Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(10)2023 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-37239833

RESUMO

Type 10 17ß-hydroxysteroid dehydrogenase (17ß-HSD10), a homo-tetrameric multifunctional protein with 1044 residues encoded by the HSD17B10 gene, is necessary for brain cognitive function. Missense mutations result in infantile neurodegeneration, an inborn error in isoleucine metabolism. A 5-methylcytosine hotspot underlying a 388-T transition leads to the HSD10 (p.R130C) mutant to be responsible for approximately half of all cases suffering with this mitochondrial disease. Fewer females suffer with this disease due to X-inactivation. The binding capability of this dehydrogenase to Aß-peptide may play a role in Alzheimer's disease, but it appears unrelated to infantile neurodegeneration. Research on this enzyme was complicated by reports of a purported Aß-peptide-binding alcohol dehydrogenase (ABAD), formerly referred to as endoplasmic-reticulum-associated Aß-binding protein (ERAB). Reports concerning both ABAD and ERAB in the literature reflect features inconsistent with the known functions of 17ß-HSD10. It is clarified here that ERAB is reportedly a longer subunit of 17ß-HSD10 (262 residues). 17ß-HSD10 exhibits L-3-hydroxyacyl-CoA dehydrogenase activity and is thus also referred to in the literature as short-chain 3-hydorxyacyl-CoA dehydrogenase or type II 3-hydorxyacyl-CoA dehydrogenase. However, 17ß-HSD10 is not involved in ketone body metabolism, as reported in the literature for ABAD. Reports in the literature referring to ABAD (i.e., 17ß-HSD10) as a generalized alcohol dehydrogenase, relying on data underlying ABAD's activities, were found to be unreproducible. Furthermore, the rediscovery of ABAD/ERAB's mitochondrial localization did not cite any published research on 17ß-HSD10. Clarification of the purported ABAD/ERAB function derived from these reports on ABAD/ERAB may invigorate this research field and encourage new approaches to the understanding and treatment of HSD17B10-gene-related disorders. We establish here that infantile neurodegeneration is caused by mutants of 17ß-HSD10 but not ABAD, and so we conclude that ABAD represents a misnomer employed in high-impact journals.


Assuntos
3-Hidroxiacil-CoA Desidrogenases , Álcool Desidrogenase , Doença de Alzheimer , Humanos , Álcool Desidrogenase/genética , Doença de Alzheimer/genética , Mutação de Sentido Incorreto
2.
Int J Mol Sci ; 24(24)2023 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-38139430

RESUMO

Type 10 17ß-hydroxysteroid dehydrogenase (17ß-HSD10) is the HSD17B10 gene product playing an appreciable role in cognitive functions. It is the main hub of exercise-upregulated mitochondrial proteins and is involved in a variety of metabolic pathways including neurosteroid metabolism to regulate allopregnanolone homeostasis. Deacetylation of 17ß-HSD10 by sirtuins helps regulate its catalytic activities. 17ß-HSD10 may also play a critical role in the control of mitochondrial structure, morphology and dynamics by acting as a member of the Parkin/PINK1 pathway, and by binding to cyclophilin D to open mitochondrial permeability pore. 17ß-HSD10 also serves as a component of RNase P necessary for mitochondrial tRNA maturation. This dehydrogenase can bind with the Aß peptide thereby enhancing neurotoxicity to brain cells. Even in the absence of Aß, its quantitative and qualitative variations can result in neurodegeneration. Since elevated levels of 17ß-HSD10 were found in brain cells of Alzheimer's disease (AD) patients and mouse AD models, it is considered to be a key factor in AD pathogenesis. Since data underlying Aß-binding-alcohol dehydrogenase (ABAD) were not secured from reported experiments, ABAD appears to be a fabricated alternative term for the HSD17B10 gene product. Results of this study would encourage researchers to solve the question why elevated levels of 17ß-HSD10 are present in brains of AD patients and mouse AD models. Searching specific inhibitors of 17ß-HSD10 may find candidates to reduce senile neurodegeneration and open new approaches for the treatment of AD.


Assuntos
17-Hidroxiesteroide Desidrogenases , Doença de Alzheimer , Animais , Humanos , Camundongos , 17-Hidroxiesteroide Desidrogenases/genética , 17-Hidroxiesteroide Desidrogenases/metabolismo , Álcool Desidrogenase/metabolismo , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo
3.
Am J Med Genet A ; 179(7): 1148-1156, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31050164

RESUMO

Instability of the FMR1 repeat, commonly observed in transmissions of premutation alleles (55-200 repeats), is influenced by the size of the repeat, its internal structure and the sex of the transmitting parent. We assessed these three factors in unstable transmissions of 14/3,335 normal (~5 to 44 repeats), 54/293 intermediate (45-54 repeats), and 1561/1,880 premutation alleles. While most unstable transmissions led to expansions, contractions to smaller repeats were observed in all size classes. For normal alleles, instability was more frequent in paternal transmissions and in alleles with long 3' uninterrupted repeat lengths. For premutation alleles, contractions also occurred more often in paternal than maternal transmissions and the frequency of paternal contractions increased linearly with repeat size. All paternal premutation allele contractions were transmitted as premutation alleles, but maternal premutation allele contractions were transmitted as premutation, intermediate, or normal alleles. The eight losses of AGG interruptions in the FMR1 repeat occurred exclusively in contractions of maternal premutation alleles. We propose a refined model of FMR1 repeat progression from normal to premutation size and suggest that most normal alleles without AGG interruptions are derived from contractions of maternal premutation alleles.


Assuntos
Alelos , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Padrões de Herança , Expansão das Repetições de Trinucleotídeos , Feminino , Síndrome do Cromossomo X Frágil/diagnóstico , Síndrome do Cromossomo X Frágil/patologia , Expressão Gênica , Frequência do Gene , Humanos , Masculino , Linhagem
4.
Genet Med ; 17(5): 358-64, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25210937

RESUMO

PURPOSE: Fragile X CGG repeat alleles often contain one or more AGG interruptions that influence allele stability and risk of a full mutation transmission from parent to child. We have examined transmissions of maternal and paternal alleles with 45-90 repeats to quantify the effect of AGG interruptions on fragile X repeat instability. METHODS: A novel FMR1 polymerase chain reaction assay was used to determine CGG repeat length and AGG interruptions for 1,040 alleles from 705 families. RESULTS: We grouped transmissions into nine categories of five repeats by parental size and found that in every size category, alleles with no AGGs had the greatest risk for instability. For maternal alleles <75 repeats, 89% (24/27) that expanded to a full mutation had no AGGs. Two contractions in maternal transmission were accompanied by loss of AGGs, suggesting a mechanism for generating alleles that lack AGG interruptions. Maternal age was examined as a factor in full mutation expansions using prenatal samples to minimize ascertainment bias, and a possible effect was observed though it was not statistically significant (P = 0.06). CONCLUSION: These results strengthen the association of AGG repeats with CGG repeat stability and provide more accurate risk estimates of full mutation expansions for women with 45-90 repeat alleles.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Heterozigoto , Mutação , Expansão das Repetições de Trinucleotídeos , Fatores Etários , Alelos , Antecipação Genética , Família , Feminino , Síndrome do Cromossomo X Frágil/diagnóstico , Testes Genéticos , Instabilidade Genômica , Humanos , Masculino , Programas de Rastreamento , Mosaicismo , Reação em Cadeia da Polimerase
5.
Hum Mutat ; 35(12): 1485-94, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25224527

RESUMO

Fragile X syndrome (FXS) is the most frequent cause of inherited intellectual disability and autism. It is caused by the absence of the fragile X mental retardation 1 (FMR1) gene product, fragile X mental retardation protein (FMRP), an RNA-binding protein involved in the regulation of translation of a subset of brain mRNAs. In Fmr1 knockout mice, the absence of FMRP results in elevated protein synthesis in the brain as well as increased signaling of many translational regulators. Whether protein synthesis is also dysregulated in FXS patients is not firmly established. Here, we demonstrate that fibroblasts from FXS patients have significantly elevated rates of basal protein synthesis along with increased levels of phosphorylated mechanistic target of rapamycin (p-mTOR), phosphorylated extracellular signal regulated kinase 1/2, and phosphorylated p70 ribosomal S6 kinase 1 (p-S6K1). The treatment with small molecules that inhibit S6K1 and a known FMRP target, phosphoinositide 3-kinase (PI3K) catalytic subunit p110ß, lowered the rates of protein synthesis in both control and patient fibroblasts. Our data thus demonstrate that fibroblasts from FXS patients may be a useful in vitro model to test the efficacy and toxicity of potential therapeutics prior to clinical trials, as well as for drug screening and designing personalized treatment approaches.


Assuntos
Biomarcadores/metabolismo , Síndrome do Cromossomo X Frágil/genética , Animais , Estudos de Casos e Controles , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Fibroblastos/citologia , Fibroblastos/metabolismo , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/tratamento farmacológico , Humanos , Leucina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Fosfatidilinositol 3-Quinases/metabolismo , Biossíntese de Proteínas , RNA Mensageiro/genética , Proteínas Quinases S6 Ribossômicas/metabolismo
6.
BMC Med Genet ; 15: 119, 2014 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-25348928

RESUMO

BACKGROUND: The fragile X syndrome (FXS) results from mutation of the FMR1 gene that prevents expression of its gene product, FMRP. We previously characterized 215 dried blood spots (DBS) representing different FMR1 genotypes and ages with a Luminex-based immunoassay (qFMRP). We found variable FMRP levels in the normal samples and identified affected males by the drastic reduction of FMRP. METHODS: Here, to establish the variability of expression of FMRP in a larger random population we quantified FMRP in 2,000 anonymous fresh newborn DBS. We also evaluated the effect of long term storage on qFMRP by retrospectively assaying 74 aged newborn DBS that had been stored for 7-84 months that included normal and full mutation individuals. These analyses were performed on 3 mm DBS disks. To identify the alleles associated with the lowest FMRP levels in the fresh DBS, we analyzed the DNA in the samples that were more than two standard deviations below the mean. RESULTS: Analysis of the fresh newborn DBS revealed a broad distribution of FMRP with a mean approximately 7-fold higher than that we previously reported for fresh DBS in normal adults and no samples whose FMRP level indicated FXS. DNA analysis of the lowest FMRP DBS showed that this was the low extreme of the normal range and included a female carrying a 165 CGG repeat premutation. In the retrospective study of aged newborn DBS, the FMRP mean of the normal samples was less than 30% of the mean of the fresh DBS. Despite the degraded signal from these aged DBS, qFMRP identified the FXS individuals. CONCLUSIONS: The assay showed that newborn DBS contain high levels of FMRP that will allow identification of males and potentially females, affected by FXS. The assay is also an effective screening tool for aged DBS stored for up to four years.


Assuntos
Teste em Amostras de Sangue Seco/métodos , Proteína do X Frágil da Deficiência Intelectual/sangue , Síndrome do Cromossomo X Frágil/sangue , Preservação de Sangue , Feminino , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Humanos , Recém-Nascido , Masculino , Estudos Retrospectivos , Fatores de Tempo
7.
BMC Biochem ; 14: 17, 2013 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-23834306

RESUMO

BACKGROUND: Hydroxysteroid (17beta) dehydrogenase X (HSD10) is a multifunctional protein encoded by the HSD17B10 gene at Xp11.2. In response to stress or hypoxia-ischemia its levels increase rapidly. Expression of this gene is also elevated significantly in colonic mucosa of the inactive ulcerative colitis patients. However, accurate information about its several transcripts is still lacking, and additional evidence for its escape from X-chromosome inactivation remains to be obtained in order to help settle a debate (He XY, Dobkin C, Yang SY: Does the HSD17B10 gene escape from X-inactivation? Eur J Hum Genet 2011, 19: 123-124). RESULTS: Two major HSD17B10 transcription start sites were identified by primer extension at -37 and -6 as well as a minor start site at -12 nucleotides from the initiation codon ATG. Epigenetic analysis of the 5'-flanking region of the HSD17B10 gene showed that there was little 5-methylcytosine (< 3%) in a normal male, and that none of CpG dinucleotides in the CpG island approached 50% methylation in females. CONCLUSION: The actual length of first exon of the HSD17B10 gene was found to be about a quarter larger than that originally reported. Its transcripts result from a slippery transcription complex. The hypomethylation of the CpG island provides additional evidence for the variable escape of the HSD17B10 gene from X-chromosome inactivation which could influence the range of severity of HSD10 deficiency, an inherited error in isoleucine metabolism, in heterozygous females.


Assuntos
3-Hidroxiacil-CoA Desidrogenases/metabolismo , Epigênese Genética , 3-Hidroxiacil-CoA Desidrogenases/genética , 5-Metilcitosina/química , 5-Metilcitosina/metabolismo , Sequência de Bases , Cromossomos Humanos X , Ilhas de CpG , Metilação de DNA , Éxons , Feminino , Humanos , Masculino , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Sítio de Iniciação de Transcrição
8.
Am J Med Genet A ; 161A(4): 771-8, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23444167

RESUMO

We investigated the effect of AGG interruptions on fragile X repeat instability upon transmission of fragile X intermediate and small premutation alleles with 45-69 CGG repeats. The FMR1 repeat structure was determined for 375 mothers, 48 fathers, and 538 offspring (457 maternal and 81 paternal transmissions) using a novel PCR assay to determine repeat length and AGG interruptions. The number of AGG interruptions and the length of uninterrupted CGG repeats at the 3' end were correlated with repeat instability on transmission. Maternal alleles with no AGGs conferred the greatest risk for unstable transmissions. All nine full mutation expansions were inherited from maternal alleles with no AGGs. Furthermore, the magnitude of repeat expansion was larger for alleles lacking AGG interruptions. Transmissions from paternal alleles with no AGGs also exhibited greater instability than those with one or more AGGs. Our results demonstrate that characterization of the AGG structure within the FMR1 repeat allows more accurate risk estimates of repeat instability and expansion to full mutations for intermediate and small premutation alleles.


Assuntos
Alelos , Síndrome do Cromossomo X Frágil/diagnóstico , Síndrome do Cromossomo X Frágil/genética , Expansão das Repetições de Trinucleotídeos , Feminino , Proteína do X Frágil da Deficiência Intelectual/genética , Instabilidade Genômica , Humanos , Padrões de Herança , Masculino , Mutação , Prognóstico , Risco
9.
J Alzheimers Dis ; 88(4): 1487-1497, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35786658

RESUMO

BACKGROUND: Mitochondrial 17ß-hydroxysteroid dehydrogenase type 10 (17ß-HSD10) is necessary for brain cognitive function, but its studies were confounded by reports of Aß-peptide binding alcohol dehydrogenase (ABAD), formerly endoplasmic reticulum-associated Aß-peptide binding protein (ERAB), for two decades so long as ABAD serves as the alternative term of 17ß-HSD10. OBJECTIVE: To determine whether those ABAD reports are true or false, even if they were published in prestigious journals. METHODS: 6xHis-tagged 17ß-HSD10 was prepared and characterized by well-established experimental procedures. RESULTS: The N-terminal 6xHis tag did not significantly interfere with the dehydrogenase activities of 17ß-HSD10, but the kinetic constants of its 3-hydroxyacyl-CoA dehydrogenase activity are drastically distinct from those of ABAD, and it was not involved in ketone body metabolism as previously reported for ABAD. Furthermore, it was impossible to measure its generalized alcohol dehydrogenase activities underlying the concept of ABAD because the experimental procedures described in ABAD reports violated basic chemical and/or biochemical principles. More incredibly, both authors and journals had not yet agreed to make any corrigenda of ABAD reports. CONCLUSION: Brain 17ß-HSD10 plays a key role in neurosteroid metabolism and further studies in this area may lead to potential treatments of neurodegeneration including AD.


Assuntos
3-Hidroxiacil-CoA Desidrogenases , Doença de Alzheimer , 17-Hidroxiesteroide Desidrogenases , 3-Hidroxiacil-CoA Desidrogenases/metabolismo , Álcool Desidrogenase , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Coenzima A , Humanos
10.
J Neuroinflammation ; 8: 52, 2011 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-21595886

RESUMO

BACKGROUND: Although the cellular mechanisms responsible for the pathogenesis of autism are not understood, a growing number of studies have suggested that localized inflammation of the central nervous system (CNS) may contribute to the development of autism. Recent evidence shows that IL-6 has a crucial role in the development and plasticity of CNS. METHODS: Immunohistochemistry studies were employed to detect the IL-6 expression in the cerebellum of study subjects. In vitro adenoviral gene delivery approach was used to over-express IL-6 in cultured cerebellar granule cells. Cell adhesion and migration assays, DiI labeling, TO-PRO-3 staining and immunofluorescence were used to examine cell adhesion and migration, dendritic spine morphology, cell apoptosis and synaptic protein expression respectively. RESULTS: In this study, we found that IL-6 was significantly increased in the cerebellum of autistic subjects. We investigated how IL-6 affects neural cell development and function by transfecting cultured mouse cerebellar granule cells with an IL-6 viral expression vector. We demonstrated that IL-6 over-expression in granule cells caused impairments in granule cell adhesion and migration but had little effect on the formation of dendritic spines or granule cell apoptosis. However, IL-6 over-expression stimulated the formation of granule cell excitatory synapses, without affecting inhibitory synapses. CONCLUSIONS: Our results provide further evidence that aberrant IL-6 may be associated with autism. In addition, our results suggest that the elevated IL-6 in the autistic brain could alter neural cell adhesion, migration and also cause an imbalance of excitatory and inhibitory circuits. Thus, increased IL-6 expression may be partially responsible for the pathogenesis of autism.


Assuntos
Transtorno Autístico/fisiopatologia , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Cerebelo/metabolismo , Interleucina-6/metabolismo , Neurônios/fisiologia , Sinapses/fisiologia , Animais , Transtorno Autístico/imunologia , Cerebelo/citologia , Criança , Pré-Escolar , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/ultraestrutura , Técnicas de Transferência de Genes , Humanos , Interleucina-6/genética , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/citologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Sinapses/ultraestrutura
11.
Am J Med Genet A ; 155A(4): 870-4, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21595002

RESUMO

Terminal deletions on the X chromosome in female patients may be detected as part of a work up for infertility, premature ovarian insufficiency (POI) or in screening for fragile X carrier status. We present the clinical, cytogenetic and molecular features of four patients with terminal deletions of chromosome X that include the FMR1 gene, and discuss biological and genetic implications of this deletion. Providers should be aware of possible identification of Xq27 deletions as a potential outcome of fragile X screening.


Assuntos
Deleção Cromossômica , Cromossomos Humanos X/genética , Proteína do X Frágil da Deficiência Intelectual/genética , Adulto , Feminino , Deleção de Genes , Humanos
12.
Prenat Diagn ; 31(10): 925-31, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21717484

RESUMO

OBJECTIVE: To determine risks of expansion for normal, intermediate, and premutation FMR1 CGG repeats. METHODS: PCR was used to compare the FMR1 alleles in prenatal (chorionic villi and amniocytes) and parental samples collected from 1991 to 2010. Prenatal diagnoses were confirmed by Southern analysis. RESULTS: Fragile X analysis of 1112 pregnancies identified 558 normal, 106 intermediate, 216 premutation, and 232 full mutation fetuses. Of 509 maternal, intermediate, and premutation alleles, 350 (68.7%) were unstable on transmission with expansions ranging from one repeat to the full mutation. The smallest premutation alleles expanding to the full mutation were in mothers with 65 and 66 repeats. Transmissions from women with or without a family history of fragile X suggested greater instability in women from families that included full mutation expansions. CONCLUSIONS: The maternal transmissions of alleles with 55 to 59 CGG repeats summarized here indicate that the risk for expansion to full mutation is substantially less than previous estimates for this size category. Most premutation alleles with no family history of fragile X exhibited less instability than those with a history of fragile X. Thus, lower risk estimates for full mutation expansion may be appropriate for women newly identified as premutation carriers through routine screening.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Testes Genéticos , Diagnóstico Pré-Natal/métodos , Expansão das Repetições de Trinucleotídeos/genética , Adulto , Feminino , Síndrome do Cromossomo X Frágil/epidemiologia , Instabilidade Genômica , Humanos , Masculino , Mães , Reação em Cadeia da Polimerase , Gravidez , Prevalência , Fatores de Risco , Estados Unidos/epidemiologia
13.
Genes (Basel) ; 12(10)2021 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-34680911

RESUMO

Fragile X syndrome results from the absence of the FMR1 gene product-Fragile X Mental Retardation Protein (FMRP). Fragile X animal research has lacked a reliable method to quantify FMRP. We report the development of an array of FMRP-specific monoclonal antibodies and their application for quantitative assessment of FMRP (qFMRPm) in mouse tissue. To characterize the assay, we determined the normal variability of FMRP expression in four brain structures of six different mouse strains at seven weeks of age. There was a hierarchy of FMRP expression: neocortex > hippocampus > cerebellum > brainstem. The expression of FMRP was highest and least variable in the neocortex, whereas it was most variable in the hippocampus. Male C57Bl/6J and FVB mice were selected to determine FMRP developmental differences in the brain at 3, 7, 10, and 14 weeks of age. We examined the four structures and found a developmental decline in FMRP expression with age, except for the brainstem where it remained stable. qFMRPm assay of blood had highest values in 3 week old animals and dropped by 2.5-fold with age. Sex differences were not significant. The results establish qFMRPm as a valuable tool due to its ease of methodology, cost effectiveness, and accuracy.


Assuntos
Teste em Amostras de Sangue Seco/métodos , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/metabolismo , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Teste em Amostras de Sangue Seco/normas , Feminino , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Imunoensaio/métodos , Imunoensaio/normas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Sensibilidade e Especificidade
15.
Brain Sci ; 10(10)2020 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-33008014

RESUMO

Fragile X syndrome (FXS) is caused by silencing of the FMR1 gene, which encodes a protein with a critical role in synaptic plasticity. The molecular abnormality underlying FMR1 silencing, CGG repeat expansion, is well characterized; however, delineation of the pathway from DNA to RNA to protein using biosamples from well characterized patients with FXS is limited. Since FXS is a common and prototypical genetic disorder associated with intellectual disability (ID) and autism spectrum disorder (ASD), a comprehensive assessment of the FMR1 DNA-RNA-protein pathway and its correlations with the neurobehavioral phenotype is a priority. We applied nine sensitive and quantitative assays evaluating FMR1 DNA, RNA, and FMRP parameters to a reference set of cell lines representing the range of FMR1 expansions. We then used the most informative of these assays on blood and buccal specimens from cohorts of patients with different FMR1 expansions, with emphasis on those with FXS (N = 42 total, N = 31 with FMRP measurements). The group with FMRP data was also evaluated comprehensively in terms of its neurobehavioral profile, which allowed molecular-neurobehavioral correlations. FMR1 CGG repeat expansions, methylation levels, and FMRP levels, in both cell lines and blood samples, were consistent with findings of previous FMR1 genomic and protein studies. They also demonstrated a high level of agreement between blood and buccal specimens. These assays further corroborated previous reports of the relatively high prevalence of methylation mosaicism (slightly over 50% of the samples). Molecular-neurobehavioral correlations confirmed the inverse relationship between overall severity of the FXS phenotype and decrease in FMRP levels (N = 26 males, mean 4.2 ± 3.3 pg FMRP/ng genomic DNA). Other intriguing findings included a significant relationship between the diagnosis of FXS with ASD and two-fold lower levels of FMRP (mean 2.8 ± 1.3 pg FMRP/ng genomic DNA, p = 0.04), in particular observed in younger age- and IQ-adjusted males (mean age 6.9 ± 0.9 years with mean 3.2 ± 1.2 pg FMRP/ng genomic DNA, 57% with severe ASD), compared to FXS without ASD. Those with severe ID had even lower FMRP levels independent of ASD status in the male-only subset. The results underscore the link between FMR1 expansion, gene methylation, and FMRP deficit. The association between FMRP deficiency and overall severity of the neurobehavioral phenotype invites follow up studies in larger patient cohorts. They would be valuable to confirm and potentially extend our initial findings of the relationship between ASD and other neurobehavioral features and the magnitude of FMRP deficit. Molecular profiling of individuals with FXS may have important implications in research and clinical practice.

16.
Neurobiol Dis ; 35(1): 52-62, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19362146

RESUMO

The pre-mRNA of the fragile X mental retardation 1 gene (FMR1) is subject to exon skipping and alternative splice site selection, which can generate up to 12 isoforms. The expression and function of these variants in vivo has not yet been fully explored. In the present study, we investigated the distribution of Fmr1 exon 12 and exon 15 isoforms. Exon 12 encodes an extension of KH(2) domain, one of the RNA binding domains in the FMR1 gene product (FMRP) and we show that exon 12 variant proteins differentially interact with kissing complex RNA. Alternative splicing at exon 15 produces FMRPs differing in RNA binding ability and each is distinguished by unique post-translational modifications. Using semiquantitative RT-PCR and Northern blotting, we found that particular Fmr1 exon 12 and exon 15 isoforms change during neuronal differentiation. Interestingly, Fmr1 exon 12 variants display tissue-specific and developmental differences, while exon 15-containing transcripts vary less. Altogether, the spatio-temporal plasticity of FMR1 mRNA is consistent with complex RNA processing that is mis-regulated in fragile X syndrome.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Regulação da Expressão Gênica/fisiologia , Neurônios/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Antineoplásicos/farmacologia , Células Cultivadas , Córtex Cerebral/citologia , Embrião de Mamíferos , Éxons/genética , Camundongos , Camundongos Mutantes , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Biológicos , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Isoformas de Proteínas/fisiologia , RNA Mensageiro/metabolismo , Sinaptossomos/metabolismo , Distribuição Tecidual , Tretinoína/farmacologia
17.
Am J Med Genet A ; 149A(10): 2152-7, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19764037

RESUMO

The fragile X mutation is an expansion of a CGG triplet repeat in the 5' untranslated region of the FMR1 gene. Expansion to >200 repeats (the "full mutation") silences FMR1 transcription and leads to the fragile X mental retardation syndrome in males and in some females. It also affects the structure of the mitotic chromosome as evidenced by a folate sensitive fragile site. Isolated cases of 45,X/46,XX (mosaic Turner syndrome) in full mutation females have been reported but an increased prevalence was not apparent from these reports. PCR and Southern analysis of the CGG repeat in 423 prenatal female samples identified 106 full mutation cases. Surprisingly five of these had 45,X/4,6XX mosaicism while none of the other 317 female fetuses did. In two of the five cases >or=50% of the cells were reported to be 45,X and in the other three,

Assuntos
Cromossomos Humanos X/genética , Proteína do X Frágil da Deficiência Intelectual/genética , Mosaicismo , Aberrações dos Cromossomos Sexuais , Síndrome de Turner/diagnóstico , Células Cultivadas , Análise Mutacional de DNA , Feminino , Proteína do X Frágil da Deficiência Intelectual/análise , Síndrome do Cromossomo X Frágil/diagnóstico , Síndrome do Cromossomo X Frágil/genética , Deleção de Genes , Frequência do Gene , Disgenesia Gonadal Mista/genética , Humanos , Masculino , Mutação/fisiologia , Gravidez , Diagnóstico Pré-Natal , Fatores de Risco , Síndrome de Turner/etiologia , Síndrome de Turner/genética
18.
Mol Cell Endocrinol ; 489: 92-97, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30321584

RESUMO

17ß-Hydroxysteroid dehydrogenases are indispensable for downstream enzyme steps of the neurosteroidogenesis. Neurosteroids are synthesized de novo in neurons and glia from cholesterol transported into mitochondria, or by conversion from proneurosteroids, e. g. dehydroepiandrosterone (DHEA) and pregnenolone, through the same metabolic pathway as revealed in the de novo neurosteroidogenesis. Hormonal steroids generated from endocrine glands are transported into brain from the circulation to exert neuronal activity via genomic pathway, whereas neurosteroids produced in brain cells without genomic targets identified could bind to cell surface targets, e.g., GABAA or NMDA receptors and elicit antidepressant, anxiolytic, anticonvulsant and anesthetic effects by regulating neuroexcitability. In a broad sense, neurosteroids include hormonal steroids in brain, and they are irrespective of origin playing important roles in brain development including neuroprotection, neurogenesis and neural plasticity. They are also a critical element in cognitive and memory functions. Mitochondrial 17ß-HSD10, encoded by the HSD17B10 gene mapping to Xp11.2, is found in various brain regions, essential for the maintenance of neurosteroid homeostasis. Mutations identified in this gene resulted in two distinct brain diseases, namely HSD10 deficiency and MRXS10, of which clinical presentations and pathogenetic mechanisms are quite different. Since elevated levels of 17ß-HSD10 was found in brains of Alzheimer's disease patients and AD mouse model, it may also act as an adverse factor in the AD pathogenesis due to an imbalance of neurosteroid metabolism.


Assuntos
17-Hidroxiesteroide Desidrogenases/metabolismo , Sistema Nervoso Central/metabolismo , Neuroesteroides/metabolismo , 17-Hidroxiesteroide Desidrogenases/genética , Animais , Homeostase , Humanos , Mutação de Sentido Incorreto/genética
19.
Am J Med Genet A ; 146A(1): 60-5, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18074381

RESUMO

Southern analysis of the FMR1 repeat region has suggested that individuals with the full mutation usually carry a heterogeneous array of FMR1 alleles in somatic tissue that can range from 200 to more than 1,000 repeats. Our studies indicate that this heterogeneity is an artifact generated by ethidium bromide commonly used in Southern analysis. When analyzed in the absence of ethidium bromide, nearly all full mutation individuals carried only one to four major alleles and did not exhibit the heterogeneity often referred to as a "smear" in the literature. Full mutations in chorionic villi, however, exhibited much greater heterogeneity. Nine transmissions from mothers with full mutation alleles to offspring indicated that the full mutations continued to expand in transmission to the next generation. In contrast, analysis of leukocyte DNA from three full mutation males revealed no change in somatic full mutation alleles over many years. Our studies support the hypothesis that the FMR1 CGG repeat instability is limited to very early embryogenesis in the soma. These studies also have clinical importance because the omission of ethidium bromide will facilitate the diagnosis of females with full mutation alleles.


Assuntos
Alelos , Artefatos , Síndrome do Cromossomo X Frágil/genética , Mutação , Expansão das Repetições de Trinucleotídeos/genética , Regiões 5' não Traduzidas , Southern Blotting , Estudos de Casos e Controles , Feminino , Síndrome do Cromossomo X Frágil/diagnóstico , Heterogeneidade Genética , Humanos , Masculino , Linhagem , Análise de Sequência de DNA
20.
Neurosci Lett ; 377(3): 141-6, 2005 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-15755515

RESUMO

The fragile X mental retardation syndrome is due to the transcriptional silence of the fragile X gene, FMR1, and to the resulting loss of the FMR1 product, FMRP. The pathogenesis of the syndrome, however, is not understood. Increased prevalence of childhood seizures is a feature of the fragile X syndrome and increased seizure susceptibility is seen in the fragile X knock out mouse model for this disorder. To investigate the increased seizure susceptibility, we examined GABA(A) receptor expression in the FVB/N fragile X mouse. Western blot analysis revealed that expression of the GABA(A) receptor beta subunit (GABA(A) beta), which is required for receptor function, was reduced in the cortex, hippocampus, diencephalon and brainstem in adult male fragile X mice. Immunohistochemical analysis of brain sections indicated a reduction in GABA(A) beta immunoreactivity. We also found increased expression of glutamic acid decarboxylase, the enzyme responsible for GABA synthesis, in the same regions that showed GABA(A) beta reduction. These results indicate that the absence of Fmrp leads to GABAergic system alterations that could account for the increased seizure susceptibility of the fragile X mouse. These alterations may also be relevant to the seizures and the abnormal behaviors in the human syndrome.


Assuntos
Síndrome do Cromossomo X Frágil/metabolismo , Receptores de GABA-A/biossíntese , Convulsões/metabolismo , Animais , Síndrome do Cromossomo X Frágil/genética , Regulação da Expressão Gênica/fisiologia , Masculino , Camundongos , Camundongos Congênicos , Camundongos Knockout , Proteínas de Ligação a RNA/biossíntese , Proteínas de Ligação a RNA/genética , Receptores de GABA-A/genética , Convulsões/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA