Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cerebellum ; 17(3): 380-385, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29248984

ABSTRACT

Autosomal recessive cerebellar ataxias (ARCAs) represent a heterogeneous group of inherited disorders. The association of early-onset cerebellar ataxia with hypogonadotropic hypogonadism is related to two syndromes, known as Gordon Holmes syndrome (GHS-ataxia and pyramidal signs with hypogonadotropic hypogonadism) and Boucher-Neuhäuser syndrome (BNS-ataxia with chorioretinal dystrophy). Mutations in the PNPLA6 gene have been identified as the cause of hereditary spastic paraplegia and complex forms of ataxia associated with retinal and endocrine manifestations. We reported two Brazilian patients with sporadic, progressive cerebellar ataxia, associated with hypogonadotropic hypogonadism, in whom the GHS and BNS were confirmed by the demonstration of compound heterozygote mutations in the PNPLA6 gene. Genetic analysis of the patient 1 revealed compound heterozygous mutations, one allele in exon 34 and the other allele in exon 29. Genetic exam of the patient 2 also demonstrated compound heterozygous mutations. Three were novel mutations. The missense mutation c.3373G> A, found in the BNS patient, was previously related to Oliver-McFarlane syndrome. These different mutations in this gene suggest a complex phenotype associated disease spectrum.


Subject(s)
Cerebellar Ataxia/genetics , Mutation , Phospholipases/genetics , Cerebellar Ataxia/diagnosis , Cerebellar Ataxia/drug therapy , Cerebellar Ataxia/physiopathology , Diagnosis, Differential , Genes, Recessive , Humans , Male , Phenotype , Young Adult
2.
J Clin Endocrinol Metab ; 100(10): E1378-85, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26207952

ABSTRACT

CONTEXT: Loss of function (LoF) mutations in more than 20 genes are now known to cause isolated GnRH deficiency (IGD) in humans. Most causal IGD mutations are typically private, ie, limited to a single individual/pedigree. However, somewhat paradoxically, four IGD genes (GNRH1, TAC3, PROKR2, and GNRHR) have been shown to harbor LoF founder mutations that are shared by multiple unrelated individuals. It is not known whether similar founder mutations occur in other IGD genes. OBJECTIVE: The objective of the study was to determine whether shared deleterious mutations in IGD-associated genes represent founder alleles. SETTING: This study was an international collaboration among academic medical centers. METHODS: IGD patients with shared mutations, defined as those documented in three or more unrelated probands in 14 IGD-associated genes, were identified from various academic institutions, the Human Gene Mutation Database, and literature reports by other international investigators. Haplotypes of single-nucleotide polymorphisms and short tandem repeats surrounding the mutations were constructed to assess genetic ancestry. RESULTS: A total of eight founder mutations in five genes, GNRHR (Q106R, R262Q, R139H), TACR3 (W275X), PROKR2 (R85H), FGFR1 (R250Q, G687R), and HS6ST1 (R382W) were identified. Most founder alleles were present at low frequency in the general population. The estimated age of these mutant alleles ranged from 1925 to 5600 years and corresponded to the time of rapid human population expansion. CONCLUSIONS: We have expanded the spectrum of founder alleles associated with IGD to a total of eight founder mutations. In contrast to the approximately 9000-year-old PROKR2 founder allele that may confer a heterozygote advantage, the rest of the founder alleles are relatively more recent in origin, in keeping with the timing of recent human population expansion and any selective heterozygote advantage of these alleles requires further evaluation.


Subject(s)
Gonadotropin-Releasing Hormone/deficiency , Gonadotropin-Releasing Hormone/genetics , Hypothalamic Diseases/genetics , Mutation , Neurokinin B/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, LHRH/genetics , Receptors, Peptide/genetics , Alleles , Haplotypes , Humans , Pedigree
3.
J Clin Endocrinol Metab ; 100(4): E646-54, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25636053

ABSTRACT

CONTEXT: Delayed puberty (DP) is a common issue and, in the absence of an underlying condition, is typically self limited. Alhough DP seems to be heritable, no specific genetic cause for DP has yet been reported. In contrast, many genetic causes have been found for idiopathic hypogonadotropic hypogonadism (IHH), a rare disorder characterized by absent or stalled pubertal development. OBJECTIVE: The objective of this retrospective study, conducted at academic medical centers, was to determine whether variants in IHH genes contribute to the pathogenesis of DP. SUBJECTS AND OUTCOME MEASURES: Potentially pathogenic variants in IHH genes were identified in two cohorts: 1) DP family members of an IHH proband previously found to have a variant in an IHH gene, with unaffected family members serving as controls, and 2) DP individuals with no family history of IHH, with ethnically matched control subjects drawn from the Exome Aggregation Consortium. RESULTS: In pedigrees with an IHH proband, the proband's variant was shared by 53% (10/19) of DP family members vs 12% (4/33) of unaffected family members (P = .003). In DP subjects with no family history of IHH, 14% (8/56) had potentially pathogenic variants in IHH genes vs 5.6% (1 907/33 855) of controls (P = .01). Potentially pathogenic variants were found in multiple DP subjects for the genes IL17RD and TAC3. CONCLUSIONS: These findings suggest that variants in IHH genes can contribute to the pathogenesis of self-limited DP. Thus, at least in some cases, self-limited DP shares an underlying pathophysiology with IHH.


Subject(s)
Hypogonadism/genetics , Puberty, Delayed/genetics , Adolescent , DNA Mutational Analysis , Extracellular Matrix Proteins/genetics , Female , Humans , Male , Nerve Tissue Proteins/genetics , Pedigree , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/genetics , Receptors, Tachykinin/genetics , Retrospective Studies , Sulfotransferases/genetics , Tachykinins/genetics
4.
Proc Natl Acad Sci U S A ; 111(50): 17953-8, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25472840

ABSTRACT

Inactivating mutations in chromodomain helicase DNA binding protein 7 (CHD7) cause CHARGE syndrome, a severe multiorgan system disorder of which Isolated gonadotropin-releasing hormone (GnRH) deficiency (IGD) is a minor feature. Recent reports have described predominantly missense CHD7 alleles in IGD patients, but it is unclear if these alleles are relevant to causality or overall genetic burden of Kallmann syndrome (KS) and normosmic form of IGD. To address this question, we sequenced CHD7 in 783 well-phenotyped IGD patients lacking full CHARGE features; we identified nonsynonymous rare sequence variants in 5.2% of the IGD cohort (73% missense and 27% splice variants). Functional analyses in zebrafish using a surrogate otolith assay of a representative set of these CHD7 alleles showed that rare sequence variants observed in controls showed no altered function. In contrast, 75% of the IGD-associated alleles were deleterious and resulted in both KS and normosmic IGD. In two families, pathogenic mutations in CHD7 coexisted with mutations in other known IGD genes. Taken together, our data suggest that rare deleterious CHD7 alleles contribute to the mutational burden of patients with both KS and normosmic forms of IGD in the absence of full CHARGE syndrome. These findings (i) implicate a unique role or preferential sensitivity for CHD7 in the ontogeny of GnRH neurons, (ii) reiterate the emerging genetic complexity of this family of IGD disorders, and (iii) demonstrate how the coordinated use of well-phenotyped cohorts, families, and functional studies can inform genetic architecture and provide insights into the developmental biology of cellular systems.


Subject(s)
DNA Helicases/genetics , DNA-Binding Proteins/genetics , Deficiency Diseases/genetics , Gonadotropin-Releasing Hormone/deficiency , Kallmann Syndrome/genetics , Phenotype , Zebrafish/genetics , Animals , Base Sequence , CHARGE Syndrome/genetics , CHARGE Syndrome/pathology , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Gene Knockdown Techniques , Gonadotropin-Releasing Hormone/genetics , Humans , Molecular Sequence Data , Mutation, Missense/genetics , Otolithic Membrane/pathology , Protein Structure, Tertiary , Sequence Analysis, DNA
5.
J Clin Endocrinol Metab ; 98(5): E943-53, 2013 May.
Article in English | MEDLINE | ID: mdl-23533228

ABSTRACT

CONTEXT: The complexity of genetic testing in Kallmann syndrome (KS) is growing and costly. Thus, it is important to leverage the clinical evaluations of KS patients to prioritize genetic screening. OBJECTIVE: The objective of the study was to determine which reproductive and nonreproductive phenotypes of KS subjects have implications for specific gene mutations. SUBJECTS: Two hundred nineteen KS patients were studied: 151 with identified rare sequence variants (RSVs) in 8 genes known to cause KS (KAL1, NELF, CHD7, HS6ST1, FGF8/FGFR1, or PROK2/PROKR2) and 68 KS subjects who remain RSV negative for all 8 genes. MAIN OUTCOME MEASURES: Reproductive and nonreproductive phenotypes within each genetic group were measured. RESULTS: Male KS subjects with KAL1 RSVs displayed the most severe reproductive phenotype with testicular volumes (TVs) at presentation of 1.5 ± 0.1 mL vs 3.7 ± 0.3 mL, P < .05 vs all non-KAL1 probands. In both sexes, synkinesia was enriched but not unique to patients with KAL1 RSVs compared with KAL1-negative probands (43% vs 12%; P < .05). Similarly, dental agenesis and digital bone abnormalities were enriched in patients with RSVs in the FGF8/FGFR1 signaling pathway compared with all other gene groups combined (39% vs 4% and 23% vs 0%; P < .05, respectively). Hearing loss marked the probands with CHD7 RSVs (40% vs 13% in non-CHD7 probands; P < .05). Renal agenesis and cleft lip/palate did not emerge as statistically significant phenotypic predictors. CONCLUSIONS: Certain clinical features in men and women are highly associated with genetic causes of KS. Synkinesia (KAL1), dental agenesis (FGF8/FGFR1), digital bony abnormalities (FGF8/FGFR1), and hearing loss (CHD7) can be useful for prioritizing genetic screening.


Subject(s)
DNA Helicases/genetics , DNA-Binding Proteins/genetics , Extracellular Matrix Proteins/genetics , Fibroblast Growth Factor 8/genetics , Kallmann Syndrome/genetics , Mutation , Nerve Tissue Proteins/genetics , Polymorphism, Genetic , Receptor, Fibroblast Growth Factor, Type 1/genetics , Adult , Anodontia/etiology , Cohort Studies , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Female , Fibroblast Growth Factor 8/metabolism , Finger Phalanges/abnormalities , Genetic Association Studies , Genetic Testing/economics , Health Care Costs , Hearing Loss/etiology , Humans , Kallmann Syndrome/economics , Kallmann Syndrome/metabolism , Kallmann Syndrome/physiopathology , Male , Massachusetts , Nerve Tissue Proteins/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Severity of Illness Index , Signal Transduction , Synkinesis/etiology
6.
Mol Cell Endocrinol ; 346(1-2): 102-9, 2011 Oct 22.
Article in English | MEDLINE | ID: mdl-21664415

ABSTRACT

As our understanding of the complexities of the various etiologies and complex genetic architecture of GnRH deficiency grows, so too does the need to apply newly-developed genetic tools in a way that: (a) is meaningful to individuals and their families; (b) integrates all of the phenotypic features of this syndrome into a rationale; and (c) provides up-to-date diagnostic technologies in a cost-effective algorithm of genetic testing. Genetic counseling aims to accomplish these goals through ascertainment of detailed family histories, targeted comprehensive phenotypic evaluations, informed selection of genetic testing, interpretation of genetic test results, and the provision of highly specific risk assessments and psychological support to individuals diagnosed with this reproductive condition. This chapter offers a guide to incorporating this rapidly evolving state of knowledge of the pedigree and phenotypes into the process of selecting and prioritizing genetic testing. In addition, the provision of risk assessment that accounts for nuanced genetic concepts such as variable expressivity, incomplete penetrance, and oligogenicity, all of which are emerging features of the genetics of this clinical syndrome, is considered. Beyond translating genetic information, genetic counseling should address the psychological impact of embarrassment, shame, anxiety, and guilt that are often seen among individuals with reproductive disorders.


Subject(s)
Genetic Counseling , Genetic Testing , Gonadotropin-Releasing Hormone/deficiency , Animals , Humans , Pedigree , Phenotype , Risk Assessment
SELECTION OF CITATIONS
SEARCH DETAIL
...