1
|
Chen PK, Tang KT, Chen DY. The NLRP3 Inflammasome as a Pathogenic Player Showing Therapeutic Potential in Rheumatoid Arthritis and Its Comorbidities: A Narrative Review. Int J Mol Sci 2024; 25:626. [PMID: 38203796 PMCID: PMC10779699 DOI: 10.3390/ijms25010626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 12/24/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune inflammatory disease characterized by chronic synovitis and the progressive destruction of cartilage and bone. RA is commonly accompanied by extra-articular comorbidities. The pathogenesis of RA and its comorbidities is complex and not completely elucidated. The assembly of the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome activates caspase-1, which induces the maturation of interleukin (IL)-1β and IL-18 and leads to the cleavage of gasdermin D with promoting pyroptosis. Accumulative evidence indicates the pathogenic role of NLRP3 inflammasome signaling in RA and its comorbidities, including atherosclerotic cardiovascular disease, osteoporosis, and interstitial lung diseases. Although the available therapeutic agents are effective for RA treatment, their high cost and increased infection rate are causes for concern. Recent evidence revealed the components of the NLRP3 inflammasome as potential therapeutic targets in RA and its comorbidities. In this review, we searched the MEDLINE database using the PubMed interface and reviewed English-language literature on the NLRP3 inflammasome in RA and its comorbidities from 2000 to 2023. The current evidence reveals that the NLRP3 inflammasome contributes to the pathogenesis of RA and its comorbidities. Consequently, the components of the NLRP3 inflammasome signaling pathway represent promising therapeutic targets, and ongoing research might lead to the development of new, effective treatments for RA and its comorbidities.
Collapse
Affiliation(s)
- Po-Ku Chen
- Rheumatology and Immunology Center, China Medical University Hospital, No. 2, Yude Road, Taichung 40447, Taiwan;
- College of Medicine, China Medical University, Taichung 40447, Taiwan
- Translational Medicine Laboratory, Rheumatology and Immunology Center, Taichung 40447, Taiwan
| | - Kuo-Tung Tang
- College of Medicine, National Chung Hsing University, Taichung 402202, Taiwan;
- Division of Allergy, Immunology, and Rheumatology, Taichung Veterans General Hospital, Taichung 40705, Taiwan
- Faculty of Medicine, National Yang-Ming University, Taipei 112304, Taiwan
| | - Der-Yuan Chen
- Rheumatology and Immunology Center, China Medical University Hospital, No. 2, Yude Road, Taichung 40447, Taiwan;
- College of Medicine, China Medical University, Taichung 40447, Taiwan
- Translational Medicine Laboratory, Rheumatology and Immunology Center, Taichung 40447, Taiwan
- College of Medicine, National Chung Hsing University, Taichung 402202, Taiwan;
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| |
Collapse
|
2
|
Hansen MS, Terslev L, Jensen MR, Brittain JM, Døhn UM, Faber C, Heegaard S, Klefter ON, Kønig EB, Subhi Y, Wiencke AK, Hamann S. Comparison of temporal artery ultrasound versus biopsy in the diagnosis of giant cell arteritis. Eye (Lond) 2023; 37:344-349. [PMID: 35094027 PMCID: PMC9873813 DOI: 10.1038/s41433-022-01947-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 01/13/2022] [Accepted: 01/17/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND/OBJECTIVES Giant cell arteritis (GCA) is a medical and ophthalmological emergency due to risk of stroke and sudden irreversible loss of vision. Fast and accurate diagnosis is important to prevent complications and long-term high dose glucocorticoids toxicity. Temporal artery biopsy is gold standard for diagnosing GCA. However, temporal artery ultrasound is a fast and non-invasive procedure which may provide a supplement or an alternative to biopsy. This study assesses the diagnostic performance of ultrasound and biopsy in the diagnosis of GCA. SUBJECTS/METHODS Examination results of patients suspected of having GCA in the period from August 2018 to June 2019 were reviewed. Patients underwent clinical examination and blood tests. Within a few days of starting glucocorticoid treatment, temporal ultrasound and unilateral biopsy were performed. Experienced physicians established the final clinical diagnosis at 6-months follow-up. RESULTS Seventy-eight patients underwent both ultrasound and biopsy. Thirty-five (45%) received the final clinical diagnosis of GCA. Compared with the final clinical diagnosis, biopsy had a sensitivity of 69% (51-83%) and a specificity of 100% (92-100%), and ultrasound a sensitivity of 63% (45-79%) and a specificity of 79% (64-94%). Area under the receiver operating characteristics curves were 0.84 and 0.71 for biopsy and ultrasound respectively (p = 0.048). False negative rate of ultrasound was 4 out of 78 (5%). CONCLUSION Sensitivity of ultrasound is almost on par with that of biopsy although the overall diagnostic accuracy of ultrasound was slightly lower. We find that ultrasound is a reliable tool for first line diagnosis of GCA.
Collapse
Affiliation(s)
- Michael Stormly Hansen
- Department of Ophthalmology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| | - Lene Terslev
- Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen, Denmark
| | - Mads Radmer Jensen
- Department of Clinical Physiology and Nuclear Medicine, Bispebjerg & Frederiksberg Hospital, Copenhagen, Denmark
| | - Jane Maestri Brittain
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Uffe Møller Døhn
- Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen, Denmark
| | - Carsten Faber
- Department of Ophthalmology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Steffen Heegaard
- Department of Ophthalmology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Eye Pathology Section, Department of Pathology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Oliver Niels Klefter
- Department of Ophthalmology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Elisabeth Bay Kønig
- Eye Pathology Section, Department of Pathology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Yousif Subhi
- Department of Ophthalmology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Anne Katrine Wiencke
- Department of Ophthalmology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Steffen Hamann
- Department of Ophthalmology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
3
|
Carta S, Gattorno M, Rubartelli A. Methods to Study NLR in Human Blood Cells. Methods Mol Biol 2023; 2696:115-122. [PMID: 37578719 DOI: 10.1007/978-1-0716-3350-2_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Autoinflammatory diseases are a group of inherited and multifactorial disorders characterized by an over-activation of innate immune response. In most cases, the clinical manifestations are due to increased activity of the NLRP3 inflammasome resulting in increased IL-1β secretion. Investigating inflammatory cells from subjects affected by autoinflammatory diseases presents a number of technical difficulties related to the rarity of the diseases, to the young age of most patients, to the difficult modulation of gene expression in primary cells. However, since cell stress is involved in the pathophysiology of these diseases, the study of freshly drawn blood monocytes from patients affected by IL-1-mediated diseases strongly increases the chances that the observed phenomena is indeed pertinent to the pathogenesis of the disease and not influenced by the long-term cell culture conditions (e.g., the high O2 tension) or gene transfection in continuous cell lines that may lead to artifacts.
Collapse
Affiliation(s)
- Sonia Carta
- Cell Biology Unit, IRCCS Azienda Ospedaliera Universitaria San Martino-IST, Genoa, Italy
| | | | | |
Collapse
|
4
|
Bertoni A, Prigione I, Chiesa S, Ceccherini I, Gattorno M, Rubartelli A. A Knock-In Mouse Model of Cryopyrin-Associated Periodic Syndromes. Methods Mol Biol 2023; 2696:281-297. [PMID: 37578730 DOI: 10.1007/978-1-0716-3350-2_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Autoinflammatory diseases are a group of distinct disorders characterized by recurrent fever and inflammatory manifestations predominantly mediated by cytokines of the innate immune system, particularly IL-1β, without involvement of autoantibodies or autoreactive T lymphocytes. Cryopyrin-associated periodic syndromes (CAPS), due to NLRP3 gene mutations, represent the prototype of these diseases. Owing to their genetic nature, most of these disorders have an early onset, ranging from the first hours to the first decade of life. Due to the rarity of CAPS patients and to the limitations of working with pediatric samples, the development of animal models of this disease is of great help for studying both pathophysiology and therapeutic strategies. In this chapter, we review the generation and characterization of a knock-in mouse bearing the NLRP3 gene with the N475K mutation, associated with CINCA, the most severe form of human CAPS.
Collapse
Affiliation(s)
- Arinna Bertoni
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy.
| | - Ignazia Prigione
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Sabrina Chiesa
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Marco Gattorno
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Anna Rubartelli
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
5
|
Increased Lipid Peroxidation May Be Linked to Ferritin Levels Elevation in Adult-Onset Still's Disease. Biomedicines 2021; 9:biomedicines9111508. [PMID: 34829738 PMCID: PMC8614840 DOI: 10.3390/biomedicines9111508] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/13/2021] [Accepted: 10/18/2021] [Indexed: 12/24/2022] Open
Abstract
Lipid peroxidation (LPO) and hyper-ferritinemia are involved in inflammatory responses. Although hyper-ferritinemia is a characteristic of AOSD, its link to LPO remains unclear. We investigated the association between LPO and ferritin expression, and evaluated the relationship between LPO-related metabolites and inflammatory parameters. Mean fluorescence intensity (MFI) of LPO (C11-Biodipy581/591)-expressing PBMCs/monocytes in AOSD patients and healthy control (HC) subjects was determined by flow-cytometry analysis. Expression of ferritin and cytokines on PBMCs/macrophages was examined by immunoblotting. Plasma levels of LPO-related metabolites and cytokines were determined by ELISA and the MULTIPLEX platform, respectively. LPO MFI on PBMCs/monocytes were significantly higher in patients (median 4456 and 9091, respectively) compared with HC (1900, p < 0.05, and 4551, p < 0.01, respectively). Patients had higher ferritin expression on PBMCs (mean fold, 1.02) than HC (0.55, p < 0.05). Their ferritin expression levels on PBMCs stimulated with LPO inducers erastin or RSL3 (2.47 or 1.61, respectively) were higher than HC (0.84, p < 0.05, or 0.74, p < 0.01). Ferritin expression on erastin-treated/IL-1β-treated macrophages from patients were higher than those from HC (p < 0.001). The elevated levels of LPO-related metabolites, including malondialdehyde and 4-hydroxyalkenals, were positively correlated with disease activity scores, suggesting LPO involvement in AOSD pathogenesis. Increased ferritin expression on PBMCs/macrophages stimulated with LPO inducers indicates a link between LPO and elevated ferritin.
Collapse
|
6
|
CARD8 SNP rs11672725 Identified as a Potential Genetic Variant for Adult-Onset Still's Disease. Life (Basel) 2021; 11:life11050382. [PMID: 33922655 PMCID: PMC8146669 DOI: 10.3390/life11050382] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/19/2021] [Accepted: 04/21/2021] [Indexed: 11/16/2022] Open
Abstract
Adult-onset Still's disease (AOSD), an autoinflammatory disorder, is related to the dysregulation of NLR3-containing a pyrin domain (NLRP3)-inflammasome signaling. We aimed to investigate the associations of genetic polymorphisms of NLRP3-inflammasome signaling with AOSD susceptibility and outcome and to examine their functional property. Fifty-three candidate single-nucleotide polymorphisms (SNPs) involved in NLRP3-inflammasome response were genotyped using Sequenom MassArray on the samples from 66 AOSD patients and 128 healthy controls. The significant SNPs were validated by direct sequencing using a TaqMan SNP analyzer. Serum levels of associated gene products were examined by ELISA. One SNP rs11672725 of CARD8 gene was identified to be significantly associated with AOSD susceptibility by using MassArray and subsequent replication validation (p = 3.57 × 10-7; odds ratio 3.02). Functional assays showed that serum CARD8 levels were significantly lower in AOSD patients (median, 10,524.6 pg/mL) compared to controls (13,964.1 pg/mL, p = 0.005), while levels of caspase-1, IL-1β and IL-18 were significantly higher in patients (107.1 pg/mL, 2.1 pg/mL, and 1495.8 pg/mL, respectively) than those in controls (99.0 pg/mL, 1.0 pg/mL, and 141.4 pg/mL, respectively). Patients carrying rs11672725CC genotype had significantly higher serum caspase-1 and IL-18 levels (121.3 pg/mL and 1748.6 pg/mL) compared to those with CT/TT genotypes (72.6 pg/mL, p = 0.019 and 609.3 pg/mL, p = 0.046). A higher proportion of patients with rs11672725CC genotype had a systemic pattern of disease outcome, which was linked to low CARD8 levels. A novel variant, rs11672725, of the CARD8 gene was identified as a potential genetic risk for AOSD. Patients carrying the rs11672725CC genotype and C allele had low CARD8 levels, and were predisposed to a systemic pattern with an elevated expression of inflammasome signaling.
Collapse
|
7
|
Huang JH, Chiang BL. Regulatory T cells induced by B cells suppress NLRP3 inflammasome activation and alleviate monosodium urate-induced gouty inflammation. iScience 2021; 24:102103. [PMID: 33615201 PMCID: PMC7881254 DOI: 10.1016/j.isci.2021.102103] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/25/2020] [Accepted: 01/21/2021] [Indexed: 01/22/2023] Open
Abstract
Regulatory T cells induced by B cells (Treg-of-B cells), a distinct Foxp3- Treg cell subset, have established the roles in the suppression of inflammatory conditions, including asthma and intestinal inflammation. However, little is known about the regulatory effects of Treg-of-B cells on innate immunity. Herein, we examined whether Treg-of-B cells could regulate macrophage function and prevent NLRP3-associated diseases, particularly inflammatory gouty arthritis. Treg-of-B cells, but not thymus-derived Treg or effector T cells, inhibited inflammasome-mediated IL-1β secretion, caspase-1 activation, and NLRP3 production by LPS/ATP stimulation in a cell contact-dependent manner. In addition, Treg-of-B cells inhibited monosodium urate-induced NLRP3 inflammasome activation in vitro via NF-κB signaling. Treg-of-B cells ameliorated gouty inflammation in a mouse air pouch model by reducing neutrophil and leukocyte influx and cytokine and chemokine production. Our results demonstrated that Treg-of-B cells exerted regulatory effects on innate immunity by suppressing NLRP3 inflammasome activation and feasible for future therapeutic applications.
Collapse
Affiliation(s)
- Jing-Hui Huang
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Bor-Luen Chiang
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan.,Department of Pediatrics, National Taiwan University Hospital, Taipei 10041, Taiwan.,Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| |
Collapse
|
8
|
Elevated Expression of C-Type Lectin Domain Family 5-Member A (CLEC5A) and Its Relation to Inflammatory Parameters and Disease Course in Adult-Onset Still's Disease. J Immunol Res 2020; 2020:9473497. [PMID: 32377540 PMCID: PMC7195645 DOI: 10.1155/2020/9473497] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/07/2020] [Indexed: 12/15/2022] Open
Abstract
C-type lectin domain family 5-member A (CLEC5A) associates with adaptor DAP12 (DNAX activation protein 12) to form receptor complexes involved in inflammatory responses. We postulated a potential role of CLEC5A in the pathogenesis of adult-onset Still's disease (AOSD) and aimed to investigate CLEC5A expression and its association with activity parameters and disease course. In 34 AOSD patients and 12 healthy controls (HC), circulating levels of CLEC5A-expressing monocytes or granulocytes were determined by flow cytometry analysis, the mRNA expression of CLEC5A and DAP12 on PBMCs by quantitative PCR, and plasma levels of proinflammatory cytokines by ELISA. AOSD patients had significantly higher percentages and mean fluorescence intensity (MFI) of CLEC5A-expressing monocytes (median 62.1% and 3.20, respectively) or granulocytes (72.6% and 3.22, respectively) compared with HC (in monocytes: 17.0% and 0.65, both p < 0.001; in granulocytes: 67.3%, p < 0.05 and 0.90, p < 0.001; respectively). Patients also had significantly higher levels of CLEC5A mRNA expression on PBMCs compared with HC (median 1.77 vs. 0.68, p < 0.05). The levels of CLEC5A-expressing monocytes or granulocytes were positively associated with activity scores and levels of IL-1β and IL-18 in AOSD patients. The patients with a systemic pattern had significantly higher levels of CLEC5A-expressing granulocytes and IL-18 compared to those with a chronic articular pattern of disease course. After 6 months of therapy, levels of CLEC5A-expressing monocytes and granulocytes significantly declined, paralleling the decrease of AOSD activity. Elevated CLEC5A levels and their positive association with activity parameters suggest that CLEC5A is involved in the pathogenesis and may serve as an activity indicator of AOSD.
Collapse
|
9
|
Pathak S, Vambutas A. Autoimmune inner ear disease patient-associated 28-kDa proinflammatory IL-1β fragment results from caspase-7-mediated cleavage in vitro. JCI Insight 2020; 5:130845. [PMID: 32051334 DOI: 10.1172/jci.insight.130845] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 12/26/2019] [Indexed: 12/14/2022] Open
Abstract
Interleukin-1β (IL-1β) is a key proinflammatory cytokine involved in the progression of many autoinflammatory and autoimmune diseases, including autoimmune inner ear disease (AIED). IL-1β inhibition has been shown to result in clinical hearing improvement in a small cohort of corticosteroid-resistant patients with AIED. Canonical processing of pro-IL-1β by caspase-1 generates an active 17-kDa fragment, capable of instigating a proinflammatory microenvironment. However, in response to LPS, PBMCs from patients with AIED uniquely express a 28-kDa IL-1β fragment, as compared with PBMCs from control subjects. We synthesized and compared the biologic activity of the 28-kDa fragment to the 17-kDa IL-1β product and the pro-IL-1 31-kDa protein. The 28-kDa IL-1β fragment induces IL-6, TNF-α, and CCL3 in PBMCs. Uniquely, only caspase-7 treatment showed a dose- and time-dependent increase in 28-kDa band generation. Mass spectrometry confirmed the putative caspase-7 cleavage site of pro-IL-1β, which was used to generate the 28-kDa fragment used for PBMC stimulation studies. Collectively, these results provide insight into the function of a poorly understood, processed 28-kDa form of IL-1β in patients with AIED that is uniquely generated by caspase-7 and is capable of activating further downstream proinflammatory cytokines. Further investigation may provide novel pharmacologic targets for the treatment of this rare disease.
Collapse
Affiliation(s)
- Shresh Pathak
- Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Department of Otolaryngology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA.,Head and Neck Surgery, Department of Otorhinolaryngology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Andrea Vambutas
- Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Department of Otolaryngology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA.,Head and Neck Surgery, Department of Otorhinolaryngology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| |
Collapse
|
10
|
Qin W, Wu D, Luo Y, Zhao M, Wang Y, Shi X, Zhou L, Yu W, Sun Y, Wang R, Zhang W, Liu M, Shen M. Neurological manifestations of autoinflammatory diseases in Chinese adult patients. Semin Arthritis Rheum 2020; 50:1500-1506. [PMID: 32115236 DOI: 10.1016/j.semarthrit.2019.12.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 11/15/2019] [Accepted: 12/30/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Systemic autoinflammatory diseases (SAIDs) are a group of disorders characterized by a dysregulation of innate immune system leading to multi-systemic inflammation. We aim to assess the neurological manifestations of Chinese adult patients with SAIDs. METHODS Eighty adult patients (≥16 years) were diagnosed as SAIDs from April 2015 to June 2019, at the center of adult autoinflammatory diseases, Department of Rheumatology, Peking Union Medical College Hospital. Clinical and genetic features of these patients were collected. All patients underwent neurologic, ophthalmologic and otolaryngologic evaluation. RESULTS Totally 31 out of 80 (38.8%) patients had neurological manifestations, including 14 familial Mediterranean fever (45.2%), 6 NLRP3-associated autoinflammatory disease (19.4%), 5 tumor necrosis factor receptor-associated periodic fever syndrome (16.1%), 5 NLRP12-associated autoinflammatory disease (16.1%), and 1 Yao syndrome (3.2%). Twenty patients (64.5%) were adult-onset. The median time of diagnosis delay was 11.7 years (0.5-50 years). The common neurological damage included headache (28 patients, 90.3%), sensorineural hearing loss (6, 19.4%), dizziness (4, 12.9%), cerebral infarction/hemorrhage (4, 12.9%), chronic aseptic meningitis (3, 9.7%), intracranial hypertension (3, 9.7%), papilledema (3, 9.7%), optic neuritis (2, 6.5%), and hydrocephalus (1, 3.2%). Severe neurological damage was observed in 8 patients (25.8%), including brain atrophy, hydrocephalus, complete hearing loss, chronic aseptic meningitis and optic neuritis. CONCLUSION Neurological damage was diverse in SAIDs patients. Neurological symptoms should be fully realized by physicians, in not only pediatric but also adult patients with SAIDs. CSF analysis and brain images should be performed promptly. Early diagnosis and appropriate treatment are essential to avoid irreversible neurological complications.
Collapse
Affiliation(s)
- Wenyi Qin
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China; The Integrated Traditional Chinese and Western Medicine Department, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Di Wu
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.
| | - Yi Luo
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.
| | - Mengzhu Zhao
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.
| | - Yi Wang
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100032, China.
| | - Xiaochun Shi
- Department of Infectious Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100032, China.
| | - Lixin Zhou
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100032, China.
| | - Weihong Yu
- Key Lab of Ocular Fundus Diseases, Chinese Academy of Medical Sciences, Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100032, China.
| | - Yang Sun
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100730, China.
| | - Rongrong Wang
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100730, China.
| | - Wen Zhang
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.
| | - Mengqi Liu
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Min Shen
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.
| |
Collapse
|
11
|
Chen PK, Lan JL, Li JP, Chang CK, Chang SH, Huang PH, Yeo KJ, Chen DY. Elevated plasma galectin-3 levels and their correlation with disease activity in adult-onset Still’s disease. Clin Rheumatol 2020; 39:1945-1952. [DOI: 10.1007/s10067-020-04946-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/23/2019] [Accepted: 01/13/2020] [Indexed: 01/13/2023]
|
12
|
Lee HA, Song YR, Park MH, Chung HY, Na HS, Chung J. Catechin ameliorates Porphyromonas gingivalis-induced inflammation via the regulation of TLR2/4 and inflammasome signaling. J Periodontol 2019; 91:661-670. [PMID: 31473995 DOI: 10.1002/jper.18-0004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Porphyromonas gingivalis is a major periodontopathogen found in patients with chronic periodontitis that can lead to alveolar bone or tooth loss. Interleukin-1β (IL-1β), a proinflammatory cytokine, is most relevant to the pathogenesis of periodontitis. Catechin is one of the main polyphenol compounds found in green tea and possesses a range of health benefits. This study examined the anti-inflammatory effects of catechin in THP-1-derived macrophages infected with P. gingivalis as well as its effects on P. gingivalis-induced periodontitis in a mouse model. METHODS The cytokine levels and relevant protein expression in THP-1 cells were measured using an enzyme-linked immunosorbent assay and Western blot analysis, respectively. An apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) pyroptosome formation was measured by confocal laser scanning microscopy. Micro-computed tomography was used to determine the level of bone loss induced by a P. gingivalis oral infection. RESULTS Catechin attenuated the production of IL-1β by inhibiting pro-IL-1β expression via the downregulation of nuclear factor-κB, p38 mitogen-activated protein kinase, and Toll-like receptor signaling. In addition, catechin inhibited the activation of inflammasomes induced by P. gingivalis, but did not affect the growth of P. gingivalis. Catechin reduced the level of alveolar bone loss in a P. gingivalis-induced periodontitis mouse model. CONCLUSION Catechin possesses anti-inflammatory properties by reducing the level of IL-1β production, suggesting that it can potentially be used for the prevention and treatment of periodontal inflammation caused by P. gingivalis.
Collapse
Affiliation(s)
- Hyun Ah Lee
- Department of Oral Microbiology, School of Dentistry, Pusan National University, Yangsan, Korea
| | - Yu Ri Song
- Department of Oral Microbiology, School of Dentistry, Pusan National University, Yangsan, Korea.,Oral Genomics Research Center, Pusan National University, Yangsan, Korea
| | - Mi Hee Park
- Department of Oral Microbiology, School of Dentistry, Pusan National University, Yangsan, Korea.,Oral Genomics Research Center, Pusan National University, Yangsan, Korea
| | - Hae-Young Chung
- Department of Biochemistry, School of Pharmacy, Pusan National University, Busan, Korea
| | - Hee Sam Na
- Department of Oral Microbiology, School of Dentistry, Pusan National University, Yangsan, Korea.,Oral Genomics Research Center, Pusan National University, Yangsan, Korea
| | - Jin Chung
- Department of Oral Microbiology, School of Dentistry, Pusan National University, Yangsan, Korea.,Oral Genomics Research Center, Pusan National University, Yangsan, Korea
| |
Collapse
|
13
|
Bertoni A, Carta S, Baldovini C, Penco F, Balza E, Borghini S, Di Duca M, Ognio E, Signori A, Nozza P, Schena F, Castellani P, Pastorino C, Perrone C, Obici L, Martini A, Ceccherini I, Gattorno M, Rubartelli A, Chiesa S. A novel knock-in mouse model of cryopyrin-associated periodic syndromes with development of amyloidosis: Therapeutic efficacy of proton pump inhibitors. J Allergy Clin Immunol 2019; 145:368-378.e13. [PMID: 31194989 DOI: 10.1016/j.jaci.2019.05.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 04/18/2019] [Accepted: 05/07/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Cryopyrin-associated periodic syndromes (CAPS) are a group of autoinflammatory diseases linked to gain-of-function mutations in the NOD-like receptor family, pyrin domain containing 3 (NLRP3) gene, which cause uncontrolled IL-1β secretion. Proton pump inhibitors (PPIs), which are commonly used as inhibitors of gastric acid production, also have anti-inflammatory properties, protect mice from sepsis, and prevent IL-1β secretion by monocytes from patients with CAPS. OBJECTIVE We sought to develop a novel Nlrp3 knock-in (KI) mouse model of CAPS to study amyloidosis, a severe CAPS complication, and test novel therapeutic approaches. METHODS We generated KI mice by engineering the N475K mutation, which is associated with the CAPS phenotype, into the mouse Nlrp3 gene. KI and wild-type mice received PPIs or PBS intraperitoneally and were analyzed for survival, inflammation, cytokine secretion, and amyloidosis development. RESULTS Mutant Nlrp3 KI mice displayed features that recapitulate the immunologic and clinical phenotype of CAPS. They showed systemic inflammation with high levels of serum proinflammatory cytokines, inflammatory infiltrates in various organs, and amyloid deposits in the spleen, liver, and kidneys. Toll-like receptor stimulated macrophages from KI mice secreted high levels of IL-1β, IL-18, and IL-1α but low amounts of IL-1 receptor antagonist. Treatment of KI mice with PPIs had a clear clinical effect, showing a reduction in inflammatory manifestations, regression of amyloid deposits, and normalization of proinflammatory and anti-inflammatory cytokine production by macrophages. CONCLUSION Nlrp3 KI mice displayed a CAPS phenotype with many characteristics of autoinflammation, including amyloidosis. The therapeutic effectiveness of PPIs associated with a lack of toxicity indicates that these drugs could represent relevant adjuvants to the anti-IL-1 drugs in patients with CAPS and other IL-1-driven diseases.
Collapse
Affiliation(s)
- Arinna Bertoni
- UOSD Centro Malattie Autoinfiammatorie ed Immunodeficienze, IRCCS Istituto G. Gaslini, Genova, Italy
| | - Sonia Carta
- Unità di Biologia Cellulare, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | - Federica Penco
- UOSD Centro Malattie Autoinfiammatorie ed Immunodeficienze, IRCCS Istituto G. Gaslini, Genova, Italy
| | - Enrica Balza
- Unità di Biologia Cellulare, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | - Marco Di Duca
- Laboratorio di Fisiopatologia dell' Uremia, IRCCS Istituto G. Gaslini, Genova, Italy
| | - Emanuela Ognio
- S.S Animal Facility, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Alessio Signori
- Dipartimento di Scienze della Salute, Sezione di Biostatistica, DISSAL, Università degli studi di Genova, Genova, Italy
| | - Paolo Nozza
- Anatomia Patologica, IRCCS Istituto G. Gaslini, Genova, Italy
| | - Francesca Schena
- UOSD Centro Malattie Autoinfiammatorie ed Immunodeficienze, IRCCS Istituto G. Gaslini, Genova, Italy
| | - Patrizia Castellani
- Unità di Biologia Cellulare, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Claudia Pastorino
- UOSD Centro Malattie Autoinfiammatorie ed Immunodeficienze, IRCCS Istituto G. Gaslini, Genova, Italy
| | - Carola Perrone
- UOSD Centro Malattie Autoinfiammatorie ed Immunodeficienze, IRCCS Istituto G. Gaslini, Genova, Italy
| | - Laura Obici
- Centro per lo Studio e la Cura delle Amiloidosi Sistemiche, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Alberto Martini
- Direzione Scientifica, IRCCS Istituto G. Gaslini, Genova, Italy
| | | | - Marco Gattorno
- UOSD Centro Malattie Autoinfiammatorie ed Immunodeficienze, IRCCS Istituto G. Gaslini, Genova, Italy; UOC Clinica Pediatrica e Reumatologica, IRCCS Istituto G. Gaslini, Genova, Italy.
| | - Anna Rubartelli
- Unità di Biologia Cellulare, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Sabrina Chiesa
- UOSD Centro Malattie Autoinfiammatorie ed Immunodeficienze, IRCCS Istituto G. Gaslini, Genova, Italy.
| |
Collapse
|
14
|
Davis MDP, van der Hilst JCH. Mimickers of Urticaria: Urticarial Vasculitis and Autoinflammatory Diseases. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2018; 6:1162-1170. [PMID: 29871797 DOI: 10.1016/j.jaip.2018.05.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 05/14/2018] [Indexed: 12/17/2022]
Abstract
A wide differential diagnosis must be considered in a patient presenting with urticarial plaques. Although acute and chronic urticaria are the commonest diagnoses, other differential diagnoses include polymorphous eruption of pregnancy, mast cell disorders, hypereosinophilic syndrome, urticarial vasculitis, pemphigoid, systemic lupus erythematosus, and autoinflammatory disease. This review will specifically address urticarial vasculitis and autoinflammatory syndromes. These entities represent contrasting examples of urticarial-like lesions resulting from either an adaptive immune complex-mediated mechanism (urticarial vasculitis) or an innate immune-mediated mechanism (autoinflammatory disorders), with differing therapeutic implications. In patients presenting with painful, persistent plaques that last more than 24 hours and resolve with bruising of the skin, consideration should be given to a diagnosis of urticarial vasculitis. A biopsy should be obtained to ascertain this diagnosis. In patients presenting with a persistent history of recurrent urticarial plaques associated with signs of systemic inflammation including fevers and elevated inflammatory markers (C-reactive protein [CRP]/serum amyloid A, leukocytosis, and negative connective tissue serologies), consideration should be given to autoinflammatory disorders: the 3 cryopyrin-associated periodic syndromes, Schnitzler syndrome, and familial cold autoinflammatory syndrome 2. Serum protein electrophoresis should be checked to rule out an underlying monoclonal gammopathy.
Collapse
Affiliation(s)
- Mark D P Davis
- Department of Dermatology, Mayo Clinic, Rochester, Minn.
| | - Jeroen C H van der Hilst
- Department of Infectious Diseases and Immunity, Jessa Hospital. BIOMED Research Institute, University of Hasselt, Hasselt, Belgium
| |
Collapse
|
15
|
Carta S, Gattorno M, Rubartelli A. NLR in Human Diseases: Role and Laboratory Findings. Methods Mol Biol 2017; 1417:247-54. [PMID: 27221496 DOI: 10.1007/978-1-4939-3566-6_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Autoinflammatory diseases are a group of inherited and multifactorial disorders characterized by an overactivation of innate immune response. In most cases, the clinical manifestations are due to increased activity of the NLRP3 inflammasome resulting in increased IL-1β secretion. Investigating inflammatory cells from subjects affected by autoinflammatory diseases presents a number of technical difficulties related to the rarity of the diseases, to the young age of most patients, and to the difficult modulation of gene expression in primary cells. However, since cell stress is involved in the pathophysiology of these diseases, the study of freshly drawn blood monocytes from patients affected by IL-1-mediated diseases strongly increases the chances that the observed phenomena is indeed pertinent to the pathogenesis of the disease and not influenced by the long-term cell culture conditions (e.g., the high O2 tension) or gene transfection in continuous cell lines that may lead to artifacts.
Collapse
Affiliation(s)
- Sonia Carta
- Cell Biology Unit, IRCCS Azienda Ospedaliera Universitaria San Martino-IST, Genoa, Italy
| | | | - Anna Rubartelli
- Cell Biology Unit, IRCCS Azienda Ospedaliera Universitaria San Martino-IST, Genoa, Italy.
| |
Collapse
|
16
|
Gulumian M, Andraos C. In Search of a Converging Cellular Mechanism in Nanotoxicology and Nanomedicine in the Treatment of Cancer. Toxicol Pathol 2017; 46:4-13. [PMID: 29034767 DOI: 10.1177/0192623317735776] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Multiple applications of nanomaterials have raised concern with regard to their toxicity. With increasing research into nanomaterial safety, mechanisms involved in the toxic effects of nanomaterials have begun to emerge. The importance of nanomaterial-induced lysosomal membrane permeabilization through overloading or direct damage of the lysosomal compartment, resulting in the blockade of autophagosome-lysosome fusion and autophagy dysfunction, as well as inflammasome activation were cited as emerging mechanisms of nanomaterial toxicity. It has recently been proposed that these very mechanisms leading to nanomaterial toxicity may be utilized in nanotherapeutics. This review discusses these nanomaterial-induced mechanisms in detail and how it has been exploited in cancer research. This review also addresses certain considerations that need to be kept in mind when using nanomaterials in therapeutics.
Collapse
Affiliation(s)
- Mary Gulumian
- 1 National Institute for Occupational Health (NIOH), Johannesburg, South Africa.,2 Haematology and Molecular Medicine Department, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Charlene Andraos
- 1 National Institute for Occupational Health (NIOH), Johannesburg, South Africa
| |
Collapse
|
17
|
Hsieh CW, Chen YM, Lin CC, Tang KT, Chen HH, Hung WT, Lai KL, Chen DY. Elevated Expression of the NLRP3 Inflammasome and Its Correlation with Disease Activity in Adult-onset Still Disease. J Rheumatol 2017; 44:1142-1150. [PMID: 28507179 DOI: 10.3899/jrheum.161354] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2017] [Indexed: 01/02/2023]
Abstract
OBJECTIVE The dysregulation of the NLRP3 (NLR containing a pyrin domain) inflammasome is involved in autoinflammatory diseases. Adult-onset Still disease (AOSD) is regarded as an autoinflammatory disease. However, the pathogenic involvement of NLRP3 inflammasome in AOSD remains unclear and NLRP3 activators in AOSD are currently unknown. METHODS The mRNA expression of NLRP3 inflammasome signaling in peripheral blood mononuclear cells (PBMC) from 34 patients with AOSD and 14 healthy subjects was determined using quantitative-PCR (qPCR). The changes in mRNA and protein levels of NLRP3 inflammasome signaling in PBMC treated with the potential activator [imiquimod (IMQ)] or inhibitor of NLRP3 were evaluated using qPCR and immunoblotting, respectively. The supernatant levels of interleukin (IL)-1β and IL-18 were determined by ELISA. RESULTS Significantly higher mRNA levels of NLRP3 inflammasome signaling were observed in patients with AOSD compared with healthy controls. NLRP3 expressions were positively correlated with disease activity in patients with AOSD. IMQ (an effective Toll-like receptor 7 ligand; 10 µg/ml and 25 µg/ml) stimulation of PBMC from patients with AOSD induced dose-dependent increases of mRNA expression of NLRP3 (mean ± standard error of the mean, 2.06 ± 0.46 and 6.05 ± 1.84, respectively), caspase-1 (1.81 ± 0.23 and 4.25 ± 0.48), IL-1β (5.68 ± 1.51 and 12.13 ± 3.71), and IL-18 (2.32 ± 0.37 and 4.81 ± 0.51) compared with controls (all p < 0.005). IMQ stimulation of PBMC from patients similarly induced greater increases in protein expressions of NLRP3 inflammasome compared with controls. The protein expressions of NLRP3, IL-1β, and IL-18 on PBMC significantly decreased after treatment with NLRP3 inhibitor in patients with AOSD. CONCLUSION Increased expression of NLRP3 inflammasome and its positive correlation with disease activity in AOSD suggest its involvement in disease pathogenesis. IMQ upregulated expressions of NLRP3 inflammasome signaling, and IMQ might be an activator of NLRP3 inflammasome in AOSD.
Collapse
Affiliation(s)
- Chia-Wei Hsieh
- From the Division of Allergy, Immunology and Rheumatology, and the Department of Medical Education and Research, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung; Faculty of Medicine, National Yang Ming University, Taipei, Taiwan.,C.W. Hsieh, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, National Chung Hsing University; Y.M. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University, and PhD Program in Translational Medicine, National Chung Hsing University; C.C. Lin, PhD, PhD Program in Translational Medicine, National Chung Hsing University, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University; K.T. Tang, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, National Chung Hsing University; H.H. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University; W.T. Hung, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital; K.L. Lai, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University; D.Y. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University, and PhD Program in Translational Medicine, National Chung Hsing University, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University. Dr. Y.M. Chen and C.C. Lin contributed equally to this work
| | - Yi-Ming Chen
- From the Division of Allergy, Immunology and Rheumatology, and the Department of Medical Education and Research, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung; Faculty of Medicine, National Yang Ming University, Taipei, Taiwan.,C.W. Hsieh, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, National Chung Hsing University; Y.M. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University, and PhD Program in Translational Medicine, National Chung Hsing University; C.C. Lin, PhD, PhD Program in Translational Medicine, National Chung Hsing University, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University; K.T. Tang, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, National Chung Hsing University; H.H. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University; W.T. Hung, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital; K.L. Lai, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University; D.Y. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University, and PhD Program in Translational Medicine, National Chung Hsing University, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University. Dr. Y.M. Chen and C.C. Lin contributed equally to this work
| | - Chi-Chen Lin
- From the Division of Allergy, Immunology and Rheumatology, and the Department of Medical Education and Research, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung; Faculty of Medicine, National Yang Ming University, Taipei, Taiwan.,C.W. Hsieh, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, National Chung Hsing University; Y.M. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University, and PhD Program in Translational Medicine, National Chung Hsing University; C.C. Lin, PhD, PhD Program in Translational Medicine, National Chung Hsing University, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University; K.T. Tang, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, National Chung Hsing University; H.H. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University; W.T. Hung, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital; K.L. Lai, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University; D.Y. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University, and PhD Program in Translational Medicine, National Chung Hsing University, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University. Dr. Y.M. Chen and C.C. Lin contributed equally to this work
| | - Kuo-Tung Tang
- From the Division of Allergy, Immunology and Rheumatology, and the Department of Medical Education and Research, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung; Faculty of Medicine, National Yang Ming University, Taipei, Taiwan.,C.W. Hsieh, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, National Chung Hsing University; Y.M. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University, and PhD Program in Translational Medicine, National Chung Hsing University; C.C. Lin, PhD, PhD Program in Translational Medicine, National Chung Hsing University, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University; K.T. Tang, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, National Chung Hsing University; H.H. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University; W.T. Hung, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital; K.L. Lai, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University; D.Y. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University, and PhD Program in Translational Medicine, National Chung Hsing University, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University. Dr. Y.M. Chen and C.C. Lin contributed equally to this work
| | - Hsin-Hua Chen
- From the Division of Allergy, Immunology and Rheumatology, and the Department of Medical Education and Research, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung; Faculty of Medicine, National Yang Ming University, Taipei, Taiwan.,C.W. Hsieh, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, National Chung Hsing University; Y.M. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University, and PhD Program in Translational Medicine, National Chung Hsing University; C.C. Lin, PhD, PhD Program in Translational Medicine, National Chung Hsing University, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University; K.T. Tang, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, National Chung Hsing University; H.H. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University; W.T. Hung, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital; K.L. Lai, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University; D.Y. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University, and PhD Program in Translational Medicine, National Chung Hsing University, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University. Dr. Y.M. Chen and C.C. Lin contributed equally to this work
| | - Wei-Ting Hung
- From the Division of Allergy, Immunology and Rheumatology, and the Department of Medical Education and Research, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung; Faculty of Medicine, National Yang Ming University, Taipei, Taiwan.,C.W. Hsieh, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, National Chung Hsing University; Y.M. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University, and PhD Program in Translational Medicine, National Chung Hsing University; C.C. Lin, PhD, PhD Program in Translational Medicine, National Chung Hsing University, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University; K.T. Tang, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, National Chung Hsing University; H.H. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University; W.T. Hung, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital; K.L. Lai, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University; D.Y. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University, and PhD Program in Translational Medicine, National Chung Hsing University, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University. Dr. Y.M. Chen and C.C. Lin contributed equally to this work
| | - Kuo-Lung Lai
- From the Division of Allergy, Immunology and Rheumatology, and the Department of Medical Education and Research, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung; Faculty of Medicine, National Yang Ming University, Taipei, Taiwan.,C.W. Hsieh, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, National Chung Hsing University; Y.M. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University, and PhD Program in Translational Medicine, National Chung Hsing University; C.C. Lin, PhD, PhD Program in Translational Medicine, National Chung Hsing University, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University; K.T. Tang, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, National Chung Hsing University; H.H. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University; W.T. Hung, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital; K.L. Lai, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University; D.Y. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University, and PhD Program in Translational Medicine, National Chung Hsing University, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University. Dr. Y.M. Chen and C.C. Lin contributed equally to this work
| | - Der-Yuan Chen
- From the Division of Allergy, Immunology and Rheumatology, and the Department of Medical Education and Research, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung; Faculty of Medicine, National Yang Ming University, Taipei, Taiwan. .,C.W. Hsieh, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, National Chung Hsing University; Y.M. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University, and PhD Program in Translational Medicine, National Chung Hsing University; C.C. Lin, PhD, PhD Program in Translational Medicine, National Chung Hsing University, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University; K.T. Tang, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and PhD Program in Translational Medicine, National Chung Hsing University; H.H. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University; W.T. Hung, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital; K.L. Lai, MD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University; D.Y. Chen, MD, PhD, Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, and Department of Medical Education and Research, Taichung Veterans General Hospital, and Faculty of Medicine, National Yang Ming University, and PhD Program in Translational Medicine, National Chung Hsing University, and Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University. Dr. Y.M. Chen and C.C. Lin contributed equally to this work.
| |
Collapse
|
18
|
La Torre F, Caparello MC, Cimaz R. Canakinumab for the treatment of TNF-receptor associated periodic syndrome. Expert Rev Clin Immunol 2017; 13:513-523. [DOI: 10.1080/1744666x.2017.1324783] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- F La Torre
- Pediatric Rheumatology Regional Center, Department of Pediatrics, Antonio Perrino Hospital, Brindisi, Puglia, Italy
| | - MC Caparello
- Pediatric Rheumatology Unit, Neurofarba Department, AOU Meyer, University of Florence, Florence, Italy
| | - R Cimaz
- Pediatric Rheumatology Unit, Neurofarba Department, AOU Meyer, University of Florence, Florence, Italy
| |
Collapse
|
19
|
Bording-Jorgensen M, Alipour M, Danesh G, Wine E. Inflammasome Activation by ATP Enhances Citrobacter rodentium Clearance through ROS Generation. Cell Physiol Biochem 2017; 41:193-204. [PMID: 28132060 DOI: 10.1159/000455988] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/22/2016] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Nod-like receptor family, pyrin domain containing 3 (NLRP3) is an important cytosolic sensor of cellular stress and infection. Once activated, NLRP3 forms a multiprotein complex (inflammasome) that triggers the maturation and secretion of interleukin (IL)-1β and IL-18. We aimed to define the consequences of NLRP3 induction, utilizing exogenous adenosine triphosphate (ATP) as an inflammasome activator, to determine if inflammasome activation increases macrophage killing of Citrobacter rodentium and define mechanisms. METHODS Bacterial survival was measured using a gentamicin protection assay. Inflammasome activation or inhibition in mouse J774A.1 macrophages were assessed by measuring IL-1β; cytokines and reactive oxygen species (ROS) were measured by ELISA and DCFDA, respectively. RESULTS Activation of the inflammasome increased bacterial killing by macrophages and its inhibition attenuated this effect with no impact on phagocytosis or cell death. Furthermore, inflammasome activation suppressed pro-inflammatory cytokines during infection, possibly due to more effective bacterial killing. While the infection increased ROS production, this effect was reduced by inflammasome inhibitors, indicating that ROS is inflammasome-dependent. ROS inhibitors increased bacterial survival in the presence of ATP, suggesting that inflammasome-induced bacterial killing is mediated, at least in part, by ROS activity. CONCLUSION Improving inflammasome activity during infection may increase bacterial clearance by macrophages and reduce subsequent microbe-induced inflammation.
Collapse
|
20
|
Torene R, Nirmala N, Obici L, Cattalini M, Tormey V, Caorsi R, Starck-Schwertz S, Letzkus M, Hartmann N, Abrams K, Lachmann H, Gattorno M. Canakinumab reverses overexpression of inflammatory response genes in tumour necrosis factor receptor-associated periodic syndrome. Ann Rheum Dis 2016; 76:303-309. [PMID: 27474763 PMCID: PMC5264306 DOI: 10.1136/annrheumdis-2016-209335] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 06/14/2016] [Accepted: 06/20/2016] [Indexed: 11/28/2022]
Abstract
Objective To explore whether gene expression profiling can identify a molecular mechanism for the clinical benefit of canakinumab treatment in patents with tumour necrosis factor receptor-associated periodic syndrome (TRAPS). Methods Blood samples were collected from 20 patients with active TRAPS who received canakinumab 150 mg every 4 weeks for 4 months in an open-label proof-of-concept phase II study, and from 20 aged-matched healthy volunteers. Gene expression levels were evaluated in whole blood samples by microarray analysis for arrays passing quality control checks. Results Patients with TRAPS exhibited a gene expression signature in blood that differed from that in healthy volunteers. Upon treatment with canakinumab, many genes relevant to disease pathogenesis moved towards levels seen in the healthy volunteers. Canakinumab downregulated the TRAPS-causing gene (TNF super family receptor 1A (TNFRSF1A)), the drug-target gene (interleukin (IL)-1B) and other inflammation-related genes (eg, MAPK14). In addition, several inflammation-related pathways were evident among the differentially expressed genes. Canakinumab treatment reduced neutrophil counts, but the observed expression differences remained after correction for this. Conclusions These gene expression data support a model in which canakinumab produces clinical benefit in TRAPS by increasing neutrophil apoptosis and reducing pro-inflammatory signals resulting from the inhibition of IL-1β. Notably, treatment normalised the overexpression of TNFRSF1A, suggesting that canakinumab has a direct impact on the main pathogenic mechanism in TRAPS. Trial registration number NCT01242813.
Collapse
Affiliation(s)
- Rebecca Torene
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA
| | - Nanguneri Nirmala
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA
| | - Laura Obici
- Amyloid Centre, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marco Cattalini
- Pediatric Clinic, University of Brescia and Spedali Civili, Brescia, Italy
| | | | | | | | - Martin Letzkus
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Nicole Hartmann
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Ken Abrams
- Novartis Pharmaceuticals, East Hanover, New Jersey, USA
| | | | | |
Collapse
|
21
|
Gattorno M, Obici L, Cattalini M, Tormey V, Abrams K, Davis N, Speziale A, Bhansali SG, Martini A, Lachmann HJ. Canakinumab treatment for patients with active recurrent or chronic TNF receptor-associated periodic syndrome (TRAPS): an open-label, phase II study. Ann Rheum Dis 2016; 76:173-178. [PMID: 27269295 PMCID: PMC5264215 DOI: 10.1136/annrheumdis-2015-209031] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 04/27/2016] [Accepted: 05/17/2016] [Indexed: 12/23/2022]
Abstract
Objective To evaluate the efficacy of canakinumab, a high-affinity human monoclonal anti-interleukin-1β antibody, in inducing complete or almost complete responses in patients with active tumour necrosis factor receptor-associated periodic syndrome (TRAPS). Methods Twenty patients (aged 7–78 years) with active recurrent or chronic TRAPS were treated with canakinumab 150 mg every 4 weeks for 4 months (2 mg/kg for those ≤40 kg) in this open-label, proof-of-concept, phase II study. Canakinumab was then withdrawn for up to 5 months, with reintroduction on relapse, and 4 weekly administration (subsequently increased to every 8 weeks) for 24 months. The primary efficacy variable was the proportion of patients achieving complete or almost complete response at day 15, defined as clinical remission (Physician's Global Assessment score ≤1) and full or partial serological remission. Results Nineteen patients (19/20, 95%; 95% CI 75.1% to 99.9%) achieved the primary efficacy variable. Responses to canakinumab occurred rapidly; median time to clinical remission 4 days (95% CI 3 to 8 days). All patients relapsed after canakinumab was withdrawn; median time to relapse 91.5 days (95% CI 65 to 117 days). On reintroduction of canakinumab, clinical and serological responses were similar to those seen during the first phase, and were sustained throughout treatment. Canakinumab was well tolerated and clinical responses were accompanied by rapid and sustained improvement in health-related quality of life. Weight normalised pharmacokinetics of canakinumab, although limited, appeared to be consistent with historical canakinumab data. Conclusions Canakinumab induces rapid disease control in patients with active TRAPS, and clinical benefits are sustained during long-term treatment. Trial registration number NCT01242813; Results.
Collapse
Affiliation(s)
- Marco Gattorno
- UO Pediatria 2, Reumatologia, Eurofever project, G Gaslini Institute, Genoa, Italy
| | - Laura Obici
- Amyloidosis Research and Treatment Centre, Biotechnology Research Laboratories, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marco Cattalini
- Pediatric Clinic, University of Brescia and Spedali Civili, Brescia, Italy
| | | | - Ken Abrams
- Novartis Pharmaceutical Corporation, East Hanover, New Jersey, USA
| | - Nicole Davis
- Novartis Pharmaceutical Corporation, East Hanover, New Jersey, USA
| | | | - Suraj G Bhansali
- Novartis Pharmaceutical Corporation, East Hanover, New Jersey, USA
| | - Alberto Martini
- UO Pediatria 2, Reumatologia, Eurofever project, G Gaslini Institute, Genoa, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, and Maternal-Fetal Medicine (DINOGMI), University of Genoa, Genoa, Italy
| | - Helen J Lachmann
- Division of Medicine, National Amyloidosis Center, University College London Medical School, London, UK
| |
Collapse
|
22
|
Naselli A, Penco F, Cantarini L, Insalaco A, Alessio M, Tommasini A, Maggio C, Obici L, Gallizi R, Cimmino M, Signa S, Lucherini OM, Carta S, Caroli F, Martini A, Rubartelli A, Ceccherini I, Gattorno M. Clinical Characteristics of Patients Carrying the Q703K Variant of the NLRP3 Gene: A 10-year Multicentric National Study. J Rheumatol 2016; 43:1093-100. [DOI: 10.3899/jrheum.150962] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2016] [Indexed: 01/05/2023]
Abstract
Objective.The aim of our study was to analyze the clinical and functional effect of the p.Q703K (p. Q705K, c. 2107C>A) variant of the NLRP3 gene in a population of patients screened for suspected cryopyrin-associated periodic syndrome (CAPS).Methods.Since 2002, 580 patients underwent molecular analysis for NLRP3. Data on clinical presentation, response to treatment, and longterm followup were collected using a uniform questionnaire. The pattern of cytokine secretion after lipopolysaccharide stimulation from isolated monocytes was analyzed in 3 patients carrying the p.Q703K variant and 1 patient with a chronic infantile neurologic, cutaneous, articular syndrome phenotype carrying both the p.M406I and p.Q703K, and compared with 7 patients with CAPS with sure pathogenic variants and 6 healthy controls.Results.The p.Q703K variant was found in 57 screened patients with an overall allelic frequency of 5%. The frequency in normal controls was 5.5%. Clinical data at the moment of molecular analysis and at followup were available in 36 patients. Two patients displayed additional mutations of NLRP3. The mean followup was 2.5 years. Thirteen patients (39%) had a final diagnosis different from the original suspicion of CAPS. The remaining 21 patients displayed a mild phenotype mainly characterized by recurrent episodes of urticarial rash and arthralgia. Only 8 patients were treated with anti-interleukin (IL)-1 treatment, with a complete response in 5 patients. The pattern of secretion of IL-1β and other cytokines (IL-6 and IL-1 receptor antagonist) in patients did not display the aberrancies observed in patients with CAPS and was similar to that observed in healthy controls.Conclusion.The present study confirms the weak clinical and functional effect of the p.Q703K variant.
Collapse
|
23
|
Kang I. Editorial: Is the NLPR3 Inflammasome “Overheated” by Pneumococcal Vaccination in Cryopyrin-Associated Periodic Syndromes? Arthritis Rheumatol 2016; 68:274-6. [DOI: 10.1002/art.39483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 10/15/2015] [Indexed: 01/02/2023]
Affiliation(s)
- Insoo Kang
- Yale University School of Medicine; New Haven Connecticut
| |
Collapse
|
24
|
Extracellular ATP mediates inflammatory responses in colitis via P2 × 7 receptor signaling. Sci Rep 2016; 6:19108. [PMID: 26739809 PMCID: PMC4703960 DOI: 10.1038/srep19108] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 12/02/2015] [Indexed: 12/19/2022] Open
Abstract
Extracellular purinergic products, particularly ATP, have recently been implicated to regulate immune cell functions and contribute to aberrant inflammatory responses of immune diseases. However, regulation of immune responses of colitis by extracellular ATP and its main receptor, P2 × 7, remains to be elucidated. In the study, we induced murine colitis by feeding mice with 4% dextran sulfate sodium (DSS), and noted dramatically heightened extracellular ATP levels in colon tissues during the progression of experimental colitis. Blockade of ATP release by carbenoxolone (CBX) treatment, or promoting ATP degradation by ATP diphosphohydrolase (apyrase), decreased extracellular ATP levels in colon tissues, attenuated DSS-induced colitis, whereas inhibition of extracellular ATP degradation by sodium metatungstate (POM-1) exacerbated tissue damage in the mice with colitis. Moreover, treatment with inhibitor of P2 × 7 receptor, A438079, decreased NFκB activation and active caspase-1 expression in lamina propria immune cells, downregulated proinflammatory cytokine production in colon tissues, and attenuated murine colitis. Collectively, these data suggest extracellular ATP participates in regulation of inflammatory responses of experimental colitis, through P2 × 7 receptor and inflammasome and NFκB signaling, which provides potential alternatives to the current clinical approaches to suppress extracellular ATP-mediated immune responsiveness.
Collapse
|
25
|
Cochlear functions in children with familial Mediterranean fever: any role of the severity of the disease? Int J Pediatr Otorhinolaryngol 2015; 79:1566-70. [PMID: 26231741 DOI: 10.1016/j.ijporl.2015.07.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 07/14/2015] [Accepted: 07/15/2015] [Indexed: 01/29/2023]
Abstract
OBJECTIVES The aim of the study was to compare the cochlear functions of children diagnosed with familial Mediterranean fever (FMF) with healthy controls and to determine their cochlear functions according to their disease severity. METHODS Seventy-three children with FMF and 30 healthy controls were included in the study. All the patients and controls were evaluated by audiologic evaluation, including high-frequency pure-tone audiometry and distortion product otoacoustic emission tests (DPOAE). The disease severity was evaluated by scoring systems adapted from those used by Pras et al. and with severity scoring systems from the Sheba Medical Center. RESULTS High-frequency pure-tone audiometry and DPOAE levels were normal in both patients and controls. Significant differences in the hearing levels of FMF patients were not found, according to both adapted severity scoring systems. CONCLUSIONS Cochlear functions in children with FMF had been evaluated by previous studies, but in our study we evaluated hearing functions according to both controls and disease severity. As a unique study comparing cochlear functions according to severity scores, no significant differences were shown between the groups and controls.
Collapse
|
26
|
Chen Y, Pitzer AL, Li X, Li PL, Wang L, Zhang Y. Instigation of endothelial Nlrp3 inflammasome by adipokine visfatin promotes inter-endothelial junction disruption: role of HMGB1. J Cell Mol Med 2015; 19:2715-27. [PMID: 26293846 PMCID: PMC4687695 DOI: 10.1111/jcmm.12657] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 06/23/2015] [Indexed: 01/24/2023] Open
Abstract
Recent studies have indicated that the inflammasome plays a critical role in the pathogenesis of vascular diseases. However, the pathological relevance of this inflammasome activation, particularly in vascular cells, remains largely unknown. Here, we investigated the role of endothelial (Nucleotide‐binding Oligomerization Domain) NOD‐like receptor family pyrin domain containing three (Nlrp3) inflammasomes in modulating inter‐endothelial junction proteins, which are associated with endothelial barrier dysfunction, an early onset of obesity‐associated endothelial injury. Our findings demonstrate that the activation of Nlrp3 inflammasome by visfatin markedly decreased the expression of inter‐endothelial junction proteins including tight junction proteins ZO‐1, ZO‐2 and occludin, and adherens junction protein VE‐cadherin in cultured mouse vascular endothelial (VE) cell monolayers. Such visfatin‐induced down‐regulation of junction proteins in endothelial cells was attributed to high mobility group box protein 1 (HMGB1) release derived from endothelial inflammasome‐dependent caspase‐1 activity. Similarly, in the coronary arteries of wild‐type mice, high‐fat diet (HFD) treatment caused a down‐regulation of inter‐endothelial junction proteins ZO‐1, ZO‐2, occludin and VE‐cadherin, which was accompanied with enhanced inflammasome activation and HMGB1 expression in the endothelium as well as transmigration of CD43+ T cells into the coronary arterial wall. In contrast, all these HFD‐induced alterations in coronary arteries were prevented in mice with Nlrp3 gene deletion. Taken together, these data strongly suggest that the activation of endothelial Nlrp3 inflammasomes as a result of the increased actions of injurious adipokines such as visfatin produces HMGB1, which act in paracrine or autocrine fashion to disrupt inter‐endothelial junctions and increase paracellular permeability of the endothelium contributing to the early onset of endothelial injury during metabolic disorders such as obesity or high‐fat/cholesterol diet.
Collapse
Affiliation(s)
- Yang Chen
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Ashley L Pitzer
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Xiang Li
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.,Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Pin-Lan Li
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Lei Wang
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Yang Zhang
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.,Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| |
Collapse
|
27
|
Varga G, Gattorno M, Foell D, Rubartelli A. Redox distress and genetic defects conspire in systemic autoinflammatory diseases. Nat Rev Rheumatol 2015; 11:670-80. [PMID: 26241183 DOI: 10.1038/nrrheum.2015.105] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inflammation is initiated by innate immune cell activation after contact with pathogens or tissue injury. An increasing number of observations have suggested that cellular stress, in the absence of infection or evident damage, can also induce inflammation. Thus, inflammation can be triggered by exogenous pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs)-so-called classic inflammation-or by endogenous stress resulting from tissue or cellular dysfunction. External triggers and cellular stress activate the same molecular pathways, possibly explaining why classic and stress-induced inflammation have similar clinical manifestations. In some systemic autoinflammatory diseases (SAIDs), inflammatory cells exhibit reduction-oxidation (redox) distress, having high levels of reactive oxygen species (ROS), which promote proinflammatory cytokine production and contribute to the subversion of mechanisms that self-limit inflammation. Thus, SAIDs can be viewed as a paradigm of stress-related inflammation, being characterized by recurrent flares or chronic inflammation (with no recognizable external triggers) and by a failure to downmodulate this inflammation. Here, we review SAID pathophysiology, focusing on the major cytokines and DAMPs, and on the key roles of redox distress. New therapeutic opportunities to tackle SAIDs by blocking stress-induced pathways and control the response to stress in patients are also discussed.
Collapse
Affiliation(s)
- Georg Varga
- Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster, Domagkstrasse 3, 48149 Münster, Germany
| | - Marco Gattorno
- Second Division of Paediatrics, G. Gaslini Institute, 16145 Genova, Italy
| | - Dirk Foell
- Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster, Domagkstrasse 3, 48149 Münster, Germany
| | - Anna Rubartelli
- Cell Biology Unit, IRCCS Azienda Ospedaliera Universitaria San Martino-IST, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| |
Collapse
|
28
|
Sarrabay G, Grandemange S, Touitou I. Diagnosis of cryopyrin-associated periodic syndrome: challenges, recommendations and emerging concepts. Expert Rev Clin Immunol 2015; 11:827-35. [DOI: 10.1586/1744666x.2015.1047765] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
29
|
Chae JJ, Park YH, Park C, Hwang IY, Hoffmann P, Kehrl JH, Aksentijevich I, Kastner DL. Connecting two pathways through Ca 2+ signaling: NLRP3 inflammasome activation induced by a hypermorphic PLCG2 mutation. Arthritis Rheumatol 2015; 67:563-7. [PMID: 25418813 DOI: 10.1002/art.38961] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 10/09/2014] [Accepted: 11/11/2014] [Indexed: 01/06/2023]
Abstract
OBJECTIVE We previously reported that p.Ser707Tyr, a novel variant in phospholipase Cγ2 (PLCγ2), is the cause of a dominantly inherited autoinflammatory disease, autoinflammation and PLCγ2-associated antibody deficiency and immune dysregulation (APLAID). The hypermorphic mutation enhances PLCγ2 activity and causes an increase in intracellular Ca2+ release from endoplasmic reticulum stores. Because increased intracellular Ca2+ signaling has been associated with NLRP3 inflammasome activation, we studied the role of the NLRP3 inflammasome in the pathogenesis of APLAID. METHODS Human peripheral blood mononuclear cells (PBMCs) were isolated from healthy control subjects and 2 patients with APLAID. Inflammasome activation was analyzed by Western blotting. Intracellular Ca2+ levels were measured with a FLIPR Calcium 4 assay kit. RESULTS Cells from the patients had elevated basal levels of intracellular Ca2+, and the intracellular Ca2+ flux triggered by extracellular CaCl2 was substantially enhanced. Patient PBMCs secreted interleukin-1β in response to lipopolysaccharide priming alone, and this effect was attenuated by treatment with a PLC inhibitor, intracellular Ca2+ blockers, or an adenylate cyclase activator. CONCLUSION Our findings suggest that the inflammation in patients with APLAID is partially driven by activation of the NLRP3 inflammasome. These data link 2 seemingly distinct molecular pathways and provide new insights into the pathogenesis of APLAID and autoinflammation.
Collapse
|
30
|
Yokoyama K, Ikeya M, Umeda K, Oda H, Nodomi S, Nasu A, Matsumoto Y, Izawa K, Horigome K, Kusaka T, Tanaka T, Saito MK, Yasumi T, Nishikomori R, Ohara O, Nakayama N, Nakahata T, Heike T, Toguchida J. Enhanced chondrogenesis of induced pluripotent stem cells from patients with neonatal-onset multisystem inflammatory disease occurs via the caspase 1-independent cAMP/protein kinase A/CREB pathway. Arthritis Rheumatol 2015; 67:302-14. [PMID: 25302486 DOI: 10.1002/art.38912] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 10/07/2014] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Neonatal-onset multisystem inflammatory disease (NOMID) is a dominantly inherited autoinflammatory disease caused by NLRP3 mutations. NOMID pathophysiology is explained by the NLRP3 inflammasome, which produces interleukin-1β (IL-1β). However, epiphyseal overgrowth in NOMID is resistant to anti-IL-1 therapy and may therefore occur independently of the NLRP3 inflammasome. This study was undertaken to investigate the effect of mutated NLRP3 on chondrocytes using induced pluripotent stem cells (iPSCs) from patients with NOMID. METHODS We established isogenic iPSCs with wild-type or mutant NLRP3 from 2 NOMID patients with NLRP3 somatic mosaicism. The iPSCs were differentiated into chondrocytes in vitro and in vivo. The phenotypes of chondrocytes with wild-type and mutant NLRP3 were compared, particularly the size of the chondrocyte tissue produced. RESULTS Mutant iPSCs produced larger chondrocyte masses than wild-type iPSCs owing to glycosaminoglycan overproduction, which correlated with increased expression of the chondrocyte master regulator SOX9. In addition, in vivo transplantation of mutant cartilaginous pellets into immunodeficient mice caused disorganized endochondral ossification. Enhanced chondrogenesis was independent of caspase 1 and IL-1, and thus the NLRP3 inflammasome. Investigation of the human SOX9 promoter in chondroprogenitor cells revealed that the CREB/ATF-binding site was critical for SOX9 overexpression caused by mutated NLRP3. This was supported by increased levels of cAMP and phosphorylated CREB in mutant chondroprogenitor cells. CONCLUSION Our findings indicate that the intrinsic hyperplastic capacity of NOMID chondrocytes is dependent on the cAMP/PKA/CREB pathway, independent of the NLRP3 inflammasome.
Collapse
|
31
|
Chen Y, Li X, Boini KM, Pitzer AL, Gulbins E, Zhang Y, Li PL. Endothelial Nlrp3 inflammasome activation associated with lysosomal destabilization during coronary arteritis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:396-408. [PMID: 25450976 DOI: 10.1016/j.bbamcr.2014.11.012] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 11/05/2014] [Accepted: 11/10/2014] [Indexed: 12/20/2022]
Abstract
Inflammasomes play a critical role in the development of vascular diseases. However, the molecular mechanisms activating the inflammasome in endothelial cells and the relevance of this inflammasome activation is far from clear. Here, we investigated the mechanisms by which an Nlrp3 inflammasome is activated to result in endothelial dysfunction during coronary arteritis by Lactobacillus casei (L. casei) cell wall fragments (LCWE) in a mouse model for Kawasaki disease. Endothelial dysfunction associated with increased vascular cell adhesion protein 1 (VCAM-1) expression and endothelial-leukocyte adhesion was observed during coronary arteritis in mice treated with LCWE. Accompanied with these changes, the inflammasome activation was also shown in coronary arterial endothelium, which was characterized by a marked increase in caspase-1 activity and IL-1β production. In cultured endothelial cells, LCWE induced Nlrp3 inflammasome formation, caspase-1 activation and IL-1β production, which were blocked by Nlrp3 gene silencing or lysosome membrane stabilizing agents such as colchicine, dexamethasone, and ceramide. However, a potassium channel blocker glibenclamide or an oxygen free radical scavenger N-acetyl-l-cysteine had no effects on LCWE-induced inflammasome activation. LCWE also increased endothelial cell lysosomal membrane permeability and triggered lysosomal cathepsin B release into cytosol. Silencing cathepsin B blocked LCWE-induced Nlrp3 inflammasome formation and activation in endothelial cells. In vivo, treatment of mice with cathepsin B inhibitor also abolished LCWE-induced inflammasome activation in coronary arterial endothelium. It is concluded that LCWE enhanced lysosomal membrane permeabilization and consequent release of lysosomal cathepsin B, resulting in activation of the endothelial Nlrp3 inflammasome, which may contribute to the development of coronary arteritis.
Collapse
Affiliation(s)
- Yang Chen
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Xiang Li
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Krishna M Boini
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Ashley L Pitzer
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Erich Gulbins
- Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| | - Yang Zhang
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA.
| | - Pin-Lan Li
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
32
|
Finetti M, Insalaco A, Cantarini L, Meini A, Breda L, Alessio M, D'Alessandro M, Picco P, Martini A, Gattorno M. Long-term efficacy of interleukin-1 receptor antagonist (anakinra) in corticosteroid-dependent and colchicine-resistant recurrent pericarditis. J Pediatr 2014; 164:1425-31.e1. [PMID: 24630353 DOI: 10.1016/j.jpeds.2014.01.065] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 12/23/2013] [Accepted: 01/30/2014] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To evaluate the long-term response and safety of interleukin-1 receptor antagonist (anakinra) in recurrent pericarditis. STUDY DESIGN Fifteen patients (12 children, 3 adults) were enrolled in a multicenter retrospective study. All the patients were corticosteroid-dependent and 14 had received colchicine. Anakinra was given at 1-2 mg/kg/d. The primary outcome of the study was a reduction of at least 70% of disease flares after anakinra treatment compared with the pretreatment period. Secondary outcomes were: (1) number of complete or partial responders to anakinra and time for complete response; (2) number of patients who discontinued other ongoing treatments (non-steroidal anti-inflammatory drugs, corticosteroid, colchicine) and time needed for discontinuation; (3) number of relapses during continuous anakinra treatment; and (4) number of relapses during anakinra tapering or discontinuation. RESULTS All patients treated had a complete response within a few days and were able to rapidly withdraw concomitant treatments, including corticosteroids. During daily treatment, no patient had a relapse of the disease; 14 patients started tapering and 6 of them experienced a relapse, with a prompt response after anakinra reintroduction. Overall, after a median follow-up of 39 months (range 6-57), a 95% reduction of flares was observed compared with pretreatment period. CONCLUSION The long-term use of anakinra in monotherapy is associated with persistent control of recurrent pericarditis.
Collapse
Affiliation(s)
| | | | - Luca Cantarini
- Policlinico Le Scotte, Università di Siena, Siena, Italy
| | | | | | | | | | - Paolo Picco
- UO Pediatria II, Istituto Giannina Gaslini, Genoa, Italy
| | - Alberto Martini
- UO Pediatria II, Istituto Giannina Gaslini, Genoa, Italy; Università di Genoa, Genoa, Italy
| | - Marco Gattorno
- UO Pediatria II, Istituto Giannina Gaslini, Genoa, Italy.
| |
Collapse
|
33
|
Federici S, Gattorno M. A practical approach to the diagnosis of autoinflammatory diseases in childhood. Best Pract Res Clin Rheumatol 2014; 28:263-76. [DOI: 10.1016/j.berh.2014.05.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
34
|
Abstract
Autoinflammatory diseases represent an expanding spectrum of genetic and non-genetic inflammatory diseases characterized by recurrent episodes of fever and systemic inflammation affecting the eyes, joints, skin, and serosal surfaces. Thus, these syndromes are recognized as disorders of innate immunity. Confirming this view, most autoinflammatory diseases are uniquely responsive to IL-1β blockade. Although many autoinflammatory diseases have a genetic cause, increasing evidence indicates that the degree of cell stress concurs to the severity of the disease phenotype. In this mini-review, I will discuss the recent advances on pathogenesis, pathophysiology and therapeutic approaches in autoinflammatory syndromes.
Collapse
|