• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 4
  • 3
  • Tagged with
  • 8
  • 8
  • 4
  • 4
  • 4
  • 4
  • 3
  • 3
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

Investigation of the genetic regulation of delayed puberty

Howard, Sasha January 2017 (has links)
The genetic control of puberty remains an important but mostly unanswered question. Late pubertal timing affects over 2% of adolescents and is associated with adverse health outcomes. Self-limited delayed puberty (DP) segregates in an autosomal dominant pattern and is highly heritable; however, its neuroendocrine pathophysiology and genetic regulation remain unclear. The genetic control of puberty remains an important but mostly unanswered question. Late pubertal timing affects over 2% of adolescents and is associated with adverse health outcomes. Self-limited delayed puberty (DP) segregates in an autosomal dominant pattern and is highly heritable; however, its neuroendocrine pathophysiology and genetic regulation remain unclear. Our large, accurately phenotyped cohort of patients with familial self-limited DP is a unique resource with a relatively homogeneous genetic composition. I have utilised this cohort to investigate the genetic variants segregating with the DP trait in these pedigrees. Whole exome sequencing in eighteen probands and their relatives, and subsequent targeted sequencing in an extended subgroup of the cohort, has revealed potential novel genetic regulators of pubertal timing. In ten unrelated probands, I identified rare mutations in IGSF10, a gene that is strongly expressed in the nasal mesenchyme during embryonic migration of gonadotropin-releasing hormone (GnRH) neurons. IGSF10 knockdown both in vitro and in a transgenic zebrafish model resulted in perturbed GnRH neuronal migration. Loss-of-function mutations in IGSF10 were also identified in five patients with absent puberty due to hypogonadotropic hypogonadism (HH). Additionally, I have identified and investigated one rare, pathogenic mutation in HS6ST1 - a gene known to cause HH - in one family with DP, and two rare variants in FTO - a gene implicated in the timing of menarche in the general population - in 3 families. Further potentially pathogenic variants have emerged from investigating candidate genes identified from microarray studies (LGR4, SEMA6A and NEGR1) and from related clinical phenotypes (IGSF1). Our large, accurately phenotyped cohort of patients with familial self-limited DP is a unique resource with a relatively homogeneous genetic composition. I have utilised this cohort to investigate the genetic variants segregating with the DP trait in these pedigrees. Whole exome sequencing in eighteen probands and their relatives, and subsequent targeted sequencing in an extended subgroup of the cohort, has revealed potential novel genetic regulators of pubertal timing. In ten unrelated probands, I identified rare mutations in IGSF10, a gene that is strongly expressed in the nasal mesenchyme during embryonic migration of gonadotropin-releasing hormone (GnRH) neurons. IGSF10 knockdown both in vitro and in a transgenic zebrafish model resulted in perturbed GnRH neuronal migration. Loss-of-function mutations in IGSF10 were also identified in five patients with absent puberty due to hypogonadotropic hypogonadism (HH). Additionally, I have identified and investigated one rare, pathogenic mutation in HS6ST1 - a gene known to cause HH - in one family with DP, and two rare variants in FTO - a gene implicated in the timing of menarche in the general population - in 3 families. Further potentially pathogenic variants have emerged from investigating candidate genes identified from microarray studies (LGR4, SEMA6A and NEGR1) and from related clinical phenotypes (IGSF1).
2

Low Estrogen Model and Percent Lamellar Bone Pre and Post Puberty

Seigenfuse, Matthew David January 2010 (has links)
INTRODUCTION: Pubertal growth is an important time during development for bone accrual and attainment of peak bone mass. Suboptimal bone gain has been observed in females with reproductive abnormalities such as primary and secondary amenorrhea and these conditions are very prevalent in female athletes. Amenorrhea is associated with decreased estradiol levels. Previous research has shown that in prepubertal animals a low estrogen environment significantly decreased mechanical strength, but there was no significant loss in bone area and actually an increase in moment of inertia. The decrease in mechanical properties may be related to the microstructure of the bone. Two types of bone are involved in growth-- woven bone, which is added for structural support in the short term, and lamellar bone , which is highly organized and has a greater contribution to overall strength. We will test the hypotheses that suppressed estradiol will result in bones with no change in cortical area and decreased strength properties but will have a larger composition of non lamellar bone as opposed to lamellar bone. PURPOSE: The goal of this study was to determine the relative amounts of woven and lamellar tissue in a bone and the relationship with the bone's mechanical strength in two models of low estrogen-- pre- and post-pubertal onset. METHODS: Fifty-Five female Sprague-Dawley rats were randomly assigned into four groups: a control group (n=14) and three experimental groups injected with gonadotropin releasing-hormone antagonist (GnRH-a)-- the Dose 1 group was injected with 1.25 mg/kg/dose daily (n=14), the Dose 2 was injected with 2.5 mg/kg/dose daily (n=14), and the Dose 3 group was injected with 5.0 mg/kg/dose, 5 days per week (n=13). All groups were sacrificed at Day 49. Additionally, twenty-nine Sprague Dawley rats were randomly assigned into three groups. The baseline day 65 group (BL 65) was sacrificed on day 65 (n=9). There was an aged match control group that was sacrificed on day 90 (n=12). Finally, there was an AMEN experiment group injected with 2.5 mg/kg/dose daily that was sacrificed on day 90 (n=9). All experimental groups for both protocols received injections of gonadotropin releasing hormone antagonists (GnRH-a) (Zentaris GmbH) intraperitoneally. Left femora were mechanically tested under 3-point bending. The right femora were dehydrated, embedded in polymethylmethacrylate, cut and ground to 100 µm thickness. Bones were analyzed under polarized light using Stereo Investigator Software (MBF Bioscience, VT). The proportion of the cortex with primary lamellar vs. non-lamellar/other primary tissue type was measured and expressed as percent of the total cortical bone area. Outcome measures included lamellar endocortical area, lamellar periosteal area, cortical area, endocortical area, % lamellar area and % non-lamellar area. RESULTS: There was a significant decrease (p<.05) in the distribution of lamellar versus non-lamellar cortical tissue type in the experimental group in the model of delayed puberty. Additionally, the pre-pubertal bones had a lower percentage of lamellar periosteal and endocortical area. The post-pubertal group showed no significant differences between the control and experimental group in any of the outcome measures. CONCLUSION: There were significant differences in relative bone distribution throughout the femoral cortex. Relative decreases in lamellar tissue distribution, especially on the periosteal surface, will result in decreased mechanical strength due to increased percentage of woven bone in pre-pubertal models. / Kinesiology / Accompanied by one .pdf file: Lamellar/Woven Database.
3

IDENTIFICATION OF MECHANISMS OF DELAYED PUBERTY ON BONE STRENGTH DEFICITS DURING DEVELOPMENT

Joshi, Rupali Narayan January 2010 (has links)
Osteoporosis which is frequently referred to as a pediatric disease with geriatric consequences (Golden, 2000) can result from a lack of optimal bone accrual during the development (NIH Consensus Development Panel on Osteoporosis Prevention, Diagnosis, and Therapy, 2001). Pubertal timing is a key factor that contributes to optimal bone accrual and strength (Bonjour et al., 1994; 21 Warren et al., 2002). Bone mass doubles during the onset of puberty and young adulthood (Katzman et al., 1991) with more than 90% of peak bone mass being accrued at the end of second decade of life (Schneider & Wade, 2000). The rate of periosteal expansion is elevated during the pubertal period (Specker et al., 1987; Bradney et al., 2000) and this expansion parallels longitudinal growth (Parfitt, 1994). Irrespective of other changes, periosteal expansion lowers fracture risk by improving the strength of long bones by increasing the moment of inertia (Orwoll, 2003). Therefore, a delay in puberty may actually increase the time available for periosteal development and positively affect bone strength. Previous animal studies have shown decreases in strength, endocortical bone formation and increases in periosteal bone formation with delayed puberty. Clinical studies report negative effects of delayed puberty on bone mass accrual suggesting that delayed puberty is a multifactorial problem affecting bone strength development. The purpose of this study was to determine the effect of delayed puberty on mechanical strength and endocortical bone marrow cells in two models: female rats treated with gonadatropin releasing hormone antagonists (GnRH-a) and energy restriction (30%). Thirty-two female Sprague Dawley rats (21 to 22 days-of-age) were received from (Charles Rivers Laboratories, Wilmington, MA, USA) and housed individually at the Temple University Central Animal Facility (Temple University Weiss Hall). Animals were randomly assigned to one of three groups; control (n=10), GnRH-a (n=10) and energy restriction (ER) (n=12). The GnRH-a group was injected with gonadotropin releasing antagonist injections (GnRH-a) (Antide, Bachem, Torrance, Ca. USA) at a dose of 2.5 mg/kg/BW. The ER group received a 30% energy restricted diet (0pen Source diet (D07100606)(Research Diets, New Brunswick, NJ). All animals were sacrificed on Day 51. One way analysis of variance testing (ANOVA) with a significance level of 0.05 was used to assess group differences. Following the two protocols the uterine weight in the GnRH-a group was 80.6% lower than control; no change in the ER group. Ovarian weight was significantly lower in the GnRH-a group (83.3%) and in the ER group (33.3%) as compared to controls. A 22.7% lower muscle weight was found in the ER group but was equal to control and GnRH-a when normalized by body weight (BW). The retro-peritoneal fat pad weight was significantly decreased by 64.95% in the ER group as compared to controls. Energy restriction did not result in any deficit in bone strength when normalized by body weight however the GnRH-a group had a 26.2% lower bone strength compared to control. Histomorphometric changes were not significantly different between groups, but the ratio for periosteal versus endocortical bone formation rates for the control group was 1.38, GnRH-a was significantly higher with a ratio of 5.54 and for ER was 3.02 indicating that periosteal BFR are almost twice endocortical BFR in the experimental groups. There was a significant decrease in the trabecular percent bone volume (BV/TV) of the lumbar vertebra in the GnRH-a group (20.2%) compared to control. However BV/TV was significantly higher in the ER (18.4%) compared to the control group. Proliferation was suppressed to 59.6% of control in the GnRH-a group but only 85.5% of control in the ER group. The alkaline phosphatase activity was 31.2% lower in the GnRH-a group and 63.9% lower in the ER group. The relative quantification (RQ) of RUNX2 gene expression was lowest in control followed by GnRH-a and highest in ER group although no statistical significance was observed between any groups. Thus our data infers that 30% energy restriction does not negatively impact bone health. Thirty percent food restriction with no deficits in micronutrients or hormone suppression may just suppress growth as indicated by the maintenance of bone strength per body weight and equivalent muscle mass per body weight in the ER group compared to control. The GnRH-a injections resulted in decreased bone strength and trabecular bone volume. Female Athlete Triad or Anorexia Nervosa are the two clinical conditions hypothesized to result from a combination of ER and estrogen deficient environment. Studies replacing estrogen in hypothalamic amenorrhea or IGF-1 in anorexia alone have failed to improve bone mineral density (BMD), but a combination of IGF-1 and estrogen has been successful in improving BMD. This suggests that estrogen dependant and independent mechanisms work in combination to protect bone. Our study investigated both mechanisms separately and indicates that ER at 30% may be protective for bone health. Since estrogen deficiency may be the extreme end of the spectrum affecting trabecular bone, treatment therapies may have to be based on age, magnitude and severity of energy restriction and presence or absence of menstrual status. / Kinesiology
4

Novas perspectivas no estudo genético do hipogonadismo hipogonadotrófico isolado (HHI) por meio da técnica de sequenciamento paralelo em larga escala / New perspectives in the genetic study of congenital isolated hypogonadotrophic hypogonadism (IHH) using targeted next-generation sequencing

Amato, Lorena Guimarães Lima 03 August 2018 (has links)
O Hipogonadismo hipogonadotrófico isolado (HHI) congênito é uma síndrome clínica rara causada por defeito na produção ou secreção do hormônio liberador de gonadotrofinas (GnRH) pelo hipotálamo ou por resistência hipofisária à ação do GnRH. O HHI é mais prevalente em homens e cerca de 50% a 60% dos indivíduos afetados apresentam anosmia ou hiposmia associada, caracterizando a síndrome de Kallmann. Diversos genes já foram associados à patogênese do HHI congênito, porém, a maioria dos casos ainda permanece sem diagnóstico molecular definido. Até recentemente, a identificação das causas genéticas dos pacientes com HHI era realizada por sequenciamento de genes candidatos, empregando a técnica de Sanger. No entanto, com o número crescente de genes descritos nos últimos anos, esse processo vem se tornando impraticável. Novas metodologias de sequenciamento (sequenciamento paralelo em larga escala) foram desenvolvidas permitindo a genotipagem simultânea de diversas regiões, com maior velocidade e menor custo relativo. O atual projeto foi desenvolvido com o objetivo de rastrear genes candidatos em pacientes portadores de HHI congênito utilizando-se o sequenciamento paralelo em larga escala, visando ampliar o conhecimento genético do HHI. Realizamos o sequenciamento paralelo em larga escala (SPLE) de 130 pacientes com HHI congênito utilizando um painel contendo 36 genes relacionados ao HHI. Inicialmente, identificamos 104 variantes, potencialmente patogênicas em 77 pacientes (59,2%). Após a filtragem inicial, foi realizada uma análise individualizada de cada variante e com isso foram mantidos 41 (31,5%) pacientes com variantes classificadas como patogênicas ou provavelmente patogênicas. Os genes KAL1, FGFR1, CHD7 e GNRHR foram os mais frequentemente afetados. Esses resultados confirmam a importância dos genes classicamente associados ao HHI congênito. Destaca-se a alta prevalência de variantes no CHD7 (10,8%), gene bastante extenso, levando à dificuldade técnica de sequenciá-lo pelos métodos tradicionais, até então sem estudos nessa coorte. O CHD7 é um gene originalmente associado à complexa síndrome de CHARGE, porém, nos últimos anos vem sendo cada vez mais associados ao HHI congênito. Dentre os resultados, ressaltamos a identificação de uma mutação inédita no gene GNRH1, causa rara de HHI, e a identificação de variantes deletérias no gene IGSF10, recentemente descrito associado ao atraso puberal, mas sem papel claro no fenótipo de HHI, em dois pacientes que tiveram reversibilidade do hipogonadismo. Variantes provavelmente patogênicas em genes com poucas descrições ou até mesmo sem relatos de associação ao fenótipo de HHI (SPRY4, FLRT3, IGSF1, NSMF, SOX10 e OTX2) também foram identificadas nessa coorte, ampliando nosso conhecimento genético do HHI. A oligogenicidade, previamente descrita com prevalência de 2,5% a 7%, em nosso estudo esteve presente em 22% dos pacientes, demonstrando uma ampliação das descrições de oligogenicidade quando comparados aos estudos prévios utilizando somente a técnica de Sanger. A nova técnica de sequenciamento genético (SPLE), utilizada em nosso estudo, foi capaz de ampliar de 22% para 31,5% (41 em 130 pacientes) a porcentagem de pacientes com diagnóstico molecular definido, quando comparado aos dados prévios utilizando a técnica de Sanger, mostrando-se rápida, confiável e eficaz / Congenital isolated hypogonadotropic hypogonadism (IHH) is a rare condition caused by GnRH deficiency, due to defective hypothalamic gonadotropin-releasing hormone (GnRH) production or secretion, or by pituitary resistance to the GnRH action. Congenital IHH is more prevalent in men and about 50% to 60% of affected individuals present with associated anosmia or hyposmia, characterizing Kallmann\'s syndrome. Several genes have already been associated with the pathogenesis of congenital IHH, but most cases still remain without a molecular diagnosis. Until recently, identification of the genetic causes of IHH was performed by sequencing candidate genes using the Sanger technique. However, with the growing number of genes and the genetic complexity of IHH, it has become almost impossible to keep the screening of all candidate genes updated using the traditional techniques. The advent of next-generation sequencing (NGS) has allowed the simultaneous genotyping of several regions, faster and with lower relative cost. The present project was developed with the objective of tracking candidate genes in patients with congenital IHH using large-scale parallel sequencing, in aiming to increase the genetic knowledge of this rare condition. A total of 130 unrelated patients with IHH was studied by targeted NGS, using a panel containing 36 IHH associated genes. Initially, 104 potentially pathogenic variants were identified in 77 patients (59.2%). However, after an individualized analysis of each variant, the number of patients considered to carry pathogenic or probably pathogenic variants dropped to 41 (31.5%). The genes KAL1, FGFR1, CHD7 and GNRHR were the most frequently affected and these results confirm the importance of genes classically associated with IHH. It is noteworthy the high prevalence of variants in CHD7 (10.8%), a rather extensive gene, leading to technical difficulty of sequencing by traditional methods, which had not been studied in this cohort. CHD7 is the causative gene of CHARGE syndrome, however it has been recently identified in a growing number of congenital IHH patients with or without additional features of the syndrome. Among the results, we emphasize a novel mutation in the GNRH1 gene, a rare cause of IHH, and the identification of deleterious variants in the IGSF10 gene, recently associated with pubertal delay but without a clear role in the IHH phenotype, in two patients with reversible hypogonadism. Probably pathogenic variants in genes with few descriptions or even no reports of association with the IHH phenotype (SPRY4, FLRT3, IGSF1, NSMF, SOX10 and OTX2) were also identified in this cohort, increasing the genetic knowledge of IHH. Oligogenicity, previously described with a prevalence of 2.5% to 7%, was observed in 22% of our patients, demonstrating an increase in oligogenicity cases when compared to previous studies using only the Sanger sequencing. In conclusion, targeted NGS was able to increase the percentage of patients with molecular diagnosis from 22% to 31.5% in our cohort when compared to the previous data using the Sanger sequencing, and has been shown to be a fast, reliable and effective tool in the molecular diagnosis of congenital IHH
5

Novas perspectivas no estudo genético do hipogonadismo hipogonadotrófico isolado (HHI) por meio da técnica de sequenciamento paralelo em larga escala / New perspectives in the genetic study of congenital isolated hypogonadotrophic hypogonadism (IHH) using targeted next-generation sequencing

Lorena Guimarães Lima Amato 03 August 2018 (has links)
O Hipogonadismo hipogonadotrófico isolado (HHI) congênito é uma síndrome clínica rara causada por defeito na produção ou secreção do hormônio liberador de gonadotrofinas (GnRH) pelo hipotálamo ou por resistência hipofisária à ação do GnRH. O HHI é mais prevalente em homens e cerca de 50% a 60% dos indivíduos afetados apresentam anosmia ou hiposmia associada, caracterizando a síndrome de Kallmann. Diversos genes já foram associados à patogênese do HHI congênito, porém, a maioria dos casos ainda permanece sem diagnóstico molecular definido. Até recentemente, a identificação das causas genéticas dos pacientes com HHI era realizada por sequenciamento de genes candidatos, empregando a técnica de Sanger. No entanto, com o número crescente de genes descritos nos últimos anos, esse processo vem se tornando impraticável. Novas metodologias de sequenciamento (sequenciamento paralelo em larga escala) foram desenvolvidas permitindo a genotipagem simultânea de diversas regiões, com maior velocidade e menor custo relativo. O atual projeto foi desenvolvido com o objetivo de rastrear genes candidatos em pacientes portadores de HHI congênito utilizando-se o sequenciamento paralelo em larga escala, visando ampliar o conhecimento genético do HHI. Realizamos o sequenciamento paralelo em larga escala (SPLE) de 130 pacientes com HHI congênito utilizando um painel contendo 36 genes relacionados ao HHI. Inicialmente, identificamos 104 variantes, potencialmente patogênicas em 77 pacientes (59,2%). Após a filtragem inicial, foi realizada uma análise individualizada de cada variante e com isso foram mantidos 41 (31,5%) pacientes com variantes classificadas como patogênicas ou provavelmente patogênicas. Os genes KAL1, FGFR1, CHD7 e GNRHR foram os mais frequentemente afetados. Esses resultados confirmam a importância dos genes classicamente associados ao HHI congênito. Destaca-se a alta prevalência de variantes no CHD7 (10,8%), gene bastante extenso, levando à dificuldade técnica de sequenciá-lo pelos métodos tradicionais, até então sem estudos nessa coorte. O CHD7 é um gene originalmente associado à complexa síndrome de CHARGE, porém, nos últimos anos vem sendo cada vez mais associados ao HHI congênito. Dentre os resultados, ressaltamos a identificação de uma mutação inédita no gene GNRH1, causa rara de HHI, e a identificação de variantes deletérias no gene IGSF10, recentemente descrito associado ao atraso puberal, mas sem papel claro no fenótipo de HHI, em dois pacientes que tiveram reversibilidade do hipogonadismo. Variantes provavelmente patogênicas em genes com poucas descrições ou até mesmo sem relatos de associação ao fenótipo de HHI (SPRY4, FLRT3, IGSF1, NSMF, SOX10 e OTX2) também foram identificadas nessa coorte, ampliando nosso conhecimento genético do HHI. A oligogenicidade, previamente descrita com prevalência de 2,5% a 7%, em nosso estudo esteve presente em 22% dos pacientes, demonstrando uma ampliação das descrições de oligogenicidade quando comparados aos estudos prévios utilizando somente a técnica de Sanger. A nova técnica de sequenciamento genético (SPLE), utilizada em nosso estudo, foi capaz de ampliar de 22% para 31,5% (41 em 130 pacientes) a porcentagem de pacientes com diagnóstico molecular definido, quando comparado aos dados prévios utilizando a técnica de Sanger, mostrando-se rápida, confiável e eficaz / Congenital isolated hypogonadotropic hypogonadism (IHH) is a rare condition caused by GnRH deficiency, due to defective hypothalamic gonadotropin-releasing hormone (GnRH) production or secretion, or by pituitary resistance to the GnRH action. Congenital IHH is more prevalent in men and about 50% to 60% of affected individuals present with associated anosmia or hyposmia, characterizing Kallmann\'s syndrome. Several genes have already been associated with the pathogenesis of congenital IHH, but most cases still remain without a molecular diagnosis. Until recently, identification of the genetic causes of IHH was performed by sequencing candidate genes using the Sanger technique. However, with the growing number of genes and the genetic complexity of IHH, it has become almost impossible to keep the screening of all candidate genes updated using the traditional techniques. The advent of next-generation sequencing (NGS) has allowed the simultaneous genotyping of several regions, faster and with lower relative cost. The present project was developed with the objective of tracking candidate genes in patients with congenital IHH using large-scale parallel sequencing, in aiming to increase the genetic knowledge of this rare condition. A total of 130 unrelated patients with IHH was studied by targeted NGS, using a panel containing 36 IHH associated genes. Initially, 104 potentially pathogenic variants were identified in 77 patients (59.2%). However, after an individualized analysis of each variant, the number of patients considered to carry pathogenic or probably pathogenic variants dropped to 41 (31.5%). The genes KAL1, FGFR1, CHD7 and GNRHR were the most frequently affected and these results confirm the importance of genes classically associated with IHH. It is noteworthy the high prevalence of variants in CHD7 (10.8%), a rather extensive gene, leading to technical difficulty of sequencing by traditional methods, which had not been studied in this cohort. CHD7 is the causative gene of CHARGE syndrome, however it has been recently identified in a growing number of congenital IHH patients with or without additional features of the syndrome. Among the results, we emphasize a novel mutation in the GNRH1 gene, a rare cause of IHH, and the identification of deleterious variants in the IGSF10 gene, recently associated with pubertal delay but without a clear role in the IHH phenotype, in two patients with reversible hypogonadism. Probably pathogenic variants in genes with few descriptions or even no reports of association with the IHH phenotype (SPRY4, FLRT3, IGSF1, NSMF, SOX10 and OTX2) were also identified in this cohort, increasing the genetic knowledge of IHH. Oligogenicity, previously described with a prevalence of 2.5% to 7%, was observed in 22% of our patients, demonstrating an increase in oligogenicity cases when compared to previous studies using only the Sanger sequencing. In conclusion, targeted NGS was able to increase the percentage of patients with molecular diagnosis from 22% to 31.5% in our cohort when compared to the previous data using the Sanger sequencing, and has been shown to be a fast, reliable and effective tool in the molecular diagnosis of congenital IHH
6

Triple A syndrome with a novel indel mutation in the AAAS gene and delayed puberty: Patient report

Bustanji, Haidar, Sahar, Bashar, Hübner, Angela, Ajlouni, Kamel, Landgraf, Dana, Hamamy, Hanan, Koehler, Katrin 23 June 2020 (has links)
Triple A syndrome, formerly known as Allgrove syndrome, is an autosomal recessive disorder characterized clinically by adrenal insufficiency, alacrima, achalasia, and neurological abnormalities. We report a 17-year-old boy presented to the endocrine clinic with delayed puberty and a 4-year’s history of fatigue and muscle weakness. He had achalasia, alacrima, and skin and mucosal hyperpigmentation. Hormonal assessment revealed isolated glucocorticoid deficiency. Clinical diagnosis of triple A syndrome was confirmed by sequencing the entire coding region including exon-intron boundaries of the AAAS gene. Analysis revealed a homozygous novel indel mutation encompassing intron 7 to intron 10 of the gene (g.16166_17813delinsTGAGGCCTGCTG; NG_016775). This is the first report of triple A syndrome in Jordan with a novel indel mutation and presenting with delayed puberty.
7

Pesquisa de mutações no gene do receptor do secretagogo de hormônio de crescimento (GHSR) em crianças com baixa estatura idiopática e deficiência isolada de hormônio de crescimento / Growth hormone secretatogue receptor gene (GHSR) analysis in patients with idiopathic short stature (ISS) and patients with isolated growth hormone deficiency

Pires, Patrícia Nascimbem Pugliese 10 October 2011 (has links)
A ghrelina, hormônio secretado principalmente por células gástricas, liga-se ao seu receptor, o receptor de secretagogo de GH (GHSR - Growth hormone secretagogue receptor), localizado no hipotálamo e na hipófise, estimulando a síntese e secreção do GH. Recentemente foram identificadas mutações no gene GHSR em crianças com baixa estatura idiopática (BEI) e com deficiência isolada de GH (DGH). No presente estudo investigamos a presença de mutações no gene GHSR em crianças com DGH isolada de causa não identificada e crianças com BEI, incluindo um subgrupo de crianças com atraso constitucional de crescimento e desenvolvimento (ACCD). Foram selecionados 14 pacientes com deficiência isolada de GH sem alterações anatômicas da região hipotálamo-hipofisária e 96 pacientes com BEI, destes 31 (32%) apresentavam ACCD. Também foram estudados 150 controles adultos e 197 crianças controle com crescimento e puberdade normais. A região codificadora do GHSR foi amplificada utilizando-se oligonucleotídeos iniciadores específicos, seguida de purificação enzimática e seqüenciamento automático. Encontramos 6 variantes alélicas em heterozigose no GHSR: nenhuma delas presente nos controles estudados, e quatro destas variantes estão localizadas em regiões conservadas do gene. Uma variante foi encontrada em uma paciente do grupo DGH (p.Val249Leu) e as outras cinco (c.-6 G>C, p.Ser84Ile, p.Val182Ala, p.Ala169Thr e p.Ala358Thr) foram encontradas em pacientes do subgrupo ACCD do grupo BEI. As variantes missense foram submetidas a estudo funcional que evidenciou que as mutações p.Ser84Ile e p.Val182Ala possuem diminuição na atividade basal associadas à diminuição da expressão do receptor na superfície celular. Adicionalmente, a mutação p.Ser84Ile também apresenta redução na atividade do GHSR induzida pelo ligante. A variante p.Val249Leu foi encontrada em uma paciente do sexo feminino com diagnóstico de DGH isolado. A falta de segregação familiar associada à ausência de déficit funcional da variante nos estudos in vitro sugere que, neste caso, a variante p.Val249Leu não é a causa do fenótipo de DGH nesta família e trata-se de uma variante alélica rara. As 5 variantes alélicas no GHSR (c.-6 G>C, p.Ser84Ile, p.Val182Ala, p.Ala169Thr e p.Ala358Thr) encontradas nos pacientes com BEI foram identificadas apenas naqueles com puberdade atrasada, ou seja, pertencentes ao subgrupo ACCD (3 do sexo masculino e 2 do sexo feminino). A freqüência de variantes neste grupo de pacientes foi de 16%, significativamente maior que nos outros grupos, e a ausência de variantes gênicas novas no grupo de crianças obesas com altura normal e mesmo no grupo de crianças com BEI sem ACCD sugere que nosso achado não foi casual e que as alterações descritas podem estar associadas ao fenótipo de ACCD. Os estudos in vitro mostraram prejuízos funcionais em 2 destas variantes (p.Ser84Ile e p.Val182Ala) porém, devido à limitação dos estudos funcionais (celulas heterólogas) não podemos afastar que as demais não tenham algum impacto funcional in vivo. Em conclusão, nossos resultados sugerem um envolvimento dos defeitos no GHSR na etiologia do atraso constitucional do crescimento e desenvolvimento em uma parcela de pacientes com esta condição / Ghrelin, hormone secreted by gastric cells, stimulates growth hormone secretion by acting on its receptor GHSR, located in the hypothalamus and pituitary. Recently, mutations in the GHSR gene were described in patients with growth hormone deficiency (GHD) and idiopathic short stature (ISS). In the present study we analyzed the GHSR gene in patients with isolated GHD and patients with ISS, including a subgroup of patients with constitutional delay of growth and puberty (CDGP). We studied 14 GHD patients with normal pituitary magnetic resonance imaging and 96 patients with ISS, 31 of them with CDGP. We also studied 150 adults and in 197 children with normal stature. The entire coding region as well as the exon-intron boundaries of GHSR were PCR amplified in all patients and control group and PCR products were bidirectionally sequenced. Six different heterozygous variants in GHSR were identified: none of them were found in the control group and four of these amino acid substitutions occurred at a conserved position within the GHSR. One variant (p.Val249Leu) was found in a GHD patient and the other five (c.-6 G>C, p.Ser84Ile, p.Val182Ala, p.Ala169Thr e p.Ala358Thr) were found in patients with CDGP. The missense variants were submitted to functional studies. Two of these variants (p.Ser84Ile and p.Val182Ala) result in a decrease in basal activity that was in part explained by a reduction in cell surface expression. The p.Ser84Ile mutation was also associated with a defect in ghrelin potency. The p.Val249Leu variant, found in a female patient with isolated GHD, did not segregate with the phenotype in the family and had no functional impairment in vitro. This suggests that p.Val249Leu is not the cause of the GHD in the family and may be a rare allelic variant. The other variants (c.-6 G>C, p.Ser84Ile, p.Val182Ala, p.Ala169Thr e p.Ala358Thr) were identified only in patients with CDGP (3 male and 2 female). The frequency of allelic variants observed in this group (16%) was higher than expected by chance in contrast with ISS and GHD children, and the absence of other GHSR mutations in the large group of control children suggests that the association between GHSR mutations and CDGP phenotype is unlikely to be fortuitous. Functional studies revealed that two of the identified missense variants (p.Ser84Ile and p.Val182Ala) are functionally significant. These functional studies were performed in heterologous cell expression systems; therefore it is not possible to completely rule out that the other identified variants might cause some unrevealed impairment on GHSR function or expression in vivo. In conclusion, our data raise the possibility that abnormalities in ghrelin receptor function may be implicated in the ethiology of CDGP in some patients
8

Pesquisa de mutações no gene do receptor do secretagogo de hormônio de crescimento (GHSR) em crianças com baixa estatura idiopática e deficiência isolada de hormônio de crescimento / Growth hormone secretatogue receptor gene (GHSR) analysis in patients with idiopathic short stature (ISS) and patients with isolated growth hormone deficiency

Patrícia Nascimbem Pugliese Pires 10 October 2011 (has links)
A ghrelina, hormônio secretado principalmente por células gástricas, liga-se ao seu receptor, o receptor de secretagogo de GH (GHSR - Growth hormone secretagogue receptor), localizado no hipotálamo e na hipófise, estimulando a síntese e secreção do GH. Recentemente foram identificadas mutações no gene GHSR em crianças com baixa estatura idiopática (BEI) e com deficiência isolada de GH (DGH). No presente estudo investigamos a presença de mutações no gene GHSR em crianças com DGH isolada de causa não identificada e crianças com BEI, incluindo um subgrupo de crianças com atraso constitucional de crescimento e desenvolvimento (ACCD). Foram selecionados 14 pacientes com deficiência isolada de GH sem alterações anatômicas da região hipotálamo-hipofisária e 96 pacientes com BEI, destes 31 (32%) apresentavam ACCD. Também foram estudados 150 controles adultos e 197 crianças controle com crescimento e puberdade normais. A região codificadora do GHSR foi amplificada utilizando-se oligonucleotídeos iniciadores específicos, seguida de purificação enzimática e seqüenciamento automático. Encontramos 6 variantes alélicas em heterozigose no GHSR: nenhuma delas presente nos controles estudados, e quatro destas variantes estão localizadas em regiões conservadas do gene. Uma variante foi encontrada em uma paciente do grupo DGH (p.Val249Leu) e as outras cinco (c.-6 G>C, p.Ser84Ile, p.Val182Ala, p.Ala169Thr e p.Ala358Thr) foram encontradas em pacientes do subgrupo ACCD do grupo BEI. As variantes missense foram submetidas a estudo funcional que evidenciou que as mutações p.Ser84Ile e p.Val182Ala possuem diminuição na atividade basal associadas à diminuição da expressão do receptor na superfície celular. Adicionalmente, a mutação p.Ser84Ile também apresenta redução na atividade do GHSR induzida pelo ligante. A variante p.Val249Leu foi encontrada em uma paciente do sexo feminino com diagnóstico de DGH isolado. A falta de segregação familiar associada à ausência de déficit funcional da variante nos estudos in vitro sugere que, neste caso, a variante p.Val249Leu não é a causa do fenótipo de DGH nesta família e trata-se de uma variante alélica rara. As 5 variantes alélicas no GHSR (c.-6 G>C, p.Ser84Ile, p.Val182Ala, p.Ala169Thr e p.Ala358Thr) encontradas nos pacientes com BEI foram identificadas apenas naqueles com puberdade atrasada, ou seja, pertencentes ao subgrupo ACCD (3 do sexo masculino e 2 do sexo feminino). A freqüência de variantes neste grupo de pacientes foi de 16%, significativamente maior que nos outros grupos, e a ausência de variantes gênicas novas no grupo de crianças obesas com altura normal e mesmo no grupo de crianças com BEI sem ACCD sugere que nosso achado não foi casual e que as alterações descritas podem estar associadas ao fenótipo de ACCD. Os estudos in vitro mostraram prejuízos funcionais em 2 destas variantes (p.Ser84Ile e p.Val182Ala) porém, devido à limitação dos estudos funcionais (celulas heterólogas) não podemos afastar que as demais não tenham algum impacto funcional in vivo. Em conclusão, nossos resultados sugerem um envolvimento dos defeitos no GHSR na etiologia do atraso constitucional do crescimento e desenvolvimento em uma parcela de pacientes com esta condição / Ghrelin, hormone secreted by gastric cells, stimulates growth hormone secretion by acting on its receptor GHSR, located in the hypothalamus and pituitary. Recently, mutations in the GHSR gene were described in patients with growth hormone deficiency (GHD) and idiopathic short stature (ISS). In the present study we analyzed the GHSR gene in patients with isolated GHD and patients with ISS, including a subgroup of patients with constitutional delay of growth and puberty (CDGP). We studied 14 GHD patients with normal pituitary magnetic resonance imaging and 96 patients with ISS, 31 of them with CDGP. We also studied 150 adults and in 197 children with normal stature. The entire coding region as well as the exon-intron boundaries of GHSR were PCR amplified in all patients and control group and PCR products were bidirectionally sequenced. Six different heterozygous variants in GHSR were identified: none of them were found in the control group and four of these amino acid substitutions occurred at a conserved position within the GHSR. One variant (p.Val249Leu) was found in a GHD patient and the other five (c.-6 G>C, p.Ser84Ile, p.Val182Ala, p.Ala169Thr e p.Ala358Thr) were found in patients with CDGP. The missense variants were submitted to functional studies. Two of these variants (p.Ser84Ile and p.Val182Ala) result in a decrease in basal activity that was in part explained by a reduction in cell surface expression. The p.Ser84Ile mutation was also associated with a defect in ghrelin potency. The p.Val249Leu variant, found in a female patient with isolated GHD, did not segregate with the phenotype in the family and had no functional impairment in vitro. This suggests that p.Val249Leu is not the cause of the GHD in the family and may be a rare allelic variant. The other variants (c.-6 G>C, p.Ser84Ile, p.Val182Ala, p.Ala169Thr e p.Ala358Thr) were identified only in patients with CDGP (3 male and 2 female). The frequency of allelic variants observed in this group (16%) was higher than expected by chance in contrast with ISS and GHD children, and the absence of other GHSR mutations in the large group of control children suggests that the association between GHSR mutations and CDGP phenotype is unlikely to be fortuitous. Functional studies revealed that two of the identified missense variants (p.Ser84Ile and p.Val182Ala) are functionally significant. These functional studies were performed in heterologous cell expression systems; therefore it is not possible to completely rule out that the other identified variants might cause some unrevealed impairment on GHSR function or expression in vivo. In conclusion, our data raise the possibility that abnormalities in ghrelin receptor function may be implicated in the ethiology of CDGP in some patients

Page generated in 0.4458 seconds