Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Int J Mol Sci ; 20(21)2019 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-31653080

RESUMO

Lysinuric protein intolerance (LPI) is a rare autosomal disease caused by defective cationic amino acid (CAA) transport due to mutations in SLC7A7, which encodes for the y+LAT1 transporter. LPI patients suffer from a wide variety of symptoms, which range from failure to thrive, hyperammonemia, and nephropathy to pulmonar alveolar proteinosis (PAP), a potentially life-threatening complication. Hyperammonemia is currently prevented by citrulline supplementation. However, the full impact of this treatment is not completely understood. In contrast, there is no defined therapy for the multiple reported complications of LPI, including PAP, for which bronchoalveolar lavages do not prevent progression of the disease. The lack of a viable LPI model prompted us to generate a tamoxifen-inducible Slc7a7 knockout mouse (Slc7a7-/-). The Slc7a7-/- model resembles the human LPI phenotype, including malabsorption and impaired reabsorption of CAA, hypoargininemia and hyperammonemia. Interestingly, the Slc7a7-/- mice also develops PAP and neurological impairment. We observed that citrulline treatment improves the metabolic derangement and survival. On the basis of our findings, the Slc7a7-/- model emerges as a promising tool to further study the complexity of LPI, including its immune-like complications, and to design evidence-based therapies to halt its progression.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/patologia , Sistema y+L de Transporte de Aminoácidos/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/tratamento farmacológico , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Sistema y+L de Transporte de Aminoácidos/genética , Aminoácidos/metabolismo , Animais , Citrulina/uso terapêutico , Modelos Animais de Doenças , Hiperamonemia/metabolismo , Hiperamonemia/patologia , Mucosa Intestinal/metabolismo , Rim/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteinose Alveolar Pulmonar/metabolismo , Proteinose Alveolar Pulmonar/patologia
2.
Am J Med Genet C Semin Med Genet ; 157C(1): 54-62, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21308987

RESUMO

Lysinuric protein intolerance (LPI) is an inherited aminoaciduria caused by defective cationic amino acid transport at the basolateral membrane of epithelial cells in intestine and kidney. LPI is caused by mutations in the SLC7A7 gene, which encodes the y(+)LAT-1 protein, the catalytic light chain subunit of a complex belonging to the heterodimeric amino acid transporter family. LPI was initially described in Finland, but has worldwide distribution. Typically, symptoms begin after weaning with refusal of feeding, vomiting, and consequent failure to thrive. Hepatosplenomegaly, hematological anomalies, neurological involvement, including hyperammonemic coma are recurrent clinical features. Two major complications, pulmonary alveolar proteinosis and renal disease are increasingly observed in LPI patients. There is extreme variability in the clinical presentation even within individual families, frequently leading to misdiagnosis or delayed diagnosis. This condition is diagnosed by urine amino acids, showing markedly elevated excretion of lysine and other dibasic amino acids despite low plasma levels of lysine, ornithine, and arginine. The biochemical diagnosis can be uncertain, requiring confirmation by DNA testing. So far, approximately 50 different mutations have been identified in the SLC7A7 gene in a group of 142 patients from 110 independent families. No genotype-phenotype correlation could be established. Therapy requires a low protein diet, low-dose citrulline supplementation, nitrogen-scavenging compounds to prevent hyperammonemia, lysine, and carnitine supplements. Supportive therapy is available for most complications with bronchoalveolar lavage being necessary for alveolar proteinosis.


Assuntos
Rim/metabolismo , Lisina/urina , Aminoacidúrias Renais/genética , Aminoacidúrias Renais/metabolismo , Sistema y+L de Transporte de Aminoácidos , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos Básicos/genética , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Células Epiteliais/metabolismo , Finlândia , Cadeias Leves da Proteína-1 Reguladora de Fusão/genética , Cadeias Leves da Proteína-1 Reguladora de Fusão/metabolismo , Estudos de Associação Genética , Humanos , Mucosa Intestinal/metabolismo , Transportador 1 de Aminoácidos Neutros Grandes/genética , Mutação , Proteinose Alveolar Pulmonar/genética , Proteinose Alveolar Pulmonar/metabolismo , Aminoacidúrias Renais/diagnóstico , Aminoacidúrias Renais/dietoterapia
3.
Hum Mol Genet ; 17(10): 1427-35, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18245780

RESUMO

The parent of origin-dependent expression of the IGF2 and H19 genes is controlled by the imprinting centre 1 (IC1) consisting in a methylation-sensitive chromatin insulator. Deletions removing part of IC1 have been found in patients affected by the overgrowth- and tumour-associated Beckwith-Wiedemann syndrome (BWS). These mutations result in the hypermethylation of the remaining IC1 region, loss of IGF2/H19 imprinting and fully penetrant BWS phenotype when maternally transmitted. We now report that 12 additional cases with IC1 hypermethylation have a similar clinical phenotype but showed neither a detectable deletion nor other mutation in the local vicinity. Likewise, no IC1 deletion was detected in 40 sporadic non-syndromic Wilms' tumours. A detailed analysis of the BWS patients showed that the hypermethylation variably affected the IC1 region and was generally mosaic. We observed that all these cases were sporadic and in at least two families affected and unaffected members shared the same maternal IC1 allele but not the abnormal maternal chromosome epigenotype. Furthermore, the chromosome with the imprinting defect derived from either the maternal grandfather or maternal grandmother. Overall, these results indicate that methylation-imprinting defects at the IGF2-H19 locus can result from inherited mutations of the IC and have high recurrence risk or arise independently from the sequence context and generally not transmitted to the progeny. Despite these differences, the epigenetic abnormalities are usually present in the patients in the mosaic form and probably acquired by post-zygotic de novo methylation. Distinguishing between these two groups of cases is important for genetic counselling.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Impressão Genômica , Fator de Crescimento Insulin-Like II/genética , RNA não Traduzido/genética , Tumor de Wilms/genética , Alelos , Síndrome de Beckwith-Wiedemann/diagnóstico , Fator de Ligação a CCCTC , Segregação de Cromossomos , Cromossomos Humanos Par 11 , Metilação de DNA , Proteínas de Ligação a DNA/genética , Feminino , Deleção de Genes , Haplótipos , Humanos , Itália , Masculino , Mutação , Linhagem , RNA Longo não Codificante , Proteínas Repressoras/genética
5.
Hum Mutat ; 29(1): 14-21, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17764084

RESUMO

Lysinuric protein intolerance (LPI) is an inherited aminoaciduria caused by defective cationic amino acid (CAA) transport at the basolateral membrane of epithelial cells in the intestine and kidney. LPI is caused by mutations in the SLC7A7 gene, which encodes the y(+)LAT-1 protein, the catalytic light chain subunit of a complex belonging to the heterodimeric amino acid transporter family. Coexpression of 4F2hc (the heavy chain subunit) and y(+)LAT-1 induces y(+)L activity (CAA transport). So far a total of 43 different mutations of the SLC7A7 gene, nine of which newly reported here, have been identified in a group of 130 patients belonging to at least 98 independent families. The mutations are distributed along the entire gene and include all different types of mutations. Five polymorphisms within the SLC7A7 coding region and two variants found in the 5'UTR have been identified. A genuine founder effect mutation has been demonstrated only in Finland, where LPI patients share the same homozygous mutation, c.895-2A>T. LPI patients show extreme variability in clinical presentation, and no genotype-phenotype correlations have been defined. This phenotypic variability and the lack of a specific clinical presentation have caused various misdiagnoses. At the biochemical level, the elucidation of SLC7A7 function will be necessary to understand precise disease mechanisms and develop more specific and effective therapies. In this review, we summarize the current knowledge of SLC7A7 mutations and their role in LPI pathogenesis.


Assuntos
Cadeias Leves da Proteína-1 Reguladora de Fusão/genética , Lisina/urina , Mutação , Aminoacidúrias Renais/genética , Sequência de Aminoácidos , Sistema y+L de Transporte de Aminoácidos , Animais , Análise Mutacional de DNA , Diagnóstico Diferencial , Cadeias Leves da Proteína-1 Reguladora de Fusão/fisiologia , Genótipo , Humanos , Modelos Animais , Dados de Sequência Molecular , Fenótipo , Polimorfismo Genético , Aminoacidúrias Renais/metabolismo
6.
Am J Physiol Cell Physiol ; 293(1): C191-8, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17376816

RESUMO

The solute carrier family 7A member 7 gene (SLC7A7) encodes the light chain of the heterodimeric carrier responsible for cationic amino acid (CAA) transport across the basolateral membranes of epithelial cells in intestine and kidney. Mutations affecting SLC7A7 cause lysinuric protein intolerance (LPI), a multiorgan disorder with clinical symptoms that include visceromegaly, growth retardation, osteoporosis, hyperammonemia, and hyperdibasicaminoaciduria. Here, we describe the consequences of inactivating Slc7a7 in a mouse model of LPI. The Slc7a7 mutation was generated by high-throughput retroviral gene-trapping in embryonic stem cells. The Slc7a7(-/-) mouse displayed intrauterine growth restriction (IUGR), commonly leading to neonatal lethality. After heavy protein ingestion, the surviving adult animals presented metabolic derangement consistent with that observed in human LPI. IUGR was investigated by examining the expression of main factors controlling fetal growth. Insulin-like growth factor 1, the dominant fetal growth regulator in late gestation, was markedly downregulated as demonstrated by quantitative real-time RT-PCR, immunostaining and Western blot analysis in fetal liver. To further explore the pathophysiology of LPI, gene expression profiling analyses were carried out by DNA microarray technology in intestine and liver of adult Slc7a7(-/-) mice. Significant upregulation or downregulation (twofold or greater) was observed for 488 transcripts in intestine, and for 521 transcripts in the liver. The largest category of differentially expressed genes corresponds to those involved in transport according to Gene Ontology classification. This mouse model offers new insights into the pathophysiology of LPI and into mechanisms linking CAA metabolic pathways and growth control.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Sistema y+ de Transporte de Aminoácidos/metabolismo , Proteínas Alimentares/metabolismo , Retardo do Crescimento Fetal/metabolismo , Expressão Gênica , Fator de Crescimento Insulin-Like I/metabolismo , Lisina/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/complicações , Erros Inatos do Metabolismo dos Aminoácidos/genética , Erros Inatos do Metabolismo dos Aminoácidos/fisiopatologia , Sistema y+ de Transporte de Aminoácidos/deficiência , Sistema y+ de Transporte de Aminoácidos/genética , Sistema y+L de Transporte de Aminoácidos , Animais , Arginina/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Retardo do Crescimento Fetal/genética , Perfilação da Expressão Gênica/métodos , Genótipo , Fator de Crescimento Insulin-Like I/genética , Mucosa Intestinal/metabolismo , Intestinos/embriologia , Fígado/embriologia , Fígado/metabolismo , Redes e Vias Metabólicas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Reação em Cadeia da Polimerase , Ureia/metabolismo
7.
J Pediatr ; 150(3): 300-5, 305.e1, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17307551

RESUMO

OBJECTIVE: To investigate the hypothalamus-pituitary-thyroid axis in patients with glycogen storage disease type 1(GSD1). STUDY DESIGN: Ten patients with GSD1a, 7 patients with GSD1b, and 34 sex- and age-matched healthy control subjects were enrolled in the study. RESULTS: The levels of serum-free thyroxine (FT4) were significantly lower in patients with GSD1a and GSD1b (P < .05), whereas thyrotropin was significantly higher compared with control subjects only in patients with GSD1b (P < .005). Thyroglobulin and thyroperoxidase auto-antibodies were significantly higher in patients with GSD1b than in patients with GSD1a and control subjects (P < .005). After thyrotropin-releasing hormone stimulation, an enhanced thyrotropin response was found in patients with GSD1a and patients with GSD1b (P < .005) compared with control subjects. The presence of a subclinical or overt hypothyroidism was found in 4 of 7 patients with GSD1b and in no patient with GSD1a (chi2 = 7.47, P < .005) or control subject (chi2 = 27.2, P < .0001). CONCLUSIONS: Patients with GSD1b have an increased prevalence of thyroid autoimmunity and hypothyroidism, although patients with GSD1a have little evidence of thyroid abnormalities. Concomitant damage at the level of the hypothalamus or pituitary gland might be hypothesized on the basis of the slightly elevated thyrotropin levels, even in patients with overt hypothyroidism.


Assuntos
Doença de Depósito de Glicogênio Tipo I/epidemiologia , Hipotireoidismo/epidemiologia , Tireoidite Autoimune/epidemiologia , Adolescente , Adulto , Distribuição por Idade , Estudos de Casos e Controles , Criança , Comorbidade , Feminino , Doença de Depósito de Glicogênio Tipo I/diagnóstico , Humanos , Hipotireoidismo/diagnóstico , Masculino , Prevalência , Probabilidade , Prognóstico , Estudos Prospectivos , Valores de Referência , Medição de Risco , Distribuição por Sexo , Estatísticas não Paramétricas , Testes de Função Tireóidea , Tireoidite Autoimune/diagnóstico , Tireotropina/sangue , Tiroxina/sangue
8.
Hum Mutat ; 28(3): 255-64, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17072863

RESUMO

Homozygosity or compound heterozygosity for the c.833T>C transition (p.I278 T) in the cystathionine beta-synthase (CBS) gene represents the most common cause of pyridoxine-responsive homocystinuria in Western Eurasians. However, the frequency of the pathogenic c.833C allele, as observed in healthy newborns from several European countries (q(c.833C) approximately equals 3.3 x 10(-3)), is approximately 20-fold higher than expected on the basis of the observed number of symptomatic homocystinuria patients carrying this mutation (q(c.833C) approximately equals 0.18 x 10(-3)), implying clinical underascertainment. Intriguingly, the c.833C mutation is also present in combination with a 68-bp insertion, c.[833C; 844_845ins68], in a substantial proportion of chromosomes from nonhomocystinuric individuals worldwide. We have sought to study the relationship between the pathogenic and nonpathogenic c.833C-bearing chromosomes and to determine whether the pathogenic c.[833C; -] chromosomes are identical-by-descent or instead arose by recurrent mutation. Initial haplotype analysis of 780 randomly selected Czech and sub-Saharan African wild-type chromosomes, employing 12 intragenic markers, revealed 29 distinct CBS haplotypes, of which 10 carried the c.[833C; 844_845ins68] combination; none carried an isolated c.833C or c.844_845ins68 mutation. Subsequent examination of 69 pathogenic c.[833C; -] chromosomes, derived from homocystinuria patients of predominantly European origin, disclosed three unrelated haplotypes that differed from their wild-type counterparts by virtue of the presence of c.833C, thereby indicating that c.833T>C transition has occurred repeatedly and independently in the past. Since c.833T does not reside within an obvious mutational hotspot, we surmise that the three pathogenic and comparatively prevalent c.[833C; -] chromosomes may have originated by recurrent gene conversion employing the common nonpathogenic c.[833C; 844_845ins68] chromosomes as templates.


Assuntos
Cistationina beta-Sintase/genética , Conversão Gênica/fisiologia , Variação Genética , Haplótipos , Homocistinúria/genética , África , Sequência de Bases , Europa (Continente) , Frequência do Gene , Testes Genéticos , Humanos , Dados de Sequência Molecular
9.
Genet Med ; 8(7): 409-16, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16845273

RESUMO

PURPOSE: We present a case-control study of seven polymorphisms of six genes involved in homocysteine/folate pathway as risk factors for Down syndrome. Gene-gene/allele-allele interactions, haplotype analysis and the association with age at conception were also evaluated. METHODS: We investigated 94 Down syndrome-mothers and 264 control-women from Campania, Italy. RESULTS: Increased risk of Down syndrome was associated with the methylenetetrahydrofolate reductase (MTHFR) 1298C allele (OR 1.46; 95% CI 1.02-2.10), the MTHFR 1298CC genotype (OR 2.29; 95% CI 1.06-4.96), the reduced-folate-carrier1 (RFC1) 80G allele (1.48; 95% CI 1.05-2.10) and the RFC1 80 GG genotype (OR 2.05; 95% CI 1.03-4.07). Significant associations were found between maternal age at conception > or = 34 years and either the MTHFR 1298C or the RFC 180G alleles. Positive interactions were found for the following genotype-pairs: MTHFR 677TT and 1298CC/CA, 1298CC/CA and RFC1 80 GG/GA, RFC1 80 GG and methylenetetrahydrofolate-dehydrogenase 1958 AA. The 677-1298 T-C haplotype at the MTHFR locus was also a risk factor for Down syndrome (P = 0.0022). The methionine-synthase-reductase A66G, the methionine-synthase A2756G and the cystathionine-beta-synthase 844ins68 polymorphisms were not associated with increased risk of Down syndrome. CONCLUSION: These results point to a role of maternal polymorphisms of homocysteine/folate pathway as risk factors for Down syndrome.


Assuntos
Síndrome de Down/genética , Ácido Fólico/metabolismo , Homocisteína/metabolismo , Proteínas de Membrana Transportadoras/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Polimorfismo Genético , Adulto , Estudos de Casos e Controles , Aberrações Cromossômicas , Feminino , Frequência do Gene , Genótipo , Haplótipos , Humanos , Idade Materna , Modelos Biológicos , Fatores de Risco
10.
Eur J Pediatr ; 165(11): 763-6, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16775724

RESUMO

INTRODUCTION: Lysinuric protein intolerance (LPI; MIM 222700) is a rare, autosomal recessive metabolic disorder caused by mutations in the SLC7A7 gene, which encodes the light chain of the cationic amino acids (CAA) transporter y+. The clinical presentation of LPI includes gastrointestinal symptoms, failure to thrive, episodes of coma, hepatosplenomegaly and osteoporosis. However, other findings have also been reported, and these suggest a multisystem involvement. DISCUSSION: We report a girl with confirmed LPI who presented with severe short stature that was unresponsive to adequate LPI treatment. The girl was found to have a classic growth hormone deficiency (GHD) and responded well to growth hormone (GH) replacement therapy. CONCLUSION: While it is not known whether the mechanisms involved in the GHD of our patient are related to LPI, this case suggests that GH/IGF-I axis should be investigated in LPI children with persistent growth failure.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/complicações , Transtornos do Crescimento/etiologia , Hormônio do Crescimento Humano/deficiência , Lisina/urina , Pré-Escolar , Feminino , Transtornos do Crescimento/tratamento farmacológico , Hormônio do Crescimento Humano/uso terapêutico , Humanos
11.
Clin Dysmorphol ; 15(1): 13-18, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16317301

RESUMO

The '13q-' syndrome shows widely variable manifestations. Investigation of the involvement of different tissues has never been reported in patients with 13q- syndrome previously. We describe a patient with mosaicism for del(13q) and clinical features of 13q- syndrome. The mother of the patient was professionally exposed to aniline colorants and glue components during the whole pregnancy. The patient had dysmorphic features, skeletal anomalies and brain malformations with agenesis of the corpus callosum, vermian hypoplasia and IVth ventricular system abnormalities. Eye examination revealed chorioretinal coloboma and irregular dispersion of retinal pigment in the right eye. The karyotype analyses and the molecular studies performed on peripheral lymphocytes, oral swab and cells of urinary tract were normal whereas a deletion of the long arm of chromosome 13 (13q13.2) was found in skin fibroblasts and in hair cells. We hypothesized that the 13q deletion arose during the third week after conception possibly due to a teratogenic effect and that tissue of mesodermal and ectodermal origin are involved. We suggest analysing a fibroblast karyotype when a diagnosis of 13q- syndrome is suspected on clinical ground. The role of teratogens in causing this type of mosaic chromosome abnormality also warrants further investigation.


Assuntos
Anormalidades Múltiplas/genética , Deleção Cromossômica , Cromossomos Humanos Par 13/genética , Mosaicismo , Anormalidades Múltiplas/induzido quimicamente , Anormalidades Múltiplas/patologia , Compostos de Anilina/efeitos adversos , Carcinógenos , Coloboma/induzido quimicamente , Coloboma/genética , Coloboma/patologia , Feminino , Retardo do Crescimento Fetal/induzido quimicamente , Retardo do Crescimento Fetal/genética , Retardo do Crescimento Fetal/patologia , Fibroblastos/patologia , Seguimentos , Cabelo/patologia , Humanos , Lactente , Exposição Materna/efeitos adversos , Mosaicismo/induzido quimicamente , Exposição Ocupacional/efeitos adversos , Pele/patologia
12.
Eur J Hum Genet ; 13(9): 1025-32, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15999116

RESUMO

Beckwith-Wiedemann Syndrome (BWS) results from mutations or epigenetic events involving imprinted genes at 11p15.5. Most BWS cases are sporadic and uniparental disomy (UPD) or putative imprinting errors predominate in this group. Sporadic cases with putative imprinting defects may be subdivided into (a) those with loss of imprinting (LOI) of IGF2 and H19 hypermethylation and silencing due to a defect in a distal 11p15.5 imprinting control element (IC1) and (b) those with loss of methylation at KvDMR1, LOI of KCNQ1OT1 (LIT1) and variable LOI of IGF2 in whom there is a defect at a more proximal imprinting control element (IC2). We investigated genotype/epigenotype-phenotype correlations in 200 cases with a confirmed molecular genetic diagnosis of BWS (16 with CDKN1C mutations, 116 with imprinting centre 2 defects, 14 with imprinting centre 1 defects and 54 with UPD). Hemihypertrophy was strongly associated with UPD (P<0.0001) and exomphalos was associated with an IC2 defect or CDKN1C mutation but not UPD or IC1 defect (P<0.0001). When comparing birth weight centile, IC1 defect cases were significantly heavier than the patients with CDKN1C mutations or IC2 defect (P=0.018). The risk of neoplasia was significantly higher in UPD and IC1 defect cases than in IC2 defect and CDKN1C mutation cases. Kaplan-Meier analysis revealed a risk of neoplasia for all patients of 9% at age 5 years, but 24% in the UPD subgroup. The risk of Wilms' tumour in the IC2 defect subgroup appears to be minimal and intensive screening for Wilms' tumour appears not to be indicated. In UPD patients, UPD extending to WT1 was associated with renal neoplasia (P=0.054). These findings demonstrate that BWS represents a spectrum of disorders. Identification of the molecular subtype allows more accurate prognostic predictions and enhances the management and surveillance of BWS children such that screening for Wilms' tumour and hepatoblastoma can be focused on those at highest risk.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Impressão Genômica/genética , Alelos , Sequência de Bases , Criança , Feminino , Genótipo , Humanos , Neoplasias/genética , Fenótipo , Dissomia Uniparental , Tumor de Wilms/genética
14.
Hum Mutat ; 25(4): 410, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15776427

RESUMO

Lysinuric protein intolerance (LPI) is an inherited hyperdibasic aminoaciduria caused by defective cationic amino acid (CAA) transport at the basolateral membrane of epithelial cells in the intestine and kidney. LPI is relatively common in Finland and a few clusters of patients are known in Italy and Japan. The SLC7A7 gene, mutated in LPI patients, encodes the y+LAT-1 protein which is the light subunit of a heterodimeric CAA transporter. We performed the mutation analysis in seven probands from five unrelated LPI families and identified five novel SLC7A7 mutations (p.M50K, p.T188I, p.R333M, p.Y457X, and c.499+?_629-?). By expression studies in X. laevis oocytes or patient's renal tubular cells, the functional analysis of altogether eight SLC7A7 mutations is here reported. Noteworthy, the p.R333M mutation, caused by a G to T transversion of the last nucleotide at 3' end of exon 7, disrupts a functional splicing motif generating misspliced transcripts. Three of the novel mutations were found in patients originating from Greece and Pakistan thus increasing the list of ethnic backgrounds where LPI mutant alleles are present. This reinforces the view that the rarity of LPI outside Finland might be ascribed to misdiagnosis of this disease.


Assuntos
Transtornos Congênitos do Transporte de Aminoácidos/genética , Membrana Celular/metabolismo , Cadeias Leves da Proteína-1 Reguladora de Fusão/genética , Lisina/química , Alelos , Transtornos Congênitos do Transporte de Aminoácidos/patologia , Sistema y+L de Transporte de Aminoácidos , Animais , Análise Mutacional de DNA , Dimerização , Cães , Células Epiteliais/metabolismo , Éxons , Feminino , Humanos , Túbulos Renais/metabolismo , Masculino , Mutação , Oócitos/metabolismo , Estrutura Terciária de Proteína , Xenopus laevis
15.
Eur J Hum Genet ; 13(5): 628-34, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15756301

RESUMO

Lysinuric protein intolerance (LPI) is an inherited aminoaciduria caused by defective cationic amino acid (CAA) transport at the basolateral membrane of epithelial cells in the intestine and kidney. The SLC7A7 gene, mutated in LPI, encodes the y(+)LAT-1 protein, which is the light subunit of the heterodimeric CAA transporter in which 4F2hc is the heavy chain subunit. Co-expression of 4F2hc and y(+)LAT-1 induces the y(+)L activity. This activity is also exerted by another complex composed of 4F2hc and y(+)LAT-2, the latter encoded by the SLC7A6 gene and more ubiquitously expressed than SLC7A7. On the basis of both the pattern of expression and the transport activity, y(+)LAT-2 might compensate for CAA transport when y(+)LAT-1 is defective. By expression in Xenopus laevis oocytes and mammalian cells, we functionally analysed two SLC7A7 mutants, E36del and F152L, respectively, the former displaying a partial dominant-negative effect. The results of the present study provide further insight into the molecular pathogenesis of LPI: a putative multiheteromeric structure of both [4F2hc/y(+)LAT-1] and [4F2hc/y(+)LAT-2], and the interference between y(+)LAT-1 and y(+)LAT-2 proteins. This interference can explain why the compensatory mechanism, that is, an increased expression of SLC7A6 as seen in lymphoblasts from LPI patients, may not be sufficient to restore the y(+)L system activity.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/genética , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Cadeias Leves da Proteína-1 Reguladora de Fusão/genética , Lisina/urina , Sistema y+L de Transporte de Aminoácidos , Animais , Arginina/metabolismo , Linhagem Celular , Pré-Escolar , Cães , Cadeias Leves da Proteína-1 Reguladora de Fusão/metabolismo , Humanos , Masculino , Mutação , Oócitos/metabolismo , Xenopus laevis
16.
Clin Dysmorphol ; 14(1): 13-18, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15602087

RESUMO

We report a father and son affected by spondylo-epi-metaphyseal dysplasia with multiple dislocations (Hall type), also called leptodactylic form. This family contributes to the delineation of the clinical and radiological phenotype of this rare condition.


Assuntos
Osteocondrodisplasias/diagnóstico , Adulto , Estatura , Deformidades Congênitas do Pé , Deformidades Congênitas da Mão , Humanos , Lactente , Masculino
17.
J Pediatr ; 145(2): 268-72, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15289783

RESUMO

We present a case of recurrent pulmonary alveolar proteinosis after heart-lung transplantation in a child with lysinuric protein intolerance. The recurrence of the pulmonary disease provides further insight regarding the possible pathogenesis of pulmonary alveolar proteinosis and therapeutic options for this complication.


Assuntos
Transtornos Congênitos do Transporte de Aminoácidos/complicações , Transplante de Coração-Pulmão , Proteinose Alveolar Pulmonar/complicações , Proteinose Alveolar Pulmonar/cirurgia , Transtornos Congênitos do Transporte de Aminoácidos/urina , Evolução Fatal , Humanos , Lactente , Lisina/urina , Masculino , Recidiva
19.
Am J Med Genet A ; 118A(1): 68-70, 2003 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-12605445

RESUMO

Lowry-Wood syndrome (LWS) is a rare condition characterized by multiple epiphyseal dysplasia (MED), microcephaly, and congenital nystagmus. A variable degree of mental retardation can also be present. It is probably inherited as an autosomal recessive trait. We report a new case of MED and microcephaly, without other additional features, suggesting a mild form of LWS. Molecular analysis of the cartilage oligomeric matrix protein (COMP) gene was performed and failed to find mutations.


Assuntos
Anormalidades Múltiplas/fisiopatologia , Anormalidades Múltiplas/genética , Proteína de Matriz Oligomérica de Cartilagem , Pré-Escolar , Análise Mutacional de DNA , Proteínas da Matriz Extracelular/genética , Genes Recessivos , Glicoproteínas/genética , Humanos , Recém-Nascido , Proteínas Matrilinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA