Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
J Mol Diagn ; 26(9): 843-850, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38925456

ABSTRACT

PMS2 is one of the DNA-mismatch repair genes included in routine genetic testing for Lynch syndrome and colorectal, ovarian, and endometrial cancers. PMS2 is also included in the American College of Medical Genetics and Genomics' List of Secondary Findings Genes in the context of clinical exome and genome sequencing. However, sequencing of PMS2 by short-read-based next-generation sequencing technologies is complicated by the presence of the pseudogene PMS2CL, and is often supplemented by long-range-based approaches, such as long-range PCR or long-read-based next-generation sequencing, which increases the complexity and cost. This article describes a bioinformatics homology triage workflow that can eliminate the need for long-read-based testing for PMS2 in the vast majority of patients undergoing exome sequencing, thus simplifying PMS2 testing and reducing the associated cost.


Subject(s)
Exome Sequencing , Exons , High-Throughput Nucleotide Sequencing , Mismatch Repair Endonuclease PMS2 , Humans , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Computational Biology/methods , Exome/genetics , Exome Sequencing/methods , Exons/genetics , Genetic Testing/methods , Genetic Variation , High-Throughput Nucleotide Sequencing/methods , Mismatch Repair Endonuclease PMS2/genetics
2.
J Appl Lab Med ; 5(3): 467-479, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32445360

ABSTRACT

BACKGROUND: Deafness and hearing loss are common conditions that can be seen independently or as part of a syndrome and are often mediated by genetic causes. We sought to develop and validate a hereditary hearing loss panel (HHLP) to detect single nucleotide variants (SNVs), insertions and deletions (indels), and copy number variants (CNVs) in 166 genes related to nonsyndromic and syndromic hearing loss. METHODS: We developed a custom-capture next-generation sequencing (NGS) reagent to detect all coding regions, ±10 flanking bp, for the 166 genes related to nonsyndromic and syndromic hearing loss. Our validation consisted of testing 52 samples to establish accuracy, reproducibility, and analytical sensitivity. In addition to NGS, supplementary methods, including multiplex ligation-dependent probe amplification, long-range PCR, and Sanger sequencing, were used to ensure coverage of regions that had high complexity or homology. RESULTS: We observed 100% positive and negative percentage agreement for detection of SNVs (n = 362), small indels (1-22 bp, n = 25), and CNVs (gains, n = 8; losses, n = 17). Finally, we showed that this assay was able to detect variants with a variant allele frequency ≥20% for SNVs and indels and ≥30% to 35% for CNVs. CONCLUSIONS: We validated an HHLP that detects SNVs, indels, and CNVs in 166 genes related to syndromic and nonsyndromic hearing loss. The results of this assay can be utilized to confirm a diagnosis of hearing loss and related syndromic disorders associated with known causal genes.


Subject(s)
Genetic Predisposition to Disease , Genetic Testing , Hearing Loss/diagnosis , Hearing Loss/genetics , High-Throughput Nucleotide Sequencing , Alleles , Amino Acid Substitution , Chromosome Mapping , Computational Biology/methods , Genetic Association Studies , Genetic Testing/methods , Genotype , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/standards , Humans , Molecular Diagnostic Techniques , Reproducibility of Results
3.
Cell Rep ; 21(1): 195-207, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28978473

ABSTRACT

Th17 cells are potent mediators in autoimmune diseases, and RORγt is required for their development. Recent studies have shown that RORγt+ Treg cells in the gut regulate intestinal inflammation by inhibiting effector T cell function. In the current study, we report that RORγt+ Treg cells were also found in lymph nodes following immunization. Not only distinct from intestinal RORγt+ Treg cells in their transcriptomes, peripheral RORγt+ Treg cells were derived from Foxp3+ thymic Treg cells in an antigen-specific manner. Development of these RORγt+ Treg cells, coined T regulatory 17 (Tr17) cells, depended on IL-6/Stat3 signaling. Tr17 cells showed suppressive activity against antigen-specific effector T cells in vitro. In addition, Tr17 cells efficiently inhibited myelin-specific Th17-cell-mediated CNS auto-inflammation in a passive EAE model. Collectively, our study demonstrates that Tr17 cells are effector Treg cells that potentially restrict autoimmunity.


Subject(s)
Autoimmunity/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Forkhead Transcription Factors/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Adoptive Transfer , Animals , Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Forkhead Transcription Factors/immunology , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Inducible T-Cell Co-Stimulator Protein/genetics , Inducible T-Cell Co-Stimulator Protein/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Peptide Fragments/administration & dosage , Receptors, CCR6/genetics , Receptors, CCR6/immunology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Signal Transduction , T-Lymphocytes, Regulatory/pathology , T-Lymphocytes, Regulatory/transplantation , Th17 Cells/pathology
4.
Cancer Cell ; 32(1): 88-100.e6, 2017 07 10.
Article in English | MEDLINE | ID: mdl-28669490

ABSTRACT

Amplification of 1q21 occurs in approximately 30% of de novo and 70% of relapsed multiple myeloma (MM) and is correlated with disease progression and drug resistance. Here, we provide evidence that the 1q21 amplification-driven overexpression of ILF2 in MM promotes tolerance of genomic instability and drives resistance to DNA-damaging agents. Mechanistically, elevated ILF2 expression exerts resistance to genotoxic agents by modulating YB-1 nuclear localization and interaction with the splicing factor U2AF65, which promotes mRNA processing and the stabilization of transcripts involved in homologous recombination in response to DNA damage. The intimate link between 1q21-amplified ILF2 and the regulation of RNA splicing of DNA repair genes may be exploited to optimize the use of DNA-damaging agents in patients with high-risk MM.


Subject(s)
Multiple Myeloma/genetics , Nuclear Factor 45 Protein/physiology , RNA Splicing/genetics , DNA Damage , DNA Repair , Homologous Recombination , Humans , Nuclear Factor 45 Protein/genetics , Nuclear Factor 45 Protein/metabolism , Splicing Factor U2AF/metabolism , Tumor Cells, Cultured , Y-Box-Binding Protein 1/metabolism
5.
PLoS One ; 11(4): e0153933, 2016.
Article in English | MEDLINE | ID: mdl-27100181

ABSTRACT

Mutational processes and signatures that drive early tumorigenesis are centrally important for early cancer prevention. Yet, to date, biomarkers and risk factors for polyps (adenomas) that inordinately and rapidly develop into colon cancer remain poorly defined. Here, we describe surprisingly high mutational profiles through whole-genome sequence (WGS) analysis in 2 of 4 pairs of benign colorectal adenoma tissue samples. Unsupervised hierarchical clustered transcriptomic analysis of a further 7 pairs of adenomas reveals distinct mutational signatures regardless of adenoma size. Transitional single nucleotide substitutions of C:G>T:A predominate in the adenoma mutational spectrum. Strikingly, we observe mutations in the TGF-ß pathway and CEA-associated genes in 4 out of 11 adenomas, overlapping with the Wnt pathway. Immunohistochemical labeling reveals a nearly 5-fold increase in CEA levels in 23% of adenoma samples with a concomitant loss of TGF-ß signaling. We also define a functional role by which the CEA B3 domain interacts with TGFBR1, potentially inactivating the tumor suppressor function of TGF-ß signaling. Our study uncovers diverse mutational processes underlying the transition from early adenoma to cancer. This has broad implications for biomarker-driven targeting of CEA/TGF-ß in high-risk adenomas and may lead to early detection of aggressive adenoma to CRC progression.


Subject(s)
Adenoma/genetics , Carcinoembryonic Antigen/genetics , Colon/metabolism , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Mutation/genetics , Transforming Growth Factor beta/genetics , Adenoma/metabolism , Adenoma/pathology , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoembryonic Antigen/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Colon/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Progression , High-Throughput Nucleotide Sequencing/methods , Humans , Immunoenzyme Techniques , Immunoprecipitation , Signal Transduction , Transforming Growth Factor beta/metabolism
6.
J Clin Invest ; 126(2): 527-42, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26784546

ABSTRACT

Beckwith-Wiedemann syndrome (BWS) is a human stem cell disorder, and individuals with this disease have a substantially increased risk (~800-fold) of developing tumors. Epigenetic silencing of ß2-spectrin (ß2SP, encoded by SPTBN1), a SMAD adaptor for TGF-ß signaling, is causally associated with BWS; however, a role of TGF-ß deficiency in BWS-associated neoplastic transformation is unexplored. Here, we have reported that double-heterozygous Sptbn1+/- Smad3+/- mice, which have defective TGF-ß signaling, develop multiple tumors that are phenotypically similar to those of BWS patients. Moreover, tumorigenesis-associated genes IGF2 and telomerase reverse transcriptase (TERT) were overexpressed in fibroblasts from BWS patients and TGF-ß-defective mice. We further determined that chromatin insulator CCCTC-binding factor (CTCF) is TGF-ß inducible and facilitates TGF-ß-mediated repression of TERT transcription via interactions with ß2SP and SMAD3. This regulation was abrogated in TGF-ß-defective mice and BWS, resulting in TERT overexpression. Imprinting of the IGF2/H19 locus and the CDKN1C/KCNQ1 locus on chromosome 11p15.5 is mediated by CTCF, and this regulation is lost in BWS, leading to aberrant overexpression of growth-promoting genes. Therefore, we propose that loss of CTCF-dependent imprinting of tumor-promoting genes, such as IGF2 and TERT, results from a defective TGF-ß pathway and is responsible at least in part for BWS-associated tumorigenesis as well as sporadic human cancers that are frequently associated with SPTBN1 and SMAD3 mutations.


Subject(s)
Beckwith-Wiedemann Syndrome/metabolism , Carrier Proteins/metabolism , Microfilament Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Repressor Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Beckwith-Wiedemann Syndrome/genetics , CCCTC-Binding Factor , Carrier Proteins/genetics , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 11/metabolism , Cyclin-Dependent Kinase Inhibitor p57/genetics , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Hep G2 Cells , Humans , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , KCNQ1 Potassium Channel/genetics , KCNQ1 Potassium Channel/metabolism , Mice , Mice, Knockout , Microfilament Proteins/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Repressor Proteins/genetics , Signal Transduction/genetics , Smad3 Protein/genetics , Smad3 Protein/metabolism , Telomerase/biosynthesis , Telomerase/genetics , Telomerase/metabolism , Transforming Growth Factor beta/genetics
7.
Breast Cancer Res ; 18(1): 13, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26818199

ABSTRACT

BACKGROUND: Despite advances in early diagnosis and treatment of cancer patients, metastasis remains the major cause of mortality. TP53 is one of the most frequently mutated genes in human cancer, and these alterations can occur during the early stages of oncogenesis or as later events as tumors progress to more aggressive forms. Previous studies have suggested that p53 plays a role in cellular pathways that govern metastasis. To investigate how p53 deficiency contributes to late-stage tumor growth and metastasis, we developed paired isogenic patient-derived xenograft (PDX) models of triple-negative breast cancer (TNBC) differing only in p53 status for longitudinal analysis. METHODS: Patient-derived isogenic human tumor lines differing only in p53 status were implanted into mouse mammary glands. Tumor growth and metastasis were monitored with bioluminescence imaging, and circulating tumor cells (CTCs) were quantified by flow cytometry. RNA-Seq was performed on p53-deficient and p53 wild-type tumors, and functional validation of a lead candidate gene was performed in vivo. RESULTS: Isogenic p53 wild-type and p53-deficient tumors metastasized out of mammary glands and colonized distant sites with similar frequency. However, p53-deficient tumors metastasized earlier than p53 wild-type tumors and grew faster in both primary and metastatic sites as a result of increased proliferation and decreased apoptosis. In addition, greater numbers of CTCs were detected in the blood of mice engrafted with p53-deficient tumors. However, when normalized to tumor mass, the number of CTCs isolated from mice bearing parental and p53-deficient tumors was not significantly different. Gene expression profiling followed by functional validation identified B cell translocation gene 2 (BTG2), a downstream effector of p53, as a negative regulator of tumor growth both at primary and metastatic sites. BTG2 expression status correlated with survival of TNBC patients. CONCLUSIONS: Using paired isogenic PDX-derived metastatic TNBC cells, loss of p53 promoted tumor growth and consequently increased tumor cell shedding into the blood, thus enhancing metastasis. Loss of BTG2 expression in p53-deficient tumors contributed to this metastatic potential by enhancing tumor growth in primary and metastatic sites. Furthermore, clinical data support conclusions generated from PDX models and indicate that BTG2 expression is a candidate prognostic biomarker for TNBC.


Subject(s)
Cell Proliferation/genetics , Immediate-Early Proteins/biosynthesis , Triple Negative Breast Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/biosynthesis , Animals , Apoptosis/genetics , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Immediate-Early Proteins/genetics , Mice , Neoplasm Metastasis , Neoplastic Cells, Circulating/pathology , Triple Negative Breast Neoplasms/pathology , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays
8.
Genome Announc ; 3(3)2015 May 21.
Article in English | MEDLINE | ID: mdl-25999555

ABSTRACT

Mycobacterium lepromatosis is a newly discovered cause of leprosy. Here, we present a near-complete genome of M. lepromatosis from strain FJ924 obtained from a patient who died of leprosy. The genome contained 3,215,823 nucleotides and matched ~87% with the Mycobacterium leprae genome. This genome is likely the smallest of all mycobacterial genomes known to date.

9.
Cancer Cell ; 27(5): 644-57, 2015 May 11.
Article in English | MEDLINE | ID: mdl-25965571

ABSTRACT

Myelodysplastic syndrome (MDS) risk correlates with advancing age, therapy-induced DNA damage, and/or shorter telomeres, but whether telomere erosion directly induces MDS is unknown. Here, we provide the genetic evidence that telomere dysfunction-induced DNA damage drives classical MDS phenotypes and alters common myeloid progenitor (CMP) differentiation by repressing the expression of mRNA splicing/processing genes, including SRSF2. RNA-seq analyses of telomere dysfunctional CMP identified aberrantly spliced transcripts linked to pathways relevant to MDS pathogenesis such as genome stability, DNA repair, chromatin remodeling, and histone modification, which are also enriched in mouse CMP haploinsufficient for SRSF2 and in CD34(+) CMML patient cells harboring SRSF2 mutation. Together, our studies establish an intimate link across telomere biology, aberrant RNA splicing, and myeloid progenitor differentiation.


Subject(s)
Cell Differentiation/genetics , Hematopoiesis/genetics , Myelodysplastic Syndromes/genetics , Telomere , Animals , Haploinsufficiency , Humans , Mice , Myelodysplastic Syndromes/pathology , Nuclear Proteins/genetics , RNA Splicing , Ribonucleoproteins/genetics , Serine-Arginine Splicing Factors
10.
Neuron ; 80(2): 415-28, 2013 Oct 16.
Article in English | MEDLINE | ID: mdl-24139042

ABSTRACT

A hexanucleotide GGGGCC repeat expansion in the noncoding region of the C9ORF72 gene is the most common genetic abnormality in familial and sporadic amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). The function of the C9ORF72 protein is unknown, as is the mechanism by which the repeat expansion could cause disease. Induced pluripotent stem cell (iPSC)-differentiated neurons from C9ORF72 ALS patients revealed disease-specific (1) intranuclear GGGGCCexp RNA foci, (2) dysregulated gene expression, (3) sequestration of GGGGCCexp RNA binding protein ADARB2, and (4) susceptibility to excitotoxicity. These pathological and pathogenic characteristics were confirmed in ALS brain and were mitigated with antisense oligonucleotide (ASO) therapeutics to the C9ORF72 transcript or repeat expansion despite the presence of repeat-associated non-ATG translation (RAN) products. These data indicate a toxic RNA gain-of-function mechanism as a cause of C9ORF72 ALS and provide candidate antisense therapeutics and candidate human pharmacodynamic markers for therapy.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Frontotemporal Dementia/metabolism , Oligonucleotides, Antisense/therapeutic use , Proteins/metabolism , RNA/toxicity , Adenosine Deaminase/metabolism , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , C9orf72 Protein , Cell Count , Dose-Response Relationship, Drug , Frontotemporal Dementia/drug therapy , Frontotemporal Dementia/genetics , Glutamic Acid/toxicity , Humans , Induced Pluripotent Stem Cells , Neurons/drug effects , Neurons/metabolism , Oligonucleotides, Antisense/pharmacology , Proteins/genetics , RNA/genetics , RNA/metabolism , RNA-Binding Proteins , Repetitive Sequences, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL