Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Allergy Clin Immunol Pract ; 9(2): 803-818.e11, 2021 02.
Article in English | MEDLINE | ID: mdl-33217613

ABSTRACT

BACKGROUND: Gain-of-function mutations in STING1 underlie a type I interferonopathy termed SAVI (STING-associated vasculopathy with onset in infancy). This severe disease is variably characterized by early-onset systemic inflammation, skin vasculopathy, and interstitial lung disease (ILD). OBJECTIVE: To describe a cohort of patients with SAVI. METHODS: Assessment of clinical, radiological and immunological data from 21 patients (17 families) was carried out. RESULTS: Patients carried heterozygous substitutions in STING1 previously described in SAVI, mainly the p.V155M. Most were symptomatic from infancy, but late onset in adulthood occurred in 1 patient. Systemic inflammation, skin vasculopathy, and ILD were observed in 19, 18, and 21 patients, respectively. Extensive tissue loss occurred in 4 patients. Severity of ILD was highly variable with insidious progression up to end-stage respiratory failure reached at teenage in 6 patients. Lung imaging revealed early fibrotic lesions. Failure to thrive was almost constant, with severe growth failure seen in 4 patients. Seven patients presented polyarthritis, and the phenotype in 1 infant mimicked a combined immunodeficiency. Extended features reminiscent of other interferonopathies were also found, including intracranial calcification, glaucoma and glomerular nephropathy. Increased expression of interferon-stimulated genes and interferon α protein was constant. Autoantibodies were frequently found, in particular rheumatoid factor. Most patients presented with a T-cell defect, with low counts of memory CD8+ cells and impaired T-cell proliferation in response to antigens. Long-term follow-up described in 8 children confirmed the clinical benefit of ruxolitinib in SAVI where the treatment was started early in the disease course, underlying the need for early diagnosis. Tolerance was reasonably good. CONCLUSION: The largest worldwide cohort of SAVI patients yet described, illustrates the core features of the disease and extends the clinical and immunological phenotype to include overlap with other monogenic interferonopathies.


Subject(s)
Lung Diseases, Interstitial , Membrane Proteins/genetics , Vascular Diseases , Adolescent , Adult , Child , Humans , Infant , Inflammation , Mutation
2.
Rheumatology (Oxford) ; 59(8): 1927-1937, 2020 08 01.
Article in English | MEDLINE | ID: mdl-31755959

ABSTRACT

OBJECTIVES: JDM and juvenile overlap myositis represent heterogeneous subtypes of juvenile idiopathic inflammatory myopathy (JIIM). Chronic evolution can occur in up to 60% of cases, and morbidity/mortality is substantial. We aimed to describe the clinical, biological, histological and type I IFN status in JIIM associated with anti-melanoma differentiation-associated protein 5 (anti-MDA5) autoantibodies at presentation (group 1) in comparison with other JIIM (group 2). METHODS: This was a retrospective and prospective study of patients with JIIM ascertained from three French paediatric rheumatology reference centres between 2013 and 2019. Muscle biopsies were reviewed. Type I interferon pathway activity was assessed by dosage of IFNα serum protein and the expression of IFN-stimulated genes. RESULTS: Sixty-four patients were included, 13 in group 1 (54% JDM and 46% juvenile overlap myositis) and 51 in group 2 (76% JDM and 24% juvenile overlap myositis). Group 1 patients demonstrated more arthritis, skin ulcerations, lupus features and interstitial lung disease, and a milder muscular involvement. Serum IFNα levels were higher in group 1 than 2, and decreased after treatment or improvement in both groups. Outcome was similar in both groups. Unconventional treatment (more than two lines) was required in order to achieve remission, especially when skin ulceration was reported. CONCLUSION: This study indicates a higher frequency of arthritis, skin ulcerations and interstitial lung disease, but milder muscular involvement, in JIIM with positive anti-MDA5 autoantibodies compared with other JIIM. Our data support an important role of systemic IFNα in disease pathology, particularly in the anti-MDA5 auto-antibody-positive subgroup. In severe and refractory forms of JIIM, IFNα may represent a therapeutic target.


Subject(s)
Autoantibodies/immunology , Interferon-Induced Helicase, IFIH1/immunology , Interferon-alpha/metabolism , Muscle, Skeletal/metabolism , Myositis/metabolism , Signal Transduction/physiology , Adolescent , Child , Child, Preschool , Female , Humans , Male , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Myositis/immunology , Myositis/pathology , Prospective Studies , Retrospective Studies
4.
J Exp Med ; 215(10): 2567-2585, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30143481

ABSTRACT

Life-threatening pulmonary influenza can be caused by inborn errors of type I and III IFN immunity. We report a 5-yr-old child with severe pulmonary influenza at 2 yr. She is homozygous for a loss-of-function IRF9 allele. Her cells activate gamma-activated factor (GAF) STAT1 homodimers but not IFN-stimulated gene factor 3 (ISGF3) trimers (STAT1/STAT2/IRF9) in response to IFN-α2b. The transcriptome induced by IFN-α2b in the patient's cells is much narrower than that of control cells; however, induction of a subset of IFN-stimulated gene transcripts remains detectable. In vitro, the patient's cells do not control three respiratory viruses, influenza A virus (IAV), parainfluenza virus (PIV), and respiratory syncytial virus (RSV). These phenotypes are rescued by wild-type IRF9, whereas silencing IRF9 expression in control cells increases viral replication. However, the child has controlled various common viruses in vivo, including respiratory viruses other than IAV. Our findings show that human IRF9- and ISGF3-dependent type I and III IFN responsive pathways are essential for controlling IAV.


Subject(s)
Alleles , Homozygote , Influenza, Human , Interferon-Stimulated Gene Factor 3, gamma Subunit/deficiency , Orthomyxoviridae/immunology , Pneumonia, Viral , Female , Humans , Infant , Influenza, Human/genetics , Influenza, Human/immunology , Influenza, Human/pathology , Interferon alpha-2/genetics , Interferon alpha-2/immunology , Interferon-Stimulated Gene Factor 3, gamma Subunit/immunology , Pneumonia, Viral/genetics , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology
5.
Nat Commun ; 8(1): 2176, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29259162

ABSTRACT

Microbial nucleic acid recognition serves as the major stimulus to an antiviral response, implying a requirement to limit the misrepresentation of self nucleic acids as non-self and the induction of autoinflammation. By systematic screening using a panel of interferon-stimulated genes we identify two siblings and a singleton variably demonstrating severe neonatal anemia, membranoproliferative glomerulonephritis, liver fibrosis, deforming arthropathy and increased anti-DNA antibodies. In both families we identify biallelic mutations in DNASE2, associated with a loss of DNase II endonuclease activity. We record increased interferon alpha protein levels using digital ELISA, enhanced interferon signaling by RNA-Seq analysis and constitutive upregulation of phosphorylated STAT1 and STAT3 in patient lymphocytes and monocytes. A hematological disease transcriptomic signature and increased numbers of erythroblasts are recorded in patient peripheral blood, suggesting that interferon might have a particular effect on hematopoiesis. These data define a type I interferonopathy due to DNase II deficiency in humans.


Subject(s)
Deoxyribonucleases/deficiency , Endodeoxyribonucleases/deficiency , Hereditary Autoinflammatory Diseases/enzymology , Interferon-alpha/immunology , Signal Transduction/immunology , Adolescent , Antiviral Agents/pharmacology , Child , Deoxyribonucleases/genetics , Deoxyribonucleases/immunology , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/immunology , Erythroblasts/immunology , Female , Gene Expression Profiling , Hematopoiesis/immunology , Hereditary Autoinflammatory Diseases/blood , Hereditary Autoinflammatory Diseases/genetics , Hereditary Autoinflammatory Diseases/immunology , Humans , Interferon-alpha/blood , Interferon-alpha/metabolism , Male , Mutation , Phosphorylation , RNA, Messenger/analysis , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Sequence Analysis, RNA , Up-Regulation/drug effects
6.
Arthritis Rheumatol ; 69(10): 2081-2091, 2017 10.
Article in English | MEDLINE | ID: mdl-28605144

ABSTRACT

OBJECTIVE: To define the molecular basis of a multisystem phenotype with progressive musculoskeletal disease of the hands and feet, including camptodactyly, subluxation, and tendon rupture, reminiscent of Jaccoud's arthropathy. METHODS: We identified 2 families segregating an autosomal-dominant phenotype encompassing musculoskeletal disease and variable additional features, including psoriasis, dental abnormalities, cardiac valve involvement, glaucoma, and basal ganglia calcification. We measured the expression of interferon (IFN)-stimulated genes in the peripheral blood and skin, and undertook targeted Sanger sequencing of the IFIH1 gene encoding the cytosolic double-stranded RNA (dsRNA) sensor melanoma differentiation-associated protein 5 (MDA-5). We also assessed the functional consequences of IFIH1 gene variants using an in vitro IFNß reporter assay in HEK 293T cells. RESULTS: We recorded an up-regulation of type I IFN-induced gene transcripts in all 5 patients tested and identified a heterozygous gain-of-function mutation in IFIH1 in each family, resulting in different substitutions of the threonine residue at position 331 of MDA-5. Both of these variants were associated with increased IFNß expression in the absence of exogenous dsRNA ligand, consistent with constitutive activation of MDA-5. CONCLUSION: These cases highlight the significant musculoskeletal involvement that can be associated with mutations in MDA-5, and emphasize the value of testing for up-regulation of IFN signaling as a marker of the underlying molecular lesion. Our data indicate that both Singleton-Merten syndrome and neuroinflammation described in the context of MDA-5 gain-of-function constitute part of the same type I interferonopathy disease spectrum, and provide possible novel insight into the pathology of Jaccoud's arthropathy.


Subject(s)
Aortic Diseases/genetics , Basal Ganglia Diseases/genetics , Calcinosis/genetics , Dental Enamel Hypoplasia/genetics , Glaucoma/genetics , Heart Valve Diseases/genetics , Interferon-Induced Helicase, IFIH1/genetics , Metacarpus/abnormalities , Muscular Diseases/genetics , Musculoskeletal Diseases/genetics , Odontodysplasia/genetics , Osteoporosis/genetics , Psoriasis/genetics , Vascular Calcification/genetics , Adolescent , Adult , Child , HEK293 Cells , Heterozygote , Humans , Middle Aged , Mutation , Syndrome
7.
Neuropediatrics ; 48(3): 166-184, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28561207

ABSTRACT

We investigated the genetic, phenotypic, and interferon status of 46 patients from 37 families with neurological disease due to mutations in ADAR1. The clinicoradiological phenotype encompassed a spectrum of Aicardi-Goutières syndrome, isolated bilateral striatal necrosis, spastic paraparesis with normal neuroimaging, a progressive spastic dystonic motor disorder, and adult-onset psychological difficulties with intracranial calcification. Homozygous missense mutations were recorded in five families. We observed a p.Pro193Ala variant in the heterozygous state in 22 of 23 families with compound heterozygous mutations. We also ascertained 11 cases from nine families with a p.Gly1007Arg dominant-negative mutation, which occurred de novo in four patients, and was inherited in three families in association with marked phenotypic variability. In 50 of 52 samples from 34 patients, we identified a marked upregulation of type I interferon-stimulated gene transcripts in peripheral blood, with a median interferon score of 16.99 (interquartile range [IQR]: 10.64-25.71) compared with controls (median: 0.93, IQR: 0.57-1.30). Thus, mutations in ADAR1 are associated with a variety of clinically distinct neurological phenotypes presenting from early infancy to adulthood, inherited either as an autosomal recessive or dominant trait. Testing for an interferon signature in blood represents a useful biomarker in this context.


Subject(s)
Adenosine Deaminase/genetics , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/immunology , Interferon Type I/metabolism , Nervous System Malformations/genetics , Nervous System Malformations/immunology , RNA-Binding Proteins/genetics , Adolescent , Adult , Autoimmune Diseases of the Nervous System/diagnostic imaging , Biomarkers/metabolism , Child , Child, Preschool , Female , Humans , Infant , Male , Mutation , Nervous System Malformations/diagnostic imaging , Phenotype , Young Adult
8.
Arthritis Rheumatol ; 69(7): 1495-1501, 2017 07.
Article in English | MEDLINE | ID: mdl-28426911

ABSTRACT

OBJECTIVE: Gain-of-function mutations in TMEM173, encoding the stimulator of interferon (IFN) genes (STING) protein, underlie a novel type I interferonopathy that is minimally responsive to conventional immunosuppressive therapies and associated with high frequency of childhood morbidity and mortality. STING gain-of-function causes constitutive oversecretion of IFN. This study was undertaken to determine the effects of a TANK-binding kinase 1 (TBK-1)/IKKɛ inhibitor (BX795) on secretion and signaling of IFN in primary peripheral blood mononuclear cells (PBMCs) from patients with mutations in STING. METHODS: PBMCs from 4 patients with STING-associated disease were treated with BX795. The effect of BX795 on IFN pathways was assessed by Western blotting and an IFNß reporter assay, as well as by quantification of IFNα in cell lysates, staining for STAT-1 phosphorylation, and measurement of IFN-stimulated gene (ISG) messenger RNA (mRNA) expression. RESULTS: Treatment of PBMCs with BX795 inhibited the phosphorylation of IFN regulatory factor 3 and IFNß promoter activity induced in HEK 293T cells by cyclic GMP-AMP or by genetic activation of STING. In vitro exposure to BX795 inhibited IFNα production in PBMCs of patients with STING-associated disease without affecting cell survival. In addition, BX795 decreased STAT-1 phosphorylation and ISG mRNA expression independent of IFNα blockade. CONCLUSION: These findings demonstrate the effect of BX795 on reducing type I IFN production and IFN signaling in cells from patients with gain-of-function mutations in STING. A combined inhibition of TBK-1 and IKKɛ therefore holds potential for the treatment of patients carrying STING mutations, and may also be relevant in other type I interferonopathies.


Subject(s)
Interferon Regulatory Factor-3/drug effects , Interferon-Stimulated Gene Factor 3, gamma Subunit/drug effects , Interferon-alpha/drug effects , Interferon-beta/drug effects , Leukocytes, Mononuclear/drug effects , Membrane Proteins/drug effects , Pyrimidines/pharmacology , Thiophenes/pharmacology , Blotting, Western , Child , HEK293 Cells , Humans , I-kappa B Kinase/antagonists & inhibitors , In Vitro Techniques , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factors/drug effects , Interferon Regulatory Factors/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interferon-alpha/immunology , Interferon-beta/immunology , Leukocytes, Mononuclear/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mutation , Nucleotides, Cyclic/pharmacology , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/drug effects , STAT1 Transcription Factor/metabolism
9.
J Allergy Clin Immunol ; 140(2): 543-552.e5, 2017 08.
Article in English | MEDLINE | ID: mdl-28087229

ABSTRACT

BACKGROUND: Gain-of-function mutations in transmembrane protein 173 (TMEM173) encoding stimulator of interferon genes (STING) underlie a recently described type I interferonopathy called STING-associated vasculopathy with onset in infancy (SAVI). OBJECTIVES: We sought to define the molecular and cellular pathology relating to 3 individuals variably exhibiting the core features of the SAVI phenotype including systemic inflammation, destructive skin lesions, and interstitial lung disease. METHODS: Genetic analysis, conformational studies, in vitro assays and ex vivo flow-cytometry were performed. RESULTS: Molecular and in vitro data demonstrate that the pathology in these patients is due to amino acid substitutions at positions 206, 281, and 284 of the human STING protein. These mutations confer cGAMP-independent constitutive activation of type I interferon signaling through TBK1 (TANK-binding kinase), independent from the alternative STING pathway triggered by membrane fusion of enveloped RNA viruses. This constitutive activation was abrogated by ex vivo treatment with the janus kinase 1/2 inhibitor ruxolitinib. CONCLUSIONS: Structural analysis indicates that the 3 disease-associated mutations at positions 206, 281, and 284 of the STING protein define a novel cluster of amino acids with functional importance in the regulation of type I interferon signaling.


Subject(s)
Inflammation/genetics , Interferon Type I/genetics , Membrane Proteins/genetics , Adolescent , Adult , Amino Acid Substitution , Child , Female , HEK293 Cells , Humans , Interferon Type I/metabolism , Male , Mutation , STAT1 Transcription Factor/metabolism , Signal Transduction
10.
J Clin Immunol ; 37(2): 123-132, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27943079

ABSTRACT

PURPOSE: Increased type I interferon is considered relevant to the pathology of a number of monogenic and complex disorders spanning pediatric rheumatology, neurology, and dermatology. However, no test exists in routine clinical practice to identify enhanced interferon signaling, thus limiting the ability to diagnose and monitor treatment of these diseases. Here, we set out to investigate the use of an assay measuring the expression of a panel of interferon-stimulated genes (ISGs) in children affected by a range of inflammatory diseases. DESIGN, SETTING, AND PARTICIPANTS: A cohort study was conducted between 2011 and 2016 at the University of Manchester, UK, and the Institut Imagine, Paris, France. RNA PAXgene blood samples and clinical data were collected from controls and symptomatic patients with a genetically confirmed or clinically well-defined inflammatory phenotype. The expression of six ISGs was measured by quantitative polymerase chain reaction, and the median fold change was used to calculate an interferon score (IS) for each subject compared to a previously derived panel of 29 controls (where +2 SD of the control data, an IS of >2.466, is considered as abnormal). Results were correlated with genetic and clinical data. RESULTS: Nine hundred ninety-two samples were analyzed from 630 individuals comprising symptomatic patients across 24 inflammatory genotypes/phenotypes, unaffected heterozygous carriers, and controls. A consistent upregulation of ISG expression was seen in 13 monogenic conditions (455 samples, 265 patients; median IS 10.73, interquartile range (IQR) 5.90-18.41), juvenile systemic lupus erythematosus (78 samples, 55 patients; median IS 10.60, IQR 3.99-17.27), and juvenile dermatomyositis (101 samples, 59 patients; median IS 9.02, IQR 2.51-21.73) compared to controls (78 samples, 65 subjects; median IS 0.688, IQR 0.427-1.196), heterozygous mutation carriers (89 samples, 76 subjects; median IS 0.862, IQR 0.493-1.942), and individuals with non-molecularly defined autoinflammation (89 samples, 69 patients; median IS 1.07, IQR 0.491-3.74). CONCLUSIONS AND RELEVANCE: An assessment of six ISGs can be used to define a spectrum of inflammatory diseases related to enhanced type I interferon signaling. If future studies demonstrate that the IS is a reactive biomarker, this measure may prove useful both in the diagnosis and the assessment of treatment efficacy.


Subject(s)
Inflammation/etiology , Inflammation/metabolism , Interferon Type I/metabolism , Signal Transduction , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Biomarkers , Case-Control Studies , Child , Child, Preschool , Female , Gene Expression Profiling , Gene Expression Regulation , Genetic Association Studies , Genotype , Humans , Infant , Infant, Newborn , Inflammation/diagnosis , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Male , Middle Aged , Phenotype , Young Adult
11.
Mol Oncol ; 8(7): 1169-80, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25266362

ABSTRACT

Fusions between the transmembrane protease serine 2 (TMPRSS2) and ETS related gene (ERG) represent one of the most specific biomarkers that define a distinct molecular subtype of prostate cancer. Studies of TMPRSS2-ERG gene fusions have seldom been performed at the protein level, primarily due to the lack of high-quality antibodies suitable for quantitative studies. Herein, we applied a recently developed PRISM (high-pressure high-resolution separations with intelligent selection and multiplexing)-SRM (selected reaction monitoring) strategy for quantifying ERG protein in prostate cancer cell lines and tumors. The highly sensitive PRISM-SRM assays provided confident detection of 6 unique ERG peptides in both TMPRSS2-ERG positive cell lines and tissues, but not in cell lines or tissues lacking the TMPRSS2-ERG rearrangement, clearly indicating that ERG protein expression is significantly increased in the presence of the TMPRSS2-ERG gene fusion. Significantly, our results provide evidence that two distinct ERG protein isoforms are simultaneously expressed in TMPRSS2-ERG positive samples as evidenced by the concomitant detection of two mutually exclusive peptides in two patient tumors and in the VCaP prostate cancer cell line. Three peptides, shared across almost all fusion protein products, were determined to be the most abundant peptides, providing "signature" peptides for detection of ERG over-expression resulting from TMPRSS2-ERG gene fusion. The PRISM-SRM assays provide valuable tools for studying TMPRSS2-ERG gene fusion protein products in prostate cancer.


Subject(s)
Oncogene Proteins, Fusion/analysis , Prostate/pathology , Prostatic Neoplasms/pathology , Amino Acid Sequence , Cell Line, Tumor , Gene Rearrangement , Humans , Male , Molecular Sequence Data , Oncogene Proteins, Fusion/genetics , Peptides/analysis , Peptides/genetics , Prostate/metabolism , Prostatic Neoplasms/genetics
12.
Cancer Res ; 74(6): 1651-60, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24473064

ABSTRACT

Impairment of double-stranded DNA break (DSB) repair is essential to many cancers. However, although mutations in DSB repair proteins are common in hereditary cancers, mechanisms of impaired DSB repair in sporadic cancers remain incompletely understood. Here, we describe the first role for a long noncoding RNA (lncRNA) in DSB repair in prostate cancer. We identify PCAT-1, a prostate cancer outlier lncRNA, which regulates cell response to genotoxic stress. PCAT-1 expression produces a functional deficiency in homologous recombination through its repression of the BRCA2 tumor suppressor, which, in turn, imparts a high sensitivity to small-molecule inhibitors of PARP1. These effects reflected a posttranscriptional repression of the BRCA2 3'UTR by PCAT-1. Our observations thus offer a novel mechanism of "BRCAness" in sporadic cancers.


Subject(s)
BRCA2 Protein/genetics , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , RNA, Long Noncoding/genetics , Recombinational DNA Repair , 3' Untranslated Regions , Animals , Antineoplastic Agents/pharmacology , BRCA2 Protein/metabolism , Cell Death/drug effects , Cell Line, Tumor , DNA Damage , Humans , Male , Mice , Mice, SCID , Phthalazines/pharmacology , Piperazines/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Prostatic Neoplasms/metabolism , RNA Interference , RNA, Long Noncoding/metabolism , Xenograft Model Antitumor Assays
13.
Science ; 342(6154): 1235587, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-24092746

ABSTRACT

Interpreting variants, especially noncoding ones, in the increasing number of personal genomes is challenging. We used patterns of polymorphisms in functionally annotated regions in 1092 humans to identify deleterious variants; then we experimentally validated candidates. We analyzed both coding and noncoding regions, with the former corroborating the latter. We found regions particularly sensitive to mutations ("ultrasensitive") and variants that are disruptive because of mechanistic effects on transcription-factor binding (that is, "motif-breakers"). We also found variants in regions with higher network centrality tend to be deleterious. Insertions and deletions followed a similar pattern to single-nucleotide variants, with some notable exceptions (e.g., certain deletions and enhancers). On the basis of these patterns, we developed a computational tool (FunSeq), whose application to ~90 cancer genomes reveals nearly a hundred candidate noncoding drivers.


Subject(s)
Genetic Variation , Molecular Sequence Annotation/methods , Neoplasms/genetics , Binding Sites/genetics , Genome, Human , Genomics , Humans , Kruppel-Like Transcription Factors/metabolism , Mutation , Polymorphism, Single Nucleotide , Population/genetics , RNA, Untranslated/genetics , Selection, Genetic
14.
Cell ; 153(3): 666-77, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23622249

ABSTRACT

The analysis of exonic DNA from prostate cancers has identified recurrently mutated genes, but the spectrum of genome-wide alterations has not been profiled extensively in this disease. We sequenced the genomes of 57 prostate tumors and matched normal tissues to characterize somatic alterations and to study how they accumulate during oncogenesis and progression. By modeling the genesis of genomic rearrangements, we identified abundant DNA translocations and deletions that arise in a highly interdependent manner. This phenomenon, which we term "chromoplexy," frequently accounts for the dysregulation of prostate cancer genes and appears to disrupt multiple cancer genes coordinately. Our modeling suggests that chromoplexy may induce considerable genomic derangement over relatively few events in prostate cancer and other neoplasms, supporting a model of punctuated cancer evolution. By characterizing the clonal hierarchy of genomic lesions in prostate tumors, we charted a path of oncogenic events along which chromoplexy may drive prostate carcinogenesis.


Subject(s)
Chromosome Aberrations , Gene Expression Regulation, Neoplastic , Genome, Human , Prostatic Neoplasms/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Cohort Studies , Genome-Wide Association Study , Humans , Male , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Prostatic Neoplasms/pathology
15.
Genes Chromosomes Cancer ; 52(6): 538-50, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23463663

ABSTRACT

Spindle cell rhabdomyosarcoma (RMS) is a rare form of RMS with different clinical characteristics between children and adult patients. Its genetic hallmark remains unknown and it remains debatable if there is pathogenetic relationship between the spindle cell and the so-called sclerosing RMS. We studied two pediatric and one adult spindle cell RMS by next generation RNA sequencing and FusionSeq data analysis to detect novel fusions. An SRF-NCOA2 fusion was detected in a spindle cell RMS from the posterior neck in a 7-month-old child. The fusion matched the tumor karyotype and was confirmed by FISH and RT-PCR, which showed fusion of SRF exon 6 to NCOA2 exon 12. Additional 14 spindle cell (from 8 children and 6 adults) and 4 sclerosing (from 2 children and 2 adults) RMS were tested by FISH for the presence of abnormalities in NCOA2, SRF, as well as for PAX3 and NCOA1. NCOA2 rearrangements were found in two additional spindle cell RMS from a 3-month-old and a 4-week-old child. In the latter tumor, TEAD1 was identified by rapid amplification of cDNA ends (RACE) to be the NCOA2 gene fusion partner. None of the adult tumors were positive for NCOA2 rearrangement. Despite similar histomorphology in adults and young children, these results suggest that spindle cell RMS is a heterogeneous disease genetically as well as clinically. Our findings also support a relationship between NCOA2-rearranged spindle cell RMS occurring in young childhood and the so-called congenital RMS, which often displays rearrangements at 8q13 locus (NCOA2).


Subject(s)
Chromosomes, Human, Pair 8/genetics , Gene Rearrangement , Nevus, Spindle Cell/pathology , Nuclear Receptor Coactivator 2/genetics , Rhabdomyosarcoma/genetics , Soft Tissue Neoplasms/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Humans , In Situ Hybridization, Fluorescence , In Vitro Techniques , Infant , Infant, Newborn , Male , Middle Aged , Prognosis , Rhabdomyosarcoma/congenital , Rhabdomyosarcoma/pathology , Soft Tissue Neoplasms/congenital , Soft Tissue Neoplasms/pathology , Young Adult
16.
Nat Genet ; 44(6): 685-9, 2012 May 20.
Article in English | MEDLINE | ID: mdl-22610119

ABSTRACT

Prostate cancer is the second most common cancer in men worldwide and causes over 250,000 deaths each year. Overtreatment of indolent disease also results in significant morbidity. Common genetic alterations in prostate cancer include losses of NKX3.1 (8p21) and PTEN (10q23), gains of AR (the androgen receptor gene) and fusion of ETS family transcription factor genes with androgen-responsive promoters. Recurrent somatic base-pair substitutions are believed to be less contributory in prostate tumorigenesis but have not been systematically analyzed in large cohorts. Here, we sequenced the exomes of 112 prostate tumor and normal tissue pairs. New recurrent mutations were identified in multiple genes, including MED12 and FOXA1. SPOP was the most frequently mutated gene, with mutations involving the SPOP substrate-binding cleft in 6-15% of tumors across multiple independent cohorts. Prostate cancers with mutant SPOP lacked ETS family gene rearrangements and showed a distinct pattern of genomic alterations. Thus, SPOP mutations may define a new molecular subtype of prostate cancer.


Subject(s)
Hepatocyte Nuclear Factor 3-alpha/genetics , Mediator Complex/genetics , Microtubule-Associated Proteins/genetics , Mutation , Prostatic Neoplasms/genetics , Exome , Humans , Male , Sequence Analysis, DNA
17.
Diagn Mol Pathol ; 21(2): 61-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22555088

ABSTRACT

Next-generation DNA and RNA sequencing requires intact nucleic acids from high-quality human tissue samples to better elucidate the molecular basis of cancer. We have developed a prostate biobanking protocol to acquire suitable samples for sequencing without compromising the accuracy of clinical diagnosis. To assess the clinical implications of implementing this protocol, we evaluated 105 consecutive radical prostatectomy specimens from November 2008 to February 2009. Alternating levels of prostate samples were submitted to Surgical Pathology as formalin-fixed, paraffin-embedded blocks and to the institutional biobank as frozen blocks. Differences in reported pathologic characteristics between clinical and procured specimens were compared. Clinical staging and grading were not affected by the biobank protocol. Tumor foci on frozen hematoxylin and eosin slides were identified and high-density tumor foci were scored and processed for DNA and RNA extractions for sequencing. Both DNA and RNA were extracted from 22 cases of 44 with high-density tumor foci. Eighty-two percent (18/22) of the samples passed rigorous quality control steps for DNA and RNA sequencing. To date, DNA extracted from 7 cases has undergone whole-genome sequencing, and RNA from 18 cases has been RNA sequenced. This protocol provides prostate tissue for high-throughput biomedical research and confirms the feasibility of actively integrating prostate cancer into The Cancer Genome Atlas Program, a member of the International Cancer Genome Consortium.


Subject(s)
Prostate/pathology , Prostatic Neoplasms/pathology , Tissue Banks , Adult , Aged , Aged, 80 and over , DNA/isolation & purification , Genes, Neoplasm , Genome, Human , Humans , International Cooperation , Male , Middle Aged , Prostatectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/surgery , RNA/isolation & purification , Sequence Analysis, RNA , Specimen Handling
18.
Proc Natl Acad Sci U S A ; 109(17): 6686-91, 2012 Apr 24.
Article in English | MEDLINE | ID: mdl-22496589

ABSTRACT

Copy number variants (CNVs) are a recently recognized class of human germ line polymorphisms and are associated with a variety of human diseases, including cancer. Because of the strong genetic influence on prostate cancer, we sought to identify functionally active CNVs associated with susceptibility of this cancer type. We queried low-frequency biallelic CNVs from 1,903 men of Caucasian origin enrolled in the Tyrol Prostate Specific Antigen Screening Cohort and discovered two CNVs strongly associated with prostate cancer risk. The first risk locus (P = 7.7 × 10(-4), odds ratio = 2.78) maps to 15q21.3 and overlaps a noncoding enhancer element that contains multiple activator protein 1 (AP-1) transcription factor binding sites. Chromosome conformation capture (Hi-C) data suggested direct cis-interactions with distant genes. The second risk locus (P = 2.6 × 10(-3), odds ratio = 4.8) maps to the α-1,3-mannosyl-glycoprotein 4-ß-N-acetylglucosaminyltransferase C (MGAT4C) gene on 12q21.31. In vitro cell-line assays found this gene to significantly modulate cell proliferation and migration in both benign and cancer prostate cells. Furthermore, MGAT4C was significantly overexpressed in metastatic versus localized prostate cancer. These two risk associations were replicated in an independent PSA-screened cohort of 800 men (15q21.3, combined P = 0.006; 12q21.31, combined P = 0.026). These findings establish noncoding and coding germ line CNVs as significant risk factors for prostate cancer susceptibility and implicate their role in disease development and progression.


Subject(s)
Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 15 , Gene Dosage , Genetic Predisposition to Disease , Prostatic Neoplasms/genetics , Case-Control Studies , Cell Line, Tumor , Cell Proliferation , Cohort Studies , Humans , Male , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Prostatic Neoplasms/pathology
19.
Pancreas ; 41(5): 685-90, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22466166

ABSTRACT

OBJECTIVES: This study aimed to evaluate microRNA (miRNA) expression in pancreatic resection specimens and fine needle aspiration biopsies and determine which, if any, miRNAs aid the distinction between benign and malignant pancreatic tumors in limited cytology material. METHODS: Resection specimens containing adenocarcinoma (n = 17), intraductal papillary mucinous neoplasms (n = 11), and nonneoplastic tissues (n = 15) were evaluated for miR-21, miR-221, miR-100, miR-155, and miR-181b expression by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR), and a subset of carcinomas and intraductal papillary mucinous neoplasms was analyzed with miRNA microarrays. Cellblocks containing carcinoma (n = 26) or benign pancreatic lesions (n = 11) from fine needle aspiration biopsies were subjected to qRT-PCR for miR-21, miR-221, miR-181b, miR-196a, and miR-217. RESULTS: Carcinomas showed higher expression of miR-21, miR-221, miR-155, miR-100, and miR-181b than benign lesions by qRT-PCR, and overexpression of miR-21, miR-221, and miR-181b was confirmed by microarray analysis. Cellblocks containing carcinoma showed higher expression of miR-21, miR-221, and miR-196a than those from benign lesions (P < 0.001, P = 0.009, and P < 0.001, respectively). CONCLUSIONS: Pancreatic ductal adenocarcinomas show differential expression of miRNAs compared to benign pancreatic lesions. A select panel of miRNAs aids the distinction between pancreatic lesions in cytology specimens.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Aged , Carcinoma, Pancreatic Ductal/diagnosis , Diagnosis, Differential , Female , Gene Expression Profiling , Humans , Logistic Models , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/diagnosis , Reverse Transcriptase Polymerase Chain Reaction
20.
Mol Syst Biol ; 7: 522, 2011 Aug 02.
Article in English | MEDLINE | ID: mdl-21811232

ABSTRACT

To study allele-specific expression (ASE) and binding (ASB), that is, differences between the maternally and paternally derived alleles, we have developed a computational pipeline (AlleleSeq). Our pipeline initially constructs a diploid personal genome sequence (and corresponding personalized gene annotation) using genomic sequence variants (SNPs, indels, and structural variants), and then identifies allele-specific events with significant differences in the number of mapped reads between maternal and paternal alleles. There are many technical challenges in the construction and alignment of reads to a personal diploid genome sequence that we address, for example, bias of reads mapping to the reference allele. We have applied AlleleSeq to variation data for NA12878 from the 1000 Genomes Project as well as matched, deeply sequenced RNA-Seq and ChIP-Seq data sets generated for this purpose. In addition to observing fairly widespread allele-specific behavior within individual functional genomic data sets (including results consistent with X-chromosome inactivation), we can study the interaction between ASE and ASB. Furthermore, we investigate the coordination between ASE and ASB from multiple transcription factors events using a regulatory network framework. Correlation analyses and network motifs show mostly coordinated ASB and ASE.


Subject(s)
Alleles , DNA-Binding Proteins/genetics , Gene Regulatory Networks , Sequence Analysis, RNA , Cell Line , Chromosome Mapping , Chromosomes, Human, X/genetics , Chromosomes, Human, Y/genetics , DNA-Binding Proteins/metabolism , Databases, Genetic , Gene Expression Regulation , Genome, Human , Humans , Molecular Sequence Annotation , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...