Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Med Genet ; 2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38719348

RESUMO

BACKGROUND: Exploring the expression of X linked disorders like haemophilia A (HA) in females involves understanding the balance achieved through X chromosome inactivation (XCI). Skewed XCI (SXCI) may be involved in symptomatic HA carriers. We aimed to develop an approach for dissecting the specific cause of SXCI and verify its value in HA. METHODS: A family involving three females (two symptomatic with severe/moderate HA: I.2, the mother, and II.1, the daughter; one asymptomatic: II.2) and two related affected males (I.1, the father and I.3, the maternal uncle) was studied. The genetic analysis included F8 mutational screening, multiplex ligation-dependent probe amplification, SNP microarray, whole exome sequencing (WES) and Sanger sequencing. XCI patterns were assessed in ectoderm/endoderm and mesoderm-derived tissues using AR-based and RP2-based systems. RESULTS: The comprehensive family analysis identifies I.2 female patient as a heterozygous carrier of F8:p.(Ser1414Ter) excluding copy number variations. A consistent XCI pattern of 99.5% across various tissues was observed. A comprehensive filtering algorithm for WES data was designed, developed and applied to I.2. A Gly58Arg missense variant in VMA21 was revealed as the cause for SXCI.Each step of the variant filtering system takes advantage of publicly available genomic databases, non-SXCI controls and case-specific molecular data, and aligns with established concepts in the theoretical background of SXCI. CONCLUSION: This study acts as a proof of concept for our genomic filtering algorithm's clinical utility in analysing X linked disorders. Our findings clarify the molecular aspects of SXCI and improve genetic diagnostics and counselling for families with X linked diseases like HA.

2.
Front Pediatr ; 11: 1210158, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37425258

RESUMO

Introduction: Hemolytic uremic syndrome (HUS) is a condition that results in acute kidney failure mainly in children, which is caused by Shiga toxin-producing Escherichia coli and inflammatory response. Although anti-inflammatory mechanisms are triggered, studies on the implication in HUS are scarce. Interleukin-10 (IL-10) regulates inflammation in vivo, and the interindividual differences in its expression are related to genetic variants. Notably, the single nucleotide polymorphism (SNP) rs1800896 -1082 (A/G), located in the IL-10 promoter, regulates cytokine expression. Methods: Plasma and peripheral blood mononuclear cells (PBMC) were collected from healthy children and HUS patients exhibiting hemolytic anemia, thrombocytopenia, and kidney damage. Monocytes identified as CD14+ cells were analyzed within PBMC by flow cytometry. IL-10 levels were quantified by ELISA, and SNP -1082 (A/G) was analyzed by allele-specific PCR. Results: Circulating IL-10 levels were increased in HUS patients, but PBMC from these patients exhibited a lower capacity to secrete this cytokine compared with those from healthy children. Interestingly, there was a negative association between the circulating levels of IL-10 and inflammatory cytokine IL-8. We observed that circulating IL-10 levels were threefold higher in HUS patients with -1082G allele in comparison to AA genotype. Moreover, there was relative enrichment of GG/AG genotypes in HUS patients with severe kidney failure. Discussion: Our results suggest a possible contribution of SNP -1082 (A/G) to the severity of kidney failure in HUS patients that should be further evaluated in a larger cohort.

3.
Haemophilia ; 29(3): 844-854, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36930806

RESUMO

INTRODUCTION: Haemophilia B (HB) is associated with pathogenic variants in F9. Hemizygous deletions encompassing the entire F9 and proximate genes may express extra-haematological clinical phenotypes. AIM: To analyse the genotype/phenotype correlations in two unrelated boys with severe early childhood obesity (SCO), global developmental delay (GDD) and similar bleeding phenotype associated with comparable Xq27 deletions spanning the entire F9 and proximate genes, and characterise the pathogenic events estimating the most likely mutational mechanism involved. METHODS: Entire F9-deletions were detected in three hemizygous unrelated probands with HB: two cases, C#1/C#2, presented SCO and GDD and a control patient (Co), who only had severe bleeding symptoms. Dense SNP-array and case-specific STS walking scan allowed characterisation of the deletion breakpoints. Extensive use of bioinformatics, statistics and clinical databases allowed the investigation of genotype-phenotype associations. RESULTS: Patients C#1/C#2 and Co resulted in a complete F9 and additional gene deletions of variable extensions on Xq26.3-Xq27.2 (C#1/C#2/Co: 4.3Mb/3.9Mb/160Kb). C#1/C#2 common deleted gene SOX3 is directly associated with SCO, GDD and pituitary hypothyroidism (PH) whilst C#2 extra-deleted gene MAGEC2 indirectly relates to anal atresia (AA). Breakpoint analysis revealed the involvement of the mechanisms of Alu/Alu recombination for the first time in HB and non-homologous or alternative end-joining. CONCLUSION: Our results represent the first report of unrelated patients with HB, SCO and GDD. This study and the literature update expand the spectrum of clinical findings and molecular insights observed in patients with HB caused by complete F9 and nearby SOX3 and MAGEC2 gene deletions, which may configure a contiguous gene syndrome.


Assuntos
Hemofilia B , Obesidade Infantil , Humanos , Hemofilia B/genética , Mutação , Fenótipo , Biologia Computacional
4.
Neuromuscul Disord ; 31(3): 253-263, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33451931

RESUMO

This work describes a family with Duchenne muscular dystrophy (DMD) with a rare case of a symptomatic pregnant woman. The main aim was to perform prenatal molecular diagnosis to provide genetic counseling. The secondary aim was to suggest the molecular mechanisms causing the complex structural variant (cxSV) identified. To accomplish this, we used a multi-technique algorithm including segregation analysis, Multiplex Ligation-dependent Probe Amplification, PCR, X-chromosome inactivation studies, microarrays, whole genome sequencing and bioinformatics. We identified a duplication of exons 38-43 in the DMD gene in all affected and obligate carrier members, proving that this was the DMD-causing mutation. We also observed a skewed X-chromosome inactivation in the symptomatic woman that explained her symptomatology. In addition, we identified a cxSV (duplication of exons 38-43 and deletion of exons 45-54) in the affected boy. The molecular characterization and bioinformatic analyses of the breakpoint junctions allowed us to identify Double Strand Breaks stimulator motifs and suggested the replication-dependent Fork Stalling and Template Switching as the most probable mechanisms leading to the duplication. In addition, the de novo deletion might have been the result of a germline inter-chromosome non-allelic recombination involving the Non-Homologous End Joining mechanism. In conclusion, the diagnostic strategy used allowed us to provide accurate molecular diagnosis and genetic counseling. In addition, the familial molecular diagnosis together with the in-depth characterization of the cxSV helped to determine the chronology of the molecular events, and propose and understand the molecular mechanisms involved in the generation of this complex rearrangement.


Assuntos
Distrofia Muscular de Duchenne/genética , Diagnóstico Pré-Natal/métodos , Adolescente , Adulto , Distrofina/genética , Éxons , Feminino , Deleção de Genes , Aconselhamento Genético , Humanos , Masculino , Reação em Cadeia da Polimerase Multiplex , Mutação , Gravidez
6.
Hum Mutat ; 41(4): 825-836, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31898853

RESUMO

Hemophilia A (HA) provides excellent models to analyze genotype-phenotype relationships and mutational mechanisms. NhF8ld's breakpoints were characterized using case-specific DNA-tags, direct- or inverse-polymerase chain reaction amplification, and Sanger sequencing. DNA-break's stimulators (n = 46), interspersed repeats, non-B-DNA, and secondary structures were analyzed around breakpoints versus null hypotheses (E-values) based on computer simulations and base-frequency probabilities. Nine of 18 (50%) severe-HA patients with nhF8lds developed inhibitors, 1/8 affecting one exon and 8/10 (80%) affecting multi-exons. NhF8lds range: 2-165 kb. Five (45%) nhF8lds involve F8-extragenic regions including three affecting vicinal genes (SMIM9 and BRCC3) but none shows an extra-phenotype not related to severe-HA. The contingency analysis of recombinogenic motifs at nhF8ld breakpoints indicated a significant involvement of several DNA-break stimulator elements. Most nhF8ld's breakpoint junctions showed microhomologies (1-7 bp). Three (27%) nhF8lds show complexities at the breakpoints: an 8-bp inverted-insertion, and the remnant two, inverted- and direct-insertions (46-68 bp) supporting replicative models microhomology-mediated break-induced replication/Fork Stalling and Template Switching. The remnant eight (73%) nhF8lds may support nonhomologous end joining/microhomology-mediated end joining models. Our study suggests the involvement of the retroposition machinery (e.g., Jurka-targets, Alu-elements, long interspersed nuclear elements, long terminal repeats), microhomologies, and secondary structures at breakpoints playing significant roles in the origin of the upmost severe phenotype in HA.


Assuntos
Fator VIII/genética , Variação Genética , Hemofilia A/genética , Pontos de Quebra do Cromossomo , Biologia Computacional/métodos , Estudos de Associação Genética , Loci Gênicos , Predisposição Genética para Doença , Hemofilia A/diagnóstico , Humanos , Masculino , Mutação , Conformação de Ácido Nucleico , Motivos de Nucleotídeos , Fenótipo , Recombinação Genética , Índice de Gravidade de Doença
7.
Eur J Hum Genet ; 27(4): 603-611, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30626931

RESUMO

Among other applications of long-distance haplotype phasing in clinical genetics, determination of linked DNA markers as surrogate for problematic structural variants (e.g., repeat-mediated rearrangements) is essential to perform diagnosis from low-quality DNA samples. We describe a next-of-kin-independent (physical) phasing approach based on inverse-PCR (iPCR) paired-end amplification (PI). This method enables typing the multialleles of the short tandem repeat (STR) F8Int21[CA]n at the F8-intron 21, as a surrogate DNA marker for the F8-intron 22 inversion (Inv22), the hemophilia A-causative hotspot, within the transmitted haplotype in informative carriers. We provide proof-of-concept by blindly validating the PI approach in 15 carrier mother/affected-son duos. Every F8Int21[CA]n STR allele determined in phase with the Inv22 allele in the female carriers from the informative duos was confirmed in the hemizygous proband (P = 0.00003). A second surrogate STR locus at the F8-IVS22 was obtained by the PI approach improving severe-HA preimplantation genetic diagnosis by augmenting heterozygosity in Inv22 carriers bypassing the requirement for family linkage analysis. The ability of the PI-assay to combine other marker pairs was demonstrated by haplotyping a SNV (F8:c.6118T > C) with a >28kb-distant F8-IVS22 STR. The PI approach has proven flexibility to target different marker pairs and has potential for multiplex characterization of iPCR products by massively parallel sequencing.


Assuntos
Hemofilia A/genética , Repetições de Microssatélites/genética , Reação em Cadeia da Polimerase/métodos , Diagnóstico Pré-Implantação , Alelos , Feminino , Marcadores Genéticos/genética , Genótipo , Haplótipos/genética , Hemofilia A/diagnóstico , Hemofilia A/fisiopatologia , Humanos , Íntrons , Masculino , Gravidez
9.
Mol Cell Endocrinol ; 381(1-2): 220-9, 2013 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-23933148

RESUMO

The objective of this study was to perform genetic analysis in three brothers of Turkish origin born from consanguineus parents and affected by congenital hypothyroidism, goiter and low levels of serum TG. The combination of sequencing of DNA, PCR mapping, quantitative real-time PCR, inverse-PCR (I-PCR), multiplex PCR and bioinformatics analysis were used in order to detect TG mutations. We demonstrated that the three affected siblings are homozygous for a DNA inversion of 16,962bp in the TG gene associated with two deleted regions at both sides of the inversion limits. The inversion region includes the first 9bp of exon 48, 1015bp of intron 47, 191bp of exon 47, 1523bp of intron 46, 135bp of exon 46 and the last 14,089bp of intron 45. The proximal deletion corresponds to 27bp of TG intron 45, while the distal deletion spans the last 230bp of TG exon 48 and the first 588bp of intergenic region downstream TG end. The parents were heterozygous carriers of the complex rearrangement. In conclusion, a novel large imperfect DNA inversion within the TG gene was identified by the strategy of I-PCR. This aberration was not detectable by normal sequencing of the exons and exon/intron boundaries. Remarkably, the finding represents the first description of a TG deficiency disease caused by a DNA inversion.


Assuntos
Hipotireoidismo Congênito/genética , Tireoglobulina/genética , Sequência de Bases , Consanguinidade , Análise Mutacional de DNA , Estudos de Associação Genética , Humanos , Íntrons , Masculino , Dados de Sequência Molecular , Linhagem , Polimorfismo de Nucleotídeo Único , Deleção de Sequência , Inversão de Sequência , Tireoglobulina/deficiência
10.
Thromb Haemost ; 109(1): 24-33, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23093250

RESUMO

In haemophilia B (HB) (factor IX [FIX] deficiency), F9 genotype largely determines clinical phenotype. Aimed to characterise Argentinian families with HB, this study presents F9 genotype frequencies and their specific FIX inhibitor risk and 10 novel F9 mutations. Ninety-one DNA samples from HB patients and relatives were subjected to a new scheme: a primary screen for large deletions, a secondary screen for point mutations using conformation sensitive gel electrophoresis, DNA-sequencing and bioinformatic analysis. Our unbiased HB population (N=52) (77% with severe, 11.5% moderate and 11.5% mild HB) showed 32 missense (61.5%), including three novel mutations predicting specific structural/functional defects in silico , seven nonsense (13.5%) (one novel), five large deletions, four splice including three novel mutations affecting predicted splicing scores, three indels (two novel) and one Leiden mutation. Our comprehensive HB population included five patients with long-lasting FIX inhibitors: three nonsense (p.E35* (novel), p.R75*, p.W240*) and two entire- F9 deletions. Another patient with an indel (p.A26Rfs*14) developed transient inhibitors. A case-control analysis, based on our global prevalence of 3.05% for developing inhibitors in HB revealed that missense mutations were associated with a low risk odds ratio (OR) of 0.05 and a prevalence of 0.39%, whereas nonsense and entire- F9 deletions had significantly higher risks (OR 11.0 and 32.7) and prevalence (14.3% and 44.5%, respectively). Our cost-effective practical approach enabled identification of the causative mutation in all 55 Argentine families with HB, analysis of the molecular pathology of novel F9 defects and determination of mutation-associated FIX inhibitor risks.


Assuntos
Fator IX/genética , Hemofilia B/genética , Hemostasia/genética , Mutação , Argentina/epidemiologia , Autoanticorpos/sangue , Biomarcadores/sangue , Estudos de Casos e Controles , Códon sem Sentido , Biologia Computacional , Análise Mutacional de DNA/métodos , Fator IX/química , Fator IX/imunologia , Feminino , Frequência do Gene , Predisposição Genética para Doença , Hemofilia B/sangue , Hemofilia B/diagnóstico , Hemofilia B/epidemiologia , Humanos , Mutação INDEL , Masculino , Mutação de Sentido Incorreto , Razão de Chances , Linhagem , Fenótipo , Mutação Puntual , Prevalência , Conformação Proteica , Fatores de Risco , Deleção de Sequência , Índice de Gravidade de Doença , Relação Estrutura-Atividade
12.
Int J Mol Sci ; 12(10): 7271-85, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22072947

RESUMO

The factor VIII gene (F8) intron 22 inversion (Inv22) is a paradigmatic duplicon-mediated rearrangement, found in about one half of patients with severe hemophilia A worldwide. The identification of this prevalent cause of hemophilia was delayed for nine years after the F8 characterization in 1984. The aim of this review is to present the wide diversity of practical approaches that have been developed for genotyping the Inv22 (and related int22h rearrangements) since discovery in 1993. The sequence- Southern blot, long distance-PCR and inverse shifting-PCR-for Inv22 genotyping is an interesting example of scientific ingenuity and evolution in order to resolve challenging molecular diagnostic problems.


Assuntos
Fator VIII/genética , Hemofilia A/genética , Southern Blotting , DNA/análise , Rearranjo Gênico , Genótipo , Hemofilia A/diagnóstico , Hemofilia A/patologia , Humanos , Reação em Cadeia da Polimerase , Inversão de Sequência
14.
Haematologica ; 92(6): 842-5, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17550859

RESUMO

Hemophilia A (HA) is caused by heterogeneous mutations in the factor VIII gene (F8). This paper reports 16 novel small F8-mutations and rearrangements in a series of 80 Argentinian families with severe-HA. Using an updated scheme for F8-analysis, we found 37 F8-inversions (46%), 10 large deletions (13%), 13 small ins/del (16%), 7 nonsense (9%) and 8 missense mutations (10%), including 4 new ones (p.T233K, p.W1942R, p.L2297P and p.L2301S). The potential changes leading to severe-HA of these latter mutations were suggested by bioinformatics. The F8-mutation was characterised in 76 families (95%). They received genetic counselling and precise information about treatment design.


Assuntos
Fator VIII/genética , Hemofilia A/genética , Mutação , Argentina , Biologia Computacional , Saúde da Família , Feminino , Rearranjo Gênico , Aconselhamento Genético , Hemofilia A/epidemiologia , Humanos , Masculino , Epidemiologia Molecular
15.
Bol. Acad. Nac. Med. B.Aires ; 83(2): 325-334, jul.-dic. 2005. tab
Artigo em Espanhol | LILACS | ID: lil-567704

RESUMO

La hemofilia A (HA) y B (HB) son enfermedades hemorrágicas hereditarias ligadas al sexo causadas por defectos de los factores VIII y IX, respectivamente. Excepto grandes inversiones recurrentes involucradas en la mitad de las HA severas, el resto de las hemofilias son causadas por distintos tipos de mutaciones grandes y pequeñas. Fueron estudiadas 70 familias con HA severa (se), 6 con seHB, 1 con HA moderada-leve (m) y 2 con mHB. Primero, en seHA, se estudio la inversión del intrón 22 (Inv22) usando un nuevo abordaje basado en PCR inversa. En los casos negativos para las inversiones se estudiaron primariamente las grandes deleciones y secundariamente las mutaciones pequeñas. En familias con HA, encontramos la Inv22 en 43 por ciento de las seHAs, una única inversión del intrón 1, 10 grandes deleciones (catorce por ciento)y 23 mutaciones pequeñas (incluyendo 10 deleciones, 3 inserciones, 4 cambios nonsense, 5 missense y 1 de splicing); y en HB, 1 deleción afectando un sitio de splicing, 4 missense y 3 nonsense. Este esquema de caracterización de mutaciones permite un estudio y análisis molecular preciso de HA y HB y beneficiará tanto al asesoramiento genético como a la provisión de información clave para el diseño del tratamiento.


Assuntos
Humanos , Masculino , Feminino , Fator VIII/genética , Hemofilia A/classificação , Hemofilia A/genética , Hemofilia B/classificação , Hemofilia B/genética , Biologia Molecular , Argentina , Sequência de Bases , Southern Blotting , Deleção Cromossômica , Íntrons/genética , Inversão Cromossômica/genética , Mutação/genética , Polimorfismo Genético , Reação em Cadeia da Polimerase/métodos
16.
Hemoglobin ; 29(2): 141-50, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15921166

RESUMO

We describe a child with ATR-16 [alpha-thalassemia (thal)/mental retardation], who was referred for genetic evaluation because of minor anomalies and developmental delay. Cytogenetic analysis demonstrated a de novo complex rearrangement of chromosome 16. Fluorescence in situ hybridization (FISH) analysis, using chromosome 16 subtelomeric probes, showed that this patient had a deletion of the distal short arm of chromosome 16 that contains the alpha-globin genes and a duplication of 16q. Analysis of the alpha-globin locus by Southern blot showed a half normal dose of the alpha-globin gene. Microsatellite marker studies revealed that the duplicated 16q region was maternal in origin. Hematological studies revealed anemia, hypochromia and occasional cells with Hb H inclusion bodies. A hematological screening for alpha-thal should be considered in patients with mild developmental delay and a suggestive phenotype of ATR-16 with microcytic hypochromic anemia and normal iron status. The stellate pattern of the iris, a new finding in our patient, may contribute to a better clinical delineation of both syndromes, ATR-16 and/or duplication of 16qter.


Assuntos
Aberrações Cromossômicas , Cromossomos Humanos Par 16/genética , Deficiência Intelectual/genética , Talassemia alfa/genética , Análise Citogenética/métodos , Análise Mutacional de DNA/métodos , Genótipo , Hemoglobina H/análise , Hemoglobinas Anormais/genética , Humanos , Hibridização in Situ Fluorescente/métodos , Lactente , Deficiência Intelectual/diagnóstico , Masculino , Mutação Puntual/genética , Deleção de Sequência , Talassemia alfa/diagnóstico
17.
Clin Chem ; 51(7): 1154-8, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15860568

RESUMO

BACKGROUND: Factor VIII intron 22 inversions (Inv22) cause 40%-45% of severe cases of hemophilia A in all human populations. Currently, Inv22 can be analyzed either by Southern blotting or by rapid long-distance-PCR-based approaches. We describe an alternative method using inverse-PCR (I-PCR). METHODS: I-PCR involved 3 steps: (a) BclI restriction; (b) self-ligation of restriction fragments, providing BclI rings; and (c) standard multiplex-PCR analysis. PCR was achieved by use of a set of 3 primers that yielded a 487-bp amplicon for the nonrearranged intragenic allele and a 559-bp amplicon for the Inv22 allele. Specific primer sites were targeted by masking relevant regions for human repeats and low-complexity DNA. Inv22 I-PCR was applied to samples from 16 individuals (8 women and 8 men) representing 24 X chromosomes previously genotyped by Southern blotting. Additionally, we evaluated the sensitivity and the ability to assess eventual Inv22 carrier mosaicisms by experiments using artificial DNA mixtures (Inv22 + no-Inv22 male samples). RESULTS: Results for previously genotyped samples agreed with results of Southern blot analyses. As expected, cell composition of the artificial mosaic was linearly reflected by the relative intensities of Inv22 signals. I-PCR was estimated to detect Inv22-positive cells at concentrations as low as approximately 5%. CONCLUSION: The proposed technique provides a rapid tool for Inv22 genotyping.


Assuntos
Inversão Cromossômica , Hemofilia A/genética , Southern Blotting , Cromossomos Humanos , Fator VIII/genética , Feminino , Genótipo , Heterozigoto , Humanos , Íntrons , Masculino , Reação em Cadeia da Polimerase/métodos
18.
Hum Mutat ; 24(5): 440, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15459970

RESUMO

Although large deletions from the coagulation factor VIII gene, F8, are responsible for 5% of severe hemophilia A (seHA), few of them have been fully characterised. A detailed description of a large partial deletion of the F8 caused by unequal recombination between homeologous AluSx-derived sequences is presented. The proband, a case of isolated hemophilia A with a high inhibitor titre (5700 BU), showed a consistent absence of PCR-amplification of exons 4 to 10, EX4_EX10del. Two approaches were used to narrow down the deletion breakpoints: a direct physical analysis based on PCR (that additionally permits carrier detection in the family); and, under the hypothesis that the mutation resulted from homologous recombination, sequence alignments of F8 intron 3 and 10. Both approaches indicate an unequal crossing over (CO) between two Alu-related sequences. Both elements involved were derived from the AluSx-subfamily consensus and demonstrate 86% sequence identity (with only single-base mismatches), with three gaps (of 2, 3 and 14-bases) and two main tracts of perfectly homologous sequence (28 and 24-bp). The short stretch of intron 10 embedded into intron 3 sequence, linked to the CO, represents a typical hallmark of homologous recombination (double-strand break repair model). A detailed description of EX4_EX10del mutation is c.[338+3485delins1687+2223_1687+2225; 338+3551_1687+2291 del]. The common involvement of unequal homologous recombination mediated by repetitive elements allowed us to suggest that our experimental design (based on intron sequence alignments) may be successfully applied to rearrangements involved in other X-linked inherited diseases. Like other Alu-rich genes throughout the human genome, Alu-mediated homologous recombination in F8 may be an important cause of hemophilia by promoting large DNA deletions.


Assuntos
Elementos Alu/genética , Hemofilia A/genética , Recombinação Genética/genética , Homologia de Sequência do Ácido Nucleico , Sequência de Bases , Quebra Cromossômica/genética , Deleção Cromossômica , Sequência Consenso/genética , Análise Mutacional de DNA , Éxons/genética , Genômica , Humanos , Íntrons/genética , Modelos Genéticos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Deleção de Sequência/genética
19.
Blood Coagul Fibrinolysis ; 15(7): 569-72, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15389123

RESUMO

Besides intron 22 factor VIII gene inversion (Inv22), intron 1 inversion (Inv1) has recently been reported as a further recurrent mutation that causes approximately 5% of severe haemophilia A (HA) cases. We analysed the presence of the Inv1 in a group of 64 severe HA-affected families from Argentina, and found only one positive case. This Inv1 patient has not developed a factor VIII inhibitor, and the screening for small mutations in the coding sequences of the factor VIII gene did not detect any additional defect in this case. The Inv1 genotyping was further applied to analyse the haemophilia carrier status of the proband's sister. In addition, we studied the accuracy of the current polymerase chain reaction-based method to investigate the Inv1, and confirmed the absence of amplimer length polymorphisms associated to the Inv1-specific polymerase chain reaction amplifications in 101 X-chromosome haplotypes from unrelated Argentinian healthy males. In order to discuss Inv1 mutation frequency in severe HA and the risk of inhibitor formation, a review of the literature was included. Our data highlight the importance of analysis of the Inv1 in Inv22-negative severe HA cases. This will benefit both genetic counselling and the study of the relationship between genotype and inhibitor development.


Assuntos
Fator VIII/genética , Hemofilia A/genética , Íntrons/genética , Mutação , Argentina , Família , Feminino , Testes Genéticos , Hemofilia A/patologia , Humanos , Masculino , Reação em Cadeia da Polimerase , Análise de Sequência de DNA
20.
Haematologica ; 89(6): 746-7, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15257926

RESUMO

In order to determine the molecular heterogeneity of the beta-thalassemia gene and to analyze the influence of immigration from the Mediterranean Basin, a total of 254 families (475 subjects) from Argentinean beta-thalassemia patients were investigated using molecular biology techniques. This allowed us to provide a simplified diagnosis and genetic counselling of this disorder in Argentina.


Assuntos
Talassemia beta/etnologia , Talassemia beta/genética , Argentina/epidemiologia , Argentina/etnologia , Emigração e Imigração , Europa (Continente)/etnologia , Saúde da Família , Humanos , Epidemiologia Molecular , Mutação Puntual , Talassemia beta/epidemiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA