Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters











Publication year range
1.
Front Genet ; 14: 1031074, 2023.
Article in English | MEDLINE | ID: mdl-37609037

ABSTRACT

Rapid-onset Obesity with Hypothalamic dysfunction, Hypoventilation and Autonomic Dysregulation (ROHHAD) is a rare, life-threatening, pediatric disorder of unknown etiology, whose diagnosis is made difficult by poor knowledge of clinical manifestation, and lack of any confirmatory tests. Children with ROHHAD usually present with rapid onset weight gain which may be followed, over months or years, by hypothalamic dysfunction, hypoventilation, autonomic dysfunction, including impaired bowel motility, and tumors of neural crest origin. Despite the lack of evidence of inheritance in ROHHAD, several studies have been conducted in recent years that have explored possible genetic origins, with unsuccessful results. In order to broaden the search for possible genetic risk factors, an attempt was made to analyse the non-coding variants in two trios (proband with parents), recruited in the Gaslini Children's Hospital in Genoa (Italy). Both patients were females, with a typical history of ROHHAD. Gene variants (single nucleotide variants, short insertions/deletions, splice variants or in tandem expansion of homopolymeric tracts) or altered genomic regions (copy number variations or structural variants) shared between the two probands were searched. Currently, we have not found any potentially pathogenic changes, consistent with the ROHHAD clinical phenotype, and involving genes, regions or pathways shared between the two trios. To definitively rule out the genetic etiology, third-generation sequencing technologies (e.g., long-reads sequencing, optical mapping) should be applied, as well as other pathways, including those associated with immunological and autoimmune disorders, should be explored, making use not only of genomics but also of different -omic datasets.

2.
Gastric Cancer ; 16(4): 596-601, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23291969

ABSTRACT

PURPOSE: Gastrointestinal stromal tumors (GISTs) are mesenchymal tumors of the gastrointestinal tract. Most (80 %) contain activating mutations in the KIT receptor tyrosine kinase, roughly 10 % in platelet-derived growth factor receptor-alpha (PDGFRA). In a small subset, BRAF mutations are an alternative molecular pathway. GISTs respond well to imatinib, but low response is seen in patients with wild-type KIT or PDGFRA. Resistance has also been reported as a result of mutations in downstream effectors such as BRAF. METHODS: We provide here a molecular characterization of a series of primary GISTs from Italian patients. Of 121 GIST cases diagnosed between 2000 and 2012, 83 were evaluated by PCR amplification and direct sequencing for mutations in KIT exons 8, 9, 11, 13, and 17, PDGFRA exons 12, 14, and 18, and BRAF exon 15. Eighty-one GISTs also underwent K-RAS testing. RESULTS: Sixty-four GISTs were positive: 55 had mutations in KIT and 9 in PDGFRA; 16 patients were mutation negative. Three samples came from NF1 patients and were KIT- and PDGFRA negative. Overall, we identified six novel mutations in KIT (p.K550_M552delinsL, p.Q556_W557delinsG p.Q556_G575del, p.W557_V559delinsQ p.P573_R588dup, p.G592_K593dup) and one novel mutation in PDGFRA (p.D842_N848delinsVDV), thus contributing to widening the spectrum of known mutations in GIST tumors and confirming the most frequently altered regions underlying GIST development. CONCLUSIONS: Among the 64 KIT- and PDGFRA-positive sporadic patients in our series, no BRAF or KRAS mutations were identified, suggesting that co-occurrence of these mutations is likely to be rare in the northwestern Italian population and not a frequent cause of primary resistance to imatinib in KIT-positive GIST patients.


Subject(s)
Gastrointestinal Stromal Tumors/genetics , Mutation/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , ras Proteins/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Biomarkers, Tumor/genetics , Female , Follow-Up Studies , Gastrointestinal Stromal Tumors/drug therapy , Humans , Imatinib Mesylate , Male , Middle Aged , Piperazines/therapeutic use , Polymerase Chain Reaction , Prognosis , Proto-Oncogene Proteins p21(ras) , Pyrimidines/therapeutic use , Retrospective Studies
3.
Fam Cancer ; 8(4): 547-53, 2009.
Article in English | MEDLINE | ID: mdl-19728162

ABSTRACT

Lynch syndrome is an inherited cancer syndrome caused by germline mutations in mismatch repair (MMR) genes MLH1, MSH2, MSH6 and PMS2. LS predisposes to high risk of early-onset colorectal, endometrial and other tumors. Patients with Lynch syndrome have also been shown to have an elevated risk for pancreatic cancer (PC). In this study, we aimed to estimate the frequency of suspected Lynch syndrome among a series of 135 PC patients. Further, we wanted to determine the frequency of MMR gene mutations in the suspected Lynch syndrome cases. We also aimed to verify the pathogenicity of any novel non-truncating variants we might detect with a functional assay. Based on personal and/or familial cancer history, 19 patients were classified as suspected Lynch syndrome cases. DNA material for mutation analysis was available for eleven of them. Four patients were found to carry a total of five MLH1 or MSH2 variants. Of these, MSH2-Q402X, MSH2-G322D, and MLH1-K618A had been previously reported, while the MSH2-E205Q and MSH2-V367I variants were novel. MSH2-Q402X is a known stop mutation and reported here for the first time here in association with PC. MLH1-K618A was found in the unaffected branch of a kindred, suggesting that it may be a polymorphism or a low penetrance variant. MSH2-G322D likely does not cause a MMR defect, although this variant has also been associated with breast cancer as indeed seen in our patient. The novel variants MSH2-E205Q and MSH2-V367I were found in the same patient. Both novel variants were however functional in the applied MMR assay. Our findings suggest that only a small subset of pancreatic cancer patients carry pathogenic MMR mutations.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Genetic Predisposition to Disease , MutS Homolog 2 Protein/genetics , Nuclear Proteins/genetics , Pancreatic Neoplasms/genetics , Adult , Base Sequence , Colorectal Neoplasms, Hereditary Nonpolyposis/complications , DNA Mismatch Repair/genetics , DNA Mutational Analysis , Female , Germ-Line Mutation , Humans , Immunohistochemistry , Italy , Male , Microsatellite Instability , Middle Aged , MutL Protein Homolog 1 , Pedigree , Reverse Transcriptase Polymerase Chain Reaction
4.
Am J Med Genet A ; 149A(7): 1539-43, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19533801

ABSTRACT

Gorlin syndrome (GS) is inherited in an autosomal dominant pattern with high-penetrance and is characterized by a range of developmental anomalies and increased risk of developing basal cell carcinoma and medulloblastoma. Between 50% and 85% of patients with GS harbor germ line mutations in the only susceptibility gene identified to date, PTCH1, a key component in the Sonic Hedgehog signaling pathway. Another component in this pathway, SUFU, is known to be involved in susceptibility to medulloblastoma but has never been reported in GS patients to date. We have identified the known c.1022 + 1G>A SUFU germ line splicing mutation in a family that was PTCH1-negative and who had signs and symptoms of GS, including medulloblastoma. This is the first report of a germ line SUFU mutation associated with GS.


Subject(s)
Basal Cell Nevus Syndrome/genetics , Germ-Line Mutation , Repressor Proteins/genetics , Adult , Basal Cell Nevus Syndrome/diagnosis , Base Sequence , Child, Preschool , Family , Female , Humans , Patched Receptors , Patched-1 Receptor , Receptors, Cell Surface/genetics
5.
Eur J Neurol ; 15(5): 520-4, 2008 May.
Article in English | MEDLINE | ID: mdl-18394049

ABSTRACT

BACKGROUND AND PURPOSE: The hereditary spastic paraplegias (HSPs) are a heterogeneous group of neurodegenerative disorders, characterized by a progressive spasticity of the lower limbs. So far, 33 different loci (SPGs) have been mapped and the 15 genes responsible have been identified. We mapped a locus responsible for a form of spastic paraplegia, complicated by bilateral cataracts, gastroesophageal reflux with persisting vomiting and amyotrophy to chromosome 10q23.3-q24.2, in an Italian family. The critical region was in a 12 cm chromosomal interval between markers D10S564 and D10S603 (SPG9, MIM601162). In the same region, two other forms of HSP have been recently mapped: SPG27 and SPG33. In the latter case, the gene responsible has been identified. MATERIALS AND METHODS: To better characterize this region, we genotyped individuals from SPG9-linked families using additional markers and reduced the candidate region to a 4.8 Mb, excluding several genes by positional cloning. RESULTS: The refined SPG9 locus is positioned completely within SPG27 and does not include the SPG33 gene. DISCUSSION: Fifty-two transcripts are present in the refined critical region and 25 strong candidates have been excluded as disease causing genes by direct sequencing. Six of them were also excluded as responsible for SPG27.


Subject(s)
Chromosomes, Human, Pair 10/genetics , Genetic Heterogeneity , Physical Chromosome Mapping/methods , Spastic Paraplegia, Hereditary/genetics , Chromosome Mapping/methods , Family Health , Female , Genetic Linkage , Genotype , Humans , Italy , Lod Score , Male , Mutation , Sequence Analysis, DNA
7.
Child Care Health Dev ; 31(3): 351-4, 2005 May.
Article in English | MEDLINE | ID: mdl-15840155

ABSTRACT

BACKGROUND: Diagnosis of Nevoid Basal Cell Carcinoma Syndrome (NBCCS) in infants may pose significant challenges to clinicians owing to its variable expressivity and age-related manifestations. METHODS: We report two paediatric cases of NBCCS who presented initially with a non-specific phenotype. RESULTS: In case 1, a diagnosis of NBCCS was possible only after the father was interviewed and found to present with two major criteria for the disease. Subsequent molecular testing confirmed the diagnosis. In case 2, molecular testing of the infant and his father had diagnostic value as neither satisfied fully the current diagnostic criteria for NBCCS. CONCLUSIONS: Presence of the few clinical manifestations of NBCCS that appear in infants, typically congenital malformations and skeletal abnormalities, should prompt clinicians to conduct in-person interviews with both parents. In general, paediatricians should refer both parents of infants who are suspected of having an inherited condition to clinical geneticists for expert examination, given the potential unreliability of reported medical history.


Subject(s)
Basal Cell Nevus Syndrome/diagnosis , Basal Cell Nevus Syndrome/genetics , Codon, Nonsense , Family Health , Fathers , Humans , Infant , Male , Phenotype
8.
Hum Mutat ; 25(3): 322-3, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15712338

ABSTRACT

Mutations in the PTCH gene, the human homolog of the Drosophila patched gene, have been found to lead to the autosomal dominant disorder termed Nevoid Basal Cell Carcinoma Syndrome (NBCCS, also called Gorlin Syndrome). Patients display an array of developmental anomalies and are prone to develop a variety of tumors, with multiple Basal Cell Carcinomas occurring frequently. We provide here the results of molecular testing of a set of Italian Nevoid Basal Cell Carcinoma Syndrome patients. Twelve familial patients belonging to 7 kindreds and 5 unaffected family members, 6 non-familial patients and an additional set of 7 patients with multiple Basal Cell Carcinoma but no other criteria for the disease were examined for mutations in the PTCH gene. All of the Nevoid Basal Cell Carcinoma Syndrome patients were found to carry variants of the PTCH gene. We detected nine novel mutations (1 of which occurring twice): 1 missense mutation (c.1436T>G [p.L479R]), 1 nonsense mutation (c.1138G>T [p.E380X]), 6 frameshift mutations (c.323_324ins2, c.2011_2012dup, c.2535_2536dup, c.2577_2583del, c.3000_3005del, c.3050_3051del), 1 novel splicing variant (c.6552A>T) and 3 mutations that have been previously reported (c.3168+5G>A, c.1526G>T [p.G509V], and c.3499G>A [p.G1167R]). None of the patients with multiple Basal Cell Carcinoma but no other criteria for the syndrome, carried germline coding region mutations.


Subject(s)
Basal Cell Nevus Syndrome/genetics , Codon, Nonsense , Frameshift Mutation , Mutation, Missense , Point Mutation , RNA Splice Sites/genetics , Receptors, Cell Surface/genetics , Adolescent , Adult , Amino Acid Sequence , Child , Consensus Sequence , DNA Mutational Analysis , Female , Humans , Italy , Male , Middle Aged , Molecular Sequence Data , Patched Receptors , Patched-1 Receptor , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid
9.
Ann Oncol ; 15(1): 70-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14679123

ABSTRACT

BACKGROUND: Roughly 40% of germinal mutations in melanoma families (MF) affect p16(INK4a) and p14(ARF). We investigated the association between INK4/ARF alterations and the occurrence of pancreatic cancer in MF and in sporadic pancreatic cancer (SPC) patients. PATIENTS AND METHODS: Forty-nine MF, 66 SPC cases and 54 controls were enrolled. The INK4/ARF locus was screened. RESULTS: As compared with the general population, the risk of pancreatic cancer (PC) was increased 9.4-fold [95% confidence interval (CI) 2.7-33.4] and 2.2-fold (95% CI 0.8-5.7) in G101W-positive and -negative MF, respectively, while mean ages at onset were 61 and 77 years, respectively. A 1.7 (95% CI 1.06-2.79) increased risk of cancer at any site was observed among first-degree relatives of SPC cases as compared with controls. The G101W founder mutation was detected in 4% of SPC cases but the rate increased to 13% when tumor clustering in either branch of families was taken into account. One G101W-positive PC patient with a melanoma in a first-degree relative harbored a germline deletion of the second allele, including exon 1B. CONCLUSIONS: The presence of a deletion including exon 1B in two PC patients points to the involvement of p14(ARF) in the development of PC and may suggest that the increased risk of PC in MF is caused by impairment of both loci.


Subject(s)
Adenocarcinoma/genetics , Genes, p16 , Germ-Line Mutation , Pancreatic Neoplasms/genetics , Tumor Suppressor Protein p14ARF/genetics , Adenocarcinoma/epidemiology , Adolescent , Adult , Aged , Aged, 80 and over , DNA Mutational Analysis , Female , Genetic Predisposition to Disease , Heterozygote , Humans , Incidence , Male , Melanoma/epidemiology , Melanoma/genetics , Melanoma/physiopathology , Middle Aged , Molecular Sequence Data , Pancreatic Neoplasms/epidemiology , Pedigree , Polymerase Chain Reaction
10.
Int J Mol Med ; 10(5): 575-8, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12373294

ABSTRACT

The objective of this study was to investigate MICA (major histocompatibility complex MHC class I chain-related genes) polymorphisms in an Italian series of patients with juvenile Behcet disease (jBD) and to compare these genetic findings with the high prevalence of inflammatory mucosal disease, which occurs in Western populations. Ten families which included at least 1 affected patient were studied. We genotyped 18 patients (13 children and 5 adults) affected with the complete or incomplete form of jBD comparing the results to those found in a population of 20 apparently healthy individuals. The MICA transmembrane polymorphism was analysed by PCR and polyacrylamide gel electrophoresis. HLA typing was assessed by SSP-PCR technique. Statistical analysis was performed using chi2 based methods. In our series the prevalence of gastrointestinal disease was high (41%). Seven of 10 patients were HLA-B51 positive. MICA A6 allele was present in 70% of probands as compared to 25% of an ethnically matched control population. On the other hand, MICA A5.1 was present in 20% of probands as compared to 60% in controls. Out of 5 A6 homozygotes, 2 probands and 2 affected relatives developed a severe gut inflammatory disease. The study of MICA gene polymorphisms disclosed an independent association with genetic risk for jBD. The combination of MICA A6 and HLA-B51 is the strongest genetic marker for this disease. Homozygous A6 patients seem to develop more severe mucosal gut involvement. This finding sheds light on the role of a receptor for MICA, named NKG2D, presented by natural killer cells, and CD8+, alphabetaT cells and gammadeltaT cells, usually localised in gut mucosa.


Subject(s)
Behcet Syndrome/genetics , Behcet Syndrome/immunology , Histocompatibility Antigens Class I/genetics , Polymorphism, Genetic , Adult , Alleles , Case-Control Studies , Child , Female , HLA-B Antigens/genetics , HLA-B51 Antigen , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Italy , Male , Risk Factors
11.
Prenat Diagn ; 21(8): 668-71, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11536268

ABSTRACT

A prenatal diagnosis of Pelizaeus-Merzbacher disease (PMD) resulting from proteolipid protein gene (PLP) duplication was performed by a quantitative fluorescent multiplex PCR method. PLP gene copy number was determined in the proband, the pregnant mother, the male fetus and two aunts. Small amounts of genomic DNA extracted from peripheral blood and from chorionic villi were used. The fetus, in common with the proband, was identified as PMD-affected being a carrier of the PLP gene duplication, inherited from the mother, while the two aunts were non-carriers. The data obtained were confirmed by segregation analysis of a PLP-associated dinucleotide-repeat polymorphism amplified by the same multiplex PCR.


Subject(s)
Myelin Proteolipid Protein/genetics , Pelizaeus-Merzbacher Disease/diagnosis , Case-Control Studies , Diagnosis, Differential , Female , Gene Duplication , Humans , Male , Pedigree , Pelizaeus-Merzbacher Disease/genetics , Polymerase Chain Reaction , Pregnancy , Pregnancy Trimester, First , Prenatal Diagnosis
12.
Hum Mutat ; 18(2): 164-5, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11462244

ABSTRACT

Mucopolysaccharidosis type II (MPS2, or Hunter syndrome), rare X-linked lysosomal storage disorder, results from deleterious mutations in the iduronate-2-sulfatase (IDS) gene. We report here the mutational analysis of a total of 40 unrelated Italian MPS II patients ranging from mild to severe phenotype. We are able to assign the genotype to 29 of them (72.5%), identifying 22 different mutations, five of which are unpublished (c.533delTT, W12X, N265I, c.1131-1142del, c.1131-1305del). A total of 55.2% of the molecularly characterised patients resulted from missense mutations, 20.7% from nonsense mutations, and another 13.8% of patients from small deletions (<20pb) or splice mutations, whereas 10.3% of the cases carried major structural alterations such as large deletion and rearrangements. The results reported here support the evidence of the mutational heterogeneity of the IDS gene as well as the difficulty to correlate genotype and phenotype in the patients with MPSII. However, the molecular characterisation of the patients is advantageous, making the carrier detection feasible for the females in the family at risk and improving the reliability of prenatal diagnosis techniques. Moreover, it provides a good foundation for therapeutic strategies.


Subject(s)
Iduronate Sulfatase/genetics , Mucopolysaccharidosis II/enzymology , Mucopolysaccharidosis II/genetics , Mutation/genetics , Cells, Cultured , Codon, Nonsense/genetics , DNA Mutational Analysis , Exons/genetics , Female , Genotype , Humans , Italy , Mucopolysaccharidosis II/diagnosis , Mucopolysaccharidosis II/physiopathology , Mutation, Missense/genetics , Phenotype , Prenatal Diagnosis , RNA Splice Sites/genetics , Sequence Deletion/genetics
13.
Hum Mutat ; 18(1): 85, 2001.
Article in English | MEDLINE | ID: mdl-11439001

ABSTRACT

Pseudoxanthoma elasticum (PXE) is a mendelian disorder characterized by calcification of elastic fibers in skin, arteries, and retina. It results in dermal lesions, arterial insufficiency and retinal hemorrhages, leading to macular degeneration. PXE is transmitted either as an autosomal dominant or recessive trait and several sporadic cases have been observed. Mutations in the ABCC6 gene have been identified very recently in patients. Here, we report on a large Italian family affected by pseudoxanthoma elasticum for which linkage analysis had pointed to a region encompassing markers D16S3069-D16S405-D16S3103; hemizygosity of marker D16S405 allowed us to detect a submicroscopic deletion of at least 900 kb involving ABCC6, ABCC1, and MYH11. Mutation analysis on the other allele of the family, as well as on two additional sporadic cases, revealed nonsense (Y227X, R518X, R1164X) and frame-shift (c.960delC) mutations in ABCC6 (MRP6) further confirming the role of this multi-drug resistance gene in the etiology of pseudoxanthoma elasticum. Furthermore, clinical re-examination of members of the family harboring the deletion led to the detection of additional features, potentially caused by the deletion of the MYH11 gene. In the course of the analysis five nonpathogenic variants were found in ABCC6: 1233T>C, 1245G>A, 1838 T>G (V614A), 1890C>G, and 3506+83C>A. Hum Mutat 18:85, 2001.


Subject(s)
Multidrug Resistance-Associated Proteins/genetics , Myosin Heavy Chains/genetics , Point Mutation/genetics , Pseudoxanthoma Elasticum/genetics , Sequence Deletion/genetics , Smooth Muscle Myosins/genetics , Adult , Aged , Alleles , Chromosome Mapping , DNA Mutational Analysis , Exons/genetics , Female , Genetic Markers/genetics , Haplotypes/genetics , Humans , Italy , Male , Pedigree , Pseudoxanthoma Elasticum/pathology
14.
Am J Med Genet ; 100(3): 214-8, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11343306

ABSTRACT

Craniosynostosis is determined by the precocious fusion of one or more calvarial sutures leading to an abnormal skull shape. Additionally, nodular heterotopia is a disorder of neuronal migration and/or proliferation. We describe a very rare multiple congenital anomalies (MCA) syndrome in which craniosynostosis is associated with bilateral periventricular nodular heterotopia (BPNH) of the gray matter and other malformations involving hands, feet, and the gut. Clinical findings and further investigations suggest the diagnosis of craniosynostosis Fontaine-Farriaux type. To the best of our knowledge, this case is only the second report of this MCA syndrome. Based on the clinical and radiological data of the two cases reported, we hypothesize that this malformative complex may be considered a new BPNH/MCA syndrome and propose to classify it as BPNH/craniosynostosis. Previous studies demonstrated that at least two BPNH/MCA syndromes have been mapped to the Xq28 chromosomal region in which a causative gene for isolated BPNH is located. The same authors hypothesized that other BPNH syndromes could be due to microrearrangements at the same Xq28 region. Our case presents several overlapping features with some BPNH/MCA syndromes and it is possible that this new complex disorder may be caused by rearrangements at the same chromosomal region that could alter expression of different genes in Xq28.


Subject(s)
Cerebral Ventricles/abnormalities , Craniosynostoses , Abnormalities, Multiple , Choristoma , Craniosynostoses/classification , Craniosynostoses/diagnosis , Craniosynostoses/genetics , Diagnosis, Differential , Genetic Linkage , Humans , Infant , Magnetic Resonance Imaging , Syndrome , X Chromosome
15.
Atherosclerosis ; 154(3): 599-605, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11257260

ABSTRACT

The proband is a 50 year-old woman born from a consanguineous marriage. She has been suffering from angina pectoris since the age of 38 and underwent coronary bypass surgery for three-vessel disease at 48. The presence of low plasma levels of total cholesterol and high density lipoprotein (HDL) cholesterol (2.4 and 0.1 mmol/l) and apo AI (<15 mg/dl), associated with corneal lesions and a mild splenomegaly suggested the diagnosis of Tangier disease. However, none of the other features of Tangier disease, including hepatomegaly, anemia and peripheral neuropathy, were present. The analysis of the dinucleotide microsatellites located in chromosome 9q31 region demonstrated that the proband was homozygous for the alleles of D9S53, D9S1784 and D9S1832. The mother and son of the proband, both with low levels of HDL cholesterol, shared one of the proband's haplotypes, whereas neither of these haplotypes was present in the normolipidemic proband's sister. The sequence of ATP-binding cassette transporter 1 (ABC1-1) cDNA obtained by reverse transcription-PCR (RT-PCR) of total RNA isolated from cultured fibroblasts showed that the proband was homozygous for a C>T transition in exon 13, which caused a tryptophane for arginine substitution (R527W). This mutation was confirmed by direct sequencing of exon 13 amplified from genomic DNA. It can be easily screened, as the nucleotide change introduces a restriction site for the enzyme Afl III. R527W substitution occurs in a highly conserved region of the NH2 cytoplasmic domain of ABC1 protein. R527W co-segregates with the low HDL phenotype in the family and was not found in 200 chromosomes from normolipidemic individuals.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Coronary Disease/genetics , Glycoproteins/genetics , Point Mutation , Tangier Disease/genetics , ATP Binding Cassette Transporter 1 , Amino Acid Sequence/genetics , Base Sequence/genetics , Chromosomes, Human, Pair 9/genetics , Coronary Disease/physiopathology , Female , Genetic Testing , Genotype , Humans , Middle Aged , Molecular Sequence Data , Mutation/genetics , Pedigree , Phenotype , Polymorphism, Genetic , Severity of Illness Index
17.
Int J Mol Med ; 7(1): 85-9, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11115614

ABSTRACT

Two families with recurrence of neuroblastoma one Italian and one British with three and two affected children respectively were genotyped using polymorphic markers on chromosome 1 spanning the p32-p36 region frequently deleted in neuroblastoma tumor cells. Linkage to this region was excluded by haplotype inspection and negative lod scores. Furthermore, the exclusion of genes involved in neurocristopathies sometimes associated with neuroblastoma was carried out by typing the Italian family with polymorphic markers located in or near the corresponding genes. Finally, linkage analysis in the two families showed negative lod scores for markers spanning the 16p12-13 chromosomal region where a locus for familial neuroblastoma has been recently mapped. Our findings indicate that different genes are involved in the pathogenesis of familial neuroblastoma.


Subject(s)
Chromosome Mapping , Genetic Predisposition to Disease/genetics , Neuroblastoma/genetics , England , Family Health , Female , Humans , Italy , Lod Score , Male , Microsatellite Repeats , Neuroblastoma/pathology , Pedigree
18.
Biotechniques ; 29(5): 1012-4, 1016-7, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11084863

ABSTRACT

In this report, we present a fluorescence-based approach to the assessment of cellular gene expression and transcription rates. Nuclear run-on was performed by supplying biotin-16-UTP to nuclei, and labeled transcripts were bound to streptavidin-coated magnetic beads. Total cDNA was then synthesized by means of random hexamer primed reverse transcription of captured molecules. To monitor transcript abundance in cDNA, both from nuclear run-on and total RNA, we propose a semiquantitative PCR approach based on the use of fluorescent primers.


Subject(s)
Biotinylation , Cell Nucleus/genetics , Drosophila Proteins , Magnetics , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Cycloheximide/pharmacology , DNA Primers , Fluorescence , Humans , Microspheres , Nucleic Acid Hybridization , Proto-Oncogene Proteins , Proto-Oncogene Proteins c-ret , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , Receptor Protein-Tyrosine Kinases , Transcription, Genetic/drug effects , Tretinoin/pharmacology , Tumor Cells, Cultured , Uridine Triphosphate/analogs & derivatives , Uridine Triphosphate/metabolism
19.
Eur J Hum Genet ; 8(11): 895-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11093280

ABSTRACT

Fechtner syndrome is an autosomal dominant disorder which has been thought to be a variant of Alport syndrome. It is characterised by nephritis, sensorineural hearing loss and eye abnormalities, as well as by macrothrombocytopenia and polymorphonuclear inclusion bodies. Recently, the Fechtner syndrome has been mapped in a 5.5 Mb region on the long arm of chromosome 22 by linkage analysis in an extended Israeli family. We describe here the genetic refinement of the Fechtner critical interval to a region less than 600 Kb by linkage analysis performed in a large Italian pedigree. The presence of several recombination events allowed the disease gene to be localised between markers D22S278 and D22S426, in a region containing only two non-recombinant markers, D22S1173 and D22S283. This interval, spanning <600 Kb on genomic DNA, has been entirely sequenced and contains six known and three putative genes.


Subject(s)
Abnormalities, Multiple/genetics , Chromosomes, Human, Pair 22/genetics , Hearing Loss, Sensorineural , Nephritis , Abnormalities, Multiple/pathology , Adult , Child , Chromosome Mapping , DNA/genetics , Eye Abnormalities , Family Health , Female , Genotype , Haplotypes , Humans , Italy , Male , Microsatellite Repeats , Middle Aged , Pedigree , Syndrome , Thrombocytopenia
20.
Eur J Hum Genet ; 8(10): 777-82, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11039578

ABSTRACT

Hereditary spastic paraplegia (HSP) is a genetically heterogeneous disorder characterised by progressive spasticity of the lower limbs. Beside 'pure' forms of HSP, 'complicated' forms are reported, where spasticity occurs associated with additional symptoms. We recently described an Italian family with a complicated dominant form of HSP (SPG9) and we mapped the gene responsible to 10q23.3-q24.2, in a 12cM interval between markers D10S564 and D10S603. The phenotypic manifestations in our family are reminiscent of those already described in a smaller British pedigree. We typed individuals from this British family using markers located in the SPG9 critical interval and haplotype reconstruction showed the disorder co-segregating with SPG9. To characterise the SPG9 region better, we constructed a contig of 22 YACs, assigned it to 18 polymorphic markers and positioned 54 ESTs. Furthermore, we searched for ESTs containing a trinucleotide repeat sequence, since anticipation of symptoms was reported in both families. Finally, analysis of a muscle biopsy specimen from one patient was normal, suggesting that, contrary to SPG7, mitochondrial disturbance could not be a primary feature of SPG9.


Subject(s)
Chromosomes, Human, Pair 10/genetics , Physical Chromosome Mapping/methods , Spastic Paraplegia, Hereditary/genetics , Trinucleotide Repeats/genetics , Biopsy , Chromosome Mapping , Contig Mapping , Expressed Sequence Tags , Female , Genotype , Humans , Male , Microsatellite Repeats , Muscles/surgery , Pedigree , Sequence Analysis, DNA , Spastic Paraplegia, Hereditary/complications
SELECTION OF CITATIONS
SEARCH DETAIL