Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 722
Filter
1.
Hum Mol Genet ; 31(24): 4207-4216, 2022 12 16.
Article in English | MEDLINE | ID: mdl-35899427

ABSTRACT

Kallmann syndrome (KS) is a congenital disorder characterized by idiopathic hypogonadotropic hypogonadism and olfactory dysfunction. KS is linked to variants in >34 genes, which are scattered across the human genome and show disparate biological functions. Although the genetic basis of KS is well studied, the mechanisms by which disruptions of these diverse genes cause the same outcome of KS are not fully understood. Here we show that disruptions of KS-linked genes affect the same biological processes, indicating convergent molecular mechanisms underlying KS. We carried out machine learning-based predictions and found that KS-linked mutations in heparan sulfate 6-O-sulfotransferase 1 (HS6ST1) are likely loss-of-function mutations. We next disrupted Hs6st1 and another KS-linked gene, fibroblast growth factor receptor 1 (Fgfr1), in mouse neuronal cells and measured transcriptome changes using RNA sequencing. We found that disruptions of Hs6st1 and Fgfr1 altered genes in the same biological processes, including the upregulation of genes in extracellular pathways and the downregulation of genes in chromatin pathways. Moreover, we performed genomics and bioinformatics analyses and found that Hs6st1 and Fgfr1 regulate gene transcription likely via the transcription factor Sox9/Sox10 and the chromatin regulator Chd7, which are also associated with KS. Together, our results demonstrate how different KS-linked genes work coordinately in a convergent signaling pathway to regulate the same biological processes, thus providing new insights into KS.


Subject(s)
Hypogonadism , Kallmann Syndrome , Receptor, Fibroblast Growth Factor, Type 1 , Sulfotransferases , Animals , Mice , Chromatin , Hypogonadism/genetics , Kallmann Syndrome/genetics , Kallmann Syndrome/metabolism , Mutation , Receptor, Fibroblast Growth Factor, Type 1/genetics , Sulfotransferases/genetics
2.
Int J Mol Sci ; 25(7)2024 Mar 31.
Article in English | MEDLINE | ID: mdl-38612730

ABSTRACT

There is very little knowledge regarding the terminal nerve, from its implications in the involvement and pathogenesis of certain conditions, to its embryological origin. With this review, we try to summarize the most important evidence on the terminal nerve, aiming to clarify its anatomy and the various functions attributed to it, to better interpret its potential involvement in pathological processes. Recent studies have also suggested its potential role in the control of human reproductive functions and behaviors. It has been hypothesized that it plays a role in the unconscious perception of specific odors that influence autonomic and reproductive hormonal systems through the hypothalamic-pituitary-gonadal axis. We used the PubMed database and found different articles which were then selected independently by three authors. We found 166 articles, of which, after careful selection, only 21 were analyzed. The terminal nerve was always thought to be unimportant in our body. It was well studied in different types of animals, but few studies have been completed in humans. For this reason, its function remains unknown. Studies suggest a possible implication in olfaction due to the anatomical proximity with the olfactive nerve. Others suggest a more important role in reproduction and sexual behaviors. New emerging information suggests a possible role in Kallmann syndrome and COVID-19.


Subject(s)
COVID-19 , Kallmann Syndrome , Animals , Humans , Smell , Nerve Endings , Autonomic Nervous System
3.
Clin Endocrinol (Oxf) ; 98(4): 554-558, 2023 04.
Article in English | MEDLINE | ID: mdl-36536529

ABSTRACT

OBJECTIVE: Kallmann's syndrome (KS) is characterized by hypogonadotropic hypogonadism and olfactory disorders. The complementary exams for evaluating of patients with hypogonadotrophic hypogonadism are important for the diagnosis and management of these patients. PATIENTS: We performed a well-established olfactory Sniffin' Stick test (SST) on 17 adult patients with KS and brain magnetic resonance imaging (MRI) to evaluate olfactory structures and further analysis by Freesurfer, a software for segmentation and volumetric evaluation of brain structures. We compared the Freesurfer results with 34 healthy patients matched for age and sex and performed correlations between the data studied. RESULTS: More than half of the patients with KS reported preserved smell but had olfactory disorders in the SST. In the MRI, 16 patients showed changes in the olfactory groove, the olfactory bulb-tract complex was altered in all of themĀ and 52% had symmetrical structural changes. Interestingly, the pituitary gland was normal in only 29%. Regarding correlations, symmetrical changes in the olfactory structures were related to anosmia in 100%, while asymmetric changes induced anosmia in only 50% (p = .0294). In Freesurfer's assessment, patients with KS, compared to controls, had lower brainstem volume. In those with aplastic anterior olfactory sulcus, the brainstem volume was lower than in hypoplasia (p = .0333). CONCLUSIONS: Olfactory assessment and MRI proved to be important auxiliary tools for the diagnosis and management of patients with KS. New studies are needed to confirm the decrease in brainstem volume found by the Freesurfer software in patients with KS. Further studies are needed to confirm the decrease in brainstem volume found by the Freesurfer software in patients with KS.


Subject(s)
Hypogonadism , Kallmann Syndrome , Klinefelter Syndrome , Olfaction Disorders , Adult , Humans , Kallmann Syndrome/diagnosis , Smell , Anosmia/pathology , Olfaction Disorders/diagnosis , Olfaction Disorders/pathology , Hypogonadism/diagnosis , Brain/pathology
4.
Reprod Biol Endocrinol ; 21(1): 23, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36859276

ABSTRACT

BACKGROUND: Kallmann syndrome (KS) is a common type of idiopathic hypogonadotropic hypogonadism. To date, more than 30 genes including ANOS1 and FGFR1 have been identified in different genetic models of KS without affirmatory genotype-phenotype correlation, and novel mutations have been found. METHODS: A total of 35 unrelated patients with clinical features of disorder of sex development were recruited. Custom-panel sequencing or whole-exome sequencing was performed to detect the pathogenic mutations. Sanger sequencing was performed to verify single-nucleotide variants. Copy number variation-sequencing (CNV-seq) was performed to determine CNVs. The pathogenicity of the identified variant was predicted in silico. mRNA transcript analysis and minigene reporter assay were performed to test the effect of the mutation on splicing. RESULTS: ANOS1 gene c.709Ā T > A and c.711Ā G > T were evaluated as pathogenic by several commonly used software, and c.1063-2Ā A > T was verified by transcriptional splicing assay. The c.1063-2Ā A > T mutation activated a cryptic splice acceptor site downstream of the original splice acceptor site and resulted in an aberrant splicing of the 24-basepair at the 5' end of exon 8, yielding a new transcript with c.1063-1086 deletion. FRFR1 gene c.1835delA was assessed as pathogenic according to the ACMG guideline. The CNV of del(8)(p12p11.22)chr8:g.36140000_38460000del was judged as pathogenic according to the ACMG & ClinGen technical standards. CONCLUSIONS: Herein, we identified three novel ANOS1 mutations and two novel FGFR1 variations in Chinese KS families. In silico prediction and functional experiment evaluated the pathogenesis of ANOS1 mutations. FRFR1 c.1835delA mutation and del(8)(p12p11.22)chr8:g.36140000_38460000del were assessed as pathogenic variations. Therefore, our study expands the spectrum of mutations associated with KS and provides diagnostic evidence for patients who carry the same mutation in the future.


Subject(s)
Extracellular Matrix Proteins , Kallmann Syndrome , Nerve Tissue Proteins , Receptor, Fibroblast Growth Factor, Type 1 , Humans , DNA Copy Number Variations , Exons , Kallmann Syndrome/genetics , Mutation , Receptor, Fibroblast Growth Factor, Type 1/genetics , RNA Splice Sites , Extracellular Matrix Proteins/genetics , Nerve Tissue Proteins/genetics
5.
Am J Med Genet A ; 191(3): 831-834, 2023 03.
Article in English | MEDLINE | ID: mdl-36454653

ABSTRACT

Kallmann syndrome (KS) is a rare genetic disease characterized by pubertal failure and olfactory defects. Although many genes associated with KS have been reported, most are rare. Recently, heterozygous inactivating mutations in the neuron-derived neurotrophic factor gene (NDNF) were reported to cause KS. Here, we present a 14-year-old Kurdish boy with KS who has a novel homozygous nonsense c.1251C>A (p.Tyr417Ter) variant in NDNF. The variant was not observed in reference population databases and was predicted to be deleterious. Segregation analysis performed with Sanger sequencing indicated the autosomal recessive inheritance of the clinical phenotype. His heterozygous parents have experienced timely pubertal development and normal reproductive features. This study reported the first homozygous truncating NDNF variant, enabling the direct observation of the clinical consequences of predictively absent NDNF function. These results support the contention that the inactivating mutations in NDNF cause KS, and provide additional evidence for the complex inheritance of KS.


Subject(s)
Kallmann Syndrome , Humans , Kallmann Syndrome/genetics , Neurons , Phenotype , Reproduction , Heterozygote , Mutation
6.
Neuroendocrinology ; 113(8): 834-843, 2023.
Article in English | MEDLINE | ID: mdl-36758531

ABSTRACT

INTRODUCTION: Congenital hypogonadotropic hypogonadism (CHH) is a rare condition caused by GnRH deficiency. More than 40 genes have been associated with the pathogenesis of CHH, but most cases still remain without a molecular diagnosis. Mutations involving the same gene (e.g., FGFR1, PROK2/PROKR2, CHD7) were found to cause normosmic CHH and Kallmann syndrome (KS), with and without associated phenotypes, illustrating the coexistence of CHH with signs of other complex syndromes. The Witteveen-Kolk syndrome (WITKOS), caused by defects of the SIN3A gene, is a heterogeneous disorder characterized by distinctive facial features, microcephaly, short stature, delayed cognitive, and motor development. Although micropenis and cryptorchidism have been reported in this syndrome, WITKOS has not been formally associated with CHH so far. PATIENTS AND METHODS: A man with KS associated with mild syndromic features (S1) and a boy with global developmental delay, syndromic short stature, micropenis and cryptorchidism (S2), in whom common genetic defects associated with CHH and short stature had been previously excluded, were studied by either chromosomal microarray analysis or whole exome sequencing. RESULTS: Rare SIN3A pathogenic variants were identified in these 2 unrelated patients with CHH phenotypic features. A 550 kb deletion at 15q24.1, including the whole SIN3A gene, was identified in S1, and a SIN3A nonsense rare variant (p.Arg471*) was detected in S2. CONCLUSION: These findings lead us to propose a link between SIN3A defects and CHH, especially in syndromic cases, based on these 2 patients with overlapping phenotypes of WITKOS and CHH.


Subject(s)
Cryptorchidism , Genital Diseases, Male , Hypogonadism , Kallmann Syndrome , Humans , Male , Hypogonadism/genetics , Kallmann Syndrome/diagnosis , Mutation
7.
Neuroendocrinology ; 113(10): 1051-1058, 2023.
Article in English | MEDLINE | ID: mdl-37497936

ABSTRACT

INTRODUCTION: Olfaction and its relation to human health is an area of growing interest. Although olfaction disorders have been considered a part of Kallmann syndrome, the role of olfactory dysfunction on spermatogenesis has not been studied yet. We studied if olfactory bulbectomy (OBX) causes dysfunction in spermatogenesis as a result of Onuf's nucleus damage. METHODS: Twenty-eight male rats were divided into three groups: six as the control (G-1; n = 6), six as the only frontal burr hole applied animals SHAM (G-2; n = 6), and 16 as the study group (G-3; n = 16) in which OBX was performed. The animals were followed for 2 months. After the decapitation of the animals, olfactory bulb (OB) volumes (mm3), the neuron density of the Onuf's nucleus (n/mm3), and sperm density (n/mm3) were estimated stereologically and analyzed. RESULTS: OB volumes (mm3), degenerated neuron density of Onuf's nucleus (n/mm3), and sperm numbers of control, SHAM, and study groups were estimated as: 4 Ā± 0.5; 6 Ā± 2 and 103.245 Ā± 10.841 in G-1; 3.5 Ā± 0.7; 14 Ā± 4 and 96.891 Ā± 9.569 in G-2; and 1.3 Ā± 0.3; 91 Ā± 17 and 73.561 Ā± 6.324 in G-3. The statistical results of degenerated neuron density of Onuf's nucleus and sperm numbers between groups are p < 0.005 for G-1/G-2; p < 0.0005 for G-2/G-3; and p < 0.00001 for G-1/G-3. DISCUSSION: This study first time indicates that Onuf's nucleus degeneration secondary to OBX seems to be responsible for reduced sperm numbers.


Subject(s)
Kallmann Syndrome , Male , Humans , Animals , Rats , Sperm Count , Smell , Semen , Spinal Cord , Spermatozoa
8.
BMC Endocr Disord ; 23(1): 213, 2023 Oct 05.
Article in English | MEDLINE | ID: mdl-37798680

ABSTRACT

BACKGROUND: Idiopathic hypogonadotropic hypogonadism (IHH) is a rare congenital or acquired genetic disorder caused by gonadotropin-releasing hormone (GnRH) deficiency. IHH patients are divided into two major groups, hyposmic or anosmic IHH (Kallmann syndrome) and normosmic IHH (nIHH), according to whether their sense of smell is intact. Here we report a case of novel compound heterozygous mutations in the GNRH1 gene in a 15-year-old male with nIHH. CASE PRESENTATION: The patient presented typical clinical symptoms of delayed testicular development, with testosterone < 3.5 mmol/L and reduced gonadotropin (follicle-stimulating hormone, luteinizing hormone) levels. Two heterozygous variants of the GNRH1 gene were detected, nonsense variant 1: c.85G > T:p.G29* and variant 2: c.1A > G:p.M1V, which disrupted the start codon. CONCLUSIONS: Two GNRH1 mutations responsible for nIHH are identified in this study. Our findings extend the mutational spectrum of GNRH1 by revealing novel causative mutations of nIHH.


Subject(s)
Gonadotropin-Releasing Hormone , Hypogonadism , Adolescent , Humans , Male , Gonadotropin-Releasing Hormone/genetics , Hypogonadism/genetics , Hypogonadism/diagnosis , Kallmann Syndrome/genetics , Mutation , Testosterone/analysis
9.
J Med Genet ; 59(2): 105-114, 2022 02.
Article in English | MEDLINE | ID: mdl-34667088

ABSTRACT

SOX10 belongs to a family of 20 SRY (sex-determining region Y)-related high mobility group box-containing (SOX) proteins, most of which contribute to cell type specification and differentiation of various lineages. The first clue that SOX10 is essential for development, especially in the neural crest, came with the discovery that heterozygous mutations occurring within and around SOX10 cause Waardenburg syndrome type 4. Since then, heterozygous mutations have been reported in Waardenburg syndrome type 2 (Waardenburg syndrome type without Hirschsprung disease), PCWH or PCW (peripheral demyelinating neuropathy, central dysmyelination, Waardenburg syndrome, with or without Hirschsprung disease), intestinal manifestations beyond Hirschsprung (ie, chronic intestinal pseudo-obstruction), Kallmann syndrome and cancer. All of these diseases are consistent with the regulatory role of SOX10 in various neural crest derivatives (melanocytes, the enteric nervous system, Schwann cells and olfactory ensheathing cells) and extraneural crest tissues (inner ear, oligodendrocytes). The recent evolution of medical practice in constitutional genetics has led to the identification of SOX10 variants in atypical contexts, such as isolated hearing loss or neurodevelopmental disorders, making them more difficult to classify in the absence of both a typical phenotype and specific expertise. Here, we report novel mutations and review those that have already been published and their functional consequences, along with current understanding of SOX10 function in the affected cell types identified through in vivo and in vitro models. We also discuss research options to increase our understanding of the origin of the observed phenotypic variability and improve the diagnosis and medical care of affected patients.


Subject(s)
Embryonic Development/genetics , Embryonic Development/physiology , SOXE Transcription Factors/genetics , SOXE Transcription Factors/physiology , Animals , Enteric Nervous System/physiology , Gene Expression Regulation, Developmental , Hearing Loss/genetics , Hirschsprung Disease/genetics , Humans , Kallmann Syndrome/genetics , Melanocytes/physiology , Mutation , Neoplasms/genetics , Neural Crest/embryology , Neural Crest/physiology , Phenotype , Waardenburg Syndrome/genetics
10.
Int J Mol Sci ; 24(8)2023 Apr 18.
Article in English | MEDLINE | ID: mdl-37108593

ABSTRACT

Congenital hypogonadotropic hypogonadism (cHH)/Kallmann syndrome (KS) is a rare genetic disorder with variable penetrance and a complex inheritance pattern. Consequently, it does not always follow Mendelian laws. More recently, digenic and oligogenic transmission has been recognized in 1.5-15% of cases. We report the results of a clinical and genetic investigation of five unrelated patients with cHH/KS analyzed using a customized gene panel. Patients were diagnosed according to the clinical, hormonal, and radiological criteria of the European Consensus Statement. DNA was analyzed using next-generation sequencing with a customized panel that included 31 genes. When available, first-degree relatives of the probands were also analyzed to assess genotype-phenotype segregation. The consequences of the identified variants on gene function were evaluated by analyzing the conservation of amino acids across species and by using molecular modeling. We found one new pathogenic variant of the CHD7 gene (c.576T>A, p.Tyr1928) and three new variants of unknown significance (VUSs) in IL17RD (c.960G>A, p.Met320Ile), FGF17 (c.208G>A, p.Gly70Arg), and DUSP6 (c.434T>G, p.Leu145Arg). All were present in the heterozygous state. Previously reported heterozygous variants were also found in the PROK2 (c.163del, p.Ile55*), CHD7 (c.c.2750C>T, p.Thr917Met and c.7891C>T, p.Arg2631*), FLRT3 (c.1106C>T, p.Ala369Val), and CCDC103 (c.461A>C, p.His154Pro) genes. Molecular modeling, molecular dynamics, and conservation analyses were performed on three out of the nine variants identified in our patients, namely, FGF17 (p.Gly70Arg), DUSP6 (p.Leu145Arg), and CHD7 p.(Thr917Met). Except for DUSP6, where the L145R variant was shown to disrupt the interaction between Ɵ6 and Ɵ3, needed for extracellular signal-regulated kinase 2 (ERK2) binding and recognition, no significant changes were identified between the wild-types and mutants of the other proteins. We found a new pathogenic variant of the CHD7 gene. The molecular modeling results suggest that the VUS of the DUSP6 (c.434T>G, p.Leu145Arg) gene may play a role in the pathogenesis of cHH. However, our analysis indicates that it is unlikely that the VUSs for the IL17RD (c.960G>A, p.Met320Ile) and FGF17 (c.208G>A, p.Gly70Arg) genes are involved in the pathogenesis of cHH. Functional studies are needed to confirm this hypothesis.


Subject(s)
Hypogonadism , Kallmann Syndrome , Humans , Hypogonadism/genetics , Hypogonadism/diagnosis , Kallmann Syndrome/genetics , Phenotype , Heterozygote , Penetrance , Mutation
11.
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
12.
Clin Endocrinol (Oxf) ; 97(6): 804-813, 2022 12.
Article in English | MEDLINE | ID: mdl-36138264

ABSTRACT

OBJECTIVE: To study phenotype-genotype data of Asian-Indian Kallmann syndrome (KS) from our center and systematically review the studies analyzing multiple congenital hypogonadotropic hypogonadism (CHH) genes in KS cohorts using next-generation sequencing. DESIGN, PATIENTS, MEASUREMENT: Five hundred twenty-twoĀ KS probands (our center n = 78, published studies n = 444) were included in this systematic review. Molecular diagnosis was considered if the likely pathogenic/pathogenic variant in known CHH gene/s was reported in the appropriate allelic state. Varsome prediction tool (following American College of Medical Genetics standards) was used to analyze the variants. RESULT: For our center, the molecular diagnosis was seen in 20.5% of probands and was seen more often with severe than partial reproductive phenotype (28.3% vs. 4%, p = .0013). Our center data adds eight novel variants. The molecular diagnosis was seen in 31% as per the systematic review and analysis. It ranged from 16.6% to 72.2% at different centers. The affected genes were FGFR1 (9.8%), ANOS1 (7.5%), PROKR2 (6.1%), CHD7 (5.4%), oligogenic (2.1%), and others <1% each (FGF8, SOX10, PROK2, SEMA3A, IL17RD, andĀ GNRHR). FGFR1 and ANOS1 were the commonly affected genes globally, whereas PROKR2 was commonest in studies from China and CHD7 from Japan, South Korea and Poland. CONCLUSION(S): This systematic review highlights that the genetic yield is 31% in KS probands, with distinct regional variations. The association of severe reproductive phenotype with the higher genetic yield needs further validation.


Subject(s)
Hypogonadism , Kallmann Syndrome , Humans , Kallmann Syndrome/diagnosis , Hypogonadism/pathology , Phenotype , Republic of Korea , China , Mutation
13.
Clin Endocrinol (Oxf) ; 97(5): 604-611, 2022 11.
Article in English | MEDLINE | ID: mdl-35274757

ABSTRACT

OBJECTIVE: Idiopathic hypogonadotropic hypogonadism (IHH) is rare and can either be associated with normal or defective olfactory sensation, classified as normosmic IHH (nIHH) or Kallmann syndrome (KS). We do not yet understand the central processing pathways in the olfactory system. We aimed to compare the resting-state structural and functional connectivity (FC) of olfactory neural pathways in patients with IHH. We hypotheses that alterations of structuralĀ connectivity and FC may exist in the olfactory cortex pathways in IHH patients. DESIGN: STRUCTURAL AND FUNCTIONAL CONNECTIVITY DATA RESULTS BETWEEN TWO GROUPS WERE ANALYZED: Patients: Twenty-five IHH patients (13 nIHH patients and 12 KS patients) were recruited from the Department of Endocrinology and were assessed. A total of 25 age-matched healthy male controls were recruited from the community. MEASUREMENTS: All subjects underwent diffusion tensor imaging and functional magnetic resonance imaging (fMRI) scans. Structural and functional connectivity data analyses were then performed. Pearson's correlation analyses were performed to investigate the correlations between the fractional anisotropy (FA) value and FC strength, showing significant differences among the three groups separately. RESULTS: Compared with the HC group, FA value in the right uncinate fasciculus (UF) decreased significantly in the IHH group. The olfactory cortex FC values of the right gyrus rectus, orbitofrontal cortex (OFC) and right middle temporal gyrus in the IHH group were decreased compared with those in the HC group. Moreover, there were significant negative correlations between right UF FA and olfactory cortex-FC to both the gyrus rectus and OFC within the HC group (p < .05). CONCLUSION: Our findings suggest that alterations of structural and FC support the presence of neurobiological disruptions in IHH patients, particularly a specific structural-functional asymmetry disruption may exist in the olfactory cortex pathways in IHH patients.


Subject(s)
Hypogonadism , Kallmann Syndrome , Diffusion Tensor Imaging , Humans , Limbic System , Male
14.
Endocr J ; 69(7): 831-838, 2022 Jul 28.
Article in English | MEDLINE | ID: mdl-35236788

ABSTRACT

Congenital hypogonadotropic hypogonadism (CHH) is a rare disorder that causes gonadotropin-releasing hormone (GnRH) deficiency and sexual immaturity. CHH may accompany an abnormal sense of smell (Kallmann syndrome, KS) or no such manifestation (normosmic-CHH). This unusual combination of manifestations is explained by the fact that GnRH neurons originate in the olfactory placode and migrate to the forebrain during embryogenesis. We describe the case of a 31-year-old man with normosmic-CHH, who also had obesity, type 2 diabetes and intellectual disability. He was noticed to have sexual immaturity (small testes with no pubic hair) at age 20 years, when diabetic ketoacidosis developed. Basal and GnRH-stimulated levels of LH (1.0Ć¢Ā†Ā’12.0 IU/L) and FSH (1.9Ć¢Ā†Ā’6.1 IU/L) were detectable but low. The results of the T&T olfactometer and the Alinamin test were definitely normal, with an anatomically normal olfactory system on MRI. Sequencing of 22 CHH-related genes was performed, and compound heterozygous PROKR2 variants were identified: one was a previously known loss-of-function variant (p.Trp178Ser) and the other was a nonsense variant (p.Trp212*). Through a literature review, we found 22 patients (including our patient) with CHH due to biallelic PROKR2 variants, which led us to recognize that most of the patients (86%) were diagnosed with KS. Clinical observations in this study indicate that, even though they have CHH, biallelic PROKR2 variant carriers may have a normal olfactory system as well as presumably normal migration of GnRH neurons. This suggests that the PROK2-PROKR2 pathway affects the function of GnRH neurons after their migration.


Subject(s)
Diabetes Mellitus, Type 2 , Hypogonadism , Kallmann Syndrome , Receptors, G-Protein-Coupled , Receptors, Peptide , Adult , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Gonadotropin-Releasing Hormone/genetics , Heterozygote , Humans , Hypogonadism/genetics , Kallmann Syndrome/diagnosis , Kallmann Syndrome/genetics , Male , Mutation , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/genetics , Young Adult
15.
Int J Mol Sci ; 23(8)2022 Apr 17.
Article in English | MEDLINE | ID: mdl-35457241

ABSTRACT

Congenital hypogonadotropic hypogonadism (CHH) is a rare reproductive endocrine disorder characterized by complete or partial failure of pubertal development and infertility due to deficiency of the gonadotropin-releasing hormone (GnRH). CHH has a significant clinical heterogeneity and can be caused by mutations in over 30 genes. The aim of this study was to investigate the genetic defect in two siblings with CHH. A woman with CHH associated with anosmia and her brother with normosmic CHH were investigated by whole exome sequencing. The genetic studies revealed a novel heterozygous missense mutation in the Fibroblast Growth Factor Receptor 1 (FGFR1) gene (NM_023110.3: c.242T>C, p.Ile81Thr) in the affected siblings and in their unaffected father. The mutation affected a conserved amino acid within the first Ig-like domain (D1) of the protein, was predicted to be pathogenic by structure and sequence-based prediction methods, and was absent in ethnically matched controls. These were consistent with a critical role for the identified missense mutation in the activity of the FGFR1 protein. In conclusion, our identification of a novel missense mutation of the FGFR1 gene associated with a variable expression and incomplete penetrance of CHH extends the known mutational spectrum of this gene and may contribute to the understanding of the pathogenesis of CHH.


Subject(s)
Hypogonadism , Kallmann Syndrome , Female , Humans , Hypogonadism/genetics , Hypogonadism/metabolism , Kallmann Syndrome/genetics , Male , Mutation , Mutation, Missense , Portugal , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism
16.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 39(11): 1275-1278, 2022 Nov 10.
Article in Zh | MEDLINE | ID: mdl-36317218

ABSTRACT

OBJECTIVE: To explore the etiology of a patient with Kallmann syndrome (congenital hypogonadism and anosmia) and a 45,X/46,XY karyotype. METHODS: Peripheral venous blood samples were collected from the proband and his parents and subjected to whole exome sequencing. Candidate variants were verified by Sanger sequencing. RESULTS: The proband was found to harbor compound heterozygous variants of the PROKR2 gene, namely c.533G>C (p.W178S) and c.308C>T (p.A103V), which were inherited from his father and mother, respectively. The two variants were respectively predicted to be likely pathogenic and variant of unknown significance, respectively. CONCLUSION: The reduced chromosomal mosaicism might have caused no particular clinical manifestations in this patient. For patients with features of Kallmann syndrome, genetic testing is conducive to early diagnosis and can provide a basis for genetic counseling and clinical treatment.


Subject(s)
Hypogonadism , Kallmann Syndrome , Humans , Genetic Testing , Hypogonadism/genetics , Kallmann Syndrome/genetics , Karyotype , Mutation , Exome Sequencing , Chromosomes, Human, X/genetics , Chromosomes, Human, Y/genetics
17.
Eat Weight Disord ; 27(5): 1903-1906, 2022 Jun.
Article in English | MEDLINE | ID: mdl-34855141

ABSTRACT

BACKGROUND: Although anorexia nervosa might delay puberty, a structured assessment at its onset remains crucial in excluding congenital delayed puberty diseases. CASE PRESENTATION: During the follow-up of a 15-year-old girl suffering from anorexia nervosa, a change of treatment has led to a thorough medical history revealing the absence of the olfactory bulb. Kallmann syndrome diagnosis was made on a blood analysis and the patient was treated with a multidisciplinary approach. CONCLUSION: After the diagnosis, our patient was relieved as it has clarified some of her symptoms including anosmia, poor height and maturational delay. Too often a delayed puberty is attributed to anorexia nervosa itself without considering medical history. This case definitely shows the importance of performing a case history and early diagnosis in pre-pubertal AN to rule out other rare diseases and avoid mid- and long-term sequelae. LEVEL OF EVIDENCE: IV (case study).


Subject(s)
Anorexia Nervosa , Kallmann Syndrome , Puberty, Delayed , Adolescent , Anorexia Nervosa/complications , Anorexia Nervosa/diagnosis , Female , Humans , Kallmann Syndrome/diagnosis
18.
Yi Chuan ; 44(12): 1158-1166, 2022 Dec 20.
Article in English | MEDLINE | ID: mdl-36927561

ABSTRACT

Hypogonadotropic hypogonadism (HH) is a disease defined by dysfunction of the hypothalamic- pituitary-gonadal hormone axis, leading to low sex hormone levels and impaired fertility. HH with anosmia or hyposmia is known as Kallmann syndrome (KS). Waardenburg syndrome (WS) is a rare autosomal dominant genetic disorder characterized by sensorineural hearing loss and abnormal pigmentation. In this report, we collected the clinical data of a patient with hypogonadotropic hypogonadism and congenital hearing loss of unknown cause. The patient had no obvious secondary sexual characteristics development after puberty, and had a heterozygous deletion (at least 419 kb) in 22q13.1 region (Chr.22:38106433-38525560), which covered the SOX10 gene. The abnormalities were not found in gene sequencing analysis of both the parents and sister of the proband. By summarizing and analyzing the characteristics of this case, we further discussed the molecular biological etiological association between HH and WS type 2. This case also enriches the clinical data of subsequent genetic studies, and provides a reference for the diagnosis and treatment of such diseases.


Subject(s)
Hypogonadism , Kallmann Syndrome , Waardenburg Syndrome , Humans , Waardenburg Syndrome/genetics , Waardenburg Syndrome/complications , Gene Deletion , Hypogonadism/genetics , Hypogonadism/complications , Kallmann Syndrome/genetics , Kallmann Syndrome/complications , SOXE Transcription Factors/genetics , Mutation
19.
Yi Chuan ; 44(10): 937-949, 2022 Oct 20.
Article in English | MEDLINE | ID: mdl-36384729

ABSTRACT

Idiopathic hypogonadotropic hypogonadism (IHH) is a rare endocrine disease characterized by gonadal dysplasia. According to whether the sense of smell is affected, this disorder is classified into Kallmann syndrome (KS) and normosmic isolated hypogonadotropic hypogonadism (nIHH). In this study, we reported a case of nIHH patient and explored the pathogenic mechanism of FGFR1 in nIHH. A FGFR1 variant (c.2008G>A, p.E670K) and a CEP290 variant (c.964G>A, p.D322N) were detected by the whole exome sequencing in this nIHH patient. Bioinformatic analysis revealed that this FGFR1 variant (c.2008G>A) causes structural perturbations in TK2 domain demonstrating that this variant result in FGFR1 loss-of-function and abnormal signaling. The identification of an additional CEP290 variant (c.964G>A) indicated that CEP290 might play a potential role in developmental abnormalities and inhibition of GnRH neuron release. A protein interaction network analysis showed that CEP290 was predicted to interact with FGFR1. In summary, our study identified the potential pathogenic mechanism(s) of the novel FGFR1 variant and indicated that CEP290 might play a role in the GnRH neuron migration route. Our findings expand the mutation spectrum of FGFR1 and CEP290 and provide a reference for clinical diagnosis and treatment of IHH.


Subject(s)
Hypogonadism , Kallmann Syndrome , Humans , Hypogonadism/genetics , Hypogonadism/diagnosis , Kallmann Syndrome/genetics , Mutation , Gonadotropin-Releasing Hormone/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Antigens, Neoplasm , Cytoskeletal Proteins/genetics , Cell Cycle Proteins/genetics
20.
J Neurosci ; 40(2): 311-326, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31767679

ABSTRACT

During mammalian development, gonadotropin-releasing-hormone-1 neurons (GnRH-1ns) migrate from the developing vomeronasal organ (VNO) into the brain asserting control of pubertal onset and fertility. Recent data suggest that correct development of the olfactory ensheathing cells (OEC) is imperative for normal GnRH-1 neuronal migration. However, the full ensemble of molecular pathways that regulate OEC development remains to be fully deciphered. Loss-of-function of the transcription factor Gli3 is known to disrupt olfactory development, however, if Gli3 plays a role in GnRH-1 neuronal development is unclear. By analyzing Gli3 extra-toe mutants (Gli3Xt/Xt), we found that Gli3 loss-of-function compromises the onset of achaete-scute family bHLH transcription factor 1 (Ascl-1)+ vomeronasal progenitors and the formation of OEC in the nasal mucosa. Surprisingly, GnRH-1 neurogenesis was intact in Gli3Xt/Xt mice but they displayed significant defects in GnRH-1 neuronal migration. In contrast, Ascl-1null mutants showed reduced neurogenesis for both vomeronasal and GnRH-1ns but less severe defects in OEC development. These observations suggest that Gli3 is critical for OEC development in the nasal mucosa and subsequent GnRH-1 neuronal migration. However, the nonoverlapping phenotypes between Ascl-1 and Gli3 mutants indicate that Ascl-1, while crucial for GnRH-1 neurogenesis, is not required for normal OEC development. Because Kallmann syndrome (KS) is characterized by abnormal GnRH-1ns migration, we examined whole-exome sequencing data from KS subjects. We identified and validated a GLI3 loss-of-function variant in a KS individual. These findings provide new insights into GnRH-1 and OECs development and demonstrate that human GLI3 mutations contribute to KS etiology.SIGNIFICANCE STATEMENT The transcription factor Gli3 is necessary for correct development of the olfactory system. However, if Gli3 plays a role in controlling GnRH-1 neuronal development has not been addressed. We found that Gli3 loss-of-function compromises the onset of Ascl-1+ vomeronasal progenitors, formation of olfactory ensheathing cells in the nasal mucosa, and impairs GnRH-1 neuronal migration to the brain. By analyzing Ascl-1null mutants we dissociated the neurogenic defects observed in Gli3 mutants from lack of olfactory ensheathing cells in the nasal mucosa, moreover, we discovered that Ascl-1 is necessary for GnRH-1 ontogeny. Analyzing human whole-exome sequencing data, we identified a GLI3 loss-of-function variant in a KS individual. Our data suggest that GLI3 is a candidate gene contributing to KS etiology.


Subject(s)
Kallmann Syndrome/genetics , Neurogenesis/physiology , Neuroglia/physiology , Neurons/physiology , Vomeronasal Organ/physiology , Zinc Finger Protein Gli3/metabolism , Animals , Cell Movement/physiology , Female , Gonadotropin-Releasing Hormone/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Olfactory Bulb/growth & development , Olfactory Mucosa/metabolism , Protein Precursors/metabolism , Zinc Finger Protein Gli3/genetics
SELECTION OF CITATIONS
SEARCH DETAIL