Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
J Exp Med ; 221(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38652464

ABSTRACT

OTULIN-related autoinflammatory syndrome (ORAS), a severe autoinflammatory disease, is caused by biallelic pathogenic variants of OTULIN, a linear ubiquitin-specific deubiquitinating enzyme. Loss of OTULIN attenuates linear ubiquitination by inhibiting the linear ubiquitin chain assembly complex (LUBAC). Here, we report a patient who harbors two rare heterozygous variants of OTULIN (p.P152L and p.R306Q). We demonstrated accumulation of linear ubiquitin chains upon TNF stimulation and augmented TNF-induced cell death in mesenchymal stem cells differentiated from patient-derived iPS cells, which confirms that the patient has ORAS. However, although the de novo p.R306Q variant exhibits attenuated deubiquitination activity without reducing the amount of OTULIN, the deubiquitination activity of the p.P152L variant inherited from the mother was equivalent to that of the wild-type. Patient-derived MSCs in which the p.P152L variant was replaced with wild-type also exhibited augmented TNF-induced cell death and accumulation of linear chains. The finding that ORAS can be caused by a dominant-negative p.R306Q variant of OTULIN furthers our understanding of disease pathogenesis.


Subject(s)
Ubiquitination , Female , Humans , Endopeptidases/genetics , Endopeptidases/metabolism , Hereditary Autoinflammatory Diseases/genetics , Hereditary Autoinflammatory Diseases/pathology , Hereditary Autoinflammatory Diseases/metabolism , Induced Pluripotent Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , Mutation , Pedigree , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/genetics , Ubiquitin/metabolism , Infant, Newborn
2.
Immunol Res ; 71(4): 578-587, 2023 08.
Article in English | MEDLINE | ID: mdl-36991303

ABSTRACT

The kidney represents an important target of systemic inflammation. Its involvement in monogenic and multifactorial autoinflammatory diseases (AIDs) vary from peculiar and relatively frequent manifestations to some rare but severe features that may end up requiring transplantation. The pathogenetic background is also very heterogeneous ranging from amyloidosis to non-amyloid related damage rooted in inflammasome activation. Kidney involvement in monogenic and polygenic AIDs may present as renal amyloidosis, IgA nephropathy, and more rarely as various forms of glomerulonephritis (GN), namely segmental glomerulosclerosis, collapsing glomerulopathy, fibrillar, or membranoproliferative GN. Vascular disorders such as thrombosis or renal aneurysms and pseudoaneurysms may be encountered in patients with Behcet's disease. Patients with AIDs should be routinely assessed for renal involvement. Screening with urinalysis, serum creatinine, 24-h urinary protein, microhematuria, and imaging studies should be carried out for early diagnosis. Awareness of drug-induced nephrotoxicity, drug-drug interactions as well as addressing the issue of proper renal adjustment of drug doses deserve a special mention and should always be considered when dealing with patients affected by AIDs. Finally, we will explore the role of IL-1 inhibitors in AIDs patients with renal involvement. Targeting IL-1 may indeed have the potential to successfully manage kidney disease and improve long-term prognosis of AIDs patients.


Subject(s)
Amyloidosis , Glomerulonephritis, IGA , Hereditary Autoinflammatory Diseases , Humans , Kidney/pathology , Glomerulonephritis, IGA/pathology , Amyloidosis/pathology , Hereditary Autoinflammatory Diseases/diagnosis , Hereditary Autoinflammatory Diseases/pathology , Interleukin-1
3.
Front Immunol ; 13: 926175, 2022.
Article in English | MEDLINE | ID: mdl-35936010

ABSTRACT

Tumor necrosis factor (TNF) receptor-associated periodic syndrome (TRAPS) is an autoinflammatory periodic fever syndrome associated with heterozygous mutations in TNFRSF1A, which encodes TNF receptor type I (TNFR1). Although possible proinflammatory mechanisms have been proposed, most previous studies were performed using in vitro overexpression models, which could lead to undesirable inflammatory responses due to artificial overexpression. It is crucial to reproduce heterozygous mutations at physiological expression levels; however, such studies remain limited. In this study, we generated TRAPS mutant mice and analyzed their phenotypes. Three Tnfrsf1a mutant strains were generated by introducing T79M, G87V, or T90I mutation. T79M is a known mutation responsible for TRAPS, whereas G87V is a TRAPS mutation that we have reported, and T90I is a variant of unknown significance. Using these murine models, we investigated whether TRAPS mutations could affect the inflammatory responses in vivo and in vitro. We found that none of the mutant mice exhibited detectable inflammatory phenotypes under standard housing conditions for 1 year. Interestingly, TRAPS mutant (T79M and G87V) mice had reduced mortality rates after the administration of lipopolysaccharide (LPS) and D-galactosamine, which induce TNFα-dependent lethal hepatitis. Moreover, TRAPS mutations strongly suppressed the development of TNFα-mediated arthritis when crossed with human TNFα transgenic mice. In in vitro primary bone marrow-derived macrophage cultures, the T79M and G87V mutations attenuated the inflammatory responses to TNFα compared with the wild-type, whereas these mutations did not alter the responsiveness of these cells to LPS. The T90I mutant macrophages behaved similarly to wild type in response to LPS and TNFα. The TNFR1 levels were increased in whole-cell lysates of TRAPS mutant macrophages, whereas the cell surface expression of TNFR1 was significantly decreased in TRAPS mutant macrophages. Taken together, TRAPS mutations did not augment the inflammatory responses to TNFα and LPS; instead, they suppressed the response to TNFα via decreased cell surface expression of TNFR1. The stimulation of lymphotoxin-α, adenosine triphosphate, and norepinephrine in primary macrophages or various stimuli in murine splenocytes did not induce detectable inflammatory responses. In conclusion, TRAPS mutations suppressed responsiveness to TNFα, and TRAPS-associated inflammation is likely induced by unconfirmed disease-specific proinflammatory factors.


Subject(s)
Hereditary Autoinflammatory Diseases/pathology , Receptors, Tumor Necrosis Factor, Type I , Tumor Necrosis Factor-alpha , Animals , Fever , Hereditary Autoinflammatory Diseases/metabolism , Humans , Lipopolysaccharides , Mice , Mice, Transgenic , Mutation , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Syndrome , Tumor Necrosis Factor-alpha/metabolism
4.
Am J Dermatopathol ; 44(6): e64-e66, 2022 Jun 01.
Article in English | MEDLINE | ID: mdl-35120036

ABSTRACT

ABSTRACT: A 15-month-old full-term boy of African descent with an asymptomatic sickle cell trait presented with episodes of transient erythematous subcutaneous nodules involving the entire body except the face, since 2 weeks of age. The skin lesions evolved to areas of lipoatrophy and hyperpigmentation. An initial skin biopsy, studied at a different department at 2 months, was initially misinterpreted as subcutaneous fat necrosis of the newborn, despite the lack of the typical radiated crystals and needle-shaped clefts characterizing that entity. At 4 months of age, he developed systemic inflammatory manifestations, including fever, a new rash, significant periorbital edema, and failure to thrive. An extensive workup showed leukocytosis, hypercalcemia, elevated inflammatory markers, hypertriglyceridemia, and transaminitis. A new skin biopsy of the eyelid was diagnosed as neutrophilic lobular panniculitis with necrotic adipocytes. An initial whole-exome sequencing did not identify any causative mutations, but a WES reanalysis focused on autoinflammatory disorders was requested based on additional clinicopathologic data and revealed a mosaic intronic mutation in IKBKG c. 671+3 G > C. This mutation encodes an mRNA missing exon 5 resulting in NF-kB essential modulator (NEMO) Δ-exon 5-autoinflammatory syndrome (NDAS). NEMO-NDAS is one of the systemic autoinflammatory diseases that may appear as an unexplained panniculitis in young children, who should be monitored for immunodeficiency and/or autoinflammatory diseases. The differential diagnosis of autoinflammatory disorders should be considered in such cases incorporating the use of the whole-genome/exome sequencing in the investigation. The inhibitor of kappa-B kinase regulatory subunit gamma (IKBKG) is located on chromosome Xq28 and encodes the NEMO, a critical molecule upstream of NF-kB activation.


Subject(s)
Hereditary Autoinflammatory Diseases , Immunologic Deficiency Syndromes , Panniculitis , Child , Child, Preschool , Hereditary Autoinflammatory Diseases/diagnosis , Hereditary Autoinflammatory Diseases/genetics , Hereditary Autoinflammatory Diseases/pathology , Humans , I-kappa B Kinase/genetics , Immunologic Deficiency Syndromes/genetics , Infant , Infant, Newborn , Male , NF-kappa B , Panniculitis/genetics , Panniculitis/pathology , Skin/pathology
5.
Nat Commun ; 12(1): 6819, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34819510

ABSTRACT

Impaired proteasome activity due to genetic variants of certain subunits might lead to proteasome-associated autoinflammatory syndromes (PRAAS). Here we report a de novo heterozygous missense variant of the PSMB9 proteasome subunit gene in two unrelated Japanese infants resulting in amino acid substitution of the glycine (G) by aspartic acid (D) at position 156 of the encoded protein ß1i. In addition to PRAAS-like manifestations, these individuals suffer from pulmonary hypertension and immunodeficiency, which are distinct from typical PRAAS symptoms. The missense variant results in impaired immunoproteasome maturation and activity, yet ubiquitin accumulation is hardly detectable in the patients. A mouse model of the heterozygous human genetic variant (Psmb9G156D/+) recapitulates the proteasome defects and the immunodeficiency phenotype of patients. Structurally, PSMB9 G156D interferes with the ß-ring-ßring interaction of the wild type protein that is necessary for 20S proteasome formation. We propose the term, proteasome-associated autoinflammatory syndrome with immunodeficiency (PRAAS-ID), to indicate a separate category of autoinflammatory diseases, similar to, but distinct from PRAAS, that describes the patients in this study.


Subject(s)
Cysteine Endopeptidases/genetics , Hereditary Autoinflammatory Diseases/genetics , Hypertension, Pulmonary/genetics , Primary Immunodeficiency Diseases/genetics , Proteasome Endopeptidase Complex/metabolism , Animals , Cysteine Endopeptidases/metabolism , Disease Models, Animal , Female , Hereditary Autoinflammatory Diseases/diagnosis , Hereditary Autoinflammatory Diseases/immunology , Hereditary Autoinflammatory Diseases/pathology , Heterozygote , Humans , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/immunology , Infant, Newborn , Male , Mice , Mice, Transgenic , Mutation, Missense , Pedigree , Primary Immunodeficiency Diseases/diagnosis , Primary Immunodeficiency Diseases/immunology , Primary Immunodeficiency Diseases/pathology , Proteasome Endopeptidase Complex/genetics , Syndrome
6.
Nat Immunol ; 22(9): 1118-1126, 2021 09.
Article in English | MEDLINE | ID: mdl-34326534

ABSTRACT

Transcription factors specialized to limit the destructive potential of inflammatory immune cells remain ill-defined. We discovered loss-of-function variants in the X-linked ETS transcription factor gene ELF4 in multiple unrelated male patients with early onset mucosal autoinflammation and inflammatory bowel disease (IBD) characteristics, including fevers and ulcers that responded to interleukin-1 (IL-1), tumor necrosis factor or IL-12p40 blockade. Using cells from patients and newly generated mouse models, we uncovered ELF4-mutant macrophages having hyperinflammatory responses to a range of innate stimuli. In mouse macrophages, Elf4 both sustained the expression of anti-inflammatory genes, such as Il1rn, and limited the upregulation of inflammation amplifiers, including S100A8, Lcn2, Trem1 and neutrophil chemoattractants. Blockade of Trem1 reversed inflammation and intestine pathology after in vivo lipopolysaccharide challenge in mice carrying patient-derived variants in Elf4. Thus, ELF4 restrains inflammation and protects against mucosal disease, a discovery with broad translational relevance for human inflammatory disorders such as IBD.


Subject(s)
DNA-Binding Proteins/genetics , Hereditary Autoinflammatory Diseases/genetics , Inflammatory Bowel Diseases/genetics , Macrophages/immunology , Transcription Factors/genetics , Animals , Calgranulin A/metabolism , Female , Gene Expression Regulation/genetics , Hereditary Autoinflammatory Diseases/immunology , Hereditary Autoinflammatory Diseases/pathology , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Interleukin 1 Receptor Antagonist Protein/immunology , Lipocalin-2/metabolism , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Th17 Cells/immunology , Transcription, Genetic/genetics , Triggering Receptor Expressed on Myeloid Cells-1/antagonists & inhibitors , Triggering Receptor Expressed on Myeloid Cells-1/metabolism
7.
Nat Rev Rheumatol ; 17(7): 387-404, 2021 07.
Article in English | MEDLINE | ID: mdl-34113018

ABSTRACT

Ankylosing spondylitis (AS) is a chronic inflammatory disorder of unknown aetiology. Unlike other systemic autoimmune diseases, in AS, the innate immune system has a dominant role characterized by aberrant activity of innate and innate-like immune cells, including γδ T cells, group 3 innate lymphoid cells, neutrophils, mucosal-associated invariant T cells and mast cells, at sites predisposed to the disease. The intestine is involved in disease manifestations, as it is at the forefront of the interaction between the mucosal-associated immune cells and the intestinal microbiota. Similarly, biomechanical factors, such as entheseal micro-trauma, might also be involved in the pathogenesis of the articular manifestation of AS, and sentinel immune cells located in the entheses could provide links between local damage, genetic predisposition and the development of chronic inflammation. Although these elements might support the autoinflammatory nature of AS, studies demonstrating the presence of autoantibodies (such as anti-CD74, anti-sclerostin and anti-noggin antibodies) and evidence of activation and clonal expansion of T cell populations support an autoimmune component to the disease. This Review presents the evidence for autoinflammation and the evidence for autoimmunity in AS and, by discussing the pathophysiological factors associated with each, aims to reconcile the two hypotheses.


Subject(s)
Autoimmune Diseases/etiology , Hereditary Autoinflammatory Diseases/etiology , Spondylitis, Ankylosing/etiology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Hereditary Autoinflammatory Diseases/immunology , Hereditary Autoinflammatory Diseases/pathology , Humans , Spondylitis, Ankylosing/immunology , Spondylitis, Ankylosing/pathology
8.
Nat Rev Rheumatol ; 17(7): 405-425, 2021 07.
Article in English | MEDLINE | ID: mdl-34035534

ABSTRACT

Monogenic autoinflammatory diseases are a group of rheumatologic disorders caused by dysregulation in the innate immune system. The molecular mechanisms of these disorders are linked to defects in inflammasome-mediated, NF-κB-mediated or interferon-mediated inflammatory signalling pathways, cytokine receptors, the actin cytoskeleton, proteasome complexes and various enzymes. As with other human disorders, disease-causing variants in a single gene can present with variable expressivity and incomplete penetrance. In some cases, pathogenic variants in the same gene can be inherited either in a recessive or dominant manner and can cause distinct and seemingly unrelated phenotypes, although they have a unifying biochemical mechanism. With an enhanced understanding of protein structure and functionality of protein domains, genotype-phenotype correlations are beginning to be unravelled. Many of the mutated proteins are primarily expressed in haematopoietic cells, and their malfunction leads to systemic inflammation. Disease presentation is also defined by a specific effect of the mutant protein in a particular cell type and, therefore, the resulting phenotype might be more deleterious in one tissue than in another. Many patients present with the expanded immunological disease continuum that includes autoinflammation, immunodeficiency, autoimmunity and atopy, which necessitate genetic testing.


Subject(s)
Hereditary Autoinflammatory Diseases/genetics , Hereditary Autoinflammatory Diseases/immunology , Hereditary Autoinflammatory Diseases/pathology , Humans , Inflammasomes/physiology , Interferons/physiology , Phenotype
10.
Eur J Med Genet ; 64(5): 104191, 2021 May.
Article in English | MEDLINE | ID: mdl-33753323

ABSTRACT

Familial periodic fever (FPF) is an uncommonly diagnosed autosomal dominant disorder caused by a genetic alteration in the TNFRSF1A gene. These patients usually present with fever which is usually under-investigated and under-diagnosed. In untreated cases, amyloidosis is a frequent complication. We present a 24 years male who had a history of fever from childhood, however, remained undiagnosed short of genetic testing. He has recurrent episodes of fever. During the episodes of fever, he was found to have leukocytosis (total leukocyte count- 25.7 x10^9/L) and neutrophilia (absolute neutrophil count- 22.7 x10^9/L) both of which came back to normal limits as the fever subsided. On further evaluation for neutrophilia, the exclusion of common causes of neutrophilia was done. Next-generation sequencing detected a missense variant in TNFRSF1A: c.215G > A (p.Cys72Tyr) which was confirmed by Sanger sequencing. This variant has been described in the literature in anecdotal cases of FPF. This is a first case report from the Indian subcontinent reporting TNFRSF1A: c.215G > A (p.Cys72Tyr) variant in a patient of FPF. Short of genetic testing, the fever would remain a diagnostic dilemma in this patient. This report highlights the importance of targeted resequencing in clinching diagnosis in such patients.


Subject(s)
Fever/genetics , Hereditary Autoinflammatory Diseases/genetics , Leukocyte Disorders/congenital , Receptors, Tumor Necrosis Factor, Type I/genetics , Fever/pathology , Hereditary Autoinflammatory Diseases/pathology , Humans , Leukocyte Disorders/genetics , Leukocyte Disorders/pathology , Male , Mutation, Missense , Young Adult
11.
Laryngoscope ; 131(7): E2149-E2152, 2021 07.
Article in English | MEDLINE | ID: mdl-33630321

ABSTRACT

OBJECTIVES/HYPOTHESIS: Tonsillectomy is an effective treatment for periodic fever, aphthous stomatitis, pharyngitis, and cervical adenitis (PFAPA) syndrome, but the role of adenoidectomy, as well as later tonsillar regrowth, is unclear. To find out if the volume of lymphoid tissue is pivotal to the efficacy, we analyzed the association between the relapse of the symptoms of PFAPA syndrome and regrowth of tonsillar tissue after tonsillectomy or adenotonsillectomy. STUDY DESIGN: Prospective cohort study of operated PFAPA pateints. METHODS: We invited all patients that had undergone tonsillectomy or adenotonsillectomy due to PFAPA syndrome at the Oulu University Hospital, Oulu, Finland, between the years 1990 and 2007, at the age of ≤12 years, to a follow-up visit, after an average period of 9.8 years after their diagnoses. Out of the 132 invited, 94 (71%) participated in the follow-up study. RESULTS: At the follow-up study visit, 5 (5%) of the 94 PFAPA syndrome cases experienced recurrent fevers. The regrowth of palatine tonsillar tissue was seen in four of them (80%) as compared to 19/89 (21%) of symptom-free patients (P = .006). Two of the patients with clear PFAPA relapse at the time of the study visit were reoperated with clear effect on the symptoms. At the time of the study visit, 59/63 (94%) of the patients who had undergone adenotonsillectomy and 30/31 of the patients (97%) who had undergone tonsillectomy earlier were free of fever flares (P = .99). CONCLUSION: Palatine tonsil regrowth was associated with PFAPA syndrome relapse after tonsillectomy. Reoperation might be a treatment option in these patients. LEVEL OF EVIDENCE: 4 Laryngoscope, 131:E2149-E2152, 2021.


Subject(s)
Hereditary Autoinflammatory Diseases/surgery , Lymphadenitis/surgery , Pharyngitis/surgery , Stomatitis, Aphthous/surgery , Tonsillectomy/adverse effects , Adenoidectomy/adverse effects , Child , Female , Finland/epidemiology , Follow-Up Studies , Hereditary Autoinflammatory Diseases/pathology , Humans , Lymphadenitis/pathology , Male , Neck/pathology , Palatine Tonsil/growth & development , Palatine Tonsil/pathology , Palatine Tonsil/surgery , Pharyngitis/pathology , Postoperative Period , Prospective Studies , Recurrence , Stomatitis, Aphthous/pathology , Syndrome , Treatment Outcome
12.
Sci Rep ; 11(1): 4172, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33603056

ABSTRACT

Binding of tumour necrosis factor α (TNFα) to its receptor (TNFR1) is critical for both survival and death cellular pathways. TNFα/TNFR1 signalling is complex and tightly regulated at different levels to control cell fate decisions. Previously, we identified TNFR1-d2, an exon 2-spliced transcript of TNFRSF1A gene encoding TNFR1, whose splicing may be modulated by polymorphisms associated with inflammatory disorders. Here, we investigated the impact of TNFRSF1A variants involved in TNFR-associated periodic syndrome (TRAPS) on TNFR1-d2 protein expression and activity. We found that TNFR1-d2 could be translated by using an internal translation initiation codon and a de novo internal ribosome entry site (IRES), which resulted in a putative TNFR1 isoform lacking its N-terminal region. The kinetic of assembly of TNFR1-d2 clusters at the cell surface was reduced as compared with full-length TNFR1. Although co-localized with the full-length TNFR1, TNFR1-d2 neither activated nuclear factor (NF)-κB signalling, nor interfered with TNFR1-induced NF-κB activation. Translation of TNFR1-d2 carrying the severe p.(Thr79Met) pathogenic variant (also known as T50M) was initiated at the mutated codon, resulting in an elongated extracellular domain, increased speed to form preassembled clusters in absence of TNFα, and constitutive NF-κB activation. Overall, TNFR1-d2 might reflect the complexity of the TNFR1 signalling pathways and could be involved in TRAPS pathophysiology of patients carrying the p.(Thr79Met) disease-causing variant.


Subject(s)
Hereditary Autoinflammatory Diseases/genetics , Hereditary Autoinflammatory Diseases/pathology , Mutation/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/genetics , Cell Line , Cell Line, Tumor , Exons/genetics , HEK293 Cells , HeLa Cells , Humans , NF-kappa B/genetics
16.
Int J Mol Sci ; 21(22)2020 Nov 23.
Article in English | MEDLINE | ID: mdl-33238371

ABSTRACT

Notch pathway is a highly conserved intracellular signaling route that modulates a vast variety of cellular processes including proliferation, differentiation, migration, cell fate and death. Recently, the presence of a strict crosstalk between Notch signaling and inflammation has been described, although the precise molecular mechanisms underlying this interplay have not yet been fully unravelled. Disruptions in Notch cascade, due both to direct mutations and/or to an altered regulation in the core components of Notch signaling, might lead to hypo- or hyperactivation of Notch target genes and signaling molecules, ultimately contributing to the onset of autoinflammatory diseases. To date, alterations in Notch signaling have been reported as associated with three autoinflammatory disorders, therefore, suggesting a possible role of Notch in the pathogenesis of the following diseases: hidradenitis suppurativa (HS), Behçet disease (BD), and giant cell arteritis (GCA). In this review, we aim at better characterizing the interplay between Notch and autoinflammatory diseases, trying to identify the role of this signaling route in the context of these disorders.


Subject(s)
Hereditary Autoinflammatory Diseases/genetics , Inflammation/genetics , Receptors, Notch/genetics , Animals , Behcet Syndrome/genetics , Behcet Syndrome/pathology , Cell Differentiation/genetics , Giant Cell Arteritis/genetics , Giant Cell Arteritis/pathology , Hereditary Autoinflammatory Diseases/classification , Hereditary Autoinflammatory Diseases/pathology , Hidradenitis Suppurativa/genetics , Hidradenitis Suppurativa/pathology , Humans , Inflammation/pathology , Mutation/genetics , Signal Transduction/genetics
17.
Clin Exp Dermatol ; 45(8): 962-966, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32901977

ABSTRACT

The systemic autoinflammatory disorders (SAIDs) or periodic fever syndromes are disorders of innate immunity, which can be inherited or acquired. They are almost all very rare and easily overlooked; typically, patients will have seen multiple specialities prior to diagnosis, so a high level of clinical suspicion is key. It is important to note that these are 'high-value' diagnoses as the majority of these syndromes can be very effectively controlled, dramatically improving quality of life and providing protection against the development of irreversible complications such as AA amyloidosis. In this article, we take an overview of SAIDs and look at the common features; in Part 2, we take a more in-depth look at the better recognized or more dermatologically relevant conditions.


Subject(s)
Amyloidosis/prevention & control , Dermatologists/statistics & numerical data , Hereditary Autoinflammatory Diseases/immunology , Immune System Diseases/immunology , Amyloidosis/etiology , Amyloidosis/pathology , Cryopyrin-Associated Periodic Syndromes/diagnosis , Cryopyrin-Associated Periodic Syndromes/metabolism , Cryopyrin-Associated Periodic Syndromes/pathology , Diagnosis, Differential , Familial Mediterranean Fever/diagnosis , Familial Mediterranean Fever/metabolism , Familial Mediterranean Fever/pathology , Fever/diagnosis , Fever/metabolism , Fever/pathology , Genetic Testing/standards , Hereditary Autoinflammatory Diseases/diagnosis , Hereditary Autoinflammatory Diseases/metabolism , Hereditary Autoinflammatory Diseases/pathology , Humans , Immune System Diseases/complications , Immune System Diseases/diagnosis , Immune System Diseases/pathology , Immunity, Innate/immunology , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Mevalonate Kinase Deficiency/diagnosis , Mevalonate Kinase Deficiency/metabolism , Mevalonate Kinase Deficiency/pathology , Quality of Life , Severity of Illness Index
18.
Clin Exp Dermatol ; 45(8): 967-973, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32882069

ABSTRACT

The systemic autoinflammatory disorders (SAIDS) or periodic fever syndromes are disorders of innate immunity, which can be inherited or acquired. They are almost all very rare and easily overlooked; typically, patients will have seen multiple specialities prior to diagnosis, so a high level of clinical suspicion is key. It is important to note that these are 'high-value' diagnoses as the majority of these syndromes can be very effectively controlled, dramatically improving quality of life and providing protection against the development of irreversible complications such as AA amyloidosis. In Part 1 of this review, we took an overview of SAIDS and described the common features; in this article, we take a more in-depth look at the better recognized or more dermatologically relevant conditions.


Subject(s)
Amyloidosis/prevention & control , Dermatologists/statistics & numerical data , Hereditary Autoinflammatory Diseases/immunology , Immune System Diseases/immunology , Receptors, Interleukin-1/deficiency , Amyloidosis/etiology , Amyloidosis/pathology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biological Products/therapeutic use , Colchicine/therapeutic use , Cryopyrin-Associated Periodic Syndromes/diagnosis , Cryopyrin-Associated Periodic Syndromes/drug therapy , Cryopyrin-Associated Periodic Syndromes/genetics , Cryopyrin-Associated Periodic Syndromes/pathology , Familial Mediterranean Fever/diagnosis , Familial Mediterranean Fever/drug therapy , Familial Mediterranean Fever/genetics , Familial Mediterranean Fever/pathology , Fever/diagnosis , Fever/drug therapy , Fever/genetics , Fever/pathology , Hereditary Autoinflammatory Diseases/diagnosis , Hereditary Autoinflammatory Diseases/drug therapy , Hereditary Autoinflammatory Diseases/genetics , Hereditary Autoinflammatory Diseases/pathology , Humans , Immune System Diseases/complications , Immune System Diseases/diagnosis , Immune System Diseases/pathology , Immunity, Innate/genetics , Immunity, Innate/immunology , Inflammation/pathology , Interleukin 1 Receptor Antagonist Protein/adverse effects , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Interleukin-1/immunology , Interleukin-1/metabolism , Mevalonate Kinase Deficiency/diagnosis , Mevalonate Kinase Deficiency/drug therapy , Mevalonate Kinase Deficiency/genetics , Mevalonate Kinase Deficiency/pathology , Receptors, Interleukin-1/drug effects , Receptors, Interleukin-1/genetics , Receptors, Interleukin-6/antagonists & inhibitors , Receptors, Interleukin-6/therapeutic use , Schnitzler Syndrome/diagnosis , Schnitzler Syndrome/drug therapy , Schnitzler Syndrome/immunology , Schnitzler Syndrome/pathology , Skin Diseases, Genetic/diagnosis , Skin Diseases, Genetic/immunology , Skin Diseases, Genetic/pathology , Steroids/therapeutic use , Tubulin Modulators/therapeutic use
19.
Immunity ; 53(3): 672-684.e11, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32750333

ABSTRACT

Autoinflammatory disease can result from monogenic errors of immunity. We describe a patient with early-onset multi-organ immune dysregulation resulting from a mosaic, gain-of-function mutation (S703I) in JAK1, encoding a kinase essential for signaling downstream of >25 cytokines. By custom single-cell RNA sequencing, we examine mosaicism with single-cell resolution. We find that JAK1 transcription was predominantly restricted to a single allele across different cells, introducing the concept of a mutational "transcriptotype" that differs from the genotype. Functionally, the mutation increases JAK1 activity and transactivates partnering JAKs, independent of its catalytic domain. S703I JAK1 is not only hypermorphic for cytokine signaling but also neomorphic, as it enables signaling cascades not canonically mediated by JAK1. Given these results, the patient was treated with tofacitinib, a JAK inhibitor, leading to the rapid resolution of clinical disease. These findings offer a platform for personalized medicine with the concurrent discovery of fundamental biological principles.


Subject(s)
Hereditary Autoinflammatory Diseases/genetics , Hereditary Autoinflammatory Diseases/pathology , Janus Kinase 1/genetics , Systemic Inflammatory Response Syndrome/genetics , Systemic Inflammatory Response Syndrome/pathology , Adolescent , COVID-19/mortality , Catalytic Domain/genetics , Cell Line , Cytokines/metabolism , Female , Gain of Function Mutation/genetics , Genotype , HEK293 Cells , Hereditary Autoinflammatory Diseases/drug therapy , Humans , Janus Kinase 1/antagonists & inhibitors , Mosaicism , Piperidines/therapeutic use , Precision Medicine/methods , Pyrimidines/therapeutic use , Signal Transduction/immunology , Systemic Inflammatory Response Syndrome/drug therapy
20.
J Evid Based Med ; 13(3): 227-245, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32627322

ABSTRACT

Disruption of innate immunity leading to systemic inflammation and multi-organ dysfunction is the basilar footprint of autoinflammatory disorders (AIDs), ranging from rare hereditary monogenic diseases to a large number of common chronic inflammatory conditions in which there is a simultaneous participation of multiple genetic components and environmental factors, sometimes combined with autoimmune phenomena and immunodeficiency. Whatever their molecular mechanism, hereditary AIDs are caused by mutations in regulatory molecules or sensors proteins leading to dysregulated production of proinflammatory cytokines or cytokine-inducing transcription factors, fever, elevation of acute phase reactants, and a portfolio of manifold inflammatory signs which might occur in a stereotyped manner, mostly with overactivity or misactivation of different inflammasomes. Symptoms might overlap in the pediatric patient, obscuring the final diagnosis of AIDs and delaying the most appropriate treatment. Actually, the fast-paced evolution of scientific knowledge has led to recognize or reclassify an overgrowing number of multifactorial diseases, which share the basic pathogenetic mechanisms with AIDs. The wide framework of classic hereditary periodic fevers, AIDs with prominent skin involvement, disorders of the ubiquitin-proteasome system, defects of actin cytoskeleton dynamics, and also idiopathic nonhereditary febrile syndromes occurring in children is herein presented. Interleukin-1 dependence of these diseases or involvement of other predominating molecules is also discussed.


Subject(s)
Autoimmune Diseases/pathology , Hereditary Autoinflammatory Diseases/pathology , Acquired Immunodeficiency Syndrome/pathology , Child , Cryopyrin-Associated Periodic Syndromes/pathology , Familial Mediterranean Fever/pathology , Humans , Inflammation/pathology , Mevalonate Kinase Deficiency/pathology , Phenotype , Skin/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...