Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Am J Med Genet A ; 179(2): 295-299, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30556293

RESUMO

Grange syndrome is an autosomal recessive condition characterized by arterial occlusions and hypertension. Syndactyly, brachydactyly, bone fragility, heart defects, and learning disabilities have also been reported. Loss-of-function variants in YY1AP1 have only recently been associated with Grange syndrome. YY1AP1 encodes for the transcription coactivator yin yang 1-associated protein 1 which regulates smooth muscle cell proliferation and differentiation. We here report on three siblings with steno-occlusive arterial disorder and syndactyly in two of them. Whole exome sequencing including near-splice regions led to the identification of two intronic YY1AP1 variants which were predicted to interfere with normal splicing. Sanger sequencing demonstrated compound-heterozygosity in all affected siblings. RT-PCR analyses confirmed skipping of exon 6 on one allele and exonization of 22 bp in intron 6 on the other. This is the first report of biallelic YY1AP1 variants in noncoding regions and just the second family with multiple affected siblings. Therefore, our report further delineates the phenotypic spectrum of Grange syndrome.


Assuntos
Arteriopatias Oclusivas/genética , Osso e Ossos/anormalidades , Braquidactilia/genética , Proteínas de Ciclo Celular/genética , Predisposição Genética para Doença , Cardiopatias Congênitas/genética , Hipertensão/genética , Sindactilia/genética , Fatores de Transcrição/genética , Adolescente , Adulto , Arteriopatias Oclusivas/fisiopatologia , Osso e Ossos/fisiopatologia , Braquidactilia/fisiopatologia , Criança , Feminino , Cardiopatias Congênitas/fisiopatologia , Humanos , Hipertensão/fisiopatologia , Masculino , Pessoa de Meia-Idade , Linhagem , Isoformas de Proteínas/genética , Sindactilia/fisiopatologia , Sequenciamento do Exoma
2.
Nat Metab ; 1(2): 222-235, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-32694784

RESUMO

Heterogeneous populations of hypothalamic neurons orchestrate energy balance via the release of specific signatures of neuropeptides. However, how specific intracellular machinery controls peptidergic identities and function of individual hypothalamic neurons remains largely unknown. The transcription factor T-box 3 (Tbx3) is expressed in hypothalamic neurons sensing and governing energy status, whereas human TBX3 haploinsufficiency has been linked with obesity. Here, we demonstrate that loss of Tbx3 function in hypothalamic neurons causes weight gain and other metabolic disturbances by disrupting both the peptidergic identity and plasticity of Pomc/Cart and Agrp/Npy neurons. These alterations are observed after loss of Tbx3 in both immature hypothalamic neurons and terminally differentiated mouse neurons. We further establish the importance of Tbx3 for body weight regulation in Drosophila melanogaster and show that TBX3 is implicated in the differentiation of human embryonic stem cells into hypothalamic Pomc neurons. Our data indicate that Tbx3 directs the terminal specification of neurons as functional components of the melanocortin system and is required for maintaining their peptidergic identity. In summary, we report the discovery of a key mechanistic process underlying the functional heterogeneity of hypothalamic neurons governing body weight and systemic metabolism.


Assuntos
Hipotálamo/metabolismo , Melanocortinas/metabolismo , Neurônios/metabolismo , Proteínas com Domínio T/metabolismo , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Animais , Peso Corporal , Metabolismo Energético , Perfilação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Hipotálamo/citologia , Camundongos , Camundongos Endogâmicos C57BL , Pró-Opiomelanocortina/genética , RNA Mensageiro/genética , Proteínas com Domínio T/genética
3.
Orphanet J Rare Dis ; 13(1): 120, 2018 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-30025539

RESUMO

BACKGROUND: Mitochondrial acyl-CoA dehydrogenase family member 9 (ACAD9) is essential for the assembly of mitochondrial respiratory chain complex I. Disease causing biallelic variants in ACAD9 have been reported in individuals presenting with lactic acidosis and cardiomyopathy. RESULTS: We describe the genetic, clinical and biochemical findings in a cohort of 70 patients, of whom 29 previously unpublished. We found 34 known and 18 previously unreported variants in ACAD9. No patients harbored biallelic loss of function mutations, indicating that this combination is unlikely to be compatible with life. Causal pathogenic variants were distributed throughout the entire gene, and there was no obvious genotype-phenotype correlation. Most of the patients presented in the first year of life. For this subgroup the survival was poor (50% not surviving the first 2 years) comparing to patients with a later presentation (more than 90% surviving 10 years). The most common clinical findings were cardiomyopathy (85%), muscular weakness (75%) and exercise intolerance (72%). Interestingly, severe intellectual deficits were only reported in one patient and severe developmental delays in four patients. More than 70% of the patients were able to perform the same activities of daily living when compared to peers. CONCLUSIONS: Our data show that riboflavin treatment improves complex I activity in the majority of patient-derived fibroblasts tested. This effect was also reported for most of the treated patients and is mirrored in the survival data. In the patient group with disease-onset below 1 year of age, we observed a statistically-significant better survival for patients treated with riboflavin.


Assuntos
Acidose/genética , Acidose/metabolismo , Acil-CoA Desidrogenase/deficiência , Erros Inatos do Metabolismo dos Aminoácidos/genética , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/metabolismo , Doenças Mitocondriais/genética , Doenças Mitocondriais/metabolismo , Debilidade Muscular/genética , Debilidade Muscular/metabolismo , Riboflavina/uso terapêutico , Acidose/patologia , Atividades Cotidianas , Acil-CoA Desidrogenase/genética , Acil-CoA Desidrogenase/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/patologia , Cardiomiopatia Hipertrófica/patologia , Complexo I de Transporte de Elétrons/metabolismo , Feminino , Humanos , Masculino , Doenças Mitocondriais/patologia , Debilidade Muscular/tratamento farmacológico , Debilidade Muscular/patologia , Prognóstico
4.
Cell Metab ; 26(4): 620-632.e6, 2017 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-28943448

RESUMO

Chronic inflammation has been proposed to contribute to the pathogenesis of diet-induced obesity. However, scarce therapeutic options are available to treat obesity and the associated immunometabolic complications. Glucocorticoids are routinely employed for the management of inflammatory diseases, but their pleiotropic nature leads to detrimental metabolic side effects. We developed a glucagon-like peptide-1 (GLP-1)-dexamethasone co-agonist in which GLP-1 selectively delivers dexamethasone to GLP-1 receptor-expressing cells. GLP-1-dexamethasone lowers body weight up to 25% in obese mice by targeting the hypothalamic control of feeding and by increasing energy expenditure. This strategy reverses hypothalamic and systemic inflammation while improving glucose tolerance and insulin sensitivity. The selective preference for GLP-1 receptor bypasses deleterious effects of dexamethasone on glucose handling, bone integrity, and hypothalamus-pituitary-adrenal axis activity. Thus, GLP-1-directed glucocorticoid pharmacology represents a safe and efficacious therapy option for diet-induced immunometabolic derangements and the resulting obesity.


Assuntos
Dexametasona/uso terapêutico , Peptídeo 1 Semelhante ao Glucagon/uso terapêutico , Glucocorticoides/uso terapêutico , Incretinas/uso terapêutico , Inflamação/tratamento farmacológico , Obesidade/tratamento farmacológico , Animais , Peso Corporal/efeitos dos fármacos , Dexametasona/análogos & derivados , Metabolismo Energético/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Glucocorticoides/química , Glucose/metabolismo , Células HEK293 , Humanos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Incretinas/química , Inflamação/complicações , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/complicações , Obesidade/metabolismo
5.
Am J Hum Genet ; 101(2): 283-290, 2017 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-28757203

RESUMO

Lipoate serves as a cofactor for the glycine cleavage system (GCS) and four 2-oxoacid dehydrogenases functioning in energy metabolism (α-oxoglutarate dehydrogenase [α-KGDHc] and pyruvate dehydrogenase [PDHc]), or amino acid metabolism (branched-chain oxoacid dehydrogenase, 2-oxoadipate dehydrogenase). Mitochondrial lipoate synthesis involves three enzymatic steps catalyzed sequentially by lipoyl(octanoyl) transferase 2 (LIPT2), lipoic acid synthetase (LIAS), and lipoyltransferase 1 (LIPT1). Mutations in LIAS have been associated with nonketotic hyperglycinemia-like early-onset convulsions and encephalopathy combined with a defect in mitochondrial energy metabolism. LIPT1 deficiency spares GCS deficiency and has been associated with a biochemical signature of combined 2-oxoacid dehydrogenase deficiency leading to early death or Leigh-like encephalopathy. We report on the identification of biallelic LIPT2 mutations in three affected individuals from two families with severe neonatal encephalopathy. Brain MRI showed major cortical atrophy with white matter abnormalities and cysts. Plasma glycine was mildly increased. Affected individuals' fibroblasts showed reduced oxygen consumption rates, PDHc, α-KGDHc activities, leucine catabolic flux, and decreased protein lipoylation. A normalization of lipoylation was observed after expression of wild-type LIPT2, arguing for LIPT2 requirement in intramitochondrial lipoate synthesis. Lipoic acid supplementation did not improve clinical condition nor activities of PDHc, α-KGDHc, or leucine metabolism in fibroblasts and was ineffective in yeast deleted for the orthologous LIP2.


Assuntos
Aciltransferases/genética , Atrofia/patologia , Encefalopatias/genética , Encéfalo/patologia , Lipoilação/genética , Mitocôndrias/metabolismo , Aminoácidos/metabolismo , Encéfalo/diagnóstico por imagem , Encefalopatias/patologia , Mapeamento Encefálico/métodos , Células Cultivadas , Metabolismo Energético/genética , Metabolismo Energético/fisiologia , Glicina/sangue , Humanos , Recém-Nascido , Imageamento por Ressonância Magnética , Mitocôndrias/genética , Consumo de Oxigênio/genética , Ligação Proteica/genética , Ácido Tióctico/metabolismo
6.
Am J Hum Genet ; 99(4): 894-902, 2016 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-27616477

RESUMO

To safeguard the cell from the accumulation of potentially harmful metabolic intermediates, specific repair mechanisms have evolved. APOA1BP, now renamed NAXE, encodes an epimerase essential in the cellular metabolite repair for NADHX and NADPHX. The enzyme catalyzes the epimerization of NAD(P)HX, thereby avoiding the accumulation of toxic metabolites. The clinical importance of the NAD(P)HX repair system has been unknown. Exome sequencing revealed pathogenic biallelic mutations in NAXE in children from four families with (sub-) acute-onset ataxia, cerebellar edema, spinal myelopathy, and skin lesions. Lactate was elevated in cerebrospinal fluid of all affected individuals. Disease onset was during the second year of life and clinical signs as well as episodes of deterioration were triggered by febrile infections. Disease course was rapidly progressive, leading to coma, global brain atrophy, and finally to death in all affected individuals. NAXE levels were undetectable in fibroblasts from affected individuals of two families. In these fibroblasts we measured highly elevated concentrations of the toxic metabolite cyclic-NADHX, confirming a deficiency of the mitochondrial NAD(P)HX repair system. Finally, NAD or nicotinic acid (vitamin B3) supplementation might have therapeutic implications for this fatal disorder.


Assuntos
Proteínas de Transporte/genética , Doenças Metabólicas/genética , Mutação , NAD/análogos & derivados , Doenças do Sistema Nervoso/genética , Racemases e Epimerases/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Pré-Escolar , Evolução Fatal , Feminino , Fibroblastos , Humanos , Lactente , Masculino , Doenças Metabólicas/metabolismo , Doenças Metabólicas/patologia , NAD/metabolismo , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/patologia , Neuroimagem , Anormalidades da Pele/genética , Anormalidades da Pele/patologia
7.
Am J Hum Genet ; 99(2): 414-22, 2016 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-27426735

RESUMO

tRNA synthetase deficiencies are a growing group of genetic diseases associated with tissue-specific, mostly neurological, phenotypes. In cattle, cytosolic isoleucyl-tRNA synthetase (IARS) missense mutations cause hereditary weak calf syndrome. Exome sequencing in three unrelated individuals with severe prenatal-onset growth retardation, intellectual disability, and muscular hypotonia revealed biallelic mutations in IARS. Studies in yeast confirmed the pathogenicity of identified mutations. Two of the individuals had infantile hepatopathy with fibrosis and steatosis, leading in one to liver failure in the course of infections. Zinc deficiency was present in all affected individuals and supplementation with zinc showed a beneficial effect on growth in one.


Assuntos
Alelos , Retardo do Crescimento Fetal/genética , Deficiência Intelectual/genética , Isoleucina-tRNA Ligase/genética , Hepatopatias/congênito , Hepatopatias/genética , Hipotonia Muscular/congênito , Hipotonia Muscular/genética , Mutação , Adolescente , Animais , Criança , Pré-Escolar , Suplementos Nutricionais , Fígado Gorduroso/genética , Feminino , Fibrose/genética , Humanos , Lactente , Recém-Nascido , Isoleucina-tRNA Ligase/deficiência , Falência Hepática/genética , Masculino , Síndrome , Peixe-Zebra/genética , Zinco/administração & dosagem , Zinco/deficiência , Zinco/uso terapêutico
8.
Eur J Hum Genet ; 24(3): 450-4, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26081641

RESUMO

Coenzyme Q10 (CoQ10) has an important role in mitochondrial energy metabolism by way of its functioning as an electron carrier in the respiratory chain. Genetic defects disrupting the endogenous biosynthesis pathway of CoQ10 may lead to severe metabolic disorders with onset in early childhood. Using exome sequencing in a child with fatal neonatal lactic acidosis and encephalopathy, we identified a homozygous loss-of-function variant in COQ9. Functional studies in patient fibroblasts showed that the absence of the COQ9 protein was concomitant with a strong reduction of COQ7, leading to a significant accumulation of the substrate of COQ7, 6-demethoxy ubiquinone10. At the same time, the total amount of CoQ10 was severely reduced, which was reflected in a significant decrease of mitochondrial respiratory chain succinate-cytochrome c oxidoreductase (complex II/III) activity. Lentiviral expression of COQ9 restored all these parameters, confirming the causal role of the variant. Our report on the second COQ9 patient expands the clinical spectrum associated with COQ9 variants, indicating the importance of COQ9 already during prenatal development. Moreover, the rescue of cellular CoQ10 levels and respiratory chain complex activities by CoQ10 supplementation points to the importance of an early diagnosis and immediate treatment.


Assuntos
Acidose Láctica/complicações , Acidose Láctica/genética , Encefalopatias/complicações , Encefalopatias/genética , Proteínas Mitocondriais/genética , Mutação/genética , Ubiquinona/genética , Encéfalo/patologia , Encefalopatias/diagnóstico por imagem , Complexo de Proteínas da Cadeia de Transporte de Elétrons/genética , Evolução Fatal , Homozigoto , Humanos , Recém-Nascido , Masculino , Ultrassonografia
9.
J Clin Endocrinol Metab ; 100(5): E776-82, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25763608

RESUMO

CONTEXT: Insulinomas represent pancreatic neuroendocrine neoplasms that cause severe morbidity attributed to their often pronounced endocrine activity. Apart from hereditary forms such as multiple endocrine neoplasia type 1 (MEN-1), genetic causes for sporadic insulinoma development had remained obscure until recently. Applying next-generation sequencing methods, disease-causing genetic alterations have been identified in various endocrine tumors. OBJECTIVE AND DESIGN: Paired tumor and blood DNA from eight patients with sporadic insulinomas (five females and two malignant tumors) were analyzed by whole-exome sequencing. After this initial analysis, Ying Yang 1 (YY1) mutation status was assessed in a larger cohort of 39 additional insulinomas (including eight malignant and one liver metastasis) from three German hospitals by targeted sequencing. The mutation status was correlated with various clinical parameters. RESULTS: A range of one to 12 somatic genetic variants were identified by exome sequencing. A recurrent somatic Thr372Arg YY1 point mutation was detected in two patients of the initial cohort and four patients of the second cohort (total, six of 47; 13%). The presence of the mutation was associated with a trend toward higher age (63.5 y; IQR, 48.0-74.0 vs 45.0 y; IQR, 33.0-63.0; P = .05), and all affected patients were females (six of six; P = .04). All other clinical parameters, including the presence of malignancy and metastatic spread, tumor localization, and hypoglycemic episodes were not different between YY1-mutated and nonmutated tumor carriers. CONCLUSIONS: The somatic Thr372Arg YY1 mutation is a relevant finding in female patients with sporadic insulinomas. The prevalence of this mutation in this Caucasian population is considerably lower compared to that of a recently described Asian cohort.


Assuntos
Insulinoma/genética , Mutação , Neoplasias Pancreáticas/genética , Fator de Transcrição YY1/genética , Adulto , Idoso , Exoma , Feminino , Humanos , Insulinoma/patologia , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/patologia
10.
J Inherit Metab Dis ; 35(6): 943-8, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22864630

RESUMO

Brown-Vialetto-Van Laere syndrome (BVVLS [MIM 211530]) is a rare neurological disorder characterized by infancy onset sensorineural deafness and ponto-bulbar palsy. Mutations in SLC52A3 (formerly C20orf54), coding for riboflavin transporter 2 (hRFT2), have been identified as the molecular genetic correlate in several individuals with BVVLS. Exome sequencing of just one single case revealed that compound heterozygosity for two pathogenic mutations in the SLC52A2 gene coding for riboflavin transporter 3 (hRFT3), another member of the riboflavin transporter family, is also associated with BVVLS. Overexpression studies confirmed that the gene products of both mutant alleles have reduced riboflavin transport activities. While mutations in SLC52A3 cause decreased plasma riboflavin levels, concordant with a role of SLC52A3 in riboflavin uptake from food, the SLC52A2-mutant individual had normal plasma riboflavin concentrations, a finding in line with a postulated function of SLC52A2 in riboflavin uptake from blood into target cells. Our results contribute to the understanding of human riboflavin metabolism and underscore its role in the pathogenesis of BVVLS, thereby providing a rational basis for a high-dose riboflavin treatment.


Assuntos
Paralisia Bulbar Progressiva/genética , Paralisia Bulbar Progressiva/metabolismo , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/metabolismo , Mutação de Sentido Incorreto , Receptores Acoplados a Proteínas G/genética , Riboflavina/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Transporte Biológico Ativo/genética , Paralisia Bulbar Progressiva/diagnóstico , Pré-Escolar , Análise Mutacional de DNA , Feminino , Perda Auditiva Neurossensorial/diagnóstico , Humanos , Proteínas de Membrana Transportadoras/deficiência , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Modelos Biológicos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/metabolismo , Homologia de Sequência de Aminoácidos , Síndrome
11.
Brain ; 135(Pt 5): 1387-94, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22492562

RESUMO

In the large group of genetically undetermined infantile-onset mitochondrial encephalopathies, multiple defects of mitochondrial DNA-related respiratory-chain complexes constitute a frequent biochemical signature. In order to identify responsible genes, we used exome-next-generation sequencing in a selected cohort of patients with this biochemical signature. In an isolated patient, we found two mutant alleles for EARS2, the gene encoding mitochondrial glutamyl-tRNA synthetase. The brain magnetic resonance imaging of this patient was hallmarked by extensive symmetrical cerebral white matter abnormalities sparing the periventricular rim and symmetrical signal abnormalities of the thalami, midbrain, pons, medulla oblongata and cerebellar white matter. Proton magnetic resonance spectroscopy showed increased lactate. We matched this magnetic resonance imaging pattern with that of a cohort of 11 previously selected unrelated cases. We found mutations in the EARS2 gene in all. Subsequent detailed clinical and magnetic resonance imaging based phenotyping revealed two distinct groups: mild and severe. All 12 patients shared an infantile onset and rapidly progressive disease with severe magnetic resonance imaging abnormalities and increased lactate in body fluids and proton magnetic resonance spectroscopy. Patients in the 'mild' group partially recovered and regained milestones in the following years with striking magnetic resonance imaging improvement and declining lactate levels, whereas those of the 'severe' group were characterized by clinical stagnation, brain atrophy on magnetic resonance imaging and persistent lactate increases. This new neurological disease, early-onset leukoencephalopathy with thalamus and brainstem involvement and high lactate, is hallmarked by unique magnetic resonance imaging features, defined by a peculiar biphasic clinical course and caused by mutations in a single gene, EARS2, expanding the list of medically relevant defects of mitochondrial DNA translation.


Assuntos
Tronco Encefálico/patologia , Glutamato-tRNA Ligase/genética , Ácido Láctico/metabolismo , Leucoencefalopatias , Mutação/genética , Tálamo/patologia , Células Cultivadas , Criança , Análise Mutacional de DNA , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Feminino , Fibroblastos/fisiologia , Humanos , Leucoencefalopatias/genética , Leucoencefalopatias/metabolismo , Leucoencefalopatias/patologia , Imageamento por Ressonância Magnética , Espectroscopia de Ressonância Magnética , Masculino , Proteínas Mitocondriais/genética , Consumo de Oxigênio/genética , Consumo de Oxigênio/fisiologia , Prótons , Pele/patologia
12.
J Am Soc Nephrol ; 21(7): 1223-32, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20558539

RESUMO

Phosphorus is an essential mineral that maintains cellular energy and mineralizes the skeleton. Because complex actions of ion transporters and regulatory hormones regulate serum phosphorus concentrations, genetic variation may determine interindividual variation in phosphorus metabolism. Here, we report a comprehensive genome-wide association study of serum phosphorus concentration. We evaluated 16,264 participants of European ancestry from the Cardiovascular Heath Study, Atherosclerosis Risk in Communities Study, Framingham Offspring Study, and the Rotterdam Study. We excluded participants with an estimated GFR <45 ml/min per 1.73 m(2) to focus on phosphorus metabolism under normal conditions. We imputed genotypes to approximately 2.5 million single-nucleotide polymorphisms in the HapMap and combined study-specific findings using meta-analysis. We tested top polymorphisms from discovery cohorts in a 5444-person replication sample. Polymorphisms in seven loci with minor allele frequencies 0.08 to 0.49 associate with serum phosphorus concentration (P = 3.5 x 10(-16) to 3.6 x 10(-7)). Three loci were near genes encoding the kidney-specific type IIa sodium phosphate co-transporter (SLC34A1), the calcium-sensing receptor (CASR), and fibroblast growth factor 23 (FGF23), proteins that contribute to phosphorus metabolism. We also identified genes encoding phosphatases, kinases, and phosphodiesterases that have yet-undetermined roles in phosphorus homeostasis. In the replication sample, five of seven top polymorphisms associate with serum phosphorous concentrations (P < 0.05 for each). In conclusion, common genetic variants associate with serum phosphorus in the general population. Further study of the loci identified in this study may help elucidate mechanisms of phosphorus regulation.


Assuntos
Loci Gênicos/genética , Variação Genética/genética , Estudo de Associação Genômica Ampla , Fósforo/sangue , Polimorfismo de Nucleotídeo Único/genética , Adulto , Idoso , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/genética , Frequência do Gene/genética , Humanos , Rim/fisiologia , Masculino , Pessoa de Meia-Idade , Receptores de Detecção de Cálcio/genética , Fatores Sexuais , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/genética , População Branca
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA