Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
mBio ; 14(5): e0206623, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37787552

ABSTRACT

IMPORTANCE: Pyrin, a unique cytosolic receptor, initiates inflammatory responses against RhoA-inactivating bacterial toxins and effectors like Yersinia's YopE and YopT. Understanding pyrin regulation is crucial due to its association with dysregulated inflammatory responses, including Familial Mediterranean Fever (FMF), linked to pyrin gene mutations. FMF mutations historically acted as a defense mechanism against plague. Negative regulation of pyrin through PKN phosphorylation is well established, with Yersinia using the YopM effector to promote pyrin phosphorylation and counteract its activity. This study highlights the importance of phosphoprotein phosphatase activity in positively regulating pyrin inflammasome assembly in phagocytic cells of humans and mice. Oligomeric murine pyrin has S205 phosphorylated before inflammasome assembly, and this study implicates the dephosphorylation of murine pyrin S205 by two catalytic subunits of PP2A in macrophages. These findings offer insights for investigating the regulation of oligomeric pyrin and the balance of kinase and phosphatase activity in pyrin-associated infectious and autoinflammatory diseases.


Subject(s)
Inflammasomes , Protein Processing, Post-Translational , Humans , Animals , Mice , Inflammasomes/metabolism , Pyrin/genetics , Pyrin/metabolism , Macrophages/metabolism , Phosphoprotein Phosphatases/genetics , Mutation
3.
Cell Death Dis ; 14(3): 213, 2023 03 25.
Article in English | MEDLINE | ID: mdl-36966139

ABSTRACT

Familial Mediterranean Fever (FMF) is the most common monogenic autoinflammatory disorder. FMF is caused by mutations in the MEFV gene, encoding pyrin, an inflammasome sensor. The best characterized pathogenic mutations associated with FMF cluster in exon 10. Yet, mutations have been described along the whole MEFV coding sequence. Exon 10 encodes the B30.2 domain of the pyrin protein, but the function of this human-specific domain remains unclear. Pyrin is an inflammasome sensor detecting RhoA GTPase inhibition following exposure to bacterial toxins such as TcdA. Here, we demonstrate that the B30.2 domain is dispensable for pyrin inflammasome activation in response to this toxin. Deletion of the B30.2 domain mimics the most typical FMF-associated mutation and confers spontaneous inflammasome activation in response to pyrin dephosphorylation. Our results indicate that the B30.2 domain is a negative regulator of the pyrin inflammasome that acts independently from and downstream of pyrin dephosphorylation. In addition, we identify the central helical scaffold (CHS) domain of pyrin, which lies immediately upstream of the B30.2 domain as a second regulatory domain. Mutations affecting the CHS domain mimic pathogenic mutations in the B30.2 domain and render the pyrin inflammasome activation under the sole control of the dephosphorylation. In addition, specific mutations in the CHS domain strongly increase the cell susceptibility to steroid catabolites, recently described to activate pyrin, in both a cell line model and in monocytes from genotype-selected FMF patients. Taken together, our work reveals the existence of two distinct regulatory regions at the C-terminus of the pyrin protein, that act in a distinct manner to regulate positively or negatively inflammasome activation. Furthermore, our results indicate that different mutations in pyrin regulatory domains have different functional impacts on the pyrin inflammasome which could contribute to the diversity of pyrin-associated autoinflammatory diseases.


Subject(s)
Familial Mediterranean Fever , Inflammasomes , Pyrin , Humans , Familial Mediterranean Fever/genetics , Familial Mediterranean Fever/metabolism , Inflammasomes/genetics , Inflammasomes/metabolism , Mutation , Pyrin/genetics , Pyrin/metabolism
4.
Cell Rep ; 41(2): 111472, 2022 10 11.
Article in English | MEDLINE | ID: mdl-36223753

ABSTRACT

The pyrin inflammasome acts as a guard of RhoA GTPases and is central to immune defenses against RhoA-manipulating pathogens. Pyrin activation proceeds in two steps. Yet, the second step is still poorly understood. Using cells constitutively activated for the pyrin step 1, a chemical screen identifies etiocholanolone and pregnanolone, two catabolites of testosterone and progesterone, acting at low concentrations as specific step 2 activators. High concentrations of these metabolites fully and rapidly activate pyrin, in a human specific, B30.2 domain-dependent manner and without inhibiting RhoA. Mutations in MEFV, encoding pyrin, cause two distinct autoinflammatory diseases pyrin-associated autoinflammation with neutrophilic dermatosis (PAAND) and familial Mediterranean fever (FMF). Monocytes from PAAND patients, and to a lower extent from FMF patients, display increased responses to these metabolites. This study identifies an unconventional pyrin activation mechanism, indicates that endogenous steroid catabolites can drive autoinflammation, through the pyrin inflammasome, and explains the "steroid fever" described in the late 1950s upon steroid injection in humans.


Subject(s)
Familial Mediterranean Fever , Inflammasomes , Pyrin , Etiocholanolone , Familial Mediterranean Fever/genetics , Familial Mediterranean Fever/metabolism , Humans , Inflammasomes/metabolism , Mutation , Pregnanolone , Progesterone , Pyrin/genetics , Pyrin/metabolism , Testosterone
5.
Methods Mol Biol ; 2523: 179-195, 2022.
Article in English | MEDLINE | ID: mdl-35759198

ABSTRACT

The pyrin inflammasome detects effectors and toxins that inhibit RhoA GTPases and triggers inflammatory cytokines release and a fast cell death termed pyroptosis. Ancient plague pandemics in the Mediterranean basin have selected in the human population pyrin variants that can trigger an autoinflammatory disease termed familial Mediterranean fever (FMF). In addition, distinct mutations in MEFV, the gene encoding pyrin, cause a different rare autoinflammatory disease termed pyrin-associated autoinflammation with neutrophilic dermatosis (PAAND). As of today, more than 385 MEFV variants have been described although for most of them, whether they are pathogenic variant or benign polymorphism is unknown.Here, we describe different methods using primary human monocytes or engineered monocytic cell lines to functionally characterize MEFV variants, determine their potential pathogenicity, and classify them as either FMF-like or PAAND-like variants.


Subject(s)
Familial Mediterranean Fever , Familial Mediterranean Fever/diagnosis , Familial Mediterranean Fever/genetics , Humans , Inflammasomes/metabolism , Monocytes/metabolism , Mutation , Pyrin/genetics , Pyroptosis
6.
J Exp Med ; 218(3)2021 03 01.
Article in English | MEDLINE | ID: mdl-33606008

ABSTRACT

Juvenile idiopathic arthritis is the most common chronic rheumatic disease in children, and its etiology remains poorly understood. Here, we explored four families with early-onset arthritis carrying homozygous loss-of-expression mutations in LACC1. To understand the link between LACC1 and inflammation, we performed a functional study of LACC1 in human immune cells. We showed that LACC1 was primarily expressed in macrophages upon mTOR signaling. We found that LACC1 deficiency had no obvious impact on inflammasome activation, type I interferon response, or NF-κB regulation. Using bimolecular fluorescence complementation and biochemical assays, we showed that autophagy-inducing proteins, RACK1 and AMPK, interacted with LACC1. Autophagy blockade in macrophages was associated with LACC1 cleavage and degradation. Moreover, LACC1 deficiency reduced autophagy flux in primary macrophages. This was associated with a defect in the accumulation of lipid droplets and mitochondrial respiration, suggesting that LACC1-dependent autophagy fuels macrophage bioenergetics metabolism. Altogether, LACC1 deficiency defines a novel form of genetically inherited juvenile arthritis associated with impaired autophagy in macrophages.


Subject(s)
Arthritis, Juvenile/metabolism , Arthritis, Juvenile/pathology , Autophagy , Intracellular Signaling Peptides and Proteins/deficiency , Macrophages/metabolism , Adenylate Kinase/metabolism , Adolescent , Amino Acid Sequence , Apoptosis/drug effects , Arthritis, Juvenile/genetics , Autophagy/drug effects , Autophagy/genetics , Autophagy-Related Proteins/metabolism , Bacteria/metabolism , Cell Differentiation/drug effects , Child , Exome/genetics , Female , Homozygote , Humans , Inflammasomes/metabolism , Inflammation/complications , Inflammation/pathology , Interferons/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Lipid Droplets/drug effects , Lipid Droplets/metabolism , Loss of Function Mutation/genetics , Lysosomes/drug effects , Lysosomes/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/drug effects , Male , Mitochondria/drug effects , Mitochondria/metabolism , Monocytes/drug effects , Monocytes/pathology , NF-kappa B/metabolism , Pedigree , Proteomics , Receptors for Activated C Kinase/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Young Adult
8.
Ann Rheum Dis ; 80(1): 128-132, 2021 01.
Article in English | MEDLINE | ID: mdl-33037005

ABSTRACT

BACKGROUND AND OBJECTIVE: Familial Mediterranean fever (FMF) is the most frequent hereditary autoinflammatory disease. Its diagnosis relies on a set of clinical criteria and a genetic confirmation on identification of biallelic pathogenic MEFV variants. MEFV encodes pyrin, an inflammasome sensor. Using a kinase inhibitor, UCN-01, we recently identified that dephosphorylation of FMF-associated pyrin mutants leads to inflammasome activation. The aim of this study was to assess whether quantifying UCN-01-mediated inflammasome activation could discriminate FMF patients from healthy donors (HD) and from patients with other inflammatory disorders (OID). METHODS: Real-time pyroptosis and IL-1ß secretion were monitored in response to UCN-01 in monocytes from FMF patients (n=67), HD (n=71) and OID patients (n=40). Sensitivity and specificity of the resulting diagnostic tests were determined by receiver operating characteristic curve analyses. RESULTS: Inflammasome monitoring in response to UCN-01 discriminates FMF patients from other individuals. Pyroptosis assessment leads to a fast FMF diagnosis while combining pyroptosis and IL-1ß dosage renders UCN-01-based assays highly sensitive and specific. UCN-01-triggered monocytes responses were influenced by MEFV gene dosage and MEFV mutations in a similar way as clinical phenotypes are. CONCLUSIONS: UCN-01-based inflammasome assays could be used to rapidly diagnose FMF, with high sensitivity and specificity.


Subject(s)
Familial Mediterranean Fever/diagnosis , Inflammasomes/drug effects , Interleukin-1beta/drug effects , Monocytes/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrin/drug effects , Pyroptosis/drug effects , Staurosporine/analogs & derivatives , Adolescent , Adult , Aged , Aged, 80 and over , Arthritis, Juvenile/diagnosis , Behcet Syndrome/diagnosis , Case-Control Studies , Child , Child, Preschool , Cryopyrin-Associated Periodic Syndromes/diagnosis , Familial Mediterranean Fever/genetics , Familial Mediterranean Fever/immunology , Female , Fever/diagnosis , Fever of Unknown Origin/diagnosis , Hereditary Autoinflammatory Diseases/diagnosis , Humans , Immunologic Tests/methods , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Lupus Erythematosus, Systemic/diagnosis , Male , Mevalonate Kinase Deficiency/diagnosis , Middle Aged , Monocytes/immunology , Monocytes/metabolism , Pyrin/genetics , Pyrin/immunology , Pyrin/metabolism , Sensitivity and Specificity , Sepsis/diagnosis , Staurosporine/pharmacology , Still's Disease, Adult-Onset/diagnosis , Young Adult
10.
EMBO Mol Med ; 11(11): e10547, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31589380

ABSTRACT

Familial Mediterranean fever (FMF) is the most frequent hereditary systemic autoinflammatory syndrome. FMF is usually caused by biallelic mutations in the MEFV gene, encoding Pyrin. Conclusive genetic evidence lacks for about 30% of patients diagnosed with clinical FMF. Pyrin is an inflammasome sensor maintained inactive by two kinases (PKN1/2). The consequences of MEFV mutations on inflammasome activation are still poorly understood. Here, we demonstrate that PKC superfamily inhibitors trigger inflammasome activation in monocytes from FMF patients while they trigger a delayed apoptosis in monocytes from healthy donors. The expression of the pathogenic p.M694V MEFV allele is necessary and sufficient for PKC inhibitors (or mutations precluding Pyrin phosphorylation) to trigger caspase-1- and gasdermin D-mediated pyroptosis. In line with colchicine efficacy in patients, colchicine fully blocks this response in FMF patients' monocytes. These results indicate that Pyrin inflammasome activation is solely controlled by Pyrin (de)phosphorylation in FMF patients while a second control mechanism restricts its activation in healthy donors/non-FMF patients. This study paves the way toward a functional characterization of MEFV variants and a functional test to diagnose FMF.


Subject(s)
Familial Mediterranean Fever/physiopathology , Inflammasomes/metabolism , Protein Processing, Post-Translational , Pyrin/metabolism , Cells, Cultured , Humans , Monocytes/drug effects , Monocytes/metabolism , Phosphorylation , Protein Kinase Inhibitors/metabolism , Pyroptosis
11.
Pathog Dis ; 76(3)2018 04 01.
Article in English | MEDLINE | ID: mdl-29718184

ABSTRACT

Numerous pathogens including Clostridium difficile and Yersinia pestis have evolved toxins or effectors targeting GTPases from the RhoA subfamily (RhoA/B/C) to inhibit or hijack the host cytoskeleton dynamics. The resulting impairment of RhoA GTPases activity is sensed by the host via an innate immune complex termed the pyrin inflammasome in which caspase-1 is activated. The cascade leading to activation of the pyrin inflammasome has been recently uncovered. In this review, following a brief presentation of RhoA GTPases-modulating toxins, we present the pyrin inflammasome and its regulatory mechanisms. Furthermore, we discuss how some pathogens have developed strategies to escape detection by the pyrin inflammasome. Finally, we present five monogenic autoinflammatory diseases associated with pyrin inflammasome deregulation. The molecular insights provided by the study of these diseases and the corresponding mutations on pyrin inflammasome regulation and activation are presented.


Subject(s)
Bacterial Toxins/immunology , Enterocolitis, Pseudomembranous/immunology , Inflammasomes/immunology , Plague/immunology , Pyrin/immunology , rhoA GTP-Binding Protein/immunology , Animals , Autoimmunity , Bacterial Toxins/biosynthesis , Clostridioides difficile/immunology , Clostridioides difficile/metabolism , Clostridioides difficile/pathogenicity , Enterocolitis, Pseudomembranous/microbiology , Enterocolitis, Pseudomembranous/pathology , Host-Pathogen Interactions/immunology , Humans , Inflammasomes/genetics , Inflammation , Isoenzymes/genetics , Isoenzymes/immunology , Plague/microbiology , Plague/pathology , Pyrin/genetics , Syndrome , Yersinia pestis/immunology , Yersinia pestis/metabolism , Yersinia pestis/pathogenicity , rhoA GTP-Binding Protein/genetics
12.
Nat Commun ; 9(1): 242, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29339744

ABSTRACT

Caspase-4/5 in humans and caspase-11 in mice bind hexa-acylated lipid A, the lipid moeity of lipopolysaccharide (LPS), to induce the activation of non-canonical inflammasome. Pathogens such as Francisella novicida express an under-acylated lipid A and escape caspase-11 recognition in mice. Here, we show that caspase-4 drives inflammasome responses to F. novicida infection in human macrophages. Caspase-4 triggers F. novicida-mediated, gasdermin D-dependent pyroptosis and activates the NLRP3 inflammasome. Inflammasome activation could be recapitulated by transfection of under-acylated LPS from different bacterial species or synthetic tetra-acylated lipid A into cytosol of human macrophage. Our results indicate functional differences between human caspase-4 and murine caspase-11. We further establish that human Guanylate-binding proteins promote inflammasome responses to under-acylated LPS. Altogether, our data demonstrate a broader reactivity of caspase-4 to under-acylated LPS than caspase-11, which may have important clinical implications for management of sepsis.


Subject(s)
Caspases, Initiator/metabolism , Caspases/metabolism , Francisella/metabolism , Lipopolysaccharides/metabolism , Macrophages/metabolism , Acylation , Animals , Caspases/genetics , Caspases, Initiator/genetics , Cells, Cultured , Cytosol/microbiology , Francisella/physiology , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , Macrophages/microbiology , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , RNA Interference , Species Specificity , U937 Cells
13.
Rheumatology (Oxford) ; 57(1): 100-111, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29040788

ABSTRACT

Objectives: FMF is the most frequent autoinflammatory disease and is associated in most patients with bi-allelic MEFV mutations. MEFV encodes Pyrin, an inflammasome sensor activated following RhoGTPase inhibition. The functional consequences of MEFV mutations on the ability of Pyrin variants to act as inflammasome sensors are largely unknown. The aim of this study was to assess whether MEFV mutations affect the ability of Pyrin to detect RhoGTPase inhibition and other inflammasome stimuli. Methods: IL-1ß and IL-18 released by monocytes from healthy donors (HDs) and FMF patients were measured upon specific engagement of the Pyrin, NLRP3 and NLRC4 inflammasomes. Cell death kinetics following Pyrin activation was monitored in real time. Results: Monocytes from FMF patients secreted significantly more IL-1ß and IL-18 and died significantly faster than HD monocytes in response to low concentrations of Clostridium difficile toxin B (TcdB), a Pyrin-activating stimulus. Monocytes from patients bearing two MEFV exon 10 pathogenic variants displayed an increased Pyrin inflammasome response compared with monocytes from patients with a single exon 10 pathogenic variant indicating a gene-dosage effect. Using a short priming step, the response of monocytes from FMF patients to NLRP3- and NLRC4-activating stimuli was normal indicating that MEFV mutations trigger a specific hypersensitivity of monocytes to low doses of a Pyrin-engaging stimulus. Conclusion: Contrary to the NLRP3 mutations described in cryopyrin-associated periodic syndrome, FMF-associated MEFV mutations do not lead to a constitutive activation of Pyrin. Rather, FMF-associated mutations are hypermorphic mutations that specifically decrease the activation threshold of the Pyrin inflammasome without affecting other canonical inflammasomes.


Subject(s)
CARD Signaling Adaptor Proteins/immunology , Calcium-Binding Proteins/immunology , Familial Mediterranean Fever/genetics , Monocytes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Pyrin/genetics , Adenosine Triphosphate/pharmacology , Adolescent , Adult , Antigens, Bacterial/pharmacology , Bacterial Proteins/pharmacology , Bacterial Toxins/pharmacology , Case-Control Studies , Cell Death , Child , Child, Preschool , Familial Mediterranean Fever/immunology , Female , Healthy Volunteers , Humans , Inflammasomes/genetics , Interleukin-18/immunology , Interleukin-1beta/immunology , Ionophores/pharmacology , Male , Middle Aged , Monocytes/drug effects , Mutation , Nigericin/pharmacology , Pyrin/immunology , Salmonella typhimurium , rho GTP-Binding Proteins
14.
Clin Rev Allergy Immunol ; 54(3): 454-479, 2018 Jun.
Article in English | MEDLINE | ID: mdl-28578473

ABSTRACT

The knowledge on systemic autoinflammatory disorders (SAID) is expanding rapidly and new signalling pathways are being decrypted. The concept of autoinflammation has been proposed since 1999, to define a group of diseases with abnormal innate immunity activation. Since then, more than 30 monogenic SAID have been described. In this review, we first describe inflammasomopathies and SAID related to the interleukin-1 pathway. Recent insights into the pathogenesis of familial Mediterranean fever and the function of Pyrin are detailed. In addition, complex or polygenic SAID, such as Still's disease or PFAPA syndrome, are also discussed. Then, major players driving autoinflammation, such as type-1 interferonopathies (including the recently described haploinsuffiency in A20 and otulipenia), TNF-associated periodic syndromes, defects in ubiquitination, and SAID with overlapping features of autoimmunity or immunodeficiency. Discoveries of the pathogenic role of mosaicism, intronic defects coupled to the likelihood to identify digenic or polygenic diseases are providing new challenges for physicians and geneticists. This comprehensive review depicts the various SAID, presenting them according to their predominant pathophysiological mechanism, with a particular emphasis on recent findings. Epidemiologic data are also presented. Finally, we propose a practical diagnostic approach to the most common monogenic SAID, based on the most characteristic clinical presentation of these disorders.


Subject(s)
Autoimmunity , Immune System Diseases/immunology , Immunologic Deficiency Syndromes/immunology , Gene-Environment Interaction , Humans , Immune System Diseases/genetics , Immunologic Deficiency Syndromes/genetics , Inflammation , Signal Transduction
15.
J Leukoc Biol ; 99(5): 761-9, 2016 05.
Article in English | MEDLINE | ID: mdl-26598380

ABSTRACT

TNFR-associated periodic syndrome is an autoinflammatory disorder caused by autosomal-dominant mutations in TNFRSF1A, the gene encoding for TNFR superfamily 1A. The lack of knowledge in the field of TNFR-associated periodic syndrome biology is clear, particularly in the context of control of immune self-tolerance. We investigated how TNF-α/TNFR superfamily 1A signaling can affect T cell biology, focusing on conventional CD4(+)CD25(-) and regulatory CD4(+)CD25(+) T cell functions in patients with TNFR-associated periodic syndrome carrying either high or low penetrance TNFRSF1A mutations. Specifically, we observed that in high penetrance TNFR-associated periodic syndrome, at the molecular level, these alterations were secondary to a hyperactivation of the ERK1/2, STAT1/3/5, mammalian target of rapamycin, and NF-κB pathways in conventional T cells. In addition, these patients had a lower frequency of peripheral regulatory T cells, which also displayed a defective suppressive phenotype. These alterations were partially found in low penetrance TNFR-associated periodic syndrome, suggesting a specific link between the penetrance of the TNFRSF1A mutation and the observed T cell phenotype. Taken together, our data envision a novel role for adaptive immunity in the pathogenesis of TNFR-associated periodic syndrome involving both CD4(+) conventional T cells and Tregs, suggesting a novel mechanism of inflammation in the context of autoinflammatory disorders.


Subject(s)
Fever/genetics , Fever/immunology , Hereditary Autoinflammatory Diseases/genetics , Hereditary Autoinflammatory Diseases/immunology , Mutation/genetics , Penetrance , Receptors, Tumor Necrosis Factor, Type I/genetics , T-Lymphocytes, Regulatory/immunology , Adolescent , Adult , Aged , Cell Proliferation , Child , Cytokines/metabolism , Demography , Female , Fever/pathology , Hereditary Autoinflammatory Diseases/pathology , Humans , Immunophenotyping , Male , Middle Aged , Receptors, Antigen, T-Cell/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Young Adult
16.
Clin Rheumatol ; 34(1): 17-28, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24953660

ABSTRACT

Autoinflammatory disorders (AIDs) are a novel class of diseases elicited by mutations in genes regulating the homeostasis of innate immune complexes, named inflammasomes, which lead to uncontrolled oversecretion of the proinflammatory cytokine interleukin-1ß. Protean inflammatory symptoms are variably associated with periodic fever, depicting multiple specific conditions. Childhood is usually the lifetime in which most hereditary AIDs start, though still a relevant number of patients may experience a delayed disease onset and receive a definite diagnosis during adulthood. As a major referral laboratory for patients with recurrent fevers, we have tested samples from 787 patients in the period September 2007-March 2014, with a total of 1,328 AID-related genes evaluated and a gene/patient ratio of 1.69. In this report, we describe our experience in the clinical approach to AIDs, highlight the most striking differences between child and adult-onset AIDs, and shed an eye-opening insight into their diagnostic process.


Subject(s)
Hereditary Autoinflammatory Diseases/diagnosis , Hereditary Autoinflammatory Diseases/therapy , Immunity, Innate/genetics , Adult , Age Factors , Child , Diagnosis, Differential , Hereditary Autoinflammatory Diseases/genetics , Humans , Italy
17.
Mod Rheumatol ; 24(3): 381-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24251993

ABSTRACT

Hereditary autoinflammatory disorders encompass manifold dysfunctions of innate immunity caused by mutations in genes coding for the main characters of the inflammatory scene: most of these conditions have an early onset, ranging from the first days of life to the first decades, and include hereditary periodic fevers, NLRP-related diseases, granulomatous and pyogenic syndromes, which are basically characterized by upturned inflammasome activity and overproduction of bioactive interleukin (IL)-1ß and other proinflammatory cytokines. The discovery of a causative link between autoinflammation and IL-1ß release has improved our understanding of the intimate mechanisms of innate immunity, and has likewise led to the identification of extraordinary treatments for many of these disorders.


Subject(s)
Cytokines/genetics , Hereditary Autoinflammatory Diseases/genetics , Immunity, Innate/genetics , Interleukin-1beta/genetics , Cytokines/metabolism , Hereditary Autoinflammatory Diseases/metabolism , Humans , Interleukin-1beta/metabolism
18.
Orphanet J Rare Dis ; 8: 196, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-24360083

ABSTRACT

BACKGROUND: Mevalonate kinase deficiency (MKD) is caused by mutations in the MVK gene, encoding the second enzyme of mevalonate pathway, which results in subsequent shortage of downstream compounds, and starts in childhood with febrile attacks, skin, joint, and gastrointestinal symptoms, sometimes induced by vaccinations. METHODS: For a history of early-onset corticosteroid-induced reduction of bone mineral density in a 14-year-old boy with MKD, who also had presented three bone fractures, we administered weekly oral alendronate, a drug widely used in the management of osteoporosis and other high bone turnover diseases, which blocks mevalonate and halts the prenylation process. RESULTS: All of the patient's MKD clinical and laboratory abnormalities were resolved after starting alendronate treatment. CONCLUSIONS: This observation appears enigmatic, since alendronate should reinforce the metabolic block characterizing MKD, but is crucial because of the ultimate improvement shown by this patient. The anti-inflammatory properties of bisphosphonates are a new question for debate among physicians across various specialties, and requires further biochemical and clinical investigation.


Subject(s)
Alendronate/therapeutic use , Mevalonate Kinase Deficiency/drug therapy , Adolescent , Alendronate/administration & dosage , Bone Density/drug effects , Bone Density Conservation Agents/therapeutic use , Humans , Male
19.
PLoS One ; 8(9): e73443, 2013.
Article in English | MEDLINE | ID: mdl-24066048

ABSTRACT

Tumor necrosis factor-receptor associated periodic syndrome (TRAPS) is a rare autosomal dominant autoinflammatory disorder characterized by recurrent episodes of long-lasting fever and inflammation in different regions of the body, such as the musculo-skeletal system, skin, gastrointestinal tract, serosal membranes and eye. Our aims were to evaluate circulating microRNAs (miRNAs) levels in patients with TRAPS, in comparison to controls without inflammatory diseases, and to correlate their levels with parameters of disease activity and/or disease severity. Expression levels of circulating miRNAs were measured by Agilent microarrays in 29 serum samples from 15 TRAPS patients carrying mutations known to be associated with high disease penetrance and from 8 controls without inflammatory diseases. Differentially expressed and clinically relevant miRNAs were detected using GeneSpring GX software. We identified a 6 miRNAs signature able to discriminate TRAPS from controls. Moreover, 4 miRNAs were differentially expressed between patients treated with the interleukin (IL)-1 receptor antagonist, anakinra, and untreated patients. Of these, miR-92a-3p and miR-150-3p expression was found to be significantly reduced in untreated patients, while their expression levels were similar to controls in samples obtained during anakinra treatment. MiR-92b levels were inversely correlated with the number of fever attacks/year during the 1(st) year from the index attack of TRAPS, while miR-377-5p levels were positively correlated with serum amyloid A (SAA) circulating levels. Our data suggest that serum miRNA levels show a baseline pattern in TRAPS, and may serve as potential markers of response to therapeutic intervention.


Subject(s)
Hereditary Autoinflammatory Diseases/genetics , Adult , Case-Control Studies , Female , Fever , Hereditary Autoinflammatory Diseases/drug therapy , Humans , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Male , MicroRNAs , Middle Aged , Receptors, Tumor Necrosis Factor, Type I/genetics
20.
Clin Exp Rheumatol ; 31(3 Suppl 77): 141-9, 2013.
Article in English | MEDLINE | ID: mdl-23899820

ABSTRACT

Tumour necrosis factor-receptor associated periodic syndrome (TRAPS) is a rare autosomal dominant autoinflammatory disorder characterised by recurrent episodes of long-lasting fever and inflammation in different regions of the body, as musculo-skeletal system, skin, gastrointestinal tube, serosal membranes and eye. Inflammatory attacks usually start in the pediatric age with initial corticosteroid-responsiveness. Most reported cases of TRAPS involve patients of European ancestry and diagnosis can be formulated by the combination of genetic analysis and a compatible phenotype. Its prognosis is strictly dependent on the appearance of amyloidosis, secondary to uncontrolled relapsing inflammation. Thanks to a better understanding of its pathogenesis, the disease is now managed with anti-interleukin (IL)-1 antagonists, rather than corticosteroids or tumour necrosis factor (TNF) inhibitors. The aim of this review is to describe the current understanding and advances of TRAPS genetic basis, pathogenesis and management options by integrating the most recent data in the medical literature.


Subject(s)
Hereditary Autoinflammatory Diseases/drug therapy , Immunosuppressive Agents/therapeutic use , Fever , Genetic Predisposition to Disease , Hereditary Autoinflammatory Diseases/genetics , Hereditary Autoinflammatory Diseases/immunology , Hereditary Autoinflammatory Diseases/physiopathology , Humans , Mutation , Phenotype , Prognosis , Receptors, Tumor Necrosis Factor, Type I/genetics , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...