Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hum Mol Genet ; 29(14): 2435-2450, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32620954

ABSTRACT

Dysfunction of the gonadotropin-releasing hormone (GnRH) axis causes a range of reproductive phenotypes resulting from defects in the specification, migration and/or function of GnRH neurons. To identify additional molecular components of this system, we initiated a systematic genetic interrogation of families with isolated GnRH deficiency (IGD). Here, we report 13 families (12 autosomal dominant and one autosomal recessive) with an anosmic form of IGD (Kallmann syndrome) with loss-of-function mutations in TCF12, a locus also known to cause syndromic and non-syndromic craniosynostosis. We show that loss of tcf12 in zebrafish larvae perturbs GnRH neuronal patterning with concomitant attenuation of the orthologous expression of tcf3a/b, encoding a binding partner of TCF12, and stub1, a gene that is both mutated in other syndromic forms of IGD and maps to a TCF12 affinity network. Finally, we report that restored STUB1 mRNA rescues loss of tcf12 in vivo. Our data extend the mutational landscape of IGD, highlight the genetic links between craniofacial patterning and GnRH dysfunction and begin to assemble the functional network that regulates the development of the GnRH axis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Gonadotropin-Releasing Hormone/genetics , Kallmann Syndrome/genetics , Ubiquitin-Protein Ligases/genetics , Zebrafish Proteins/genetics , Adult , Aged , Animals , Disease Models, Animal , Female , Genes, Dominant/genetics , Gonadotropin-Releasing Hormone/deficiency , Haploinsufficiency/genetics , Humans , Kallmann Syndrome/pathology , Male , Middle Aged , Mutation/genetics , Neurons/metabolism , Neurons/pathology , Phenotype , Zebrafish/genetics
2.
Hum Mol Genet ; 27(8): 1343-1352, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29432577

ABSTRACT

CHARGE syndrome is an autosomal dominant malformation disorder caused by heterozygous loss of function mutations in the chromatin remodeler CHD7. Chd7 regulates the expression of Sema3a, which also contributes to the pathogenesis of Kallmann syndrome, a heterogeneous condition with the typical features hypogonadotropic hypogonadism and an impaired sense of smell. Both features are common in CHARGE syndrome suggesting that SEMA3A may provide a genetic link between these syndromes. Indeed, we find evidence that SEMA3A plays a role in the pathogenesis of CHARGE syndrome. First, Chd7 is enriched at the Sema3a promotor in neural crest cells and loss of function of Chd7 inhibits Sema3a expression. Second, using a Xenopus CHARGE model, we show that human SEMA3A rescues Chd7 loss of function. Third, to elucidate if SEMA3A mutations in addition to CHD7 mutations also contribute to the severity of the CHARGE phenotype, we screened 31 CHD7-positive patients and identified one patient with a heterozygous non-synonymous SEMA3A variant, c.2002A>G (p.I668V). By analyzing protein expression and processing, we did not observe any differences of the p.I668V variant compared with wild-type SEMA3A, while a pathogenic SEMA3A variant p.R66W recently described in a patient with Kallmann syndrome did affect protein secretion. Furthermore, the p.I668V variant, but not the pathogenic p.R66W variant, rescues Chd7 loss of function in Xenopus, indicating that the p.I668V variant is likely benign. Thus, SEMA3A is part of an epigenetic loop that plays a role in the pathogenesis of CHARGE syndrome, however, it seems not to act as a common direct modifier.


Subject(s)
CHARGE Syndrome/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Epigenesis, Genetic , Neural Crest/metabolism , Semaphorin-3A/genetics , Animals , CHARGE Syndrome/metabolism , CHARGE Syndrome/pathology , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Disease Models, Animal , Embryo, Nonmammalian , Genetic Complementation Test , HEK293 Cells , Homeobox Protein Nkx-2.5/genetics , Homeobox Protein Nkx-2.5/metabolism , Humans , Kallmann Syndrome/genetics , Kallmann Syndrome/metabolism , Kallmann Syndrome/pathology , Mutation , Neural Crest/pathology , Promoter Regions, Genetic , Semaphorin-3A/metabolism , Severity of Illness Index , Xenopus laevis
3.
Clin Genet ; 97(5): 696-703, 2020 05.
Article in English | MEDLINE | ID: mdl-32060892

ABSTRACT

Isolated hypogonadotropic hypogonadism (IHH) is a rare disorder characterized by impaired sexual development and infertility, caused by the deficiency of hypothalamic gonadotropin-releasing hormone neurons. IHH is named Kallmann's syndrome (KS) or normosmic IHH (nIHH) when associated with a defective or normal sense of smell. Variants in SEMA3A have been recently identified in patients with KS. In this study, we screened SEMA3A variants in a cohort of Chinese patients with IHH by whole exome sequencing. Three novel heterozygous SEMA3A variants (R197Q, R617Q and V458I) were identified in two nIHH and one KS patients, respectively. Functional studies indicated that R197Q and R617Q variants were ineffective in activating the phosphorylation of FAK (focal adhesion kinase) in GN11 cells, despite normal production and secretion in HEK293T cells. The V458I SEMA3A had defect in secretion as it was not detected in the conditioned medium from HEK293T cells. Compared with wild type SEMA3A protein, all three SEMA3A mutant proteins were ineffective in inducing the migration of GN11 cells. Our study further showed the contribution of SEMA3A loss-of-function variants to the pathogenesis of IHH.


Subject(s)
Hypogonadism/genetics , Infertility/genetics , Kallmann Syndrome/genetics , Semaphorin-3A/genetics , Adult , Cell Movement/genetics , Female , Focal Adhesion Kinase 1/genetics , Gonadotropin-Releasing Hormone/genetics , HEK293 Cells , Heterozygote , Humans , Hypogonadism/pathology , Infertility/pathology , Kallmann Syndrome/pathology , Male , Mutation/genetics , Pedigree , Phenotype , Exome Sequencing
4.
Cereb Cortex ; 29(8): 3561-3576, 2019 07 22.
Article in English | MEDLINE | ID: mdl-30272120

ABSTRACT

Seven unrelated individuals (four pediatric, three adults) with the TUBB3 E410K syndrome, harboring identical de novo heterozygous TUBB3 c.1228 G>A mutations, underwent neuropsychological testing and neuroimaging. Despite the absence of cortical malformations, they have intellectual and social disabilities. To search for potential etiologies for these deficits, we compared their brain's structural and white matter organization to 22 controls using structural and diffusion magnetic resonance imaging. Diffusion images were processed to calculate fractional anisotropy (FA) and perform tract reconstructions. Cortical parcellation-based network analysis and gyral topology-based FA analyses were performed. Major interhemispheric, projection and intrahemispheric tracts were manually segmented. Subjects had decreased corpus callosum volume and decreased network efficiency. While only pediatric subjects had diffuse decreases in FA predominantly affecting mid- and long-range tracts, only adult subjects had white matter volume loss associated with decreased cortical surface area. All subjects showed aberrant corticospinal tract trajectory and bilateral absence of the dorsal language network long segment. Furthermore, pediatric subjects had more tracts with decreased FA compared with controls than did adult subjects. These findings define a TUBB3 E410K neuroimaging endophenotype and lead to the hypothesis that the age-related changes are due to microscopic intrahemispheric misguided axons that are pruned during maturation.


Subject(s)
Autism Spectrum Disorder/diagnostic imaging , Cerebral Cortex/diagnostic imaging , Corpus Callosum/diagnostic imaging , Intellectual Disability/diagnostic imaging , Pyramidal Tracts/diagnostic imaging , Tubulin/genetics , White Matter/diagnostic imaging , Adult , Age Factors , Anisotropy , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Autism Spectrum Disorder/physiopathology , Brain/diagnostic imaging , Brain/pathology , Case-Control Studies , Cerebral Cortex/pathology , Child , Corpus Callosum/pathology , Diffusion Magnetic Resonance Imaging , Diffusion Tensor Imaging , Endophenotypes , Female , Fibrosis/diagnostic imaging , Fibrosis/genetics , Fibrosis/pathology , Fibrosis/physiopathology , Heterozygote , Humans , Intellectual Disability/genetics , Intellectual Disability/pathology , Intellectual Disability/physiopathology , Kallmann Syndrome/diagnostic imaging , Kallmann Syndrome/genetics , Kallmann Syndrome/pathology , Kallmann Syndrome/physiopathology , Male , Mutation , Neural Pathways/diagnostic imaging , Neural Pathways/pathology , Neuropsychological Tests , Ophthalmoplegia/diagnostic imaging , Ophthalmoplegia/genetics , Ophthalmoplegia/pathology , Ophthalmoplegia/physiopathology , Organ Size , Pyramidal Tracts/pathology , Syndrome , White Matter/pathology , Young Adult
5.
J Assist Reprod Genet ; 36(12): 2481-2484, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31741254

ABSTRACT

PURPOSE: To present the first case proposing the use of preimplantation genetic testing for monogeneic disorders for Kallmann syndrome, providing comprehensive care in the genomic era of precision medicine. METHODS: Gonadotropin therapy was used for spermatogenesis, followed by in vitro fertilization by intracytoplasmic sperm injection and embryo transfer. Cross-generational targeted next-generation sequencing was then done for genes known to cause Kallmann syndrome. RESULTS: A heterozygous mutation at codon 102 of the FGFR1 gene was found in the patient, but the father was found to have the same mutation yet is unaffected by Kallmann syndrome. Since no causative mutation was found, a de novo or sporadic mutation was suspected as the cause of Kallmann syndrome in this case. CONCLUSIONS: Comprehensive care must be available for male Kallmann syndrome patients, as treatment should not stop at spermatogenesis, but continue with genetic counseling due to possible inheritance.


Subject(s)
Hypogonadism/drug therapy , Kallmann Syndrome/diagnosis , Preimplantation Diagnosis , Receptor, Fibroblast Growth Factor, Type 1/genetics , Embryo Transfer , Female , Fertilization in Vitro , Gonadotropins/administration & dosage , High-Throughput Nucleotide Sequencing , Humans , Hypogonadism/genetics , Hypogonadism/pathology , Kallmann Syndrome/drug therapy , Kallmann Syndrome/genetics , Kallmann Syndrome/pathology , Live Birth/epidemiology , Live Birth/genetics , Male , Mutation , Pedigree , Precision Medicine , Pregnancy , Pregnancy, Multiple , Spermatogenesis/drug effects
6.
Hum Mol Genet ; 25(10): 1912-1922, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26931467

ABSTRACT

Mutations in FGFR1 have recently been associated with Hartsfield syndrome, a clinically distinct syndromic form of holoprosencephaly (HPE) with ectrodactly, which frequently includes combinations of craniofacial, limb and brain abnormalities not typical for classical HPE. Unrelated clinical conditions generally without craniofacial or multi-system malformations include Kallmann syndrome and idiopathic hypogonadotropic hypogonadism. FGFR1 is a principal cause for these less severe diseases as well. Here we demonstrate that of the nine FGFR1 mutations recently detected in our screen of over 200 HPE probands by next generation sequencing, only five distinct mutations in the kinase domain behave as dominant-negative mutations in zebrafish over-expression assays. Three FGFR1 mutations seen in HPE probands behave identical to wild-type FGFR1 in rescue assays, including one apparent de novo variation. Interestingly, in one HPE family, a deleterious FGFR1 allele was transmitted from one parent and a loss-of-function allele in FGF8 from the other parent to both affected daughters. This family is one of the clearest examples to date of gene:gene synergistic interactions causing HPE in humans.


Subject(s)
Cleft Lip/genetics , Cleft Palate/genetics , Fingers/abnormalities , Genetic Predisposition to Disease , Hand Deformities, Congenital/genetics , Holoprosencephaly/genetics , Hypogonadism/genetics , Intellectual Disability/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Alleles , Animals , Child , Child, Preschool , Cleft Lip/physiopathology , Cleft Palate/physiopathology , Disease Models, Animal , Female , Fingers/physiopathology , Gene Expression Regulation , Genotype , Hand Deformities, Congenital/physiopathology , High-Throughput Nucleotide Sequencing , Holoprosencephaly/physiopathology , Humans , Hypogonadism/pathology , Infant , Intellectual Disability/physiopathology , Kallmann Syndrome/genetics , Kallmann Syndrome/pathology , Male , Mutation , Pedigree , Severity of Illness Index , Zebrafish/genetics
7.
Clin Endocrinol (Oxf) ; 87(6): 757-766, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28833369

ABSTRACT

CONTEXT: The diagnosis of congenital hypogonadotropic hypogonadism (CHH) in prepuberty has always been challenging. Here, we aimed at studying the clinical and genetic features of paediatric CHH, especially the phenotype of hypospadias and dual defects (patients showing hypothalamic and/or pituitary defects and testicular hypoplasia), so as to have a better understanding of CHH. DESIGN: The clinical and genetic features of patients with CHH were analysed, and the relationships between hypospadias, dual defects and genetics were investigated. PATIENTS: Patients who visited Beijing Children's Hospital and were positively diagnosed with CHH. MEASUREMENTS: The collected data included sex hormones, MRI of the olfactory bulb, human chorionic gonadotrophin (hCG) test and genetic testing. We analysed clinical features and genetic results, especially hypospadias and dual defects, and compared the stimulated testosterone (T) levels in patients with and without cryptorchidism. RESULTS: Sixty-four patients were positively diagnosed, and forty-seven (73.4%) had Kallmann syndrome (KS). Four patients (6.3%) had hypospadias, including 2 KS. Micropenis combined with cryptorchidism was the most common phenotype (39%). Approximately two-third of patients showed a poor response to hCG; 15 cases were diagnosed with dual defects, and there were no significant differences between those with and without cryptorchidism. Twenty-six cases (51%) of 51 patients were identified as having classical HH mutations, affecting 10 different genes, with oligogenic mutations in 5 cases (9.8%). The most common mutations were in PROKR2 (17.6%), FGFR1 (13.7%) and CHD7 (7.8%). The frequency of PROKR2 mutations was higher in dual HH when compared to other HH cases (6/15 vs 3/36, P = .021). CONCLUSIONS: Micropenis and/or cryptorchidism can serve as important signs for the diagnosis of HH in paediatrics, and the coexistence of hypospadias does not exclude the diagnosis of CHH, including KS or normosmic isolated HH (nHH). Testicular function may be impaired earlier than expected, and PROKR2 mutations need to be evaluated to identify presumed dual defects.


Subject(s)
Hypogonadism/genetics , Hypogonadism/pathology , Adolescent , Child , Child, Preschool , Chorionic Gonadotropin/metabolism , Cryptorchidism/genetics , Cryptorchidism/pathology , Gastrointestinal Hormones/metabolism , Genital Diseases, Male/genetics , Genital Diseases, Male/pathology , Humans , Hypospadias/genetics , Hypospadias/pathology , Infant , Kallmann Syndrome/genetics , Kallmann Syndrome/pathology , Magnetic Resonance Imaging , Male , Mutation/genetics , Penis/abnormalities , Penis/pathology , Phenotype , Testosterone/metabolism
8.
Am J Hum Genet ; 92(5): 707-24, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23643381

ABSTRACT

Transcription factor SOX10 plays a role in the maintenance of progenitor cell multipotency, lineage specification, and cell differentiation and is a major actor in the development of the neural crest. It has been implicated in Waardenburg syndrome (WS), a rare disorder characterized by the association between pigmentation abnormalities and deafness, but SOX10 mutations cause a variable phenotype that spreads over the initial limits of the syndrome definition. On the basis of recent findings of olfactory-bulb agenesis in WS individuals, we suspected SOX10 was also involved in Kallmann syndrome (KS). KS is defined by the association between anosmia and hypogonadotropic hypogonadism due to incomplete migration of neuroendocrine gonadotropin-releasing hormone (GnRH) cells along the olfactory, vomeronasal, and terminal nerves. Mutations in any of the nine genes identified to date account for only 30% of the KS cases. KS can be either isolated or associated with a variety of other symptoms, including deafness. This study reports SOX10 loss-of-function mutations in approximately one-third of KS individuals with deafness, indicating a substantial involvement in this clinical condition. Study of SOX10-null mutant mice revealed a developmental role of SOX10 in a subpopulation of glial cells called olfactory ensheathing cells. These mice indeed showed an almost complete absence of these cells along the olfactory nerve pathway, as well as defasciculation and misrouting of the nerve fibers, impaired migration of GnRH cells, and disorganization of the olfactory nerve layer of the olfactory bulbs.


Subject(s)
Deafness/genetics , Genetic Predisposition to Disease/genetics , Kallmann Syndrome/genetics , Neuroglia/pathology , Olfactory Pathways/pathology , SOXE Transcription Factors/genetics , Animals , DNA Mutational Analysis , Deafness/pathology , Female , France , Galactosides , HeLa Cells , Humans , Indoles , Kallmann Syndrome/pathology , Male , Mice , Mutation/genetics , Plasmids/genetics
9.
Clin Endocrinol (Oxf) ; 85(1): 100-9, 2016 07.
Article in English | MEDLINE | ID: mdl-26708526

ABSTRACT

BACKGROUND: Congenital isolated hypogonadotropic hypogonadism (IHH) is caused due to defect in GnRH neuronal development, migration and action. Although genetic aetiology of IHH is increasingly being studied, Asian Indian data on phenotypic spectrum and genetic basis are scarce. OBJECTIVE: To investigate the phenotypic and genotypic spectrum of IHH in Asian Indian subjects. DESIGN, SETTING AND SUBJECTS: A cohort of 135 IHH probands were characterized phenotypically for reproductive and nonreproductive features and screened for rare sequence variations (RSVs) in five genes KAL1, FGFR1, FGF8, GNRHR and KISS1R. RESULT: Of 135 probands [56 normosmic IHH (nIHH) and 79 Kallmann syndrome (KS)], 20 were familial cases. KS group had more male dominance (M:F ratio of 8:1) as compared to nIHH group (M:F ratio of 1·5:1). Complete absence of puberty was more prevalent in KS probands (81% in KS vs 46% in nIHH). The prevalence of MRI abnormalities was more in anosmic group (92·8%) as compared to hyposmic (37·5%) and normosmic groups (15·4%). No particular nonreproductive phenotypic predominance was seen in any group. Genotyping revealed rare sequence variation (RSV) detection rate of 15·5% in five genes studied: (KAL1 - 4·4%, FGFR1 - 4·4%, GNRHR - 6·7%, oligogenicity - 1·5%). Prevalence of RSV was more common in familial cases (35%) as compared to sporadic (12·2%). GNRHR RSV p.C279Y (not reported in patients of ethnicities other than south Asians) was recurring in four unrelated patients. CONCLUSION: In our cohort, 60% were KS with majority of males and a severe reproductive phenotype as against nIHH. Contribution of the genetic burden for the five genes studied was 15·5%. RSV p.C279Y in GNRHR may have a founder effect originating from south Asia. This study provides a model for molecular and phenotypic representation of Asian Indian subjects with IHH.


Subject(s)
Genotype , Hypogonadism/genetics , Kallmann Syndrome/genetics , Phenotype , Asia/ethnology , Base Sequence , Family Health , Female , Founder Effect , Genetic Variation , Humans , Hypogonadism/pathology , India/epidemiology , Kallmann Syndrome/pathology , Male , Molecular Epidemiology , Pedigree , Reproduction
10.
J Comput Assist Tomogr ; 40(1): 39-42, 2016.
Article in English | MEDLINE | ID: mdl-26571055

ABSTRACT

PURPOSE: We sought to characterize the magnetic resonance imaging (MRI) findings in patients with Kallmann syndrome (KS). MATERIALS AND METHODS: Fourteen patients with KS and a comparison group of 20 matched people with normal MRI were analyzed with optimized voxel-based morphometry. Coronal T1- and T2-weighted images from the anterior margin of the frontal sinus to the hypothalamus were obtained. The olfactory sulci, bulbs, and bundles were assessed as normal, hypoplastic, or absent. The pituitary gland was also evaluated. RESULTS: Four of the 14 patients came from 1 family. Ten patients had low levels of GnRH and gonadal hormone, 11 had hyposmia, and 3 had anosmia. On MRI, the olfactory bulbs (OBs) and bundles were absent bilaterally in 8 patients. Two patients exhibited absence of the OBs and bundles on the left and hypoplasia on the right. Four patients displayed bilateral hypoplastic OBs and bundles. The olfactory sulci were absent in 5 and hypoplastic in 9 of these patients. The anterior pituitary was hypoplastic in 6 patients. CONCLUSIONS: Kallmann syndrome has distinctive features on MRI. Magnetic resonance imaging may aid in the diagnosis of KS in patients with ambiguous clinical findings.


Subject(s)
Hypothalamus/pathology , Kallmann Syndrome/pathology , Magnetic Resonance Imaging , Olfactory Bulb/pathology , Pituitary Gland/pathology , Prefrontal Cortex/pathology , Adolescent , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult
11.
Neuroimage ; 104: 52-8, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25300200

ABSTRACT

Among male patients affected by Kallmann syndrome, a genetically determined disease due to defective neural migration leading to hypogonadropic hypogonadism and hypo/anosmia, about 40% present the peculiar phenomenon of mirror movements, i.e. involuntary movements mirroring contralateral voluntary hand movements. Several pathogenic hypotheses have been proposed, but the ultimate neurological mechanisms are still elusive. The aim of the present study was to investigate brain anatomical substrates of mirror movements in Kallmann syndrome by means of a panel of quantitative MRI analyses. Forty-nine male Kallmann syndrome patients underwent brain MRI. The study protocol included 3D-T1-weighted gradient echo, fluid attenuated inversion recovery and diffusion tensor imaging. Voxel-based morphometry, sulcation, curvature and cortical thickness analyses and tract based spatial statistics were performed using SPM8, Freesurfer and FSL. All patients underwent a complete physical and neurological examination including the evaluation of mirror movements (according to the Woods and Teuber criteria). Kallmann syndrome patients presenting with mirror movements (16/49, 32%) displayed the following brain changes: 1) increased gray matter density in the depth of the left precentral sulcus behind the middle frontal gyrus; 2) decreased cortical thickness in the precentral gyrus bilaterally, in the depth of right precentral sulcus and in the posterior portion of the right superior frontal gyrus; and 3) decreased fractional anisotropy in the left hemisphere involving the temporal lobe and peritrigonal white matter. No differences were shown by cortical curvature and sulcation analyses. The composite array of brain changes observed in Kallmann syndrome patients with mirror movements likely represents the anatomical-structural underpinnings leading to the peculiar derangement of the complex circuitry committed to unilateral hand voluntary movements.


Subject(s)
Brain/pathology , Brain/physiopathology , Kallmann Syndrome/pathology , Kallmann Syndrome/physiopathology , Adolescent , Adult , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Diffusion Tensor Imaging , Globus Pallidus/pathology , Globus Pallidus/physiopathology , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Middle Aged , Motor Cortex/pathology , Motor Cortex/physiopathology , Psychomotor Performance/physiology , Pyramidal Tracts/pathology , Pyramidal Tracts/physiopathology , Young Adult
12.
Andrologia ; 43(6): 422-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21486418

ABSTRACT

Testosterone replacement therapy is the mainstay of treatment in male patients with isolated hypogonadotrophic hypogonadism (HH) to achieve virilisation. However, responsiveness of pilosebaceous unit (PSU) to testosterone replacement therapy in these patients is quite variable. Androgen action is inversely proportional to the number of CAG repeats in exon 1 of androgen receptor gene; therefore, we hypothesised that CAG repeat length contributes to testosterone responsiveness in patients with HH. The CAG repeat length in 21 well-virilised men (hair score > 30, responders) and 25 poorly virilised men (hair score ≤ 30, non-responders) with HH on optimal testosterone replacement therapy at least for a period of 1 year was analysed. Serum LH, FSH, testosterone and 17 ß oestradiol were estimated. Polymerase chain reaction (PCR) amplification of exon 1 of androgen receptor gene was performed from genomic DNA, and these PCR-amplified products were sequenced for the number of CAG repeats. The difference between number of CAG repeats in responders and non-responders was statistically significant (19.19 ± 3.25 and 22.24 ± 2.65, P = 0.001) and showed a strong negative correlation with total body hair score (r = -0.538 and P = 0.0001). In conclusion, these results suggest that the number of CAG repeats influences the responsiveness of PSU to testosterone treatment in patients with HH.


Subject(s)
Hormone Replacement Therapy , Kallmann Syndrome/drug therapy , Testosterone/therapeutic use , Adult , Base Sequence , DNA Primers , Estradiol/blood , Exons , Follicle Stimulating Hormone/blood , Humans , Kallmann Syndrome/pathology , Luteinizing Hormone/blood , Male , Polymerase Chain Reaction , Receptors, Androgen/genetics , Testosterone/administration & dosage , Testosterone/blood , Trinucleotide Repeats
13.
Genes (Basel) ; 12(6)2021 06 05.
Article in English | MEDLINE | ID: mdl-34198905

ABSTRACT

Kallmann syndrome (KS) is a combination of isolated hypogonadotropic hypogonadism (IHH) with olfactory dysfunction, representing a heterogeneous disorder with a broad phenotypic spectrum. The genetic background of KS has not yet been fully established. This study was conducted on 46 Polish KS subjects (41 males, 5 females; average age: 29 years old). The studied KS patients were screened for defects in a 38-gene panel with next-generation sequencing (NGS) technology. The analysis revealed 27 pathogenic and likely pathogenic (P/LP) variants, and 21 variants of uncertain significance (VUS). The P/LP variants were detected in 20 patients (43.5%). The prevalence of oligogenic P/LP defects in selected genes among KS patients was 26% (12/46), whereas the co-occurrence of other variants was detected in 43% (20 probands). The examined KS patients showed substantial genotypic and phenotypic variability. A marked difference in non-reproductive phenotypes, involving defects in genes responsible for GnRH neuron development/migration and genes contributing to pituitary development and signaling, was observed. A comprehensive gene panel for IHH testing enabled the detection of clinically relevant variants in the majority of KS patients, which makes targeted NGS an effective molecular tool. The significance of oligogenicity and the high incidence of alterations in selected genes should be further elucidated.


Subject(s)
Hypothalamo-Hypophyseal System/metabolism , Kallmann Syndrome/genetics , Mutation , Neurogenesis , Phenotype , Adolescent , Adult , Cell Movement , Female , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Humans , Hypothalamo-Hypophyseal System/cytology , Hypothalamo-Hypophyseal System/growth & development , Kallmann Syndrome/metabolism , Kallmann Syndrome/pathology , Male , Middle Aged , Neurons/cytology , Neurons/metabolism , Neurons/physiology , Signal Transduction
14.
J Clin Endocrinol Metab ; 106(9): e3312-e3326, 2021 08 18.
Article in English | MEDLINE | ID: mdl-33725720

ABSTRACT

CONTEXT: Men with congenital hypogonadotropic hypogonadism (CHH) and Kallmann syndrome (KS) have both low circulating testosterone and estradiol levels. Whether bone structure is affected remains unknown. OBJECTIVE: To characterize bone geometry, volumetric density and microarchitecture in CHH/KS. METHODS: This cross-sectional study, conducted at a single French tertiary academic medical center, included 51 genotyped CHH/KS patients and 40 healthy volunteers. Among CHH/KS men, 98% had received testosterone and/or combined gonadotropins. High-resolution peripheral quantitative computed tomography (HR-pQCT), dual-energy x-ray absorptiometry (DXA), and measurement of serum bone markers were used to determine volumetric bone mineral density (vBMD) and cortical and trabecular microarchitecture. RESULTS: CHH and controls did not differ for age, body mass index, and levels of vitamin D and PTH. Despite long-term hormonal treatment (10.8 ± 6.8 years), DXA showed lower areal bone mineral density (aBMD) in CHH/KS at lumbar spine, total hip, femoral neck, and distal radius. Consistent with persistently higher serum bone markers, HR-pQCT revealed lower cortical and trabecular vBMD as well as cortical thickness at the tibia and the radius. CHH/KS men had altered trabecular microarchitecture with a predominant decrease of trabecular thickness. Moreover, CHH/KS men exhibited lower cortical bone area, whereas total and trabecular areas were higher only at the tibia. Earlier treatment onset (before age 19 years) conferred a significant advantage for trabecular bone volume/tissue volume and trabecular vBMD at the tibia. CONCLUSION: Both vBMD and bone microarchitecture remain impaired in CHH/KS men despite long-term hormonal treatment. Treatment initiation during adolescence is associated with enhanced trabecular outcomes, highlighting the importance of early diagnosis.


Subject(s)
Bone Density , Bone and Bones/pathology , Gonadotropins/deficiency , Hypogonadism/pathology , Absorptiometry, Photon , Adolescent , Adult , Cross-Sectional Studies , Early Diagnosis , Estradiol/blood , Genotype , Hormone Replacement Therapy , Humans , Hypogonadism/congenital , Hypogonadism/drug therapy , Kallmann Syndrome/pathology , Male , Middle Aged , Testosterone/blood , Tomography, X-Ray Computed , Young Adult
15.
Front Endocrinol (Lausanne) ; 11: 592831, 2020.
Article in English | MEDLINE | ID: mdl-33597923

ABSTRACT

Introduction: Kallmann syndrome (KS) is idiopathic hypogonadotropic hypogonadism with olfactory loss or decline. Waardenburg syndrome type II (WS2) is a clinically and genetically heterogeneous disease, characterized by congenital sensorineural deafness and abnormal pigmentation of the iris, hair, and skin. Recently, mutations in the well-known WS pathogenic gene SOX10 have been found in some KS patients with deafness, but whether SOX10 is a co-pathogenic gene of KS and WS remains uncertain. Here, we report a rare case of KS and WS2 co-occurrence due to SOX10 mutations. Methods: Detailed histories were collected through questionnaires and physical examination. Blood samples of the patient and his family members were collected after obtaining informed consents. Suspected mutations were amplified and verified by Sanger sequencing after the next generation sequencing of related genes. The raw sequence data were compared to the known gene sequence data in publicly available sequence data bases using Burrows-Wheeler Aligner software (BWA, 0.7.12-r1039). Results: A 28-year-old male patient sought treatment for hypogonadism and the absence of secondary sexual characteristics. In addition, he showed signs of obesity, hyposmia, sensorineural hearing loss, and blue iris. Magnetic resonance imaging (MRI) of the olfactory bulb showed small bilateral olfactory bulbs and tracts and diaphragma cerebri. MRI of the pituitary gland revealed a flat pituitary gland in the sella. Laboratory examination demonstrated hypogonadotropic hypogonadism, pituitary hypothyroidism, subclinical hypothyroidism, and the presence of insulin resistance with normal blood glucose levels. Sequencing of the SOX10 gene showed a 20 bp insertion in between coding bases 1,179 and 1,180 (c.1179_1180insACTATGGCTCAGCCTTCCCC). This results in a frame-shifting mutation of the 394th amino acid serine in exon4 with the resulting the amino acid sequence of the protein predicted to be TMAQPSP PSPAPSLTTL TISPQDPIMA TRARPLASTR PSPIWGPRSG PSTRPSLTPA PQGPSPTAPH TGSSQYIRHC PGPKGGPVAT TPRPAPAPSL CALFLAHLRP GGGSGGG*. Conclusion: SOX10 plays an important role in some critical stages of neural crest cell development and SOX10 mutation may be a common pathogenic factor for both KS and WS. Therefore, SOX10 mutation analysis should be considered for KS patients with combined WS clinical manifestations, especially deafness.


Subject(s)
Heterozygote , Kallmann Syndrome/pathology , Mutation , SOXE Transcription Factors/genetics , Waardenburg Syndrome/pathology , Adult , Humans , Kallmann Syndrome/complications , Kallmann Syndrome/genetics , Male , Waardenburg Syndrome/complications , Waardenburg Syndrome/genetics
16.
J Clin Endocrinol Metab ; 105(3)2020 03 01.
Article in English | MEDLINE | ID: mdl-31628846

ABSTRACT

CONTEXT: Kallmann syndrome (KS) is a rare, genetically heterogeneous Mendelian disorder. Structural defects in KS patients have helped define the genetic architecture of gonadotropin-releasing hormone (GnRH) neuronal development in this condition. OBJECTIVE: Examine the functional role a novel structural defect affecting a long noncoding RNA (lncRNA), RMST, found in a KS patient. DESIGN: Whole genome sequencing, induced pluripotent stem cells and derived neural crest cells (NCC) from the KS patient were contrasted with controls. SETTING: The Harvard Reproductive Sciences Center, Massachusetts General Hospital Center for Genomic Medicine, and Singapore Genome Institute. PATIENT: A KS patient with a unique translocation, t(7;12)(q22;q24). INTERVENTIONS/MAIN OUTCOME MEASURE/RESULTS: A novel translocation was detected affecting the lncRNA, RMST, on chromosome 12 in the absence of any other KS mutations. Compared with controls, the patient's induced pluripotent stem cells and NCC provided functional information regarding RMST. Whereas RMST expression increased during NCC differentiation in controls, it was substantially reduced in the KS patient's NCC coincident with abrogated NCC morphological development and abnormal expression of several "downstream" genes essential for GnRH ontogeny (SOX2, PAX3, CHD7, TUBB3, and MKRN3). Additionally, an intronic single nucleotide polymorphism in RMST was significantly implicated in a genome-wide association study associated with age of menarche. CONCLUSIONS: A novel deletion in RMST implicates the loss of function of a lncRNA as a unique cause of KS and suggests it plays a critical role in the ontogeny of GnRH neurons and puberty.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Kallmann Syndrome/genetics , Kallmann Syndrome/pathology , RNA, Long Noncoding/genetics , Translocation, Genetic , Adult , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 7/genetics , Genome-Wide Association Study , Gonadotropin-Releasing Hormone/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Male , Neural Crest/metabolism , Neural Crest/pathology , Prognosis
17.
Asian J Androl ; 22(4): 390-395, 2020.
Article in English | MEDLINE | ID: mdl-31464203

ABSTRACT

Partial congenital hypogonadotropic hypogonadism (PCHH) is caused by an insufficiency in, but not a complete lack of, gonadotropin secretion. This leads to reduced testosterone production, mild testicular enlargement, and partial pubertal development. No studies have shown the productivity of spermatogenesis in patients with PCHH. We compared the outcomes of gonadotropin-induced spermatogenesis between patients with PCHH and those with complete congenital hypogonadotropic hypogonadism (CCHH). This retrospective study included 587 patients with CHH who were treated in Peking Union Medical College Hospital (Beijing, China) from January 2008 to September 2016. A total of 465 cases were excluded from data analysis for testosterone or gonadotropin-releasing hormone treatment, cryptorchidism, poor compliance, or incomplete medical data. We defined male patients with PCHH as those with a testicular volume of ≥4 ml and patients with a testicular volume of <4 ml as CCHH. A total of 122 compliant, noncryptorchid patients with PCHH or CCHH received combined human chorionic gonadotropin and human menopausal gonadotropin and were monitored for 24 months. Testicular size, serum luteinizing hormone levels, follicle-stimulating hormone levels, serum total testosterone levels, and sperm count were recorded at each visit. After gonadotropin therapy, patients with PCHH had a higher spermatogenesis rate (92.3%) than did patients with CCHH (74.7%). During 24-month combined gonadotropin treatment, the PCHH group took significantly less time to begin producing sperm compared with the CCHH group (median time: 11.7 vs 17.8 months, P < 0.05). In conclusion, after combined gonadotropin treatment, patients with PCHH have a higher spermatogenesis success rate and sperm concentrations and require shorter treatment periods for sperm production.


Subject(s)
Chorionic Gonadotropin/therapeutic use , Hypogonadism/drug therapy , Kallmann Syndrome/drug therapy , Menotropins/therapeutic use , Sperm Count , Testis/pathology , Adolescent , Drug Therapy, Combination , Follicle Stimulating Hormone/blood , High-Throughput Nucleotide Sequencing , Humans , Hypogonadism/congenital , Hypogonadism/genetics , Hypogonadism/pathology , Kallmann Syndrome/genetics , Kallmann Syndrome/pathology , Kaplan-Meier Estimate , Luteinizing Hormone/blood , Male , Organ Size , Severity of Illness Index , Spermatogenesis , Testosterone/blood , Time Factors , Treatment Outcome , Young Adult
18.
Anat Rec (Hoboken) ; 302(3): 394-404, 2019 03.
Article in English | MEDLINE | ID: mdl-29663690

ABSTRACT

Originally discovered in elasmobranchs by Fritsh in 1878, the nervus terminalis has been found in virtually all species, including humans. After more than one-century debate on its nomenclature, it is nowadays recognized as cranial pair zero. The nerve mostly originates in the olfactory placode, although neural crest contribution has been also proposed. Developmentally, the nervus terminalis is clearly observed in human embryos; subsequently, during the fetal period loses some of its ganglion cells, and it is less recognizable in adults. Fibers originating in the nasal cavity passes into the cranium through the middle area of the cribiform plate of the ethmoid bone. Intracranially, fibers joint the telencephalon at several sites including the olfactory trigone and the primordium of the hippocampus to reach preoptic and precommissural regions. The nervus terminalis shows ganglion cells, that sometimes form clusters, normally one or two located at the base of the crista galli, the so-called ganglion of the nervus terminalis. Its function is uncertain. It has been described that its fibers facilitates migration of luteinizing hormone-releasing hormone cells to the hypothalamus thus participating in the development of the hypothalamic-gonadal axis, which alteration may provoke Kallmann's syndrome in humans. This review summarizes current knowledge on this structure, incorporating original illustrations of the nerve at different developmental stages, and focuses on its anatomical and clinical relevance. Anat Rec, 302:394-404, 2019. © 2018 Wiley Periodicals, Inc.


Subject(s)
Cranial Nerves/anatomy & histology , Kallmann Syndrome/pathology , Nasal Mucosa/anatomy & histology , Nerve Endings/chemistry , Animals , Cranial Nerves/metabolism , Humans , Kallmann Syndrome/metabolism , Luteinizing Hormone/metabolism , Nasal Mucosa/metabolism , Nerve Endings/metabolism
19.
Pituitary ; 11(1): 109-12, 2008.
Article in English | MEDLINE | ID: mdl-17530415

ABSTRACT

Kallmann syndrome (KS) is a developmental disease characterized by the association of isolated hypogonadotropic hypogonadism and anosmia/hyposmia. We report an unusual presentation of two females with KS and empty sella. These females, aged at 20 and 29-year-old, presented primary amenorrhea with prepubertal estradiol and low gonadotropin levels. No other significant clinical signs were observed. Empty sella was observed on MRI in both cases. Sequencing of FGFR1 gene, recently implicated in autosomal form of KS, was performed and one splicing mutation (IVS14 + 1G > A) was identified in one patient.


Subject(s)
Empty Sella Syndrome/complications , Kallmann Syndrome/complications , Adult , Amenorrhea/etiology , DNA Mutational Analysis , Empty Sella Syndrome/pathology , Estradiol/blood , Female , Follicle Stimulating Hormone, Human/blood , Humans , Hypogonadism/etiology , Kallmann Syndrome/blood , Kallmann Syndrome/genetics , Kallmann Syndrome/pathology , Luteinizing Hormone/blood , Magnetic Resonance Imaging , Mutation , Olfaction Disorders/etiology , Receptor, Fibroblast Growth Factor, Type 1/genetics , Young Adult
20.
J Clin Endocrinol Metab ; 92(2): 604-7, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17090639

ABSTRACT

CONTEXT: Hypogonadotropic hypogonadism (HH) can occur at any stage of life as an isolated congenital or acquired abnormality or within a more generalized pituitary or hypothalamic impairment. However, the defect in patients with idiopathic HH is still unknown. OBJECTIVE: The aim of this study was to investigate the prevalence of antipituitary antibodies (APA) in a group of HH patients with or without Kallmann's syndrome and to characterize their pituitary target. DESIGN: We conducted a cross-sectional cohort study. SETTING: The study was performed at the Endocrinology Unit of the Second University of Naples. PATIENTS: Twenty-one HH patients with normal sense of smell (group 1), 10 patients with Kallmann's syndrome (group 2), 13 patients with HH associated with other pituitary hormone deficiencies (group 3), and 50 normal controls were studied. MAIN OUTCOME MEASURES: APA were evaluated in patients and in controls by indirect immunofluorescence. Moreover, a magnetic resonance imaging (MRI) of the hypothalamic-pituitary region was performed in all three groups of patients. RESULTS: APA were detected at high titer in eight out of 21 patients in group 1 (38%) and in five of 13 in group 3 (38.4%), and at low titers in two out of 10 in group 2 (20%) and in three of 50 controls (6%). In patients of group 1, APA immunostained selectively gonadotropin-secreting cells, whereas in those of group 3, they immunostained other pituitary hormone-secreting cells also. None of patients in group 1 showed alterations on MRI, whereas all patients in group 2 showed aplasia/hypoplasia of the olfactory bulbs/tracts and/or of olfactory sulci. Among the five APA-positive patients in group 3, three had normal MRI, one had findings of empty sella, and one had findings of autoimmune hypophysitis. CONCLUSIONS: Our results suggest that some apparently idiopathic cases of HH, both isolated and associated with other pituitary impairment, can be caused by an early autoimmune process involving the gonadotrophs at pituitary level. Future longitudinal studies are needed to clarify the natural history of this process and the possible effect of early corticosteroid therapy.


Subject(s)
Autoantibodies/blood , Gonadotropins, Pituitary/immunology , Kallmann Syndrome/epidemiology , Kallmann Syndrome/immunology , Pituitary Gland/immunology , Adult , Animals , Antibody Specificity , Cohort Studies , Cross-Sectional Studies , Fluorescent Antibody Technique, Indirect , Gonadotropins, Pituitary/deficiency , Gonadotropins, Pituitary/metabolism , Humans , Kallmann Syndrome/pathology , Magnetic Resonance Imaging , Male , Olfaction Disorders/epidemiology , Olfaction Disorders/immunology , Olfaction Disorders/pathology , Olfactory Bulb/immunology , Olfactory Bulb/pathology , Papio , Pituitary Gland/pathology , Seroepidemiologic Studies
SELECTION OF CITATIONS
SEARCH DETAIL