Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters

Publication year range
1.
Cell ; 167(4): 1099-1110.e14, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27814507

ABSTRACT

As part of the Human Functional Genomics Project, which aims to understand the factors that determine the variability of immune responses, we investigated genetic variants affecting cytokine production in response to ex vivo stimulation in two independent cohorts of 500 and 200 healthy individuals. We demonstrate a strong impact of genetic heritability on cytokine production capacity after challenge with bacterial, fungal, viral, and non-microbial stimuli. In addition to 17 novel genome-wide significant cytokine QTLs (cQTLs), our study provides a comprehensive picture of the genetic variants that influence six different cytokines in whole blood, blood mononuclear cells, and macrophages. Important biological pathways that contain cytokine QTLs map to pattern recognition receptors (TLR1-6-10 cluster), cytokine and complement inhibitors, and the kallikrein system. The cytokine QTLs show enrichment for monocyte-specific enhancers, are more often located in regions under positive selection, and are significantly enriched among SNPs associated with infections and immune-mediated diseases. PAPERCLIP.


Subject(s)
Cytokines/genetics , Cytokines/immunology , Infections/immunology , Adolescent , Adult , Aged , Blood/immunology , Female , Genome-Wide Association Study , Human Genome Project , Humans , Infections/microbiology , Infections/virology , Leukocytes, Mononuclear/immunology , Macrophages/immunology , Male , Middle Aged , Polymorphism, Single Nucleotide , Quantitative Trait Loci
2.
Immunogenetics ; 70(6): 373-379, 2018 06.
Article in English | MEDLINE | ID: mdl-29256176

ABSTRACT

In the last decade, autosomal recessive interleukin-12 receptor ß1 (IL-12Rß1) deficiency, the most common cause of Mendelian susceptibility to mycobacterial disease (MSMD), has been diagnosed in a few children and adults with severe tuberculosis in Iran. Here, we report three cases referred to the Immunology, Asthma and Allergy ward at the National Research Institute of Tuberculosis and Lung Diseases (NRITLD) at Masih Daneshvari Hospital from 2012 to 2017 with Mycobacterium tuberculosis and non-tuberculous mycobacteria infections due to defects in IL-12Rß1 but with different clinical manifestations. All three were homozygous for either an IL-12Rß1 missense or nonsense mutation that caused the IL-12Rß1 protein not to be expressed on the cell membrane and completely abolished the cellular response to recombinant IL-12. Our findings suggest that the presence of IL-12Rß1 deficiency should be determined in children with mycobacterial infections at least in countries with a high prevalence of parental consanguinity and in areas endemic for TB like Iran.


Subject(s)
Mutation , Mycobacterium Infections/genetics , Receptors, Interleukin-12/genetics , Child , Child, Preschool , Female , Genetic Predisposition to Disease , Humans , Iran , Male , Mycobacterium/isolation & purification , Mycobacterium Infections/immunology , Mycobacterium tuberculosis/isolation & purification , Pedigree , Receptors, Interleukin-12/immunology , Tuberculosis/genetics , Tuberculosis/microbiology
3.
Hum Mutat ; 38(10): 1286-1296, 2017 10.
Article in English | MEDLINE | ID: mdl-28744922

ABSTRACT

IFN-γ signaling is essential for the innate immune defense against mycobacterial infections. IFN-γ signals through the IFN-γ receptor, which consists of a tetramer of two IFN-γR1 chains in complex with two IFN-γR2 chains, where IFN-γR1 is the ligand-binding chain of the interferon-γ receptor and IFN-γR2 is the signal-transducing chain of the IFN-γ receptor. Germline mutations in the gene IFNGR1 encoding the IFN-γR1 cause a primary immunodeficiency that mainly leads to mycobacterial infections. Here, we review the molecular basis of this immunodeficiency in the 130 individuals described to date, and report mutations in five new individuals, bringing the total number to 135 individuals from 98 kindreds. Forty unique IFNGR1 mutations have been reported and they exert either an autosomal dominant or an autosomal recessive effect. Mutations resulting in premature stopcodons represent the majority of IFNGR1 mutations (60%; 24 out of 40), followed by amino acid substitutions (28%, 11 out of 40). All known mutations, as well as 287 other variations, have been deposited in the online IFNGR1 variation database (www.LOVD.nl/IFNGR1). In this article, we review the function of IFN-γR1 and molecular genetics of human IFNGR1.


Subject(s)
Germ-Line Mutation/genetics , Immunologic Deficiency Syndromes/genetics , Mycobacterium Infections/genetics , Receptors, Interferon/genetics , Amino Acid Substitution/genetics , Genetic Predisposition to Disease , Humans , Interferon-gamma/genetics , Mycobacterium Infections/microbiology , Interferon gamma Receptor
4.
Clin Infect Dis ; 65(3): 518-521, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28430999

ABSTRACT

Nontuberculous mycobacterial infections due to autoantibodies targeting interferon-γ are an emerging medical problem. However, case finding is hampered due to highly complex diagnostic procedures not available in routine laboratories. We show that QuantiFERON assays can be exploited as a simple screening tool that may facilitate adequate and timely treatment.


Subject(s)
Antibodies, Neutralizing/blood , Autoantibodies/blood , Interferon-gamma Release Tests/methods , Interferon-gamma/immunology , Mycobacterium Infections, Nontuberculous , Antibodies, Neutralizing/immunology , Autoantibodies/immunology , Female , Humans , Middle Aged , Mycobacterium Infections, Nontuberculous/diagnosis , Mycobacterium Infections, Nontuberculous/immunology , Nontuberculous Mycobacteria
5.
Clin Immunol ; 180: 111-119, 2017 07.
Article in English | MEDLINE | ID: mdl-28487087

ABSTRACT

Elderly with late-onset recurrent respiratory tract infections (RRTI) often have specific anti-polysaccharide antibody deficiency (SPAD). We hypothesized that late-onset RRTI is caused by mild immunodeficiencies, such as SPAD, that remain hidden through adult life. We analyzed seventeen elderly RRTI patients and matched controls. We determined lymphocyte subsets, expression of BAFF receptors, serum immunoglobulins, complement pathways, Pneumovax-23 vaccination response and genetic variations in BAFFR and MBL2. Twelve patients (71%) and ten controls (59%) had SPAD. IgA was lower in patients than in controls, but other parameters did not differ. However, a high percentage of both patients (53%) and controls (65%) were MBL deficient, much more than in the general population. Often, MBL2 secretor genotypes did not match functional deficiency, suggesting that functional MBL deficiency can be an acquired condition. In conclusion, we found SPAD and MBL deficiency in many elderly, and conjecture that at least the latter arises with age.


Subject(s)
Aging/immunology , Immunologic Deficiency Syndromes/immunology , Respiratory Tract Infections/immunology , Aged , Aged, 80 and over , Aging/blood , Aging/genetics , B-Cell Activation Factor Receptor/genetics , B-Cell Activation Factor Receptor/immunology , Cell Differentiation , Complement Pathway, Alternative , Complement Pathway, Classical , Complement System Proteins/analysis , Female , Humans , Immunoglobulins/blood , Immunologic Deficiency Syndromes/blood , Immunologic Deficiency Syndromes/genetics , Lymphocytes/cytology , Lymphocytes/immunology , Male , Mannose-Binding Lectin/blood , Mannose-Binding Lectin/deficiency , Mannose-Binding Lectin/genetics , Mannose-Binding Lectin/immunology , Metabolism, Inborn Errors/blood , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/immunology , Middle Aged , Pneumococcal Vaccines/therapeutic use , Recurrence , Respiratory Tract Infections/blood , Respiratory Tract Infections/genetics , Vaccination
6.
Infection ; 45(6): 917-920, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28822097

ABSTRACT

CASE PRESENTATION: We analysed a 38-year-old woman with disseminated histoplasmosis for primary immunodeficiency. Her blood showed no IFN-γ response while her peripheral blood mononuclear cells (PBMCs) did. We identified IFN-γ autoantibodies of the IgG class in her serum. CONCLUSION: IFN-γ autoantibodies leading to infections were so far mainly detected in people from Asian descent, where it was found to be associated with certain HLA types. This may be the first patient of African descent, and without the typical HLA types that predispose to this problem, that produces IFN-γ autoantibodies.


Subject(s)
Autoantibodies/immunology , Histoplasma/immunology , Histoplasmosis/diagnosis , Interferon-gamma/immunology , Lymphadenitis/diagnosis , Adult , Female , Histoplasma/isolation & purification , Histoplasmosis/immunology , Histoplasmosis/microbiology , Humans , Leukocytes, Mononuclear/immunology , Lymphadenitis/immunology , Lymphadenitis/microbiology , Netherlands
7.
J Clin Immunol ; 36(3): 195-203, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26931784

ABSTRACT

PURPOSE: Complete interferon-γ receptor 1 (IFN-γR1) deficiency is a primary immunodeficiency causing predisposition to severe infection due to intracellular pathogens. Only 36 cases have been reported worldwide. The purpose of this article is to describe a large novel deletion found in 3 related cases, which resulted in the complete removal of the IFNGR1 gene. METHODS: Whole blood from three patients was stimulated with lipopolysaccharide (LPS) and IFN-γ to determine production of tumor necrosis factor (TNF), interleukin-12 p40 (IL-12p40) and IL-10. Expression of IFN-γR1 on the cell membrane of patients' monocytes was assessed using flow cytometry. IFNGR1 transcript was analyzed in RNA and the gene and adjacent regions were analyzed in DNA. Finally, IL22RA2 transcript levels were analyzed in whole blood cells and dendritic cells. RESULTS: There was no expression of the IFN-γR1 on the monocytes. Consistent with this finding, there was no IFN-γ response in the whole blood assay as measured by effect on LPS-induced IL-12p40, TNF and IL-10 production. A 119.227 nt homozygous deletion on chromosome 6q23.3 was identified, removing the IFNGR1 gene completely and ending 117 nt upstream of the transcription start of the IL22RA2 gene. Transcript levels of IL22RA2 were similar in patient and control. CONCLUSIONS: We identified the first large genomic deletion of IFNGR1 causing complete IFN-γR1 deficiency. Despite the deletion ending very close to the IL22RA2 gene, it does not appear to affect IL22RA2 transcription and, therefore, may not have any additional clinical consequence.


Subject(s)
Gene Deletion , Immunologic Deficiency Syndromes/genetics , Opportunistic Infections/genetics , RNA, Messenger/genetics , Receptors, Interferon/genetics , Receptors, Interleukin/genetics , Adult , Blood Cells/drug effects , Blood Cells/immunology , Blood Cells/pathology , Child, Preschool , Chromosomes, Human, Pair 6 , Dendritic Cells/immunology , Dendritic Cells/pathology , Female , Gene Expression Regulation , Homozygote , Humans , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/physiopathology , Infant , Interferon-gamma/pharmacology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-12 Subunit p40/genetics , Interleukin-12 Subunit p40/immunology , Lipopolysaccharides/pharmacology , Opportunistic Infections/immunology , Opportunistic Infections/physiopathology , Pedigree , Primary Cell Culture , RNA, Messenger/immunology , Receptors, Interferon/deficiency , Receptors, Interferon/immunology , Receptors, Interleukin/immunology , Sequence Analysis, DNA , Transcription, Genetic , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Interferon gamma Receptor
8.
PLoS Pathog ; 10(10): e1004485, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25356988

ABSTRACT

The anti-tuberculosis-vaccine Bacillus Calmette-Guérin (BCG) is the most widely used vaccine in the world. In addition to its effects against tuberculosis, BCG vaccination also induces non-specific beneficial effects against certain forms of malignancy and against infections with unrelated pathogens. It has been recently proposed that the non-specific effects of BCG are mediated through epigenetic reprogramming of monocytes, a process called trained immunity. In the present study we demonstrate that autophagy contributes to trained immunity induced by BCG. Pharmacologic inhibition of autophagy blocked trained immunity induced in vitro by stimuli such as ß-glucans or BCG. Single nucleotide polymorphisms (SNPs) in the autophagy genes ATG2B (rs3759601) and ATG5 (rs2245214) influenced both the in vitro and in vivo training effect of BCG upon restimulation with unrelated bacterial or fungal stimuli. Furthermore, pharmacologic or genetic inhibition of autophagy blocked epigenetic reprogramming of monocytes at the level of H3K4 trimethylation. Finally, we demonstrate that rs3759601 in ATG2B correlates with progression and recurrence of bladder cancer after BCG intravesical instillation therapy. These findings identify a key role of autophagy for the nonspecific protective effects of BCG.


Subject(s)
Autophagy , BCG Vaccine/therapeutic use , Mycobacterium bovis/immunology , Polymorphism, Single Nucleotide , Urinary Bladder Neoplasms/drug therapy , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/therapeutic use , Administration, Intravesical , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Autophagy/genetics , Autophagy/immunology , Autophagy-Related Protein 5 , Autophagy-Related Proteins , BCG Vaccine/administration & dosage , Cytokines/metabolism , Humans , Kaplan-Meier Estimate , Microtubule-Associated Proteins/genetics , Monocytes/immunology , Neoplasm Recurrence, Local , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/virology , Vaccination , Vesicular Transport Proteins/genetics , beta-Glucans/metabolism
9.
Cytokine ; 77: 196-202, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26364993

ABSTRACT

Coxiella burnetii, the causative agent of Q fever, is recognized by TLR2. TLR10 can act as an inhibitory receptor on TLR2-derived immune responses. Therefore, we investigated the role of TLR10 on C. burnetii-induced cytokine production and assessed whether genetic polymorphisms in TLR10 influences the development of chronic Q fever. HEK293 cells, transfected with TLR2, TLR10 or TLR2/TLR10, and human peripheral blood mononuclear cells (PBMCs) in the presence of anti-TLR10, were stimulated with C. burnetii. In both assays, the absence of TLR10 resulted in increased cytokine responses after C. burnetii stimulation. In addition, the effect of single nucleotide polymorphisms (SNPs) in TLR10 was examined in healthy volunteers whose PBMCs were stimulated with C. burnetii Nine Mile or the Dutch outbreak isolate C. burnetii 3262. Individuals bearing SNPs in TLR10 displayed increased cytokine production upon C. burnetii 3262 stimulation. Furthermore, 139 chronic Q fever patients and 220 controls were genotyped for TLR10 N241H, I775V and I369L. None of these polymorphisms were associated with increased susceptibility to chronic Q fever. In conclusion, TLR10 has an inhibitory effect on in vitro cytokine production by C. burnetii, but the presence of TLR10 polymorphisms does not lead to an increased risk of developing chronic Q fever.


Subject(s)
Cytokines/metabolism , Polymorphism, Single Nucleotide , Q Fever/genetics , Toll-Like Receptor 10/genetics , Adult , Aged , Cells, Cultured , Coxiella burnetii/classification , Coxiella burnetii/physiology , Female , Gene Frequency , Genotype , HEK293 Cells , Host-Pathogen Interactions , Humans , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/microbiology , Male , Middle Aged , Q Fever/metabolism , Q Fever/microbiology , Risk Factors , Species Specificity , Young Adult
10.
J Infect Dis ; 212(5): 818-29, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-25722298

ABSTRACT

BACKGROUND: Q fever is an infection caused by Coxiella burnetii. Persistent infection (chronic Q fever) develops in 1%-5% of patients. We hypothesize that inefficient recognition of C. burnetii and/or activation of host-defense in individuals carrying genetic variants in pattern recognition receptors or adaptors would result in an increased likelihood to develop chronic Q fever. METHODS: Twenty-four single-nucleotide polymorphisms in genes encoding Toll-like receptors, nucleotide-binding oligomerization domain-like receptor-2, αvß3 integrin, CR3, and adaptors myeloid differentiation primary response protein 88 (MyD88), and Toll interleukin 1 receptor domain-containing adaptor protein (TIRAP) were genotyped in 139 patients with chronic Q fever and in 220 controls with cardiovascular risk-factors and previous exposure to C. burnetii. Associations between these single-nucleotide polymorphisms and chronic Q fever were assessed by means of univariate logistic regression models. Cytokine production in whole-blood stimulation assays was correlated with relevant genotypes. RESULTS: Polymorphisms in TLR1 (R80T), NOD2 (1007fsX1), and MYD88 (-938C>A) were associated with chronic Q fever. No association was observed for polymorphisms in TLR2, TLR4, TLR6, TLR8, ITGAV, ITGB3, ITGAM, and TIRAP. No correction for multiple testing was performed because only genes with a known role in initial recognition of C. burnetii were included. In the whole-blood assays, individuals carrying the TLR1 80R-allele showed increased interleukin 10 production with C. burnetii exposure. CONCLUSIONS: Polymorphisms in TLR1 (R80T), NOD2 (L1007fsX1), and MYD88 (-938C>A) are associated with predisposition to development of chronic Q fever. For TLR1, increased interleukin 10 responses to C. burnetii in individuals carrying the risk allele may contribute to the increased risk of chronic Q fever.


Subject(s)
Genetic Predisposition to Disease , Membrane Glycoproteins/genetics , Myeloid Differentiation Factor 88/genetics , Polymorphism, Single Nucleotide , Q Fever/immunology , Receptors, Interleukin-1/genetics , Receptors, Pattern Recognition/genetics , Aged , Coxiella burnetii/immunology , Female , Genetic Association Studies , Genotype , Humans , Male , Middle Aged
11.
J Clin Immunol ; 34(4): 436-43, 2014 May.
Article in English | MEDLINE | ID: mdl-24682681

ABSTRACT

OBJECTIVE: Patients with hypomorphic mutations in Nuclear Factor-κB Essential Modulator (NEMO) are immunodeficient (ID) and most display ectodermal dysplasia and anhidrosis (EDA). We compared cytokine production by NEMO-ID patients with and without EDA. METHODS: PBMCs of NEMO-ID patients, four with EDA carrying E315A, C417R, D311N and Q403X, and three without EDA carrying E315A, E311_L333del and R254G, were cultured with PHA, PHA plus IL-12p70, LPS, LPS plus IFN-γ, TNF and IL-1ß. The production of various cytokines was measured in the supernatants. Fifty-nine healthy individuals served as controls. RESULTS: PBMCs of NEMO-ID patients without EDA produce subnormal amounts of IFN-γ after stimulation with PHA, but normal amounts of IFN-γ after PHA plus IL-12p70. In contrast, IFN-γ production by patients with EDA was low in both cases. Patients with EDA also generate lower PHA-stimulated IL-10 and IL-1ß than controls, whereas the production of these cytokines by patients without EDA was normal. CONCLUSION: Responses of PBMCs in NEMO-ID patients with EDA to PHA with and without IL-12p70 appear less robust than in NEMO-ID patients without EDA. This possibly indicates a better preserved NEMO function in our patients without EDA.


Subject(s)
Ectodermal Dysplasia/immunology , Genetic Diseases, X-Linked/immunology , I-kappa B Kinase/immunology , Immunologic Deficiency Syndromes/immunology , Interferon-gamma/biosynthesis , Interleukin-12/pharmacology , Leukocytes, Mononuclear/drug effects , Adult , Case-Control Studies , Ectodermal Dysplasia/complications , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/pathology , Female , Genetic Diseases, X-Linked/complications , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/pathology , Humans , I-kappa B Kinase/genetics , Immunologic Deficiency Syndromes/complications , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/pathology , Interleukin-10/biosynthesis , Interleukin-1beta/biosynthesis , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/pathology , Lipopolysaccharides/pharmacology , Male , Middle Aged , Phytohemagglutinins/pharmacology , Primary Cell Culture , Primary Immunodeficiency Diseases , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology
12.
J Clin Immunol ; 34(1): 84-93, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24254535

ABSTRACT

PURPOSE: Poly-ostotic Langerhans Cell Histiocytosis (LCH) can be difficult to distinguish clinically and histologically from disseminated infection in manifesting specific subtypes of Mendelian Susceptibility to Mycobacterial Disease (MSMD). In MSMD-patients, dominant negative germline mutations in the IFN-γR1 gene, in particular in exon 6, lead to autosomal dominant IFN-γ receptor 1 deficiency (ADIFNGR1) and can mimic LCH. We hypothesized that similar defects might underlie the pathogenesis of LCH. METHODS: IFN-γR1 expression was immunohistochemically determined at disease onset in biopsies from 11 LCH-patients and four ADIFNGR1-patients. IFN-γR1 function was analyzed in 18 LCH-patients and 13 healthy controls by assessing the IFN-γ-induced upregulation of Fc-gamma-receptor I (FcγRI) expression on monocytes. Pro-inflammatory cytokine production was measured after stimulation of whole blood with LPS and IFN-γ. Exon 6 of the IFN-γR1 gene was sequenced in 67 LCH-patients to determine whether mutations were present. RESULTS: IFN-γR1 expression was high in three LCH-affected biopsies, similar to ADIFNGR1-affected biopsies, but varied from negative to moderate in eight other LCH-affected biopsies. No functional differences in IFN-γ signaling were detected between LCH-patients with active or non-active disease and healthy controls. No germline mutations in exon 6 of the IFN-γR1 gene were detected in any of the 67 LCH-patients. CONCLUSIONS: In contrast to ADIFNGR1-patients, IFN-γ signaling is fully functional in LCH-patients. Either performed before, during or after treatment, these non-invasive functional assays can distinguish LCH-patients from ADIFNGR1-patients and thereby facilitate correct therapy regimens for patients with recurrent osteolytic lesions.


Subject(s)
Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/metabolism , Mycobacterium Infections/genetics , Mycobacterium Infections/metabolism , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Child, Preschool , Diagnosis, Differential , Exons , Gene Expression , Genetic Predisposition to Disease , Germ Cells , Histiocytosis, Langerhans-Cell/diagnosis , Humans , Interferon-gamma/metabolism , Male , Mutation , Mycobacterium Infections/diagnosis , Organ Specificity , Signal Transduction , Interferon gamma Receptor
13.
Pediatr Dermatol ; 31(2): 236-40, 2014.
Article in English | MEDLINE | ID: mdl-23004925

ABSTRACT

Defects in the interleukin 12 (IL-12)/interferon gamma (IFN-γ) pathway result in Mendelian susceptibility to mycobacterial disease (MSMD). IL-12 receptor beta 1 (IL-12Rß1) deficiency, the most common form of MSMD, is associated with weakly virulent mycobacteria and salmonella. Infections in patients with this deficiency are extraintestinal, or septicemic, recurrent infections with nontyphoid salmonellae. Here we report a case of an IL-12Rß1 deficiency with cutaneous leukocytoclastic vasculitis due to Salmonella enteritidis.


Subject(s)
Receptors, Interleukin-12/deficiency , Salmonella enteritidis/isolation & purification , Vasculitis, Leukocytoclastic, Cutaneous/microbiology , Anti-Bacterial Agents/therapeutic use , Biopsy , Ceftriaxone/therapeutic use , Child, Preschool , Female , Genetic Predisposition to Disease , Humans , Vasculitis, Leukocytoclastic, Cutaneous/drug therapy , Vasculitis, Leukocytoclastic, Cutaneous/genetics
14.
Hum Mutat ; 34(10): 1329-39, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23864330

ABSTRACT

IL-12Rß1 deficiency is an autosomal recessive disorder characterized by predisposition to recurrent and/or severe infections caused by otherwise poorly pathogenic mycobacteria and salmonella. IL-12Rß1 is a receptor chain of both the IL-12 and the IL-23 receptor and deficiency of IL-12Rß1 thus abolishes both IL-12 and IL-23 signaling. IL-12Rß1 deficiency is caused by bi-allelic mutations in the IL12RB1 gene. Mutations resulting in premature stop codons, such as nonsense, frame shift, and splice site mutations, represent the majority of IL-12Rß1 deficiency causing mutations (66%; 46/70). Also every other morbid mutation completely inactivates the IL-12Rß1 protein. In addition to disease-causing mutations, rare and common variations with unknown functional effect have been reported in IL12RB1. All these variants have been deposited in the online IL12RB1 variation database (www.LOVD.nl/IL12RB1). In this article, we review the function of IL-12Rß1 and molecular genetics of human IL12RB1.


Subject(s)
Databases, Genetic , Mutation , Receptors, Interleukin-12/deficiency , Receptors, Interleukin-12/genetics , Founder Effect , Genes, Recessive , Genetic Association Studies , Genetic Predisposition to Disease , Genetic Testing , Humans , Penetrance , Polymorphism, Genetic , Receptors, Interleukin-12/metabolism
15.
Cytokine ; 61(2): 645-55, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23299081

ABSTRACT

Interleukin-12 (IL-12), IL-23 and interferon-γ (IFN-γ) are pivotal cytokines acting in concert with tumor necrosis factor (TNF) and IL-1ß to shape type I immune responses against bacterial pathogens. Recently, several groups reported that type I immunity can be inhibited by IFN-α/ß. Here we show the extent of the inhibitory effects of IFN-α and IFN-ß on the responsiveness of human monocytes to Toll like receptor-ligands and IFN-γ. Both IFN-α and IFN-ß strongly reduced the production of IL-12p40, IL-1ß and TNF and the IFN-γ induced CD54 and CD64 expression. High IFN-γ concentrations could not counterbalance the inhibitions and IFN-α still inhibited monocytes 24h after stimulation in vitro as well as in vivo in patients undergoing IFN-α treatment. Next, we explored the mechanism of inhibition. We confirm that IFN-α/ß interferes with the IFN-γR1 expression, by studying the kinetics of IFN-γR1 downregulation. However, IFN-γR1 downregulation occurred only after two hours of IFN-α/ß stimulation and was transient, which cannot explain the IFN-γ unresponsiveness observed directly and late after IFN-α/ß stimulation. Additional experiments indeed indicate that other mechanisms are involved. IFN-α may interfere with IFN-γ-elicited phosphorylation of signal transducer and activator of transcription 1 (STAT1). IFN-α may also activate methyltransferases which in turn reduce, at least partly, the TNF and IL-1ß production and CD54 expression. IFN-α also induces the protein inhibitor of activated STAT1 (PIAS1). In conclusion, IFN-α and IFN-ß strongly inhibit the IFN-γ responsiveness and the production of type I cytokines of monocytes, probably via various mechanisms. Our findings indicate that IFN-α/ß play a significant role in the immunopathogenesis of bacterial infections, for example Mycobacterium tuberculosis infection.


Subject(s)
Immunity/drug effects , Interferon-alpha/pharmacology , Interferon-beta/pharmacology , Monocytes/drug effects , Monocytes/immunology , Down-Regulation/drug effects , Humans , Intercellular Adhesion Molecule-1/metabolism , Interferon-beta/biosynthesis , Interferon-gamma/pharmacology , Interleukin-12 Subunit p40/biosynthesis , Ligands , Phosphorylation/drug effects , Protein Inhibitors of Activated STAT/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Receptors, IgG/metabolism , Repressor Proteins/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , Small Ubiquitin-Related Modifier Proteins/metabolism , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
16.
J Infect Dis ; 205(6): 934-43, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22301633

ABSTRACT

BACKGROUND: Candidemia is a severe invasive fungal infection with high mortality. Recognition of Candida species is mediated through pattern recognition receptors such as Toll-like receptors (TLRs). This study assessed whether genetic variation in TLR signaling influences susceptibility to candidemia. METHODS: Thirteen mostly nonsynonymous single nucleotide polymorphisms (SNPs) in genes encoding TLRs and signaling adaptors MyD88 and Mal/TIRAP were genotyped in 338 patients (237 white, 93 African American, 8 other race) with candidemia and 351 noninfected controls (263 white, 88 African American). The SNPs significant in univariate analysis were further analyzed with multivariable logistic regression to determine association with clinical outcomes. Functional consequences of these polymorphisms were assessed via in vitro stimulation assays. RESULTS: Analyses of TLR SNPs revealed that 3 TLR1 SNPs (R80T, S248N, I602S) were significantly associated with candidemia susceptibility in whites. This association was not found in African Americans, likely due to lower power in this smaller study population. Furthermore, these TLR1 polymorphisms displayed impaired cytokine release by primary monocytes. No associations with susceptibility to candidemia were observed for SNPs in TLR2, TLR4, TLR6, TLR9, MyD88, or TIRAP. CONCLUSIONS: Nonsynonymous SNPs in TLR1 are associated with impaired TLR1 function, decreased cytokine responses, and predisposition to candidemia in whites.


Subject(s)
Candidemia/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Toll-Like Receptor 1/genetics , Adult , Black or African American/genetics , Aged , Candidemia/ethnology , Female , Humans , Logistic Models , Male , Middle Aged , Multivariate Analysis , Prospective Studies , Signal Transduction , White People/genetics
17.
Clin Infect Dis ; 54(4): 502-10, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22144535

ABSTRACT

BACKGROUND: Candida bloodstream infections cause significant morbidity and mortality among hospitalized patients. Although clinical and microbiological factors affecting prognosis have been identified, the impact of genetic variation in the innate immune responses mediated by cytokines on outcomes of infection remains to be studied. METHODS: A cohort of 338 candidemia patients and 351 noninfected controls were genotyped for single-nucleotide polymorphisms (SNPs) in 6 cytokine genes (IFNG, IL10, IL12B, IL18, IL1ß, IL8) and 1 cytokine receptor gene (IL12RB1). The association of SNPs with both candidemia susceptibility and outcome were assessed. Concentrations of pro- and antiinflammatory cytokines were measured in in vitro peripheral blood mononuclear cell stimulation assays and in serum from infected patients. RESULTS: None of the cytokine SNPs studied were associated with susceptibility to candidemia. Persistent fungemia occurred in 13% of cases. In the multivariable model, persistent candidemia was significantly associated with (odds ratio [95% confidence interval]): total parenteral nutrition (2.79 [1.26-6.17]), dialysis dependence (3.76 [1.46-8.64]), and the SNPs IL10 rs1800896 (3.45 [1.33-8.93]) and IL12B rs41292470 (5.36 [1.51-19.0]). In vitro production capacity of interleukin-10 and interferon-γ was influenced by these polymorphisms, and significantly lower proinflammatory cytokine concentrations were measured in serum from patients with persistent fungemia. CONCLUSIONS: Polymorphisms in IL10 and IL12B that result in low production of proinflammatory cytokines are associated with persistent fungemia in candidemia patients. This provides insights for future targeted management strategies for patients with Candida bloodstream infections.


Subject(s)
Candidiasis, Invasive/immunology , Candidiasis, Invasive/pathology , Cytokines/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Adult , Aged , Candidiasis, Invasive/mortality , Cells, Cultured , Cohort Studies , Cytokines/metabolism , Female , Genetic Association Studies , Humans , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Prospective Studies , Treatment Outcome
20.
BMC Med Genet ; 13: 5, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22239941

ABSTRACT

BACKGROUND: There is reason to expect strong genetic influences on the risk of developing active pulmonary tuberculosis (TB) among latently infected individuals. Many of the genome wide linkage and association studies (GWAS) to date have been conducted on African populations. In order to identify additional targets in genetically dissimilar populations, and to enhance our understanding of this disease, we performed a multi-stage GWAS in a Southeast Asian cohort from Indonesia. METHODS: In stage 1, we used the Affymetrix 100 K SNP GeneChip marker set to genotype 259 Indonesian samples. After quality control filtering, 108 cases and 115 controls were analyzed for association of 95,207 SNPs. In stage 2, we attempted validation of 2,453 SNPs with promising associations from the first stage, in 1,189 individuals from the same Indonesian cohort, and finally in stage 3 we selected 251 SNPs from this stage to test TB association in an independent Caucasian cohort (n = 3,760) from Russia. RESULTS: Our study suggests evidence of association (P = 0.0004-0.0067) for 8 independent loci (nominal significance P < 0.05), which are located within or near the following genes involved in immune signaling: JAG1, DYNLRB2, EBF1, TMEFF2, CCL17, HAUS6, PENK and TXNDC4. CONCLUSIONS: Mechanisms of immune defense suggested by some of the identified genes exhibit biological plausibility and may suggest novel pathways involved in the host containment of infection with TB.


Subject(s)
Tuberculosis, Pulmonary/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Cohort Studies , Female , Genetic Predisposition to Disease , Genome, Human , Genome-Wide Association Study/methods , Genotype , Humans , Indonesia , Male , Middle Aged , Phenotype , Polymorphism, Single Nucleotide , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL