Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Clin Exp Immunol ; 215(3): 291-301, 2024 02 19.
Article in English | MEDLINE | ID: mdl-37583360

ABSTRACT

Innate immune activity fuels intestinal inflammation in Crohn's disease (CD), an inflammatory bowel disease. Identification and targeting of new molecular regulators of the innate activity are warranted to control the disease. Inhibitor of apoptosis proteins (IAPs) regulate both cell survival and inflammatory signaling. We investigated the effects of IAP inhibition by second mitochondria-derived activator of caspases (SMAC) mimetics (SMs) on innate responses and cell death to pathogen-associated molecular patterns in peripheral blood mononuclear cells (PBMCs) and monocytes. IAPs inhibited lipopolysaccharide (LPS)-induced expression of proinflammatory interleukin (IL)-1ß, IL-6. Likewise, LPS (but not muramyl dipeptide or Escherichia coli) induced TNF-α was inhibited in CD and control PBMCs. The SM effect was partially reversed by inhibition of receptor-interacting serine/threonine-protein kinase 1 (RIPK1). The effect was mainly cell death independent. Thus, IAP inhibition by SMs leads to reduced production of proinflammatory cytokines and may be considered in the efforts to develop new therapeutic strategies to control CD.


Subject(s)
Crohn Disease , Humans , Lipopolysaccharides , Leukocytes, Mononuclear/metabolism , Healthy Volunteers , Cytokines/metabolism , Carrier Proteins , Tumor Necrosis Factor-alpha/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
2.
Pulm Pharmacol Ther ; 79: 102201, 2023 04.
Article in English | MEDLINE | ID: mdl-36841351

ABSTRACT

Activated PI3Kδ Syndrome (APDS) is a rare inherited inborn error of immunity caused by mutations that constitutively activate the p110 delta isoform of phosphoinositide 3-kinase (PI3Kδ), resulting in recurring pulmonary infections. Currently no licensed therapies are available. Here we report the results of an open-label trial in which five subjects were treated for 12 weeks with nemiralisib, an inhaled inhibitor of PI3Kδ, to determine safety, systemic exposure, together with lung and systemic biomarker profiles (Clinicaltrial.gov: NCT02593539). Induced sputum was captured to measure changes in phospholipids and inflammatory mediators, and blood samples were collected to assess pharmacokinetics of nemiralisib, and systemic biomarkers. Nemiralisib was shown to have an acceptable safety and tolerability profile, with cough being the most common adverse event, and no severe adverse events reported during the study. No meaningful changes in phosphatidylinositol (3,4,5)-trisphosphate (PIP3; the enzyme product of PI3Kδ) or downstream inflammatory markers in induced sputum, were observed following nemiralisib treatment. Similarly, there were no meaningful changes in blood inflammatory markers, or lymphocytes subsets. Systemic levels of nemiralisib were higher in subjects in this study compared to previous observations. While nemiralisib had an acceptable safety profile, there was no convincing evidence of target engagement in the lung following inhaled dosing and no downstream effects observed in either the lung or blood compartments. We speculate that this could be explained by nemiralisib not being retained in the lung for sufficient duration, suggested by the increased systemic exposure, perhaps due to pre-existing structural lung damage. In this study investigating a small number of subjects with APDS, nemiralisib appeared to be safe and well-tolerated. However, data from this study do not support the hypothesis that inhaled treatment with nemiralisib would benefit patients with APDS.


Subject(s)
Antineoplastic Agents , Phosphatidylinositol 3-Kinases , Humans , Administration, Inhalation , Protein Kinase Inhibitors , Phosphatidylinositol 3-Kinase
3.
Front Immunol ; 13: 982870, 2022.
Article in English | MEDLINE | ID: mdl-36045673

ABSTRACT

Topoisomerase 2ß (TOP2B) introduces transient double strand breaks in the DNA helix to remove supercoiling structures and unwind entangled DNA strains. Advances in genomic technologies have enabled the discovery of novel functions for TOP2B in processes such as releasing of the paused RNA polymerase II and maintaining the genome organization through DNA loop domains. Thus, TOP2B can regulate transcription directly by acting on transcription elongation and indirectly by controlling interactions between enhancer and promoter regions through genome folding. The identification of TOP2B mutations in humans unexpectedly revealed a unique role of TOP2B in B-cell progenitors. Here we discuss the functions of TOP2B and the mechanisms leading to the B-cell development defect in patients with TOP2B deficiency.


Subject(s)
DNA Topoisomerases, Type II , DNA-Binding Proteins , DNA , DNA Topoisomerases, Type II/chemistry , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/genetics , Humans , Poly-ADP-Ribose Binding Proteins
4.
J Crohns Colitis ; 15(11): 1908-1919, 2021 Nov 08.
Article in English | MEDLINE | ID: mdl-33891011

ABSTRACT

BACKGROUND AND AIMS: Very early onset inflammatory bowel disease [VEOIBD] is characterized by intestinal inflammation affecting infants and children less than 6 years of age. To date, over 60 monogenic aetiologies of VEOIBD have been identified, many characterized by highly penetrant recessive or dominant variants in underlying immune and/or epithelial pathways. We sought to identify the genetic cause of VEOIBD in a subset of patients with a unique clinical presentation. METHODS: Whole exome sequencing was performed on five families with ten patients who presented with a similar constellation of symptoms including medically refractory infantile-onset IBD, bilateral sensorineural hearing loss and, in the majority, recurrent infections. Genetic aetiologies of VEOIBD were assessed and Sanger sequencing was performed to confirm novel genetic findings. Western analysis on peripheral blood mononuclear cells and functional studies with epithelial cell lines were employed. RESULTS: In each of the ten patients, we identified damaging heterozygous or biallelic variants in the Syntaxin-Binding Protein 3 gene [STXBP3], a protein known to regulate intracellular vesicular trafficking in the syntaxin-binding protein family of molecules, but not associated to date with either VEOIBD or sensorineural hearing loss. These mutations interfere with either intron splicing or protein stability and lead to reduced STXBP3 protein expression. Knock-down of STXBP3 in CaCo2 cells resulted in defects in cell polarity. CONCLUSION: Overall, we describe a novel genetic syndrome and identify a critical role for STXBP3 in VEOIBD, sensorineural hearing loss and immune dysregulation.


Subject(s)
Hearing Loss, Sensorineural/genetics , Immune System Diseases/genetics , Inflammatory Bowel Diseases/genetics , Qa-SNARE Proteins/analysis , Age of Onset , Female , Genetic Variation/genetics , Hearing Loss, Sensorineural/epidemiology , Humans , Immune System Diseases/epidemiology , Infant, Newborn , Inflammatory Bowel Diseases/epidemiology , Male , Qa-SNARE Proteins/genetics , Exome Sequencing
5.
Tuberculosis (Edinb) ; 124: 101983, 2020 09.
Article in English | MEDLINE | ID: mdl-32829077

ABSTRACT

The ability to genetically engineer pathogenic mycobacteria has increased significantly over the last decades due to the generation of new molecular tools. Recently, the application of the Streptococcus pyogenes and the Streptococcus thermophilus CRISPR-Cas9 systems in mycobacteria has enabled gene editing and efficient CRISPR interference-mediated transcriptional regulation. Here, we converted CRISPR interference into an efficient genome editing tool for mycobacteria. We demonstrate that the Streptococcus thermophilus CRISPR1-Cas9 (Sth1Cas9) is functional in Mycobacterium marinum and Mycobacterium tuberculosis, enabling highly efficient and precise DNA breaks and indel formation, without any off-target effects. In addition, with dual sgRNAs this system can be used to generate two indels simultaneously or to create specific deletions. The ability to use the power of the CRISPR-Cas9-mediated gene editing toolbox in M. tuberculosis with a single step will accelerate research into this deadly pathogen.


Subject(s)
CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing , Mycobacterium marinum/genetics , Mycobacterium tuberculosis/genetics , Streptococcus thermophilus/genetics , Antitubercular Agents/pharmacology , Bacterial Proteins/genetics , CRISPR-Associated Protein 9/metabolism , Catalase/genetics , Drug Resistance, Bacterial/drug effects , Gene Deletion , Gene Expression Regulation, Bacterial , INDEL Mutation , Isoniazid/pharmacology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/growth & development , RNA, Guide, Kinetoplastida/genetics , Streptococcus thermophilus/enzymology
7.
Nat Commun ; 10(1): 3765, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31434886

ABSTRACT

Of the 1.8 billion people worldwide infected with Mycobacterium tuberculosis, 5-15% will develop active tuberculosis (TB). Approximately half will progress to active TB within the first 18 months after infection, presumably because they fail to mount an effective initial immune response. Here, in a genome-wide genetic study of early TB progression, we genotype 4002 active TB cases and their household contacts in Peru. We quantify genetic heritability ([Formula: see text]) of early TB progression to be 21.2% (standard error 0.08). This suggests TB progression has a strong genetic basis, and is comparable to traits with well-established genetic bases. We identify a novel association between early TB progression and variants located in a putative enhancer region on chromosome 3q23 (rs73226617, OR = 1.18; P = 3.93 × 10-8). With in silico and in vitro analyses we identify rs73226617 or rs148722713 as the likely functional variant and ATP1B3 as a potential causal target gene with monocyte specific function.


Subject(s)
Disease Progression , Mycobacterium tuberculosis/pathogenicity , Tuberculosis/genetics , Adult , Female , Gene Expression , Genetic Loci , Genome-Wide Association Study , Genotype , Humans , Male , Monocytes , Mycobacterium tuberculosis/genetics , Peru , Sodium-Potassium-Exchanging ATPase/genetics
8.
J Allergy Clin Immunol ; 144(5): 1364-1376, 2019 11.
Article in English | MEDLINE | ID: mdl-31201888

ABSTRACT

BACKGROUND: CCAAT enhancer-binding protein epsilon (C/EBPε) is a transcription factor involved in late myeloid lineage differentiation and cellular function. The only previously known disorder linked to C/EBPε is autosomal recessive neutrophil-specific granule deficiency leading to severely impaired neutrophil function and early mortality. OBJECTIVE: The aim of this study was to molecularly characterize the effects of C/EBPε transcription factor Arg219His mutation identified in a Finnish family with previously genetically uncharacterized autoinflammatory and immunodeficiency syndrome. METHODS: Genetic analysis, proteomics, genome-wide transcriptional profiling by means of RNA-sequencing, chromatin immunoprecipitation (ChIP) sequencing, and assessment of the inflammasome function of primary macrophages were performed. RESULTS: Studies revealed a novel mechanism of genome-wide gain-of-function that dysregulated transcription of 464 genes. Mechanisms involved dysregulated noncanonical inflammasome activation caused by decreased association with transcriptional repressors, leading to increased chromatin occupancy and considerable changes in transcriptional activity, including increased expression of NLR family, pyrin domain-containing 3 protein (NLRP3) and constitutively expressed caspase-5 in macrophages. CONCLUSION: We describe a novel autoinflammatory disease with defective neutrophil function caused by a homozygous Arg219His mutation in the transcription factor C/EBPε. Mutated C/EBPε acts as a regulator of both the inflammasome and interferome, and the Arg219His mutation causes the first human monogenic neomorphic and noncanonical inflammasomopathy/immunodeficiency. The mechanism, including widely dysregulated transcription, is likely not unique for C/EBPε. Similar multiomics approaches should also be used in studying other transcription factor-associated diseases.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Gain of Function Mutation/genetics , Immunologic Deficiency Syndromes/genetics , Inflammasomes/genetics , Inflammation/genetics , Macrophages/metabolism , Neutrophils/physiology , Aged , Caspases/genetics , Caspases/metabolism , Cells, Cultured , Female , Gene Expression Profiling , Humans , Inflammasomes/metabolism , Macrophages/pathology , Male , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pedigree , Sequence Analysis, RNA , Up-Regulation
9.
J Pediatr Hematol Oncol ; 41(4): 328-333, 2019 05.
Article in English | MEDLINE | ID: mdl-29939941

ABSTRACT

X-linked severe combined immunodeficiency disease (SCID) is caused by mutations in the interleukin (IL)-2 receptor γ (IL2RG) gene and patients usually present with a TBNK SCID phenotype. Nevertheless, a minority of these patients present with a TBNK phenotype, similar to the IL-7R-deficient patients. We report a patient with a novel missense p.Glu297Gly mutation in the IL2RG gene presenting with a leaky TBNK SCID with delayed onset, moderate susceptibility to infections, and nodular regenerative hyperplasia. He presents with preserved STAT5 tyrosine phosphorylation in response to IL-15 stimulation but not in response to IL-2 and IL-7, resulting in the NK phenotype.


Subject(s)
Interleukin Receptor Common gamma Subunit/genetics , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/immunology , X-Linked Combined Immunodeficiency Diseases/pathology , B-Lymphocytes/immunology , Child, Preschool , Humans , Hyperplasia/pathology , Interleukin-15/metabolism , Killer Cells, Natural/immunology , Male , Mutation, Missense , Phenotype , Phosphorylation , STAT5 Transcription Factor/metabolism , T-Lymphocytes/immunology
10.
J Allergy Clin Immunol ; 143(4): 1482-1495, 2019 04.
Article in English | MEDLINE | ID: mdl-30170123

ABSTRACT

BACKGROUND: Caspase activation and recruitment domain 11 (CARD11) encodes a scaffold protein in lymphocytes that links antigen receptor engagement with downstream signaling to nuclear factor κB, c-Jun N-terminal kinase, and mechanistic target of rapamycin complex 1. Germline CARD11 mutations cause several distinct primary immune disorders in human subjects, including severe combined immune deficiency (biallelic null mutations), B-cell expansion with nuclear factor κB and T-cell anergy (heterozygous, gain-of-function mutations), and severe atopic disease (loss-of-function, heterozygous, dominant interfering mutations), which has focused attention on CARD11 mutations discovered by using whole-exome sequencing. OBJECTIVES: We sought to determine the molecular actions of an extended allelic series of CARD11 and to characterize the expanding range of clinical phenotypes associated with heterozygous CARD11 loss-of-function alleles. METHODS: Cell transfections and primary T-cell assays were used to evaluate signaling and function of CARD11 variants. RESULTS: Here we report on an expanded cohort of patients harboring novel heterozygous CARD11 mutations that extend beyond atopy to include other immunologic phenotypes not previously associated with CARD11 mutations. In addition to (and sometimes excluding) severe atopy, heterozygous missense and indel mutations in CARD11 presented with immunologic phenotypes similar to those observed in signal transducer and activator of transcription 3 loss of function, dedicator of cytokinesis 8 deficiency, common variable immunodeficiency, neutropenia, and immune dysregulation, polyendocrinopathy, enteropathy, X-linked-like syndrome. Pathogenic variants exhibited dominant negative activity and were largely confined to the CARD or coiled-coil domains of the CARD11 protein. CONCLUSION: These results illuminate a broader phenotypic spectrum associated with CARD11 mutations in human subjects and underscore the need for functional studies to demonstrate that rare gene variants encountered in expected and unexpected phenotypes must nonetheless be validated for pathogenic activity.


Subject(s)
CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/immunology , Guanylate Cyclase/genetics , Guanylate Cyclase/immunology , Immune System Diseases/genetics , Immune System Diseases/immunology , Adult , Female , Humans , Male , Mutation , Phenotype
11.
Am J Med Genet B Neuropsychiatr Genet ; 177(7): 641-657, 2018 10.
Article in English | MEDLINE | ID: mdl-30325587

ABSTRACT

Individuals with psychiatric disorders have elevated rates of autoimmune comorbidity and altered immune signaling. It is unclear whether these altered immunological states have a shared genetic basis with those psychiatric disorders. The present study sought to use existing summary-level data from previous genome-wide association studies to determine if commonly varying single nucleotide polymorphisms are shared between psychiatric and immune-related phenotypes. We estimated heritability and examined pair-wise genetic correlations using the linkage disequilibrium score regression (LDSC) and heritability estimation from summary statistics methods. Using LDSC, we observed significant genetic correlations between immune-related disorders and several psychiatric disorders, including anorexia nervosa, attention deficit-hyperactivity disorder, bipolar disorder, major depression, obsessive compulsive disorder, schizophrenia, smoking behavior, and Tourette syndrome. Loci significantly mediating genetic correlations were identified for schizophrenia when analytically paired with Crohn's disease, primary biliary cirrhosis, systemic lupus erythematosus, and ulcerative colitis. We report significantly correlated loci and highlight those containing genome-wide associations and candidate genes for respective disorders. We also used the LDSC method to characterize genetic correlations among the immune-related phenotypes. We discuss our findings in the context of relevant genetic and epidemiological literature, as well as the limitations and caveats of the study.


Subject(s)
Autoimmune Diseases/genetics , Mental Disorders/genetics , Autoimmune Diseases/physiopathology , Comorbidity , Databases, Factual , Female , Genetic Predisposition to Disease , Genome-Wide Association Study/methods , Humans , Linkage Disequilibrium , Male , Mental Disorders/physiopathology , Multifactorial Inheritance , Polymorphism, Single Nucleotide , White People/genetics
12.
Nat Commun ; 9(1): 3174, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30093657

ABSTRACT

Streptococcus pneumoniae is a major cause of pneumonia and a leading cause of death world-wide. Antibody-mediated immune responses can confer protection against repeated exposure to S. pneumoniae, yet vaccines offer only partial protection. Patients with Activated PI3Kδ Syndrome (APDS) are highly susceptible to S. pneumoniae. We generated a conditional knock-in mouse model of this disease and identify a CD19+B220- B cell subset that is induced by PI3Kδ signaling, resides in the lungs, and is correlated with increased susceptibility to S. pneumoniae during early phases of infection via an antibody-independent mechanism. We show that an inhaled PI3Kδ inhibitor improves survival rates following S. pneumoniae infection in wild-type mice and in mice with activated PI3Kδ. These results suggest that a subset of B cells in the lung can promote the severity of S. pneumoniae infection, representing a potential therapeutic target.


Subject(s)
B-Lymphocytes/immunology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Pneumococcal Infections/immunology , Animals , Antigens, CD19/metabolism , B-Lymphocytes/cytology , Class I Phosphatidylinositol 3-Kinases , Enzyme Activation , Female , Gene Knock-In Techniques , Genotype , Humans , Interleukin-10/metabolism , Lung/immunology , Male , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Signal Transduction , Species Specificity , Streptococcus pneumoniae
13.
Science ; 361(6404): 810-813, 2018 08 24.
Article in English | MEDLINE | ID: mdl-30026316

ABSTRACT

RIPK1 (receptor-interacting serine/threonine kinase 1) is a master regulator of signaling pathways leading to inflammation and cell death and is of medical interest as a drug target. We report four patients from three unrelated families with complete RIPK1 deficiency caused by rare homozygous mutations. The patients suffered from recurrent infections, early-onset inflammatory bowel disease, and progressive polyarthritis. They had immunodeficiency with lymphopenia and altered production of various cytokines revealed by whole-blood assays. In vitro, RIPK1-deficient cells showed impaired mitogen-activated protein kinase activation and cytokine secretion and were prone to necroptosis. Hematopoietic stem cell transplantation reversed cytokine production defects and resolved clinical symptoms in one patient. Thus, RIPK1 plays a critical role in the human immune system.


Subject(s)
Arthritis/genetics , Inflammatory Bowel Diseases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Severe Combined Immunodeficiency/genetics , Alleles , Arthritis/immunology , Cytokines/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Inflammatory Bowel Diseases/immunology , Lymphopenia/genetics , Male , Mitogen-Activated Protein Kinases/metabolism , Pedigree , Severe Combined Immunodeficiency/immunology
14.
Front Immunol ; 9: 369, 2018.
Article in English | MEDLINE | ID: mdl-29535736

ABSTRACT

Activated PI3 kinase delta syndrome (APDS) is a primary immunodeficiency caused by dominant mutations that increase activity of phosphoinositide-3-kinase δ (PI3Kδ). APDS can be caused by mutations in the PIK3CD gene that encodes PI3Kδ catalytic subunit p110δ (APDS1) or mutations in the PIK3R1 gene that encodes regulatory subunit p85α (APDS2). APDS research advanced rapidly after the initial discovery in 2013. More than 200 APDS patients have been identified around the world. Multiple novel APDS mutations were reported and molecular mechanisms leading to PI3Kδ activation have been elucidated. The finding of APDS significantly increased our understanding of the role of PI3Kδ in the human immune system. Perhaps most importantly, discovery of the molecular basis of this primary immunodeficiency suggested that APDS patients, who previously received only non-specific therapy, could be treated by a novel class of drugs that inhibits PI3Kδ activity. This led to the ongoing clinical trials of selective PI3Kδ inhibitors in APDS patients. Overall, the APDS story provides an excellent example of translational research, beginning with patients who had an unknown disease cause and leading to a novel specific knowledge-based treatment.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/genetics , Immunologic Deficiency Syndromes/genetics , Mutation/genetics , Phosphatidylinositol 3-Kinases/genetics , Animals , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class Ia Phosphatidylinositol 3-Kinase , Clinical Trials as Topic , Humans , Immunologic Deficiency Syndromes/therapy , Molecular Targeted Therapy , Phenotype , Phosphoinositide-3 Kinase Inhibitors , Primary Immunodeficiency Diseases
15.
Front Immunol ; 9: 2863, 2018.
Article in English | MEDLINE | ID: mdl-30619256

ABSTRACT

Background: The auto-inflammation and phospholipase Cγ2 (PLCγ2)-associated antibody deficiency and immune dysregulation (APLAID) syndrome is a rare primary immunodeficiency caused by a gain-of-function mutation S707Y in the PLCG2 gene previously described in two patients from one family. The APLAID patients presented with early-onset blistering skin lesions, posterior uveitis, inflammatory bowel disease (IBD) and recurrent sinopulmonary infections caused by a humoral defect, but lacked circulating autoantibodies and had no cold-induced urticaria, contrary to the patients with the related PLAID syndrome. Case: We describe a new APLAID patient who presented with vesiculopustular rash in the 1st weeks of life, followed by IBD, posterior uveitis, recurrent chest infections, interstitial pneumonitis, and also had sensorineural deafness and cutis laxa. Her disease has been refractory to most treatments, including IL1 blockers and a trial with ruxolitinib has been attempted. Results: In this patient, we found a unique de novo heterozygous missense L848P mutation in the PLCG2 gene, predicted to affect the PLCγ2 structure. Similarly to S707Y, the L848P mutation led to the increased basal and EGF-stimulated PLCγ2 activity in vitro. Whole blood assays showed reduced production of IFN-γ and IL-17 in response to polyclonal T-cell stimulation and reduced production of IL-10 and IL-1ß after LPS stimulation. Reduced IL-1ß levels and the lack of clinical response to treatment with IL-1 blockers argue against NLRP3 inflammasome hyperactivation being the main mechanism mediating the APLAID pathogenesis. Conclusion: Our findings indicate that L848P is novel a gain-of-function mutation that leads to PLCγ2 activation and suggest cutis laxa as a possible clinical manifestations of the APLAID syndrome.


Subject(s)
Cutis Laxa/genetics , Hereditary Autoinflammatory Diseases/genetics , Immunologic Deficiency Syndromes/genetics , Mutation, Missense , Phospholipase C gamma/genetics , Amino Acid Sequence , Base Sequence , Cutis Laxa/complications , Cutis Laxa/enzymology , DNA Mutational Analysis , Female , Hereditary Autoinflammatory Diseases/complications , Hereditary Autoinflammatory Diseases/enzymology , Humans , Immunologic Deficiency Syndromes/complications , Immunologic Deficiency Syndromes/enzymology , Infant, Newborn , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/genetics , Male , Pedigree , Phospholipase C gamma/chemistry , Phospholipase C gamma/metabolism , Sequence Homology, Amino Acid
16.
Nat Genet ; 49(7): 986-992, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28530675

ABSTRACT

SNP heritability, the proportion of phenotypic variance explained by SNPs, has been reported for many hundreds of traits. Its estimation requires strong prior assumptions about the distribution of heritability across the genome, but current assumptions have not been thoroughly tested. By analyzing imputed data for a large number of human traits, we empirically derive a model that more accurately describes how heritability varies with minor allele frequency (MAF), linkage disequilibrium (LD) and genotype certainty. Across 19 traits, our improved model leads to estimates of common SNP heritability on average 43% (s.d. 3%) higher than those obtained from the widely used software GCTA and 25% (s.d. 2%) higher than those from the recently proposed extension GCTA-LDMS. Previously, DNase I hypersensitivity sites were reported to explain 79% of SNP heritability; using our improved heritability model, their estimated contribution is only 24%.


Subject(s)
Genome-Wide Association Study/methods , Models, Genetic , Multifactorial Inheritance/genetics , Polymorphism, Single Nucleotide/genetics , Alleles , Case-Control Studies , Chromosome Fragile Sites , Cohort Studies , Computer Simulation , Deoxyribonuclease I , Gene Frequency , Genetic Association Studies , Humans , Inheritance Patterns , Quantitative Trait, Heritable
17.
Nat Commun ; 7: 13992, 2016 12 23.
Article in English | MEDLINE | ID: mdl-28008925

ABSTRACT

Mutations in genes encoding components of the immune system cause primary immunodeficiencies. Here, we study a patient with recurrent atypical mycobacterial infection and early-onset metastatic bladder carcinoma. Exome sequencing identified two homozygous missense germline mutations, P733L and P832S, in the JAK1 protein that mediates signalling from multiple cytokine receptors. Cells from this patient exhibit reduced JAK1 and STAT phosphorylation following cytokine stimulations, reduced induction of expression of interferon-regulated genes and dysregulated cytokine production; which are indicative of signalling defects in multiple immune response pathways including Interferon-γ production. Reconstitution experiments in the JAK1-deficient cells demonstrate that the impaired JAK1 function is mainly attributable to the effect of the P733L mutation. Further analyses of the mutant protein reveal a phosphorylation-independent role of JAK1 in signal transduction. These findings clarify JAK1 signalling mechanisms and demonstrate a critical function of JAK1 in protection against mycobacterial infection and possibly the immunological surveillance of cancer.


Subject(s)
Alleles , Janus Kinase 1/genetics , Mutation/genetics , Mycobacterium Infections/enzymology , Mycobacterium Infections/genetics , Amino Acid Sequence , Base Sequence , Blood Cells/metabolism , Child , Child, Preschool , Cytokines/blood , Disease Susceptibility , Female , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Humans , Interferon-alpha/pharmacology , Interferon-gamma/pharmacology , Janus Kinase 1/chemistry , Male , Pedigree , Phosphorylation/drug effects , Protein Domains , STAT Transcription Factors/metabolism , Signal Transduction/genetics , TYK2 Kinase/metabolism , Young Adult
18.
Nat Rev Immunol ; 16(11): 702-714, 2016 11.
Article in English | MEDLINE | ID: mdl-27616589

ABSTRACT

Primary immunodeficiencies are inherited disorders of the immune system, often caused by the mutation of genes required for lymphocyte development and activation. Recently, several studies have identified gain-of-function mutations in the phosphoinositide 3-kinase (PI3K) genes PIK3CD (which encodes p110δ) and PIK3R1 (which encodes p85α) that cause a combined immunodeficiency syndrome, referred to as activated PI3Kδ syndrome (APDS; also known as p110δ-activating mutation causing senescent T cells, lymphadenopathy and immunodeficiency (PASLI)). Paradoxically, both loss-of-function and gain-of-function mutations that affect these genes lead to immunosuppression, albeit via different mechanisms. Here, we review the roles of PI3Kδ in adaptive immunity, describe the clinical manifestations and mechanisms of disease in APDS and highlight new insights into PI3Kδ gleaned from these patients, as well as implications of these findings for clinical therapy.


Subject(s)
Immunologic Deficiency Syndromes/etiology , Immunologic Deficiency Syndromes/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cellular Senescence/genetics , Cellular Senescence/immunology , Enzyme Activation , Gene Expression Regulation , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Immunity , Immunologic Deficiency Syndromes/therapy , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocytes/cytology , Lymphocytes/immunology , Lymphocytes/metabolism , Molecular Targeted Therapy , Mutation , Phosphatidylinositol 3-Kinases/chemistry , Phosphoinositide-3 Kinase Inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , Signal Transduction
19.
J Allergy Clin Immunol ; 138(1): 210-218.e9, 2016 07.
Article in English | MEDLINE | ID: mdl-27221134

ABSTRACT

BACKGROUND: Activated phosphoinositide 3-kinase δ syndrome (APDS) 2 (p110δ-activating mutations causing senescent T cells, lymphadenopathy, and immunodeficiency [PASLI]-R1), a recently described primary immunodeficiency, results from autosomal dominant mutations in PIK3R1, the gene encoding the regulatory subunit (p85α, p55α, and p50α) of class IA phosphoinositide 3-kinases. OBJECTIVES: We sought to review the clinical, immunologic, and histopathologic phenotypes of APDS2 in a genetically defined international patient cohort. METHODS: The medical and biological records of 36 patients with genetically diagnosed APDS2 were collected and reviewed. RESULTS: Mutations within splice acceptor and donor sites of exon 11 of the PIK3R1 gene lead to APDS2. Recurrent upper respiratory tract infections (100%), pneumonitis (71%), and chronic lymphoproliferation (89%, including adenopathy [75%], splenomegaly [43%], and upper respiratory tract lymphoid hyperplasia [48%]) were the most common features. Growth retardation was frequently noticed (45%). Other complications were mild neurodevelopmental delay (31%); malignant diseases (28%), most of them being B-cell lymphomas; autoimmunity (17%); bronchiectasis (18%); and chronic diarrhea (24%). Decreased serum IgA and IgG levels (87%), increased IgM levels (58%), B-cell lymphopenia (88%) associated with an increased frequency of transitional B cells (93%), and decreased numbers of naive CD4 and naive CD8 cells but increased numbers of CD8 effector/memory T cells were predominant immunologic features. The majority of patients (89%) received immunoglobulin replacement; 3 patients were treated with rituximab, and 6 were treated with rapamycin initiated after diagnosis of APDS2. Five patients died from APDS2-related complications. CONCLUSION: APDS2 is a combined immunodeficiency with a variable clinical phenotype. Complications are frequent, such as severe bacterial and viral infections, lymphoproliferation, and lymphoma similar to APDS1/PASLI-CD. Immunoglobulin replacement therapy, rapamycin, and, likely in the near future, selective phosphoinositide 3-kinase δ inhibitors are possible treatment options.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/genetics , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/etiology , Phenotype , Adolescent , Adult , Alleles , Biopsy , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Child , Child, Preschool , Cohort Studies , Female , Gene Frequency , Genotype , Humans , Immunologic Deficiency Syndromes/mortality , Male , Middle Aged , Mutation , RNA Splice Sites , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Young Adult
20.
Nat Genet ; 48(3): 318-22, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26829749

ABSTRACT

Mycobacterium tuberculosis infections cause 9 million new tuberculosis cases and 1.5 million deaths annually. To identify variants conferring risk of tuberculosis, we tested 28.3 million variants identified through whole-genome sequencing of 2,636 Icelanders for association with tuberculosis (8,162 cases and 277,643 controls), pulmonary tuberculosis (PTB) and M. tuberculosis infection. We found association of three variants in the region harboring genes encoding the class II human leukocyte antigens (HLAs): rs557011[T] (minor allele frequency (MAF) = 40.2%), associated with M. tuberculosis infection (odds ratio (OR) = 1.14, P = 3.1 × 10(-13)) and PTB (OR = 1.25, P = 5.8 × 10(-12)), and rs9271378[G] (MAF = 32.5%), associated with PTB (OR = 0.78, P = 2.5 × 10(-12))--both located between HLA-DQA1 and HLA-DRB1--and a missense variant encoding p.Ala210Thr in HLA-DQA1 (MAF = 19.1%, rs9272785), associated with M. tuberculosis infection (P = 9.3 × 10(-9), OR = 1.14). We replicated association of these variants with PTB in samples of European ancestry from Russia and Croatia (P < 5.9 × 10(-4)). These findings show that the HLA class II region contributes to genetic risk of tuberculosis, possibly through reduced presentation of protective M. tuberculosis antigens to T cells.


Subject(s)
HLA-DQ alpha-Chains/genetics , HLA-DRB1 Chains/genetics , Mycobacterium tuberculosis/pathogenicity , Tuberculosis, Pulmonary/genetics , Alleles , Gene Frequency , Genetic Predisposition to Disease , Genetic Variation , Genome, Human , Genome-Wide Association Study , HLA-DQ alpha-Chains/immunology , HLA-DRB1 Chains/immunology , Humans , Iceland , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/immunology , Risk Factors , T-Lymphocytes/immunology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , White People
SELECTION OF CITATIONS
SEARCH DETAIL
...