Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 24(5): 814-826, 2023 05.
Article in English | MEDLINE | ID: mdl-36997670

ABSTRACT

Missense mutations in PLCG2 can cause autoinflammation with phospholipase C gamma 2-associated antibody deficiency and immune dysregulation (APLAID). Here, we generated a mouse model carrying an APLAID mutation (p.Ser707Tyr) and found that inflammatory infiltrates in the skin and lungs were only partially ameliorated by removing inflammasome function via the deletion of caspase-1. Also, deleting interleukin-6 or tumor necrosis factor did not fully prevent APLAID mutant mice from autoinflammation. Overall, these findings are in accordance with the poor response individuals with APLAID have to treatments that block interleukin-1, JAK1/2 or tumor necrosis factor. Cytokine analysis revealed increased granulocyte colony-stimulating factor (G-CSF) levels as the most distinct feature in mice and individuals with APLAID. Remarkably, treatment with a G-CSF antibody completely reversed established disease in APLAID mice. Furthermore, excessive myelopoiesis was normalized and lymphocyte numbers rebounded. APLAID mice were also fully rescued by bone marrow transplantation from healthy donors, associated with reduced G-CSF production, predominantly from non-hematopoietic cells. In summary, we identify APLAID as a G-CSF-driven autoinflammatory disease, for which targeted therapy is feasible.


Subject(s)
Bone Marrow Transplantation , Granulocyte Colony-Stimulating Factor , Animals , Mice , Cytokines , Interleukin-1 , Tumor Necrosis Factor-alpha/genetics , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/metabolism
2.
Nat Immunol ; 15(8): 738-48, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24952504

ABSTRACT

Assembly of the NLRP3 inflammasome activates caspase-1 and mediates the processing and release of the leaderless cytokine IL-1ß and thereby serves a central role in the inflammatory response and in diverse human diseases. Here we found that upon activation of caspase-1, oligomeric NLRP3 inflammasome particles were released from macrophages. Recombinant oligomeric protein particles composed of the adaptor ASC or the p.D303N mutant form of NLRP3 associated with cryopyrin-associated periodic syndromes (CAPS) stimulated further activation of caspase-1 extracellularly, as well as intracellularly after phagocytosis by surrounding macrophages. We found oligomeric ASC particles in the serum of patients with active CAPS but not in that of patients with other inherited autoinflammatory diseases. Our findings support a model whereby the NLRP3 inflammasome, acting as an extracellular oligomeric complex, amplifies the inflammatory response.


Subject(s)
Carrier Proteins/immunology , Caspase 1/immunology , Inflammasomes/immunology , Inflammation/immunology , Macrophages/immunology , Animals , Apoptosis Regulatory Proteins , CARD Signaling Adaptor Proteins , Carrier Proteins/blood , Carrier Proteins/genetics , Caspase 1/genetics , Caspases/genetics , Caspases/immunology , Caspases, Initiator , Cells, Cultured , Cryopyrin-Associated Periodic Syndromes/blood , Cytoskeletal Proteins/blood , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/immunology , HEK293 Cells , Humans , Inflammasomes/blood , Interleukin-1beta/blood , Interleukin-1beta/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Phagocytosis/immunology , Signal Transduction/immunology
3.
Article in English | MEDLINE | ID: mdl-38552317

ABSTRACT

OBJECTIVE: The vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic (VEXAS) syndrome is a complex immune disorder consequence of somatic UBA1 variants. Most reported pathogenic UBA1 variants are missense or splice site mutations directly impairing the translational start site at p. Met41, with recent studies showing that these variants are frequent causes of recurrent inflammation in older individuals. Here we aimed to characterize a novel UBA1 variant found in two patients clinically presenting with VEXAS syndrome. METHODS: Patients' data were collected from direct assessments and from their medical charts. Genomics analyses were performed by both Sanger and amplicon-based deep sequencing, mRNA studies were performed by both cDNA subcloning and mRNA sequencing. RESULTS: We report a novel, somatic variant in a canonical splice site of the UBA1 gene (c.346-2A>G), which was identified in two unrelated adult male patients with late-onset, unexplained inflammatory manifestations including recurrent fever, Sweet syndrome-like neutrophilic dermatosis, and lung inflammation responsive only to glucocorticoids. RNA analysis from patients' samples demonstrated aberrant mRNA splicing leading to multiple in-frame transcripts, including a transcript retaining the full sequence of intron 4 and a different transcript with the deletion of the first 15 nucleotides of exon 5. CONCLUSION: Here we describe the abnormal UBA1 transcription as a consequence of the novel c.346-2A>G variant identified in two patients with clinical features compatible with VEXAS syndrome. Overall, these results further demonstrate the expanding spectrum of variants in UBA1 leading to pathology and support for a complete gene evaluation in those candidate patients for VEXAS syndrome.

4.
Nat Chem Biol ; 17(3): 361, 2021 03.
Article in English | MEDLINE | ID: mdl-33495649

ABSTRACT

An Addendum to this paper has been published: https://doi.org/10.1038/s41589-021-00741-6.

5.
Australas J Dermatol ; 64(2): 260-267, 2023 May.
Article in English | MEDLINE | ID: mdl-36797819

ABSTRACT

Monoallelic NLRC4 gain-of-function variants cause an inflammasomopathy with diverse clinical forms including infantile enterocolitis, recurrent macrophage activation syndrome, cold-induced urticaria-like lesions (or familial-cold autoinflammatory syndrome, FCAS4), and painful subcutaneous nodules. Here, we identified a large family with six consecutive generations affected. Genetic analyses detected the heterozygous p.Ser445Pro NLRC4 variant in three patients, which has been previously reported in a Dutch family with FCAS4. We aimed to describe the clinicopathological features and the functional consequences of the detected NLRC4 variant. Patients presented an early-onset (3 months-6 years) inflammatory disease characterized by recurrent panniculitis, fever and arthralgia. Histopathological examination showed perivascular and interstitial lymphohistiocytic infiltrates in the dermis and mixed panniculitis. Functional analysis supported the conclusion that the p.Ser445Pro NLRC4 variant leads to a constitutive activation of NLRC4-inflammasome and increased plasma levels of IL-18. Prompt recognition of early-onset panniculitis through clinicopathological examination and laboratory biomarkers may allow targeted therapies.


Subject(s)
CARD Signaling Adaptor Proteins , Panniculitis , Humans , Virulence , CARD Signaling Adaptor Proteins/genetics , Syndrome , Panniculitis/genetics , Phenotype , Calcium-Binding Proteins/genetics
6.
J Clin Immunol ; 42(7): 1421-1432, 2022 10.
Article in English | MEDLINE | ID: mdl-35716229

ABSTRACT

Pathogenic RIPK1 variants have been described as the cause of two different inborn errors of immunity. Biallelic loss-of-function variants cause the recessively inherited RIPK1 deficiency, while monoallelic variants impairing the caspase-8-mediated RIPK1 cleavage provoke a novel autoinflammatory disease (AID) called cleavage-resistant RIPK1-induced autoinflammatory (CRIA) syndrome. The aim of this study was to characterize the pathogenicity of two novel RIPK1 variants located at the cleavage site of caspase-8 detected in patients with dominantly-inherited, early-onset undefined AID. RIPK1 genotyping was performed by Sanger and next-generation sequencing. Clinical and analytical data were collected from medical charts, and in silico and in vitro assays were performed to evaluate the functional consequences. Genetic analyses identified two novel heterozygous RIPK1 variants at the caspase-8 cleavage site (p.Leu321Arg and p.Asp324Gly), which displayed a perfect intrafamilial phenotype-genotype segregation following a dominant inheritance pattern. Structural analyses suggested that these variants disrupt the normal RIPK1 structure, probably making it less accessible to and/or less cleavable by caspase-8. In vitro experiments confirmed that the p.Leu321Arg and p.Asp324Gly RIPK1 variants were resistant to caspase-8-mediated cleavage and induced a constitutive activation of necroptotic pathway in a similar manner that previously characterized RIPK1 variants causing CRIA syndrome. All these results strongly supported the pathogenicity of the two novel RIPK1 variants and the diagnosis of CRIA syndrome in all enrolled patients. Moreover, the evidences here collected expand the phenotypic and genetic diversity of this recently described AID, and provide interesting data about effectiveness of treatments that may benefit future patients.


Subject(s)
Apoptosis , Hereditary Autoinflammatory Diseases , Humans , Caspase 8/genetics , Caspase 8/metabolism , Hereditary Autoinflammatory Diseases/diagnosis , Hereditary Autoinflammatory Diseases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
7.
Allergol Immunopathol (Madr) ; 50(1): 25-30, 2022.
Article in English | MEDLINE | ID: mdl-34873893

ABSTRACT

Familial Mediterranean fever (FMF) is the most frequent autoinflammatory disorder characterized by short, repeated, and self-limiting crises of fever and serositis. The disease was described as autosomal recessive hereditary transmission secondary to variants of the MEFV (MEditerranean FeVer) gene, even though a variable proportion of patients only present a heterozygous variant. FMF is very common in certain ethnic groups (Turkish, Armenian, Arab, and Jewish), even though it has been described throughout the Mediterranean and elsewhere in the world. The clinical manifestations are variable, with secondary amyloidosis being the most serious complication of the disorder. Treatment and prophylaxis are mainly based on the administration of colchicine, which prevents the crises and avoids complications in most cases. This study reviews the course of seven pediatric patients diagnosed with FMF during the period 2010-2018 at a district hospital. Most of the patients were of Caucasian origin, with onset at an early age in the form of fever as the main symptom, and some patients moreover presented less frequent manifestations (pericardial effusion, sensorineural hearing loss). Two cases presented plasmatic amyloid A protein elevation that subsided with the treatment. All the patients initially received colchicine, and one of them required prescription of anakinra, which was replaced by canakinumab due to a serious adverse reaction. There were no cases of consanguinity, and all the patients were of Mediterranean origin. The subjects showed a favorable course over the years, which was attributed to the early diagnosis and treatment provided.


Subject(s)
Familial Mediterranean Fever , Child , Colchicine/therapeutic use , Familial Mediterranean Fever/diagnosis , Familial Mediterranean Fever/drug therapy , Familial Mediterranean Fever/epidemiology , Humans , Interleukin 1 Receptor Antagonist Protein , Pyrin/genetics
8.
Clin Immunol ; 227: 108750, 2021 06.
Article in English | MEDLINE | ID: mdl-33945870

ABSTRACT

The co-existence of an autoinflammatory syndrome with a demyelinating disorder is a very rare occurrence raising the question whether there is a pathophysiological connection between them. We describe the case of a man with symptoms of cryopyrin-associated periodic syndrome (CAPS) since infancy who later developed multiple sclerosis (MS). As CAPS was genetically confirmed, the inhibition of interleukin-1 (IL-1) with anakinra led to a swift resolution of the CAPS symptoms and also, in combination with teriflunomide, to a clinical and imaging improvement of MS. In vitro studies showed that, upon a CAPS flare, the patient's peripheral neutrophils released neutrophil extracellular traps (NETs) decorated with IL-1ß, while NET release was markedly decreased following anakinra-induced remission of CAPS. Taking into account the growing evidence on the involvement of IL-1ß in experimental models of MS, this rare patient case suggests that the role of neutrophils/NETs and IL-1ß in MS should be further studied.


Subject(s)
Cryopyrin-Associated Periodic Syndromes/immunology , Extracellular Traps/immunology , Interleukin-1beta/immunology , Multiple Sclerosis/immunology , Neutrophils/immunology , Adult , Antirheumatic Agents/therapeutic use , Brain/diagnostic imaging , Crotonates/therapeutic use , Cryopyrin-Associated Periodic Syndromes/complications , Cryopyrin-Associated Periodic Syndromes/drug therapy , Cryopyrin-Associated Periodic Syndromes/physiopathology , Humans , Hydroxybutyrates/therapeutic use , Immunosuppressive Agents/therapeutic use , In Vitro Techniques , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Magnetic Resonance Imaging , Male , Multiple Sclerosis/complications , Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/drug therapy , Nitriles/therapeutic use , Toluidines/therapeutic use
9.
Br J Haematol ; 193(2): 299-306, 2021 04.
Article in English | MEDLINE | ID: mdl-33200406

ABSTRACT

The presence of a serum monoclonal component has been associated with poor outcomes in some lymphomas. However, data in follicular lymphoma (FL) are scarce. We studied 311 FL patients diagnosed at a single institution, for whom information on serum immunofixation electrophoresis (sIFE) at diagnosis was available. Baseline characteristics and outcomes were compared between patients with a positive (+sIFE) and a negative sIFE (-sIFE). sIFE was positive in 82 patients (26%). Baseline features were comparable between both groups, except for an older age and higher proportion of elevated ß2 -microglobulin levels in the +sIFE group. With a median follow-up of 4.6 years, a +sIFE was associated with a higher risk of early relapse (POD24, 27% vs. 15%, P = 0·02), shorter progression-free survival (PFS; 42% vs. 52% at 5 years, P = 0·008), and shorter overall survival (OS; 59% vs. 77% at 10 years, P = 0·046). In patients >60 years, a +sIFE was an independent predictor of OS [hazard ratio (HR) = 2·4, 95% confidence interval (CI): 1·2-5·0; P = 0·02]. Approximately one quarter of patients with FL has a +sIFE at diagnosis, which is a predictor of poor outcome. These findings encourage further investigation of its relationship with B-cell biology and the tumour microenvironment.


Subject(s)
Blood Protein Electrophoresis/methods , Lymphoma, Follicular/metabolism , Monoclonal Gammopathy of Undetermined Significance/blood , beta 2-Microglobulin/blood , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cyclophosphamide , Doxorubicin , Female , Follow-Up Studies , Humans , Lymphoma, Follicular/drug therapy , Lymphoma, Follicular/mortality , Lymphoma, Follicular/pathology , Male , Middle Aged , Neoplasm Grading/methods , Prednisone , Prognosis , Progression-Free Survival , Rituximab , Tumor Microenvironment , Vincristine , Watchful Waiting
10.
Nat Chem Biol ; 15(6): 560-564, 2019 06.
Article in English | MEDLINE | ID: mdl-31086329

ABSTRACT

NLRP3 (NOD-like receptor pyrin domain-containing protein 3) is an innate immune sensor that contributes to the development of different diseases, including monogenic autoinflammatory syndromes, gout, atherosclerosis, and Alzheimer's disease. The molecule sulfonylurea MCC950 is a NLRP3 inflammasome inhibitor with potential clinical utility. However, the mechanism of action of MCC950 remains unknown. Here, we characterize the mechanism of action of MCC950 in both wild-type and autoinflammatory-related NLRP3 mutants, and demonstrate that MCC950 closes the 'open' conformation of active NLRP3.


Subject(s)
Heterocyclic Compounds, 4 or More Rings/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Sulfones/pharmacology , Binding Sites/drug effects , Furans , HEK293 Cells , Heterocyclic Compounds, 4 or More Rings/chemistry , Humans , Indenes , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Conformation , Sulfonamides , Sulfones/chemistry
11.
Dermatology ; 237(3): 365-371, 2021.
Article in English | MEDLINE | ID: mdl-33171462

ABSTRACT

INTRODUCTION: Hidradenitis suppurativa (HS) is a chronic, inflammatory, recurrent disorder of the pilosebaceous unit. Currently, several attempts have been made to classify this disease according to its pathogenesis and clinical manifestations. We attempted at classifying 103 patients using two-step cluster analysis. METHODS: The final model included body mass index, C-reactive protein (CRP), and serum concentrations of IL-1, IL-6, IL-17, and IL-10 as continuous variables, and sex, later/early onset, anterior/posterior lesion sites, presence/absence of sinus tracts, nodules and abscesses, positive/negative history of pilonidal sinus, and presence/absence of mutations in gamma-secretase subunits (APH1A, APH1B, MEFV, NCSTN, PSEN1, PSEN2, PSENEN, PSTPIP1) as qualitative variables. RESULTS: The resultant model defined two groupings or clusters: cluster 1 (64.9% of patients) characterized by nonobese males, with nodular lesions in posterior sites, early-onset HS, higher IL-10, presence of gamma-secretase mutations, and history of pilonidal sinus; and cluster 2 (35.1% of patients) characterized by obese females or males, with lesions in anterior sites, more presence of sinus tracts and abscesses and less nodules, later-onset HS, and higher concentrations of IL-1, CRP, IL-17, and IL-6. Severity measures (Hurley, HS-PGA, and IHS4) and tobacco use were discarded because the analysis found them to be less relevant for clustering. CONCLUSION: Our resultant model confirms the clinical impression that HS is a disease spectrum with two pathogenic poles defining two clusters or endotypes. The probability of having severe disease was equally distributed in the two clusters. The variable with the highest predictive value for clustering was involvement of typical anterior sites (axillae, submammary) or atypical posterior sites (back, gluteal). Serum concentrations of interleukins, tobacco use, and sex had a lower predictive power for clustering.


Subject(s)
Hidradenitis Suppurativa/classification , Hidradenitis Suppurativa/etiology , Amyloid Precursor Protein Secretases/genetics , Body Mass Index , C-Reactive Protein/metabolism , Cluster Analysis , Female , Hidradenitis Suppurativa/diagnosis , Humans , Interleukins/blood , Male , Pilonidal Sinus/complications , Severity of Illness Index
12.
J Clin Immunol ; 40(7): 987-1000, 2020 10.
Article in English | MEDLINE | ID: mdl-32671674

ABSTRACT

Autoinflammatory diseases (AIDs) were first described as clinical disorders characterized by recurrent episodes of seemingly unprovoked sterile inflammation. In the past few years, the identification of novel AIDs expanded their phenotypes toward more complex clinical pictures associating vasculopathy, autoimmunity, or immunodeficiency. Herein, we describe two unrelated patients suffering since the neonatal period from a complex disease mainly characterized by severe sterile inflammation, recurrent bacterial infections, and marked humoral immunodeficiency. Whole-exome sequencing detected a novel, de novo heterozygous PLCG2 variant in each patient (p.Ala708Pro and p.Leu845_Leu848del). A clear enhanced PLCγ2 activity for both variants was demonstrated by both ex vivo calcium responses of the patient's B cells to IgM stimulation and in vitro assessment of PLC activity. These data supported the autoinflammation and PLCγ2-associated antibody deficiency and immune dysregulation (APLAID) diagnosis in both patients. Immunological evaluation revealed a severe decrease of immunoglobulins and B cells, especially class-switched memory B cells, with normal T and NK cell counts. Analysis of bone marrow of one patient revealed a reduced immature B cell fraction compared with controls. Additional investigations showed that both PLCG2 variants activate the NLRP3-inflammasome through the alternative pathway instead of the canonical pathway. Collectively, the evidences here shown expand APLAID diversity toward more severe phenotypes than previously reported including dominantly inherited agammaglobulinemia, add novel data about its genetic basis, and implicate the alternative NLRP3-inflammasome activation pathway in the basis of sterile inflammation.


Subject(s)
Agammaglobulinemia/diagnosis , Agammaglobulinemia/genetics , Hereditary Autoinflammatory Diseases/diagnosis , Hereditary Autoinflammatory Diseases/genetics , Mutation , Phospholipase C gamma/genetics , Adolescent , Agammaglobulinemia/therapy , Autoimmunity/genetics , Biomarkers , Caspase 1/metabolism , Child , Cytokines/metabolism , DNA Mutational Analysis , Female , Genetic Association Studies , Genetic Predisposition to Disease , Hereditary Autoinflammatory Diseases/therapy , Humans , Inflammasomes/metabolism , Male , Pedigree , Phenotype , Phospholipase C gamma/chemistry , Phospholipase C gamma/metabolism , Structure-Activity Relationship
13.
Blood ; 131(9): 974-981, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29284595

ABSTRACT

To date, the pathogenic mechanisms underlying Schnitzler syndrome remain obscure, in particular, the interplay between the monoclonal protein and increased interleukin-1ß (IL-1ß) production, although interest in the contribution of genetic factors has been fueled by detection of somatic NLRP3 mosaicism in 2 patients with the variant-type Schnitzler syndrome. At 2 specialist UK centers, we have identified 21 patients who fulfilled diagnostic criteria for Schnitzler syndrome with urticarial rash, fever, arthralgia, and bone pain; 47% reported weight loss, 40% fatigue, and 21% lymphadenopathy. An immunoglobulin M (IgM) κ paraprotein was detected in 86%; the remainder had IgM λ or IgG κ. Patients underwent searches for germ line and somatic mutations using next-generation sequencing technology. Moreover, we designed a panel consisting of 32 autoinflammatory genes to explore genetic susceptibility factor(s) to Schnitzler syndrome. Genetic analysis revealed neither germ line nor somatic NLRP3, TNFRSF1A, NLRC4, or NOD2 mutations, apart from 1 patient with a germ line NLRP3 p.V198M substitution. The proinflammatory cytokines and extracellular apoptosis-associated speck-like protein with caspase recruitment domain (ASC) measured in the serum of Schnitzler syndrome patients during active disease were significantly higher than healthy controls. Ninety-five percent of our cohort achieved a complete response to recombinant IL-1 receptor antagonist (anakinra). Our findings do not support a role for somatic NLRP3 mosaicism in disease pathogenesis; although elevated levels of ASC, IL-6, and IL-18 in patients' serum, and the response to anakinra, suggest that Schnitzler syndrome is associated with upregulated inflammasome activation. Despite its rarity, Schnitzler syndrome is an important diagnosis as treatment with IL-1 antagonists dramatically improves quality of life for patients.


Subject(s)
Germ-Line Mutation , Interleukin 1 Receptor Antagonist Protein/administration & dosage , Schnitzler Syndrome , Adult , Aged , Amino Acid Substitution , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Female , Humans , Interleukin-18/blood , Interleukin-18/genetics , Interleukin-6/blood , Interleukin-6/genetics , Male , Middle Aged , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Schnitzler Syndrome/blood , Schnitzler Syndrome/drug therapy , Schnitzler Syndrome/genetics
14.
J Allergy Clin Immunol ; 143(1): 359-368, 2019 01.
Article in English | MEDLINE | ID: mdl-30273710

ABSTRACT

BACKGROUND: Postzygotic de novo mutations lead to the phenomenon of gene mosaicism. The 3 main types are called somatic, gonadal, and gonosomal mosaicism, which differ in terms of the body distribution of postzygotic mutations. Mosaicism has been reported occasionally in patients with primary immunodeficiency diseases (PIDs) since the early 1990s, but its real involvement has not been systematically addressed. OBJECTIVE: We sought to investigate the incidence of gene mosaicism in patients with PIDs. METHODS: The amplicon-based deep sequencing method was used in the 3 parts of the study that establish (1) the allele frequency of germline variants (n = 100), (2) the incidence of parental gonosomal mosaicism in families with PIDs with de novo mutations (n = 92), and (3) the incidence of mosaicism in families with PIDs with moderate-to-high suspicion of gene mosaicism (n = 36). Additional investigations evaluated body distribution of postzygotic mutations, their stability over time, and their characteristics. RESULTS: The range of allele frequency (44.1% to 55.6%) was established for germline variants. Those with minor allele frequencies of less than 44.1% were assumed to be postzygotic. Mosaicism was detected in 30 (23.4%) of 128 families with PIDs, with a variable minor allele frequency (0.8% to 40.5%). Parental gonosomal mosaicism was detected in 6 (6.5%) of 92 families with de novo mutations, and a high incidence of mosaicism (63.9%) was detected among families with moderate-to-high suspicion of gene mosaicism. In most analyzed cases mosaicism was found to be both uniformly distributed and stable over time. CONCLUSION: This study represents the largest performed to date to investigate mosaicism in patients with PIDs, revealing that it affects approximately 25% of enrolled families. Our results might have serious consequences regarding treatment and genetic counseling and reinforce the use of next-generation sequencing-based methods in the routine analyses of PIDs.


Subject(s)
Alleles , Gene Frequency , Immunologic Deficiency Syndromes/genetics , Mosaicism , Family , Female , High-Throughput Nucleotide Sequencing , Humans , Immunologic Deficiency Syndromes/immunology , Male
15.
Acta Derm Venereol ; 99(12): 1071-1077, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31408184

ABSTRACT

Acquired cold urticaria (ACU) is characterized by the development of itchy wheals after cold exposure. Generalized urticarial skin rashes triggered by cold exposure characterize certain monogenic autoinflammatory diseases (AIDs). The objective of this study is to investigate the presence of variants in genes causing AIDs that present with cold-induced urticarial skin rashes in patients clinically diagnosed with ACU, in order to look for susceptibility factors for the disease. Fifty patients with primary ACU were studied. Germline and post-zygotic variants on the NLRP3, NLRP12, NLRC4 and PLCG2 genes were investigated using next-generation sequencing technology. Seven patients (14%) carried 8 heterozygous germline variants in the following genes: NLRP3 (n = 1), NLRP12 (n = 3), NLRC4 (n = 1), PLCG2 (n = 3). No pathogenic or likely pathogenic variants were detected, and deep analyses of the sequences obtained did not identify any post-zygotic variant. In conclusion, ACU is not related to post-zygotic or germline pathogenic variants in the NLRP3, NLRP12, NLRC4 and PLCG2 genes.


Subject(s)
Cold Temperature/adverse effects , Genetic Variation , Hereditary Autoinflammatory Diseases/genetics , Urticaria/genetics , Adolescent , Adult , Aged , CARD Signaling Adaptor Proteins/genetics , Calcium-Binding Proteins/genetics , Child , Child, Preschool , Female , Genetic Predisposition to Disease , Hereditary Autoinflammatory Diseases/diagnosis , Hereditary Autoinflammatory Diseases/immunology , Humans , Infant , Intracellular Signaling Peptides and Proteins/genetics , Male , Middle Aged , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Phenotype , Phospholipase C gamma/genetics , Prospective Studies , Risk Factors , Spain , Tertiary Care Centers , Urticaria/diagnosis , Urticaria/immunology , Young Adult
16.
J Med Genet ; 55(8): 530-537, 2018 08.
Article in English | MEDLINE | ID: mdl-29599418

ABSTRACT

BACKGROUND: Hereditary recurrent fevers (HRFs) are rare inflammatory diseases sharing similar clinical symptoms and effectively treated with anti-inflammatory biological drugs. Accurate diagnosis of HRF relies heavily on genetic testing. OBJECTIVES: This study aimed to obtain an experts' consensus on the clinical significance of gene variants in four well-known HRF genes: MEFV, TNFRSF1A, NLRP3 and MVK. METHODS: We configured a MOLGENIS web platform to share and analyse pathogenicity classifications of the variants and to manage a consensus-based classification process. Four experts in HRF genetics submitted independent classifications of 858 variants. Classifications were driven to consensus by recruiting four more expert opinions and by targeting discordant classifications in five iterative rounds. RESULTS: Consensus classification was reached for 804/858 variants (94%). None of the unsolved variants (6%) remained with opposite classifications (eg, pathogenic vs benign). New mutational hotspots were found in all genes. We noted a lower pathogenic variant load and a higher fraction of variants with unknown or unsolved clinical significance in the MEFV gene. CONCLUSION: Applying a consensus-driven process on the pathogenicity assessment of experts yielded rapid classification of almost all variants of four HRF genes. The high-throughput database will profoundly assist clinicians and geneticists in the diagnosis of HRFs. The configured MOLGENIS platform and consensus evolution protocol are usable for assembly of other variant pathogenicity databases. The MOLGENIS software is available for reuse at http://github.com/molgenis/molgenis; the specific HRF configuration is available at http://molgenis.org/said/. The HRF pathogenicity classifications will be published on the INFEVERS database at https://fmf.igh.cnrs.fr/ISSAID/infevers/.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Genetic Variation , Hereditary Autoinflammatory Diseases/diagnosis , Hereditary Autoinflammatory Diseases/genetics , Workflow , Alleles , Computational Biology/methods , Consensus , Databases, Genetic , Disease Management , Genetic Association Studies/methods , Genetic Testing , Humans , Phenotype , Practice Guidelines as Topic , Web Browser
18.
Pediatr Dermatol ; 35(6): e337-e340, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30168189

ABSTRACT

We report two unrelated infants in whom chronic urticaria was the first clinical manifestation of cryopyrin-associated periodic syndrome, which should be suspected in infants with early-onset chronic urticaria, especially if there is a neutrophil-rich infiltrate in the skin biopsy. Early diagnosis of cryopyrin-associated periodic syndrome may lead to early and successful treatment with anti-interleukin-1 medications.


Subject(s)
Cryopyrin-Associated Periodic Syndromes/diagnosis , Urticaria/etiology , Chronic Disease , Cryopyrin-Associated Periodic Syndromes/complications , Cryopyrin-Associated Periodic Syndromes/genetics , Female , Genetic Testing , Humans , Infant , Mutation , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Skin/pathology
19.
J Allergy Clin Immunol ; 139(1): 202-211.e6, 2017 01.
Article in English | MEDLINE | ID: mdl-27394913

ABSTRACT

BACKGROUND: Inflammasomes are cytosolic multiprotein complexes in macrophages. They assemble after infection- or stress-associated stimuli, activating both caspase-1-mediated inflammatory cytokine secretion and pyroptosis. Increased inflammasome activity resulting from gene mutations is related to monogenic autoinflammatory syndromes. However, variable penetrance among patients with the same gene mutations suggests involvement of additional mechanisms associated with inflammasome gene regulation. OBJECTIVE: We sought to investigate the role of DNA demethylation in activating inflammasome genes during macrophage differentiation and monocyte activation in healthy control subjects and patients with autoinflammatory syndrome. METHODS: Inflammasome-related genes were tested for DNA methylation and mRNA levels by using bisulfite pyrosequencing and quantitative RT-PCR in monocytes in vitro differentiated to macrophages and exposed to inflammatory conditions. The contribution of Tet methylcytosine dioxygenase 2 (TET2) and nuclear factor κB to DNA demethylation was tested by using chromatin immunoprecipitation, small interfering RNA-mediated downregulation, and pharmacologic inhibition. RESULTS: We observed that inflammasome-related genes are rapidly demethylated in both monocyte-to-macrophage differentiation and on monocyte activation. Demethylation associates with increased gene expression, and both mechanisms are impaired when TET2 and nuclear factor κB are downregulated. We analyzed DNA methylation levels of inflammasome-related genes in patients with cryopyrin-associated periodic syndromes (CAPS) and familial Mediterranean fever, 2 archetypical monogenic autoinflammatory syndromes. Under the above conditions, monocytes from untreated patients with CAPS undergo more efficient DNA demethylation than those of healthy subjects. Interestingly, patients with CAPS treated with anti-IL-1 drugs display methylation levels similar to those of healthy control subjects. CONCLUSION: Our study is the first to demonstrate the involvement of DNA methylation-associated alterations in patients with monogenic autoinflammatory disease and opens up possibilities for novel clinical markers.


Subject(s)
Cryopyrin-Associated Periodic Syndromes/genetics , DNA Methylation/genetics , Inflammasomes/genetics , Cryopyrin-Associated Periodic Syndromes/metabolism , Cytokines/genetics , Cytokines/metabolism , DNA-Binding Proteins/genetics , Dioxygenases , Familial Mediterranean Fever/genetics , Familial Mediterranean Fever/metabolism , Humans , Macrophages/metabolism , Monocytes/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins/genetics
20.
Biol Blood Marrow Transplant ; 23(8): 1269-1275, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28434927

ABSTRACT

The emergence of oligoclonal bands (OB) in patients with multiple myeloma achieving a complete remission (CR) after autologous stem cell transplantation (ASCT) and the use of novel agents is a well-recognized event. The presence of OB is associated with favorable outcome. However, the emergence of OB in light-chain (AL) amyloidosis has never been investigated. The aim of the study was to determine the incidence, natural history, and prognostic impact of OB in 50 patients with AL amyloidosis who achieved at least a partial response either after upfront ASCT (20 patients [40%]) or after conventional treatment in patients ineligible for transplantation (30 patients [60%]). OB were observed in 60% of the patients, with IgG-kappa (30.7%) the most frequently detected isotype. This phenomenon was more prevalent in patients achieving CR than those in other response categories (88% versus 32%, P = .0001). The landmark analysis at 1 year after diagnosis demonstrates a significantly longer progression-free survival and an improvement trend in overall survival (P = .04 and P = .06, respectively). This prognostic impact was also observed in patients who achieved CR and in patients with more advanced stage. In summary, this is the first report of OB in patients with AL amyloidosis. Although its biological meaning remains unclear, it could reflect a more robust humoral immune response.


Subject(s)
Hematopoietic Stem Cell Transplantation , Immunoglobulin Light-chain Amyloidosis , Adult , Aged , Aged, 80 and over , Autografts , Disease-Free Survival , Female , Follow-Up Studies , Humans , Immunoglobulin Light-chain Amyloidosis/blood , Immunoglobulin Light-chain Amyloidosis/mortality , Immunoglobulin Light-chain Amyloidosis/therapy , Incidence , Male , Middle Aged , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL