Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
J Allergy Clin Immunol ; 137(4): 1189-1196.e2, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26607704

ABSTRACT

BACKGROUND: Data on patients affected by chronic mucocutaneous candidiasis underscore the preponderant role of IL-17 receptor A (IL-17RA) in preserving mucocutaneous immunity. Little is known about the role of adenosine deaminase (ADA) 2 in regulation of immune responses, although recent reports linked ADA2 deficiency with inflammation and vasculitis. OBJECTIVE: We sought to investigate the mechanisms of chronic inflammation and vasculitis in a child lacking IL-17RA and ADA2 to identify therapeutic targets. METHODS: We report a family with 2 siblings who have had recurrent mucocutaneous infections with Candida albicans and Staphylococcus aureus and chronic inflammatory disease and vasculitis since early childhood, which were refractory to classical treatments. Array-based comparative genomic hybridization analysis showed that both siblings are homozygous for a 770-kb deletion on chr22q11.1 encompassing both IL17RA and cat eye critical region 1 (CECR1). Immunologic studies were carried out by means of flow cytometry, ELISA, and RIA. RESULTS: As expected, in the affected child we found a lack of IL-17RA expression, which implies a severe malfunction in the IL-17 signaling pathway, conferring susceptibility to recurrent mucocutaneous infections. Surprisingly, we detected an in vitro and in vivo upregulation of proinflammatory cytokines, notably IL-1ß and TNF-α, which is consistent with the persistent systemic inflammation. CONCLUSIONS: This work emphasizes the utility of whole-genome analyses combined with immunologic investigation in patients with unresolved immunodeficiency. This approach is likely to provide an insight into immunologic pathways and mechanisms of disease. It also provides molecular evidence for more targeted therapies. In addition, our report further corroborates a potential role of ADA2 in modulating immunity and inflammation.


Subject(s)
Adenosine Deaminase/deficiency , Adenosine Deaminase/genetics , Candidiasis, Chronic Mucocutaneous/genetics , Inflammation/genetics , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Receptors, Interleukin-17/deficiency , Receptors, Interleukin-17/genetics , Vasculitis/genetics , Adenosine Deaminase/immunology , Adolescent , Candidiasis, Chronic Mucocutaneous/complications , Candidiasis, Chronic Mucocutaneous/immunology , Child , Child, Preschool , Chronic Disease , Comparative Genomic Hybridization , Fatal Outcome , Female , Humans , Inflammation/complications , Inflammation/immunology , Intercellular Signaling Peptides and Proteins/immunology , Receptors, Interleukin-17/immunology , Sequence Deletion , Siblings , Vasculitis/complications , Vasculitis/immunology
2.
J Neurosci ; 35(15): 6093-106, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25878282

ABSTRACT

We have identified and characterized a spontaneous Brown Norway from Janvier rat strain (BN-J) presenting a progressive retinal degeneration associated with early retinal telangiectasia, neuronal alterations, and loss of retinal Müller glial cells resembling human macular telangiectasia type 2 (MacTel 2), which is a retinal disease of unknown cause. Genetic analyses showed that the BN-J phenotype results from an autosomal recessive indel novel mutation in the Crb1 gene, causing dislocalization of the protein from the retinal Müller glia (RMG)/photoreceptor cell junction. The transcriptomic analyses of primary RMG cultures allowed identification of the dysregulated pathways in BN-J rats compared with wild-type BN rats. Among those pathways, TGF-ß and Kit Receptor Signaling, MAPK Cascade, Growth Factors and Inflammatory Pathways, G-Protein Signaling Pathways, Regulation of Actin Cytoskeleton, and Cardiovascular Signaling were found. Potential molecular targets linking RMG/photoreceptor interaction with the development of retinal telangiectasia are identified. This model can help us to better understand the physiopathologic mechanisms of MacTel 2 and other retinal diseases associated with telangiectasia.


Subject(s)
Ependymoglial Cells/pathology , Eye Proteins/genetics , Mutation/genetics , Retinal Degeneration , Telangiectasis/complications , Telangiectasis/genetics , Age Factors , Animals , Animals, Newborn , Cells, Cultured , Disease Models, Animal , Electroretinography , Ependymoglial Cells/metabolism , Ependymoglial Cells/ultrastructure , Eye Proteins/metabolism , Fluorescein Angiography , Glial Fibrillary Acidic Protein/metabolism , Neurons/pathology , Neurons/ultrastructure , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Rats , Rats, Mutant Strains , Retinal Degeneration/etiology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Vessels/pathology , Retinal Vessels/ultrastructure , Signal Transduction/physiology , Visual Pathways/pathology , Visual Pathways/ultrastructure
3.
EMBO Mol Med ; 7(4): 380-93, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25724200

ABSTRACT

There is a widespread agreement from patient and professional organisations alike that the safety of stem cell therapeutics is of paramount importance, particularly for ex vivo autologous gene therapy. Yet current technology makes it difficult to thoroughly evaluate the behaviour of genetically corrected stem cells before they are transplanted. To address this, we have developed a strategy that permits transplantation of a clonal population of genetically corrected autologous stem cells that meet stringent selection criteria and the principle of precaution. As a proof of concept, we have stably transduced epidermal stem cells (holoclones) obtained from a patient suffering from recessive dystrophic epidermolysis bullosa. Holoclones were infected with self-inactivating retroviruses bearing a COL7A1 cDNA and cloned before the progeny of individual stem cells were characterised using a number of criteria. Clonal analysis revealed a great deal of heterogeneity among transduced stem cells in their capacity to produce functional type VII collagen (COLVII). Selected transduced stem cells transplanted onto immunodeficient mice regenerated a non-blistering epidermis for months and produced a functional COLVII. Safety was assessed by determining the sites of proviral integration, rearrangements and hit genes and by whole-genome sequencing. The progeny of the selected stem cells also had a diploid karyotype, was not tumorigenic and did not disseminate after long-term transplantation onto immunodeficient mice. In conclusion, a clonal strategy is a powerful and efficient means of by-passing the heterogeneity of a transduced stem cell population. It guarantees a safe and homogenous medicinal product, fulfilling the principle of precaution and the requirements of regulatory affairs. Furthermore, a clonal strategy makes it possible to envision exciting gene-editing technologies like zinc finger nucleases, TALENs and homologous recombination for next-generation gene therapy.


Subject(s)
Collagen Type VII , Epidermolysis Bullosa Dystrophica/therapy , Genetic Therapy/methods , Stem Cells/metabolism , Transduction, Genetic , Adult , Animals , Cells, Cultured , Collagen Type VII/biosynthesis , Collagen Type VII/genetics , Epidermis , Epidermolysis Bullosa Dystrophica/genetics , Epidermolysis Bullosa Dystrophica/metabolism , Epidermolysis Bullosa Dystrophica/pathology , Female , Heterografts , Humans , Infant, Newborn , Male , Mice , Mice, SCID , Retroviridae/genetics , Stem Cell Transplantation , Stem Cells/pathology
5.
Am J Med Genet A ; 164A(2): 346-52, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24376213

ABSTRACT

Frontonasal dysplasia (FND) is a genetically heterogeneous malformation spectrum with marked hypertelorism, broad nasal tip and bifid nose. Only a small number of genes have been associated with FND phenotypes until now, the first gene being EFNB1, related to craniofrontonasal syndrome (CFNS) with craniosynostosis in addition, and more recently the aristaless-like homeobox genes ALX3, ALX4, and ALX1, which have been related with distinct phenotypes named FND1, FND2, and FND3 respectively. We here report on a female patient presenting with severe FND features along with partial alopecia, hypogonadism and intellectual disability. While molecular investigations did not reveal mutations in any of the known genes, ALX4, ALX3, ALX1 and EFNB1, comparative genomic hybridization (array CGH) techniques showed a large heterozygous de novo deletion at 11p11.12p12, encompassing the ALX4 gene. Deletions in this region have been described in patients with Potocki-Shaffer syndrome (PSS), characterized by biparietal foramina, multiple exostoses, and intellectual disability. Although the patient reported herein manifests some overlapping features of FND and PPS, it is likely that the observed phenotype maybe due to a second unidentified mutation in the ALX4 gene. The phenotype will be discussed in view of the deleted region encompassing the ALX4 gene.


Subject(s)
Chromosome Disorders/genetics , Craniofacial Abnormalities/genetics , DNA-Binding Proteins/genetics , Exostoses, Multiple Hereditary/genetics , Face/abnormalities , Phenotype , Sequence Deletion , Transcription Factors/genetics , Chromosome Deletion , Chromosome Disorders/diagnosis , Chromosomes, Human, Pair 11/genetics , Comparative Genomic Hybridization , Craniofacial Abnormalities/diagnosis , Exons , Exostoses, Multiple Hereditary/diagnosis , Facial Bones/abnormalities , Facies , Female , Heterozygote , Humans , Imaging, Three-Dimensional/methods , Polymorphism, Single Nucleotide , Young Adult
6.
Am J Hum Genet ; 93(5): 798-811, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24140112

ABSTRACT

Copy-number variants (CNVs) represent a significant interpretative challenge, given that each CNV typically affects the dosage of multiple genes. Here we report on five individuals with coloboma, microcephaly, developmental delay, short stature, and craniofacial, cardiac, and renal defects who harbor overlapping microdeletions on 8q24.3. Fine mapping localized a commonly deleted 78 kb region that contains three genes: SCRIB, NRBP2, and PUF60. In vivo dissection of the CNV showed discrete contributions of the planar cell polarity effector SCRIB and the splicing factor PUF60 to the syndromic phenotype, and the combinatorial suppression of both genes exacerbated some, but not all, phenotypic components. Consistent with these findings, we identified an individual with microcephaly, short stature, intellectual disability, and heart defects with a de novo c.505C>T variant leading to a p.His169Tyr change in PUF60. Functional testing of this allele in vivo and in vitro showed that the mutation perturbs the relative dosage of two PUF60 isoforms and, subsequently, the splicing efficiency of downstream PUF60 targets. These data inform the functions of two genes not associated previously with human genetic disease and demonstrate how CNVs can exhibit complex genetic architecture, with the phenotype being the amalgam of both discrete dosage dysfunction of single transcripts and also of binary genetic interactions.


Subject(s)
DNA Copy Number Variations , Membrane Proteins/genetics , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Adolescent , Alleles , Animals , Child , Child, Preschool , Chromosome Mapping , Chromosomes, Human, Pair 8/genetics , Developmental Disabilities/genetics , Female , Gene Deletion , Gene Knockdown Techniques , HeLa Cells , Humans , Intellectual Disability/genetics , Male , Microcephaly/genetics , Phenotype , RNA Splicing Factors , Zebrafish/genetics
7.
PLoS One ; 8(3): e58048, 2013.
Article in English | MEDLINE | ID: mdl-23554873

ABSTRACT

The limited ability of common variants to account for the genetic contribution to complex disease has prompted searches for rare variants of large effect, to partly explain the 'missing heritability'. Analyses of genome-wide genotyping data have identified genomic structural variants (GSVs) as a source of such rare causal variants. Recent studies have reported multiple GSV loci associated with risk of obesity. We attempted to replicate these associations by similar analysis of two familial-obesity case-control cohorts and a population cohort, and detected GSVs at 11 out of 18 loci, at frequencies similar to those previously reported. Based on their reported frequencies and effect sizes (OR≥25), we had sufficient statistical power to detect the large majority (80%) of genuine associations at these loci. However, only one obesity association was replicated. Deletion of a 220 kb region on chromosome 16p11.2 has a carrier population frequency of 2×10(-4) (95% confidence interval [9.6×10(-5)-3.1×10(-4)]); accounts overall for 0.5% [0.19%-0.82%] of severe childhood obesity cases (P = 3.8×10(-10); odds ratio = 25.0 [9.9-60.6]); and results in a mean body mass index (BMI) increase of 5.8 kg.m(-2) [1.8-10.3] in adults from the general population. We also attempted replication using BMI as a quantitative trait in our population cohort; associations with BMI at or near nominal significance were detected at two further loci near KIF2B and within FOXP2, but these did not survive correction for multiple testing. These findings emphasise several issues of importance when conducting rare GSV association, including the need for careful cohort selection and replication strategy, accurate GSV identification, and appropriate correction for multiple testing and/or control of false discovery rate. Moreover, they highlight the potential difficulty in replicating rare CNV associations across different populations. Nevertheless, we show that such studies are potentially valuable for the identification of variants making an appreciable contribution to complex disease.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 16/genetics , Genetic Loci , Obesity/genetics , Adolescent , Adult , Aged , Body Mass Index , Child , Child, Preschool , Cohort Studies , Female , Forkhead Transcription Factors/genetics , Genome-Wide Association Study , Humans , Kinesins/genetics , Male , Middle Aged
8.
J Med Genet ; 49(10): 660-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23054248

ABSTRACT

BACKGROUND: The recurrent ~600 kb 16p11.2 BP4-BP5 deletion is among the most frequent known genetic aetiologies of autism spectrum disorder (ASD) and related neurodevelopmental disorders. OBJECTIVE: To define the medical, neuropsychological, and behavioural phenotypes in carriers of this deletion. METHODS: We collected clinical data on 285 deletion carriers and performed detailed evaluations on 72 carriers and 68 intrafamilial non-carrier controls. RESULTS: When compared to intrafamilial controls, full scale intelligence quotient (FSIQ) is two standard deviations lower in carriers, and there is no difference between carriers referred for neurodevelopmental disorders and carriers identified through cascade family testing. Verbal IQ (mean 74) is lower than non-verbal IQ (mean 83) and a majority of carriers require speech therapy. Over 80% of individuals exhibit psychiatric disorders including ASD, which is present in 15% of the paediatric carriers. Increase in head circumference (HC) during infancy is similar to the HC and brain growth patterns observed in idiopathic ASD. Obesity, a major comorbidity present in 50% of the carriers by the age of 7 years, does not correlate with FSIQ or any behavioural trait. Seizures are present in 24% of carriers and occur independently of other symptoms. Malformations are infrequently found, confirming only a few of the previously reported associations. CONCLUSIONS: The 16p11.2 deletion impacts in a quantitative and independent manner FSIQ, behaviour and body mass index, possibly through direct influences on neural circuitry. Although non-specific, these features are clinically significant and reproducible. Lastly, this study demonstrates the necessity of studying large patient cohorts ascertained through multiple methods to characterise the clinical consequences of rare variants involved in common diseases.


Subject(s)
Child Development Disorders, Pervasive/genetics , Chromosome Deletion , Chromosomes, Human, Pair 16 , Developmental Disabilities/genetics , Phenotype , Adolescent , Adult , Body Mass Index , Child , Child Development Disorders, Pervasive/diagnosis , Developmental Disabilities/diagnosis , Female , Gene Order , Heterozygote , Humans , Intelligence Tests , Male , Syndrome , Young Adult
9.
Am J Med Genet A ; 158A(9): 2152-61, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22847950

ABSTRACT

We describe 19 unrelated individuals with submicroscopic deletions involving 10p15.3 characterized by chromosomal microarray (CMA). Interestingly, to our knowledge, only two individuals with isolated, submicroscopic 10p15.3 deletion have been reported to date; however, only limited clinical information is available for these probands and the deleted region has not been molecularly mapped. Comprehensive clinical history was obtained for 12 of the 19 individuals described in this study. Common features among these 12 individuals include: cognitive/behavioral/developmental differences (11/11), speech delay/language disorder (10/10), motor delay (10/10), craniofacial dysmorphism (9/12), hypotonia (7/11), brain anomalies (4/6) and seizures (3/7). Parental studies were performed for nine of the 19 individuals; the 10p15.3 deletion was de novo in seven of the probands, not maternally inherited in one proband and inherited from an apparently affected mother in one proband. Molecular mapping of the 19 individuals reported in this study has identified two genes, ZMYND11 (OMIM 608668) and DIP2C (OMIM 611380; UCSC Genome Browser), mapping within 10p15.3 which are most commonly deleted. Although no single gene has been identified which is deleted in all 19 individuals studied, the deleted region in all but one individual includes ZMYND11 and the deleted region in all but one other individual includes DIP2C. There is not a clearly identifiable phenotypic difference between these two individuals and the size of the deleted region does not generally predict clinical features. Little is currently known about these genes complicating a direct genotype/phenotype correlation at this time. These data however, suggest that ZMYND11 and/or DIP2C haploinsufficiency contributes to the clinical features associated with 10p15 deletions in probands described in this study.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 10 , Telomere , Child , Female , Humans , Infant , Infant, Newborn , Male
10.
Acta Neuropathol ; 123(6): 841-52, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22249618

ABSTRACT

Glioblastoma (GBM) is a morphologically heterogeneous tumor type with a median survival of only 15 months in clinical trial populations. However, survival varies greatly among patients. As part of a central pathology review, we addressed the question if patients with GBM displaying distinct morphologic features respond differently to combined chemo-radiotherapy with temozolomide. Morphologic features were systematically recorded for 360 cases with particular focus on the presence of an oligodendroglioma-like component and respective correlations with outcome and relevant molecular markers. GBM with an oligodendroglioma-like component (GBM-O) represented 15% of all confirmed GBM (52/339) and was not associated with a more favorable outcome. GBM-O encompassed a pathogenetically heterogeneous group, significantly enriched for IDH1 mutations (19 vs. 3%, p = 0.003) and EGFR amplifications (71 vs. 48%, p = 0.04) compared with other GBM, while co-deletion of 1p/19q was found in only one case and the MGMT methylation frequency was alike (47 vs. 46%). Expression profiles classified most of the GBM-O into two subtypes, 36% (5/14 evaluable) as proneural and 43% as classical GBM. The detection of pseudo-palisading necrosis (PPN) was associated with benefit from chemotherapy (p = 0.0002), while no such effect was present in the absence of PPN (p = 0.86). In the adjusted interaction model including clinical prognostic factors and MGMT status, PPN was borderline nonsignificant (p = 0.063). Taken together, recognition of an oligodendroglioma-like component in an otherwise classic GBM identifies a pathogenetically mixed group without prognostic significance. However, the presence of PPN may indicate biological features of clinical relevance for further improvement of therapy.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Oligodendroglioma/pathology , Adolescent , Adult , Aged , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Chemoradiotherapy , Clinical Trials, Phase III as Topic , DNA Methylation , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Glioblastoma/genetics , Glioblastoma/therapy , Humans , Male , Middle Aged , Mutation , Oligodendroglioma/genetics , Oligodendroglioma/therapy , Prognosis , Survival Analysis , Temozolomide , Treatment Outcome , Young Adult
11.
Hum Mutat ; 33(1): 165-79, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21948486

ABSTRACT

We report 24 unrelated individuals with deletions and 17 additional cases with duplications at 10q11.21q21.1 identified by chromosomal microarray analysis. The rearrangements range in size from 0.3 to 12 Mb. Nineteen of the deletions and eight duplications are flanked by large, directly oriented segmental duplications of >98% sequence identity, suggesting that nonallelic homologous recombination (NAHR) caused these genomic rearrangements. Nine individuals with deletions and five with duplications have additional copy number changes. Detailed clinical evaluation of 20 patients with deletions revealed variable clinical features, with developmental delay (DD) and/or intellectual disability (ID) as the only features common to a majority of individuals. We suggest that some of the other features present in more than one patient with deletion, including hypotonia, sleep apnea, chronic constipation, gastroesophageal and vesicoureteral refluxes, epilepsy, ataxia, dysphagia, nystagmus, and ptosis may result from deletion of the CHAT gene, encoding choline acetyltransferase, and the SLC18A3 gene, mapping in the first intron of CHAT and encoding vesicular acetylcholine transporter. The phenotypic diversity and presence of the deletion in apparently normal carrier parents suggest that subjects carrying 10q11.21q11.23 deletions may exhibit variable phenotypic expressivity and incomplete penetrance influenced by additional genetic and nongenetic modifiers.


Subject(s)
Abnormalities, Multiple/genetics , Chromosome Aberrations , Nerve Growth Factors/genetics , Segmental Duplications, Genomic/genetics , Sequence Deletion , Vesicular Acetylcholine Transport Proteins/genetics , Child , Child, Preschool , Chromosome Mapping , Chromosomes, Human, Pair 10 , DNA Copy Number Variations , Developmental Disabilities/complications , Developmental Disabilities/genetics , Female , Genetic Variation , Homologous Recombination , Humans , In Situ Hybridization, Fluorescence , Infant , Intellectual Disability/complications , Intellectual Disability/genetics , Male , Oligonucleotide Array Sequence Analysis , Penetrance
12.
Nature ; 478(7367): 97-102, 2011 Aug 31.
Article in English | MEDLINE | ID: mdl-21881559

ABSTRACT

Both obesity and being underweight have been associated with increased mortality. Underweight, defined as a body mass index (BMI) ≤ 18.5 kg per m(2) in adults and ≤ -2 standard deviations from the mean in children, is the main sign of a series of heterogeneous clinical conditions including failure to thrive, feeding and eating disorder and/or anorexia nervosa. In contrast to obesity, few genetic variants underlying these clinical conditions have been reported. We previously showed that hemizygosity of a ∼600-kilobase (kb) region on the short arm of chromosome 16 causes a highly penetrant form of obesity that is often associated with hyperphagia and intellectual disabilities. Here we show that the corresponding reciprocal duplication is associated with being underweight. We identified 138 duplication carriers (including 132 novel cases and 108 unrelated carriers) from individuals clinically referred for developmental or intellectual disabilities (DD/ID) or psychiatric disorders, or recruited from population-based cohorts. These carriers show significantly reduced postnatal weight and BMI. Half of the boys younger than five years are underweight with a probable diagnosis of failure to thrive, whereas adult duplication carriers have an 8.3-fold increased risk of being clinically underweight. We observe a trend towards increased severity in males, as well as a depletion of male carriers among non-medically ascertained cases. These features are associated with an unusually high frequency of selective and restrictive eating behaviours and a significant reduction in head circumference. Each of the observed phenotypes is the converse of one reported in carriers of deletions at this locus. The phenotypes correlate with changes in transcript levels for genes mapping within the duplication but not in flanking regions. The reciprocal impact of these 16p11.2 copy-number variants indicates that severe obesity and being underweight could have mirror aetiologies, possibly through contrasting effects on energy balance.


Subject(s)
Body Mass Index , Chromosomes, Human, Pair 16/genetics , Gene Dosage/genetics , Obesity/genetics , Phenotype , Thinness/genetics , Adolescent , Adult , Aged , Aging , Body Height/genetics , Case-Control Studies , Child , Child, Preschool , Cohort Studies , Comparative Genomic Hybridization , Developmental Disabilities/genetics , Energy Metabolism/genetics , Europe , Female , Gene Duplication/genetics , Gene Expression Profiling , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Head/anatomy & histology , Heterozygote , Humans , Infant , Infant, Newborn , Male , Mental Disorders/genetics , Middle Aged , Mutation/genetics , North America , RNA, Messenger/analysis , RNA, Messenger/genetics , Sequence Deletion/genetics , Transcription, Genetic , Young Adult
13.
Nucleic Acids Res ; 39(15): e104, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21652640

ABSTRACT

Gene transfer and expression in eukaryotes is often limited by a number of stably maintained gene copies and by epigenetic silencing effects. Silencing may be limited by the use of epigenetic regulatory sequences such as matrix attachment regions (MAR). Here, we show that successive transfections of MAR-containing vectors allow a synergistic increase of transgene expression. This finding is partly explained by an increased entry into the cell nuclei and genomic integration of the DNA, an effect that requires both the MAR element and iterative transfections. Fluorescence in situ hybridization analysis often showed single integration events, indicating that DNAs introduced in successive transfections could recombine. High expression was also linked to the cell division cycle, so that nuclear transport of the DNA occurs when homologous recombination is most active. Use of cells deficient in either non-homologous end-joining or homologous recombination suggested that efficient integration and expression may require homologous recombination-based genomic integration of MAR-containing plasmids and the lack of epigenetic silencing events associated with tandem gene copies. We conclude that MAR elements may promote homologous recombination, and that cells and vectors can be engineered to take advantage of this property to mediate highly efficient gene transfer and expression.


Subject(s)
Matrix Attachment Regions , Recombination, Genetic , Transfection , Transgenes , Active Transport, Cell Nucleus , Animals , Cell Cycle , Cell Line , Cell Nucleus/metabolism , DNA/chemistry , DNA/metabolism , Gene Dosage , Gene Expression , Genetic Vectors , Plasmids/genetics , Sequence Homology, Nucleic Acid
14.
Pediatr Crit Care Med ; 12(6): e427-32, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21572369

ABSTRACT

OBJECTIVE: Report of a 16q24.1 deletion in a premature newborn, demonstrating the usefulness of array-based comparative genomic hybridization in persistent pulmonary hypertension of the newborn and multiple congenital malformations. DESIGN: Descriptive case report. SETTING: Genetic department and neonatal intensive care unit of a tertiary care children's hospital. INTERVENTIONS: None. PATIENT: We report the case of a preterm male infant, born at 26 wks of gestation. A cardiac malformation and bilateral hydronephrosis were diagnosed at 19 wks of gestation. Karyotype analysis was normal, and a 22q11.2 microdeletion was excluded by fluorescence in situ hybridization analysis. A cesarean section was performed due to fetal distress. The patient developed persistent pulmonary hypertension unresponsive to mechanical ventilation and nitric oxide treatment and expired at 16 hrs of life. MEASUREMENTS AND MAIN RESULTS: An autopsy revealed partial atrioventricular canal malformation and showed bilateral dilation of the renal pelvocaliceal system with bilateral ureteral stenosis and annular pancreas. Array-based comparative genomic hybridization analysis (Agilent oligoNT 44K, Agilent Technologies, Santa Clara, CA) showed an interstitial microdeletion encompassing the forkhead box gene cluster in 16q24.1. Review of the pulmonary microscopic examination showed the characteristic features of alveolar capillary dysplasia with misalignment of pulmonary veins. Some features were less prominent due to the gestational age. CONCLUSIONS: Our review of the literature shows that alveolar capillary dysplasia with misalignment of pulmonary veins is rare but probably underreported. Prematurity is not a usual presentation, and histologic features are difficult to interpret. In our case, array-based comparative genomic hybridization revealed a 16q24.1 deletion, leading to the final diagnosis of alveolar capillary dysplasia with misalignment of pulmonary veins. It emphasizes the usefulness of array-based comparative genomic hybridization analysis as a diagnostic tool with implications for both prognosis and management decisions in newborns with refractory persistent pulmonary hypertension and multiple congenital malformations.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 16/genetics , Hypertension, Pulmonary/pathology , Persistent Fetal Circulation Syndrome/pathology , Pulmonary Veins/abnormalities , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Comparative Genomic Hybridization , Humans , Infant, Newborn , Karyotype , Male , Persistent Fetal Circulation Syndrome/genetics , Pulmonary Alveoli/abnormalities , Pulmonary Alveoli/pathology
15.
PLoS One ; 6(4): e18369, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21494657

ABSTRACT

Cancer genomes frequently contain somatic copy number alterations (SCNA) that can significantly perturb the expression level of affected genes and thus disrupt pathways controlling normal growth. In melanoma, many studies have focussed on the copy number and gene expression levels of the BRAF, PTEN and MITF genes, but little has been done to identify new genes using these parameters at the genome-wide scale. Using karyotyping, SNP and CGH arrays, and RNA-seq, we have identified SCNA affecting gene expression ('SCNA-genes') in seven human metastatic melanoma cell lines. We showed that the combination of these techniques is useful to identify candidate genes potentially involved in tumorigenesis. Since few of these alterations were recurrent across our samples, we used a protein network-guided approach to determine whether any pathways were enriched in SCNA-genes in one or more samples. From this unbiased genome-wide analysis, we identified 28 significantly enriched pathway modules. Comparison with two large, independent melanoma SCNA datasets showed less than 10% overlap at the individual gene level, but network-guided analysis revealed 66% shared pathways, including all but three of the pathways identified in our data. Frequently altered pathways included WNT, cadherin signalling, angiogenesis and melanogenesis. Additionally, our results emphasize the potential of the EPHA3 and FRS2 gene products, involved in angiogenesis and migration, as possible therapeutic targets in melanoma. Our study demonstrates the utility of network-guided approaches, for both large and small datasets, to identify pathways recurrently perturbed in cancer.


Subject(s)
DNA Copy Number Variations/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks/genetics , Genes, Neoplasm/genetics , Melanoma/genetics , Melanoma/pathology , Signal Transduction/genetics , Cell Line, Tumor , Comparative Genomic Hybridization , Databases, Genetic , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Neoplasm Metastasis , Polymorphism, Single Nucleotide/genetics , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
16.
Eur J Hum Genet ; 19(4): 400-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21248748

ABSTRACT

The genomic architecture of the 10q22q23 region is characterised by two low-copy repeats (LCRs3 and 4), and deletions in this region appear to be rare. We report the clinical and molecular characterisation of eight novel deletions and six duplications within the 10q22.3q23.3 region. Five deletions and three duplications occur between LCRs3 and 4, whereas three deletions and three duplications have unique breakpoints. Most of the individuals with the LCR3-4 deletion had developmental delay, mainly affecting speech. In addition, macrocephaly, mild facial dysmorphisms, cerebellar anomalies, cardiac defects and congenital breast aplasia were observed. For congenital breast aplasia, the NRG3 gene, known to be involved in early mammary gland development in mice, is a putative candidate gene. For cardiac defects, BMPR1A and GRID1 are putative candidate genes because of their association with cardiac structure and function. Duplications between LCRs3 and 4 are associated with variable phenotypic penetrance. Probands had speech and/or motor delays and dysmorphisms including a broad forehead, deep-set eyes, upslanting palpebral fissures, a smooth philtrum and a thin upper lip. In conclusion, duplications between LCRs3 and 4 on 10q22.3q23.2 may lead to a distinct facial appearance and delays in speech and motor development. However, the phenotypic spectrum is broad, and duplications have also been found in healthy family members of a proband. Reciprocal deletions lead to speech and language delay, mild facial dysmorphisms and, in some individuals, to cerebellar, breast developmental and cardiac defects.


Subject(s)
Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Chromosomes, Human, Pair 10/genetics , Segmental Duplications, Genomic/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Body Dysmorphic Disorders/genetics , Body Dysmorphic Disorders/pathology , Bone Morphogenetic Protein Receptors, Type I/genetics , Child , Chromosome Deletion , DNA Copy Number Variations , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Female , Humans , Language Development Disorders/genetics , Male , Megalencephaly/genetics , Megalencephaly/pathology , Mice , Natural Cytotoxicity Triggering Receptor 3/genetics , Phenotype
17.
Sci Transl Med ; 3(64): 64ra1, 2011 Jan 05.
Article in English | MEDLINE | ID: mdl-21209411

ABSTRACT

Fragile X syndrome (FXS) is an X-linked condition associated with intellectual disability and behavioral problems. It is caused by expansion of a CGG repeat in the 5' untranslated region of the fragile X mental retardation 1 (FMR1) gene. This mutation is associated with hypermethylation at the FMR1 promoter and resultant transcriptional silencing. FMR1 silencing has many consequences, including up-regulation of metabotropic glutamate receptor 5 (mGluR5)-mediated signaling. mGluR5 receptor antagonists have shown promise in preclinical FXS models and in one small open-label study of FXS. We examined whether a receptor subtype-selective inhibitor of mGluR5, AFQ056, improves the behavioral symptoms of FXS in a randomized, double-blind, two-treatment, two-period, crossover study of 30 male FXS patients aged 18 to 35 years. We detected no significant effects of treatment on the primary outcome measure, the Aberrant Behavior Checklist-Community Edition (ABC-C) score, at day 19 or 20 of treatment. In an exploratory analysis, however, seven patients with full FMR1 promoter methylation and no detectable FMR1 messenger RNA improved, as measured with the ABC-C, significantly more after AFQ056 treatment than with placebo (P < 0.001). We detected no response in 18 patients with partial promoter methylation. Twenty-four patients experienced an adverse event, which was mostly mild to moderately severe fatigue or headache. If confirmed in larger and longer-term studies, these results suggest that blockade of the mGluR5 receptor in patients with full methylation at the FMR1 promoter may show improvement in the behavioral attributes of FXS.


Subject(s)
Epigenesis, Genetic/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Adult , DNA Methylation/genetics , Fragile X Syndrome/drug therapy , Humans , Male , Middle Aged , Promoter Regions, Genetic/genetics , Receptor, Metabotropic Glutamate 5 , Young Adult
18.
Clin Cancer Res ; 17(2): 255-66, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21097691

ABSTRACT

PURPOSE: Quantitative methylation-specific tests suggest that not all cells in a glioblastoma with detectable promoter methylation of the O6-methylguanine DNA methyltransferase (MGMT) gene carry a methylated MGMT allele. This observation may indicate cell subpopulations with distinct MGMT status, raising the question of the clinically relevant cutoff of MGMT methylation therapy. Epigenetic silencing of the MGMT gene by promoter methylation blunts repair of O6-methyl guanine and has been shown to be a predictive factor for benefit from alkylating agent therapy in glioblastoma. EXPERIMENTAL DESIGN: Ten paired samples of glioblastoma and respective glioblastoma-derived spheres (GS), cultured under stem cell conditions, were analyzed for the degree and pattern of MGMT promoter methylation by methylation-specific clone sequencing, MGMT gene dosage, chromatin status, and respective effects on MGMT expression and MGMT activity. RESULTS: In glioblastoma, MGMT-methylated alleles ranged from 10% to 90%. In contrast, methylated alleles were highly enriched (100% of clones) in respective GS, even when 2 MGMT alleles were present, with 1 exception (<50%). The CpG methylation patterns were characteristic for each glioblastoma exhibiting 25% to 90% methylated CpGs of 28 sites interrogated. Furthermore, MGMT promoter methylation was associated with a nonpermissive chromatin status in accordance with very low MGMT transcript levels and undetectable MGMT activity. CONCLUSIONS: In MGMT-methylated glioblastoma, MGMT promoter methylation is highly enriched in GS that supposedly comprise glioma-initiating cells. Thus, even a low percentage of MGMT methylation measured in a glioblastoma sample may be relevant and predict benefit from an alkylating agent therapy.


Subject(s)
Brain Neoplasms/genetics , DNA Methylation , Glioblastoma/genetics , O(6)-Methylguanine-DNA Methyltransferase/genetics , Adult , Aged , Aged, 80 and over , Chromatin/ultrastructure , Female , Gene Frequency , Humans , Male , Middle Aged , Promoter Regions, Genetic , Tumor Cells, Cultured
19.
Rev Med Suisse ; 6(237): 390-2, 394-6, 2010 Feb 24.
Article in French | MEDLINE | ID: mdl-20383968

ABSTRACT

Structural genomic abnormalities play a key role in the pathogenesis of human disorders and represent one of the first causes of mental impairment, complex syndromes and tumors. In order to detect these chromosomal abnormalities, many methodologies have been developed with limits. The new ARRAY based Comparative Genomic Hybridization (ARRAY CGH) is a revolutionary approach which allows to characterize very small genetic abnormalities undetectable by the standard approaches and in the absence of any associated clinical information. The aim of this article is to describe why the application of a new array CGH methodology is necessary in the etiological search for genetic diseases, what the limits of the standard approaches are and to whom arrayCGH analyses can be applied in a pediatric environment. Examples of our practice will be presented.


Subject(s)
Comparative Genomic Hybridization , Chromosome Aberrations , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/genetics , Humans
20.
Am J Med Genet A ; 149A(12): 2661-5, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19938077

ABSTRACT

We report on two familial cases from a non-consanguineous marriage, presenting multiple intestinal and choanal atresia. Massive hydramnios and dilatation of the bowel were observed at 29 weeks of gestation during routine ultrasound scan of a healthy mother. The fetal karyotype was normal and cystic fibrosis screening was negative. Regular scans were performed throughout the pregnancy. The child was born at 34 weeks gestation. Choanal atresia was diagnosed at birth and abdominal investigations showed multiple atresia interesting both the small bowel and the colon. Further interventions were necessary because of recurrent obstructions. During the following pregnancy, a dilatation of the fetal intestinal tract was detected by ultrasonography at 27 weeks of gestation. Pregnancy was interrupted. Post-mortem examination of the fetus confirmed the stenosis of long segments of the small intestine associated with areas of colonic atresia. In both cases, histology and distribution were consistent with those reported in hereditary multiple intestinal atresia (HMIA). An association between multiple intestinal and choanal atresia has never been reported. We suggest it could correspond to a new autosomal recessive entity for which cytogenetic investigations and high-resolution array CGH revealed no visible anomalies.


Subject(s)
Abnormalities, Multiple/epidemiology , Abnormalities, Multiple/pathology , Choanal Atresia/complications , Choanal Atresia/epidemiology , Intestinal Atresia/complications , Intestinal Atresia/epidemiology , Family , Female , Fetus/abnormalities , Fetus/pathology , Hair/abnormalities , Hair/ultrastructure , Humans , Infant, Newborn , Intestinal Atresia/pathology , Male , Pedigree , Pregnancy , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL
...