treatment-naïve patients with congenital hypogonadism had an increased status of oxidative stress.


Assuntos
Catalase/sangue , Glutationa Peroxidase/sangue , Hipogonadismo/sangue , Malondialdeído/sangue , Estresse Oxidativo , Testosterona/sangue , Biomarcadores/sangue , Estudos de Casos e Controles , Eritrócitos/enzimologia , Hormônio Foliculoestimulante/sangue , Humanos , Hipogonadismo/congênito , Lipídeos/sangue , Hormônio Luteinizante/sangue , Masculino , Superóxido Dismutase/sangue , Adulto Jovem
16.
Gynecol Endocrinol ; 32(12): 947-950, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27809610

RESUMO

Turner syndrome and idiopathic congenital hypogonadism including Kallmann syndrome are conditions associated to a large number of widely known comorbidities that need a medical support forever. One of the characteristics shared by both conditions is the lack of sexual development that influencing the sexuality functioning and quality of life of the affected women. Few studies have been conducted to assess these topics, but they need to be considered in the treatment to all women with hypogonadism. This review on the major medical issues and psychological aspects, also focus in the present knowledge about sexual function and quality of life of women with Turner syndrome and idiopathic congenital hypogonadism, which aims to help in the comprehensive management of these patients.


Assuntos
Hipogonadismo/complicações , Infertilidade Feminina/etiologia , Qualidade de Vida , Disfunções Sexuais Fisiológicas/etiologia , Síndrome de Turner/complicações , Feminino , Humanos , Hipogonadismo/congênito , Hipogonadismo/psicologia , Infertilidade Feminina/psicologia , Disfunções Sexuais Fisiológicas/psicologia , Síndrome de Turner/psicologia
17.
Genet Med ; 17(8): 651-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25394172

RESUMO

PURPOSE: Congenital hypogonadotropic hypogonadism (CHH) and split hand/foot malformation (SHFM) are two rare genetic conditions. Here we report a clinical entity comprising the two. METHODS: We identified patients with CHH and SHFM through international collaboration. Probands and available family members underwent phenotyping and screening for FGFR1 mutations. The impact of identified mutations was assessed by sequence- and structure-based predictions and/or functional assays. RESULTS: We identified eight probands with CHH with (n = 3; Kallmann syndrome) or without anosmia (n = 5) and SHFM, seven of whom (88%) harbor FGFR1 mutations. Of these seven, one individual is homozygous for p.V429E and six individuals are heterozygous for p.G348R, p.G485R, p.Q594*, p.E670A, p.V688L, or p.L712P. All mutations were predicted by in silico analysis to cause loss of function. Probands with FGFR1 mutations have severe gonadotropin-releasing hormone deficiency (absent puberty and/or cryptorchidism and/or micropenis). SHFM in both hands and feet was observed only in the patient with the homozygous p.V429E mutation; V429 maps to the fibroblast growth factor receptor substrate 2α binding domain of FGFR1, and functional studies of the p.V429E mutation demonstrated that it decreased recruitment and phosphorylation of fibroblast growth factor receptor substrate 2α to FGFR1, thereby resulting in reduced mitogen-activated protein kinase signaling. CONCLUSION: FGFR1 should be prioritized for genetic testing in patients with CHH and SHFM because the likelihood of a mutation increases from 10% in the general CHH population to 88% in these patients.


Assuntos
Hipogonadismo/congênito , Hipogonadismo/genética , Deformidades Congênitas dos Membros/genética , Mutação , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Aminoácidos , Animais , Sequência Conservada , Feminino , Estudos de Associação Genética , Humanos , Hipogonadismo/metabolismo , Deformidades Congênitas dos Membros/metabolismo , Sistema de Sinalização das MAP Quinases , Masculino , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , Linhagem , Fosforilação , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo
18.
Endocr J ; 62(7): 605-13, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25924666

RESUMO

Patients with hypogonadism have poor cardiovascular and metabolic outcomes, and the effect of testosterone replacement therapy (TRT) is not clear. We investigated the presence of inflammation, insulin resistance and endothelial dysfunction in an unconfounded population of congenital hypogonadotrophic hypogonadism (CHH) and the effect of TRT on these subjects. A total of 60 patients with CHH (mean age 21.82±2.22 years) and 70 healthy control subjects (mean age 21.32±1.13 years) were enrolled. The demographic parameters, Asymmetric dimethylarginine (ADMA), TNF-like weak inducer of apoptosis (TWEAK), high sensitive C reactive protein (hs-CRP) and homeostatic model assessment of insulin resistance (HOMA-IR) levels were measured before and after TRT. The patients had higher Waist Circumferences (WC) (p=0.009), Diastolic Blood Pressures (p=0.02), Triglycerides (p=0.03), ADMA, insulin and HOMA-IR levels (p<0.001 for all) and lower TWEAK levels (p<0.001), compared to the healthy controls. After 5.56 ± 2.04 months of TRT, the patients had significantly elevated systolic blood pressures (p=0.01), body mass indexes and WC (p<0.001 and p=0.001 respectively) and decreased total and HDL cholesterol levels (p=0.032 and p<0.001 respectively). ADMA levels significantly increased (p=0.003), while the alterations in TWEAK, hsCRP and HOMA-IR were not significant. The results of the present study show that endothelial dysfunction, inflammation and insulin resistance are prevalent even in the very young subjects with CHH, who have no metabolic or cardiac problems at present. This increased cardiometabolic risk however, do not improve but even get worse after six months of TRT. Long term follow-up studies are warranted to investigate the unfavorable cardiometabolic effects of TRT.


Assuntos
Endotélio Vascular/fisiopatologia , Terapia de Reposição Hormonal , Hipogonadismo/fisiopatologia , Resistência à Insulina/fisiologia , Testosterona/uso terapêutico , Adulto , Glicemia , Índice de Massa Corporal , Endotélio Vascular/efeitos dos fármacos , Humanos , Hipogonadismo/sangue , Hipogonadismo/congênito , Hipogonadismo/tratamento farmacológico , Inflamação/tratamento farmacológico , Inflamação/fisiopatologia , Insulina/sangue , Masculino , Fatores de Risco , Testosterona/farmacologia , Resultado do Tratamento , Triglicerídeos/sangue , Circunferência da Cintura , Adulto Jovem
19.
Pediatr Res ; 75(5): 641-4, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24522099

RESUMO

BACKGROUND: Congenital hypogonadotropic hypogonadism (HH), a rare disorder characterized by absent, partial, or delayed puberty, can be caused by the lack or deficient number of hypothalamic gonadotropin-releasing hormone (GnRH) neurons. SEMA3A was recently implicated in the etiology of the disorder, and Sema7A-deficient mice have a reduced number of GnRH neurons in their brains. METHODS: SEMA3A and SEMA7A were screened by Sanger sequencing in altogether 50 Finnish HH patients (34 with Kallmann syndrome (KS; HH with hyposmia/anosmia) and 16 with normosmic HH (nHH)). In 20 patients, mutation(s) had already been found in genes known to be implicated in congenital HH. RESULTS: Three heterozygous variants (c.458A>G (p.Asn153Ser), c.1253A>G (p.Asn418Ser), and c.1303G>A (p.Val435Ile)) were found in SEMA3A in three KS patients, two of which also had a mutation in FGFR1. Two rare heterozygous variants (c.442C>T (p.Arg148Trp) and c.1421G>A (p.Arg474Gln)) in SEMA7A were found in one male nHH patient with a previously identified KISS1R nonsense variant and one male KS patient with a previously identified mutation in KAL1, respectively. CONCLUSION: Our results suggest that heterozygous missense variants in SEMA3A and SEMA7A may modify the phenotype of KS but most likely are not alone sufficient to cause the disorder.


Assuntos
Antígenos CD/genética , Hipogonadismo/congênito , Hipogonadismo/genética , Síndrome de Kallmann/genética , Semaforina-3A/genética , Semaforinas/genética , Análise Mutacional de DNA , Feminino , Finlândia , Proteínas Ligadas por GPI/genética , Variação Genética , Hormônio Liberador de Gonadotropina/metabolismo , Heterozigoto , Humanos , Masculino , Mutação , Mutação de Sentido Incorreto , Neurônios/metabolismo , Fenótipo
20.
Gynecol Endocrinol ; 30(2): 111-6, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24256370

RESUMO

OBJECTIVES: To define the patterns and causes of hearing decline associated to Turner's syndrome (TS). METHODS: An observational study with three cohorts was designed: 31 TS patients, 15 women with other congenital hypogonadims (OCH) and 41 healthy age-matched women taking contraception. Microotoscopy, standard pure-tone audiometry brain auditory evoked potentials (BAEP) were performed to study hearing function. RESULTS: Up to 87% of TS subjects suffered from some degree of hearing loss (HL) in the audiograms, compared with 20% OCH and 27% controls. Sensorineural hearing loss (SNHL) was the most frequent type of hypoacusia found in TS group. BAEP study demonstrated that 61% of TS women showed HL compared to 20% in OCH patients. No significant differences in latencies, amplitudes, and interpeaks of waves I, III and V were found between TS and OCH, nor when compared to reference population. Worse results were observed among the oldest TS patients, those with pure monosomy or isochromosome, and those with a history of recurrent otitis. CONCLUSIONS: More than a half of TS females presented HL. SNHL is the most frequent pattern among middle-aged women with TS. Old age, karyotype and recurrent otitis are predisposition factors to produce HL, while oestrogens play a minor role.


Assuntos
Perda Auditiva/complicações , Hipogonadismo/complicações , Síndrome de Turner/complicações , Adulto , Audiometria de Tons Puros , Feminino , Perda Auditiva/fisiopatologia , Humanos , Hipogonadismo/congênito , Hipogonadismo/fisiopatologia , Pessoa de Meia-Idade , Síndrome de Turner/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA