Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters

Publication year range
1.
J Autoimmun ; 144: 103183, 2024 04.
Article in English | MEDLINE | ID: mdl-38401466

ABSTRACT

Chronic nonbacterial osteomyelitis (CNO), an autoinflammatory bone disease primarily affecting children, can cause pain, hyperostosis and fractures, affecting quality-of-life and psychomotor development. This study investigated CNO-associated variants in P2RX7, encoding for the ATP-dependent trans-membrane K+ channel P2X7, and their effects on NLRP3 inflammasome assembly. Whole exome sequencing in two related transgenerational CNO patients, and target sequencing of P2RX7 in a large CNO cohort (N = 190) were conducted. Results were compared with publicly available datasets and regional controls (N = 1873). Findings were integrated with demographic and clinical data. Patient-derived monocytes and genetically modified THP-1 cells were used to investigate potassium flux, inflammasome assembly, pyroptosis, and cytokine release. Rare presumably damaging P2RX7 variants were identified in two related CNO patients. Targeted P2RX7 sequencing identified 62 CNO patients with rare variants (32.4%), 11 of which (5.8%) carried presumably damaging variants (MAF <1%, SIFT "deleterious", Polyphen "probably damaging", CADD >20). This compared to 83 of 1873 controls (4.4%), 36 with rare and presumably damaging variants (1.9%). Across the CNO cohort, rare variants unique to one (Median: 42 versus 3.7) or more (≤11 patients) participants were over-represented when compared to 190 randomly selected controls. Patients with rare damaging variants more frequently experienced gastrointestinal symptoms and lymphadenopathy while having less spinal, joint and skin involvement (psoriasis). Monocyte-derived macrophages from patients, and genetically modified THP-1-derived macrophages reconstituted with CNO-associated P2RX7 variants exhibited altered potassium flux, inflammasome assembly, IL-1ß and IL-18 release, and pyroptosis. Damaging P2RX7 variants occur in a small subset of CNO patients, and rare P2RX7 variants may represent a CNO risk factor. Observations argue for inflammasome inhibition and/or cytokine blockade and may allow future patient stratification and individualized care.


Subject(s)
Inflammasomes , Osteomyelitis , Humans , Cytokines , Inflammasomes/genetics , Inflammasomes/metabolism , Osteomyelitis/genetics , Potassium , Pyroptosis , Receptors, Purinergic P2X7/genetics
2.
J Immunol ; 207(1): 55-64, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34135066

ABSTRACT

Effector CD4+ T lymphocytes contribute to inflammation and tissue damage in psoriasis, but the underlying molecular mechanisms remain poorly understood. The transcription factor CREMα controls effector T cell function in people with systemic autoimmune diseases. The inhibitory surface coreceptor PD-1 plays a key role in the control of effector T cell function and its therapeutic inhibition in patients with cancer can cause psoriasis. In this study, we show that CD4+ T cells from patients with psoriasis and psoriatic arthritis exhibit increased production of IL-17 but decreased expression of IL-2 and PD-1. In genetically modified mice and Jurkat T cells CREMα expression was linked to low PD-1 levels. We demonstrate that CREMα is recruited to the proximal promoter of PDCD1 in which it trans-represses gene expression and corecruits DNMT3a-mediating DNA methylation. As keratinocytes limit inflammation by PD-1 ligand expression and, in this study, reported reduced expression of PD-1 on CD4+ T cells is linked to low IL-2 and high IL-17A production, our studies reveal a molecular pathway in T cells from people with psoriasis that can deserve clinical exploitation.


Subject(s)
Arthritis, Psoriatic/immunology , CD4-Positive T-Lymphocytes/immunology , Cyclic AMP Response Element Modulator/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Humans , Mice , Mice, Inbred C57BL
3.
Clin Immunol ; 238: 108998, 2022 05.
Article in English | MEDLINE | ID: mdl-35398286

ABSTRACT

Deciphering signaling pathways that regulate the complex interplay between inflammation and cell death is a key challenge in understanding innate immune responses. Over recent years, receptor interacting protein (RIP) kinases have been described to regulate the interplay between inflammation and cell death. Whereas RIP1 and 3, the most well described members of the RIP kinase family, play important roles in necroptosis, RIP2's involvement in regulating inflammation, cell death processes and cancer is less well described and controversially discussed. Here, we demonstrate that RIP2 exerts immune regulatory functions by regulating mitochondrial damage and mitochondrial superoxide production in response to SV40 LT-induced genotoxic stress by the induction of ULK1-phosphorylation, therefore regulating the expression of interferon stimulated genes (ISGs) and NLRP3-inflammasome dependent IL-1ß release. Because RIP2 is upregulated and/or activated in autoimmune/inflammatory disease and cancer, observations from this study promise implications of RIP kinases in human disease.


Subject(s)
Inflammation , Receptor-Interacting Protein Serine-Threonine Kinase 2 , DNA Damage , Homeostasis , Humans , Reactive Oxygen Species/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism
4.
J Immunol ; 203(11): 2807-2816, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31653682

ABSTRACT

Effector CD4+ T cells with increased IL-17A and reduced IL-2 production contribute to tissue inflammation and organ damage in systemic lupus erythematosus (SLE). Increased expression of the transcription factor cAMP response element modulator (CREM) α promotes altered cytokine expression in SLE. The aim of this study was to investigate CREMα-mediated events favoring effector CD4+ T cells in health and disease. Using CRISPR/Cas9 genome editing and lentiviral transduction, we generated CREMα-deficient and CREMα-overexpressing Jurkat T cells. Gene expression and regulatory events were assessed using luciferase reporter assays and chromatin immunoprecipitation. Interaction between CREMα and p300 was investigated using proximity ligation assays, coimmunoprecipitation, and knockdown of p300. Gene expression profiles of modified cells were compared with CD4+ T cells from patients with juvenile-onset SLE. We show that CREMα induces dual specificity protein phosphatase (DUSP) 4 in effector CD4+ T cells through corecruitment of p300. The transcriptional coactivator p300 mediates histone acetylation at DUSP4, prompting increased gene expression. Using DUSP4 transfection models and genetically modified CREM-deficient and CREMα-overexpressing T cells, we demonstrate the molecular underpinnings by which DUSP4 induces IL-17A while limiting IL-2 expression. We demonstrate that CD4+ T cells from patients with juvenile-onset SLE share phenotypical features with CREMα-overexpressing CD4+ T cells, including increased DUSP4 expression and imbalanced IL-17A and IL-2 production. Taken together, we describe CREMα-mediated mechanisms that involve the transcriptional upregulation of DUSP4, leading to imbalanced cytokine production by effector T cells. Our findings identify the CREMα/DUSP4 axis as a promising candidate in the search for biomarkers and therapeutic targets in SLE.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Cyclic AMP Response Element Modulator/metabolism , Dual-Specificity Phosphatases/metabolism , Lupus Erythematosus, Systemic/metabolism , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Humans , Jurkat Cells/metabolism , Species Specificity
5.
J Allergy Clin Immunol ; 142(6): 1956-1967.e6, 2018 12.
Article in English | MEDLINE | ID: mdl-29778503

ABSTRACT

BACKGROUND: Monogenic autoinflammatory disorders are characterized by dysregulation of the innate immune system, for example by gain-of-function mutations in inflammasome-forming proteins, such as NOD-like receptor family CARD-containing 4 protein (NLRC4). OBJECTIVE: Here we investigate the mechanism by which a novel mutation in the leucine-rich repeat (LRR) domain of NLRC4 (c.G1965C, p.W655C) contributes to autoinflammatory disease. METHODS: We studied 2 unrelated patients with early-onset macrophage activation syndrome harboring the same de novo mutation in NLRC4. In vitro inflammasome complex formation was quantified by using flow cytometric analysis of apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) specks. Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 techniques and lentiviral transduction were used to generate THP-1 cells with either wild-type or mutant NLRC4 cDNA. Cell death and release of IL-1ß/IL-18 were quantified by using flow cytometry and ELISA, respectively. RESULTS: The p.W655C NLRC4 mutation caused increased ASC speck formation, caspase-1-dependent cell death, and IL-1ß/IL-18 production. ASC contributed to p.W655C NLRC4-mediated cytokine release but not cell death. Mutation of p.W655 activated the NLRC4 inflammasome complex by engaging with 2 interfaces on the opposing LRR domain of the oligomer. One key set of residues (p.D1010, p.D1011, p.L1012, and p.I1015) participated in LRR-LRR oligomerization when triggered by mutant NLRC4 or type 3 secretion system effector (PrgI) stimulation of the NLRC4 inflammasome complex. CONCLUSION: This is the first report of a mutation in the LRR domain of NLRC4 causing autoinflammatory disease. c.G1965C/p.W655C NLRC4 increased inflammasome activation in vitro. Data generated from various NLRC4 mutations provides evidence that the LRR-LRR interface has an important and previously unrecognized role in oligomerization of the NLRC4 inflammasome complex.


Subject(s)
CARD Signaling Adaptor Proteins/genetics , Calcium-Binding Proteins/genetics , Inflammasomes/genetics , Leucine , CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/immunology , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/immunology , Female , HEK293 Cells , Humans , Infant , Infant, Newborn , Inflammasomes/chemistry , Inflammasomes/immunology , Macrophage Activation , Male , Protein Domains , Syndrome , THP-1 Cells
6.
Curr Osteoporos Rep ; 15(6): 542-554, 2017 12.
Article in English | MEDLINE | ID: mdl-29080202

ABSTRACT

PURPOSE OF REVIEW: Chronic non-bacterial osteomyelitis (CNO) with its most severe form chronic recurrent multifocal osteomyelitis (CRMO) is an autoinflammatory bone disorder. We summarize the clinical presentation, diagnostic approaches, most recent advances in understanding the pathophysiology, and available treatment options and outcomes in CNO/CRMO. RECENT FINDINGS: Though the exact molecular pathophysiology of CNO/CRMO remains somewhat elusive, it appears likely that variable defects in the TLR4/MAPK/inflammasome signaling cascade result in an imbalance between pro- and anti-inflammatory cytokine expressions in monocytes from CNO/CRMO patients. In this context, we present previously unpublished data on cytokine and chemokine expression in monocytes and tissues. CNO/CRMO is an autoinflammatory bone disorder resulting from imbalanced cytokine expression from innate immune cells. Though the exact molecular pathophysiology remains unclear, variable molecular defects appear to result in inflammasome activation and pro-inflammatory cytokine expression in monocytes from CNO/CRMO patients. Recent advances suggest signaling pathways and single molecules as biomarkers for CNO/CRMO as well as future treatment targets.


Subject(s)
Cytokines/immunology , Inflammasomes/immunology , Mitogen-Activated Protein Kinases/immunology , Monocytes/immunology , Osteomyelitis/immunology , Toll-Like Receptor 4/immunology , Adrenal Cortex Hormones/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Bone Density Conservation Agents/therapeutic use , Chemokine CCL2/immunology , Chemokine CCL4/immunology , Chemokine CCL5/immunology , Chemokines/immunology , Diphosphonates/therapeutic use , Disease Models, Animal , Humans , Immunosuppressive Agents/therapeutic use , Interleukin-12/immunology , Interleukin-6/immunology , Methotrexate/therapeutic use , Mice , Osteomyelitis/diagnosis , Osteomyelitis/therapy , Receptors, Interleukin-2/immunology , Signal Transduction , Sulfasalazine/therapeutic use , Tumor Necrosis Factor-alpha/antagonists & inhibitors
7.
J Immunol ; 192(9): 4379-85, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24706726

ABSTRACT

The proinflammatory enzyme caspase-1 plays an important role in the innate immune system and is involved in a variety of inflammatory conditions. Rare naturally occurring human variants of the caspase-1 gene (CASP1) lead to different protein expression and structure and to decreased or absent enzymatic activity. Paradoxically, a significant number of patients with such variants suffer from febrile episodes despite decreased IL-1ß production and secretion. In this study, we investigate how variant (pro)caspase-1 can possibly contribute to inflammation. In a transfection model, such variant procaspase-1 binds receptor interacting protein kinase 2 (RIP2) via Caspase activation and recruitment domain (CARD)/CARD interaction and thereby activates NF-κB, whereas wild-type procaspase-1 reduces intracellular RIP2 levels by enzymatic cleavage and release into the supernatant. We approach the protein interactions by coimmunoprecipitation and confocal microscopy and show that NF-κB activation is inhibited by anti-RIP2-short hairpin RNA and by the expression of a RIP2 CARD-only protein. In conclusion, variant procaspase-1 binds RIP2 and thereby activates NF-κB. This pathway could possibly contribute to proinflammatory signaling.


Subject(s)
Caspase 1/genetics , Fever/genetics , Inflammation/genetics , NF-kappa B/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Blotting, Western , Caspase 1/metabolism , Fever/enzymology , Fluorescent Antibody Technique , Gene Knockdown Techniques , Genetic Variation , HEK293 Cells , Humans , Immunoprecipitation , Inflammation/immunology , Inflammation/metabolism , Signal Transduction/physiology , Transduction, Genetic , Transfection
8.
Clin Immunol ; 160(2): 172-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26025004

ABSTRACT

Subcellular localization studies and life cell imaging approaches usually benefit from fusion-reporter proteins, such as enhanced green fluorescent protein (EGFP) and mCherry to the proteins of interest. However, such manipulations have several risks, including protein misfolding, altered protein shuttling, or functional impairment when compared to the wild-type proteins. Here, we demonstrate altered subcellular distribution and function of the pro-inflammatory enzyme procaspase-1 as a result of fusion with the reporter protein mCherry. Our observations are of central importance to further investigations of subcellular behavior and possible protein-protein interactions of naturally occurring genetic variants of human procaspase-1 which have recently been linked to autoinflammatory disorders.


Subject(s)
Caspase 1/metabolism , Fluorescent Dyes/metabolism , Green Fluorescent Proteins/metabolism , Luminescent Agents/metabolism , Luminescent Proteins/metabolism , Protein Transport , Enzyme Assays , Fluorescent Antibody Technique , HEK293 Cells , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Red Fluorescent Protein
9.
Curr Rheumatol Rep ; 17(7): 45, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26003867

ABSTRACT

Caspase-1 is a proinflammatory enzyme that is essential in many inflammatory conditions including infectious, autoimmune, and autoinflammatory disorders. The inflammation is mainly mediated by the generation of inflammasomes that activate caspase-1 and subsequently interleukin (IL)-1ß and IL-18. In addition, homotypic CARD/CARD interaction of procaspase-1 with RIP2 and thereby activation of the NF-κB pathways may play some role in the inflammation. However, normally, this pathway seems to become downregulated rapidly by the cleavage and excretion of RIP2 by active (pro-)caspase-1. In patients with unexplained recurrent systemic inflammation, CASP1 variants were detected, which often destabilized the caspase-1 dimer interface. Obviously, the resulting decreased or abrogated enzymatic activity and IL-1ß production did not prevent the febrile episodes. As an unexpected finding, the inactive procaspase-1 variants significantly enhanced proinflammatory signaling by increasing RIP2 mediated NF-κB activation in an in vitro cell transfection model. A likely reason is the failure of inactive procaspase-1 to cleave bound RIP2 and also to mediate its excretion out of the intracelluar space thereby keeping the RIP2-NF-κB pathway upregulated. Hence, proinflammatory effects of enzymatically inactive procaspase-1 variants may partially explain the inflammatory episodes of the patients.


Subject(s)
Autoimmune Diseases/genetics , Caspase 1/genetics , Fever/genetics , Inflammation/genetics , Autoimmune Diseases/metabolism , Caspase 1/metabolism , Fever/metabolism , Genetic Variation , Humans , Immunity, Innate , Inflammation/metabolism , Recurrence
10.
Hum Mutat ; 34(1): 122-31, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22833538

ABSTRACT

Caspase-1 (Interleukin-1 Converting Enzyme, ICE) is a proinflammatory enzyme that plays pivotal roles in innate immunity and many inflammatory conditions such as periodic fever syndromes and gout. Inflammation is often mediated by enzymatic activation of interleukin (IL)-1ß and IL-18. We detected seven naturally occurring human CASP1 variants with different effects on protein structure, expression, and enzymatic activity. Most mutations destabilized the caspase-1 dimer interface as revealed by crystal structure analysis and homology modeling followed by molecular dynamics simulations. All variants demonstrated decreased or absent enzymatic and IL-1ß releasing activity in vitro, in a cell transfection model, and as low as 25% of normal ex vivo in a whole blood assay of samples taken from subjects with variant CASP1, a subset of whom suffered from unclassified autoinflammation. We conclude that decreased enzymatic activity of caspase-1 is compatible with normal life and does not prevent moderate and severe autoinflammation.


Subject(s)
Caspase 1/genetics , Caspase 1/metabolism , Genetic Variation , Interleukin-1beta/metabolism , Biocatalysis , Caspase 1/chemistry , Cell Line , Crystallography, X-Ray , Cytokines/blood , Cytokines/metabolism , DNA Mutational Analysis , Genetic Predisposition to Disease/genetics , HEK293 Cells , Humans , Inflammation/enzymology , Inflammation/genetics , Models, Molecular , Mutation , Protein Multimerization , Protein Structure, Tertiary
11.
J Clin Ultrasound ; 41(3): 187-90, 2013.
Article in English | MEDLINE | ID: mdl-23475499

ABSTRACT

Absence of the ductus venosus (ADV) is a rare vascular anomaly. Its prognosis depends on the pathway of the umbilical flow to the systemic venous circulation, and the presence or absence of associated structural or chromosomal anomalies, sometimes resulting in hydrops fetalis. In cases with isolated ADV in the absence of associated anomalies, survival rates are as high as 85%, depending on the shunt situation. Here, we report a patient with ADV and extrahepatic umbilical vein drainage with favorable outcome after intrauterine reversal of early signs of cardiac failure. Diagnosis was made after the appearance of moderate cardiomegaly in the 25th gestational week. Thus, in the case of cardiomegaly with or without further signs of cardiac failure, ultrasound imaging of the venous duct should be considered.


Subject(s)
Cardiomegaly/etiology , Heart Failure/etiology , Ultrasonography, Doppler , Ultrasonography, Prenatal , Umbilical Veins/abnormalities , Vascular Malformations/diagnostic imaging , Adult , Cardiomegaly/diagnostic imaging , Female , Heart Failure/diagnostic imaging , Humans , Infant, Newborn , Male , Pregnancy , Remission, Spontaneous , Umbilical Veins/diagnostic imaging , Vascular Malformations/complications
12.
Front Immunol ; 14: 1245876, 2023.
Article in English | MEDLINE | ID: mdl-37662940

ABSTRACT

Background: Psoriasis is an autoimmune/inflammatory disorder primarily affecting the skin. Chronic joint inflammation triggers the diagnosis of psoriatic arthritis (PsA) in approximately one-third of psoriasis patients. Although joint disease typically follows the onset of skin psoriasis, in around 15% of cases it is the initial presentation, which can result in diagnostic delays. The pathophysiological mechanisms underlying psoriasis and PsA are not yet fully understood, but there is evidence pointing towards epigenetic dysregulation involving CD4+ and CD8+ T-cells. Objectives: The aim of this study was to investigate disease-associated DNA methylation patterns in CD4+ T-cells from psoriasis and PsA patients that may represent potential diagnostic and/or prognostic biomarkers. Methods: PBMCs were collected from 12 patients with chronic plaque psoriasis and 8 PsA patients, and 8 healthy controls. CD4+ T-cells were separated through FACS sorting, and DNA methylation profiling was performed (Illumina EPIC850K arrays). Bioinformatic analyses, including gene ontology (GO) and KEGG pathway analysis, were performed using R. To identify genes under the control of interferon (IFN), the Interferome database was consulted, and DNA Methylation Scores were calculated. Results: Numbers and proportions of CD4+ T-cell subsets (naïve, central memory, effector memory, CD45RA re-expressing effector memory cells) did not vary between controls, skin psoriasis and PsA patients. 883 differentially methylated positions (DMPs) affecting 548 genes were identified between controls and "all" psoriasis patients. Principal component and partial least-squares discriminant analysis separated controls from skin psoriasis and PsA patients. GO analysis considering promoter DMPs delivered hypermethylation of genes involved in "regulation of wound healing, spreading of epidermal cells", "negative regulation of cell-substrate junction organization" and "negative regulation of focal adhesion assembly". Comparing controls and "all" psoriasis, a majority of DMPs mapped to IFN-related genes (69.2%). Notably, DNA methylation profiles also distinguished skin psoriasis from PsA patients (2,949 DMPs/1,084 genes) through genes affecting "cAMP-dependent protein kinase inhibitor activity" and "cAMP-dependent protein kinase regulator activity". Treatment with cytokine inhibitors (IL-17/TNF) corrected DNA methylation patterns of IL-17/TNF-associated genes, and methylation scores correlated with skin disease activity scores (PASI). Conclusion: DNA methylation profiles in CD4+ T-cells discriminate between skin psoriasis and PsA. DNA methylation signatures may be applied for quantification of disease activity and patient stratification towards individualized treatment.


Subject(s)
Arthritis, Psoriatic , Autoimmune Diseases , Psoriasis , Humans , Arthritis, Psoriatic/diagnosis , Arthritis, Psoriatic/genetics , Interleukin-17 , DNA Methylation , CD8-Positive T-Lymphocytes , Psoriasis/genetics , Cyclic AMP-Dependent Protein Kinases , CD4-Positive T-Lymphocytes
13.
Clin Immunol ; 145(1): 69-76, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22940633

ABSTRACT

Chronic recurrent multifocal osteomyelitis (CRMO) is an autoinflammatory bone disorder of unknown origin. We previously demonstrated that monocytes from CRMO patients fail to express the immune-modulatory cytokine interleukin-10 (IL-10) in a chromatin dependent manner. Here, we demonstrate that attenuated extracellular-signal regulated kinase (ERK)1 and 2 signaling in response to TLR4 activation results in failure to induce IL-10 expression in monocytes from CRMO patients. Attenuated ERK1/2 activation results in 1) reduced levels of Sp-1, a transcription factor that induces IL-10 expression in monocytes, and 2) impaired H3S10 phosphorylation of the IL10 promoter, an activating epigenetic mark. The pro-inflammatory cytokines tumor necrosis factor (TNF)α and IL-6 are not negatively affected, resulting in an imbalance towards pro-inflammatory cytokines. Thus, impaired ERK1/2 signaling with subsequently reduced Sp-1 expression and H3S10 phosphorylation of the IL10 promoter may centrally contribute to the pathophysiology of CRMO.


Subject(s)
Interleukin-10/metabolism , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Osteomyelitis/metabolism , Toll-Like Receptor 4/metabolism , Chromatin/genetics , Chromatin/immunology , Gene Expression , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-6/biosynthesis , Interleukin-6/immunology , MAP Kinase Signaling System/immunology , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Monocytes/immunology , Monocytes/metabolism , Osteomyelitis/genetics , Osteomyelitis/immunology , Phosphorylation , Primary Cell Culture , Promoter Regions, Genetic , Protein Kinases/genetics , Protein Kinases/immunology , Protein Kinases/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
14.
Fetal Diagn Ther ; 32(4): 256-61, 2012.
Article in English | MEDLINE | ID: mdl-22722771

ABSTRACT

INTRODUCTION: Idiopathic dilatation of the right atrium (IDRA) is a rare abnormality usually detected by chance at any time between antenatal and adult life. It is defined as isolated enlargement of the right atrium in the absence of other cardiac lesions causing right atrial dilatation. IDRA can be associated with atrial arrhythmia and systemic embolism. The clinical presentation shows high variability ranging from the lack of any symptoms up to cardiac failure. METHODS/RESULTS: We describe 2 children with antenatally diagnosed IDRA, the intrauterine course in 1 case, the postnatal management and its long-term follow-up. There has been no need for surgical intervention so far because of the lack of arrhythmias and no further progression of right atrial diameters. Thrombus formation in the right atrium, which is a potential risk for pulmonary embolism, led us to initiate anticoagulation in our cases to prevent such complications. Furthermore, we suggest one possible pathomechanism of congenital right atrial dilatation. CONCLUSION: Optimal management of severe IDRA depends on the individual case. Long-term follow-up of these patients is necessary to monitor a possible further progression of right atrial size and occurrence of arrhythmias. As a possible pathomechanism, a functional partial anomalous pulmonary venous insertion may imitate a structural abnormal pulmonary vein connection in some idiopathic cases of congenital right atrial dilatation.


Subject(s)
Heart Atria/abnormalities , Abnormalities, Multiple/diagnostic imaging , Abnormalities, Multiple/pathology , Abnormalities, Multiple/therapy , Cardiomegaly/etiology , Child Development , Dilatation, Pathologic/congenital , Dilatation, Pathologic/diagnostic imaging , Dilatation, Pathologic/physiopathology , Dilatation, Pathologic/therapy , Female , Foramen Ovale, Patent/diagnostic imaging , Foramen Ovale, Patent/pathology , Foramen Ovale, Patent/therapy , Heart Atria/diagnostic imaging , Heart Atria/pathology , Humans , Infant, Newborn , Pregnancy , Ultrasonography, Prenatal
15.
Front Cell Dev Biol ; 9: 746145, 2021.
Article in English | MEDLINE | ID: mdl-34746142

ABSTRACT

Background: Psoriasis is a T cell-mediated chronic autoimmune/inflammatory disease. While some patients experience disease limited to the skin (skin psoriasis), others develop joint involvement (psoriatic arthritis; PsA). In the absence of disease- and/or outcome-specific biomarkers, and as arthritis can precede skin manifestations, diagnostic and therapeutic delays are common and contribute to disease burden and damage accrual. Objective: Altered epigenetic marks, including DNA methylation, contribute to effector T cell phenotypes and altered cytokine expression in autoimmune/inflammatory diseases. This project aimed at the identification of disease-/outcome-specific DNA methylation signatures in CD8+ T cells from patients with psoriasis and PsA as compared to healthy controls. Method: Peripheral blood CD8+ T cells from nine healthy controls, 10 psoriasis, and seven PsA patients were collected to analyze DNA methylation marks using Illumina Human Methylation EPIC BeadChips (>850,000 CpGs per sample). Bioinformatic analysis was performed using R (minfi, limma, ChAMP, and DMRcate packages). Results: DNA methylation profiles in CD8+ T cells differentiate healthy controls from psoriasis patients [397 Differentially Methylated Positions (DMPs); 9 Differentially Methylated Regions (DMRs) when ≥CpGs per DMR were considered; 2 DMRs for ≥10 CpGs]. Furthermore, patients with skin psoriasis can be discriminated from PsA patients [1,861 DMPs, 20 DMRs (≥5 CpGs per region), 4 DMRs (≥10 CpGs per region)]. Gene ontology (GO) analyses considering genes with ≥1 DMP in their promoter delivered methylation defects in skin psoriasis and PsA primarily affecting the BMP signaling pathway and endopeptidase regulator activity, respectively. GO analysis of genes associated with DMRs between skin psoriasis and PsA demonstrated an enrichment of GABAergic neuron and cortex neuron development pathways. Treatment with cytokine blockers associated with DNA methylation changes [2,372 DMPs; 1,907 DMPs within promoters, 7 DMRs (≥5 CpG per regions)] affecting transforming growth factor beta receptor and transmembrane receptor protein serine/threonine kinase signaling pathways. Lastly, a methylation score including TNF and IL-17 pathway associated DMPs inverse correlates with skin disease activity scores (PASI). Conclusion: Patients with skin psoriasis exhibit DNA methylation patterns in CD8+ T cells that allow differentiation from PsA patients and healthy individuals, and reflect clinical activity of skin disease. Thus, DNA methylation profiling promises potential as diagnostic and prognostic tool to be used for molecular patient stratification toward individualized treatment.

16.
Cell Rep ; 30(8): 2501-2511.e5, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32101731

ABSTRACT

Pro-inflammatory caspase-1 is a key player in innate immunity. Caspase-1 processes interleukin (IL)-1ß and IL-18 to their mature forms and triggers pyroptosis. These caspase-1 functions are linked to its enzymatic activity. However, loss-of-function missense mutations in CASP1 do not prevent autoinflammation in patients, despite decreased IL-1ß production. In vitro data suggest that enzymatically inactive caspase-1 drives inflammation via enhanced nuclear factor κB (NF-κB) activation, independent of IL-1ß processing. Here, we report two mouse models of enzymatically inactive caspase-1-C284A, demonstrating the relevance of this pathway in vivo. In contrast to Casp1-/- mice, caspase-1-C284A mice show pronounced hypothermia and increased levels of the pro-inflammatory cytokines tumor necrosis factor alpha (TNF-α) and IL-6 when challenged with lipopolysaccharide (LPS). Caspase-1-C284A signaling is RIP2 dependent and mediated by TNF-α but independent of the NLRP3 inflammasome. LPS-stimulated whole blood from patients carrying loss-of-function missense mutations in CASP1 secretes higher amounts of TNF-α. Taken together, these results reveal non-canonical caspase-1 signaling in vivo.


Subject(s)
Caspase 1/metabolism , Inflammation/pathology , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Adolescent , Adult , Animals , Child , Child, Preschool , Genetic Loci , Genotype , HEK293 Cells , Heterozygote , Humans , Mice, Inbred C57BL , Mutation/genetics , Young Adult
17.
Mol Cell Biol ; 26(17): 6623-32, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16914744

ABSTRACT

Cybr (also known as Cytip, CASP, and PSCDBP) is an interleukin-12-induced gene expressed exclusively in hematopoietic cells and tissues that associates with Arf guanine nucleotide exchange factors known as cytohesins. Cybr levels are dynamically regulated during T-cell development in the thymus and upon activation of peripheral T cells. In addition, Cybr is induced in activated dendritic cells and has been reported to regulate dendritic cell (DC)-T-cell adhesion. Here we report the generation and characterization of Cybr-deficient mice. Despite the selective expression in hematopoietic cells, there was no intrinsic defect in T- or B-cell development or function in Cybr-deficient mice. The adoptive transfer of Cybr-deficient DCs showed that they migrated efficiently and stimulated proliferation and cytokine production by T cells in vivo. However, competitive stem cell repopulation experiments showed a defect in the abilities of Cybr-deficient T cells to develop in the presence of wild-type precursors. These data suggest that Cybr is not absolutely required for hematopoietic cell development or function, but stem cells lacking Cybr are at a developmental disadvantage compared to wild-type cells. Collectively, these data demonstrate that despite its selective expression in hematopoietic cells, the role of Cybr is limited or largely redundant. Previous in vitro studies using overexpression or short interfering RNA inhibition of the levels of Cybr protein appear to have overestimated its immunological role.


Subject(s)
Carrier Proteins/metabolism , Cell Differentiation , Cross-Priming/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Membrane Proteins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Carrier Proteins/genetics , Cell Differentiation/drug effects , Cytokines/pharmacology , Dendritic Cells/drug effects , Exons/genetics , Gene Expression Regulation/drug effects , Gene Targeting , Humans , Immunity, Innate/immunology , Lipopolysaccharides/immunology , Lymphocyte Subsets/immunology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Myeloid Cells/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocytes/drug effects
18.
Cardiol Young ; 19(5): 530-3, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19674502

ABSTRACT

Congenital arteriovenous malformations are rare causes of congestive cardiac failure in neonates. The most common sites are in the head and liver, but other sites include the thorax, the abdomen and the limbs. The onset of failure is usually not in the immediate neonatal period, but later on in life, albeit that lesions such as the arteriovenous malformation of the vein of Galen, and other arteriovenous malformations in different locations which produce high flow can present early. We describe here the first case, to the best of our knowledge, of prenatal detection of an intrathoracic arteriovenous malformation producing neonatal cardiac failure, which was successfully treated by surgery postnatally.


Subject(s)
Arteriovenous Fistula/complications , Arteriovenous Fistula/diagnosis , Brachiocephalic Veins , Heart Failure/etiology , Subclavian Artery , Humans , Infant, Newborn , Male
19.
Mol Cell Biol ; 24(11): 5039-49, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15143194

ABSTRACT

Janus kinases (Jaks) play an essential role in cytokine signaling and have been reported to regulate plasma membrane expression of their cognate receptors. In this study, we examined whether Jak3 and the common gamma chain (gamma(c)) reciprocally regulate their plasma membrane expression. In contrast to interleukin-2Ralpha, gamma(c) localized poorly to the plasma membrane and accumulated in endosomal-lysosomal compartments. However, gamma(c) was expressed at comparable levels on the surface of cells lacking Jak3, and plasma membrane turnover of gamma(c) was independent of Jak3. Nonetheless, overexpression of Jak3 enhanced accumulation of gamma(c) at the plasma membrane. Without gamma(c), Jak3 localized in the cytosol, whereas in the presence of the receptor, it colocalized with gamma(c) in endosomes and at the plasma membrane. Although the Jak FERM domain is necessary and sufficient for receptor binding, the requirement for full-length Jak3 in gamma(c) membrane trafficking was remarkably stringent; using truncation and deletion mutants, we showed that the entire Jak3 molecule was required, although kinase activity was not. Thus, unlike other cytokine receptors, gamma(c) does not require Jak3 for receptor membrane expression. However, full-length Jak3 is required for normal trafficking of this cytokine receptor/Jak pair, a finding that has important structural and clinical implications.


Subject(s)
Immunoglobulin gamma-Chains/metabolism , Molecular Chaperones/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/metabolism , Animals , COS Cells , HeLa Cells , Humans , Janus Kinase 3 , Protein Structure, Tertiary , Time Factors
20.
J Rheumatol ; 43(11): 1956-1964, 2016 11.
Article in English | MEDLINE | ID: mdl-27585682

ABSTRACT

Chronic nonbacterial osteomyelitis (CNO) is an autoinflammatory bone disorder, covering a clinical spectrum with asymptomatic inflammation of single bones at the one end, and chronic recurrent multifocal osteomyelitis (CRMO) at the other end. The exact molecular pathophysiology of CNO remains largely unknown. Provided familial clusters and the association with inflammatory disorders of the skin and intestine suggest a genetic predisposition. Recently, profound dysregulation of cytokine responses was demonstrated in CRMO. Failure to produce antiinflammatory cytokines interleukin (IL)-10 and IL-19 contributes to activation of inflammasomes and subsequent IL-1ß release. In IL-10-deficient and in CNO-prone chronic multifocal osteomyelitis mice, IL-1ß was linked to bone inflammation. Further, alterations to the gut microbiome were suggested in contributing to IL-1ß release from innate immune cells in mice, offering an interesting target in the search for molecular mechanisms in CNO. Here, we summarize clinical presentation and treatment options in CNO/CRMO, current pathophysiological concepts, available mouse models, and promising future scientific directions.


Subject(s)
Cytokines/metabolism , Osteomyelitis/etiology , Animals , Disease Models, Animal , Humans , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Mice , Osteomyelitis/drug therapy , Osteomyelitis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL