101
|
Verweyen EL, Schulert GS. Interfering with interferons: targeting the JAK-STAT pathway in complications of systemic juvenile idiopathic arthritis (SJIA). Rheumatology (Oxford) 2021; 61:926-935. [PMID: 34459891 PMCID: PMC9123899 DOI: 10.1093/rheumatology/keab673] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/27/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
Systemic JIA (SJIA) is distinguished from other forms of JIA by the prevalence of the severe, life-threatening complications macrophage activation syndrome (SJIA-MAS) and lung disease (SJIA-LD). Alternative therapeutics are urgently needed, as disease pathogenesis diverges from what is observed in SJIA, and currently available biologics are insufficient. SJIA-MAS, defined by a cytokine storm and dysregulated proliferation of T-lymphocytes, and SJIA-LD which presents with lymphocytic interstitial inflammation and pulmonary alveolar proteinosis, are both thought to be driven by IFNs, in particular the type II IFN-γ. Involvement of IFNs and a possible crosstalk of type I IFNs with existing biologics indicate a distinct role for the JAK-STAT signalling pathway in the pathogenesis of SJIA-MAS and SJIA-LD. Here, we review this role of JAK-STATs and IFNs in SJIA complications and discuss how new insights of ongoing research are shaping future therapeutic advances in the form of JAK inhibitors and antibodies targeting IFNs.
Collapse
Affiliation(s)
- Emely L Verweyen
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center
| | - Grant S Schulert
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA,Correspondence to: Grant Schulert, Division of Rheumatology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, MLC 4010, 3333 Burnet Avenue, Cincinnati, OH 45208, USA.
E-mail:
| |
Collapse
|
102
|
Adverse effects of biologic anti-inflammatory agents on the respiratory system: A review. Afr J Thorac Crit Care Med 2021; 27. [PMID: 34430870 PMCID: PMC8327682 DOI: 10.7196/ajtccm.2021.v27i2.117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2021] [Indexed: 12/12/2022] Open
Abstract
The therapy of autoimmune rheumatological conditions has undergone significant changes with the introduction of biologic antiinflammatory agents including cytokine antagonists and agents that interfere with the function of T and B cells or those that inhibit
intracellular enzymes such as Janus kinase (JAK). Although useful to control inflammation, these agents may be associated with druginduced lung disease, which may be difficult to differentiate from pulmonary disorders caused by the underlying autoimmune diseases.
This review aims to provide a description of lung disease, both infectious and non-infectious, that may be induced by the administration of
biologic anti-inflammatory agents with emphasis on inhibitors of tumour necrosis factor, interleukin-1, interleukin-6 and JAK.
Collapse
|
103
|
Atypical Presentation of Systemic Arthritis in a Toddler with Down Syndrome. Case Rep Pediatr 2021; 2021:6567770. [PMID: 34422425 PMCID: PMC8373510 DOI: 10.1155/2021/6567770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/06/2021] [Indexed: 12/17/2022] Open
Abstract
Systemic juvenile idiopathic arthritis (sJIA) is a chronic, inflammatory disease of childhood, which is characterized by the combination of arthritis, serositis, daily, high-spiking fevers, and evanescent macular rash and can present with the life-threatening complication of macrophage activation syndrome (MAS). Children with Down syndrome (DS) have complex medical challenges related to abnormalities in their immune system, which can cause a broad spectrum of disease manifestations, which can occur atypically. Children with DS are at increased risk for arthritis and interstitial lung disease (ILD) associated with sJIA that has high mortality. This case report outlines an atypical presentation of sJIA in a 21-month-old male with DS in which fever was not part of the initial presentation of sJIA and then later developed MAS and ILD. Due to broad spectrum of disease and atypical presentation in children with DS, this case report was created to increase awareness of atypical presentations of rheumatic disease in children with DS.
Collapse
|
104
|
Gao DK, Salomonis N, Henderlight M, Woods C, Thakkar K, Grom AA, Thornton S, Jordan MB, Wikenheiser-Brokamp KA, Schulert GS. IFN-γ is essential for alveolar macrophage driven pulmonary inflammation in macrophage activation syndrome. JCI Insight 2021; 6:e147593. [PMID: 34314387 PMCID: PMC8492332 DOI: 10.1172/jci.insight.147593] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 07/22/2021] [Indexed: 11/17/2022] Open
Abstract
Macrophage activation syndrome (MAS) is a life-threatening cytokine storm complicating systemic juvenile idiopathic arthritis (SJIA) driven by IFNγ. SJIA and MAS are associated with an unexplained emerging lung disease (SJIA-LD), with our recent work supporting pulmonary activation of IFNγ pathways pathologically linking SJIA-LD and MAS. Our objective was to mechanistically define the novel observation of pulmonary inflammation in the TLR9 mouse model of MAS. In acute MAS, lungs exhibit mild but diffuse CD4-predominant, perivascular interstitial inflammation with elevated IFNγ, IFN-induced chemokines, and AMΦ expression of IFNγ-induced genes. Single-cell RNA-sequencing confirmed IFN-driven transcriptional changes across lung cell types with myeloid expansion and detection of MAS-specific macrophage populations. Systemic MAS resolution was associated with increased AMΦ and interstitial lymphocytic infiltration. AMΦ transcriptomic analysis confirmed IFNγ-induced proinflammatory polarization during acute MAS, which switches towards an anti-inflammatory phenotype after systemic MAS resolution. Interestingly, recurrent MAS led to increased alveolar inflammation and lung injury, and reset AMΦ polarization towards a proinflammatory state. Furthermore, in mice bearing macrophages insensitive to IFNγ, both systemic feature of MAS and pulmonary inflammation were attenuated. These findings demonstrate that experimental MAS induces IFNγ-driven pulmonary inflammation replicating key features of SJIA-LD, and provides a model system for testing novel treatments directed towards SJIA-LD.
Collapse
Affiliation(s)
- Denny K Gao
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Nathan Salomonis
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Maggie Henderlight
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Christopher Woods
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Kairavee Thakkar
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Alexei A Grom
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Sherry Thornton
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Michael B Jordan
- Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | | | - Grant S Schulert
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| |
Collapse
|
105
|
Montano L, Donato F, Bianco PM, Lettieri G, Guglielmino A, Motta O, Bonapace IM, Piscopo M. Air Pollution and COVID-19: A Possible Dangerous Synergy for Male Fertility. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18136846. [PMID: 34202243 PMCID: PMC8297116 DOI: 10.3390/ijerph18136846] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 12/11/2022]
Abstract
Several studies indicate that semen quality has strongly declined in the last decades worldwide. Air pollution represents a significant co-factor with the COVID-19 impact and has negative effects on the male reproductive system, through pro-oxidant, inflammatory and immune-dysregulating mechanisms. It has recently been reported that chronic exposure to PM2.5 causes overexpression of the alveolar ACE2 receptor, the entry route of SARS-CoV-2 into the organism shared by the lungs and testis where expression is highest in the body. In the testis, the ACE2/Ang-(1-7)/MasR pathway plays an important role in the regulation of spermatogenesis and an indirect mechanism of testicular damage could be due to the blockade of the ACE2 receptor by SARS-CoV-2. This prevents the conversion of specific angiotensins, and their excess causes inflammation with the overproduction of cytokines. PM2.5-induced overexpression of the alveolar ACE2 receptor, in turn, could increase local viral load in patients exposed to pollutants, producing ACE2 receptor depletion and compromising host defenses. By presenting an overall view of epidemiological data and molecular mechanisms, this manuscript aims to interpret the possible synergistic effects of both air pollution and COVID-19 on male reproductive function, warning that the spread of SARS-CoV-2 in the fertile years may represent a significant threat to global reproductive health. All of this should be of great concern, especially for men of the age of maximum reproductive capacity, and an important topic of debate for policy makers. Altered environmental conditions, together with the direct and indirect short- and long-term effects of viral infection could cause a worsening of semen quality with important consequences for male fertility, especially in those areas with higher environmental impact.
Collapse
Affiliation(s)
- Luigi Montano
- Andrology Unit and Service of Lifestyle Medicine in UroAndrology, Local Health Authority (ASL) Salerno, Coordination Unit of the Network for Environmental and Reproductive Health (EcoFoodFertility Project), “Oliveto Citra Hospital”, 84020 Oliveto Citra, Italy
- PhD Program in Evolutionary Biology and Ecology, Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
- Correspondence: (L.M.); (I.M.B.); (M.P.); Tel.: +39-0331-339452 (I.M.B.); +39-0816-79081 (M.P.)
| | - Francesco Donato
- Unit of Hygiene, Epidemiology, and Public Health, Department of Medical and Surgical Specialties Radiological Sciences and Public Health, University of Brescia, 21100 Brescia, Italy;
| | - Pietro Massimiliano Bianco
- ISPRA, Italian Institute for Environmental Protection and Research, Via Vitaliano Brancati 60, 00144 Roma, Italy;
| | - Gennaro Lettieri
- Department of Biology, University of Naples Federico II, 80126 Napoli, Italy;
| | | | - Oriana Motta
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Salerno, Italy;
| | - Ian Marc Bonapace
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
- Correspondence: (L.M.); (I.M.B.); (M.P.); Tel.: +39-0331-339452 (I.M.B.); +39-0816-79081 (M.P.)
| | - Marina Piscopo
- Department of Biology, University of Naples Federico II, 80126 Napoli, Italy;
- Correspondence: (L.M.); (I.M.B.); (M.P.); Tel.: +39-0331-339452 (I.M.B.); +39-0816-79081 (M.P.)
| |
Collapse
|
106
|
Adult-onset Still's disease in focus: Clinical manifestations, diagnosis, treatment, and unmet needs in the era of targeted therapies. Semin Arthritis Rheum 2021; 51:858-874. [PMID: 34175791 DOI: 10.1016/j.semarthrit.2021.06.004] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Adult-onset Still's disease (AOSD) is a rare systemic inflammatory disorder of unknown etiology, characterized by a clinical triad of high spiking fever, arthralgia (± arthritis), and evanescent skin rash. Management of AOSD poses several challenges, including difficulty in diagnosis and limited therapeutic options. In this review, we examined whether AOSD and systemic juvenile idiopathic arthritis (SJIA) represent a continuum of the same disease. We also explored the latest available evidence related to prevalence, clinical and laboratory manifestations, complications, diagnostic challenges, novel biomarkers, and treatment options in the era of biologics and identified the unmet needs of patients with AOSD. METHODS A comprehensive systematic literature search was performed in the Embase and MEDLINE (via PubMed) literature databases. The search was limited to human studies published in English from inception up to March 2020. Additionally, abstracts presented at various conferences were screened and hand searches were performed. Publications were processed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. RESULTS A total of 123 publications were identified through the literature search, majority of which were case series and retrospective observational studies. AOSD and SJIA are widely considered part of the same disease spectrum owing to similarities in their clinical and biological features. The clinical presentation of AOSD is highly variable, accompanied by a broad spectrum of disease manifestations. Recent evidence suggests that the AOSD disease course can be classified into two distinct categories: "systemic" and "articular." Furthermore, AOSD patients may experience various life-threatening complications, such as macrophage activation syndrome - reported in as high as 23% of AOSD patients and considered to be the most severe complication characterized by a high mortality rate. The ambiguity in presentation and lack of serologic markers make the diagnosis of AOSD difficult, often leading to a delay in diagnosis. Given these limitations, the Yamaguchi and Fautrel criteria are the most widely used diagnostic tools in clinical practice. It has been observed that a clinical diagnosis of AOSD is generally reached by exclusion while investigating a patient with fever of unknown origin. Recent advances have demonstrated a major role of proinflammatory cytokines, such as interleukin (IL)-1, IL-6, IL-18, and IL-37, and other biomarkers in the pathogenesis and management of AOSD. Owing to the rarity of the disease, there are very limited clinical trials evaluating management strategies for AOSD. The current AOSD treatment paradigm includes non-steroidal anti-inflammatory drugs (NSAIDs) and glucocorticoids initially, conventional synthetic disease-modifying anti-rheumatic drugs in steroid-refractory patients, and biologics in those resistant to conventional treatment. Only a few country-specific guidelines for the management of AOSD have been published, and a treat-to-target approach, as previously recommended for SJIA, is still lacking. Canakinumab is the only FDA-approved biologic for the treatment of AOSD. CONCLUSION Emerging evidence supports that AOSD and SJIA represent a continuum of the same disease entity. Despite advancements in the understanding of AOSD, it continues to pose a substantial burden on patients and the healthcare systems, and substantial unmet needs exist across key domains such as the pathway to diagnosis, use of biomarkers in clinical practice, and standardized treatment strategies. Further research and collaboration is crucial for optimizing the diagnosis and management of AOSD patients.
Collapse
|
107
|
Gower WA, Vece TJ. Pediatric pulmonology 2019 year in review: rare and diffuse lung disease. Pediatr Pulmonol 2021; 56:1324-1331. [PMID: 33559960 DOI: 10.1002/ppul.25297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 12/31/2020] [Accepted: 01/20/2021] [Indexed: 11/07/2022]
Abstract
Pediatric Pulmonology publishes original research, review articles, and case reports on topics related to a wide range of children's respiratory disorders. Here we review manuscripts published in 2019 in this journal and others on (1) anatomic lung, airway, and vascular malformations, (2) children's interstitial lung disease, and (3) primary ciliary dyskinesia and non-cystic fibrosis bronchiectasis.
Collapse
Affiliation(s)
- William A Gower
- Division of Pediatric Pulmonology, Program for Rare and Interstitial Lung Disease, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Timothy J Vece
- Division of Pediatric Pulmonology, Program for Rare and Interstitial Lung Disease, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|
108
|
Di Cola I, Bruno F, Berardicurti O, Monti R, Conforti A, Di Sibio A, Pavlych V, Masciocchi C, Barile A, Cipriani P, Ruscitti P. Evaluating the multivisceral involvement on adult-onset Still's disease to retrieve imaging-based differences in patients with and without macrophage activation syndrome: results from a single-centre observational study. Clin Rheumatol 2021; 40:3971-3978. [PMID: 33855668 DOI: 10.1007/s10067-021-05727-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 03/24/2021] [Accepted: 03/30/2021] [Indexed: 11/25/2022]
Abstract
In this study, we aimed at describing the multivisceral involvement on adult-onset Still's disease (AOSD) to retrieve imaging-based differences in patients with and without macrophage activation syndrome (MAS). From our historical cohort, patients were assessed among those who underwent a total body CT scan. Clinical and CT scan characteristics of AOSD patients with and without MAS were compared. Out of 39 AOSD patients evaluated, 14 were complicated by MAS. These patients showed higher values of ferritin and systemic score. AOSD patients with MAS presented a higher prevalence of lung disease, hepatomegaly, splenomegaly, abdominal effusions, and lymph node enlargement than others without this complication. In addition, the presence of these manifestations significantly correlated with the systemic score, furtherly reinforcing its prognostic value. Due to the specific design of our study, our findings could be burdened by a selection bias since assessing those patients underwent a total body CT scan. Thus, these data should be prudently generalised suggesting the need of further studies to fully elucidate this issue. Our findings showed a higher prevalence of multiorgan involvement in AOSD patients with MAS, suggesting imaging-based differences, although other studies are needed to fully assess this issue. Pulmonary disease, hepatomegaly, splenomegaly, lymph node enlargement, and abdominal effusions were associated with a more aggressive subset of AOSD. Key Points •The importance of an accurate assessment AOSD multivisceral involvement is suggested since it is associated with life-threatening complications. •A higher prevalence of multiorgan involvement in AOSD patients with MAS could be recognised, than others without this complication, suggesting imaging-based differences. •AOSD multivisceral involvement may correlate with the systemic score, furtherly reinforcing its prognostic value.
Collapse
Affiliation(s)
- Ilenia Di Cola
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| | - Federico Bruno
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| | - Onorina Berardicurti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| | - Riccardo Monti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| | - Alessandro Conforti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| | - Alessandra Di Sibio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| | - Viktoriya Pavlych
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| | - Carlo Masciocchi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| | - Antonio Barile
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| | - Paola Cipriani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| | - Piero Ruscitti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy.
| |
Collapse
|
109
|
Wan L, Gao Y, Gu J, Chi H, Wang Z, Hu Q, Jia J, Liu T, Li B, Teng J, Liu H, Cheng X, Ye J, Su Y, Yang C, Shi H, Zhang M. Total metabolic lesion volume of lymph nodes measured by 18F-FDG PET/CT: a new predictor of macrophage activation syndrome in adult-onset Still's disease. Arthritis Res Ther 2021; 23:97. [PMID: 33785060 PMCID: PMC8008587 DOI: 10.1186/s13075-021-02482-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 03/17/2021] [Indexed: 12/15/2022] Open
Abstract
Background To investigate the potential utility of quantitative parameters obtained by 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) in the assessment of disease severity and the occurrence of macrophage activation syndrome (MAS) in adult-onset Still’s disease (AOSD). Methods Fifty-seven patients with AOSD who underwent pre-treatment 18F-FDG PET/CT were recruited in this study and compared with 60 age- and sex-matched healthy controls. Clinical features and laboratory data were recorded. The systemic score was assessed to determine the disease severity. The maximal standardized uptake value (SUVmax), metabolic lesion volume (MLV), and total lesion glycolysis (TLG) were used to evaluate the involved organs and tissues that abnormally accumulated 18F-FDG. Multivariate analysis was performed to identify the PET/CT-derived risk factors contributing to the AOSD-related MAS, and their diagnostic efficiency was evaluated. Results High 18F-FDG accumulation was observed in the bone marrow (SUVmax median, 5.10), spleen (SUVmax median, 3.70), and lymph nodes (LNs, SUVmax median, 5.55). The SUVmax of the bone marrow (rho = 0.376, p = 0.004), SUVmax of the spleen (rho = 0.450, p < 0.001), TLGtotal of LNs (rho = 0.386, p = 0.017), and MLVtotal of LNs (rho = 0.391, p = 0.015) were correlated with the systemic score. The SUVmax of the spleen (p = 0.017), TLGtotal of LNs (p = 0.045), and MLVtotal of LNs (p = 0.012) were higher in patients with MAS than in those without MAS. A MLVtotal of LNs > 62.2 (OR 27.375, p = 0.042) was an independent predictive factor for MAS with a sensitivity of 80.0% and a specificity of 93.9%. Conclusions The glucose metabolic level of the spleen could be an effective and easy-to-use imaging indicator of disease severity, and MLVtotal of LNs > 62.2 was a strong predictor of MAS occurrence in patients with AOSD.
Collapse
Affiliation(s)
- Liyan Wan
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuting Gao
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Jieyu Gu
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Huihui Chi
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhihong Wang
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qiongyi Hu
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jinchao Jia
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Tingting Liu
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Jialin Teng
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Honglei Liu
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaobing Cheng
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Junna Ye
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yutong Su
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chengde Yang
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hui Shi
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Min Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| |
Collapse
|
110
|
Hosseinikhah SM, Barani M, Rahdar A, Madry H, Arshad R, Mohammadzadeh V, Cucchiarini M. Nanomaterials for the Diagnosis and Treatment of Inflammatory Arthritis. Int J Mol Sci 2021; 22:3092. [PMID: 33803502 PMCID: PMC8002885 DOI: 10.3390/ijms22063092] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/04/2021] [Accepted: 03/16/2021] [Indexed: 12/12/2022] Open
Abstract
Nanomaterials have received increasing attention due to their unique chemical and physical properties for the treatment of rheumatoid arthritis (RA), the most common complex multifactorial joint-associated autoimmune inflammatory disorder. RA is characterized by an inflammation of the synovium with increased production of proinflammatory cytokines (IL-1, IL-6, IL-8, and IL-10) and by the destruction of the articular cartilage and bone, and it is associated with the development of cardiovascular disorders such as heart attack and stroke. While a number of imaging tools allow for the monitoring and diagnosis of inflammatory arthritis, and despite ongoing work to enhance their sensitivity and precision, the proper assessment of RA remains difficult particularly in the early stages of the disease. Our goal here is to describe the benefits of applying various nanomaterials as next-generation RA imaging and detection tools using contrast agents and nanosensors and as improved drug delivery systems for the effective treatment of the disease.
Collapse
Affiliation(s)
- Seyedeh Maryam Hosseinikhah
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91886-17871, Iran;
| | - Mahmood Barani
- Department of Chemistry, Shahid Bahonar University of Kerman, Kerman 761691411, Iran;
| | - Abbas Rahdar
- Department of Physics, Faculty of Science, University of Zabol, Zabol 538-9861, Iran
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, D-66421 Homburg/Saar, Germany;
| | - Rabia Arshad
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan;
| | - Vahideh Mohammadzadeh
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Science, Mashhad 91886-17871, Iran;
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, D-66421 Homburg/Saar, Germany;
| |
Collapse
|
111
|
Sustained remission after haploidentical bone marrow transplantation in a child with refractory systemic juvenile idiopathic arthritis. Pediatr Rheumatol Online J 2021; 19:27. [PMID: 33712044 PMCID: PMC7953742 DOI: 10.1186/s12969-021-00523-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 03/04/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Some patients with systemic juvenile idiopathic arthritis (SJIA) and severe, refractory disease achieved remission through intensive immunosuppressive treatment followed by autologous hematopoietic stem cell transplantation (HSCT). However, disease relapsed in most cases. More recently selected SJIA patients received allogenic HSCT from a HLA-identical sibling or a HLA matched unrelated donor. While most transplanted patients achieved sustained SJIA remission off-treatment, the procedure-related morbidity was high. CASE REPORT A girl presented SJIA with a severe disease course since the age of 15 months. She was refractory to the combination of methotrexate and steroids to anti-interleukin (IL)-1, then anti-IL-6, tumor necrosis factor alpha inhibitors, and thalidomide. Given the high disease burden and important treatment-related toxicity the indication for a haploidentical HSCT from her mother was validated, as no HLA matched donor was available. The patient received a T replete bone marrow graft at the age of 3.7 years. Conditioning regimen contained Rituximab, Alemtuzumab, Busulfan, and Fludarabine. Cyclophosphamide at D + 3 and + 4 post HSCT was used for graft-versus-host-disease prophylaxis, followed by Cyclosporin A and Mycophenolate Mofetil. Post HSCT complications included severe infections, grade 3 intestinal graft-versus-host-disease, autoimmune thyroiditis, and immune thrombocytopenia. Three years after HSCT, the child is alive and well, notwithstanding persistent hypothyroidy requiring substitution. Immune thrombocytopenia had resolved. Most importantly, SJIA was in complete remission, off immunosuppressive drugs. CONCLUSION Allogenic HSCT may be a therapeutic option, even with a HLA haplo-identical alternative donor, in patients with inflammatory diseases such as SJIA. Despite increased experience with this treatment, the risk of life-threatening complications restrains its indication to selected patients with severe, refractory disease.
Collapse
|
112
|
Malengier-Devlies B, Decaesteker T, Dekoster K, Vanstapel A, Ahmadzadeh K, Poosti F, Mitera T, Seldeslachts L, Verbeken E, Wouters C, Vande Velde G, Vanoirbeek J, Matthys P. Lung Functioning and Inflammation in a Mouse Model of Systemic Juvenile Idiopathic Arthritis. Front Immunol 2021; 12:642778. [PMID: 33777039 PMCID: PMC7996094 DOI: 10.3389/fimmu.2021.642778] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/10/2021] [Indexed: 11/13/2022] Open
Abstract
Systemic juvenile idiopathic arthritis (sJIA) is an immune disorder characterized by fever, skin rash, arthritis and splenomegaly. Recently, increasing number of sJIA patients were reported having lung disease. Here, we explored lung abnormalities in a mouse model for sJIA relying on injection of IFN-γ deficient (IFN-γ KO) mice with complete Freund's adjuvant (CFA). Monitoring of lung changes during development of sJIA using microcomputer tomography revealed a moderate enlargement of lungs, a decrease in aerated and increase in non-aerated lung density. When lung function and airway reactivity to methacholine was assessed, gender differences were seen. While male mice showed an increased tissue hysteresivity, female animals were characterized by an increased airway hyperactivity, mirroring ongoing inflammation. Histologically, lungs of sJIA-like mice showed subpleural and parenchymal cellular infiltrates and formation of small granulomas. Flow cytometric analysis identified immature and mature neutrophils, and activated macrophages as major cell infiltrates. Lung inflammation in sJIA-like mice was accompanied by augmented expression of IL-1β and IL-6, two target cytokines in the treatment of sJIA. The increased expression of granulocyte colony stimulating factor, a potent inducer of granulopoiesis, in lungs of mice was striking considering the observed neutrophilia in patients. We conclude that development of sJIA in a mouse model is associated with lung inflammation which is distinct to the lung manifestations seen in sJIA patients. Our observations however underscore the importance of monitoring lung disease during systemic inflammation and the model provides a tool to explore the underlying mechanism of lung pathology in an autoinflammatory disease context.
Collapse
Affiliation(s)
- Bert Malengier-Devlies
- Laboratory of Immunobiology, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Tatjana Decaesteker
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Kaat Dekoster
- Biomedical MRI, Department of Imaging & Pathology, KU Leuven, Leuven, Belgium
| | - Arno Vanstapel
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Kourosh Ahmadzadeh
- Laboratory of Immunobiology, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Fariba Poosti
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Tania Mitera
- Laboratory of Immunobiology, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Laura Seldeslachts
- Biomedical MRI, Department of Imaging & Pathology, KU Leuven, Leuven, Belgium
| | - Erik Verbeken
- Morphology and Molecular Pathology Section, University Hospitals Leuven, Leuven, Belgium
| | - Carine Wouters
- Laboratory of Immunobiology, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium.,Division of Pediatric Rheumatology, University Hospitals Leuven, Leuven, Belgium.,European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) at University Hospital Leuven, Leuven, Belgium
| | - Greetje Vande Velde
- Biomedical MRI, Department of Imaging & Pathology, KU Leuven, Leuven, Belgium
| | - Jeroen Vanoirbeek
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Patrick Matthys
- Laboratory of Immunobiology, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| |
Collapse
|
113
|
Tomaras S, Goetzke CC, Kallinich T, Feist E. Adult-Onset Still's Disease: Clinical Aspects and Therapeutic Approach. J Clin Med 2021; 10:733. [PMID: 33673234 PMCID: PMC7918550 DOI: 10.3390/jcm10040733] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/27/2021] [Accepted: 02/07/2021] [Indexed: 12/22/2022] Open
Abstract
Adult-onset Still's disease (AoSD) is a rare systemic autoinflammatory disease characterized by arthritis, spiking fever, skin rash and elevated ferritin levels. The reason behind the nomenclature of this condition is that AoSD shares certain symptoms with Still's disease in children, currently named systemic-onset juvenile idiopathic arthritis. Immune dysregulation plays a central role in AoSD and is characterized by pathogenic involvement of both arms of the immune system. Furthermore, the past two decades have seen a large body of immunological research on cytokines, which has attributed to both a better understanding of AoSD and revolutionary advances in treatment. Additionally, recent studies have introduced a new approach by grouping patients with AoSD into only two phenotypes: one with predominantly systemic features and one with a chronic articular disease course. Diagnosis presupposes an extensive diagnostic workup to rule out infections and malignancies. The severe end of the spectrum of this disease is secondary haemophagocytic lymphohistiocytosis, better known as macrophage activation syndrome. In this review, we discuss current research conducted on the pathogenesis, diagnosis, classification, biomarkers and complications of AoSD, as well as the treatment strategy at each stage of the disease course. We also highlight the similarities and differences between AoSD and systemic-onset juvenile idiopathic arthritis. There is a considerable need for large multicentric prospective trials.
Collapse
Affiliation(s)
- Stylianos Tomaras
- Department of Rheumatology, Helios Clinic Vogelsang-Gommern, 39245 Gommern, Germany;
| | - Carl Christoph Goetzke
- Department of Pediatrics, Division of Pulmonology, Immunology and Critical Care Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany; (C.C.G.); (T.K.)
- German Rheumatism Research Center (DRFZ), Leibniz Association, 10117 Berlin, Germany
- Berlin Institute of Health, 10178 Berlin, Germany
| | - Tilmann Kallinich
- Department of Pediatrics, Division of Pulmonology, Immunology and Critical Care Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany; (C.C.G.); (T.K.)
- German Rheumatism Research Center (DRFZ), Leibniz Association, 10117 Berlin, Germany
- Berlin Institute of Health, 10178 Berlin, Germany
| | - Eugen Feist
- Department of Rheumatology, Helios Clinic Vogelsang-Gommern, 39245 Gommern, Germany;
| |
Collapse
|
114
|
Abstract
Children with rheumatic disease have rare pulmonary manifestations that may cause significant morbidity and mortality. These children are often clinically asymptomatic until disease has significantly progressed, so they should be screened for pulmonary involvement. There has been recent recognition of a high mortality-related lung disease in systemic-onset juvenile idiopathic arthritis; risk factors include onset of juvenile idiopathic arthritis less than 2 years of age, history of macrophage activation syndrome, presence of trisomy 21, and history of anaphylactic reaction to biologic therapy. Early recognition and treatment of lung disease in children with rheumatic diseases may improve outcomes.
Collapse
|
115
|
Verweyen EL, Pickering A, Grom AA, Schulert GS. Distinct Gene Expression Signatures Characterize Strong Clinical Responders Versus Nonresponders to Canakinumab in Children With Systemic Juvenile Idiopathic Arthritis. Arthritis Rheumatol 2021; 73:1334-1340. [PMID: 33452871 DOI: 10.1002/art.41640] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/04/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Canakinumab is a human anti-interleukin-1β (anti-IL-1β) blocking agent that effectively neutralizes IL-1β-mediated signaling for treatment of systemic juvenile idiopathic arthritis (JIA). While many patients have dramatic clinical response to IL-1 blockade, approximately one-third fail to respond, but there are currently no validated clinical or immunologic predictors of response. We undertook this study to characterize distinct gene signatures for treatment response and nonresponse to canakinumab in systemic JIA patients. METHODS We performed a secondary analysis of whole-blood gene expression microarrays using blood samples obtained from healthy controls and systemic JIA patients at baseline and on day 3 after canakinumab treatment (GEO accession no. GSE80060). Patients were considered strong clinical responders if they met the ACR90 response (exhibited ≥90% improvement in the American College of Rheumatology [ACR] JIA response criteria; nonresponders were those who met ACR30 [exhibiting ≤30% improvement in the ACR JIA response criteria]). A random-effects model with patient identity as the random variable was used for differential expression analysis. RESULTS We identified a distinct gene expression signature in patients with a strong clinical response to canakinumab treatment as compared to nonresponders, mediated by up-regulation of neutrophil- and IL-1-associated genes and characterized by increasing divergence from control transcriptomes with increasing clinical response. We also identified a signature including up-regulated CD163 expression that was associated with canakinumab nonresponse. Intriguingly, canakinumab treatment induced either up- or down-regulation of type I interferon (IFN) genes, independent of clinical response. CONCLUSION Here, we identify a gene signature in systemic JIA patients prior to receiving treatment that distinguishes strong responders to canakinumab from nonresponders. Further prospective studies are needed to assess the utility of these insights for treatment decisions in systemic JIA and to track the association of up-regulated type I IFN signatures with systemic JIA complications.
Collapse
Affiliation(s)
| | | | - Alexei A Grom
- Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Grant S Schulert
- Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
116
|
Ramsubeik K, Motilal S, Sanchez-Ramos L, Ramrattan LA, Kaeley GS, Singh JA. Diagnostic accuracy of salivary gland ultrasound in Sjögren's syndrome: A systematic review and meta-analysis. Ther Adv Musculoskelet Dis 2020; 12:1759720X20973560. [PMID: 33281953 PMCID: PMC7682247 DOI: 10.1177/1759720x20973560] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/25/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND To systematically review the diagnostic accuracy of salivary gland ultrasound in primary Sjögren's syndrome (pSS). METHODS PubMed, Embase, CINAHL, Cochrane Central and Scopus and ClinicalTrials.gov were searched to identify diagnostic or validation studies in patients with pSS meeting the diagnostic criteria. A diagnostic test meta-analysis was performed using a bivariate model to calculate the pooled sensitivity, specificity, positive/negative likelihood ratios, and the diagnostic odds ratio. Meta-regression analyses were done for several pSS covariates. RESULTS Sixty-five studies met our criteria for the qualitative review. Fifty-four studies with a total of 6087 patients were included in the meta-analysis. Pooled sensitivity for salivary gland ultrasound was 80% [95% confidence interval (CI): 77-83%; I 2 = 78%], and specificity was 90% (95% CI: 87-92%; I 2 = 76%). The pooled positive and negative likelihood ratios were 8 (95% CI: 6.4-10) and 0.22 (95% CI: 0.19-0.25), respectively. The corresponding pooled diagnostic odds ratio (DOR) was 37 (95% CI: 28-48). Separate meta-regression models resulted in similar diagnostic estimates: (a) adjusted for mean age: sensitivity 81% (95% CI:77-84%; I 2 = 99%) and specificity 90% (95% CI: 87-93%; I2 = 99%); (b) adjusted for mean disease duration, sensitivity 79% (95% CI:72-84%; I 2 = 99%), and specificity 90% (89-94%; I 2 = 99%). The diagnostic estimates were robust to sensitivity analyses by quality criteria, pSS diagnostic criteria and ultrasound scoring systems. CONCLUSION Salivary gland ultrasound is a valuable modality for the diagnosis of Sjögren's syndrome. It is plausible that salivary gland ultrasound can be used as an important criterion for the diagnosis of pSS.
Collapse
Affiliation(s)
- Karishma Ramsubeik
- Division of Rheumatology, University of Florida College of Medicine, Jacksonville, Florida, USA
| | - Shastri Motilal
- Department of Para Clinical Sciences, The University of the West Indies, St Augustine, Trinidad and Tobago
| | - Luis Sanchez-Ramos
- Division of Maternal and Fetal Medicine, University of Florida College of Medicine, Jacksonville, Florida, USA
| | - Laurie Ann Ramrattan
- Division of Rheumatology, University of Florida College of Medicine, Jacksonville, Florida, USA
| | - Gurjit S. Kaeley
- Division of Rheumatology, University of Florida College of Medicine, Jacksonville, Florida, USA
| | - Jasvinder A. Singh
- Department of Medicine, Birmingham VA Medical Center, Faculty Office Tower 805B, 510, 20th Street South, Birmingham, AL 35233, USA
| |
Collapse
|
117
|
Murray GM, Vastert SJ, Ramanan AV. Aiming high: quantifying inflammation in systemic onset juvenile idiopathic arthritis (sJIA), a multi-faceted and complex inflammatory disease. Rheumatology (Oxford) 2020; 59:3124-3126. [PMID: 32844227 DOI: 10.1093/rheumatology/keaa394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/08/2020] [Indexed: 01/01/2023] Open
Affiliation(s)
- Grainne M Murray
- Department of Paediatric Rheumatology, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Sebastiaan J Vastert
- Department of Paediatric Rheumatology and Immunology and Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Athimalaipet V Ramanan
- Department of Paediatric Rheumatology, University Hospitals Bristol NHS Foundation Trust, Bristol, UK.,Translational Health Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
118
|
Chau AS, Cole BL, Debley JS, Nanda K, Rosen ABI, Bamshad MJ, Nickerson DA, Torgerson TR, Allenspach EJ. Heme oxygenase-1 deficiency presenting with interstitial lung disease and hemophagocytic flares. Pediatr Rheumatol Online J 2020; 18:80. [PMID: 33066778 PMCID: PMC7565350 DOI: 10.1186/s12969-020-00474-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 10/06/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Heme oxygenase-1 (HMOX1) catalyzes the metabolism of heme into carbon monoxide, ferrous iron, and biliverdin. Through biliverdin reductase, biliverdin becomes bilirubin. HMOX1-deficiency is a rare autosomal recessive disorder with hallmark features of direct antibody negative hemolytic anemia with normal bilirubin, hyperinflammation and features similar to macrophage activation syndrome. Clinical findings have included asplenia, nephritis, hepatitis, and vasculitis. Pulmonary features and evaluation of the immune response have been limited. CASE PRESENTATION We present a young boy who presented with chronic respiratory failure due to nonspecific interstitial pneumonia following a chronic history of infection-triggered recurrent hyperinflammatory flares. Episodes included hemolysis without hyperbilirubinemia, immunodeficiency, hepatomegaly with mild transaminitis, asplenia, leukocytosis, thrombocytosis, joint pain and features of macrophage activation with negative autoimmune serologies. Lung biopsy revealed cholesterol granulomas. He was found post-mortem by whole exome sequencing to have a compound heterozygous paternal frame shift a paternal frame shift HMOX1 c.264_269delCTGG (p.L89Sfs*24) and maternal splice donor HMOX1 (c.636 + 2 T > A) consistent with HMOX1 deficiency. Western blot analysis confirmed lack of HMOX1 protein upon oxidant stimulation of the patient cells. CONCLUSIONS Here, we describe a phenotype expansion for HMOX1-deficiency to include not only asplenia and hepatomegaly, but also interstitial lung disease with cholesterol granulomas and inflammatory flares with hemophagocytosis present in the bone marrow.
Collapse
Affiliation(s)
- Alice S. Chau
- grid.34477.330000000122986657Division of Allergy & Infectious Disease, Department of Medicine, University of Washington, Seattle, Washington USA ,grid.240741.40000 0000 9026 4165Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Jack MacDonald Building – 6th floor, 1900 9th Avenue, Seattle, Washington 98101 USA
| | - Bonnie L. Cole
- grid.34477.330000000122986657Department of Pathology and Laboratory Medicine, University of Washington, Seattle, Washington USA ,grid.507913.9Brotman Baty Institute for Precision Medicine, Seattle, Washington USA
| | - Jason S. Debley
- grid.240741.40000 0000 9026 4165Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Jack MacDonald Building – 6th floor, 1900 9th Avenue, Seattle, Washington 98101 USA ,grid.34477.330000000122986657Department of Pediatrics, University of Washington, Seattle, Washington USA
| | - Kabita Nanda
- grid.34477.330000000122986657Department of Pediatrics, University of Washington, Seattle, Washington USA
| | - Aaron B. I. Rosen
- grid.240741.40000 0000 9026 4165Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Jack MacDonald Building – 6th floor, 1900 9th Avenue, Seattle, Washington 98101 USA
| | - Michael J. Bamshad
- grid.507913.9Brotman Baty Institute for Precision Medicine, Seattle, Washington USA ,grid.34477.330000000122986657Department of Pediatrics, University of Washington, Seattle, Washington USA ,grid.34477.330000000122986657Genome Sciences, University of Washington, Seattle, Washington USA
| | - Deborah A. Nickerson
- grid.507913.9Brotman Baty Institute for Precision Medicine, Seattle, Washington USA ,grid.34477.330000000122986657Genome Sciences, University of Washington, Seattle, Washington USA
| | - Troy R. Torgerson
- grid.507729.eExperimental Immunology, Allen Institute, Seattle, Washington USA
| | - Eric J. Allenspach
- grid.240741.40000 0000 9026 4165Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Jack MacDonald Building – 6th floor, 1900 9th Avenue, Seattle, Washington 98101 USA ,grid.507913.9Brotman Baty Institute for Precision Medicine, Seattle, Washington USA ,grid.34477.330000000122986657Department of Pediatrics, University of Washington, Seattle, Washington USA
| |
Collapse
|
119
|
Kilinc AA, Arslan A, Yildiz M, Kucur M, Adrovic A, Barut K, Sahin S, Cokugras H, Kasapcopur O. Serum KL-6 level as a biomarker of interstitial lung disease in childhood connective tissue diseases: a pilot study. Rheumatol Int 2020; 40:1701-1706. [PMID: 31784789 DOI: 10.1007/s00296-019-04485-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 11/23/2019] [Indexed: 12/24/2022]
Abstract
Krebs von den Lungen-6 (KL-6) has been described as a promising biomarker in the diagnosis and determining the severity of interstitial lung disease in adults with connective tissue disease. This study was performed to determine whether the serum KL-6 level is useful as a biomarker for detecting the interstitial lung disease (ILD) in pediatric cases of connective tissue disease (CTD). In total, 88 patients [36 patients with systemic juvenile idiopathic arthritis (sJIA), 27 patients with juvenile systemic lupus erythematosus (JSLE), 14 patients with juvenile dermatomyositis (JDM), and 11 patients with juvenile systemic sclerosis (JSSc)] and 68 healthy controls were included in this study. Age, sex, and duration of CTD and ILD (if any) were recorded. Blood samples from all the patients and controls were examined by ELISA. Eleven of the 88 patients with CTD (12.5%) had ILD and all of them were symptomatic. Subgroup analysis indicated that eight patients had JSSc, two had JSLE, and one had systemic JIA. The median serum KL-6 level was 1450.5 U/mL (interquartile range (IQR) 742.9-2603.2) in the CTD with ILD group, 415.9 U/mL (IQR 233.4-748.4) in the CTD without ILD group, and 465.9 U/mL (IQR 273.6-1036) in the control group. KL-6 levels were significantly higher in the CTD with ILD group than the CTD without ILD group and the control group (p = 0.003). At a cut-off of 712.5 U/ml identified by ROC curve, serum KL-6 yielded a sensitivity of 81% and specificity of 72% for CTD with ILD group. There was no significant difference in serum KL-6 level among the disease subgroups (sJIA, JSLE, JSSc, JDM), in either the CTD with ILD group or the CTD without ILD group (p > 0.05). In conclusion, KL-6 is a useful biomarker of CTD with ILD in pediatric patients.
Collapse
Affiliation(s)
- Ayse Ayzit Kilinc
- Department of Pediatric Pulmonology, Cerrahpasa Faculty of Medicine, İstanbul University-Cerrahpasa, İstanbul, Turkey
| | - Asli Arslan
- Department of Pediatric Pulmonology, Cerrahpasa Faculty of Medicine, İstanbul University-Cerrahpasa, İstanbul, Turkey
| | - Mehmet Yildiz
- Department of Pediatric Rheumatology, Cerrahpasa Faculty of Medicine, İstanbul University-Cerrahpasa, İstanbul, Turkey
| | - Mine Kucur
- Department of Biochemistry, Cerrahpasa Faculty of Medicine, İstanbul University-Cerrahpasa, İstanbul, Turkey
| | - Amra Adrovic
- Department of Pediatric Rheumatology, Cerrahpasa Faculty of Medicine, İstanbul University-Cerrahpasa, İstanbul, Turkey
| | - Kenan Barut
- Department of Pediatric Rheumatology, Cerrahpasa Faculty of Medicine, İstanbul University-Cerrahpasa, İstanbul, Turkey
| | - Sezgin Sahin
- Department of Pediatric Rheumatology, Cerrahpasa Faculty of Medicine, İstanbul University-Cerrahpasa, İstanbul, Turkey
| | - Haluk Cokugras
- Department of Pediatric Pulmonology, Cerrahpasa Faculty of Medicine, İstanbul University-Cerrahpasa, İstanbul, Turkey
| | - Ozgur Kasapcopur
- Department of Pediatric Rheumatology, Cerrahpasa Faculty of Medicine, İstanbul University-Cerrahpasa, İstanbul, Turkey.
| |
Collapse
|
120
|
Abstract
PURPOSE OF REVIEW We review the epidemiology, pathophysiology, and management of pericarditis most commonly complicating autoimmune and autoinflammatory conditions. RECENT FINDINGS Typically, pericarditis occurs in the context of a systemic flare of the underlying disease but infrequently, it is the presenting manifestation requiring a high index of suspicion to unravel the indolent cause. Pericardial involvement in rheumatic diseases encompasses a clinical spectrum to include acute, recurrent and incessant pericarditis, constrictive pericarditis, asymptomatic pericardial effusion, and pericardial tamponade. Direct evidence on the pathophysiology of pericarditis in the context of rheumatic diseases is scant. It is theorized that immune perturbations within pericardial tissue result from the underlying central immunopathology of the respective autoimmune or autoinflammatory disease. Pericarditis management depends on acuity, the underlying cause and epidemiological features such as patient's immune status and geographic prevalence of infections such as tuberculosis. Immunosuppressive medications including biologics such as interleukin 1 blockers emerge as possible steroid sparing agents for pericarditis treatment.
Collapse
Affiliation(s)
- Apostolos Kontzias
- Division of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Stony Brook University School of Medicine, Stony Brook, NY, USA.
| | - Amir Barkhodari
- Division of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Stony Brook University School of Medicine, Stony Brook, NY, USA
| | - QingPing Yao
- Division of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Stony Brook University School of Medicine, Stony Brook, NY, USA
| |
Collapse
|
121
|
Ruscitti P, Bruno F, Berardicurti O, Acanfora C, Pavlych V, Palumbo P, Conforti A, Carubbi F, Di Cola I, Di Benedetto P, Cipriani P, Grassi D, Masciocchi C, Iagnocco A, Barile A, Giacomelli R. Lung involvement in macrophage activation syndrome and severe COVID-19: results from a cross-sectional study to assess clinical, laboratory and artificial intelligence-radiological differences. Ann Rheum Dis 2020; 79:1152-1155. [PMID: 32719039 PMCID: PMC7456556 DOI: 10.1136/annrheumdis-2020-218048] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVES To evaluate the clinical pictures, laboratory tests and imaging of patients with lung involvement, either from severe COVID-19 or macrophage activation syndrome (MAS), in order to assess how similar these two diseases are. METHODS The present work has been designed as a cross-sectional single-centre study to compare characteristics of patients with lung involvement either from MAS or severe COVID-19. Chest CT scans were assessed by using an artificial intelligence (AI)-based software. RESULTS Ten patients with MAS and 47 patients with severe COVID-19 with lung involvement were assessed. Although all patients showed fever and dyspnoea, patients with MAS were characterised by thrombocytopaenia, whereas patients with severe COVID-19 were characterised by lymphopaenia and neutrophilia. Higher values of H-score characterised patients with MAS when compared with severe COVID-19. AI-reconstructed images of chest CT scan showed that apical, basal, peripheral and bilateral distributions of ground-glass opacities (GGOs), as well as apical consolidations, were more represented in severe COVID-19 than in MAS. C reactive protein directly correlated with GGOs extension in both diseases. Furthermore, lymphopaenia inversely correlated with GGOs extension in severe COVID-19. CONCLUSIONS Our data could suggest laboratory and radiological differences between MAS and severe COVID-19, paving the way for further hypotheses to be investigated in future confirmatory studies.
Collapse
Affiliation(s)
- Piero Ruscitti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Abruzzo, Italy
| | - Federico Bruno
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Abruzzo, Italy
| | - Onorina Berardicurti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Abruzzo, Italy
| | - Chiara Acanfora
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Abruzzo, Italy
| | - Viktoriya Pavlych
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Abruzzo, Italy
| | - Pierpaolo Palumbo
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Abruzzo, Italy
| | - Alessandro Conforti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Abruzzo, Italy
| | - Francesco Carubbi
- Department of Medicine, ASL 1 Avezzano Sulmona L'Aquila, L'Aquila, Abruzzo, Italy
| | - Ilenia Di Cola
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Abruzzo, Italy
| | - Paola Di Benedetto
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Abruzzo, Italy
| | - Paola Cipriani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Abruzzo, Italy
| | - Davide Grassi
- Department of Life, Health, and Environmental Sciences, University of L'Aquila, L'Aquila, Abruzzo, Italy
| | - Carlo Masciocchi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Abruzzo, Italy
| | - Annamaria Iagnocco
- Dipartimento Scienze Cliniche e Biologiche, Università degli Studi di Torino, Torino, Piemonte, Italy
| | - Antonio Barile
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Abruzzo, Italy
| | - Roberto Giacomelli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Abruzzo, Italy
| |
Collapse
|
122
|
Lopez-Rodriguez E, Ochs M. Reply to: Comments on “Air Space Distension Precedes Spontaneous Fibrotic Remodeling and Impaired Cholesterol Metabolism in the Absence of Surfactant Protein C”. Am J Respir Cell Mol Biol 2020; 63:399-402. [DOI: 10.1165/rcmb.2020-0158le] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
123
|
Alehashemi S, Goldbach-Mansky R. Human Autoinflammatory Diseases Mediated by NLRP3-, Pyrin-, NLRP1-, and NLRC4-Inflammasome Dysregulation Updates on Diagnosis, Treatment, and the Respective Roles of IL-1 and IL-18. Front Immunol 2020; 11:1840. [PMID: 32983099 PMCID: PMC7477077 DOI: 10.3389/fimmu.2020.01840] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/08/2020] [Indexed: 12/19/2022] Open
Abstract
Recent research has led to novel findings in inflammasome biology and genetics that altered the diagnosis and management of patients with autoinflammatory syndromes caused by NLRP3-, Pyrin-, NLRP1-, and NLRC4-inflammasomes and spurred the development of novel treatments. The use of next-generation sequencing in clinical practice allows for rapid diagnosis and the detection of somatic mutations that cause autoinflammatory diseases. Clinical differences in patients with NLRP3, pyrin, and NLRP1 inflammasomopathies, and the constitutive elevation of unbound free serum IL-18 that predisposes to the development of macrophage activation syndrome (MAS) in patients with gain-of function mutations in NLRC4 led to the screening and the characterization of novel diseases presenting with constitutively elevated serum IL-18 levels, and start to unravel the biology of "high IL-18 states" that translate into the use of biomarkers that improve diagnosis and monitoring of disease activity and investigations of treatments that target IL-18 and IFN-gamma which promise to improve the management and outcome of these conditions. Lastly, advances in structural modeling by cryo-electron microscopy (cryo-EM) of gasdermin, and of NLRP3- and NLRC4-inflammasome assembly, and the characterization of post-translational modifications (PTM) that regulate inflammasome activation, coupled with high-throughput screening (HTS) of libraries of inflammasome-inhibiting compounds, promise a new generation of treatments for patients with inflammasome-mediated diseases.
Collapse
Affiliation(s)
- Sara Alehashemi
- Translational Autoinflammatory Diseases Section (TADS), Laboratory of Clinical Immunology and Microbiology (LCIM), National Institutes of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Raphaela Goldbach-Mansky
- Translational Autoinflammatory Diseases Section (TADS), Laboratory of Clinical Immunology and Microbiology (LCIM), National Institutes of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
124
|
Canna SW, Schulert GS, de Jesus A, Pickering A, Brunner H, Gadina M, Levine S, Goldbach-Mansky R, Boutelle J, Sinha R, DeBenedetti F, Grom A. Proceedings from the 2 nd Next Gen Therapies for Systemic Juvenile Idiopathic Arthritis and Macrophage Activation Syndrome symposium held on October 3-4, 2019. Pediatr Rheumatol Online J 2020; 18:53. [PMID: 32664935 PMCID: PMC7360380 DOI: 10.1186/s12969-020-00444-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 06/17/2020] [Indexed: 02/07/2023] Open
Abstract
For reasons poorly understood, and despite the availability of biological medications blocking IL-1 and IL-6 that have markedly improved overall disease control, children with Systemic Juvenile Idiopathic Arthritis (SJIA) are now increasingly diagnosed with life-threatening chronic complications, including hepatitis and lung disease (SJIA-LD). On October 3-4, 2019, a two-day meeting, NextGen Therapies for Systemic Juvenile Idiopathic Arthritis (SJIA) & macrophage activation syndrome (MAS) organized by the Systemic JIA Foundation ( www.systemicjia.org/ ) in Washington, DC brought together scientists, clinicians, parents and FDA representatives with the objectives (1) to integrate clinical and research findings in MAS and SJIA-LD, and (2) to develop a shared understanding of this seemingly new pulmonary complication of SJIA. The current manuscript summarizes discussions and conclusions of the meeting.
Collapse
Affiliation(s)
| | - Grant S Schulert
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, USA.
| | - Adriana de Jesus
- Intramural Research Program, National Institute of Allergy and Infectious Diseases, NIH, North Bethesda, USA
| | | | - Hermine Brunner
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, USA
| | - Massimo Gadina
- Intramural Research Program, National Institute of Allergy and Infectious Diseases, NIH, North Bethesda, USA
| | - Stewart Levine
- Intramural Research Program, National Institute of Allergy and Infectious Diseases, NIH, North Bethesda, USA
| | - Raphaela Goldbach-Mansky
- Intramural Research Program, National Institute of Allergy and Infectious Diseases, NIH, North Bethesda, USA
| | | | | | | | - Alexei Grom
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, USA.
| |
Collapse
|
125
|
Ohmura SI, Uehara K, Yamabe T, Tamechika S, Maeda S, Naniwa T. Successful use of short-term add-on tocilizumab for refractory adult-onset still's disease with macrophage activation syndrome despite treatment with high-dose glucocorticoids, cyclosporine, and etoposide. Mod Rheumatol Case Rep 2020; 4:202-207. [PMID: 33086994 DOI: 10.1080/24725625.2020.1741073] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/07/2020] [Indexed: 06/11/2023]
Abstract
Macrophage activation syndrome (MAS) is a form of secondary hemophagocytic lymphohistiocytosis (HLH) and is a life-threatening complication of adult-onset Still disease. MAS has been usually treated with high-dose glucocorticoid with additional immunosuppressive agents, such as cyclosporine. Etoposide has been used for the treatment of severe refractory MAS based on the successful results of HLH-2004 protocol in patients with mostly primary form of HLH. We herein describe a case of severe refractory MAS secondary to adult-onset Still disease in an elderly woman that inadequately responded to etoposide but remarkably responded to additional tocilizumab. Furthermore, short-term tocilizumab led her into remission and enabled tapering off glucocorticoids after 15 months. Tocilizumab may be effective for the treatment of refractory HLH after the failure of the etoposide-containing induction regimen.
Collapse
Affiliation(s)
- Shin-Ichiro Ohmura
- Division of Rheumatology, Department of Internal Medicine, Nagoya City University Hospital, Nagoya, Japan
| | - Koji Uehara
- Division of Rheumatology, Department of Internal Medicine, Nagoya City University Hospital, Nagoya, Japan
| | - Toru Yamabe
- Division of Rheumatology, Department of Internal Medicine, Nagoya City University Hospital, Nagoya, Japan
| | - Shinya Tamechika
- Division of Rheumatology, Department of Internal Medicine, Nagoya City University Hospital, Nagoya, Japan
| | - Shinji Maeda
- Division of Rheumatology, Department of Internal Medicine, Nagoya City University Hospital, Nagoya, Japan
| | - Taio Naniwa
- Division of Rheumatology, Department of Internal Medicine, Nagoya City University Hospital, Nagoya, Japan
| |
Collapse
|
126
|
Parenchymal lung disease in adult onset Still's disease: an emergent marker of disease severity-characterisation and predictive factors from Gruppo Italiano di Ricerca in Reumatologia Clinica e Sperimentale (GIRRCS) cohort of patients. Arthritis Res Ther 2020; 22:151. [PMID: 32571407 PMCID: PMC7310010 DOI: 10.1186/s13075-020-02245-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/12/2020] [Indexed: 02/07/2023] Open
Abstract
Background Adult-onset Still’s disease (AOSD) is a systemic inflammatory disorder of unknown aetiology usually affecting young adults. Interestingly, recent evidence from the juvenile counterpart of AOSD suggested the emergent high fatality rate of lung disease (LD) in these patients. In this work, we aimed to characterise LD in AOSD, to identify associated clinical features and predictive factors, and to describe long-term outcomes of the disease comparing patients with LD and those without. Methods A retrospective assessment of prospectively followed patients, from January 2001 to December 2019, was provided to describe the rate of LD in AOSD, associated clinical features and predictive factors, and long-term outcomes. Patients with AOSD, who were included in Gruppo Italiano di Ricerca in Reumatologia Clinica e Sperimentale (GIRRCS) cohort, were assessed. Results Out of 147 patients included in GIRRCS cohort, 18 (12.25%) patients were reported to be affected by LD, at the time of diagnosis of AOSD, who were characterised by older age, a higher prevalence of myalgia, of lymph node involvement, of pleuritis, and abdominal pain. Furthermore, patients with LD showed higher values of systemic score and ferritin. Among those clinical variables, older age and systemic score were also independently predictors of LD. Chest CT scans were also obtained, and the most common finding was the peripheral consolidations in 8 (44.4%) patients. Finally, a higher mortality rate, of 38.9%, was registered in patients with LD than others, since it was associated with a significant decreased survival rate. Conclusions The presence of LD could suggest an emergent cause of mortality in AOSD, as observed in juvenile counterpart recognising a further marker of severity and poor prognosis to be careful evaluated. Patients with LD were also characterised by some clinical features, higher values of systemic score and ferritin than the others, identifying a subset of patients mostly burdened by systemic signs and symptoms. Although specific designed future studies are needed to fully elucidate the significance of LD in AOSD, a more accurate evaluation and management of this feature could improve the long-term outcomes of these patients.
Collapse
|
127
|
Gerges Harb J, Noureldine HA, Chedid G, Eldine MN, Abdallah DA, Chedid NF, Nour-Eldine W. SARS, MERS and COVID-19: clinical manifestations and organ-system complications: a mini review. Pathog Dis 2020; 78:ftaa033. [PMID: 32633327 PMCID: PMC7454523 DOI: 10.1093/femspd/ftaa033] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/03/2020] [Indexed: 12/21/2022] Open
Abstract
Middle East Respiratory Syndrome (MERS), Severe Acute Respiratory Syndrome (SARS) and Coronavirus Disease 2019 (COVID-19) are caused by three distinct coronaviruses belonging to the same genus. COVID-19 and its two predecessors share many important features in their clinical presentations, and in their propensity for progression to severe disease which is marked by high rates of morbidity and mortality. However, comparison of the three viral illnesses also reveals a number of specific differences in clinical manifestations and complications, which suggest variability in the disease process. This narrative review delineates the pulmonary, cardiac, renal, gastrointestinal, hepatic, neurological and hematologic complications associated with these three respiratory coronaviruses. It further describes the mechanisms of immune hyperactivation-particularly cytokine release syndrome-implicated in the multi-organ system injury seen in severe cases of MERS, SARS and COVID-19.
Collapse
Affiliation(s)
- Jad Gerges Harb
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Hussein A Noureldine
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Georges Chedid
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Mariam Nour Eldine
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Dany Abou Abdallah
- Lebanese University, Faculty of Medical Sciences, Rafik Hariri University Campus, Hadath, Lebanon
| | - Nancy Falco Chedid
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Wared Nour-Eldine
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| |
Collapse
|
128
|
Abstract
Systemic juvenile idiopathic arthritis (sJIA) is a severe disorder now linked more to autoinflammation than to autoimmunity. Clinical and laboratory evidence support the pathogenetic role of interleukin-1 (IL-1), and blockade of this cytokine has proved to be very effective in the treatment of sJIA. There are now several agents that block IL-1 available on the market. This article reviews the efficacy and safety of these drugs for the treatment of sJIA on the basis of published data, and offers the current view on treating sJIA according to its different phenotypes. There are no head to head trials among the different IL-1 inhibitors, and although efficacy has been demonstrated for all of them, it is still unknown which one would be more appropriate for which particular situation. The presence of synovitis in addition to active systemic features might be relevant for these choices. In addition, complications such as macrophage activation syndrome can be important since, on one hand, it has been associated with biologic therapy administration and, on another, there have been some reports of this complication being treated with anti-IL-1. Current recommendations by the American College of Rheumatology are now outdated, and new ones are being prepared. In the meantime, basic and clinical research is advancing in order to identify new treatment targets and to evaluate the different protocols currently in use.
Collapse
|
129
|
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: 11.6] [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
|
130
|
Alunno A, Carubbi F, Rodríguez-Carrio J. Storm, typhoon, cyclone or hurricane in patients with COVID-19? Beware of the same storm that has a different origin. RMD Open 2020; 6:rmdopen-2020-001295. [PMID: 32423970 PMCID: PMC7299508 DOI: 10.1136/rmdopen-2020-001295] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 12/24/2022] Open
Abstract
Some of the articles being published during the severe acute respiratory syndrome-coronavirus (SARS-CoV)-2 pandemic highlight a link between severe forms of coronavirus disease 2019 (COVID-19) and the so-called cytokine storm, also with increased ferritin levels. However, this scenario is more complex than initially thought due to the heterogeneity of hyperinflammation. Some patients with coronavirus 2019 disease (COVID-19) develop a fully blown secondary haemophagocytic lymphohistiocytosis (sHLH), whereas others, despite a consistent release of pro-inflammatory cytokines, do not fulfil sHLH criteria but still show some features resembling the phenotype of the hyperferritinemic syndrome. Despite the final event (the cytokine storm) is shared by various conditions leading to sHLH, the aetiology, either infectious, autoimmune or neoplastic, accounts for the differences in the various phases of this process. Moreover, the evidence of a hyperinflammatory microenvironment provided the rationale to employ immunomodulating agents for therapeutic purposes in severe COVID-19. This viewpoint aims at discussing the pitfalls and issues to be considered with regard to the use of immunomodulating agents in COVID-19, such as timing of treatment based on the viral load and the extent of cytokine/ferritin overexpression. Furthermore, it encompasses recent findings in the paediatric field about a novel multisystem inflammatory disease resembling toxic shock syndrome and atypical Kawasaki disease observed in children with proven SARS-CoV2 infection. Finally, it includes arguments in favour of adding COVID-19 to the spectrum of the recently defined 'hyperferritinemic syndrome', which already includes adult-onset Still's disease, macrophage activation syndrome, septic shock and catastrophic anti-phospholipid syndrome.
Collapse
Affiliation(s)
- Alessia Alunno
- Department of Medicine, Rheumatology Unit, University of Perugia, Perugia, Italy
| | - Francesco Carubbi
- Department of Biotechnological and Applied Clinical Sciences, Rheumatology Unit, University of L'Aquila, L'Aquila, Italy
| | - Javier Rodríguez-Carrio
- University of Oviedo, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| |
Collapse
|
131
|
SARS-CoV-2 Inflammatory Syndrome. Clinical Features and Rationale for Immunological Treatment. Int J Mol Sci 2020. [PMID: 32397684 DOI: 10.3390/ijms21093377.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The current pandemic coronavirus, SARS-CoV-2, is a global health emergency because of its highly contagious nature, the great number of patients requiring intensive care therapy, and the high fatality rate. In the absence of specific antiviral drugs, passive prophylaxis, or a vaccine, the treatment aim in these patients is to prevent the potent virus-induced inflammatory stimuli from leading to the acute respiratory distress syndrome (ARDS), which has a severe prognosis. Here, the mechanism of action and the rationale for employing immunological strategies, which range from traditional chemically synthesized drugs, anti-cytokine antibodies, human immunoglobulin for intravenous use, to vaccines, are reviewed.
Collapse
|
132
|
Prete M, Favoino E, Catacchio G, Racanelli V, Perosa F. SARS-CoV-2 Inflammatory Syndrome. Clinical Features and Rationale for Immunological Treatment. Int J Mol Sci 2020; 21:ijms21093377. [PMID: 32397684 PMCID: PMC7247005 DOI: 10.3390/ijms21093377] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 12/15/2022] Open
Abstract
The current pandemic coronavirus, SARS-CoV-2, is a global health emergency because of its highly contagious nature, the great number of patients requiring intensive care therapy, and the high fatality rate. In the absence of specific antiviral drugs, passive prophylaxis, or a vaccine, the treatment aim in these patients is to prevent the potent virus-induced inflammatory stimuli from leading to the acute respiratory distress syndrome (ARDS), which has a severe prognosis. Here, the mechanism of action and the rationale for employing immunological strategies, which range from traditional chemically synthesized drugs, anti-cytokine antibodies, human immunoglobulin for intravenous use, to vaccines, are reviewed.
Collapse
Affiliation(s)
- Marcella Prete
- Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Science and Human Oncology (DIMO), University of Bari Medical School, Piazza G. Cesare 11, I-70124 Bari, Italy; (M.P.); (E.F.); (G.C.)
| | - Elvira Favoino
- Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Science and Human Oncology (DIMO), University of Bari Medical School, Piazza G. Cesare 11, I-70124 Bari, Italy; (M.P.); (E.F.); (G.C.)
| | - Giacomo Catacchio
- Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Science and Human Oncology (DIMO), University of Bari Medical School, Piazza G. Cesare 11, I-70124 Bari, Italy; (M.P.); (E.F.); (G.C.)
| | - Vito Racanelli
- Unit of Internal Medicine, Department of Biomedical Sciences and Human Oncology (DIMO), University of Bari Medical School, Piazza G. Cesare 11, I-70124 Bari, Italy;
| | - Federico Perosa
- Systemic Rheumatic and Autoimmune Diseases Unit, Department of Biomedical Science and Human Oncology (DIMO), University of Bari Medical School, Piazza G. Cesare 11, I-70124 Bari, Italy; (M.P.); (E.F.); (G.C.)
- Correspondence: ; Tel.: +39-80-547-88-91; Fax: +39-80-547-88-20
| |
Collapse
|
133
|
Griffin G, Shenoi S, Hughes GC. Hemophagocytic lymphohistiocytosis: An update on pathogenesis, diagnosis, and therapy. Best Pract Res Clin Rheumatol 2020; 34:101515. [PMID: 32387063 DOI: 10.1016/j.berh.2020.101515] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hemophagocytic lymphohistiocytosis (HLH) is a rare, life-threatening state of immune hyperactivation that arises in the setting of genetic mutations and infectious, inflammatory, or neoplastic triggers. Sustained, aberrant activation of cytotoxic CD8+ T cells and resultant inflammatory cytokine release are core pathogenic mechanisms. Key clinical features include high persistent fever, hepatosplenomegaly, blood cytopenia, elevated aminotransferase and ferritin levels, and coagulopathy. HLH is likely under-recognized, and mortality remains high, especially in adults; thus, prompt diagnosis and treatment are essential. Familial forms of HLH are currently treated with chemotherapy as a bridge to hematopoietic stem cell transplantation. HLH occurring in rheumatic disease (macrophage activation syndrome) is treated with glucocorticoids, IL-1 blockade, or cyclosporine A. In other forms of HLH, addressing the underlying trigger is essential. There remains a pressing need for more sensitive, context-specific diagnostic tools. Safer, more effective therapies will arise with improved understanding of the cellular and molecular mechanisms of HLH.
Collapse
Affiliation(s)
- Georgia Griffin
- Division of Rheumatology, Seattle Children's Hospital, Seattle, WA, USA.
| | - Susan Shenoi
- Division of Rheumatology, Seattle Children's Hospital, Seattle, WA, USA
| | - Grant C Hughes
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| |
Collapse
|
134
|
Henderson LA, Hoyt KJ, Lee PY, Rao DA, Jonsson AH, Nguyen JP, Rutherford K, Julé AM, Charbonnier LM, Case S, Chang MH, Cohen EM, Dedeoglu F, Fuhlbrigge RC, Halyabar O, Hazen MM, Janssen E, Kim S, Lo J, Lo MS, Meidan E, Son MBF, Sundel RP, Stoll ML, Nusbaum C, Lederer JA, Chatila TA, Nigrovic PA. Th17 reprogramming of T cells in systemic juvenile idiopathic arthritis. JCI Insight 2020; 5:132508. [PMID: 32213704 DOI: 10.1172/jci.insight.132508] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 02/26/2020] [Indexed: 12/21/2022] Open
Abstract
Systemic juvenile idiopathic arthritis (sJIA) begins with fever, rash, and high-grade systemic inflammation but commonly progresses to a persistent afebrile arthritis. The basis for this transition is unknown. To evaluate a role for lymphocyte polarization, we characterized T cells from patients with acute and chronic sJIA using flow cytometry, mass cytometry, and RNA sequencing. Acute and chronic sJIA each featured an expanded population of activated Tregs uncommon in healthy controls or in children with nonsystemic JIA. In acute sJIA, Tregs expressed IL-17A and a gene expression signature reflecting Th17 polarization. In chronic sJIA, the Th17 transcriptional signature was identified in T effector cells (Teffs), although expression of IL-17A at the protein level remained rare. Th17 polarization was abrogated in patients responding to IL-1 blockade. These findings identify evolving Th17 polarization in sJIA that begins in Tregs and progresses to Teffs, likely reflecting the impact of the cytokine milieu and consistent with a biphasic model of disease pathogenesis. The results support T cells as a potential treatment target in sJIA.
Collapse
Affiliation(s)
- Lauren A Henderson
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kacie J Hoyt
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Pui Y Lee
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, and
| | - Deepak A Rao
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, and
| | - A Helena Jonsson
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, and
| | - Jennifer P Nguyen
- Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Kayleigh Rutherford
- Harvard Bioinformatics Core, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Amélie M Julé
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Louis-Marie Charbonnier
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Siobhan Case
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Margaret H Chang
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, and
| | - Ezra M Cohen
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Fatma Dedeoglu
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert C Fuhlbrigge
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Rheumatology, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Olha Halyabar
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Melissa M Hazen
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Erin Janssen
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Susan Kim
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeffrey Lo
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mindy S Lo
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Esra Meidan
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mary Beth F Son
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert P Sundel
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew L Stoll
- Division of Pediatric Rheumatology, Department of Pediatrics, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Chad Nusbaum
- Broad Technology Labs, Broad Institute, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA
| | - James A Lederer
- Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Talal A Chatila
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Peter A Nigrovic
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, and
| |
Collapse
|
135
|
Could Sars-coronavirus-2 trigger autoimmune and/or autoinflammatory mechanisms in genetically predisposed subjects? Autoimmun Rev 2020; 19:102524. [PMID: 32220633 PMCID: PMC7271072 DOI: 10.1016/j.autrev.2020.102524] [Citation(s) in RCA: 224] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 03/23/2020] [Indexed: 12/04/2022]
|
136
|
|