Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Pediatr Dermatol ; 35(2): 188-197, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29334134

ABSTRACT

BACKGROUND: Historically, diagnosis of epidermolysis bullosa has required skin biopsies for electron microscopy, direct immunofluorescence to determine which gene(s) to choose for genetic testing, or both. METHODS: To avoid these invasive tests, we developed a high-throughput next-generation sequencing (NGS)-based diagnostic assay called EBSEQ that allows simultaneous detection of mutations in 21 genes with known roles in epidermolysis bullosa pathogenicity. Mutations are confirmed with traditional Sanger sequencing. RESULTS: We present our EBSEQ assay and preliminary studies on the first 43 subjects tested. We identified 11 cases of epidermolysis bullosa simplex, five cases of junctional epidermolysis bullosa, 11 cases of dominant dystrophic epidermolysis bullosa, 15 cases of recessive dystrophic epidermolysis bullosa, and one case that remains without diagnosis. We also found an additional 52 variants of uncertain clinical significance in 17 of the 21 epidermolysis bullosa-associated genes tested. Three of the variants of uncertain clinical significance were also found in three other patients, for a total of 49 unique variants of uncertain clinical significance. We found the clinical sensitivity of the assay to be 75% to 98% and the analytical sensitivity to be 99% in identifying base substitutions and small deletions and duplications. Turnaround time was 3 to 6 weeks. CONCLUSIONS: EBSEQ is a sensitive, relatively rapid, minimally invasive, comprehensive genetic assay for the diagnosis of epidermolysis bullosa.


Subject(s)
Epidermolysis Bullosa/genetics , High-Throughput Nucleotide Sequencing/methods , Adolescent , Adult , Aged , Child , Child, Preschool , Epidermolysis Bullosa/diagnosis , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Mutation , Ohio , Skin/pathology , Young Adult
2.
Pharmacogenomics ; 18(13): 1199-1213, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28745549

ABSTRACT

AIM: To determine parents' use of their children's CYP2D6 research result. We hypothesized that perceived utility, likelihood of sharing and actual sharing of results would differ between parents with children previously exposed (cases) or unexposed (controls) to opioids. METHODS: We returned results by phone (baseline). We surveyed parents about perceived utility and likelihood of sharing their child's research result at baseline, and actual sharing at 3 and 12 months. RESULTS: Cases were more likely than controls to agree that they (p = 0.022) and the doctors (p = 0.041) could use the results to care for their child, to report higher likelihood of sharing (p = 0.042) and to actually share results with the child's doctor (p = 0.026). CONCLUSION: Prior opioid exposure influenced perceived clinical utility and sharing behaviors.


Subject(s)
Analgesics, Opioid/adverse effects , Cytochrome P-450 CYP2D6/genetics , Adolescent , Adult , Child , Female , Humans , Male , Middle Aged , Parents , Perception/physiology , Surveys and Questionnaires
3.
J Infect Dis ; 213(7): 1180-8, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26597256

ABSTRACT

BACKGROUND: Severe H1N1 influenza can be lethal in otherwise healthy individuals and can have features of reactive hemophagocytic lymphohistiocytosis (HLH). HLH is associated with mutations in lymphocyte cytolytic pathway genes, which have not been previously explored in H1N1 influenza. METHODS: Sixteen cases of fatal influenza A(H1N1) infection, 81% with histopathologic hemophagocytosis, were identified and analyzed for clinical and laboratory features of HLH, using modified HLH-2004 and macrophage activation syndrome (MAS) criteria. Fourteen specimens were subject to whole-exome sequencing. Sequence alignment and variant filtering detected HLH gene mutations and potential disease-causing variants. Cytolytic function of the PRF1 p.A91V mutation was tested in lentiviral-transduced NK-92 natural killer (NK) cells. RESULTS: Despite several lacking variables, cases of influenza A(H1N1) infection met 44% and 81% of modified HLH-2004 and MAS criteria, respectively. Five subjects (36%) carried one of 3 heterozygous LYST mutations, 2 of whom also possessed the p.A91V PRF1 mutation, which was shown to decrease NK cell cytolytic function. Several patients also carried rare variants in other genes previously observed in MAS. CONCLUSIONS: This cohort of fatal influenza A(H1N1) infections confirms the presence of hemophagocytosis and HLH pathology. Moreover, the high percentage of HLH gene mutations suggests they are risk factors for mortality among individuals with influenza A(H1N1) infection.


Subject(s)
Exome , Genetic Predisposition to Disease , Influenza A Virus, H1N1 Subtype , Influenza, Human/genetics , Lymphohistiocytosis, Hemophagocytic/genetics , Macrophage Activation Syndrome/genetics , Cohort Studies , Female , Genotype , HEK293 Cells , Humans , Influenza, Human/mortality , Killer Cells, Natural/physiology , Male , Mutation , Perforin/genetics , Perforin/metabolism , Sequence Analysis, DNA
4.
Biol Blood Marrow Transplant ; 22(3): 482-6, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26616742

ABSTRACT

Invasive fungal infections are a significant cause of morbidity and mortality in recipients of hematopoietic stem cell transplantation (HSCT), warranting antifungal prophylaxis as a standard of care in these patients. Voriconazole is commonly used in this setting because of its broad-spectrum activity and available dosage forms. There is wide well-known inter- and intrapatient variability in voriconazole concentrations, in part because concentrations are affected by common CYP2C19 polymorphisms. In 2 successive studies we have optimized voriconazole dosing to achieve target voriconazole serum concentrations using a genotype-specific dosing algorithm for antifungal prophylaxis in the post-HSCT period. In our pilot study all patients undergoing HSCT who received voriconazole antifungal prophylaxis were prospectively followed. Voriconazole concentrations were monitored weekly and doses adjusted until concentrations reached between 1 and 5.5 µg/L. The most common CYP2C19 polymorphisms were determined and correlated with voriconazole dose and time required to reach the target concentration range. In the subsequent study patients receiving voriconazole prophylaxis were dosed based on their CYP2C19 genotype and followed prospectively. In the pilot study 25 patients received voriconazole as antifungal prophylaxis for a median of 49 days (range, 15 to 196 days). The median time to reach the target concentration was 34 days for extensive metabolizers and 11 days for poor metabolizers. Three patients were genotyped as intermediate metabolizers; they reached the target concentration in a median of 56 days. Similarly, 2 patients who were genotyped as ultrarapid metabolizers reached the target range in 18 and 25 days. The time and dose required to reach the adequate concentration showed a trend toward correlation with individual CYP2C19 genotype, although voriconazole concentrations showed large interpatient variability in wild-type patients (extensive metabolizers). In our follow-up study, 20 patients received voriconazole prophylaxis prospectively dosed based on their CYP2C19 genotype. The median times to reach the target concentration using genotype-guided dosing were 9, 6.5, and 4 days for ultrarapid, extensive, and intermediate metabolizers, respectively. Overall, the median time to reach the target concentration with genotype-guided dosing was 6.5 days compared with a median time of 29 days when all patients were started on the same dose regardless of CYP2C19 genotype (P < .001). Our data show that traditional voriconazole dosing does not lead to timely achievement of target levels for fungal prophylaxis. However, a genotype-directed dosing algorithm allows patients to reach the voriconazole target range significantly sooner, providing better prophylaxis against fungal infections in the immediate post-transplant period.


Subject(s)
Algorithms , Cytochrome P-450 CYP2C19/genetics , Genotype , Hematopoietic Stem Cell Transplantation , Mycoses , Polymorphism, Genetic , Voriconazole , Adolescent , Allografts , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Male , Mycoses/genetics , Mycoses/prevention & control , Precision Medicine , Voriconazole/administration & dosage , Voriconazole/pharmacokinetics
5.
Science ; 349(6246): 436-40, 2015 Jul 24.
Article in English | MEDLINE | ID: mdl-26206937

ABSTRACT

Mutations in the LRBA gene (encoding the lipopolysaccharide-responsive and beige-like anchor protein) cause a syndrome of autoimmunity, lymphoproliferation, and humoral immune deficiency. The biological role of LRBA in immunologic disease is unknown. We found that patients with LRBA deficiency manifested a dramatic and sustained improvement in response to abatacept, a CTLA4 (cytotoxic T lymphocyte antigen-4)-immunoglobulin fusion drug. Clinical responses and homology of LRBA to proteins controlling intracellular trafficking led us to hypothesize that it regulates CTLA4, a potent inhibitory immune receptor. We found that LRBA colocalized with CTLA4 in endosomal vesicles and that LRBA deficiency or knockdown increased CTLA4 turnover, which resulted in reduced levels of CTLA4 protein in FoxP3(+) regulatory and activated conventional T cells. In LRBA-deficient cells, inhibition of lysosome degradation with chloroquine prevented CTLA4 loss. These findings elucidate a mechanism for CTLA4 trafficking and control of immune responses and suggest therapies for diseases involving the CTLA4 pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autoimmune Diseases/drug therapy , CTLA-4 Antigen/deficiency , Common Variable Immunodeficiency/drug therapy , Immunoconjugates/therapeutic use , Abatacept , Adaptor Proteins, Signal Transducing/genetics , Adolescent , Autoimmune Diseases/metabolism , CTLA-4 Antigen/genetics , Child , Chloroquine/pharmacology , Common Variable Immunodeficiency/metabolism , Endosomes/metabolism , Female , Forkhead Transcription Factors/analysis , Gene Knockdown Techniques , HEK293 Cells , Humans , Lung Diseases, Interstitial/drug therapy , Lung Diseases, Interstitial/metabolism , Lymphocyte Activation , Lysosomes/metabolism , Male , Proteolysis , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Young Adult
6.
Blood ; 124(8): 1331-4, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-24916509

ABSTRACT

Several molecules (LYST, AP3, RAB27A, STX11, STXBP2, MUNC13-4, and PRF1) have been associated with the function of cytotoxic lymphocytes. Biallelic defects in all of these molecules have been associated with familial hemophagocytic lymphohistiocytosis (FHL). We retrospectively reviewed the genetic and immunology test results from 2701 patients with a clinically suspected diagnosis of hemophagocytic lymphohistiocytosis and found 28 patients with single heterozygous mutations in 2 FHL-associated genes. Of these patients, 21 had mutations within PRF1 and a degranulation gene, and 7 were found to have mutations within 2 genes involved in the degranulation pathway. In patients with combination defects involving 2 genes in the degranulation pathway, CD107a degranulation was decreased, comparable to patients with biallelic mutations in one of the genes in the degranulation pathway. This suggests a potential digenic mode of inheritance of FHL as a result of a synergistic function effect within genes involved in cytotoxic lymphocyte degranulation.


Subject(s)
Cell Degranulation , Epistasis, Genetic , Lymphocytes/immunology , Lymphohistiocytosis, Hemophagocytic , Lysosomal-Associated Membrane Protein 1 , Models, Genetic , Mutation , Pore Forming Cytotoxic Proteins , Adolescent , Adult , Cell Degranulation/genetics , Cell Degranulation/immunology , Child , Child, Preschool , Epistasis, Genetic/genetics , Epistasis, Genetic/immunology , Female , Humans , Infant , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/immunology , Lysosomal-Associated Membrane Protein 1/genetics , Lysosomal-Associated Membrane Protein 1/immunology , Male , Perforin , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/immunology , Retrospective Studies
7.
Pediatr Blood Cancer ; 61(6): 1034-40, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24470399

ABSTRACT

BACKGROUND: The mutations in UNC13D are responsible for familial hemophagocytic lymphohistiocytosis (FHL) type 3. A 253-kb inversion and two deep intronic mutations, c.118-308C > T and c.118-307G > A, in UNC13D were recently reported in European and Asian FHL3 patients. We sought to determine the prevalence of these three non-coding mutations in North American FHL patients and evaluate the significance of examining these new mutations in genetic testing. PROCEDURE: We performed DNA sequencing of UNC13D and targeted analysis of these three mutations in 1,709 North American patients with a suspected clinical diagnosis of hemophagocytic lymphohistiocytosis (HLH). RESULTS: The 253-kb inversion, intronic mutations c.118-308C > T and c.118-307G > A were found in 11, 15, and 4 patients, respectively, in which the genetic basis (bi-allelic mutations) explained 25 additional patients. Taken together with previously diagnosed FHL3 patients in our HLH patient registry, these three non-coding mutations were found in 31.6% (25/79) of the FHL3 patients. The 253-kb inversion, c.118-308C > T and c.118-307G > A accounted for 7.0%, 8.9%, and 1.3% of mutant alleles, respectively. Significantly, eight novel mutations in UNC13D are being reported in this study. To further evaluate the expression level of the newly reported intronic mutation c.118-307G > A, reverse transcription PCR and Western blot analysis revealed a significant reduction of both RNA and protein levels suggesting that the c.118-307G > A mutation affects transcription. CONCLUSIONS: These specified non-coding mutations were found in a significant number of North American patients and inclusion of them in mutation analysis will improve the molecular diagnosis of FHL3.


Subject(s)
Lymphohistiocytosis, Hemophagocytic/genetics , Membrane Proteins/genetics , Adolescent , Adult , Black or African American/genetics , Arabs/genetics , Asian/genetics , Child , Chromosome Inversion , Consanguinity , DNA Mutational Analysis , Female , Genetic Testing , Hispanic or Latino/genetics , Humans , Infant , Infant, Newborn , Introns/genetics , Lymphohistiocytosis, Hemophagocytic/ethnology , Male , Membrane Proteins/chemistry , Membrane Proteins/physiology , North America/epidemiology , Point Mutation , Sequence Analysis, DNA , White People/genetics , Young Adult
8.
Otolaryngol Head Neck Surg ; 149(3): 478-87, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23770805

ABSTRACT

OBJECTIVE: To identify the genetic cause of prelingual sensorineural hearing loss in Pakistani families using a next-generation sequencing (NGS)-based mutation screening test named OtoSeq. STUDY DESIGN: Prospective study. SETTING: Research laboratory. SUBJECTS AND METHODS: We used 3 fluorescently labeled short tandem repeat (STR) markers for each of the known autosomal recessive nonsyndromic (DFNB) and Usher syndrome (USH) locus to perform a linkage analysis of 243 multigenerational Pakistani families segregating prelingual hearing loss. After genotyping, we focused on 34 families with potential linkage to MYO7A, CDH23, and SLC26A4. We screened affected individuals from a subset of these families using the OtoSeq platform to identify underlying genetic variants. Sanger sequencing was performed to confirm and study the segregation of mutations in other family members. For novel mutations, normal hearing individuals from ethnically matched backgrounds were also tested. RESULTS: Hearing loss was found to co-segregate with locus-specific STR markers for MYO7A in 32 families, CDH23 in 1 family, and SLC26A4 in 1 family. Using the OtoSeq platform, a microdroplet PCR-based enrichment followed by NGS, we identified mutations in 28 of the 34 families including 11 novel mutations. Sanger sequencing of these mutations showed 100% concordance with NGS data and co-segregation of the mutant alleles with the hearing loss phenotype in the respective families. CONCLUSION: Using NGS-based platforms like OtoSeq in families segregating hearing loss will contribute to the identification of common and population-specific mutations, early diagnosis, genetic counseling, and molecular epidemiology.


Subject(s)
Cadherins/genetics , Genetic Testing/methods , Hearing Loss, Sensorineural/genetics , Membrane Transport Proteins/genetics , Myosins/genetics , Alleles , Cadherin Related Proteins , Female , Genetic Linkage , Genetic Predisposition to Disease , Genotype , Humans , Male , Microsatellite Repeats , Mutation , Myosin VIIa , Pakistan , Phenotype , Polymerase Chain Reaction , Prospective Studies , Sulfate Transporters , Usher Syndromes/genetics
9.
Otolaryngol Head Neck Surg ; 148(6): 1007-16, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23525850

ABSTRACT

OBJECTIVE: To evaluate the performance of a next-generation sequencing (NGS)-based targeted resequencing genetic test, OtoSeq, to identify the sequence variants in the genes causing sensorineural hearing loss (SNHL). STUDY DESIGN: Retrospective study. SETTING: Tertiary children's hospital. SUBJECTS AND METHODS: A total of 8 individuals presenting with prelingual hearing loss were used in this study. The coding and flanking intronic regions of 24 well-studied SNHL genes were enriched using microdroplet polymerase chain reaction and sequenced on an Illumina HiSeq 2000 sequencer. The filtered high-quality sequence reads were mapped to reference sequence, and variants were detected using NextGENe software. RESULTS: A total of 1148 sequence variants were detected in 8 samples in 24 genes. Using in-house developed NGS data analysis criteria, we classified 810 (~71%) of these variants as potential true variants that include previously detected pathogenic mutations in 5 patients. To validate our strategy, we Sanger sequenced the target regions of 5 of the 24 genes, accounting for about 29.2% of all target sequence. Our results showed >99.99% concordance between NGS and Sanger sequencing in these 5 genes, resulting in an analytical sensitivity and specificity of 100% and 99.997%, respectively. We were able to successfully detect single base substitutions, small deletions, and insertions of up to 22 nucleotides. CONCLUSION: This study demonstrated that our NGS-based mutation screening strategy is highly sensitive and specific in detecting sequence variants in the SNHL genes. Therefore, we propose that this NGS-based targeted sequencing method would be an alternative to current technologies for identifying the multiple genetic causes of SNHL.


Subject(s)
Genetic Predisposition to Disease , Hearing Loss, Sensorineural/genetics , High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, DNA/methods , Adolescent , Age Factors , Child , Child, Preschool , Cohort Studies , Female , Follow-Up Studies , Genetic Testing/methods , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/epidemiology , Hospitals, Pediatric , Humans , Incidence , Infant , Male , Polymerase Chain Reaction/methods , Retrospective Studies , Risk Assessment , Sensitivity and Specificity , Sex Factors , United States
10.
Blood ; 118(22): 5794-8, 2011 Nov 24.
Article in English | MEDLINE | ID: mdl-21881043

ABSTRACT

Familial hemophagocytic lymphohistiocytosis (HLH) is a rare primary immunodeficiency disorder characterized by defects in cell-mediated cytotoxicity that results in fever, hepatosplenomegaly, and cytopenias. Familial HLH is well recognized in children but rarely diagnosed in adults. We conducted a retrospective review of genetic and immunologic test results in patients who developed HLH in adulthood. Included in our study were 1531 patients with a clinical diagnosis of HLH; 175 patients were 18 years or older. Missense and splice-site sequence variants in PRF1, MUNC13-4, and STXBP2 were found in 25 (14%) of the adult patients. The A91V-PRF1 genotype was found in 12 of these patients (48%). The preponderance of hypomorphic mutations in familial HLH-causing genes correlates with the later-onset clinical symptoms and the more indolent course in adult patients. We conclude that late-onset familial HLH occurs more commonly than was suspected previously.


Subject(s)
Lymphohistiocytosis, Hemophagocytic/genetics , Membrane Proteins/genetics , Munc18 Proteins/genetics , Mutation , Pore Forming Cytotoxic Proteins/genetics , Adolescent , Adult , Age of Onset , Aged , DNA Mutational Analysis , Female , Gene Frequency , Humans , Lymphohistiocytosis, Hemophagocytic/epidemiology , Male , Membrane Proteins/physiology , Middle Aged , Munc18 Proteins/physiology , Mutation/physiology , Perforin , Pore Forming Cytotoxic Proteins/physiology , Young Adult
11.
BMC Med Genet ; 11: 53, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20356410

ABSTRACT

BACKGROUND: Mitochondrial function is impaired in Parkinson's disease (PD) and may contribute to the pathogenesis of PD, but the causes of mitochondrial impairment in PD are unknown. Mitochondrial dysfunction is recapitulated in cell lines expressing mitochondrial DNA (mtDNA) from PD patients, implicating mtDNA variants or mutations, though the role of mtDNA variants or mutations in PD risk remains unclear. We investigated the potential contribution of mtDNA variants or mutations to the risk of PD. METHODS: We examined the possibility of a maternal inheritance bias as well as the association between mitochondrial haplogroups and maternal inheritance and disease risk in a case-control study of 168 multiplex PD families in which the proband and one parent were diagnosed with PD. 2-tailed Fisher Exact Tests and McNemar's tests were used to compare allele frequencies, and a t-test to compare ages of onset. RESULTS: The frequency of affected mothers of the proband with PD (83/167, 49.4%) was not significantly different from the frequency of affected females of the proband generation (115/259, 44.4%) (Odds Ratio 1.22; 95%CI 0.83-1.81). After correcting for multiple tests, there were no significant differences in the frequencies of mitochondrial haplogroups or of the 10398G complex I gene polymorphism in PD patients compared to controls, and no significant associations with age of onset of PD. Mitochondrial haplogroup and 10398G polymorphism frequencies were similar in probands having an affected father as compared to probands having an affected mother. CONCLUSIONS: These data fail to demonstrate a bias towards maternal inheritance in familial PD. Consistent with this, we find no association of common haplogroup-defining mtDNA variants or for the 10398G variant with the risk of PD. However, these data do not exclude a role for mtDNA variants in other populations, and it remains possible that other inherited mitochondrial DNA variants, or somatic mDNA mutations, contribute to the risk of familial PD.


Subject(s)
DNA, Mitochondrial/chemistry , Parkinson Disease/genetics , Adult , Age of Onset , Aged , Aged, 80 and over , Case-Control Studies , Family , Female , Gene Frequency , Genetic Variation , Genotype , Haplotypes , Humans , Inheritance Patterns , Male , Middle Aged , Odds Ratio , Polymorphism, Genetic , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...