Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
J Agric Food Chem ; 72(13): 7089-7099, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38512774

ABSTRACT

Breast cancer patients undergoing chemotherapy often experience muscle wasting and weakness, which impact their quality of life. A potential solution lies in customizing amino acid compositions based on exome-derived formulations (ExAAs). The study hypothesized that tailoring dietary amino acids using ExAAs could enhance muscle health. Theoretical amino acid requirements were calculated from the genome's exome region, and a breast cancer mouse model undergoing paclitaxel treatment was established. The mice were supplemented with a cancer-specific nutritional formula (QJS), and the effects of QJS and amino acid-adjusted QJS (adjQJS) were compared. Both formulations improved the nutritional status without compromising tumor growth. Notably, adjQJS significantly enhanced muscle strength compared to QJS (1.51 ± 0.25 vs. 1.30 ± 0.08 fold change, p < 0.05). Transcriptome analysis revealed alterations in complement and coagulation cascades, with an observed upregulation of C3 gene expression in adjQJS. Immune regulation also changed, showing a decrease in B cells and an increase in monocytes in skeletal muscle with adjQJS. Importantly, adjQJS resulted in a notable increase in Alistipes abundance compared to QJS (10.19 ± 0.04% vs. 5.03 ± 1.75%). This study highlights the potential of ExAAs as valuable guide for optimizing amino acid composition in diets for breast cancer patients undergoing chemotherapy.


Subject(s)
Breast Neoplasms , Exome , Humans , Animals , Mice , Female , Exome/genetics , Quality of Life , Amino Acids/metabolism , Diet , Muscle Strength , Muscle, Skeletal/metabolism , Dietary Supplements , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism
2.
Nutrients ; 14(20)2022 Oct 16.
Article in English | MEDLINE | ID: mdl-36297015

ABSTRACT

Habitual coffee consumption is an addictive behavior with unknown genetic variations and has raised public health issues about its potential health-related outcomes. We performed exome-wide association studies to identify rare risk variants contributing to habitual coffee consumption utilizing the newly released UK Biobank exome dataset (n = 200,643). A total of 34,761 qualifying variants were imported into SKAT to conduct gene-based burden and robust tests with minor allele frequency <0.01, adjusting the polygenic risk scores (PRS) of coffee intake to exclude the effect of common coffee-related polygenic risk. The gene-based burden and robust test of the exonic variants found seven exome-wide significant associations, such as OR2G2 (PSKAT = 1.88 × 10−9, PSKAT-Robust = 2.91 × 10−17), VEZT1 (PSKAT = 3.72 × 10−7, PSKAT-Robust = 1.41 × 10−7), and IRGC (PSKAT = 2.92 × 10−5, PSKAT-Robust = 1.07 × 10−7). These candidate genes were verified in the GWAS summary data of coffee intake, such as rs12737801 (p = 0.002) in OR2G2, and rs34439296 (p = 0.008) in IRGC. This study could help to extend genetic insights into the pathogenesis of coffee addiction, and may point to molecular mechanisms underlying health effects of habitual coffee consumption.


Subject(s)
Coffee , Receptors, Odorant , Humans , Exome Sequencing , Receptors, Odorant/genetics , Exome/genetics , Hyperphagia/genetics , Genome-Wide Association Study , Polymorphism, Single Nucleotide
3.
J Allergy Clin Immunol ; 150(4): 947-954, 2022 10.
Article in English | MEDLINE | ID: mdl-35753512

ABSTRACT

BACKGROUND: Prospective genetic evaluation of patients at this referral research hospital presents clinical research challenges. OBJECTIVES: This study sought not only a single-gene explanation for participants' immune-related presentations, but viewed each participant holistically, with the potential to have multiple genetic contributions to their immune phenotype and other heritable comorbidities relevant to their presentation and health. METHODS: This study developed a program integrating exome sequencing, chromosomal microarray, phenotyping, results return with genetic counseling, and reanalysis in 1505 individuals from 1000 families with suspected or known inborn errors of immunity. RESULTS: Probands were 50.8% female, 71.5% were ≥18 years, and had diverse immune presentations. Overall, 327 of 1000 probands (32.7%) received 361 molecular diagnoses. These included 17 probands with diagnostic copy number variants, 32 probands with secondary findings, and 31 probands with multiple molecular diagnoses. Reanalysis added 22 molecular diagnoses, predominantly due to new disease-gene associations (9 of 22, 40.9%). One-quarter of the molecular diagnoses (92 of 361) did not involve immune-associated genes. Molecular diagnosis was correlated with younger age, male sex, and a higher number of organ systems involved. This program also facilitated the discovery of new gene-disease associations such as SASH3-related immunodeficiency. A review of treatment options and ClinGen actionability curations suggest that at least 251 of 361 of these molecular diagnoses (69.5%) could translate into ≥1 management option. CONCLUSIONS: This program contributes to our understanding of the diagnostic and clinical utility whole exome analysis on a large scale.


Subject(s)
Exome , Genetic Testing , Exome/genetics , Female , Genetic Testing/methods , Genomics , Humans , Male , Phenotype , Prospective Studies
4.
Orphanet J Rare Dis ; 16(1): 474, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34772435

ABSTRACT

BACKGROUND: Diagnosis, treatment, and care of patients with rare diseases require multidisciplinary cooperation between medical and paramedical specialities and with patients and families. Innovative genetic diagnostics, whole exome and whole genome sequencing (WES, WGS) has enlarged the diagnostic toolkit but also increased the complexity of the endeavour. Structured multidisciplinary clinical pathways (CPW) can guide diagnosis, treatment, and care of patients with rare diseases, link scientific evidence to clinical practice and optimise clinical outcomes whilst maximising clinical efficiency. RESULTS: In contrast to the common approach of appending disease-specific CPWs to disease-specific guidelines, we suggest a generic CPW manoeuvring the patient along the way of finding the correct diagnosis by applying the best diagnostic strategy into an appropriate system of treatment and care. Available guidelines can be integrated into the generic CPW in the course of its application. The approach also applies to situations where a diagnosis remains unsolved. The backbone of the generic CPW is a set of multidisciplinary structured case conferences projecting and evaluating diagnostic and/or therapeutic steps, enforcing to integrate best scientific evidence with clinical experience. The generic CPW is stated as a flowchart and a checklist which can be used to record and document parsimoniously the structure, process and results of a patient's pathway, but also as a data model for research. It was applied in a multicentre setting with 587 cases each with a presumptive diagnosis of a rare disease. In 369 cases (62.8%) a diagnosis could be confirmed, and multidisciplinary treatment and/or care was initiated. The median process time from first contact until confirmation of diagnosis by WES was 109 days and much shorter than diagnostic delays reported in the literature. Application of the CPW is illustrated by two case reports. CONCLUSIONS: Our model is a tool to change the diagnostic odyssey into an organised and trackable route. It can also be used to inform patients and families about the stages of their individual route, to update health care providers only partially involved or attending specialised treatment and care, like the patient's or family's primary physician, and finally to train novices in the field.


Subject(s)
Critical Pathways , Rare Diseases , Exome , Humans , Rare Diseases/diagnosis , Exome Sequencing
5.
Heredity (Edinb) ; 127(3): 253-265, 2021 09.
Article in English | MEDLINE | ID: mdl-34331028

ABSTRACT

Tuber dormancy and sprouting are commercially important potato traits as long-term tuber storage is necessary to ensure year-round availability. Premature dormancy release and sprout growth in tubers during storage can result in a significant deterioration in product quality. In addition, the main chemical sprout suppressant chlorpropham has been withdrawn in Europe, necessitating alternative approaches for controlling sprouting. Breeding potato cultivars with longer dormancy and slower sprout growth is a desirable goal, although this must be tempered by the needs of the seed potato industry, where dormancy break and sprout vigour are required for rapid emergence. We have performed a detailed genetic analysis of tuber sprout growth using a diploid potato population derived from two highly heterozygous parents. A dual approach employing conventional QTL analysis allied to a combined bulk-segregant analysis (BSA) using a novel potato whole-exome capture (WEC) platform was evaluated. Tubers were assessed for sprout growth in storage at six time-points over two consecutive growing seasons. Genetic analysis revealed the presence of main QTL on five chromosomes, several of which were consistent across two growing seasons. In addition, phenotypic bulks displaying extreme sprout growth phenotypes were subjected to WEC sequencing for performing BSA. The combined BSA and WEC approach corroborated QTL locations and served to narrow the associated genomic regions, while also identifying new QTL for further investigation. Overall, our findings reveal a very complex genetic architecture for tuber sprouting and sprout growth, which has implications both for potato and other root, bulb and tuber crops where long-term storage is essential.


Subject(s)
Solanum tuberosum , Diploidy , Exome , Plant Breeding , Plant Tubers/genetics , Solanum tuberosum/genetics
6.
Sci Rep ; 10(1): 11411, 2020 07 10.
Article in English | MEDLINE | ID: mdl-32651428

ABSTRACT

Anorexia nervosa is a complex eating disorder with genetic, metabolic, and psychosocial underpinnings. Using genome-wide methods, recent studies have associated many genes with the disorder. We characterized these genes by projecting them into reference transcriptomic atlases of the prenatal and adult human brain to determine where these genes are expressed in fine detail. We found that genes from an induced stem cell study of anorexia nervosa cases are expressed at higher levels in the lateral parabrachial nucleus. Although weaker, expression enrichment of the adult lateral parabrachial is also found with genes from independent genetic studies. Candidate causal genes from the largest genetic study of anorexia nervosa to date were enriched for expression in the arcuate nucleus of the hypothalamus. We also found an enrichment of anorexia nervosa associated genes in the adult and fetal raphe and ventral tegmental areas. Motivated by enrichment of these feeding circuits, we tested if these genes respond to fasting in mice hypothalami, which highlighted the differential expression of Rps26 and Dalrd3. This work improves our understanding of the neurobiology of anorexia nervosa by suggesting disturbances in subcortical appetitive circuits.


Subject(s)
Anorexia Nervosa/genetics , Gene Expression Profiling , Transcriptome , Adult , Animals , Brain/metabolism , Exome , Female , Genetic Markers , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Hypothalamus/metabolism , Induced Pluripotent Stem Cells/cytology , Male , Mice , Microglia/metabolism , Oligonucleotide Array Sequence Analysis , Ribosomal Proteins/genetics , tRNA Methyltransferases/genetics
7.
Am J Med Genet A ; 182(9): 2129-2132, 2020 09.
Article in English | MEDLINE | ID: mdl-32627382

ABSTRACT

YY1 mutations cause Gabriele-de Vries syndrome, a recently described condition involving cognitive impairment, facial dysmorphism and intrauterine growth restriction. Movement disorders were reported in 5/10 cases of the original series, but no detailed description was provided. Here we present a 21-year-old woman with a mild intellectual deficit, facial dysmorphism and a complex movement disorder including an action tremor, cerebellar ataxia, dystonia, and partial ocular apraxia as the presenting and most striking feature. Whole-exome sequencing revealed a novel heterozygous de novo mutation in YY1 [NM: 003403.4 (YY1): c.907 T > C; p.(Cys303Arg)], classified as pathogenic according to the ACMG guidelines.


Subject(s)
Movement Disorders/genetics , Neurodevelopmental Disorders/genetics , YY1 Transcription Factor/genetics , Child , Child, Preschool , Exome/genetics , Female , Genetic Predisposition to Disease , Heterozygote , Humans , Infant , Infant, Newborn , Intellectual Disability/genetics , Intellectual Disability/pathology , Movement Disorders/pathology , Neurodevelopmental Disorders/pathology , Phenotype , Exome Sequencing
8.
Immunology ; 159(4): 373-383, 2020 04.
Article in English | MEDLINE | ID: mdl-31821535

ABSTRACT

The anti-tumor immune response is considered to be due to the T-cell receptor (TCR) binding to tumor antigens, which can be either wild-type, early stem cell proteins, presumably foreign to a developed immune system; or mutant peptides, foreign to the immune system because of a mutant amino acid (aa) or otherwise somatically altered aa sequence. Recently, very large numbers of TCR complementarity-determining region-3 (CDR3) aa sequences obtained from tumor specimens have become available. We developed a novel algorithm for assessing the complementarity of tumor mutant peptides and TCR CDR3s, based on the retrieval of TCR CDR3 aa sequences from both tumor specimen and patient blood exomes and by using an automated process of assessing CDR3 and mutant aa electrical charges. Results indicated many instances where high electrostatic complementarity was associated with a higher survival rate. In particular, our approach led to the identification of specific genes contributing significantly to the complementary, TCR CDR3-mutant aa. These results suggest a novel approach to tumor immunoscoring and may lead to the identification of high-priority neo-antigen, peptide vaccines; or to the identification of ex vivo stimulants of tumor-infiltrating lymphocytes.


Subject(s)
Algorithms , Antigens, Neoplasm/chemistry , Breast Neoplasms/genetics , Complementarity Determining Regions/chemistry , Lung Neoplasms/genetics , Receptor-CD3 Complex, Antigen, T-Cell/chemistry , Skin Neoplasms/genetics , Amino Acid Sequence , Amino Acids , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Binding Sites , Breast Neoplasms/diagnosis , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Exome , Female , Gene Expression , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Male , Mutation , Prognosis , Protein Binding , Receptor-CD3 Complex, Antigen, T-Cell/genetics , Receptor-CD3 Complex, Antigen, T-Cell/immunology , Research Design , Skin Neoplasms/diagnosis , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Static Electricity , Survival Rate , T-Lymphocytes/immunology , T-Lymphocytes/pathology
9.
JAMA Ophthalmol ; 137(12): 1444-1448, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31647501

ABSTRACT

Importance: Greater understanding of molecular features of conjunctival melanoma (CM) may improve its clinical management. Objective: To evaluate molecular features of CM and application of this information into clinical care. Design, Setting, and Participants: In a prospective case series of CM with integrative exome and transcriptome analysis, 8 patients at an academic ocular oncology setting were evaluated. The study was conducted from November 2015 to March 2018. Interventions/Exposures: Integrative exome and transcriptome analysis of CMs and clinical management of a patient's care by using this information. Main Outcomes and Measures: Molecular characterization of CM and its potential clinical application. Results: In the 8 patients (4 men) included in analysis, 4 subgroups of CM were observed, including the BRAF V600E mutation in 1 tumor, NRAS Q61R mutation in 3 tumors, NF1 mutations (Q1188X, R440X, or M1215K+ S15fs) in 3 tumors, and triple-wild type (triple-WT) in 1 tumor. The triple-WT case had CCND1 amplification and mutation in the CIC gene (Q1508X). Five tumors, including the triple-WT, also harbored mutations in MAPK genes. In addition to the genes linked to mitogen-activated protein kinase and phosphoinositol 3-kinase pathways, those involved in cell cycle and/or survival, ubiquitin-mediated protein degradation, and chromatin remodeling/epigenetic regulation (ATRX being the most frequently mutated: noted in 5 tumors) may play an important role. Other frequently mutated genes included PREX2 (n = 3), APOB (n = 4), and RYR1/2 (n = 4), although their relevance remains to be determined. The mutation burden ranged from 1.1 to 15.6 mutations per megabase (Mut/Mb) and was 3.3 Mut/Mb or less in 3 tumors and more than 10 Mut/Mb in 2 tumors. A patient with a large tumor and BRAF V600E mutation was treated with combined systemic BRAF (dabrafenib) and MEK (trametinib) inhibitors. After 3 months of therapy, her CM responded substantially and the residual tumor was removed by local surgical excision. Conclusions and Relevance: The NRAS Q61R and NF1 mutations were more common than the BRAF V600E mutation in this series. Although small tumors (where incisional biopsy is not indicated) are treated with surgical excision regardless of mutational profile, in large tumors carrying the BRAF V600E mutation, neoadjuvant therapy with combined systemic BRAF and MEK inhibitors followed by local excision may be used as an alternative to exenteration. Integrative omics analysis of CM may be informative and guide clinical management and treatment in selected cases.


Subject(s)
Antineoplastic Agents/therapeutic use , Conjunctival Neoplasms/genetics , Exome/genetics , GTP Phosphohydrolases/genetics , Melanoma/genetics , Membrane Proteins/genetics , Neurofibromin 1/genetics , Proto-Oncogene Proteins B-raf/genetics , Adult , Aged , Aged, 80 and over , Conjunctival Neoplasms/drug therapy , Conjunctival Neoplasms/pathology , DNA Mutational Analysis , Female , Gene Expression Profiling , Humans , Imidazoles/therapeutic use , Male , Melanoma/drug therapy , Melanoma/pathology , Oximes/therapeutic use , Precision Medicine , Prospective Studies , Protein Kinase Inhibitors/therapeutic use , Pyridones/therapeutic use , Pyrimidinones/therapeutic use
10.
Am J Med Genet A ; 179(4): 645-649, 2019 04.
Article in English | MEDLINE | ID: mdl-30680869

ABSTRACT

TBC1D24-related disorders are rare neurodevelopmental disorders that show a broad range of neuropsychiatric deficits and are mostly inherited in an autosomal recessive manner. Here we describe a case with early-onset epileptic encephalopathy, in whom exome sequencing detected a novel pathogenic homozygous c.442G>A, p.(Glu148Lys) variant in TBC1D24. She showed severe developmental delay, congenital sensorineural hearing loss and seizures, but the combination of a high dose phenobarbital and potassium bromide was very effective for the seizures. Sanger sequencing revealed that her mother was a heterozygous carrier of the TBC1D24 variant, but her father showed only wild-type alleles. Homozygosity mapping analysis using exome data showed loss of the heterozygosity region at 16p13.3-p13.13 encompassing TBC1D24. Genotyping analysis using rare variants within loss of the heterozygosity region indicated that the patient has a homozygous haplotype inherited from her mother, indicating maternal segmental uniparental isodisomy (UPiD). These data clearly show that exome sequencing is a powerful tool to perform comprehensive genetic analysis.


Subject(s)
GTPase-Activating Proteins/genetics , Homozygote , Mutation , Spasms, Infantile/etiology , Uniparental Disomy/pathology , Exome , Female , Humans , Infant , Prognosis , Spasms, Infantile/pathology , Uniparental Disomy/genetics
11.
Biomed Res Int ; 2018: 1250721, 2018.
Article in English | MEDLINE | ID: mdl-30584530

ABSTRACT

BACKGROUND AND AIM: ROHHADNET (rapid-onset obesity with hypoventilation, hypothalamic, autonomic dysregulation, neuroendocrine tumor) syndrome is a rare disease with grave outcome. Although early recognition is essential, prompt diagnosis may be challenging due to its extreme rarity. This study aimed to systematically review its clinical manifestation and to identify genetic causes. MATERIALS AND METHODS: We firstly conducted a systematic review on ROHHAD/NET. Electronic databases were searched using related terms. We secondly performed whole exome sequencing (WES) and examined copy number variation (CNV) in two patients to identify genetic causes. RESULTS: In total, 46 eligible studies including 158 patients were included. There were 36 case reports available for individual patient data (IPD; 48 patients, 23 ROHHAD, and 25 ROHHADNET) and 10 case series available for aggregate patient data (APD; 110 patients, 71 ROHHAD, and 39 ROHHADNET). The median age at onset calculated from IPD was 4 years. Gender information was available in 100 patients (40 from IPD and 60 from APD) in which 65 females and 35 males were showing female preponderance. Earliest manifestation was rapid obesity, followed by hypothalamic symptoms. Most common types of neuroendocrine tumors were ganglioneuromas. Patients frequently had dysnatremia and hyperprolactinemia. Two patients were available for WES. Rare variants were identified in PIK3R3, SPTBN5, and PCF11 in one patient and SRMS, ZNF83, and KMT2B in another patient, respectively. However, there was no surviving variant shared by the two patients after filtering. CONCLUSIONS: This study systematically reviewed the phenotype of ROHHAD/NET aiming to help early recognition and reducing morbidity. The link of variants identified in the present WES requires further investigation.


Subject(s)
Autonomic Nervous System Diseases/genetics , Hypothalamus/pathology , Hypoventilation/genetics , Neuroendocrine Tumors/genetics , Obesity/genetics , Age of Onset , Child , Child, Preschool , DNA Copy Number Variations/genetics , Exome/genetics , Female , Humans , Male , Meta-Analysis as Topic , Phenotype , Syndrome
12.
Toxicol Pathol ; 46(6): 706-718, 2018 08.
Article in English | MEDLINE | ID: mdl-30045675

ABSTRACT

Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide; however, the mutational properties of HCC-associated carcinogens remain largely uncharacterized. We hypothesized that mechanisms underlying chemical-induced HCC can be characterized by evaluating the mutational spectra of these tumors. To test this hypothesis, we performed exome sequencing of B6C3F1/N HCCs that arose either spontaneously in vehicle controls ( n = 3) or due to chronic exposure to gingko biloba extract (GBE; n = 4) or methyleugenol (MEG; n = 3). Most archived tumor samples are available as formalin-fixed paraffin-embedded (FFPE) blocks, rather than fresh-frozen (FF) samples; hence, exome sequencing from paired FF and FFPE samples was compared. FF and FFPE samples showed 63% to 70% mutation concordance. Multiple known (e.g., Ctnnb1T41A, BrafV637E) and novel (e.g., Erbb4C559S, Card10A700V, and Klf11P358L) mutations in cancer-related genes were identified. The overall mutational burden was greater for MEG than for GBE or spontaneous HCC samples. To characterize the mutagenic mechanisms, we analyzed the mutational spectra in the HCCs according to their trinucleotide motifs. The MEG tumors clustered closest to Catalogue of Somatic Mutations in Cancer signatures 4 and 24, which are, respectively, associated with benzo(a)pyrene- and aflatoxin-induced HCCs in humans. These results establish a novel approach for classifying liver carcinogens and understanding the mechanisms of hepatocellular carcinogenesis.


Subject(s)
Carcinogens/toxicity , Exome/genetics , Gene Expression Profiling , Liver Neoplasms, Experimental/genetics , Liver/drug effects , Mutation , Sequence Analysis, DNA/methods , Animals , Cryopreservation , DNA, Neoplasm/genetics , Eugenol/analogs & derivatives , Eugenol/toxicity , Female , Formaldehyde/chemistry , Ginkgo biloba , Liver/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/pathology , Male , Mice, Inbred Strains , Paraffin Embedding , Plant Extracts/toxicity , Reproducibility of Results , Tissue Fixation
13.
Intern Med J ; 48(7): 786-794, 2018 07.
Article in English | MEDLINE | ID: mdl-29607586

ABSTRACT

BACKGROUND: Technology has progressed from single gene panel to large-scale genomic sequencing. This is raising expectations from clinicians and patients alike. The utility and performance of this technology in a clinical setting needs to be evaluated. AIM: This pilot study investigated the feasibility of using exome-scale sequencing (ESS) to identify molecular drivers within cancers in real-time for Precision Oncology in the clinic. METHODS: Between March 2014 and March 2015, the Victorian Comprehensive Cancer Centre Alliance explored the feasibility and utility of ESS in a pilot study. DNA extracted from the tumour specimens underwent both ESS and targeted 'hotspot' sequencing (TS). Blood was taken for germline analysis. A multi-disciplinary molecular tumour board determined the clinical relevance of identified mutations; in particular, whether they were 'actionable' and/or 'druggable'. RESULTS: Of 23 patients screened, 15 (65%) met the tissue requirements for genomic analysis. TS and ESS were successful in all cases. ESS identified pathogenic somatic variants in 73% (11/15 cases) versus 53% (8/15 cases) using TS. Clinically focused ESS identified 63 variants, consisting of 30 somatic variants (including all 13 identified by TS) and 33 germline variants. Overall, there were 48 unique variants. ESS had a clinical impact in 53% (8/15 cases); 47% (7/15 cases) were referred to the familial cancer clinic, and 'druggable' targets were identified in 53% (8/15 cases). CONCLUSION: ESS of tumour DNA impacted clinical decision-making in 53%, with 20% more pathogenic variants identified through ESS than TS. The identification of germline variants in 47% was an unexpected finding.


Subject(s)
Exome/genetics , Neoplasms/genetics , Sequence Analysis, DNA , Adolescent , Adult , Aged , DNA, Neoplasm/analysis , Female , Genetic Markers , Germ-Line Mutation , High-Throughput Nucleotide Sequencing , Humans , Infant , Male , Middle Aged , Pilot Projects , Precision Medicine , Young Adult
14.
J Clin Invest ; 128(3): 1043-1056, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29431731

ABSTRACT

Congenital long QT syndrome (LQTS) is an inherited channelopathy associated with life-threatening arrhythmias. LQTS type 2 (LQT2) is caused by mutations in KCNH2, which encodes the potassium channel hERG. We hypothesized that modifier genes are partly responsible for the variable phenotype severity observed in some LQT2 families. Here, we identified contributors to variable expressivity in an LQT2 family by using induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) and whole exome sequencing in a synergistic manner. We found that iPSC-CMs recapitulated the clinical genotype-phenotype discordance in vitro. Importantly, iPSC-CMs derived from the severely affected LQT2 patients displayed prolonged action potentials compared with cells from mildly affected first-degree relatives. The iPSC-CMs derived from all patients with hERG R752W mutation displayed lower IKr amplitude. Interestingly, iPSC-CMs from severely affected mutation-positive individuals exhibited greater L-type Ca2+ current. Whole exome sequencing identified variants of KCNK17 and the GTP-binding protein REM2, providing biologically plausible explanations for this variable expressivity. Genome editing to correct a REM2 variant reversed the enhanced L-type Ca2+ current and prolonged action potential observed in iPSC-CMs from severely affected individuals. Thus, our findings showcase the power of combining complementary physiological and genomic analyses to identify genetic modifiers and potential therapeutic targets of a monogenic disorder. Furthermore, we propose that this strategy can be deployed to unravel myriad confounding pathologies displaying variable expressivity.


Subject(s)
Long QT Syndrome/genetics , Monomeric GTP-Binding Proteins/genetics , Mutation , Potassium Channels, Tandem Pore Domain/genetics , Action Potentials , Adolescent , Adult , Animals , Arrhythmias, Cardiac/metabolism , CHO Cells , Calcium/metabolism , Cricetinae , Cricetulus , Exome , Family Health , Female , Genes, Modifier , Genetic Association Studies , Genome , Genomics , Humans , Male , Middle Aged , Myocytes, Cardiac/cytology , Pedigree , Phenotype , Sequence Analysis, DNA
15.
Arch Dermatol Res ; 309(10): 773-785, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28913623

ABSTRACT

Syndromic ichthyosis is rare inherited disorders of cornification with varied disease complications. This disorder appears in seventeen subtypes associated with severe systematic manifestations along with medical, cosmetic and social problems. Syndromic ichthyosis with prominent hair abnormalities covers five major subtypes: Netherton syndrome, trichothiodystrophy, ichthyosis hypotrichosis syndrome, ichthyosis hypotrichosis sclerosing cholangitis and ichthyosis follicularis atrichia photophobia syndrome. These syndromes mostly prevail in high consanguinity states, with distinctive clinical features. The known pathogenic molecules involved in ichthyosis syndromes with prominent hair abnormalities include SPINK5, ERCC2, ERCC3, GTF2H5, MPLKIP, ST14, CLDN1 and MBTPS2. Despite underlying genetic origin, most of the health professionals solely rely on phenotypic expression of these disorders that leads to improper management of patients, hence making these patients living an orphanage life. After dermal features, association of other systems such as nervous system, skeletal system, hair abnormalities or liver problems may sometimes give clues for diagnosis but still leaving place for molecular screening for efficient diagnosis. In this paper, we have presented a review of ichthyosis syndrome with prominent hair abnormalities, with special emphasis on their updated genetic consequences and disease management. Additionally, we aim to update health professionals about the practice of molecular screening in ichthyosis syndromes for appropriate diagnosis and treatment.


Subject(s)
Hair Diseases/therapy , Hair/abnormalities , Ichthyosis/therapy , Photophobia/therapy , Rare Diseases/therapy , Consanguinity , Dermatologic Agents/therapeutic use , Exome/genetics , Genetic Testing/methods , Hair Diseases/diagnosis , Hair Diseases/etiology , Hair Diseases/mortality , High-Throughput Nucleotide Sequencing , Humans , Ichthyosis/diagnosis , Ichthyosis/etiology , Ichthyosis/mortality , Mutation , Phenotype , Photophobia/diagnosis , Photophobia/etiology , Photophobia/mortality , Phototherapy/methods , Rare Diseases/diagnosis , Rare Diseases/etiology , Rare Diseases/mortality , Syndrome
16.
Am J Chin Med ; 45(5): 1075-1092, 2017.
Article in English | MEDLINE | ID: mdl-28659030

ABSTRACT

Glomerular mesangial cells (GMCs) activation is implicated in the pathogenesis of diabetic nephropathy (DN). Our previous study revealed that high glucose (HG)-treated glomerular endothelial cells (GECs) produce an increased number of TGF-[Formula: see text]1-containing exosomes to activate GMCs through the TGF-[Formula: see text]1/Smad3 signaling pathway. We also identified that Tongxinluo (TXL), a traditional Chinese medicine, has beneficial effects on the treatment of DN in DN patients and type 2 diabetic mice. However, it remained elusive whether TXL could ameliorate renal structure and function through suppression of intercellular transfer of TGF-[Formula: see text]1-containing exosomes from GECs to GMCs. In this study, we demonstrate that TXL can inhibit the secretion of TGF-[Formula: see text]1-containing exosomes from HG-treated GECs. Furthermore, exosomes produced by HG induced-GECs treated with TXL cannot trigger GMC activation, proliferation and extracellular matrix (ECM) overproduction both in vitro and in vivo. These results suggest that TXL can prevent the transfer of TGF-[Formula: see text]1 from GECs to GMCs via exosomes, which may be one of the mechanisms of TXL in the treatment of DN.


Subject(s)
Cell Communication/drug effects , Cell Communication/genetics , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/genetics , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Endothelial Cells/metabolism , Exome/genetics , Kidney Glomerulus/cytology , Kidney/pathology , Mesangial Cells/metabolism , Phytotherapy , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Animals , Cells, Cultured , Diabetic Nephropathies/pathology , Disease Models, Animal , Fibrosis , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Signal Transduction/drug effects , Signal Transduction/genetics
17.
Oncol Rep ; 37(4): 2215-2226, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28350084

ABSTRACT

Hepatocellular carcinoma (HCC) is the most common malignancy of the liver. Genomic analysis is conducted to identify genetic alterations in driver genes which are all druggable targets for cancer therapy. In the present study, we performed an exome sequencing of 45 driver genes in 100 paired samples from HCC patients including tumors and matched adjacent normal tissues using Illumina HiSeq 2000 platform. Non-synonymous mutations were ascertained using the iPLEX MassARRAY system and Sanger sequencing. Clinicopathological relevance with genetic variations was assessed using SPSS software. The prognostic analyses of patients with gene mutation status were summarized using Kaplan-Meier curves. Sixty-one non-synonymous somatic mutations were identified in 43% of the HCC patients. The most frequent mutations were: TP53 (20%), RET (6%), PLCE1 (5%), PTEN (4%) and VEGFR2 (3%). Patients with mutations in TP53 had a lower overall survival (OS) (P=0.002) than those without mutations. Recurrent mutations in the Ret proto­oncogene (RET) were associated with poor outcomes for both disease­free survival (DFS) (P=0.028) and OS (P=0.001) in HCC patients. The mutational status of sorafenib-targeted genes were associated with decreased DFS (P=0.039), and decreased OS (P=0.15) without statistical significance. Mutual exclusion of TP53 and RET mutations were observed in the present study. In conclusion, patients with TP53 mutations, RET mutations and sorafenib-targeted gene mutations were demonstrated to be associated with poor HCC prognosis, which suggests that both TP53 and RET may serve as biomarkers of prognostic evaluation and targeted therapy in HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , High-Throughput Nucleotide Sequencing/methods , Liver Neoplasms/pathology , Mutation , Proto-Oncogene Proteins c-ret/genetics , Sequence Analysis, DNA/methods , Tumor Suppressor Protein p53/genetics , Adult , Aged , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Exome , Female , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Male , Middle Aged , Molecular Targeted Therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds , Prognosis , Sorafenib , Survival Analysis
18.
Eur J Hum Genet ; 25(6): 783-787, 2017 06.
Article in English | MEDLINE | ID: mdl-28295041

ABSTRACT

Pediatric cardiomyopathy is a rare but severe disease with high morbidity and mortality. The causes are poorly understood and can only be established in one-third of cases. Recent advances in genetic technologies, specifically next-generation sequencing, now allow for the detection of genetic causes of cardiomyopathy in a systematic and unbiased manner. This is particularly important given the large clinical variability among pediatric cardiomyopathy patients and the large number of genes (>100) implicated in the disorder. We report on the performance of whole-exome sequencing in members of a consanguineous family with a history of pediatric hypertrophic cardiomyopathy and sudden cardiac death, which led to the identification of a homozygous stop variant in the SLC22A5 gene, implicated in primary carnitine deficiency, as the likely genetic cause. Targeted carnitine tandem mass spectrometry analysis in the patient revealed complete absence of plasma-free carnitine and only trace levels of total carnitine, further supporting the causality of the SLC22A5 variant. l-carnitine supplementation in the proband led to a rapid and marked clinical improvement. This case illustrates the use of exome sequencing as a systematic and unbiased diagnostic tool in pediatric cardiomyopathy, providing an efficient route to the identification of the underlying cause, which lead to appropriate treatment and prevention of premature death.


Subject(s)
Cardiomyopathies/genetics , Carnitine/deficiency , Codon, Terminator/genetics , Organic Cation Transport Proteins/genetics , Adult , Cardiomyopathies/diagnosis , Cardiomyopathies/drug therapy , Carnitine/blood , Carnitine/therapeutic use , Death, Sudden , Exome , Female , Humans , Infant , Male , Mutation , Pedigree , Solute Carrier Family 22 Member 5
19.
Brain Dev ; 39(5): 422-425, 2017 May.
Article in English | MEDLINE | ID: mdl-28063748

ABSTRACT

BACKGROUND: Aspartylglucosaminuria (AGU) is an autosomal recessive lysosomal storage disorder caused by a deficiency of the lysosomal enzyme, aspartylglucosaminidase (AGA). This disorder is rare in the general population except in Finland. Since the most characteristic feature of this disorder is a progressive developmental regression, patients often show no specific symptoms in the initial stages, and thus early diagnosis is often challenging. CASE REPORT: We encountered a 16-year-old boy who began to show difficulties in his speech at the age of 6years. Due to a mild regression in his development, he gradually lost common daily abilities. His diagnosis was first obtained through exome sequencing that identified a novel homozygous mutation in the AGA gene. This result was reasonable because of parental consanguinity. Reduced enzymatic activity of AGA was then confirmed. His urine was retrospectively screened by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS) and a specific pattern of abnormal metabolites was identified. CONCLUSIONS: Because both exome sequencing and MALDI-TOF-MS screening are adaptable and comprehensive, future combinatory use of these methods would be useful for diagnosis of rare inborn errors of metabolism such as AGU.


Subject(s)
Aspartylglucosaminuria/genetics , Aspartylglucosylaminase/genetics , Mutation/genetics , Adolescent , Aspartylglucosaminuria/diagnostic imaging , Aspartylglucosylaminase/metabolism , Exome/genetics , Humans , Japan , Magnetic Resonance Imaging , Male , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Thalamus/diagnostic imaging
20.
Brain Dev ; 39(5): 426-430, 2017 May.
Article in English | MEDLINE | ID: mdl-28063749

ABSTRACT

Symmetrical thalamic calcification or bilateral symmetrical thalamic gliosis presents at delivery with hypertonia, fixed flexion contractures and prominent bulbar signs, without preceding perinatal asphyxia. At post-mortem, there is evidence of bilateral symmetrical selective thalamic neuronal encrustation and gliosis. To date, 27 cases are published with no underlying diagnosis identified. Two affected children from singleton pregnancies were reported and therefore, a genetic cause proposed. No previous reports have performed genetic testing to confirm or reject this hypothesis. We report three additional cases of this rare condition, expanding the clinical and pathological phenotype. We performed trio whole exome sequencing, the first in this cohort of patients, and did not identify a pathogenic variant. As postulated in the original report, the likely underlying mechanism is antenatal hypoxia in the third trimester.


Subject(s)
Brain Diseases/complications , Brain Diseases/pathology , Calcinosis/etiology , Thalamus/pathology , Brain Diseases/diagnostic imaging , Brain Diseases/genetics , Calcinosis/diagnostic imaging , Calcinosis/genetics , Exome/genetics , Female , Genetic Testing , Humans , Infant , Magnetic Resonance Imaging , Male , Thalamus/diagnostic imaging
SELECTION OF CITATIONS
SEARCH DETAIL