Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Oxf Med Case Reports ; 2023(2): omad005, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36860961

ABSTRACT

Hyper-IgM syndrome (HIGM) is a rare immunodeficiency phenotype that is usually accompanied by serious infections. We present a curious case of the incidental detection of HIGM in a 45-year-old male with complement C1q deficiency. He had relatively mild sinopulmonary infections, recurrent skin infections and lipomas in his adulthood. Investigations revealed normal enumeration of total peripheral blood B cells and reduced expression of CD40L on his CD4+ T cells. C1q was noted to be absent, due to a peripheral inhibitor such as an autoantibody. Genomic sequencing of the patient and his parents revealed a novel, de novo heterozygous mutation in the ATM (ataxia telangiectasia mutated) gene although he displayed no clinical evidence of ataxia telangiectasia. This is a rare case of HIGM and acquired C1q deficiency. We present full phenotyping data that contributes to the growing understanding to these interesting immunodeficiencies.

2.
Genome Med ; 13(1): 32, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33632298

ABSTRACT

Whole genome sequencing (WGS) has the potential to outperform clinical microarrays for the detection of structural variants (SV) including copy number variants (CNVs), but has been challenged by high false positive rates. Here we present ClinSV, a WGS based SV integration, annotation, prioritization, and visualization framework, which identified 99.8% of simulated pathogenic ClinVar CNVs > 10 kb and 11/11 pathogenic variants from matched microarrays. The false positive rate was low (1.5-4.5%) and reproducibility high (95-99%). In clinical practice, ClinSV identified reportable variants in 22 of 485 patients (4.7%) of which 35-63% were not detectable by current clinical microarray designs. ClinSV is available at https://github.com/KCCG/ClinSV .


Subject(s)
DNA Copy Number Variations/genetics , Software , Whole Genome Sequencing , Gene Frequency/genetics , Humans , Molecular Sequence Annotation , Mutation/genetics , Reproducibility of Results
3.
Eur J Hum Genet ; 29(5): 760-770, 2021 05.
Article in English | MEDLINE | ID: mdl-33437033

ABSTRACT

Autosomal Dominant Polycystic Kidney Disease (ADPKD) is common, with a prevalence of 1/1000 and predominantly caused by disease-causing variants in PKD1 or PKD2. Clinical diagnosis is usually by age-dependent imaging criteria, which is challenging in patients with atypical clinical features, without family history, or younger age. However, there is increasing need for definitive diagnosis of ADPKD with new treatments available. Sequencing is complicated by six pseudogenes that share 97% homology to PKD1 and by recently identified phenocopy genes. Whole-genome sequencing can definitively diagnose ADPKD, but requires validation for clinical use. We initially performed a validation study, in which 42 ADPKD patients underwent sequencing of PKD1 and PKD2 by both whole-genome and Sanger sequencing, using a blinded, cross-over method. Whole-genome sequencing identified all PKD1 and PKD2 germline pathogenic variants in the validation study (sensitivity and specificity 100%). Two mosaic variants outside pipeline thresholds were not detected. We then examined the first 144 samples referred to a clinically-accredited diagnostic laboratory for clinical whole-genome sequencing, with targeted-analysis to a polycystic kidney disease gene-panel. In this unselected, diagnostic cohort (71 males :73 females), the diagnostic rate was 70%, including a diagnostic rate of 81% in patients with typical ADPKD (98% with PKD1/PKD2 variants) and 60% in those with atypical features (56% PKD1/PKD2; 44% PKHD1/HNF1B/GANAB/ DNAJB11/PRKCSH/TSC2). Most patients with atypical disease did not have clinical features that predicted likelihood of a genetic diagnosis. These results suggest clinicians should consider diagnostic genomics as part of their assessment in polycystic kidney disease, particularly in atypical disease.


Subject(s)
Gene Frequency , Genetic Testing/methods , Polycystic Kidney Diseases/genetics , Whole Genome Sequencing/methods , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Genetic Testing/standards , Glucosidases/genetics , HSP40 Heat-Shock Proteins/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Humans , Infant , Male , Middle Aged , Polycystic Kidney Diseases/diagnosis , Receptors, Cell Surface/genetics , Sensitivity and Specificity , TRPP Cation Channels/genetics , Tuberous Sclerosis Complex 2 Protein/genetics , Whole Genome Sequencing/standards
5.
Nat Commun ; 9(1): 5319, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30552336

ABSTRACT

Disorders of sex development (DSDs) are conditions affecting development of the gonads or genitalia. Variants in two key genes, SRY and its target SOX9, are an established cause of 46,XY DSD, but the genetic basis of many DSDs remains unknown. SRY-mediated SOX9 upregulation in the early gonad is crucial for testis development, yet the regulatory elements underlying this have not been identified in humans. Here, we identified four DSD patients with overlapping duplications or deletions upstream of SOX9. Bioinformatic analysis identified three putative enhancers for SOX9 that responded to different combinations of testis-specific regulators. All three enhancers showed synergistic activity and together drive SOX9 in the testis. This is the first study to identify SOX9 enhancers that, when duplicated or deleted, result in 46,XX or 46,XY sex reversal, respectively. These enhancers provide a hitherto missing link by which SRY activates SOX9 in humans, and establish SOX9 enhancer mutations as a significant cause of DSD.


Subject(s)
Disorders of Sex Development/genetics , Gene Duplication/genetics , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Sequence Deletion/genetics , Sex-Determining Region Y Protein/genetics , 46, XX Disorders of Sex Development/genetics , Animals , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Disorder of Sex Development, 46,XY/genetics , Female , Gene Knockout Techniques , Genitalia/metabolism , Gonads/metabolism , Humans , Male , Mice , Mutation , RNA Splicing Factors/metabolism , Regulatory Sequences, Nucleic Acid/genetics , SOX Transcription Factors/genetics , Sex Differentiation , Testis/growth & development , Testis/metabolism
6.
Sex Dev ; 12(1-3): 19-29, 2018.
Article in English | MEDLINE | ID: mdl-29145200

ABSTRACT

Despite considerable research effort and significant advances in sequencing technologies, the majority of disorders of sex development (DSD) cases still lack a molecular genetic diagnosis. While coding variants have been discovered in known and candidate DSD genes, comparatively little is known about copy number variations (CNVs) affecting both coding and noncoding regions. Due to rapidly falling costs of whole genome sequencing, many more CNVs in individuals with DSD will be identified. These CNVs may explain a significant number of hitherto undiagnosed cases of DSD. In this review, we provide an overview of CNVs that are known to cause DSD and discuss approaches to identify and verify causative CNVs.


Subject(s)
DNA Copy Number Variations/genetics , Disorders of Sex Development/genetics , Chromosome Mapping , High-Throughput Nucleotide Sequencing , Humans , Open Reading Frames/genetics , Regulatory Sequences, Nucleic Acid/genetics
7.
Hum Mutat ; 39(1): 124-139, 2018 01.
Article in English | MEDLINE | ID: mdl-29027299

ABSTRACT

Variants in the NR5A1 gene encoding SF1 have been described in a diverse spectrum of disorders of sex development (DSD). Recently, we reported the use of a targeted gene panel for DSD where we identified 15 individuals with a variant in NR5A1, nine of which are novel. Here, we examine the functional effect of these changes in relation to the patient phenotype. All novel variants tested had reduced trans-activational activity, while several had altered protein level, localization, or conformation. In addition, we found evidence of new roles for SF1 protein domains including a region within the ligand binding domain that appears to contribute to SF1 regulation of Müllerian development. There was little correlation between the severity of the phenotype and the nature of the NR5A1 variant. We report two familial cases of NR5A1 deficiency with evidence of variable expressivity; we also report on individuals with oligogenic inheritance. Finally, we found that the nature of the NR5A1 variant does not inform patient outcomes (including pubertal androgenization and malignancy risk). This study adds nine novel pathogenic NR5A1 variants to the pool of diagnostic variants. It highlights a greater need for understanding the complexity of SF1 function and the additional factors that contribute.


Subject(s)
Disorders of Sex Development/diagnosis , Disorders of Sex Development/genetics , Genetic Association Studies , Genetic Variation , Phenotype , Steroidogenic Factor 1/genetics , Alleles , Amino Acid Sequence , Disorder of Sex Development, 46,XY/diagnosis , Disorder of Sex Development, 46,XY/genetics , Female , Genetic Association Studies/methods , Genotype , Humans , Male , Models, Anatomic , Mutation , Protein Conformation , Protein Domains/genetics , RNA Splice Sites , Sequence Analysis, DNA , Steroidogenic Factor 1/chemistry
8.
PLoS One ; 12(10): e0186331, 2017.
Article in English | MEDLINE | ID: mdl-29053721

ABSTRACT

Remarkable progress has been achieved in understanding the mechanisms controlling sex determination, yet the cause for many Disorders of Sex Development (DSD) remains unknown. Of particular interest is a rare XX DSD subtype in which individuals are negative for SRY, the testis determining factor on the Y chromosome, yet develop testes or ovotestes, and both of these phenotypes occur in the same family. This is a naturally occurring disorder in humans (Homo sapiens) and dogs (C. familiaris). Phenotypes in the canine XX DSD model are strikingly similar to those of the human XX DSD subtype. The purposes of this study were to identify 1) a variant associated with XX DSD in the canine model and 2) gene expression alterations in canine embryonic gonads that could be informative to causation. Using a genome wide association study (GWAS) and whole genome sequencing (WGS), we identified a variant on C. familiaris autosome 9 (CFA9) that is associated with XX DSD in the canine model and in affected purebred dogs. This is the first marker identified for inherited canine XX DSD. It lies upstream of SOX9 within the canine ortholog for the human disorder, which resides on 17q24. Inheritance of this variant indicates that XX DSD is a complex trait in which breed genetic background affects penetrance. Furthermore, the homozygous variant genotype is associated with embryonic lethality in at least one breed. Our analysis of gene expression studies (RNA-seq and PRO-seq) in embryonic gonads at risk of XX DSD from the canine model identified significant RSPO1 downregulation in comparison to XX controls, without significant upregulation of SOX9 or other known testis pathway genes. Based on these data, a novel mechanism is proposed in which molecular lesions acting upstream of RSPO1 induce epigenomic gonadal mosaicism.


Subject(s)
Chromosome Aberrations , Down-Regulation , Thrombospondins/genetics , Animals , Dogs , Genome-Wide Association Study
10.
Article in English | MEDLINE | ID: mdl-28620497

ABSTRACT

46,XX disorders of sexual development (DSDs) occur rarely and result from disruptions of the genetic pathways underlying gonadal development and differentiation. We present a case of a young phenotypic male with 46,XX SRY-negative ovotesticular DSD resulting from a duplication upstream of SOX9 presenting with a painful testicular mass resulting from ovulation into an ovotestis. We present a literature review of ovulation in phenotypic men and discuss the role of SRY and SOX9 in testicular development, including the role of SOX9 upstream enhancer region duplication in female-to-male sex reversal. LEARNING POINTS: In mammals, the early gonad is bipotent and can differentiate into either a testis or an ovary. SRY is the master switch in testis determination, responsible for differentiation of the bipotent gonad into testis.SRY activates SOX9 gene, SOX9 as a transcription factor is the second major gene involved in male sex determination. SOX9 drives the proliferation of Sertoli cells and activates AMH/MIS repressing the ovary. SOX9 is sufficient to induce testis formation and can substitute for SRY function.Assessing karyotype and then determination of the presence or absence of Mullerian structures are necessary serial investigations in any case of DSD, except for mixed gonadal dysgenesis identified by karyotype alone.Treatment is ideal in a multidisciplinary setting with considerations to genetic (implications to family and reproductive recurrence risk), psychological aspects (sensitive individualized counseling including patient gender identity and preference), endocrinological (hormone replacement), surgical (cosmetic, prophylactic gonadectomy) fertility preservation and reproductive opportunities and metabolic health (cardiovascular and bones).

11.
Birth Defects Res C Embryo Today ; 108(4): 337-350, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28033663

ABSTRACT

Disorders of Sex Development (DSDs) are a major paediatric concern and are estimated to occur in around 1.7% of all live births (Fausto-Sterling, Sexing the Body: Gender Politics and the Construction of Sexuality, Basic Books, New York, 2000). They are often caused by the breakdown in the complex genetic mechanisms that underlie gonadal development and differentiation. Having a genetic diagnosis can be important for patients with a DSD: it can increase acceptance of a disorder often surrounded by stigma, alter clinical management and it can assist in reproductive planning. While Massively Parallel Sequencing (MPS) is advancing the genetic diagnosis of rare Mendelian disorders, it is not yet clear which MPS assay is best suited for the clinical diagnosis of DSD patients and to what extent other established methods are still relevant. To complicate matters, DSDs represent a wide spectrum of disorders caused by an array of different genetic changes, many of which are yet unknown. Here we discuss the different genetic lesions that are known to contribute to different DSDs, and review the utility of a range of MPS approaches for diagnosing DSD patients. Birth Defects Research (Part C) 108:337-350, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Disorders of Sex Development/diagnosis , Disorders of Sex Development/genetics , Animals , DNA Copy Number Variations , Disorders of Sex Development/metabolism , Genetic Association Studies , Genetic Predisposition to Disease , Genetic Testing , Genomics/methods , Gonadal Disorders/genetics , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/statistics & numerical data , Humans , Sexual Development/genetics
12.
Genome Biol ; 17(1): 243, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27899157

ABSTRACT

BACKGROUND: Disorders of sex development (DSD) are congenital conditions in which chromosomal, gonadal, or phenotypic sex is atypical. Clinical management of DSD is often difficult and currently only 13% of patients receive an accurate clinical genetic diagnosis. To address this we have developed a massively parallel sequencing targeted DSD gene panel which allows us to sequence all 64 known diagnostic DSD genes and candidate genes simultaneously. RESULTS: We analyzed DNA from the largest reported international cohort of patients with DSD (278 patients with 46,XY DSD and 48 with 46,XX DSD). Our targeted gene panel compares favorably with other sequencing platforms. We found a total of 28 diagnostic genes that are implicated in DSD, highlighting the genetic spectrum of this disorder. Sequencing revealed 93 previously unreported DSD gene variants. Overall, we identified a likely genetic diagnosis in 43% of patients with 46,XY DSD. In patients with 46,XY disorders of androgen synthesis and action the genetic diagnosis rate reached 60%. Surprisingly, little difference in diagnostic rate was observed between singletons and trios. In many cases our findings are informative as to the likely cause of the DSD, which will facilitate clinical management. CONCLUSIONS: Our massively parallel sequencing targeted DSD gene panel represents an economical means of improving the genetic diagnostic capability for patients affected by DSD. Implementation of this panel in a large cohort of patients has expanded our understanding of the underlying genetic etiology of DSD. The inclusion of research candidate genes also provides an invaluable resource for future identification of novel genes.


Subject(s)
Chromosome Aberrations , Disorders of Sex Development/diagnosis , Disorders of Sex Development/genetics , High-Throughput Nucleotide Sequencing , Cohort Studies , Disorders of Sex Development/pathology , Female , Genetic Association Studies , Genetic Predisposition to Disease , Genetic Variation , Gonads/growth & development , Gonads/pathology , Humans , Male , Mutation/genetics , Ovary/growth & development , Ovary/pathology , Pedigree , Phenotype , Testis/growth & development , Testis/pathology
13.
PLoS Genet ; 12(5): e1006067, 2016 05.
Article in English | MEDLINE | ID: mdl-27227454

ABSTRACT

Most humans harbor both CD177neg and CD177pos neutrophils but 1-10% of people are CD177null, placing them at risk for formation of anti-neutrophil antibodies that can cause transfusion-related acute lung injury and neonatal alloimmune neutropenia. By deep sequencing the CD177 locus, we catalogued CD177 single nucleotide variants and identified a novel stop codon in CD177null individuals arising from a single base substitution in exon 7. This is not a mutation in CD177 itself, rather the CD177null phenotype arises when exon 7 of CD177 is supplied entirely by the CD177 pseudogene (CD177P1), which appears to have resulted from allelic gene conversion. In CD177 expressing individuals the CD177 locus contains both CD177P1 and CD177 sequences. The proportion of CD177hi neutrophils in the blood is a heritable trait. Abundance of CD177hi neutrophils correlates with homozygosity for CD177 reference allele, while heterozygosity for ectopic CD177P1 gene conversion correlates with increased CD177neg neutrophils, in which both CD177P1 partially incorporated allele and paired intact CD177 allele are transcribed. Human neutrophil heterogeneity for CD177 expression arises by ectopic allelic conversion. Resolution of the genetic basis of CD177null phenotype identifies a method for screening for individuals at risk of CD177 isoimmunisation.


Subject(s)
Isoantigens/biosynthesis , Neutropenia/immunology , Neutrophils/immunology , Pseudogenes/genetics , Receptors, Cell Surface/biosynthesis , Antibodies, Antineutrophil Cytoplasmic/biosynthesis , Antibodies, Antineutrophil Cytoplasmic/immunology , Blood Transfusion, Autologous/adverse effects , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , Gene Expression Regulation , Genetic Heterogeneity , Humans , Isoantigens/blood , Isoantigens/genetics , Isoantigens/immunology , Neutropenia/pathology , Neutrophils/metabolism , Polymorphism, Single Nucleotide , Pseudogenes/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Thrombocytopenia, Neonatal Alloimmune
14.
Sex Dev ; 10(2): 59-65, 2016.
Article in English | MEDLINE | ID: mdl-27078861

ABSTRACT

Despite recent advances in our understanding, most cases of disorders of sex development (DSD) cannot be explained by mutations in known genes. In genome-wide screens of DSD patients, we and others detected duplications or deletions of potential regulatory regions of known or suspected DSD genes. It is therefore likely that a significant proportion of DSD cases may be explained by disrupted transcriptional regulation of gonad genes. Despite many recent technological advances, limited availability of relevant tissues - especially human embryonic material - can make the identification of long-range regulatory elements extremely difficult. In an attempt to overcome this limitation, we evaluated the usefulness of publicly available DNaseI hypersensitivity data from the Roadmap Epigenomics Project. For this feasibility study we used the 'gene desert' around the SOX9 gene and a genomic locus downstream of GATA4. Over 60% of our selected candidate regions had significant enhancer activity in luciferase assays. We show that this approach facilitates the detection of strong enhancer candidates worthy of further analysis.


Subject(s)
Databases, Genetic , Disorders of Sex Development/genetics , Gene Expression Regulation/physiology , Female , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Gene Expression Regulation/genetics , Genome, Human/genetics , Humans , Male , Regulatory Sequences, Nucleic Acid/genetics , Regulatory Sequences, Nucleic Acid/physiology , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism
15.
Nat Rev Endocrinol ; 10(11): 673-83, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25246082

ABSTRACT

Sex-specific gonadal development starts with formation of the bipotential gonad, which then differentiates into either a mature testis or an ovary. This process is dependent on activation of either the testis-specific or the ovary-specific pathway while the opposite pathway is continuously repressed. A network of transcription factors tightly regulates initiation and maintenance of these distinct pathways; disruption of these networks can lead to disorders of sex development in humans and male-to-female or female-to-male sex reversal in mice. Sry is the Y-linked master switch that is both required and sufficient to drive the testis-determining pathway. Another key component of the testis pathway is Sox9, which acts immediately downstream of Sry. In contrast to the testis pathway, no single sex-determining factor has been identified in the ovary pathway; however, multiple genes, such as Foxl2, Rspo1, Ctnnb1, and Wnt4, seem to work synergistically and in parallel to ensure proper ovary development. Our understanding of the regulatory networks that underpin testis and ovary development has grown substantially over the past two decades.


Subject(s)
Gene Expression Regulation, Developmental , MicroRNAs/genetics , Ovary/embryology , Sex Determination Processes/genetics , Testis/embryology , Transcription Factors/genetics , Animals , Female , Humans , Male
16.
PLoS One ; 9(7): e101811, 2014.
Article in English | MEDLINE | ID: mdl-25003592

ABSTRACT

The chicken embryo has long been a useful model organism for studying development, including sex determination and gonadal differentiation. However, manipulating gene expression specifically in the embryonic avian gonad has been difficult. The viral vector RCASBP can be readily used for embryo-wide transgene expression; however global mis-expression using this method can cause deleterious off-target effects and embryo-lethality. In an attempt to develop vectors for the over-expression of sequences in chicken embryonic urogenital tissues, the viral vector RCANBP was engineered to contain predicted promoter sequences of gonadal-expressed genes. Several promoters were analysed and it was found that although the SF1 promoter produced a tissue-restricted expression pattern that was highest in the mesonephros and liver, it was also higher in the gonads compared to the rest of the body. The location of EGFP expression from the SF1 promoter overlapped with several key gonad-expressed sex development genes; however expression was generally low-level and was not seen in all gonadal cells. To further validate this sequence the key testis determinant DMRT1 was over-expressed in female embryos, which due to insufficient levels had no effect on gonad development. The female gene aromatase was then over-expressed in male embryos, which disrupted the testis pathway as demonstrated by a reduction in AMH protein. Taken together, although these data showed that the SF1 promoter can be used for functional studies in ovo, a stronger promoter sequence would likely be required for the functional analysis of gonad genes that require high-level expression.


Subject(s)
Gene Expression , Gonads/metabolism , Animals , Aromatase/genetics , Aromatase/metabolism , Chick Embryo , Female , Gene Order , Genes, Reporter , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Organ Specificity/genetics , Promoter Regions, Genetic , Reproducibility of Results , Retroviridae/genetics , Sex Differentiation/genetics , Transcription Factors/genetics , Transduction, Genetic
17.
Sex Dev ; 8(5): 262-72, 2014.
Article in English | MEDLINE | ID: mdl-24504012

ABSTRACT

One of the defining events during human embryonic development with the most far-reaching effects for the individual is whether the embryo develops as male or female. The crucial step in this process is the differentiation of the bipotential embryonic gonads into either testes or ovaries. If the embryo inherits X and Y sex chromosomes, the Y-linked SRY (sex determining region in Y) gene initiates a network of genes that results in a functional testis and ultimately a male phenotype. By contrast, in an embryo with 2 X chromosomes, the undifferentiated gonad develops as an ovary resulting in a female phenotype. Perturbation of any of the genes in either the testicular or ovarian developmental pathway can result in individuals with disorders of sex development. In this review, we provide a summary of known components of testicular or ovarian pathways and their antagonistic actions and give a brief overview of new technologies currently used to identify the missing pieces of the sex development network.


Subject(s)
Sex Differentiation/physiology , Sexual Development/physiology , Chromosomes, Human, X/genetics , Chromosomes, Human, Y/genetics , Disorders of Sex Development/genetics , Disorders of Sex Development/physiopathology , Female , Gonads/embryology , Gonads/metabolism , Gonads/physiopathology , Humans , Male , Sex Determination Processes , Sex Differentiation/genetics , Sexual Development/genetics
18.
Methods Mol Biol ; 786: 201-10, 2012.
Article in English | MEDLINE | ID: mdl-21938628

ABSTRACT

DNaseI-hypersensitive sites within chromatin are indicative of genomic loci with regulatory function. Several techniques have been described for analyzing these regions, but are either laborious, offer low-throughput possibilities, or are expensive. We have developed a new approach based on a modified version of multiplex ligation-dependent probe amplification (MLPA). Using this method, it is possible to analyse up to 50 defined genomic regions for DNaseI-hypersensitivity in a single PCR-based reaction. This chapter outlines the approach and discusses the critical features of each step of the procedure.


Subject(s)
Chromatin/genetics , Deoxyribonuclease I/metabolism , Nucleic Acid Amplification Techniques/methods , Binding Sites/genetics , Genome/genetics
19.
PLoS One ; 6(3): e17793, 2011 Mar 07.
Article in English | MEDLINE | ID: mdl-21408189

ABSTRACT

Disorders of sex development (DSD), ranging in severity from mild genital abnormalities to complete sex reversal, represent a major concern for patients and their families. DSD are often due to disruption of the genetic programs that regulate gonad development. Although some genes have been identified in these developmental pathways, the causative mutations have not been identified in more than 50% 46,XY DSD cases. We used the Affymetrix Genome-Wide Human SNP Array 6.0 to analyse copy number variation in 23 individuals with unexplained 46,XY DSD due to gonadal dysgenesis (GD). Here we describe three discrete changes in copy number that are the likely cause of the GD. Firstly, we identified a large duplication on the X chromosome that included DAX1 (NR0B1). Secondly, we identified a rearrangement that appears to affect a novel gonad-specific regulatory region in a known testis gene, SOX9. Surprisingly this patient lacked any signs of campomelic dysplasia, suggesting that the deletion affected expression of SOX9 only in the gonad. Functional analysis of potential SRY binding sites within this deleted region identified five putative enhancers, suggesting that sequences additional to the known SRY-binding TES enhancer influence human testis-specific SOX9 expression. Thirdly, we identified a small deletion immediately downstream of GATA4, supporting a role for GATA4 in gonad development in humans. These CNV analyses give new insights into the pathways involved in human gonad development and dysfunction, and suggest that rearrangements of non-coding sequences disturbing gene regulation may account for significant proportion of DSD cases.


Subject(s)
DNA Copy Number Variations/genetics , Gonadal Dysgenesis, 46,XY/genetics , Algorithms , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Chromosome Aberrations , Chromosome Deletion , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 8/genetics , Female , GATA4 Transcription Factor/genetics , Gene Expression Regulation , Gene Rearrangement/genetics , Gonads/embryology , Gonads/metabolism , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/metabolism , Humans , Male , Mice , Oligonucleotide Array Sequence Analysis , SOX9 Transcription Factor/genetics
20.
Methods Mol Biol ; 687: 193-205, 2011.
Article in English | MEDLINE | ID: mdl-20967609

ABSTRACT

Multiplex Ligation-dependent Probe Amplification (MLPA) is a PCR-based technique that was developed for identifying deletions and duplications in genomic DNA. The simplicity and sensitivity of this approach has led to it being implemented in many laboratories around the world. Since the original publication, there have been several variants of MLPA described, allowing the quantitative analysis of mRNA transcript levels, CpG methylation, complex genomic regions, and DNaseI hypersensitive sites. This chapter outlines the basic MLPA protocol, describes the different modifications and applications that have been published, and discusses the critical points during each of the steps.


Subject(s)
Polymerase Chain Reaction/methods , Base Sequence , DNA Primers
SELECTION OF CITATIONS
SEARCH DETAIL
...