Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Cancers (Basel) ; 13(20)2021 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-34680366

RESUMO

Survival rates of childhood cancer patients have improved over the past four decades, although cancer treatments increase the risk of developing chronic diseases typical of aging. Thus, we aimed to identify molecular/metabolic cellular alterations responsible for early aging in childhood cancer survivors (CCS). Biochemical, proteomic, and molecular biology analyses were conducted on mononuclear cells (MNCs) isolated from peripheral blood of 196 CCS, the results being compared with those obtained on MNCs of 154 healthy subjects. CCS-MNCs showed inefficient oxidative phosphorylation associated with low energy status, and increased lipid peroxidation and lactate fermentation compared with age-matched normal controls. According to a mathematical model based on biochemical parameters, CCS-MNCs showed significantly higher metabolic ages than their real ages. The dysfunctional metabolism of CCS-MNCs is associated with lower expression levels of genes and proteins involved in mitochondrial biogenesis and metabolism regulation, such as CLUH, PGC1-alpha, and SIRT6 in CCS, not observed in the age-matched healthy or elderly subjects. In conclusion, our study identified some biochemical and molecular alterations possibly contributing to the pathophysiology of aging and metabolic deficiencies in CCS. These results identify new targets for pharmacological interventions to restore mitochondrial function, slowing down the aging-associated pathologies in CCS.

2.
Sci Rep ; 9(1): 10347, 2019 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-31316102

RESUMO

Aging is a physiological process in which multifactorial processes determine a progressive decline. Several alterations contribute to the aging process, including telomere shortening, oxidative stress, deregulated autophagy and epigenetic modifications. In some cases, these alterations are so linked with the aging process that it is possible predict the age of a person on the basis of the modification of one specific pathway, as proposed by Horwath and his aging clock based on DNA methylation. Because the energy metabolism changes are involved in the aging process, in this work, we propose a new aging clock based on the modifications of glucose catabolism. The biochemical analyses were performed on mononuclear cells isolated from peripheral blood, obtained from a healthy population with an age between 5 and 106 years. In particular, we have evaluated the oxidative phosphorylation function and efficiency, the ATP/AMP ratio, the lactate dehydrogenase activity and the malondialdehyde content. Further, based on these biochemical markers, we developed a machine learning-based mathematical model able to predict the age of an individual with a mean absolute error of approximately 9.7 years. This mathematical model represents a new non-invasive tool to evaluate and define the age of individuals and could be used to evaluate the effects of drugs or other treatments on the early aging or the rejuvenation.


Assuntos
Envelhecimento/metabolismo , Glucose/metabolismo , Modelos Biológicos , Trifosfato de Adenosina/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/metabolismo , Criança , Pré-Escolar , Metabolismo Energético , Feminino , Humanos , Leucócitos Mononucleares/metabolismo , Aprendizado de Máquina , Masculino , Malondialdeído/metabolismo , Pessoa de Meia-Idade , Mitocôndrias/metabolismo , Fosforilação Oxidativa , Adulto Jovem
4.
PLoS One ; 10(9): e0138680, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26417990

RESUMO

BACKGROUND: Cord blood contains high number of hematopoietic cells that after birth disappear. In this paper we have studied the functional properties of the umbilical cord blood progenitor cells collected from term and preterm neonates to establish whether quantitative and/or qualitative differences exist between the two groups. METHODS AND RESULTS: Our results indicate that the percentage of total CD34+ cells was significantly higher in preterm infants compared to full term: 0.61% (range 0.15-4.8) vs 0.3% (0.032-2.23) p = 0.0001 and in neonates <32 weeks of gestational age (GA) compared to those ≥32 wks GA: 0.95% (range 0.18-4.8) and 0.36% (0.15-3.2) respectively p = 0.0025. The majority of CD34+ cells co-expressed CD71 antigen (p<0.05 preterm vs term) and grew in vitro large BFU-E, mostly in the second generation. The subpopulations CD34+CD38- and CD34+CD45- resulted more represented in preterm samples compared to term, conversely, Side Population (SP) did not show any difference between the two group. The absolute number of preterm colonies (CFCs/10microL) resulted higher compared to term (p = 0.004) and these progenitors were able to grow until the third generation maintaining an higher proportion of CD34+ cells (p = 0.0017). The number of colony also inversely correlated with the gestational age (Pearson r = -0.3001 p<0.0168). CONCLUSIONS: We found no differences in the isolation and expansion capacity of Endothelial Colony Forming Cells (ECFCs) from cord blood of term and preterm neonates: both groups grew in vitro large number of endothelial cells until the third generation and showed a transitional phenotype between mesenchymal stem cells and endothelial progenitors (CD73, CD31, CD34 and CD144)The presence, in the cord blood of preterm babies, of high number of immature hematopoietic progenitors and endothelial/mesenchymal stem cells with high proliferative potential makes this tissue an important source of cells for developing new cells therapies.


Assuntos
Aldeído Desidrogenase/metabolismo , Células Endoteliais/citologia , Células Precursoras Eritroides/citologia , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/citologia , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Contagem de Células Sanguíneas , Feminino , Humanos , Recém-Nascido , Recém-Nascido Prematuro/sangue , Gravidez , Nascimento Prematuro/sangue , Receptores da Transferrina/metabolismo , Nascimento a Termo/sangue
5.
Nephrol Dial Transplant ; 29 Suppl 4: iv80-6, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25165188

RESUMO

BACKGROUND: Mutations of INF2 represent the major cause of familial autosomal dominant (AD) focal segmental glomerulosclerosis (FSGS). A few patients present neurological symptoms of Charcot-Marie-Tooth (CMT) disease but the prevalence of the association has not been assessed yet. METHODS: We screened 28 families with AD FSGS and identified 8 INF2 mutations in 9 families (32 patients overall), 3 of which were new. Mutations were in all cases localized in the diaphanous-inhibitory domain (DID) of the protein. RESULTS: Clinical features associated with INF2 mutations in our patient cohort included mild proteinuria (1.55 g/L; range 1-2.5) and haematuria as a unique symptom that was recognized at a median age of 21.75 years (range 8-30). Eighteen patients developed end-stage renal disease during their third decade of life; 12 patients presented a creatinine range between 1.2 and 1.5 mg/dL and 2 were healthy at 45 and 54 years of age. CMT was diagnosed in four cases (12.5%); one of these patients presented an already known mutation on exon 2 of INF2, whereas the other patients presented the same mutation on exon 4, a region that was not previously associated with CMT. CONCLUSIONS: We confirmed the high incidence of INF2 mutations in families with AD FSGS. The clinical phenotype was mild at the onset of the disease, but evolution to ESRD was frequent. The incidence of CMT has, for the first time, been calculated here to be 12.5% of mutation carriers. Our findings support INF2 gene analysis in families in which renal failure and/or neuro-sensorial defects are inherited following an AD model.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Glomerulosclerose Segmentar e Focal/genética , Falência Renal Crônica/genética , Proteínas dos Microfilamentos/genética , Mutação/genética , Adolescente , Adulto , Sequência de Aminoácidos , Criança , Estudos de Coortes , Análise Mutacional de DNA , Primers do DNA/química , Primers do DNA/genética , Feminino , Forminas , Humanos , Itália , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Linhagem , Fenótipo , Reação em Cadeia da Polimerase , Homologia de Sequência de Aminoácidos , Adulto Jovem
6.
Biochem Med (Zagreb) ; 24(1): 151-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24627724

RESUMO

INTRODUCTION: Congenital analbuminemia is a rare autosomal recessive disorder manifested by the presence of a very low amount of circulating serum albumin. It is an allelic heterogeneous defect, caused by variety of mutations within the albumin gene in homozygous or compound heterozygous state. Herein we report the clinical and molecular characterization of a new case of congenital analbuminemia diagnosed in a female newborn of consanguineous (first degree cousins) parents from Ankara, Turkey, who presented with a low albumin concentration (< 8 g/L) and severe clinical symptoms. MATERIALS AND METHODS: The albumin gene of the index case was screened by single-strand conformation polymorphism, heteroduplex analysis, and direct DNA sequencing. The effect of the splicing mutation was evaluated by examining the cDNA obtained by reverse transcriptase - polymerase chain reaction (RT-PCR) from the albumin mRNA extracted from proband's leukocytes. RESULTS: DNA sequencing revealed that the proband is homozygous, and both parents are heterozygous, for a novel G>A transition at position c.1652+1, the first base of intron 12, which inactivates the strongly conserved GT dinucleotide at the 5' splice site consensus sequence of this intron. The splicing defect results in the complete skipping of the preceding exon (exon 12) and in a frame-shift within exon 13 with a premature stop codon after the translation of three mutant amino acid residues. CONCLUSIONS: Our results confirm the clinical diagnosis of congenital analbuminemia in the proband and the inheritance of the trait and contribute to shed light on the molecular genetics of analbuminemia.


Assuntos
Processamento Alternativo/genética , Hipoalbuminemia/congênito , Hipoalbuminemia/genética , Mutação/genética , Albumina Sérica/genética , Adolescente , Adulto , Sequência de Bases , Feminino , Análise Heteroduplex , Humanos , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Linhagem , Polimorfismo Conformacional de Fita Simples , Albumina Sérica/deficiência , Albumina Sérica Humana , Adulto Jovem
7.
N Engl J Med ; 369(7): 621-9, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23862974

RESUMO

BACKGROUND: Congenital abnormalities of the kidney and the urinary tract are the most common cause of pediatric kidney failure. These disorders are highly heterogeneous, and the etiologic factors are poorly understood. METHODS: We performed genomewide linkage analysis and whole-exome sequencing in a family with an autosomal dominant form of congenital abnormalities of the kidney or urinary tract (seven affected family members). We also performed a sequence analysis in 311 unrelated patients, as well as histologic and functional studies. RESULTS: Linkage analysis identified five regions of the genome that were shared among all affected family members. Exome sequencing identified a single, rare, deleterious variant within these linkage intervals, a heterozygous splice-site mutation in the dual serine-threonine and tyrosine protein kinase gene (DSTYK). This variant, which resulted in aberrant splicing of messenger RNA, was present in all affected family members. Additional, independent DSTYK mutations, including nonsense and splice-site mutations, were detected in 7 of 311 unrelated patients. DSTYK is highly expressed in the maturing epithelia of all major organs, localizing to cell membranes. Knockdown in zebrafish resulted in developmental defects in multiple organs, which suggested loss of fibroblast growth factor (FGF) signaling. Consistent with this finding is the observation that DSTYK colocalizes with FGF receptors in the ureteric bud and metanephric mesenchyme. DSTYK knockdown in human embryonic kidney cells inhibited FGF-stimulated phosphorylation of extracellular-signal-regulated kinase (ERK), the principal signal downstream of receptor tyrosine kinases. CONCLUSIONS: We detected independent DSTYK mutations in 2.3% of patients with congenital abnormalities of the kidney or urinary tract, a finding that suggests that DSTYK is a major determinant of human urinary tract development, downstream of FGF signaling. (Funded by the National Institutes of Health and others.).


Assuntos
Mutação , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Sistema Urinário/anormalidades , Anormalidades Urogenitais/genética , Adulto , Animais , Sequência de Bases , Criança , Exoma , Feminino , Técnicas de Silenciamento de Genes , Ligação Genética , Estudo de Associação Genômica Ampla , Heterozigoto , Humanos , Lactente , Rim/anormalidades , Masculino , Camundongos , Dados de Sequência Molecular , Linhagem , RNA Interferente Pequeno , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Sistema Urinário/crescimento & desenvolvimento , Sistema Urinário/metabolismo , Adulto Jovem
8.
Eur J Clin Invest ; 43(1): 72-8, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23176518

RESUMO

BACKGROUND: Analbuminemia (OMIM # 103600) is a rare autosomal recessive disorder manifested by the absence or severe reduction of circulating serum albumin in homozygous or compound heterozygous subjects. The trait is caused by a variety of mutations within the albumin gene. DESIGN: We report here the clinical and molecular characterisation of two new cases of congenital analbuminemia diagnosed in two members of the Druze population living in a Galilean village (Northern Israel) on the basis of their low level of circulating albumin. The albumin gene was screened by single-strand conformation polymorphism and heteroduplex analysis, and the mutated region was submitted to DNA sequencing. RESULTS: Both the analbuminemic subjects resulted homozygous for a previously unreported c.1 A>C transversion, for which we suggest the name Afula from the hospital where the two cases were investigated. This mutation causes the loss of the primary start codon ATG for Met1, which is replaced by a - then untranslated - triplet CTG for Leu. (p.Met1Leu). The use of an alternative downstream ATG codon would probably give rise to a completely aberrant polypeptide chain, leading to a misrouted intracellular transport and a premature degradation. CONCLUSIONS: The discovery of this new ALB mutation, probably inherited from a common ancestor, sheds light on the molecular mechanism underlying the analbuminemic trait and may serve in the development of a rapid genetic test for the identification of a-symptomatic heterozygous carriers in the Druze population in the Galilee.


Assuntos
Transtornos Cromossômicos/etnologia , Mutação , Albumina Sérica/genética , Criança , Análise Mutacional de DNA , Análise Heteroduplex , Humanos , Israel , Masculino , Pessoa de Meia-Idade , Polimorfismo Conformacional de Fita Simples , Albumina Sérica/deficiência
11.
Mol Genet Metab ; 105(3): 479-83, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22227324

RESUMO

Analbuminemia is a rare autosomal recessive disorder manifested by the absence or severe reduction of circulating serum albumin in homozygous or compound heterozygous subjects. It is an allelic heterogeneous defect, caused by a variety of mutations within the albumin gene. The analbuminemic condition was suspected in a Portuguese boy who presented with low albumin level (about 3.8 g/L) and a significant hypercholesterolemia, but with no clinical findings. The albumin gene was screened by single strand conformational polymorphism and heteroduplex analysis and submitted to direct DNA sequencing. The proband was found to be homozygous for a previously unreported G>A change at position c.1289+1, the first base of intron 10, which inactivates the strongly conserved GT dinucleotide at the 5' splice site consensus sequence of the intron. The effect of this mutation was evaluated by examining the cDNA obtained by RT-PCR from the albumin mRNA extracted from proband's leukocytes. The splicing defect results in the skipping of the preceding exon. The subsequent reading frame-shift in exon 11 produces a premature stop codon located 33 codons downstream the 5' end of the exon. This extensive cDNA alteration is responsible for the analbuminemic trait. Both parents were found to be heterozygous for the same mutation. DNA and cDNA sequence analysis established the diagnosis of congenital analbuminemia in the proband. The effects of the so far identified splice-site mutations in the albumin gene are discussed.


Assuntos
Hipoalbuminemia/genética , Splicing de RNA , Albumina Sérica/deficiência , Albumina Sérica/genética , Adolescente , Sequência de Bases , Mutação da Fase de Leitura , Predisposição Genética para Doença , Humanos , Masculino , Polimorfismo de Nucleotídeo Único , Portugal , Análise de Sequência de DNA , Albumina Sérica Humana
12.
Int J Mol Sci ; 12(11): 7314-22, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22174600

RESUMO

Analbuminemia is a rare autosomal recessive disorder manifested by the absence, or severe reduction, of circulating serum albumin (ALB). We report here a new case diagnosed in a 45 years old man of Southwestern Asian origin, living in Switzerland, on the basis of his low ALB concentration (0.9 g/L) in the absence of renal or gastrointestinal protein loss, or liver dysfunction. The clinical diagnosis was confirmed by a mutational analysis of the albumin (ALB) gene, carried out by single-strand conformational polymorphism (SSCP), heteroduplex analysis (HA), and DNA sequencing. This screening of the ALB gene revealed that the proband is homozygous for two mutations: the insertion of a T in a stretch of eight Ts spanning positions c.1289 + 23-c.1289 + 30 of intron 10 and a c.802 G > T transversion in exon 7. Whereas the presence of an additional T in the poly-T tract has no direct deleterious effect, the latter nonsense mutation changes the codon GAA for Glu244 to the stop codon TAA, resulting in a premature termination of the polypeptide chain. The putative protein product would have a length of only 243 amino acid residues instead of the normal 585 found in the mature serum albumin, but no evidence for the presence in serum of such a truncated polypeptide chain could be obtained by two dimensional electrophoresis and western blotting analysis.


Assuntos
Códon sem Sentido , Patologia Molecular/métodos , Albumina Sérica/deficiência , Albumina Sérica/genética , Povo Asiático/genética , Éxons , Análise Heteroduplex , Humanos , Masculino , Pessoa de Meia-Idade , Polimorfismo Conformacional de Fita Simples , Análise de Sequência de DNA , Suíça
13.
Kidney Int ; 80(4): 389-96, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21697813

RESUMO

To identify gene loci associated with steroid-resistant nephrotic syndrome (SRNS), we utilized homozygosity mapping and exome sequencing in a consanguineous pedigree with three affected siblings. High-density genotyping identified three segments of homozygosity spanning 33.6 Mb on chromosomes 5, 10, and 15 containing 296 candidate genes. Exome sequencing identified two homozygous missense variants within the chromosome 15 segment; an A159P substitution in myosin 1E (MYO1E), encoding a podocyte cytoskeletal protein; and an E181K substitution in nei endonuclease VIII-like 1 (NEIL1), encoding a base-excision DNA repair enzyme. Both variants disrupt highly conserved protein sequences and were absent in public databases, 247 healthy controls, and 286 patients with nephrotic syndrome. The MYO1E A159P variant is noteworthy, as it is expected to impair ligand binding and actin interaction in the MYO1E motor domain. The predicted loss of function is consistent with the previous demonstration that Myo1e inactivation produces nephrotic syndrome in mice. Screening 71 additional patients with SRNS, however, did not identify independent NEIL1 or MYO1E mutations, suggesting larger sequencing efforts are needed to uncover which mutation is responsible for the phenotype. Our findings demonstrate the utility of exome sequencing for rapidly identifying candidate genes for human SRNS.


Assuntos
DNA Glicosilases/genética , Análise Mutacional de DNA , Exoma , Miosina Tipo I/genética , Síndrome Nefrótica/congênito , Estudos de Casos e Controles , Cromossomos Humanos Par 15 , DNA Glicosilases/química , Frequência do Gene , Estudos de Associação Genética , Predisposição Genética para Doença , Homozigoto , Humanos , Itália , Modelos Moleculares , Mutação de Sentido Incorreto , Miosina Tipo I/química , Síndrome Nefrótica/genética , Cidade de Nova Iorque , Linhagem , Fenótipo , Conformação Proteica , Relação Estrutura-Atividade
14.
Hum Mutat ; 31(12): 1352-9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20960469

RESUMO

Congenital anomalies of the kidney and the urinary tract (CAKUT) represent a major source of morbidity and mortality in children. Several factors (PAX, SOX,WNT, RET, GDFN, and others) play critical roles during the differentiation process that leads to the formation of nephron epithelia. We have identified mutations in SOX17, an HMG-box transcription factor and Wnt signaling antagonist, in eight patients with CAKUT (seven vesico-ureteric reflux, one pelvic obstruction). One mutation, c.775T>A (p.Y259N), recurred in six patients. Four cases derived from two small families; renal scars with urinary infection represented the main symptom at presentation in all but two patients. Transfection studies indicated a 5-10-fold increase in the levels of the mutant protein relative to wild-type SOX17 in transfected kidney cells. Moreover we observed a corresponding increase in the ability of SOX17 p.Y259N to inhibit Wnt/ß-catenin transcriptional activity, which is known to regulate multiple stages of kidney and urinary tract development. In conclusion, SOX17 p.Y259N mutation is recurrent in patients with CAKUT. Our data shows that this mutation correlates with an inappropriate accumulation of SOX17-p.Y259N protein and inhibition of the ß-catenin/Wnt signaling pathway. These data indicate a role of SOX17 in human kidney and urinary tract development and implicate the SOX17-p.Y259N mutation as a causative factor in CAKUT.


Assuntos
Anormalidades Congênitas/genética , Rim/anormalidades , Mutação/genética , Fatores de Transcrição SOXF/genética , Sistema Urinário/anormalidades , Pré-Escolar , Hibridização Genômica Comparativa , Família , Feminino , Regulação da Expressão Gênica , Células HEK293 , Humanos , Hibridização in Situ Fluorescente , Masculino , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Linhagem , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição SOXF/metabolismo
15.
Clin Chim Acta ; 411(21-22): 1711-5, 2010 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-20638375

RESUMO

BACKGROUND: Analbuminemia is a rare autosomal recessive disorder manifested by the absence, or severe reduction, of circulating serum albumin. The analbuminemic trait was diagnosed in a young Turkish woman on the basis of her clinical symptoms (bilateral lower limb edema) and biochemical findings (minimal albumin amount and variable increases in other protein fractions). METHODS: Total DNA from the analbuminemic proband and her parents was PCR-amplified using oligonucleotide primers designed to amplify the 14 exons of the albumin gene (ALB) and the flanking intron regions. The products were screened for mutations by single-strand conformation polymorphism (SSCP) and heteroduplex analyses (HA). RESULTS: HA allowed the identification of the mutation site in exon 12. Direct DNA sequencing of this abnormal fragment revealed that the analbuminemic trait was caused by a homozygous CA deletion at nucleotide positions c. 1614-1615 in the codons for Cys538 and Thr539. The subsequent frameshift should give rise to a putative truncated albumin variant in which the sequence Cys(538)-Thr-Leu-Ser has been changed to Cys(538)-Thr-Phe-Stop. The parents were heterozygous for the same mutation. CONCLUSIONS: Gel-based mutation detection and DNA sequencing substantiate the clinical diagnosis of congenital analbuminemia in our patient and show that the condition is caused by a novel mutation within the ALB gene. These results contribute to shed light on the molecular basis of this rare condition.


Assuntos
Mutação da Fase de Leitura , Albumina Sérica/deficiência , Albumina Sérica/genética , Códon sem Sentido , Análise Mutacional de DNA , Feminino , Homozigoto , Humanos , Deleção de Sequência , Turquia , Adulto Jovem
16.
Clin J Am Soc Nephrol ; 5(4): 698-702, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20150449

RESUMO

BACKGROUND AND OBJECTIVES: Wilms tumor-suppressor gene-1 (WT1) plays a key role in kidney development and function. WT1 mutations usually occur in exons 8 and 9 and are associated with Denys-Drash, or in intron 9 and are associated with Frasier syndrome. However, overlapping clinical and molecular features have been reported. Few familial cases have been described, with intrafamilial variability. Sporadic cases of WT1 mutations in isolated diffuse mesangial sclerosis or focal segmental glomerulosclerosis have also been reported. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: Molecular analysis of WT1 exons 8 and 9 was carried out in five members on three generations of a family with late-onset isolated proteinuria. The effect of the detected amino acid substitution on WT1 protein's structure was studied by bioinformatics tools. RESULTS: Three family members reached end-stage renal disease in full adulthood. None had genital abnormalities or Wilms tumor. Histologic analysis in two subjects revealed focal segmental glomerulosclerosis. The novel sequence variant c.1208G>A in WT1 exon 9 was identified in all of the affected members of the family. CONCLUSIONS: The lack of Wilms tumor or other related phenotypes suggests the expansion of WT1 gene analysis in patients with focal segmental glomerulosclerosis, regardless of age or presence of typical Denys-Drash or Frasier syndrome clinical features. Structural analysis of the mutated protein revealed that the mutation hampers zinc finger-DNA interactions, impairing target gene transcription. This finding opens up new issues about WT1 function in the maintenance of the complex gene network that regulates normal podocyte function.


Assuntos
Genes do Tumor de Wilms , Glomerulosclerose Segmentar e Focal/genética , Falência Renal Crônica/genética , Mutação , Proteínas WT1/genética , Adolescente , Adulto , Idade de Início , Idoso , Substituição de Aminoácidos , Biópsia , Biologia Computacional , Análise Mutacional de DNA , Progressão da Doença , Éxons , Predisposição Genética para Doença , Glomerulosclerose Segmentar e Focal/metabolismo , Glomerulosclerose Segmentar e Focal/patologia , Humanos , Itália , Falência Renal Crônica/metabolismo , Falência Renal Crônica/patologia , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Linhagem , Fenótipo , Podócitos/metabolismo , Podócitos/patologia , Conformação Proteica , Proteinúria/genética , Fatores de Risco , Relação Estrutura-Atividade , Proteínas WT1/química , Proteínas WT1/metabolismo
19.
Clin J Am Soc Nephrol ; 4(6): 1065-72, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19406966

RESUMO

BACKGROUND AND OBJECTIVES: Mutations in nephrin (NPHS1) and podocin (NPHS2) genes represent a major cause of idiopathic nephrotic syndrome (NS) in children. It is not yet clear whether the presence of a single mutation acts as a modifier of the clinical course of NS. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: We reviewed the clinical features of 40 patients with NS associated with heterozygous mutations or variants in NPHS1 (n = 7) or NPHS2 (n = 33). Long-term renal survival probabilities were compared with those of a concurrent cohort with idiopathic NS. RESULTS: Patients with a single mutation in NPHS1 received a diagnosis before those with potentially nongenetic NS and had a good response to therapies. Renal function was normal in all cases. For NPHS2, six patients had single heterozygous mutations, six had a p.P20L variant, and 21 had a p.R229Q variant. Age at diagnosis and the response to drugs were comparable in all NS subgroups. Overall, they had similar renal survival probabilities as non-NPHS1/NPHS2 cases (log-rank chi(2) 0.84, P = 0.656) that decreased in presence of resistance to therapy (P < 0.001) and in cases with renal lesions of glomerulosclerosis and IgM deposition (P < 0.001). Cox regression confirmed that the only significant predictor of dialysis was resistance to therapy. CONCLUSIONS: Our data indicate that single mutation or variant in NPHS1 and NPHS2 does not modify the outcome of primary NS. These patients should be treated following consolidated schemes and have good chances for a good long-term outcome.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Síndrome Nefrótica/genética , Síndrome Nefrótica/mortalidade , Adolescente , Criança , Pré-Escolar , Feminino , Seguimentos , Variação Genética , Heterozigoto , Humanos , Lactente , Estimativa de Kaplan-Meier , Masculino , Mutação Puntual
20.
Clin Chim Acta ; 396(1-2): 89-92, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18602380

RESUMO

BACKGROUND: Analbuminemia is a rare autosomal recessive disorder manifested by the absence, or severe reduction, of circulating serum albumin. Here we report the first case of hereditary analbuminemia in the ethnic Swedish population, and we define the molecular defect that causes the analbuminemic trait. METHODS: Total DNA, extracted from peripheral blood samples from the analbuminemic proband and his parents, was PCR-amplified using oligonucleotide primers designed to amplify the 14 exons, the exon-intron splice junctions, and the 5' and 3' untranslated regions of the albumin gene. The products were screened for mutations by single-strand conformation polymorphism and heteroduplex analyses. The latter allowed the identification of the abnormal fragment, which was then sequenced. RESULTS: The analbuminemic trait of the proband was caused by a homozygous AT deletion at nucleotides c. 228-229, the 91st and 92nd bases of exon 3. This defect, previously identified as Kayseri mutation [M. Galliano, M. Campagnoli, A. Rossi, et al. Molecular diagnosis of analbuminemia: a novel mutation identified in two Amerindian and two Turkish families. Clin Chem 2002;48: 844-849.], produces a frameshift leading to a premature stop, two codons downstream. CONCLUSIONS: The Kayseri mutation appears to be the most common cause of analbuminemia in humans, and is found in individuals belonging to geographically distant, and apparently unrelated ethnic groups.


Assuntos
Hipoalbuminemia/sangue , Hipoalbuminemia/genética , Mutação/genética , Adulto , Sequência de Bases , Eletroforese em Gel Bidimensional , Éxons/genética , Genoma Humano/genética , Humanos , Masculino , Suécia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...