1
|
Nguyen TTT, Kim YT, Jeong G, Jin M. Immunopathology of and potential therapeutics for secondary hemophagocytic lymphohistiocytosis/macrophage activation syndrome: a translational perspective. Exp Mol Med 2024; 56:559-569. [PMID: 38448692 PMCID: PMC10984945 DOI: 10.1038/s12276-024-01182-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 11/21/2023] [Accepted: 12/19/2023] [Indexed: 03/08/2024] Open
Abstract
Secondary hemophagocytic lymphohistiocytosis/macrophage activation syndrome (sHLH/MAS) is a life-threatening immune disorder triggered by rheumatic disease, infections, malignancies, or medications. Characterized by the presence of hemophagocytic macrophages and a fulminant cytokine storm, sHLH/MAS leads to hyperferritinemia and multiorgan failure and rapidly progresses to death. The high mortality rate and the lack of specific treatments necessitate the development of a new drug. However, the complex and largely unknown immunopathologic mechanisms of sHLH/MAS, which involve dysfunction of various immune cells, diverse etiologies, and different clinical contexts make this effort challenging. This review introduces the terminology, diagnosis, and clinical features of sHLH/MAS. From a translational perspective, this review focuses on the immunopathological mechanisms linked to various etiologies, emphasizing potential drug targets, including key molecules and signaling pathways. We also discuss immunomodulatory biologics, existing drugs under clinical evaluation, and novel therapies in clinical trials. This systematic review aims to provide insights and highlight opportunities for the development of novel sHLH/MAS therapeutics.
Collapse
Affiliation(s)
- Tram T T Nguyen
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Yoon Tae Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Geunyeol Jeong
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Mirim Jin
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea.
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea.
- Department of Microbiology, College of Medicine, Gachon University, Incheon, Republic of Korea.
| |
Collapse
|
2
|
Burlaka AP, Liubenko DL, Burlaka AA, Yevtushenko OI, Ganusevich II. CORONAVIRUS SARS-COV-2 MODIFIES ANTITUMOR REDOX STATUS OF BLOOD AND INTERCELLULAR MATRIX IN METASTATIC COLORECTAL CANCER PATIENTS (A PILOT STUDY). Exp Oncol 2024; 45:483-492. [PMID: 38328841 DOI: 10.15407/exp-oncology.2023.04.483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Indexed: 02/09/2024]
Abstract
BACKGROUND The current studies demonstrate that SARS-CoV-2 infection results in increasing complications incidence and the total risk of death in cancer patients. SARS-CoV-2 infection triggers oxidative stress representing one of the major factors of the inflammation contributing to the complicated course of the diseases including cancer. AIM To assess the effect of hypoxia caused by SARS-CoV-2 infection on the redox status of blood in patients with metastatic colorectal cancer (mCRC). MATERIALS AND METHODS 10 patients with SARS-CoV-2, 11 mCRC patients with metachronous liver disease, and 14 mCRC patients with preceding SARS-CoV-2 infection were included in the study. The data on blood biochemistry (C-reactive protein, ferritin, transferrin, and free iron) were analyzed. The levels of superoxide radicals (ROS) in blood cells were determined by electron paramagnetic resonance (EPR) using the spin trap technique. The metalloproteinase activity was measured by polyacrylamide gel zymography with the addition of gelatin as a substrate. RESULTS In mCRC patients with prior SARS-CoV-2 infection, a 1.26-fold increase in ROS-generating activity of blood neutrophils was observed compared to mCRC patients with no history of SARS-CoV-2 infection. The blood content of C-reactive protein, transferrin, and free iron in mCRC patients with prior SARS-CoV-2 infection increased by 2, 6, and 1.4 times, respectively. The total activity of gelatinases in platelets and neutrophils in the blood of mCRC patients with prior SARS-CoV-2 infection was 1.4 and 1.2 times higher compared to mCRC patients with no history of SARS-CoV-2 infection. CONCLUSION mCRC patients with prior COVID-19 have a higher risk of exacerbation of inflammatory reactions. SARS-CoV-2 infection results in redox dіsbalance, which may contribute to the unfavorable course of the disease.
Collapse
Affiliation(s)
- A P Burlaka
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, Kyiv, Ukraine
| | - D L Liubenko
- Bogomolets National Medical University, Kyiv, Ukraine
| | - A A Burlaka
- State Non-commercial Enterprise "National Cancer Institute", Kyiv, Ukraine
| | - O I Yevtushenko
- Shupyk National Healthcare University of Ukraine, Kyiv, Ukraine
| | - I I Ganusevich
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, Kyiv, Ukraine
| |
Collapse
|
3
|
Ruscitti P, Cantarini L, Nigrovic PA, McGonagle D, Giacomelli R. Recent advances and evolving concepts in Still's disease. Nat Rev Rheumatol 2024; 20:116-132. [PMID: 38212542 DOI: 10.1038/s41584-023-01065-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2023] [Indexed: 01/13/2024]
Abstract
Still's disease is a rare inflammatory syndrome that encompasses systemic juvenile idiopathic arthritis and adult-onset Still's disease, both of which can exhibit life-threatening complications, including macrophage activation syndrome (MAS), a secondary form of haemophagocytic lymphohistiocytosis. Genetic insights into Still's disease involve both HLA and non-HLA susceptibility genes, suggesting the involvement of adaptive immune cell-mediated immunity. At the same time, phenotypic evidence indicates the involvement of autoinflammatory processes. Evidence also implicates the type I interferon signature, mechanistic target of rapamycin complex 1 signalling and ferritin in the pathogenesis of Still's disease and MAS. Pathological entities associated with Still's disease include lung disease that could be associated with biologic DMARDs and with the occurrence of MAS. Historically, monophasic, recurrent and persistent Still's disease courses were recognized. Newer proposals of alternative Still's disease clusters could enable better dissection of clinical heterogeneity on the basis of immune cell profiles that could represent diverse endotypes or phases of disease activity. Therapeutically, data on IL-1 and IL-6 antagonism and Janus kinase inhibition suggest the importance of early administration in Still's disease. Furthermore, there is evidence that patients who develop MAS can be treated with IFNγ antagonism. Despite these developments, unmet needs remain that can form the basis for the design of future studies leading to improvement of disease management.
Collapse
Affiliation(s)
- Piero Ruscitti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Luca Cantarini
- Department of Medical Sciences, Surgery and Neurosciences, Research Center of Systemic Autoinflammatory Diseases and Behçet's Disease Clinic, University of Siena, Siena, Italy
| | - Peter A Nigrovic
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, UK
- National Institute for Health Research (NIHR) Leeds Biomedical Research Centre (BRC), Leeds Teaching Hospitals, Leeds, UK
| | - Roberto Giacomelli
- Clinical and research section of Rheumatology and Clinical Immunology, Fondazione Policlinico Campus Bio-Medico, Rome, Italy
- Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| |
Collapse
|
4
|
Berardicurti O, Navarini L, Vomero M, Currado D, Biaggi A, Marino A, Bearzi P, Corberi E, Rigon A, Arcarese L, Vadacca M, Mattei A, Agrò FE, Incalzi RA, Sambuco F, Travaglino F, Ruscitti P, Cipriani P, Giacomelli R. The similar expression of both ferritin and scavenger receptors activation genes in patients with COVID19 and AOSD support their role in the pathogenesis of these diseases and identify a common mechanism at the basis of the "hyperferritinemic syndromes". Autoimmun Rev 2023; 22:103309. [PMID: 36889656 PMCID: PMC9986116 DOI: 10.1016/j.autrev.2023.103309] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023]
Abstract
A role for COVID19 in "hyperferritinemic syndromes" has been proposed based on its clinical and serological characteristics and its similarities with AOSD. To better understand the molecular pathways responsible of these similarities, we evaluated in the PBMCs of 4 active AOSD patients, 2 COVID19 patients with ARDS, and 2 HCs the expression of genes associated with iron metabolisms, with monocyte/macrophages activation, and finally with NETs formation.
Collapse
Affiliation(s)
- Onorina Berardicurti
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policiclinico Campus Biomedico, Via Alvaro del Portillo 200, 00128 Rome, Italy; Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy.
| | - Luca Navarini
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policiclinico Campus Biomedico, Via Alvaro del Portillo 200, 00128 Rome, Italy; Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Marta Vomero
- Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Damiano Currado
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policiclinico Campus Biomedico, Via Alvaro del Portillo 200, 00128 Rome, Italy; Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Alice Biaggi
- Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Annalisa Marino
- Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Pietro Bearzi
- Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Erika Corberi
- Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Amelia Rigon
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policiclinico Campus Biomedico, Via Alvaro del Portillo 200, 00128 Rome, Italy
| | - Luisa Arcarese
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policiclinico Campus Biomedico, Via Alvaro del Portillo 200, 00128 Rome, Italy
| | - Marta Vadacca
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policiclinico Campus Biomedico, Via Alvaro del Portillo 200, 00128 Rome, Italy
| | - Alessia Mattei
- Operative Research Unit of Anaesthesia, Intensive Care and Pain Management, Fondazione Policiclinico Campus Biomedico, Via Alvaro del Portillo 200, 00128 Rome, Italy
| | - Felice Eugenio Agrò
- Operative Research Unit of Anaesthesia, Intensive Care and Pain Management, Fondazione Policiclinico Campus Biomedico, Via Alvaro del Portillo 200, 00128 Rome, Italy; Anesthesia, Intensive Care and Pain Management, Department of Medicine, University of Rome "Campus Bio-Medico", Rome, Italy
| | - Raffaele Antonelli Incalzi
- Unit of Geriatrics, Campus Bio-Medico University of Rome, Rome, Italy; Internal Medicine, Fondazione Policiclinico Campus Biomedico, Via Alvaro del Portillo 200, 00128, Rome, Italy
| | - Federica Sambuco
- Emergency Department, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Roma, Italy
| | - Francesco Travaglino
- Emergency Department, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Roma, Italy
| | - Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Paola Cipriani
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Roberto Giacomelli
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policiclinico Campus Biomedico, Via Alvaro del Portillo 200, 00128 Rome, Italy; Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| |
Collapse
|
5
|
Ruscitti P, Ursini F, Berardicurti O, Masedu F, Bozzalla Cassione E, Naldi S, Di Cola I, Di Muzio C, De Stefano L, Di Nino E, Navarini L, Vomero M, Bugatti S, Valenti M, Mariani E, Iagnocco A, Montecucco C, Giacomelli R, Cipriani P. Cytokine profile, ferritin and multi-visceral involvement characterize macrophage activation syndrome during adult-onset Still's disease. Rheumatology (Oxford) 2022; 62:321-329. [PMID: 35438139 DOI: 10.1093/rheumatology/keac247] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/01/2022] [Accepted: 04/01/2022] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVES To multidimensionally characterize macrophage activation syndrome (MAS) complicating adult-onset Still's disease (AOSD) considering cytokine profile, inflammatory markers and multi-visceral involvement of the disease. To perform a high-dimensional phenotypic analysis of circulating immune cells in AOSD patients with and without MAS. To assess interferon (IFN)-related pathways in AOSD synovial tissues by a bulky RNA sequencing. METHODS Clinical and biologic data were collected and compared in AOSD patients with and without MAS. Sera biomolecules were analysed by Luminex multiplexing technology. Mass cytometry (CyTOF) was used to characterize circulating immune cells. A bulky RNA sequencing was performed in AOSD synovial tissues. RESULTS Forty consecutive AOSD patients were assessed, 14 complicated with MAS. Paralleling with increases of systemic score and ferritin, MAS patients showed higher levels of IL-1α, IL-1β, IL-1Ra, IL-2Ra, IL-6, IL-10, IL-17A, IFN-γ, G-CSF, MCP-1, MIP-1α and SCF. Combining the discriminatory ability of these data in identifying MAS, the best model was composed by systemic score, ferritin, IFN-γ and IL-10. By CyTOF analysis, MAS patients showed an increase of circulating 'classical monocytes' and a reduction of total NK cells. Our assessment showed 3477 IFN-related genes (IRGs) were differently expressed in AOSD synovial tissues. CONCLUSIONS A multidimensional characterization of AOSD patients suggested that IFN-γ, IL-10, ferritin and systemic score discriminated the occurrence of cytokine storm syndrome associated with MAS. The inflammatory milieu of AOSD and MAS may be related to a signature of circulating immune cells. Finally, our results about IRGs reinforced the role of IFN-γ in these patients.
Collapse
Affiliation(s)
- Piero Ruscitti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila
| | - Francesco Ursini
- Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli.,Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, Bologna
| | - Onorina Berardicurti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila
| | - Francesco Masedu
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila.,Academy of Sciences of Abruzzo Region, Abruzzo
| | | | - Susanna Naldi
- Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli.,Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, Bologna
| | - Ilenia Di Cola
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila
| | - Claudia Di Muzio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila
| | - Ludovico De Stefano
- Rheumatology Department, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia
| | - Elena Di Nino
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila
| | - Luca Navarini
- Rheumatology and Immunology Unit, Department of Medicine, University of Rome Campus Biomedico, Rome
| | - Marta Vomero
- Rheumatology and Immunology Unit, Department of Medicine, University of Rome Campus Biomedico, Rome
| | - Serena Bugatti
- Rheumatology Department, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia
| | - Marco Valenti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila
| | - Erminia Mariani
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna.,Laboratory of Immunorheumatology and Tissue Regeneration, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna
| | - Annamaria Iagnocco
- Academic Rheumatology Centre, Ospedale Mauriziano - Dipartimento Scienze Cliniche e Biologiche, Università degli Studi di Torino, Turin, Italy
| | | | - Roberto Giacomelli
- Rheumatology and Immunology Unit, Department of Medicine, University of Rome Campus Biomedico, Rome
| | - Paola Cipriani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila
| |
Collapse
|
6
|
Tsuboi H, Segawa S, Yagishita M, Toko H, Honda F, Kitada A, Miki H, Ohyama A, Hagiwara S, Kondo Y, Matsumoto I, Sumida T. Activation mechanisms of monocytes/macrophages in adult-onset Still disease. Front Immunol 2022; 13:953730. [PMID: 36090971 PMCID: PMC9461677 DOI: 10.3389/fimmu.2022.953730] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/02/2022] [Indexed: 12/23/2022] Open
Abstract
Adult onset Still disease (AOSD) is a systemic inflammatory disorder characterized by skin rash, spiking fever, arthritis, sore throat, lymphadenopathy, and hepatosplenomegaly. Although the etiology of this disease has not been fully clarified, both innate and acquired immune responses could contribute to its pathogenesis. Hyperactivation of macrophages and neutrophils along with low activation of natural killer (NK) cells in innate immunity, as well as hyperactivation of Th1 and Th17 cells, whereas low activation of regulatory T cells (Tregs) in acquired immunity are involved in the pathogenic process of AOSD. In innate immunity, activation of monocytes/macrophages might play central roles in the development of AOSD and macrophage activation syndrome (MAS), a severe life-threating complication of AOSD. Regarding the activation mechanisms of monocytes/macrophages in AOSD, in addition to type II interferon (IFN) stimulation, several pathways have recently been identified, such as the pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs)-pattern recognition receptors (PRRs) axis, and neutrophil extracellular traps (NETs)-DNA. These stimulations on monocytes/macrophages cause activation of the nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain (NLRP) 3 inflammasomes, which trigger capase-1 activation, resulting in conversion of pro-IL-1β and pro-IL-18 into mature forms. Thereafter, IL-1β and IL-18 produced by activated monocytes/macrophages contribute to various clinical features in AOSD. We identified placenta-specific 8 (PLAC8) as a specifically increased molecule in monocytes of active AOSD, which correlated with serum levels of CRP, ferritin, IL-1β, and IL-18. Interestingly, PLAC8 could suppress the synthesis of pro-IL-1β and pro-IL-18 via enhanced autophagy; thus, PLAC8 seems to be a regulatory molecule in AOSD. These findings for the activation mechanisms of monocytes/macrophages could shed light on the pathogenesis and development of a novel therapeutic strategy for AOSD.
Collapse
|
7
|
Rao S, Tsang LSL, Zhao M, Shi W, Lu Q. Adult-onset Still’s disease: A disease at the crossroad of innate immunity and autoimmunity. Front Med (Lausanne) 2022; 9:881431. [PMID: 36072947 PMCID: PMC9442343 DOI: 10.3389/fmed.2022.881431] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 07/27/2022] [Indexed: 01/12/2023] Open
Abstract
Adult-onset Still’s disease (AOSD) is a rare disease affecting multiple systems and organs with unknown etiology, and the clinical symptoms are usually described as spiking fever, arthritis, evanescent salmon-pink eruptions, lymphadenopathy, splenomegaly, and other manifestations. The laboratory indicators are not specific, often presenting as increased leukocyte counts and neutrophil percentage, elevated erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP), hyperferritinemia, and increased inflammatory factors. ANA, ENA, and RF are negative. According to those unspecific clinical presentations and laboratory findings, infection, tumor, connective tissue disease, and other diseases must be ruled out before diagnosis. The diagnosis of AOSD is a great challenge for clinicians. The mechanism of AOSD pathogenesis is complicated and still being studied. There is a new opinion that atypical persistent skin eruptions (APSEs) with specific histological manifestations are unique for AOSD, and APSEs might be on a spectrum with classical evanescent eruptions. Studies on APSEs showed that IL-1β and IFN-γ are strongly correlated with the pathogenesis of necrosis keratinocytes in APSEs. IL-1β is strongly involved in inflammatory disease when it is abnormal, and plays an important role in the pathogenesis of neutrophil dermatosis. In the early stage of AOSD, skin lesions appear to be evanescent urticaria-like eruptions accompanied by fever, and only neutrophils infiltrate around the blood vessels in the dermis pathologically. As the course of the disease progresses, IL-1β is gradually released. Through the stimulation of other inflammatory factors and the influence of unknown factors, IL-1β gradually infiltrates into the stratum corneum and finally accumulates around the necrotic keratinocytes of the stratum corneum. However, the detailed mechanism is still unknown. IFN-γ could play a pro-inflammatory or regulatory role in some disorders. IL-1β can enhance the expression of IFN-γ, and IFN-γ can cause keratinocyte apoptosis by activating the autocrine of caspase. Also, several pieces of evidence indicate that adaptive immunity is also involved in the pathogenesis of AOSD. Increased α-soluble receptors of IL-2 may suggest T-cell activation and proliferation in AOSD patients. Increased IL-4- and IFN-γ-producing T cells were found in active AOSD and related to disease severity. Frequencies of Treg cells in AOSD were significantly lower and were inversely correlated with disease severity. According to these, more and more researchers have reached a consensus that AOSD is a disease at the crossroads of innate immunity and autoimmunity. In this review, we will provide a comprehensive insight into AOSD, describing research progress and the immunological mechanism contribution to the disease. In the meantime, different treatment options and the efficacy and safety of various biologic agents are also discussed. A further understanding of AOSD requires closer cooperation among doctors from different departments, and this review will provide a new idea for diagnosis and therapeutic options.
Collapse
Affiliation(s)
- Shijia Rao
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
| | - Lemuel Shui-Lun Tsang
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Ming Zhao
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
| | - Wei Shi
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Wei Shi,
| | - Qianjin Lu
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China
- Qianjin Lu,
| |
Collapse
|
8
|
Ruscitti P, Di Cola I, Di Muzio C, Italiano N, Ursini F, Giacomelli R, Cipriani P. Expanding the spectrum of the hyperferritinemic syndrome, from pathogenic mechanisms to clinical observations, and therapeutic implications. Autoimmun Rev 2022; 21:103114. [PMID: 35595050 DOI: 10.1016/j.autrev.2022.103114] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/15/2022] [Indexed: 01/19/2023]
Abstract
From the introduction of hyperferritinemic syndrome concept, a growing body of evidence has suggested the role of ferritin as a pathogenic mediator and a relevant clinical feature in the management of patients with inflammatory diseases. From a pathogenic point of view, ferritin may directly stimulate the aberrant immune response by triggering the production of pro-inflammatory mediators in inducing a vicious pathogenic loop and contributing to the occurrence of cytokine storm syndrome. The latter has been recently defined as a clinical picture characterised by elevated circulating cytokine levels, acute systemic inflammatory symptoms, and secondary organ dysfunction beyond that which could be attributed to a normal response to a pathogen It is noteworthy that the occurrence of hyperferritinemia may be correlated with the development of the cytokine storm syndrome in the context of an inflammatory disease. In addition to adult onset Still's disease, macrophage activation syndrome, catastrophic anti-phospholipids syndrome, and septic shock, recent evidence has suggested this association between ferritin and life-threatening evolution in patients with systemic lupus erythematosus, with anti-MDA5 antibodies in the context of poly-dermatomyositis, with severe COVID-19, and with multisystem inflammatory syndrome. The possible underlying common inflammatory mechanisms, associated with hyperferritinemia, may led to the similar clinical picture observed in these patients. Furthermore, similar therapeutic strategies could be suggested inhibiting pro-inflammatory cytokines and improving long-term outcomes in these disorders. Thus, it could be possible to expand the spectrum of the hyperferritinemic syndrome to those diseases burdened by a dreadful clinical picture correlated with hyperferritinemia and the occurrence of the cytokine storm syndrome. In addition, the assessment of ferritin may provide useful information to the physicians in clinical practice to manage these patients. Therefore, ferritin may be considered a relevant clinical feature to be used as biomarker in dissecting the unmet needs in the management of these disorders. Novel evidence may thus support an expansion of the spectrum of the hyperferritinemic syndrome to these diseases burdened by a life-threatening clinical picture correlated with hyperferritinemia and the occurrence of the cytokine storm syndrome.
Collapse
Affiliation(s)
- Piero Ruscitti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Ilenia Di Cola
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Claudia Di Muzio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Noemi Italiano
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Francesco Ursini
- Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy; Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Roberto Giacomelli
- Rheumatology and Immunology Unit, Department of Medicine, University of Rome Campus Biomedico, Rome, Italy
| | - Paola Cipriani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
9
|
Ruscitti P, Berardicurti O, Di Cola I, Di Muzio C, Di Nino E, Giacomelli R, Cipriani P. The hyper-expression of NLRP4 characterizes the occurrence of macrophage activation syndrome assessing STING pathway in adult-onset Still's disease. Clin Exp Immunol 2022; 208:95-102. [PMID: 35467709 PMCID: PMC9113323 DOI: 10.1093/cei/uxac014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/21/2022] [Accepted: 01/27/2022] [Indexed: 02/04/2023] Open
Abstract
To assess stimulator of interferon genes (STING) pathway in patients with adult-onset Still's disease (AOSD) who were complicated or not by macrophage activation syndrome (MAS), evaluating peripheral blood mononuclear cells (PBMCs), and synovial tissues. The relative mRNA expression of key molecules of the STING pathway (i.e. CGAS, NLRP4, PKDC, STING1, XRCC5, and XRCC6) and interferon (IFN)-γ was assessed in PBMCs obtained from patients with AOSD, who were complicated or not by MAS, and healthy controls (HCs). A bulky RNA sequencing was performed in synovial tissues from two patients with AOSD. Finally, the ability of heavy ferritin subunit (FeH) to induce the expression of NLRP4 was evaluated in cultured macrophages. Twenty patients with AOSD were analysed. Out of them, seven patients were complicated by MAS. Assessing mRNA relative expression in PBMCs, STING1, NLRP4, XRCC6, and IFN-γ were significantly expressed in AOSD than HCs. The mRNA relative expression of CGAS, PKDC, and XRCC5 did not differ between patients and HCs. Furthermore, NLRP4 and IFN-γ resulted to be significantly increased in patients with AOSD complicated by MAS than others. By RNA-sequencing analysis, we observed that Nlrp4 gene was significantly up-regulated in patients with AOSD. Following the stimulation with FeH, an increased expression of NLRP4 was observed in cultured macrophages. In conclusion, an increased expression of some key molecules of STING pathway characterized patients with AOSD. In addition, our results suggested that a hyper-activity of NLRP4 may be observed in patients with MAS. Furthermore, FeH increased the expression of NLRP4 in cultured macrophages.
Collapse
Affiliation(s)
- Piero Ruscitti
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Onorina Berardicurti
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Ilenia Di Cola
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Claudia Di Muzio
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Elena Di Nino
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Roberto Giacomelli
- Rheumatology and Immunology Unit, Department of Medicine, University of Rome Campus Biomedico, Rome, Italy
| | - Paola Cipriani
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
10
|
Moreno-Fernandez J, Ochoa JJ, De Paco Matallana C, Caño A, Martín-Alvarez E, Sanchez-Romero J, Toledano JM, Puche-Juarez M, Prados S, Ruiz-Duran S, Diaz-Meca L, Carrillo MP, Diaz-Castro J. COVID-19 during Gestation: Maternal Implications of Evoked Oxidative Stress and Iron Metabolism Impairment. Antioxidants (Basel) 2022; 11:184. [PMID: 35204067 PMCID: PMC8868249 DOI: 10.3390/antiox11020184] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/15/2022] [Accepted: 01/17/2022] [Indexed: 12/23/2022] Open
Abstract
COVID-19 has reached pandemic proportions worldwide, with considerable consequences for both health and the economy. In pregnant women, COVID-19 can alter the metabolic environment, iron metabolism, and oxygen supply of trophoblastic cells, and therefore have a negative influence on essential mechanisms of fetal development. The purpose of this study was to investigate, for the first time, the effects of COVID-19 infection during pregnancy with regard to the oxidative/antioxidant status in mothers' serum and placenta, together with placental iron metabolism. Results showed no differences in superoxide dismutase activity and placental antioxidant capacity. However, antioxidant capacity decreased in the serum of infected mothers. Catalase activity decreased in the COVID-19 group, while an increase in 8-hydroxy-2'-deoxyguanosine, hydroperoxides, 15-FT-isoprostanes, and carbonyl groups were recorded in this group. Placental vitamin D, E, and Coenzyme-Q10 also showed to be increased in the COVID-19 group. As for iron-related proteins, an up-regulation of placental DMT1, ferroportin-1, and ferritin expression was recorded in infected women. Due to the potential role of iron metabolism and oxidative stress in placental function and complications, further research is needed to explain the pathogenic mechanism of COVID-19 that may affect pregnancy, so as to assess the short-term and long-term outcomes in mothers' and infants' health.
Collapse
Affiliation(s)
- Jorge Moreno-Fernandez
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.-F.); (J.M.T.); (M.P.-J.); (J.D.-C.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain
- Clinical Medicine and Public Health Ph.D. Program, University of Granada, 18071 Granada, Spain
| | - Julio J. Ochoa
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.-F.); (J.M.T.); (M.P.-J.); (J.D.-C.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain
| | - Catalina De Paco Matallana
- Department of Obstetrics and Gynecology, Hospital Clínico Universitario ‘Virgen de la Arrixaca’, El Palmar, 30120 Murcia, Spain; (J.S.-R.); (L.D.-M.)
- Institute for Biomedical Research of Murcia, IMIB-Arrixaca, El Palmar, 30120 Murcia, Spain
| | - Africa Caño
- Department of Obstetrics and Gynaecology, San Cecilio Universitary Hospital, 18071 Granada, Spain; (A.C.); (S.P.)
| | - Estefania Martín-Alvarez
- Unit of Neonatology, Pediatric Service, Hospital Universitario Materno-Infantil Virgen de las Nieves, 18014 Granada, Spain;
| | - Javier Sanchez-Romero
- Department of Obstetrics and Gynecology, Hospital Clínico Universitario ‘Virgen de la Arrixaca’, El Palmar, 30120 Murcia, Spain; (J.S.-R.); (L.D.-M.)
- Institute for Biomedical Research of Murcia, IMIB-Arrixaca, El Palmar, 30120 Murcia, Spain
| | - Juan M. Toledano
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.-F.); (J.M.T.); (M.P.-J.); (J.D.-C.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain
- Nutrition and Food Sciences Ph.D. Program, University of Granada, 18071 Granada, Spain
| | - Maria Puche-Juarez
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.-F.); (J.M.T.); (M.P.-J.); (J.D.-C.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain
- Nutrition and Food Sciences Ph.D. Program, University of Granada, 18071 Granada, Spain
| | - Sonia Prados
- Department of Obstetrics and Gynaecology, San Cecilio Universitary Hospital, 18071 Granada, Spain; (A.C.); (S.P.)
| | - Susana Ruiz-Duran
- Department of Obstetrics & Gynaecology, Virgen de las Nieves University Hospital, 18014 Granada, Spain; (S.R.-D.); (M.P.C.)
| | - Lucia Diaz-Meca
- Department of Obstetrics and Gynecology, Hospital Clínico Universitario ‘Virgen de la Arrixaca’, El Palmar, 30120 Murcia, Spain; (J.S.-R.); (L.D.-M.)
- Institute for Biomedical Research of Murcia, IMIB-Arrixaca, El Palmar, 30120 Murcia, Spain
| | - María Paz Carrillo
- Department of Obstetrics & Gynaecology, Virgen de las Nieves University Hospital, 18014 Granada, Spain; (S.R.-D.); (M.P.C.)
| | - Javier Diaz-Castro
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.-F.); (J.M.T.); (M.P.-J.); (J.D.-C.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS), 18012 Granada, Spain
| |
Collapse
|
11
|
Jia F, Liu H, Kang S. NCOA4-Mediated Ferritinophagy: A Vicious Culprit in COVID-19 Pathogenesis? Front Mol Biosci 2022; 8:761793. [PMID: 34977155 PMCID: PMC8714652 DOI: 10.3389/fmolb.2021.761793] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/30/2021] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a global pandemic that has caused widespread loss of life. Notably, in this disease, severe inflammatory reactions characterized by cytokine storms are caused by severe acute respiratory syndrome coronavirus 2. The cytokine storms may promote hyper-ferritinemia which can further intensify the inflammation. Moreover, elevated ferritin levels trigger nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy, in which ferritin is degraded and iron is released. Excess iron released from ferritinophagy can promote ferroptosis and cellular damage. Therefore, we propose that NCOA4-mediated ferritinophagy can be targeted to limit the ferroptosis and prevent the multi-organ damage and severity in COVID-19 patients.
Collapse
Affiliation(s)
- Fengju Jia
- School of Nursing, Qingdao University, Qingdao, China
| | - Hongxia Liu
- Yantai Ludong Hospital (Shandong Provincial Hospital Group), Yantai, China
| | - Shan Kang
- Department of Laboratory, Qingdao Eighth People's Hospital, Qingdao, China
| |
Collapse
|
12
|
Ruscitti P, Berardicurti O, Giacomelli R, Cipriani P. The clinical heterogeneity of adult onset Still's disease may underlie different pathogenic mechanisms. Implications for a personalised therapeutic management of these patients. Semin Immunol 2021; 58:101632. [PMID: 35787972 DOI: 10.1016/j.smim.2022.101632] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Adult-onset Still's disease (AOSD) is a rare inflammatory disease of unknown aetiology usually affecting young adults and manifesting with a clinical triad of spiking fever, arthritis, and evanescent cutaneous rash. AOSD may be considered a highly heterogeneous disease, despite a similar clinical presentation, the disease course may be completely different. Some patients may have a single episode of the disease whereas others may evolve toward a chronic course and experience life-threatening complications. On these bases, to dissect the clinical heterogeneity of this disease, four different subsets were identified combining the manifestations at the beginning with possible diverse outcomes over time. Each one of these derived subsets would be characterised by a prominent different clinical feature from others, thus proposing dissimilar underlying pathogenic mechanisms, at least partially. Consequently, a distinct management of AOSD may be suggested to appropriately tailor the therapeutic strategy to these patients, according to principles of the precision medicine. These findings would also provide the rationale to recognise a different genetic and molecular profile of patients with AOSD. Taking together these findings, the basis for a precision medicine approach may be suggested in AOSD, which would drive a tailored therapeutic approach in these patients. A better patient stratification may also help in arranging specific designed studies to improve the management of patients with AOSD. Behind these different clinical phenotypes, distinct endotypes of AOSD may be suggested, probably differing in pathogenesis, outcomes, and response to therapies.
Collapse
Affiliation(s)
- Piero Ruscitti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Onorina Berardicurti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Roberto Giacomelli
- Unit of Rheumatology and Clinical Immunology, University of Rome "Campus Biomedico", Rome, Italy
| | - Paola Cipriani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
13
|
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
|
14
|
An Update on the Pathogenic Role of Macrophages in Adult-Onset Still's Disease and Its Implication in Clinical Manifestations and Novel Therapeutics. J Immunol Res 2021; 2021:8998358. [PMID: 34239943 PMCID: PMC8238602 DOI: 10.1155/2021/8998358] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/23/2021] [Accepted: 05/28/2021] [Indexed: 12/18/2022] Open
Abstract
Increasing evidence indicates a pivotal role of macrophages in innate immunity, which contributes to the pathogenesis of adult-onset Still's disease (AOSD). Despite the available reviews that summarized the pathogenic role of proinflammatory cytokines in AOSD, a systematic approach focusing on the crucial role of macrophages in this disease is still lacking. This review summarizes the updated functions of macrophages in AOSD and their implication in clinical manifestations and therapeutics. We searched the MEDLINE database using the PubMed interface and reviewed the English-language literature as of 31 March 2021, from 1971 to 2021. We focus on the existing evidence on the pathogenic role of macrophages in AOSD and its implication in clinical characteristics and novel therapeutics. AOSD is an autoinflammatory disease mainly driven by the innate immune response. Among the innate immune responses, macrophage activation is a hallmark of AOSD pathogenesis. The pattern recognition receptors (PRRs) on macrophages recognize pathogen-associated molecular patterns and damage-associated molecular patterns and subsequently cause overproduction of proinflammatory cytokines and recruit adaptive immunity. Some biomarkers, such as ferritin and gasdermin D, reflecting macrophage activation were elevated and correlated with AOSD activity. Given that macrophage activation with the overproduction of proinflammatory cytokines plays a pathogenic role in AOSD, these inflammatory mediators would be the therapeutic targets. Accordingly, the inhibitors to interleukin- (IL-) 1, IL-6, and IL-18 have been shown to be effective in AOSD treatment. Gaining insights into the pathogenic role of macrophages in AOSD can aid in identifying disease biomarkers and therapeutic agents for this disease.
Collapse
|
15
|
The joint involvement in adult onset Still's disease is characterised by a peculiar magnetic resonance imaging and a specific transcriptomic profile. Sci Rep 2021; 11:12455. [PMID: 34127696 PMCID: PMC8203668 DOI: 10.1038/s41598-021-91613-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 05/21/2021] [Indexed: 02/06/2023] Open
Abstract
Adult onset Still's disease (AOSD) is a rare systemic autoinflammatory disease, characterised by fever, arthritis, and skin rash, and joint involvement is one of its clinical manifestations. The aims of this work were to assess joint involvement, to describe main patterns of involvement, and associated clinical characteristics. In this work, we aimed at assessing the joint involvement in AOSD by using MRI, to describe main patterns and associated clinical characteristics. In addition, we aimed at assessing the global transcriptomic profile of synovial tissues in AOSD to elucidate possible pathogenic pathways involved. We also evaluated the global transcriptomic profile of synovial tissues to elucidate possible pathogenic pathways involved in the disease. Thus, AOSD patients, who underwent to MRI exam on joints, were assessed to describe patterns of joint involvement and associated clinical characteristics. Some synovial tissues were collected for RNA-sequencing purposes. The most common MRI finding was the presence of synovitis on 60.5%, mainly in peripheral affected joints, with low to intermediate signal intensity on T1-weighted images and intermediate to high signal intensity on T2-fat-saturated weighted and STIR images. Bone oedema and MRI-bone erosions were reported on 34.9% and 25.6% MRI exams, respectively. Patients with MRI-bone erosions showed a higher prevalence of splenomegaly, a more frequent chronic disease course, lower levels of erythrocyte sedimentation rate, and ferritin. In AOSD synovial tissues, a hyper-expression of interleukin (IL)-1, IL-6, and TNF pathways was shown together with ferritin genes. In conclusion, in AOSD patients, the most common MRI-finding was the presence of synovitis, characterised by intermediate to high signal intensity on T2-fat-saturated weighted and STIR images. MRI-bone erosions and bone oedema were also observed. In AOSD synovial tissues, IL-1, IL-6, and TNF pathways together with ferritin genes resulted to be hyper-expressed.
Collapse
|
16
|
Liu Q, Wu J, Zhang X, Wu X, Zhao Y, Ren J. Iron homeostasis and disorders revisited in the sepsis. Free Radic Biol Med 2021; 165:1-13. [PMID: 33486088 DOI: 10.1016/j.freeradbiomed.2021.01.025] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/31/2020] [Accepted: 01/11/2021] [Indexed: 12/26/2022]
Abstract
Sepsis is a life-threatening condition caused by a dysregulated host-response to inflammation, although it currently lacks a fully elucidated pathobiology. Iron is a crucial trace element that is essential for fundamental processes in both humans and bacteria. During sepsis, iron metabolism is altered, including increased iron transport and uptake into cells and decreased iron export. The intracellular sequestration of iron limits its availability to circulating pathogens, which serves as a conservative strategy against the pathogens. Although iron retention has been showed to have protective protect effects, an increase in labile iron may cause oxidative injury and cell death (e.g., pyroptosis, ferroptosis) as the condition progresses. Moreover, iron disorders are substantial and correlate with the severity of sepsis. This also suggests that iron may be useful as a diagnostic marker for evaluating the severity and predicting the outcome of the disease. Further knowledge about these disorders could help in evaluating how drugs targeting iron homeostasis can be optimally applied to improve the treatment of patients with sepsis. Here, we present a comprehensive review of recent advances in the understanding of iron metabolism, focusing on the regulatory mechanisms and iron-mediated injury in sepsis.
Collapse
Affiliation(s)
- Qinjie Liu
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China.
| | - Jie Wu
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210002, PR China.
| | - Xufei Zhang
- Research Institute of General Surgery, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, PR China.
| | - Xiuwen Wu
- Research Institute of General Surgery, Jinling Hospital, Nanjing, 210002, PR China.
| | - Yun Zhao
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210002, PR China.
| | - Jianan Ren
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China; Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210002, PR China; Research Institute of General Surgery, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, PR China.
| |
Collapse
|
17
|
Perricone C, Bartoloni E, Bursi R, Cafaro G, Guidelli GM, Shoenfeld Y, Gerli R. COVID-19 as part of the hyperferritinemic syndromes: the role of iron depletion therapy. Immunol Res 2020. [PMID: 32681497 DOI: 10.22541/au.158880283.34604328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
SARS-CoV-2 infection is characterized by a protean clinical picture that can range from asymptomatic patients to life-threatening conditions. Severe COVID-19 patients often display a severe pulmonary involvement and develop neutrophilia, lymphopenia, and strikingly elevated levels of IL-6. There is an over-exuberant cytokine release with hyperferritinemia leading to the idea that COVID-19 is part of the hyperferritinemic syndrome spectrum. Indeed, very high levels of ferritin can occur in other diseases including hemophagocytic lymphohistiocytosis, macrophage activation syndrome, adult-onset Still's disease, catastrophic antiphospholipid syndrome and septic shock. Numerous studies have demonstrated the immunomodulatory effects of ferritin and its association with mortality and sustained inflammatory process. High levels of free iron are harmful in tissues, especially through the redox damage that can lead to fibrosis. Iron chelation represents a pillar in the treatment of iron overload. In addition, it was proven to have an anti-viral and anti-fibrotic activity. Herein, we analyse the pathogenic role of ferritin and iron during SARS-CoV-2 infection and propose iron depletion therapy as a novel therapeutic approach in the COVID-19 pandemic.
Collapse
Affiliation(s)
- Carlo Perricone
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy
| | - Elena Bartoloni
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy
| | - Roberto Bursi
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy
| | - Giacomo Cafaro
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy
| | | | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Aviv University, 5265601, Tel-Hashomer, Israel
- The Mosaic of Autoimmunity Project, Saint Petersburg University, Saint Petersburg, Russia
- Ministry of Health of the Russian Federation, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Roberto Gerli
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy.
| |
Collapse
|
18
|
Perricone C, Bartoloni E, Bursi R, Cafaro G, Guidelli GM, Shoenfeld Y, Gerli R. COVID-19 as part of the hyperferritinemic syndromes: the role of iron depletion therapy. Immunol Res 2020; 68:213-224. [PMID: 32681497 PMCID: PMC7366458 DOI: 10.1007/s12026-020-09145-5] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
SARS-CoV-2 infection is characterized by a protean clinical picture that can range from asymptomatic patients to life-threatening conditions. Severe COVID-19 patients often display a severe pulmonary involvement and develop neutrophilia, lymphopenia, and strikingly elevated levels of IL-6. There is an over-exuberant cytokine release with hyperferritinemia leading to the idea that COVID-19 is part of the hyperferritinemic syndrome spectrum. Indeed, very high levels of ferritin can occur in other diseases including hemophagocytic lymphohistiocytosis, macrophage activation syndrome, adult-onset Still's disease, catastrophic antiphospholipid syndrome and septic shock. Numerous studies have demonstrated the immunomodulatory effects of ferritin and its association with mortality and sustained inflammatory process. High levels of free iron are harmful in tissues, especially through the redox damage that can lead to fibrosis. Iron chelation represents a pillar in the treatment of iron overload. In addition, it was proven to have an anti-viral and anti-fibrotic activity. Herein, we analyse the pathogenic role of ferritin and iron during SARS-CoV-2 infection and propose iron depletion therapy as a novel therapeutic approach in the COVID-19 pandemic.
Collapse
Affiliation(s)
- Carlo Perricone
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy
| | - Elena Bartoloni
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy
| | - Roberto Bursi
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy
| | - Giacomo Cafaro
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy
| | | | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Aviv University, 5265601, Tel-Hashomer, Israel
- The Mosaic of Autoimmunity Project, Saint Petersburg University, Saint Petersburg, Russia
- Ministry of Health of the Russian Federation, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Roberto Gerli
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy.
| |
Collapse
|
19
|
Ruscitti P, Di Benedetto P, Berardicurti O, Panzera N, Grazia N, Lizzi AR, Cipriani P, Shoenfeld Y, Giacomelli R. Pro-inflammatory properties of H-ferritin on human macrophages, ex vivo and in vitro observations. Sci Rep 2020; 10:12232. [PMID: 32699419 PMCID: PMC7376151 DOI: 10.1038/s41598-020-69031-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 07/06/2020] [Indexed: 12/14/2022] Open
Abstract
Ferritin is an iron-binding molecule, which comprises 24 subunits, heavy (FeH) and light (FeL) subunits, suggested to have a pathogenic role by the 'hyperferritinemic syndrome'. In this work, we tested (1) FeH and FeL in bone marrow (BM) and sera in patients with macrophage activation syndrome (MAS); (2) pro-inflammatory effects of ferritin, FeL, and FeH on macrophages; (3) ability of FeH-stimulated macrophages to stimulate the proliferation of peripheral blood mononuclear cells (PBMCs); (4) production of mature IL-1β and IL-12p70 in extracellular compartments of FeH-stimulated macrophages. Immunofluorescence analysis and liquid chromatography mass spectrometry (LC-MS/MS) based proteomics were performed to identify FeL and FeH in BM and sera, respectively, in the same patients. Macrophages were stimulated with ferritin, FeH, and FeL to assess pro-inflammatory effects by RT-PCR and western blot. The proliferation of co-cultured PBMCs with FeH-stimulated macrophages was tested. Immunofluorescence showed an increased FeH expression in BMs, whereas LC-MS/MS identified that FeL was mainly represented in sera. FeH induced a significant increase of gene expressions of IL-1β, IL-6, IL-12, and TNF-α, more marked with FeH, which also stimulated NLRP3. FeH-stimulated macrophages enhanced the proliferation of PBMCs. The ELISA assays showed that mature form of IL-1β and IL-12p70 were increased, in extracellular compartments of FeH-stimulated macrophages. Our results showed FeH in BM biopsies of MAS patients, whereas, LC-MS/MS identified FeL in the sera. FeH showed pro-inflammatory effects on macrophages, stimulated NLRP3, and increased PBMCs proliferation.
Collapse
Affiliation(s)
- Piero Ruscitti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy.
| | - Paola Di Benedetto
- Clinical Pathology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Onorina Berardicurti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| | - Noemi Panzera
- Clinical Pathology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Nicolò Grazia
- Clinical Pathology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Anna Rita Lizzi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Paola Cipriani
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel HaShomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel.,Laboratory of the Mosaics of Autoimmunity, Saint Petersburg State University, Saint Petersburg, Russia
| | - Roberto Giacomelli
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| |
Collapse
|
20
|
Ruscitti P, Berardicurti O, Di Benedetto P, Cipriani P, Iagnocco A, Shoenfeld Y, Giacomelli R. Severe COVID-19, Another Piece in the Puzzle of the Hyperferritinemic Syndrome. An Immunomodulatory Perspective to Alleviate the Storm. Front Immunol 2020; 11:1130. [PMID: 32574264 PMCID: PMC7270352 DOI: 10.3389/fimmu.2020.01130] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/08/2020] [Indexed: 12/11/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19), an acute respiratory disease caused by severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2), has been declared as a worldwide public health emergency. Interestingly, severe COVID-19 is characterized by fever, hyperferritinemia, and a hyper-inflammatory process with a massive release of pro-inflammatory cytokines, which may be responsible for the high rate of mortality. These findings may advocate for a similarity between severe COVID-19 and some challenging rheumatic diseases, such as adult onset Still's disease, secondary hemophagocytic lymphohistiocytosis, and catastrophic anti-phospholipid syndrome, which have been included in the “hyperferritinemic syndrome” category. Furthermore, as performed in these hyper-inflammatory states, severe COVID-19 may benefit from immunomodulatory therapies.
Collapse
Affiliation(s)
- Piero Ruscitti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Onorina Berardicurti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Paola Di Benedetto
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Paola Cipriani
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Annamaria Iagnocco
- Academic Rheumatology Centre, Università degli Studi di Torino, Turin, Italy
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel HaShomer, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel.,Laboratory of the Mosaics of Autoimmunity, Saint Petersburg State University, Saint Petersburg, Russia
| | - Roberto Giacomelli
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
21
|
Giemza-Stokłosa J, Islam MA, Kotyla PJ. Hyperferritinaemia: An Iron Sword of Autoimmunity. Curr Pharm Des 2020; 25:2909-2918. [PMID: 31686632 DOI: 10.2174/1381612825666190709202804] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 06/30/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Ferritin is a molecule that plays many roles being the storage for iron, signalling molecule, and modulator of the immune response. METHODS Different electronic databases were searched in a non-systematic way to find out the literature of interest. RESULTS The level of ferritin rises in many inflammatory conditions including autoimmune disorders. However, in four inflammatory diseases (i.e., adult-onset Still's diseases, macrophage activation syndrome, catastrophic antiphospholipid syndrome, and sepsis), high levels of ferritin are observed suggesting it as a remarkable biomarker and pathological involvement in these diseases. Acting as an acute phase reactant, ferritin is also involved in the cytokine-associated modulator of the immune response as well as a regulator of cytokine synthesis and release which are responsible for the inflammatory storm. CONCLUSION This review article presents updated information on the role of ferritin in inflammatory and autoimmune diseases with an emphasis on hyperferritinaemic syndrome.
Collapse
Affiliation(s)
| | - Md Asiful Islam
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Przemysław J Kotyla
- Department of Internal Medicine, Rheumatology and Clinical Immunology, Faculty in Katowice, Medical University of Silesia, 40-635 Katowice, Poland
| |
Collapse
|
22
|
Ruscitti P, Conforti A, Pavlych V, Giacomelli R. Inhibiting inflammatory cytokines in adult onset Still’s disease. Current trends and new therapeutic perspectives. Expert Opin Orphan Drugs 2019. [DOI: 10.1080/21678707.2019.1701431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Piero Ruscitti
- Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Alessandro Conforti
- Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Viktoriya Pavlych
- Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Roberto Giacomelli
- Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
23
|
Wang MY, Jia JC, Yang CD, Hu QY. Pathogenesis, disease course, and prognosis of adult-onset Still's disease: an update and review. Chin Med J (Engl) 2019; 132:2856-2864. [PMID: 31856058 PMCID: PMC6940076 DOI: 10.1097/cm9.0000000000000538] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Adult-onset Still's disease (AOSD) is a rare but clinically well-known polygenic systemic autoinflammatory disease. In this review, we aim to present frontiers in the pathogenesis, clinical features, diagnosis, biomarkers, disease course, prognosis, and treatment in AOSD. DATA SOURCES We retrieved information from the PubMed database up to July 2019, using various search terms and relevant words, including AOSD and Still's disease. STUDY SELECTION We included data from peer-reviewed journals. Both basic and clinical studies were selected. RESULTS Pathogenesis of AOSD involves genetic background, infectious triggers, and immunopathogenesis, mainly the activation of macrophages and neutrophils followed by a cytokine storm. Diagnosis and prognosis evaluation of AOSD is still challenging; therefore, there is an urgent need to identify better biomarkers. Biologic agents, including interleukin (IL)-1β, IL-6, and tumor necrosis factor-α antagonists in the treatment of AOSD, have good prospect. CONCLUSION This review highlights the advances in pathogenesis, potential biomarkers, disease course, and treatment in AOSD.
Collapse
Affiliation(s)
- Meng-Yan Wang
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | | | | | | |
Collapse
|
24
|
Zeng F, Deng G, Luo H, Zuo X, Xie Y. Macrophage activation syndrome and pulmonary arterial hypertension in a patient with adult-onset Still disease: A case report. Medicine (Baltimore) 2019; 98:e17427. [PMID: 31689747 PMCID: PMC6946370 DOI: 10.1097/md.0000000000017427] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
INTRODUCTION Macrophage activation syndrome (MAS) and pulmonary arterial hypertension (PAH) are rare and life-threatening complications of adult-onset Still disease (AOSD). PATIENT CONCERNS We reported an interesting case of a 25-year-old AOSD patient with MAS and PAH, and the patient was found to have right anomalous pulmonary venous connection accompanied by an atrial septal defect. DIAGNOSIS MAS was diagnosed as a complication of AOSD. PAH was contributed probably by right anomalous pulmonary venous connection. INTERVENTIONS The patient dramatically improved with methylprednisolone (80 mg I.V. daily) plus supportive treatments, without interleukin (IL) inhibitors or ciclosporin A given. OUTCOMES The patient's serum hepatic enzyme levels dropped and hemocytes rose within 1 week. CONCLUSION Other causes need to be excluded carefully before giving a diagnosis of PAH with AOSD. Early diagnosis and aggressive treatments are pivotal to improve the quality of life and the survival of patients.
Collapse
Affiliation(s)
- Furong Zeng
- Department of Rheumatology and Immunology, Xiangya Hospital, The Institution of Rheumatology and Immunology
| | - Guangtong Deng
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hui Luo
- Department of Rheumatology and Immunology, Xiangya Hospital, The Institution of Rheumatology and Immunology
| | - Xiaoxia Zuo
- Department of Rheumatology and Immunology, Xiangya Hospital, The Institution of Rheumatology and Immunology
| | - Yanli Xie
- Department of Rheumatology and Immunology, Xiangya Hospital, The Institution of Rheumatology and Immunology
| |
Collapse
|
25
|
Rao S, Li Q, Wu H, Zhao M, Wang A, Zhang G, Li J, Lu L, Shi W, Lu Q. Juxtaposition of IL-1β and IFN-γ expression and apoptosis of keratinocytes in adult-onset Still's disease. Expert Rev Clin Immunol 2019; 15:1341-1350. [PMID: 31661988 DOI: 10.1080/1744666x.2020.1685876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Backgroud: Recently, atypical persistent skin eruptions (APSEs) have been documented as a new manifestation of adult-onset Still's disease (AOSD), with a unique pathological feature of necrotic keratinocytes in the upper third of the epidermis, but the mechanism has not been elucidated. The aim of this study was to explore the potential mechanism of the unique pathological phenomenon of APSEs.Methods: Clinical and pathological data from 26 AOSD patients with APSEs and 6 with evanescent skin eruptions (ESEs) were reviewed. Fourteen APSE biopsies and 6 ESE biopsies were selected for multi-spectrum immunohistochemistry with 5 disease controls and 5 healthy controls.Results: The unique pathological manifestation was present in all APSE patients but was hardly found in ESE patients. There were more CD4 + T-cells infiltrated in the dermis of APSEs than in the dermis of ESEs. IL-1β and IFN-γ were specifically expressed in the upper third of the epidermis and were juxtaposed to the loci of the necrotic keratinocytes.Conclusion: Our findings showed important cellular and molecular derangements related to the APSE-specific pathological phenomena and helped to understand the pathogenesis of dyskeratosis in the epidermis. The findings could also pave a way to explore an effective intervention to this potentially life-threatening disorder.
Collapse
Affiliation(s)
- Shijia Rao
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, PR China
| | - Qianwen Li
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, PR China
| | - Haijing Wu
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, PR China
| | - Ming Zhao
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, PR China
| | - Alun Wang
- Department of Pathology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Guiying Zhang
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, PR China
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Lixia Lu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Wei Shi
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Qianjin Lu
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, PR China
| |
Collapse
|
26
|
Vitale A, Cavalli G, Colafrancesco S, Priori R, Valesini G, Argolini LM, Baldissera E, Bartoloni E, Cammelli D, Canestrari G, Sota J, Cavallaro E, Massaro MG, Ruscitti P, Cipriani P, De Marchi G, De Vita S, Emmi G, Ferraccioli G, Frassi M, Gerli R, Gremese E, Iannone F, Lapadula G, Lopalco G, Manna R, Mathieu A, Montecucco C, Mosca M, Piazza I, Piga M, Pontikaki I, Romano M, Rossi S, Rossini M, Silvestri E, Stagnaro C, Talarico R, Tincani A, Viapiana O, Vitiello G, Galozzi P, Sfriso P, Gaggiano C, Rigante D, Dagna L, Giacomelli R, Cantarini L. Long-Term Retention Rate of Anakinra in Adult Onset Still's Disease and Predictive Factors for Treatment Response. Front Pharmacol 2019; 10:296. [PMID: 31001115 PMCID: PMC6454864 DOI: 10.3389/fphar.2019.00296] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Accepted: 03/11/2019] [Indexed: 12/13/2022] Open
Abstract
Background: Anakinra (ANA) is an effective treatment choice in patients with adult onset Still’s disease (AOSD). Variables affecting treatment survival include loss of efficacy or adverse events, but also the decision to discontinue treatment after long-term clinical remission. Objectives: Aims of this study were: (i) to assess the drug retention rate (DRR) of ANA during a long-term follow-up looking for any difference related to the line of biologic treatment, the concomitant use of conventional disease modifying anti-rheumatic drugs (cDMARDs) and the different type of AOSD (systemic versus chronic articular); (ii) to identify predictive factors of lack of efficacy, loss of efficacy, and ANA withdrawal owing to long-term remission. Methods: AOSD patients classified according with Yamaguchi criteria and treated with ANA were retrospectively enrolled in 18 Italian tertiary Centers. Demographic, laboratory, clinical and therapeutic data related to the start of ANA (baseline), the 3-month assessment and the last follow-up visit while on ANA treatment were retrospectively collected and statistically analyzed. Results: One hundred and forty-one AOSD patients (48 males, 93 females) treated with ANA for a mean period of 35.96 ± 36.05 months were enrolled. The overall DRR of ANA was 44.6 and 30.5% at the 60- and 120-month assessments, respectively, with no significant differences between: (i) biologic naïve patients and those previously treated with other biologics (log-rank p = 0.97); (ii) monotherapy and concomitant use of cDMARDs (log-rank p = 0.45); (iii) systemic and chronic articular types of AOSD (log-rank p = 0.67). No variables collected at baseline could predict primary inefficacy, while the number of swollen joints at baseline was significantly associated with secondary inefficacy (p = 0.01, OR = 1.194, C.I. 1.043–1.367). The typical AOSD skin rash was negatively related with ANA withdrawal owing to long-term remission (p = 0.03, OR = 0.224, C.I. 0.058–0.863). Conclusion: Long-term DRR of ANA has been found excellent and is not affected by different lines of biologic treatment, concomitant use of cDMARDs, or type of AOSD. The risk of losing ANA efficacy increases along with the number of swollen joints at the start of therapy, while the typical skin rash is a negative predictor of ANA withdrawal related to sustained remission.
Collapse
Affiliation(s)
- Antonio Vitale
- Research Centre of Systemic Autoinflammatory Diseases, Behçet's Disease Clinic and Rheumatology-Ophthalmology Collaborative Uveitis Centre, Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Giulio Cavalli
- Vita-Salute San Raffaele University, Milan, Italy.,Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Serena Colafrancesco
- Rheumatology Unit, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Roberta Priori
- Rheumatology Unit, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Guido Valesini
- Rheumatology Unit, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | | | - Elena Baldissera
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elena Bartoloni
- Rheumatology Unit, Department of Medicine, University of Perugia, Perugia, Italy
| | - Daniele Cammelli
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
| | - Giovanni Canestrari
- Institute of Rheumatology and Affine Sciences, Division of Rheumatology, Catholic University of the Sacred Heart, Rome, Italy
| | - Jurgen Sota
- Research Centre of Systemic Autoinflammatory Diseases, Behçet's Disease Clinic and Rheumatology-Ophthalmology Collaborative Uveitis Centre, Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Elena Cavallaro
- Department of Medical and Biological Sciences, Rheumatology Clinic, University of Udine, Udine, Italy
| | - Maria Grazia Massaro
- Periodic Fever Research Center, Institute of Internal Medicine, Catholic University of the Sacred Heart, Fondazione Policlinico A. Gemelli, Rome, Italy
| | - Piero Ruscitti
- Department of Biotechnological and Applied Clinical Science, Division of Rheumatology, University of L'Aquila, L'Aquila, Italy
| | - Paola Cipriani
- Department of Biotechnological and Applied Clinical Science, Division of Rheumatology, University of L'Aquila, L'Aquila, Italy
| | - Ginevra De Marchi
- Department of Medical and Biological Sciences, Rheumatology Clinic, University of Udine, Udine, Italy
| | - Salvatore De Vita
- Department of Medical and Biological Sciences, Rheumatology Clinic, University of Udine, Udine, Italy
| | - Giacomo Emmi
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
| | - Gianfranco Ferraccioli
- Institute of Rheumatology and Affine Sciences, Division of Rheumatology, Catholic University of the Sacred Heart, Rome, Italy
| | - Micol Frassi
- Rheumatology and Clinical Immunology, Spedali Civili and Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Roberto Gerli
- Rheumatology Unit, Department of Medicine, University of Perugia, Perugia, Italy
| | - Elisa Gremese
- Institute of Rheumatology and Affine Sciences, Division of Rheumatology, Catholic University of the Sacred Heart, Rome, Italy
| | - Florenzo Iannone
- Rheumatology Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Giovanni Lapadula
- Rheumatology Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Giuseppe Lopalco
- Rheumatology Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Raffaele Manna
- Periodic Fever Research Center, Institute of Internal Medicine, Catholic University of the Sacred Heart, Fondazione Policlinico A. Gemelli, Rome, Italy
| | - Alessandro Mathieu
- Rheumatology Unit, Department of Medical Sciences, University and AOU of Cagliari, Cagliari, Italy
| | - Carlomaurizio Montecucco
- Department of Rheumatology, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
| | - Marta Mosca
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Ilaria Piazza
- Rheumatology Unit, Department of Medicine, University of Verona, Verona, Italy
| | - Matteo Piga
- Rheumatology Unit, Department of Medical Sciences, University and AOU of Cagliari, Cagliari, Italy
| | | | - Micol Romano
- Division of Rheumatology, ASST Gaetano Pini, Milan, Italy
| | - Silvia Rossi
- Department of Rheumatology, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
| | - Maurizio Rossini
- Rheumatology Unit, Department of Medicine, University of Verona, Verona, Italy
| | - Elena Silvestri
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
| | - Chiara Stagnaro
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Rosaria Talarico
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Angela Tincani
- Rheumatology and Clinical Immunology, Spedali Civili and Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Ombretta Viapiana
- Rheumatology Unit, Department of Medicine, University of Verona, Verona, Italy
| | - Gianfranco Vitiello
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
| | - Paola Galozzi
- Department of Medicine DIMED, Rheumatology Unit, University of Padua, Padua, Italy
| | - Paolo Sfriso
- Department of Medicine DIMED, Rheumatology Unit, University of Padua, Padua, Italy
| | - Carla Gaggiano
- Clinical Pediatrics, Department of Molecular Medicine and Development, University of Siena, Siena, Italy
| | - Donato Rigante
- Institute of Pediatrics, Università Cattolica Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli I.R.C.C.S., Rome, Italy
| | - Lorenzo Dagna
- Vita-Salute San Raffaele University, Milan, Italy.,Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Roberto Giacomelli
- Department of Biotechnological and Applied Clinical Science, Division of Rheumatology, University of L'Aquila, L'Aquila, Italy
| | - Luca Cantarini
- Research Centre of Systemic Autoinflammatory Diseases, Behçet's Disease Clinic and Rheumatology-Ophthalmology Collaborative Uveitis Centre, Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| |
Collapse
|
27
|
Ruscitti P, Cipriani P, Liakouli V, Guggino G, Carubbi F, Berardicurti O, Ciccia F, Giacomelli R. Managing Adult-onset Still's disease: The effectiveness of high-dosage of corticosteroids as first-line treatment in inducing the clinical remission. Results from an observational study. Medicine (Baltimore) 2019; 98:e15123. [PMID: 30985672 PMCID: PMC6485786 DOI: 10.1097/md.0000000000015123] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
To assess the effectiveness of the treatment with high dosage of corticosteroids (CCSs), as first-line therapy, in inducing remission in naïve Adult-onset Still's disease (AOSD) patients compared with low dosage of CCSs, after 6 months. To further evaluate the rate of patients maintaining the remission and the rate of CCSs discontinuation, after additional 12 months of follow-up.A retrospective evaluation of patients prospectively followed was designed to compare the rate of clinical remission in naïve AOSD patients treated with high dosages of CCSs (0.8-1 mg/kg/day of prednisone-equivalent) or low dosage of CCSs (0.2-0.3 mg/kg/day of prednisone-equivalent), after 6 months. An additional analysis was performed to compare the rate of monocyclic pattern between these groups, after further 12 months of follow-up.The clinical remission was achieved in a higher percentage of patients treated with the first-line treatment with high dosage of CCSs than treated the first-line treatment with low dosage of CCSs. At the end of 18 months of follow-up, a larger percentage of patients treated the first-line treatment with high dosage of CCSs was classified as monocyclic pattern and discontinued CCSs when compared with patients treated the first-line treatment with low dosage of CCSs. Patients defined as CCSs non-responder were treated with methotrexate (MTX)+CCSs or with combination therapy CCSs+MTX+biologic drug. The clinical remission was observed in a percentage of these patients.We showed the effectiveness of the first-line treatment with high dosage of CCSs in inducing clinical remission in naïve AOSD patients when compared with the first-line treatment with low dosage of CCSs. The first-line treatment with high dosage of CCSs was also associated with the achievement of monocyclic pattern and CCSs discontinuation, after 18 months of follow-up.
Collapse
Affiliation(s)
- Piero Ruscitti
- Rheumatology, Department of Biotechnological and Applied Clinical Science, University of L’Aquila, L’Aquila
| | - Paola Cipriani
- Rheumatology, Department of Biotechnological and Applied Clinical Science, University of L’Aquila, L’Aquila
| | - Vasiliki Liakouli
- Rheumatology, Department of Biotechnological and Applied Clinical Science, University of L’Aquila, L’Aquila
| | - Giuliana Guggino
- Department of Clinical and Experimental Medicine, Rheumatology Section, University of Campania “Luigi Vanvitelli,” Naples, Italy
| | - Francesco Carubbi
- Rheumatology, Department of Biotechnological and Applied Clinical Science, University of L’Aquila, L’Aquila
| | - Onorina Berardicurti
- Rheumatology, Department of Biotechnological and Applied Clinical Science, University of L’Aquila, L’Aquila
| | - Francesco Ciccia
- Department of Clinical and Experimental Medicine, Rheumatology Section, University of Campania “Luigi Vanvitelli,” Naples, Italy
| | - Roberto Giacomelli
- Rheumatology, Department of Biotechnological and Applied Clinical Science, University of L’Aquila, L’Aquila
| |
Collapse
|
28
|
Ruscitti P, Giacomelli R. Pathogenesis of adult onset still’s disease: current understanding and new insights. Expert Rev Clin Immunol 2018; 14:965-976. [DOI: 10.1080/1744666x.2018.1533403] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Piero Ruscitti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Roberto Giacomelli
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
29
|
Giacomelli R, Ruscitti P, Shoenfeld Y. A comprehensive review on adult onset Still's disease. J Autoimmun 2018; 93:24-36. [PMID: 30077425 DOI: 10.1016/j.jaut.2018.07.018] [Citation(s) in RCA: 245] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 07/26/2018] [Accepted: 07/26/2018] [Indexed: 02/06/2023]
Abstract
Adult-onset Still's disease (AOSD) is a systemic inflammatory disorder of unknown etiology usually affecting young adults; spiking fever, arthritis and evanescent rash are commonly observed during the disease. Other frequently observed clinical features include sore throat, hepatomegaly, splenomegaly, lymphadenopathy and serositis. Furthermore, AOSD patients may experience different life-threating complications. Macrophage activation syndrome (MAS) has been reported up to 15% of AOSD patients and it is considered to be the most severe complication of the disease being characterised by high mortality rate. During AOSD, laboratory tests reflect the systemic inflammatory process showing high levels of erythrocyte sedimentation rate and C-reactive protein. In addition, the ferritin levels are typically higher than those observed in other autoimmune, inflammatory, infectious, or neoplastic diseases. Analysing AOSD disease course, 3 different clinical patterns of AOSD have been identified: i. monocyclic pattern, characterised by a systemic single episode; ii. polycyclic pattern, characterised by multiple, ≤ 1 year lasting, flares, alternating with remissions; iii. chronic pattern, related to a persistently active disease with associated polyarthritis. At present, AOSD therapeutic strategy is aimed at targeting pro-inflammatory signs and symptoms, preventing organ damage and life-threating complications and minimising adverse effects of treatment. However, the treatment of AOSD remains largely empirical, lacking controlled clinical trials. High dosages of corticosteroids are usually the first line therapy when the systemic symptoms predominate. Despite this treatment, a large percentage of patients experiences several flares with an evolution toward the chronic disease course and up to 16% of patients die during the follow up, due to AOSD-related complications. On these bases, in the last years, biological agents have been successfully used in refractory cases. Finally, multiple recent lines of evidence have suggested new insights in AOSD pathogenesis unmasking further therapeutic targets. In fact, small molecules, used in experimental MAS models, might represent new therapeutic options.
Collapse
Affiliation(s)
- Roberto Giacomelli
- Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Piero Ruscitti
- Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
30
|
Enfermedad de Still del adulto. Med Clin (Barc) 2018; 150:348-353. [DOI: 10.1016/j.medcli.2017.10.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/24/2017] [Accepted: 10/25/2017] [Indexed: 11/17/2022]
|
31
|
Ruscitti P, Iacono D, Ciccia F, Emmi G, Cipriani P, Grembiale RD, Perosa F, Emmi L, Triolo G, Giacomelli R, Valentini G. Macrophage Activation Syndrome in Patients Affected by Adult-onset Still Disease: Analysis of Survival Rates and Predictive Factors in the Gruppo Italiano di Ricerca in Reumatologia Clinica e Sperimentale Cohort. J Rheumatol 2018; 45:864-872. [PMID: 29657144 DOI: 10.3899/jrheum.170955] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2018] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Macrophage activation syndrome (MAS) is a reactive form of hemophagocytic lymphohistiocytosis, which can complicate adult-onset Still disease (AOSD). We investigated AOSD clinical features at the time of diagnosis, to assess predictors of MAS occurrence. Further, we analyzed the outcomes of patients with AOSD who experience MAS. METHODS Patients with AOSD admitted to any Gruppo Italiano di Ricerca in Reumatologia Clinica e Sperimentale center were retrospectively analyzed for features typical of AOSD, MAS occurrence, and their survival rate. RESULTS Of 119 patients with AOSD, 17 experienced MAS (12 at admission and 5 during followup). Twelve patients with MAS at first admission differed from the remaining 107 in prevalence of lymphadenopathy and liver involvement at the time of diagnosis. In addition, serum ferritin levels and systemic score values were significantly higher in the patients presenting with MAS. At the time of diagnosis, the 5 patients who developed MAS differed from the remaining 102 in the prevalence of abdominal pain, and they showed increased systemic score values. In the multivariate analysis, lymphadenopathy (OR 7.22, 95% CI 1.49-34.97, p = 0.014) and abdominal pain (OR 4.36, 95% CI 1.24-15.39, p = 0.022) were predictive of MAS occurrence. Finally, MAS occurrence significantly reduced the survival rate of patients with AOSD (p < 0.0001). CONCLUSION MAS occurrence significantly reduced the survival rate in patients with AOSD. Patients with MAS at baseline presented an increased prevalence of lymphadenopathy and liver involvement, as well as high serum ferritin levels and systemic score values. The presence of lymphadenopathy and abdominal pain was associated with MAS occurrence.
Collapse
Affiliation(s)
- Piero Ruscitti
- From the Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila; Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples, Naples; Rheumatology Section, Department of Internal Medicine, University of Palermo, Palermo; Department of Experimental and Clinical Medicine, University of Florence, Florence; Department of Health Sciences, University of Catanzaro "Magna Graecia," Catanzaro; Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy. .,P. Ruscitti, MD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; D. Iacono, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples; F. Ciccia, MD, PhD, Rheumatology Section, Department of Internal Medicine, University of Palermo; G. Emmi, MD, PhD, Department of Experimental and Clinical Medicine, University of Florence; P. Cipriani, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; R.D. Grembiale, MD, Department of Health Sciences, University of Catanzaro "Magna Graecia"; F. Perosa, MD, PhD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; L. Emmi, MD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; G. Triolo, MD, Rheumatology Section, Department of Internal Medicine, University of Palermo; R. Giacomelli, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; G. Valentini, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples.
| | - Daniela Iacono
- From the Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila; Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples, Naples; Rheumatology Section, Department of Internal Medicine, University of Palermo, Palermo; Department of Experimental and Clinical Medicine, University of Florence, Florence; Department of Health Sciences, University of Catanzaro "Magna Graecia," Catanzaro; Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy.,P. Ruscitti, MD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; D. Iacono, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples; F. Ciccia, MD, PhD, Rheumatology Section, Department of Internal Medicine, University of Palermo; G. Emmi, MD, PhD, Department of Experimental and Clinical Medicine, University of Florence; P. Cipriani, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; R.D. Grembiale, MD, Department of Health Sciences, University of Catanzaro "Magna Graecia"; F. Perosa, MD, PhD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; L. Emmi, MD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; G. Triolo, MD, Rheumatology Section, Department of Internal Medicine, University of Palermo; R. Giacomelli, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; G. Valentini, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples
| | - Francesco Ciccia
- From the Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila; Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples, Naples; Rheumatology Section, Department of Internal Medicine, University of Palermo, Palermo; Department of Experimental and Clinical Medicine, University of Florence, Florence; Department of Health Sciences, University of Catanzaro "Magna Graecia," Catanzaro; Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy.,P. Ruscitti, MD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; D. Iacono, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples; F. Ciccia, MD, PhD, Rheumatology Section, Department of Internal Medicine, University of Palermo; G. Emmi, MD, PhD, Department of Experimental and Clinical Medicine, University of Florence; P. Cipriani, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; R.D. Grembiale, MD, Department of Health Sciences, University of Catanzaro "Magna Graecia"; F. Perosa, MD, PhD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; L. Emmi, MD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; G. Triolo, MD, Rheumatology Section, Department of Internal Medicine, University of Palermo; R. Giacomelli, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; G. Valentini, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples
| | - Giacomo Emmi
- From the Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila; Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples, Naples; Rheumatology Section, Department of Internal Medicine, University of Palermo, Palermo; Department of Experimental and Clinical Medicine, University of Florence, Florence; Department of Health Sciences, University of Catanzaro "Magna Graecia," Catanzaro; Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy.,P. Ruscitti, MD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; D. Iacono, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples; F. Ciccia, MD, PhD, Rheumatology Section, Department of Internal Medicine, University of Palermo; G. Emmi, MD, PhD, Department of Experimental and Clinical Medicine, University of Florence; P. Cipriani, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; R.D. Grembiale, MD, Department of Health Sciences, University of Catanzaro "Magna Graecia"; F. Perosa, MD, PhD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; L. Emmi, MD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; G. Triolo, MD, Rheumatology Section, Department of Internal Medicine, University of Palermo; R. Giacomelli, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; G. Valentini, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples
| | - Paola Cipriani
- From the Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila; Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples, Naples; Rheumatology Section, Department of Internal Medicine, University of Palermo, Palermo; Department of Experimental and Clinical Medicine, University of Florence, Florence; Department of Health Sciences, University of Catanzaro "Magna Graecia," Catanzaro; Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy.,P. Ruscitti, MD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; D. Iacono, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples; F. Ciccia, MD, PhD, Rheumatology Section, Department of Internal Medicine, University of Palermo; G. Emmi, MD, PhD, Department of Experimental and Clinical Medicine, University of Florence; P. Cipriani, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; R.D. Grembiale, MD, Department of Health Sciences, University of Catanzaro "Magna Graecia"; F. Perosa, MD, PhD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; L. Emmi, MD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; G. Triolo, MD, Rheumatology Section, Department of Internal Medicine, University of Palermo; R. Giacomelli, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; G. Valentini, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples
| | - Rosa Daniela Grembiale
- From the Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila; Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples, Naples; Rheumatology Section, Department of Internal Medicine, University of Palermo, Palermo; Department of Experimental and Clinical Medicine, University of Florence, Florence; Department of Health Sciences, University of Catanzaro "Magna Graecia," Catanzaro; Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy.,P. Ruscitti, MD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; D. Iacono, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples; F. Ciccia, MD, PhD, Rheumatology Section, Department of Internal Medicine, University of Palermo; G. Emmi, MD, PhD, Department of Experimental and Clinical Medicine, University of Florence; P. Cipriani, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; R.D. Grembiale, MD, Department of Health Sciences, University of Catanzaro "Magna Graecia"; F. Perosa, MD, PhD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; L. Emmi, MD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; G. Triolo, MD, Rheumatology Section, Department of Internal Medicine, University of Palermo; R. Giacomelli, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; G. Valentini, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples
| | - Federico Perosa
- From the Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila; Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples, Naples; Rheumatology Section, Department of Internal Medicine, University of Palermo, Palermo; Department of Experimental and Clinical Medicine, University of Florence, Florence; Department of Health Sciences, University of Catanzaro "Magna Graecia," Catanzaro; Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy.,P. Ruscitti, MD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; D. Iacono, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples; F. Ciccia, MD, PhD, Rheumatology Section, Department of Internal Medicine, University of Palermo; G. Emmi, MD, PhD, Department of Experimental and Clinical Medicine, University of Florence; P. Cipriani, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; R.D. Grembiale, MD, Department of Health Sciences, University of Catanzaro "Magna Graecia"; F. Perosa, MD, PhD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; L. Emmi, MD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; G. Triolo, MD, Rheumatology Section, Department of Internal Medicine, University of Palermo; R. Giacomelli, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; G. Valentini, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples
| | - Lorenzo Emmi
- From the Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila; Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples, Naples; Rheumatology Section, Department of Internal Medicine, University of Palermo, Palermo; Department of Experimental and Clinical Medicine, University of Florence, Florence; Department of Health Sciences, University of Catanzaro "Magna Graecia," Catanzaro; Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy.,P. Ruscitti, MD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; D. Iacono, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples; F. Ciccia, MD, PhD, Rheumatology Section, Department of Internal Medicine, University of Palermo; G. Emmi, MD, PhD, Department of Experimental and Clinical Medicine, University of Florence; P. Cipriani, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; R.D. Grembiale, MD, Department of Health Sciences, University of Catanzaro "Magna Graecia"; F. Perosa, MD, PhD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; L. Emmi, MD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; G. Triolo, MD, Rheumatology Section, Department of Internal Medicine, University of Palermo; R. Giacomelli, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; G. Valentini, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples
| | - Giovanni Triolo
- From the Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila; Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples, Naples; Rheumatology Section, Department of Internal Medicine, University of Palermo, Palermo; Department of Experimental and Clinical Medicine, University of Florence, Florence; Department of Health Sciences, University of Catanzaro "Magna Graecia," Catanzaro; Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy.,P. Ruscitti, MD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; D. Iacono, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples; F. Ciccia, MD, PhD, Rheumatology Section, Department of Internal Medicine, University of Palermo; G. Emmi, MD, PhD, Department of Experimental and Clinical Medicine, University of Florence; P. Cipriani, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; R.D. Grembiale, MD, Department of Health Sciences, University of Catanzaro "Magna Graecia"; F. Perosa, MD, PhD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; L. Emmi, MD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; G. Triolo, MD, Rheumatology Section, Department of Internal Medicine, University of Palermo; R. Giacomelli, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; G. Valentini, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples
| | - Roberto Giacomelli
- From the Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila; Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples, Naples; Rheumatology Section, Department of Internal Medicine, University of Palermo, Palermo; Department of Experimental and Clinical Medicine, University of Florence, Florence; Department of Health Sciences, University of Catanzaro "Magna Graecia," Catanzaro; Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy.,P. Ruscitti, MD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; D. Iacono, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples; F. Ciccia, MD, PhD, Rheumatology Section, Department of Internal Medicine, University of Palermo; G. Emmi, MD, PhD, Department of Experimental and Clinical Medicine, University of Florence; P. Cipriani, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; R.D. Grembiale, MD, Department of Health Sciences, University of Catanzaro "Magna Graecia"; F. Perosa, MD, PhD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; L. Emmi, MD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; G. Triolo, MD, Rheumatology Section, Department of Internal Medicine, University of Palermo; R. Giacomelli, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; G. Valentini, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples
| | - Gabriele Valentini
- From the Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila; Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples, Naples; Rheumatology Section, Department of Internal Medicine, University of Palermo, Palermo; Department of Experimental and Clinical Medicine, University of Florence, Florence; Department of Health Sciences, University of Catanzaro "Magna Graecia," Catanzaro; Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy.,P. Ruscitti, MD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; D. Iacono, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples; F. Ciccia, MD, PhD, Rheumatology Section, Department of Internal Medicine, University of Palermo; G. Emmi, MD, PhD, Department of Experimental and Clinical Medicine, University of Florence; P. Cipriani, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; R.D. Grembiale, MD, Department of Health Sciences, University of Catanzaro "Magna Graecia"; F. Perosa, MD, PhD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; L. Emmi, MD, Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School; G. Triolo, MD, Rheumatology Section, Department of Internal Medicine, University of Palermo; R. Giacomelli, MD, PhD, Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila; G. Valentini, MD, Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples
| |
Collapse
|
32
|
Ruscitti P, Cipriani P, Di Benedetto P, Liakouli V, Berardicurti O, Carubbi F, Ciccia F, Guggino G, Triolo G, Giacomelli R. H-ferritin and proinflammatory cytokines are increased in the bone marrow of patients affected by macrophage activation syndrome. Clin Exp Immunol 2017; 191:220-228. [PMID: 28960260 DOI: 10.1111/cei.13057] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2017] [Indexed: 12/12/2022] Open
Abstract
Macrophage activation syndrome (MAS) is hyperinflammatory life-threatening syndrome, associated typically with high levels of serum ferritin. This is an iron storage protein including heavy (H) and light (L) subunits, categorized on their molecular weight. The H-/L subunits ratio may be different in tissues, depending on the specific tissue and pathophysiological status. In this study, we analysed the bone marrow (BM) biopsies of adult MAS patients to assess the presence of: (i) H-ferritin and L-ferritin; (ii) CD68+ /H-ferritin+ and CD68+ /L-ferritin+ ; and (iii) interleukin (IL)-1β, tumour necrosis factor (TNF) and interferon (IFN)-γ. We also explored possible correlations of these results with clinical data. H-ferritin, IL-1β, TNF and IFN-γ were increased significantly in MAS. Furthermore, an increased number of CD68+ /H-ferritin+ cells and an infiltrate of cells co-expressing H-ferritin and IL-12, suggesting an infiltrate of M1 macrophages, were observed. H-ferritin levels and CD68+ /H-ferritin+ cells were correlated with haematological involvement of the disease, serum ferritin and C-reactive protein. L-ferritin and CD68+ /L-ferritin+ cells did not correlate with these parameters. In conclusion, during MAS, H-ferritin, CD68+ /H-ferritin+ cells and proinflammatory cytokines were increased significantly in the BM inflammatory infiltrate, pointing out a possible vicious pathogenic loop. To date, H-ferritin and CD68+ /H-ferritin+ were associated significantly with haematological involvement of the disease, suggesting biomarkers assessing severity of clinical picture.
Collapse
Affiliation(s)
- P Ruscitti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - P Cipriani
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - P Di Benedetto
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - V Liakouli
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - O Berardicurti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - F Carubbi
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - F Ciccia
- Division of Rheumatology, Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - G Guggino
- Division of Rheumatology, Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - G Triolo
- Division of Rheumatology, Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - R Giacomelli
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
33
|
Ruscitti P, Ursini F, Cipriani P, De Sarro G, Giacomelli R. Biologic drugs in adult onset Still's disease: a systematic review and meta-analysis of observational studies. Expert Rev Clin Immunol 2017; 13:1089-1097. [PMID: 28870100 DOI: 10.1080/1744666x.2017.1375853] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Biological drugs, mainly interleukin (IL)-1 and IL-6 antagonists, but also tumor necrosis factor (TNF) inhibitors, have been used in the treatment of adult onset Still's disease patients (AOSD). METHODS We summarised the available evidence for the effectiveness of biologic drugs in AOSD. A systematic review of the literature was performed in order to identify all the available data concerning the effectiveness of biologic drugs in AOSD. The proportion of patients achieving complete remission or any clinical response was calculated. The meta-analysis was thus performed using a random-effects model accounting for the expected high level of heterogeneity. RESULTS Nineteen observational published studies were included in the meta-analysis. The pooled analysis under a random-effects model showed an overall rate of clinical response of 0.85 (95% CI: 0.77-0.91, p < 0.0001) and an overall rate of complete remission of 0.66 (95% CI: 0.54-0.77, p = 0.01). The heterogeneity across studies was high (Q = 59.82 with df = 19.0, p < 0.0001, I2 = 68.23%). CONCLUSIONS Our meta-analysis suggests that AOSD patients may experience a clinical response and/or a complete remission when treated with biologic drugs. Specifically designed and powered studies are needed to fully investigate the role of such medications in the management of AOSD patients.
Collapse
Affiliation(s)
- Piero Ruscitti
- a Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences , University of L'Aquila , L'Aquila , Italy
| | - Francesco Ursini
- b Department of Health Sciences , University of Catanzaro "Magna Graecia" , Catanzaro , Italy
| | - Paola Cipriani
- a Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences , University of L'Aquila , L'Aquila , Italy
| | - Giovambattista De Sarro
- b Department of Health Sciences , University of Catanzaro "Magna Graecia" , Catanzaro , Italy
| | - Roberto Giacomelli
- a Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences , University of L'Aquila , L'Aquila , Italy
| |
Collapse
|
34
|
Ruscitti P, Rago C, Breda L, Cipriani P, Liakouli V, Berardicurti O, Carubbi F, Di Battista C, Verrotti A, Giacomelli R. Macrophage activation syndrome in Still’s disease: analysis of clinical characteristics and survival in paediatric and adult patients. Clin Rheumatol 2017; 36:2839-2845. [DOI: 10.1007/s10067-017-3830-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/28/2017] [Accepted: 09/05/2017] [Indexed: 01/10/2023]
|
35
|
Ruscitti P, Cipriani P, Di Benedetto P, Liakouli V, Carubbi F, Berardicurti O, Ciccia F, Guggino G, Triolo G, Giacomelli R. Advances in immunopathogenesis of macrophage activation syndrome during rheumatic inflammatory diseases: toward new therapeutic targets? Expert Rev Clin Immunol 2017; 13:1041-1047. [PMID: 28837367 DOI: 10.1080/1744666x.2017.1372194] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Macrophage activation syndrome (MAS) is a severe, hyperinflammatory life-threatening syndrome, generally complicating different rheumatic diseases. Despite the severity of the disease, little is known about the pathogenic mechanisms and, thus, possible targeted therapies in the management of these patients. Areas covered: In this review, we aimed to update the current pathogenic knowledge of MAS, during rheumatic diseases, focusing mainly on immunologic abnormalities and on new possible therapeutic strategies. Expert commentary: The difficult pathogenic scenario of MAS, in which genetic defects, predisposing diseases, and triggers are mixed together with the high mortality rate, make it difficult to manage these patients. Although most efforts have been focused on investigating the disease in children, in recent years, several studies are trying to elucidate the possible pathogenic mechanism in adult MAS patients. In this context, genetic and immunological studies might lead to advances in the knowledge of pathogenic mechanisms and possible new therapeutic targets. In the future, the results of ongoing clinical trials are awaited in order to improve the management and, thus, the survival of these patients.
Collapse
Affiliation(s)
- Piero Ruscitti
- a Division of Rheumatology , University of L'Aquila , L'Aquila , Italy
| | - Paola Cipriani
- a Division of Rheumatology , University of L'Aquila , L'Aquila , Italy
| | | | - Vasiliky Liakouli
- a Division of Rheumatology , University of L'Aquila , L'Aquila , Italy
| | - Francesco Carubbi
- a Division of Rheumatology , University of L'Aquila , L'Aquila , Italy
| | | | - Francesco Ciccia
- b Division of Rheumatology , University of Palermo , Palermo , Italy
| | - Giuliana Guggino
- b Division of Rheumatology , University of Palermo , Palermo , Italy
| | - Giovanni Triolo
- b Division of Rheumatology , University of Palermo , Palermo , Italy
| | | |
Collapse
|
36
|
Narváez Garcia FJ, Pascual M, López de Recalde M, Juarez P, Morales-Ivorra I, Notario J, Jucglà A, Nolla JM. Adult-onset Still's disease with atypical cutaneous manifestations. Medicine (Baltimore) 2017; 96:e6318. [PMID: 28296747 PMCID: PMC5369902 DOI: 10.1097/md.0000000000006318] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The diagnosis of adult-onset Still's disease (AOSD) can be very difficult. There are no specific tests available, and diagnosis is usually based on a symptom complex and the well-described typical evanescent rash seen in the majority of patients. However, in recent years, other atypical cutaneous manifestations of AOSD have been reported. These atypical skin eruptions often present in addition to the typical evanescent rash but may also be the only skin manifestation, resulting in delayed diagnosis because of under-recognition.In this study, we present 3 new cases of AOSD with atypical cutaneous manifestations diagnosed during a 30-year period in our department and review 78 additional cases previously reported (PubMed 1990-2016). These 81 patients form the basis of the present analysis.The overall prevalence of atypical cutaneous manifestations in our AOSD population was 14%. These manifestations may appear at any time over the course of the disease, and usually occur in patients who have persistent and severe disease, with a considerable frequency of clinical complications (23%), including serositis, myopericarditis, lung involvement, abdominal pain, neurologic involvement, and reactive hemophagocytic syndrome.The most representative and frequent lesion among the nonclassical skin rashes is the development of persistent pruritic papules and/or plaques. Interestingly, these lesions show a distinctive histological pattern. Other, less frequently observed lesions include urticaria and urticaria-like eruptions, generalized or widespread non-pruritic persistent erythema, vesiculopustular eruptions, a widespread peau d'orange appearance of the skin, and edema of the eyelids mimicking dermatomyositis without any accompanying skin lesion.The great majority of these patients required medium or high doses of glucocorticoids (including intravenous methylprednisolone pulse therapy in some cases) and, in nearly 40%, a more potent or maintenance immunotherapy with immunosuppressant drugs and/or biologic agents (mainly anakinra or tocilizumab) to control or manage symptoms because of a polycyclic or chronic course. The development of atypical cutaneous manifestations seems to be associated with a potentially worse prognosis, with a mortality rate reaching 8% primarily because of infectious complications related to immunosuppressive therapy.In conclusion, the appearance of atypical cutaneous manifestations is not uncommon in AOSD. Recognition of this clinical variant is crucial for the early diagnosis of AOSD, as it might imply persistent disease activity and the need for more aggressive treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Jaime Notario
- Department of Dermatology, Hospital Universitario de Bellvitge-IDIBELL, Barcelona, Spain
| | - Anna Jucglà
- Department of Dermatology, Hospital Universitario de Bellvitge-IDIBELL, Barcelona, Spain
| | | |
Collapse
|
37
|
Ruscitti P, Cipriani P, Masedu F, Iacono D, Ciccia F, Liakouli V, Guggino G, Carubbi F, Berardicurti O, Di Benedetto P, Valenti M, Triolo G, Valentini G, Giacomelli R. Adult-onset Still's disease: evaluation of prognostic tools and validation of the systemic score by analysis of 100 cases from three centers. BMC Med 2016; 14:194. [PMID: 27903264 PMCID: PMC5131497 DOI: 10.1186/s12916-016-0738-8] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 11/03/2016] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Adult-onset Still's disease (AOSD) is rare inflammatory disease of unknown etiology that usually affects young adults. The more common clinical manifestations are spiking fevers, arthritis, evanescent rash, elevated liver enzymes, lymphadenopathy, hepatosplenomegaly, and serositis. The multi-visceral involvement of the disease and the different complications, such as macrophage activation syndrome, may strongly decrease the life expectancy of AOSD patients. METHODS This study aimed to identify the positive and negative features correlated with the outcome of patients. A retrospective analysis of AOSD patients prospectively admitted to three rheumatologic centers was performed to identify the clinical features present at the time of diagnosis and to predict the possible outcome. Furthermore, we investigated the as yet to be validated prognostic value of the systemic score previously proposed. RESULTS One hundred consecutive AOSD patients were enrolled. The mean systemic score showed that the majority of patients had a multi-organ involvement. Sixteen patients showed different complications, mainly the macrophage activation syndrome. A strong increase of inflammatory markers was observed. All patients received steroids at different dosages, 55 patients in association with immunosuppressive drugs and 32 in association with biologic agents. Sixteen patients died during the follow-up. Regression analysis showed that the higher values of the systemic score and the presence of AOSD-related complications, assessed at the time of diagnosis, were significantly correlated with patient mortality. A prognostic impact of the systemic score of ≥ 7.0 was reported. CONCLUSIONS Our study showed that a higher systemic score and the presence of AOSD-related complications at the time of diagnosis were significantly associated with mortality. Of note, a cut-off at 7.0 of the systemic score showed a strong prognostic impact in identifying patients at risk of AOSD-related death.
Collapse
Affiliation(s)
- Piero Ruscitti
- Rheumatology Section, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy.
| | - Paola Cipriani
- Rheumatology Section, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| | - Francesco Masedu
- Medical Statistic Section, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, L'Aquila, Italy
| | - Daniela Iacono
- Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples, Naples, Italy
| | - Francesco Ciccia
- Rheumatology Section, Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - Vasiliki Liakouli
- Rheumatology Section, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| | - Giuliana Guggino
- Rheumatology Section, Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - Francesco Carubbi
- Rheumatology Section, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| | - Onorina Berardicurti
- Rheumatology Section, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| | - Paola Di Benedetto
- Rheumatology Section, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| | - Marco Valenti
- Medical Statistic Section, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, L'Aquila, Italy
| | - Giovanni Triolo
- Rheumatology Section, Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - Gabriele Valentini
- Rheumatology Section, Department of Clinical and Experimental Medicine, Second University of Naples, Naples, Italy
| | - Roberto Giacomelli
- Rheumatology Section, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| |
Collapse
|
38
|
Ruscitti P, Cipriani P, Ciccia F, Masedu F, Liakouli V, Carubbi F, Berardicurti O, Guggino G, Di Benedetto P, Di Bartolomeo S, Valenti M, Triolo G, Giacomelli R. Prognostic factors of macrophage activation syndrome, at the time of diagnosis, in adult patients affected by autoimmune disease: Analysis of 41 cases collected in 2 rheumatologic centers. Autoimmun Rev 2016; 16:16-21. [PMID: 27664384 DOI: 10.1016/j.autrev.2016.09.016] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 07/29/2016] [Indexed: 12/21/2022]
Abstract
Macrophage activation syndrome (MAS) is a rare, life-threatening disease in which early diagnosis and aggressive therapeutic strategy may improve the outcome. Due to its rarity, epidemiologic data are still lacking. Hyperferritinemia is frequently associated with MAS and might modulate the cytokine storm, which is involved in the development of multiple organ failure. In this paper, we investigated clinical data, treatments, and outcome of a homogeneous cohort of 41 adult MAS patients, complicating autoimmune rheumatic diseases. MAS-related death occurred in 17 patients (42.5%) during the follow-up, and older age and increased serum ferritin levels, at the time of diagnosis, were significantly associated with mortality. In conclusion, adult MAS is associated with high mortality rate. Some clinical features at diagnosis may be predictive of MAS-associated death.
Collapse
Affiliation(s)
- Piero Ruscitti
- Division of Rheumatology, University of L'Aquila, L'Aquila, Italy
| | - Paola Cipriani
- Division of Rheumatology, University of L'Aquila, L'Aquila, Italy
| | | | - Francesco Masedu
- Division of Medical Statistic Unit, University of L'Aquila, L'Aquila, Italy
| | | | | | | | | | | | | | - Marco Valenti
- Division of Medical Statistic Unit, University of L'Aquila, L'Aquila, Italy
| | - Giovanni Triolo
- Division of Rheumatology, University of Palermo, Palermo, Italy
| | | |
Collapse
|