Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 169
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 15(8): 727-37, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24952505

ABSTRACT

Microbes or danger signals trigger inflammasome sensors, which induce polymerization of the adaptor ASC and the assembly of ASC specks. ASC specks recruit and activate caspase-1, which induces maturation of the cytokine interleukin 1ß (IL-1ß) and pyroptotic cell death. Here we found that after pyroptosis, ASC specks accumulated in the extracellular space, where they promoted further maturation of IL-1ß. In addition, phagocytosis of ASC specks by macrophages induced lysosomal damage and nucleation of soluble ASC, as well as activation of IL-1ß in recipient cells. ASC specks appeared in bodily fluids from inflamed tissues, and autoantibodies to ASC specks developed in patients and mice with autoimmune pathologies. Together these findings reveal extracellular functions of ASC specks and a previously unknown form of cell-to-cell communication.


Subject(s)
Apoptosis/immunology , Caspase 1/immunology , Cytoskeletal Proteins/immunology , Inflammation/immunology , Interleukin-1beta/immunology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Antibodies/immunology , Apoptosis Regulatory Proteins , Autoantibodies/immunology , Autoimmune Diseases/immunology , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Caspase 1/genetics , Caspase Inhibitors/pharmacology , Cell Communication/immunology , Cytoskeletal Proteins/genetics , Humans , Inflammasomes/immunology , Lysosomes/pathology , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Phagocytosis/immunology , Prions/chemistry , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Signal Transduction/immunology
2.
Ann Rheum Dis ; 83(2): 184-193, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-37890976

ABSTRACT

OBJECTIVES: Early diagnosis of inflammatory arthritis is critical to prevent joint damage and functional incapacities. However, the discrepancy between recommendations of early diagnosis and reality is remarkable. The Rheuma-VOR study aimed to improve the time to diagnosis of patients with early arthritis by coordinating cooperation between primary care physicians, specialists and patients in Germany. METHODS: This prospective non-randomised multicentre study involved 2340 primary care physicians, 72 rheumatologists, 4 university hospitals and 4 rheumatology centres in 4 German Federal States. The two coprimary endpoints (time to diagnosis and screening performance of primary care physicians) were evaluated for early versus late implementation phase. Additionally, time to diagnosis and secondary endpoints (decrease of disease activity, increase in quality of life and overall well-being, improvement of fatigue, depression, functional ability, and work ability, reduction in drug and medical costs and hospitalisation) were compared with a reference cohort of the German Rheumatism Research Centre (DRFZ) reflecting standard care. RESULTS: A total of 7049 patients were enrolled in the coordination centres and 1537 patients were diagnosed with a rheumatic disease and consented to further participation. A follow-up consultation after 1 year was realised in 592 patients. The time to diagnosis endpoint and the secondary endpoints were met. In addition, the calculation of cost-effectiveness shows that Rheuma-VOR has a dominant cost-benefit ratio compared with standard care. DISCUSSION: Rheuma-VOR has shown an improvement in rheumatological care, patient-reported outcome parameters and cost savings by coordinating the cooperation of primary care physicians, rheumatologists and patients, in a nationwide approach.


Subject(s)
Arthritis, Rheumatoid , Rheumatic Diseases , Humans , Arthritis, Rheumatoid/diagnosis , Quality of Life , Prospective Studies , Rheumatic Diseases/diagnosis , Rheumatic Diseases/therapy , Delivery of Health Care
3.
Int Arch Allergy Immunol ; 183(3): 337-349, 2022.
Article in English | MEDLINE | ID: mdl-34619682

ABSTRACT

INTRODUCTION: Primary immunodeficiencies (PIDs) are a heterogeneous group of disorders characterized by increased susceptibility to infections, immune dysregulation, and/or malignancy. Genetic studies, especially during the last decade, led to a better understanding of the pathogenesis of primary immunodeficiencies and contributed to their classification into distinct monogenic disorders falling under one of the >430 currently known inborn errors of immunity (IEI). The growing availability of molecular genetic testing resulted in the increasing identification of patients with IEI. Here, we evaluated the diagnostic yield and the clinical consequences of targeted next-generation sequencing (tNGS) in a cohort of 294 primary immunodeficiency patients, primarily consisting of cases with sporadic primary antibody deficiency. METHOD: We have custom designed a tNGS panel to sequence a cohort of PID patients. Agilent's HaloPlex Target Enrichment System for Illumina was used for DNA target enrichment. RESULTS: tNGS identified a definite or predicted pathogenic variant in 15.3% of patients. The highest diagnostic rate was observed among patients with combined immunodeficiency or immune dysregulation, for whom genetic diagnosis may affect therapeutic decision-making. CONCLUSION: Next-generation sequencing has changed diagnostic assignment and paved the way for targeted therapeutic intervention with agents directed at reverting the disease-causing molecular abnormality or its pathophysiological consequences. Therefore, such targeted therapies and identifying the genetic basis of PID can be essential for patients with manifested immune dysregulation as conventional immunomodulatory regimens may exert an immunosuppressive effect, aggravating their immunodeficiency or may only inadequately control autoimmune or lymphoproliferative manifestations.


Subject(s)
Immunologic Deficiency Syndromes , Primary Immunodeficiency Diseases , Cohort Studies , High-Throughput Nucleotide Sequencing , Humans , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/therapy , Primary Immunodeficiency Diseases/diagnosis , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/therapy
4.
Ann Rheum Dis ; 80(3): 392-399, 2021 03.
Article in English | MEDLINE | ID: mdl-33046446

ABSTRACT

OBJECTIVES: Treatment of rheumatic diseases requires immunomodulatory agents which can compromise antibody production. However, even in case of agents directly targeting B cells, a minority of patients develop hypogammaglobulinaemia, suggesting a genetic predisposition, which has not been investigated so far. The phenotypic overlap between primary immunodeficiency disorders (PIDs) and rheumatic diseases suggests a shared genetic basis, especially in case of patients with rheumatic diseases with hypogammaglobulinaemia. METHODS: 1008 patients with rheumatic diseases visiting the outpatient clinics of the Hannover University Hospital were screened for hypogammaglobulinaemia. Those with persistent hypogammaglobulinaemia and an equal number of patients without it underwent targeted next-generation sequencing, searching for variations in genes linked with hypogammaglobulinaemia in the context of PIDs. RESULTS: We identified 33 predicted pathogenic variants in 30/64 (46.9%) patients with persistent secondary hypogammaglobulinaemia. All 33 variants were monoallelic and 10 of them in 10/64 (15.6%) patients were found in genes associated with autosomal dominant PIDs. 2/64 (3.1%) patients harboured variants which were previously reported to cause PIDs. In the group without hypogammaglobulinaemia we identified seven monoallelic variants in 7/64 (10.9%), including a variant in a gene associated with an autosomal dominant PID. CONCLUSIONS: Approximately half of patients with persistent secondary hypogammaglobulinaemia harboured at least a variant in a PID gene. Despite the fact that previous immunomodulatory treatment is an exclusion criterion in the diagnosis of PIDs, we identified genetic variants that can account for PID in patients with clear rheumatic phenotypes who developed hypogammaglobulinaemia after the introduction of immunomodulatory treatment. Our data suggest the common genetic causes of primary and secondary hypogammaglobulinaemia.


Subject(s)
Agammaglobulinemia , Rheumatic Diseases , Agammaglobulinemia/genetics , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Phenotype , Rheumatic Diseases/genetics
5.
Ann Rheum Dis ; 80(12): 1537-1544, 2021 12.
Article in English | MEDLINE | ID: mdl-34226189

ABSTRACT

OBJECTIVES: The monoclonal anti-CD20 antibody rituximab is frequently applied in the treatment of lymphoma as well as autoimmune diseases and confers efficient depletion of recirculating B cells. Correspondingly, B cell-depleted patients barely mount de novo antibody responses during infections or vaccinations. Therefore, efficient immune responses of B cell-depleted patients largely depend on protective T cell responses. METHODS: CD8+ T cell expansion was studied in rituximab-treated rheumatoid arthritis (RA) patients and B cell-deficient mice on vaccination/infection with different vaccines/pathogens. RESULTS: Rituximab-treated RA patients vaccinated with Influvac showed reduced expansion of influenza-specific CD8+ T cells when compared with healthy controls. Moreover, B cell-deficient JHT mice infected with mouse-adapted Influenza or modified vaccinia virus Ankara showed less vigorous expansion of virus-specific CD8+ T cells than wild type mice. Of note, JHT mice do not have an intrinsic impairment of CD8+ T cell expansion, since infection with vaccinia virus induced similar T cell expansion in JHT and wild type mice. Direct type I interferon receptor signalling of B cells was necessary to induce several chemokines in B cells and to support T cell help by enhancing the expression of MHC-I. CONCLUSIONS: Depending on the stimulus, B cells can modulate CD8+ T cell responses. Thus, B cell depletion causes a deficiency of de novo antibody responses and affects the efficacy of cellular response including cytotoxic T cells. The choice of the appropriate vaccine to vaccinate B cell-depleted patients has to be re-evaluated in order to efficiently induce protective CD8+ T cell responses.


Subject(s)
Antirheumatic Agents/adverse effects , Arthritis, Rheumatoid/drug therapy , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunogenicity, Vaccine/immunology , Influenza Vaccines/immunology , Interferon Type I/immunology , Rituximab/adverse effects , Animals , Case-Control Studies , Cytokines/immunology , Histocompatibility Antigens Class I/immunology , Humans , Influenza Vaccines/therapeutic use , Influenza, Human/prevention & control , Mice , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/immunology , Vaccinia/immunology , Vaccinia virus/immunology
6.
J Immunol ; 203(4): 795-800, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31292215

ABSTRACT

Protein arginine deiminase (PAD) enzymes catalyze the conversion of protein-bound arginine into citrulline, an irreversible posttranslational modification with loss of a positive charge that can influence protein-protein interactions and protein structure. Protein arginine deiminase activity depends on high intracellular calcium concentrations occurring in dying cells. In this study, we demonstrate that protein citrullination is common during pyroptotic cell death in macrophages and that inhibition of PAD enzyme activity by Cl-amidine, a pan-PAD inhibitor, blocks NLRP3 inflammasome assembly and proinflammatory IL-1ß release in macrophages. Genetic deficiency of either PAD2 or PAD4 alone in murine macrophages does not impair IL-1ß release; however, pharmacological inhibition or small interfering RNA knockdown of PAD2 within PAD4-/- macrophages does. Our results suggest that PAD2 and 4 activity in macrophages is required for optimal inflammasome assembly and IL-1ß release, a finding of importance for autoimmune diseases and inflammation.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , Protein-Arginine Deiminases/metabolism , Animals , Citrullination/physiology , Humans , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis/physiology
7.
J Allergy Clin Immunol ; 146(4): 901-911, 2020 10.
Article in English | MEDLINE | ID: mdl-32278790

ABSTRACT

BACKGROUND: An increasing number of NFKB1 variants are being identified in patients with heterogeneous immunologic phenotypes. OBJECTIVE: To characterize the clinical and cellular phenotype as well as the management of patients with heterozygous NFKB1 mutations. METHODS: In a worldwide collaborative effort, we evaluated 231 individuals harboring 105 distinct heterozygous NFKB1 variants. To provide evidence for pathogenicity, each variant was assessed in silico; in addition, 32 variants were assessed by functional in vitro testing of nuclear factor of kappa light polypeptide gene enhancer in B cells (NF-κB) signaling. RESULTS: We classified 56 of the 105 distinct NFKB1 variants in 157 individuals from 68 unrelated families as pathogenic. Incomplete clinical penetrance (70%) and age-dependent severity of NFKB1-related phenotypes were observed. The phenotype included hypogammaglobulinemia (88.9%), reduced switched memory B cells (60.3%), and respiratory (83%) and gastrointestinal (28.6%) infections, thus characterizing the disorder as primary immunodeficiency. However, the high frequency of autoimmunity (57.4%), lymphoproliferation (52.4%), noninfectious enteropathy (23.1%), opportunistic infections (15.7%), autoinflammation (29.6%), and malignancy (16.8%) identified NF-κB1-related disease as an inborn error of immunity with immune dysregulation, rather than a mere primary immunodeficiency. Current treatment includes immunoglobulin replacement and immunosuppressive agents. CONCLUSIONS: We present a comprehensive clinical overview of the NF-κB1-related phenotype, which includes immunodeficiency, autoimmunity, autoinflammation, and cancer. Because of its multisystem involvement, clinicians from each and every medical discipline need to be made aware of this autosomal-dominant disease. Hematopoietic stem cell transplantation and NF-κB1 pathway-targeted therapeutic strategies should be considered in the future.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Heterozygote , Mutation , NF-kappa B p50 Subunit/genetics , Phenotype , Adult , Aged , Autoimmunity/genetics , Biological Variation, Population , Biomarkers , Disease Management , Female , Fluorescent Antibody Technique , Genetic Association Studies/methods , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Magnetic Resonance Imaging , Male , Middle Aged , Prognosis , Tomography, X-Ray Computed
8.
Clin Immunol ; 210: 108269, 2020 01.
Article in English | MEDLINE | ID: mdl-31683054

ABSTRACT

Genetic studies have led to identification of an increasing number of monogenic primary immunodeficiency disorders. Monoallelic pathogenic gain-of-function (GOF) variants in NFKBIA, the gene encoding IκBα, result in an immunodeficiency disorder, typically accompanied by anhidrotic ectodermal dysplasia (EDA). So far, 14 patients with immunodeficiency due to NFKBIA GOF mutations have been reported. In this study we report three patients from the same family with immunodeficiency, presenting with recurrent respiratory tract infections, bronchiectasis and viral skin conditions due to a novel pathogenic NFKBIA variant (c.106 T > G, p.Ser36Ala), which results in reduced IκBα degradation. Immunological investigations revealed inadequate antibody responses against vaccine antigens, despite hypergammaglobulinemia. Interestingly, none of the studied patients displayed features of EDA. Therefore, missense NFKBIA variants substituting serine 36 of IκBα, differ from the rest of pathogenic GOF NFKBIA variants in that they cause combined immunodeficiency, even in the absence of EDA.


Subject(s)
Gain of Function Mutation/genetics , Immunologic Deficiency Syndromes/diagnosis , Leukocytes, Mononuclear/immunology , Meningitis, Bacterial/diagnosis , NF-KappaB Inhibitor alpha/genetics , Neisseria meningitidis/physiology , Papillomaviridae/physiology , Pseudomonas Infections/diagnosis , Pseudomonas aeruginosa/physiology , Virus Diseases/immunology , Adult , Arthritis, Juvenile , Azithromycin/therapeutic use , Bronchiectasis , Cell Proliferation , Cells, Cultured , Child , Ectodermal Dysplasia , Gentamicins/therapeutic use , Humans , Immunologic Deficiency Syndromes/genetics , Male , Meningitis, Bacterial/drug therapy , Pedigree , Pseudomonas Infections/drug therapy , Virus Diseases/diagnosis , Warts , Young Adult
9.
Eur J Immunol ; 48(4): 696-704, 2018 04.
Article in English | MEDLINE | ID: mdl-29277896

ABSTRACT

IgG Fc receptors (FcγRs) and the C5a anaphylatoxin receptor (C5aR) were identified as key regulators of type II autoimmune injury in mice. However, and with respect to C5aR, the relative importance of C5a for IgG autoantibody-induced cellular destruction remained unclear. Using an experimental model of autoimmune hemolytic anemia (AIHA), we here report marked differences in the development of AIHA between mice lacking C5aR and C5-deficient (Hc0 ) strain, indicating a limited role of C5 in this type of C5aR-regulated disease. Ex-vivo-analyses of liver homogenates from anemic Hc0 mice demonstrate C5a-independent C5aR activation, upregulation of FcγR expression and amplification of erythrophagocytosis by macrophages. As assessed by pharmacological inhibition studies, targeting of C5aR, but not of C5, is effective in treating experimental AIHA. Collectively, these results define a previously unrecognized disease mechanism of C5aR activation in AIHA that does not necessarily involve C5 and C5a.


Subject(s)
Anemia, Hemolytic, Autoimmune/immunology , Autoimmunity/immunology , Complement C5a/deficiency , Kupffer Cells/immunology , Receptor, Anaphylatoxin C5a/metabolism , Receptors, IgG/immunology , Animals , Erythrocytes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/immunology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/genetics , Receptors, IgG/biosynthesis
10.
Clin Immunol ; 179: 1-7, 2017 06.
Article in English | MEDLINE | ID: mdl-28216420

ABSTRACT

Here we describe novel mutations in recombination activation gene 1 (RAG1) in a compound heterozygous male patient with combined T and B cell immunodeficiency (CID). Clinical manifestations besides antibody deficiency included airway infections, granulomatosis and autoimmune features. He died at the age of 37 due to PML caused by JC virus infection. By targeted next-generation sequencing we detected post mortem in this patient three mutations in RAG1. One allele harbored two novel mutations (c.1123C>G, p.H375D and c.1430delC, p.F478Sfs*14), namely a missense variant and a frameshift deletion, of which the latter leads to a truncated RAG1 protein. The other allele revealed a previously described missense mutation (c.1420C>T, p.R474C, rs199474678). Functional analysis of the p.R474C variant in an in vitro V(D)J recombination assay exhibited reduced recombination activity compared to a wild-type control. Our findings suggest that mutations in RAG1, specifically the p.R474C variant, can be associated with relatively mild clinical symptoms or delayed occurrence of T cell and B cell deficiencies but may predispose to PML.


Subject(s)
Homeodomain Proteins/genetics , Immunologic Deficiency Syndromes/genetics , Leukoencephalopathy, Progressive Multifocal/genetics , Adult , B-Lymphocytes/immunology , Biopsy , Brain/diagnostic imaging , Brain/pathology , Cell Proliferation , Humans , Immunoglobulins/blood , Immunologic Deficiency Syndromes/blood , Immunologic Deficiency Syndromes/diagnostic imaging , Immunologic Deficiency Syndromes/immunology , Leukoencephalopathy, Progressive Multifocal/blood , Leukoencephalopathy, Progressive Multifocal/diagnostic imaging , Leukoencephalopathy, Progressive Multifocal/immunology , Lymphocyte Count , Magnetic Resonance Imaging , Male , Mutation , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , V(D)J Recombination
11.
Cytokine ; 96: 71-74, 2017 08.
Article in English | MEDLINE | ID: mdl-28324805

ABSTRACT

We used whole exome sequencing to determine the genetic background of CVID in two non-consanguineous German families. We identified IFNK (interferon-kappa) as the only candidate gene that harbored truncating mutations in affected members from both families. One family segregated c.30_31insTGTT, a known frameshift variant, while the other family segregated the novel IFNK mutation p.K199X that creates a premature stop codon. We sequenced the whole coding region of IFNK in a further series of 167 CVID patients and 192 healthy controls. Frameshift mutation c.30_31insTGTT was identified in 12 cases and 17 controls (OR 0.79, 95% CI 0.33-1.81, p=0.79), whereas the p.K199X mutation remained restricted to the original family. No additional truncating variants were found. We conclude that, given their frequent occurrence in non-diseased family members and controls, it is unlikely that truncating variants in IFNK constitute a major factor in the development of CVID.


Subject(s)
Common Variable Immunodeficiency/genetics , Common Variable Immunodeficiency/immunology , Frameshift Mutation , Interferon Type I/genetics , Adult , Aged , Common Variable Immunodeficiency/congenital , Female , Genome, Human , Germany , Humans , Male , Middle Aged , Mutation , Sequence Analysis, DNA , Exome Sequencing
13.
Eur J Immunol ; 45(7): 2143-53, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25912155

ABSTRACT

Stromal interaction molecule 1 (STIM1)-dependent store operated calcium-entry (SOCE) through Orai1-mediated calcium (Ca(2+) ) influx is considered a major pathway of Ca(2+) signaling, serving T-cell, mast cell, and platelet responses. Here, we show that Orai1 is critical for neutrophil function. Orai1-deficient neutrophils present defects in fMLP and complement C5a-induced Ca(2+) influx and migration, although they respond normally to another chemoattractant, CXCL2. Up until now, no specific contribution of Orai1 independent from STIM1 or SOCE has been recognized in immune cells. Here, we observe that Orai1-deficient neutrophils exhibit normal STIM1-dependent SOCE and STIM1-deficient neutrophils respond to fMLP and C5a efficiently. Despite substantial cytokine production, Orai1(-/-) chimeric mice show impaired neutrophil recruitment in LPS-induced peritonitis. Moreover, Orai1 deficiency results in profoundly defective C5a-triggered neutrophil lung recruitment in hypersensitivity pneumonitis. Comparative evaluation of inflammation in Stim1(-/-) chimeras reveals a distinct pathogenic contribution of STIM1, including its involvement in IgG-induced C5a production. Our data establish Orai1 as key signal mediator of C5aR activation, contributing to inflammation by a STIM1-independent pathway of Ca(2+) -influx in neutrophils.


Subject(s)
Calcium Channels/immunology , Complement C5a/immunology , Inflammation/immunology , Neutrophil Infiltration/immunology , Animals , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , ORAI1 Protein , Receptor, Anaphylatoxin C5a/immunology
14.
Int Arch Allergy Immunol ; 171(2): 136-140, 2016.
Article in English | MEDLINE | ID: mdl-27902982

ABSTRACT

AIM OF STUDY: We used a triplex real-time polymerase chain reaction (PCR) to classify our common variable immunodeficiency (CVID) patients into distinct groups according to the amount of their T-cell receptor excision circles (TRECs) and κ-deleting recombination excision circles (KRECs). MATERIALS AND METHODS: TREC and KREC analysis was performed using a multiplex real-time PCR assay. The T- and B-lymphocyte subsets were measured by flow cytometry. RESULTS: The copy number of TRECs and KRECs was significantly reduced in CVID patients compared to healthy controls. The TREC copy number was inversely correlated with age in both healthy subjects and patients; however, the KREC copy number was inversely correlated with age only in CVID patients. Moreover, no association was seen between TREC/KREC copy number and clinical manifestations such as bronchiectasis, splenomegaly, granulomata, autoimmune cytopenias, organ-specific autoimmunity, enteropathy and lymphoid hyperplasia. CONCLUSION: TREC and KREC quantification might be a useful tool to differentiate between CVID and combined immunodeficiency, but considering the results of this study a classification of CVID patients in certain groups is hardly possible.


Subject(s)
Common Variable Immunodeficiency/genetics , Multiplex Polymerase Chain Reaction , Real-Time Polymerase Chain Reaction , Receptors, Antigen, T-Cell/genetics , Recombination, Genetic , Adult , Aged , Aged, 80 and over , Case-Control Studies , Common Variable Immunodeficiency/immunology , DNA Copy Number Variations , Female , Humans , Male , Middle Aged , Young Adult
15.
Virol J ; 13: 67, 2016 Apr 18.
Article in English | MEDLINE | ID: mdl-27091211

ABSTRACT

BACKGROUND: HIV-HCV co-infection is associated with accelerated progression to hepatic fibrosis, cirrhosis and hepatocellular carcinoma than HCV mono-infection. The contribution of innate immunity during HIV-HCV co-infection has been a relatively under-investigated area. Natural killer (NK) cells are pivotal sentinels of innate immunity against viruses and tumour cells. In this study we evaluated the effect of HIV-HCV co-infection on peripheral blood NK cell subsets with emphasis on the phenotype of CD56(bright) NK cells. METHODS: Sixty patients were included in the study; HIV mono-infected (n = 12), HCV mono-infected (n = 15), HCV-HIV co-infected (n = 21) and healthy controls (n = 16). PBMCs were isolated and immunophenotyping of NK cells was performed by flowcytometry. RESULTS: We observed an expansion of CD56(bright) NK cell subset in HIV-HCV co-infection as compared to healthy controls and HIV mono-infected group. All the infected groups had an upregulated expression of the activating receptor NKG2D on CD56(bright) NK cells in comparison to healthy controls while not differing amongst themselves. The expression of NKp46 in HIV-HCV co-infected group was significantly upregulated as compared to both HIV as well as HCV mono-infections while NKp30 expression in the HIV-HCV co-infected group significantly differed as compared to HIV mono-infection. The CD56(bright) NK cell subset was activated in HIV-HCV co-infection as assessed by the expression of CD69 as compared to healthy controls but was significantly downregulated in comparison to HIV mono-infection. CD95 expression on CD56(bright) NK cells followed the same pattern where there was an increased expression of CD95 in HIV mono-infection and HIV-HCV co-infection as compared to healthy controls. In contrast to CD69 expression, CD95 expression in HCV mono-infection was decreased when compared to HIV mono-infection and HIV-HCV co-infection. Finally, expression of CXCR3 on CD56(bright) NK cells was increased in HIV-HCV co-infection in comparison to HIV mono-infection while remaining similar to HCV mono-infection. CONCLUSION: Thus, HIV-HCV co-infection is able to modulate the phenotype of CD56(bright) NK cell subset in a unique way such that NKp46 and CXCR3 expressions are distinct for co-infection while both mono-infections have an additive effect on CD56(bright), CD69 with CD95 expressions. HCV mono-infection has a dominant effect on NKp30 expression while NKG2D and CD127 expressions remained same in all the groups.


Subject(s)
Antigens, CD/analysis , Coinfection/pathology , HIV Infections/complications , HIV Infections/pathology , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/pathology , Killer Cells, Natural/immunology , Adult , Aged , Female , Flow Cytometry , Humans , Immunophenotyping , Male , Middle Aged , Natural Cytotoxicity Triggering Receptor 1/analysis , Phenotype , Receptors, CXCR3/analysis
16.
Cell Mol Life Sci ; 72(16): 3037-49, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25939268

ABSTRACT

Natural killer (NK) cells are innate immune effectors that provide first line of defence against viruses. Human NK cells are heterogeneous in nature, and their functions rely on a dynamic balance between germ-line-encoded activating and inhibitory receptors. HIV-1 infection results in altered NK cell receptor repertoire and impaired effector functions including the ability to lyse virus-infected cells and secretion of antiviral cytokine IFN-γ. Over the last decade, additional NK cell subset-specific molecules have been identified, leading to emergence of a more complex cellular diversity than previously thought. Herein, we discuss NK cell subset redistribution, altered receptor repertoire and influence of interaction of polymorphic leucocyte antigen (HLA) and killer cell immunoglobulin-like receptors (KIR) on HIV-1 disease progression.


Subject(s)
HIV Infections/immunology , HIV Infections/physiopathology , HIV-1 , Immunity, Innate/immunology , Killer Cells, Natural/cytology , Receptors, KIR/immunology , Receptors, Natural Cytotoxicity Triggering/immunology , HLA Antigens/metabolism , Humans , Killer Cells, Natural/immunology , Lectins, C-Type/metabolism , Receptors, KIR/metabolism
17.
Nat Genet ; 39(9): 1065-7, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17660818

ABSTRACT

TREX1 acts in concert with the SET complex in granzyme A-mediated apoptosis, and mutations in TREX1 cause Aicardi-Goutières syndrome and familial chilblain lupus. Here, we report monoallelic frameshift or missense mutations and one 3' UTR variant of TREX1 present in 9/417 individuals with systemic lupus erythematosus but absent in 1,712 controls (P = 4.1 x 10(-7)). We demonstrate that two mutant TREX1 alleles alter subcellular targeting. Our findings implicate TREX1 in the pathogenesis of SLE.


Subject(s)
Exodeoxyribonucleases/genetics , Lupus Erythematosus, Cutaneous/genetics , Mutation , Phosphoproteins/genetics , 3' Untranslated Regions/genetics , Endosomes/metabolism , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/metabolism , Frameshift Mutation , Gene Frequency , Genetic Predisposition to Disease , Genotype , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Lupus Erythematosus, Cutaneous/enzymology , Mutation, Missense , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
18.
Article in German | MEDLINE | ID: mdl-27090247

ABSTRACT

BACKGROUND: Currently only vague estimates exist for the seroprevalence and vaccination status for viral hepatitis B (HBV) in refugees arriving in Germany during the current refugee crisis. OBJECTIVES: To assess the prevalence of hepatitis B in refugees arriving in northern Germany in 2015. METHODS: In a cross-sectional study in 793 patients from all age groups tests for serological markers of hepatitis B virus infection (HBsAg, anti-HBc) and liver enzymes (ALT, AST, bilirubin, γGT, alkaline phosphatase) were performed in August 2015 at six reception centers in northern Germany. In 258 patients anti-HBs antibodies were assessed additionally. RESULTS: Of the tested refugees, 76.7 % were male, the median age was 28.8 ± 11.4 years, and 7.8 % were children under the age of 18. The overall prevalence of HBsAg and total anti-HBc was 2.3 % and 14.0 % respectively (2.5 % and 14.5 % in men; 1.2 % and 13.5 % in women). Prevalence was highest in 35 to 49-year-old patients for HBsAg (3.1 %) and for refugees over 50 years for anti-HBc (38 %). No immunity to Hepatitis B was found in 62 %, 18.6 % had been vaccinated against Hepatitis B, while 50 % of children aged up to 15 years (n = 12) had been vaccinated. Positive predictive values of elevated AST and ALT for detection of HBsAg was 0 and 0.016, respectively. Only two patients with a positive HBsAg had elevated transaminases. CONCLUSIONS: This study showed a high prevalence of HBsAg in a German refugee sample in comparison to the general German population. Liver enzymes are not an appropriate tool for screening for hepatitis B virus infection.


Subject(s)
Hepatitis B/diagnosis , Hepatitis B/epidemiology , Refugees/statistics & numerical data , Transaminases/blood , Vaccination/statistics & numerical data , Adolescent , Adult , Age Distribution , Aged , Aged, 80 and over , Biomarkers/blood , Child , Child, Preschool , Clinical Enzyme Tests/statistics & numerical data , Female , Germany/epidemiology , Hepatitis B/prevention & control , Hepatitis B Vaccines/therapeutic use , Humans , Infant , Infant, Newborn , Liver Function Tests/methods , Liver Function Tests/statistics & numerical data , Male , Mass Screening/statistics & numerical data , Middle Aged , Prevalence , Prognosis , Reproducibility of Results , Risk Factors , Sensitivity and Specificity , Seroepidemiologic Studies , Sex Distribution , Young Adult
20.
Eur J Immunol ; 44(12): 3717-28, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25229755

ABSTRACT

Human blood NK cells exert strong cytotoxicity against transformed cells and produce different cytokines and chemokines with an important role in modulating immune responses. However, the nature of NK-cell function depends on NK-cell interaction with other immune cells. One type of immune cells that communicate with NK cells are 6-sulfo LacNAc DCs (slanDCs), which comprise a major subpopulation of proinflammatory human blood DCs. In this study, we investigated the molecular mechanisms by which slanDCs interact with NK cells. Our in vitro studies demonstrate that LPS-stimulated slanDCs enhance activation and function of NK cells essentially via membrane-bound TNF-α (mTNF-α). LPS stimulation upregulates expression of mTNF-α in slanDCs, and surface TNF receptor 2 (TNFR2) is upregulated on NK cells after coincubation with slanDCs. IL-12 secreted by slanDCs increases surface expression of TNFR2 in NK cells. TNFR2 signaling in NK cells leads to activation of NF-kB, a transcription factor for cytokines such as GM-CSF. GM-CSF provided by NK cells is responsible for enhancing IL-12 secretion in slanDCs. In conclusion, TNFR2 and IL-12 signaling, which support one another, enables slanDCs to enhance NK-cell function through mTNF-α, thereby regulating immune responses.


Subject(s)
Cell Membrane/immunology , Dendritic Cells/immunology , Interleukin-12/immunology , Killer Cells, Natural/immunology , Receptors, Tumor Necrosis Factor, Type II/immunology , Tumor Necrosis Factor-alpha/immunology , Amino Sugars/immunology , Dendritic Cells/cytology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Killer Cells, Natural/cytology , Lipopolysaccharides/pharmacology , Male , NF-kappa B/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Up-Regulation/drug effects , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL