1
|
Chen F, Ye W, Wang Q, Zhao L, Liang M, Zheng S, Zhao T, Xuan D, Zhu Z, Yu Y, Kong N, Jiang L, Yang X, Zhu X, Wan W, Zou H, Xue Y. BAricitinib in patients with SystemIC Sclerosis (BASICS): a prospective, open-label, randomised trial. Clin Rheumatol 2025:10.1007/s10067-025-07433-9. [PMID: 40381085 DOI: 10.1007/s10067-025-07433-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/27/2025] [Accepted: 04/07/2025] [Indexed: 05/19/2025]
Abstract
BACKGROUND AND AIMS Despite advances in approaches to treatment for patients with systemic sclerosis (SSc), the effects have been modest at best. This investigator-initiated study aimed to evaluate the therapeutic benefit, safety and the genetic characteristics related to effect of baricitinib in SSc. METHODS In this 24-week study, eligible SSc patients were randomised to baricitinib 4 mg, 2 mg or control group. The primary outcome was the change in modified Rodnan skin score (mRSS) from baseline to week 12. Secondary outcomes included changes in the American College of Rheumatology Combined Response Index in Systemic Sclerosis (ACR-CRISS) score, forced vital capacity (FVC), Systemic Sclerosis Score, tender and swollen joint counts, digital ulcers, EQ5D (EuroQol five-dimensions) and safety at week 12 and 24. Transcriptome differences in blood samples from patients before and after baricitinib treatment were compared. Gene Ontology enrichment analysis was performed to identify potential biological functions and canonical pathways. RESULTS Between April 2021 to January 2022, 48 patients were randomly assigned to three groups. Mean change in mRSS score from baseline to week 12 was - 8.9 in 4 mg group, - 3.8 in 2 mg group, and - 3.6 in control group (P = 0.019). At week 12, the ACR-CRISS scores were 0.5 and 0.3 in baricitinib 4 mg and 2 mg group, as compared with 0.2 among those in control group (P = 0.171). FVC (%), digital ulcers and EQ5D in 4 mg baricitinib group showed favorable responses over 24 weeks. There were no significant differences in adverse events among groups. The differentially expressed genes (DEGs) identified in our analysis were significantly enriched in gene ontology (GO) terms related to the positive regulation of cytokine production, immune response-activating signaling pathway, and activation of immune response pathway. Interleukin- 1 Receptor Like 1 (IL- 1RL1 or ST2) and synaptotagmin- 17 (SYT17) were downregulated and upregulated respectively, after baricitinib treatment. CONCLUSIONS The therapy with baricitinib 4 mg appeared to improve mRSS of SSc patients, probably by influencing mechanisms of immune inflammation, and had an acceptable safety profile. This study paves the way for further investigations into Janus kinase (JAK) 1/2 inhibition with baricitinib as a prospective treatment for SSc. Key Points • The baricitinib 4 mg seems to improve clinical outcomes of SSc patients with safety profiles. • The mechanism of JAK inhibitors has been confirmed to be related to anti-inflammatory pathways and molecules in clinical samples.
Collapse
Affiliation(s)
- Fangfang Chen
- Department of Rheumatology, Huashan Hospital, Institute of Rheumatology, Immunology and Allergy, Fudan University, No. 12, Middle Urumqi Road, Jing'an District, Shanghai, 200000, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- International Network of Scleroderma Clinic and Research, Shandong, China
| | - Wenjing Ye
- Department of Rheumatology, Huashan Hospital, Institute of Rheumatology, Immunology and Allergy, Fudan University, No. 12, Middle Urumqi Road, Jing'an District, Shanghai, 200000, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- International Network of Scleroderma Clinic and Research, Shandong, China
| | - Qian Wang
- Department of Rheumatology, Huashan Hospital, Institute of Rheumatology, Immunology and Allergy, Fudan University, No. 12, Middle Urumqi Road, Jing'an District, Shanghai, 200000, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- International Network of Scleroderma Clinic and Research, Shandong, China
| | - Li Zhao
- Department of Rheumatology, Huashan Hospital, Institute of Rheumatology, Immunology and Allergy, Fudan University, No. 12, Middle Urumqi Road, Jing'an District, Shanghai, 200000, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- International Network of Scleroderma Clinic and Research, Shandong, China
| | - Minrui Liang
- Department of Rheumatology, Huashan Hospital, Institute of Rheumatology, Immunology and Allergy, Fudan University, No. 12, Middle Urumqi Road, Jing'an District, Shanghai, 200000, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- International Network of Scleroderma Clinic and Research, Shandong, China
| | - Shucong Zheng
- Department of Rheumatology, Huashan Hospital, Institute of Rheumatology, Immunology and Allergy, Fudan University, No. 12, Middle Urumqi Road, Jing'an District, Shanghai, 200000, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- International Network of Scleroderma Clinic and Research, Shandong, China
| | - Tianyi Zhao
- Department of Rheumatology, Huashan Hospital, Institute of Rheumatology, Immunology and Allergy, Fudan University, No. 12, Middle Urumqi Road, Jing'an District, Shanghai, 200000, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- International Network of Scleroderma Clinic and Research, Shandong, China
| | - Dandan Xuan
- Department of Rheumatology, Huashan Hospital, Institute of Rheumatology, Immunology and Allergy, Fudan University, No. 12, Middle Urumqi Road, Jing'an District, Shanghai, 200000, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- International Network of Scleroderma Clinic and Research, Shandong, China
| | - Zaihua Zhu
- Department of Rheumatology, Huashan Hospital, Institute of Rheumatology, Immunology and Allergy, Fudan University, No. 12, Middle Urumqi Road, Jing'an District, Shanghai, 200000, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- International Network of Scleroderma Clinic and Research, Shandong, China
| | - Yiyun Yu
- Department of Rheumatology, Huashan Hospital, Institute of Rheumatology, Immunology and Allergy, Fudan University, No. 12, Middle Urumqi Road, Jing'an District, Shanghai, 200000, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- International Network of Scleroderma Clinic and Research, Shandong, China
| | - Ning Kong
- Department of Rheumatology, Huashan Hospital, Institute of Rheumatology, Immunology and Allergy, Fudan University, No. 12, Middle Urumqi Road, Jing'an District, Shanghai, 200000, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- International Network of Scleroderma Clinic and Research, Shandong, China
| | - Li Jiang
- International Network of Scleroderma Clinic and Research, Shandong, China
- Department of Rheumatology, Linyi People's Hospital, Shandong, China
| | - Xue Yang
- Department of Rheumatology, Huashan Hospital, Institute of Rheumatology, Immunology and Allergy, Fudan University, No. 12, Middle Urumqi Road, Jing'an District, Shanghai, 200000, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- International Network of Scleroderma Clinic and Research, Shandong, China
| | - Xiaoxia Zhu
- Department of Rheumatology, Huashan Hospital, Institute of Rheumatology, Immunology and Allergy, Fudan University, No. 12, Middle Urumqi Road, Jing'an District, Shanghai, 200000, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- International Network of Scleroderma Clinic and Research, Shandong, China
| | - Weiguo Wan
- Department of Rheumatology, Huashan Hospital, Institute of Rheumatology, Immunology and Allergy, Fudan University, No. 12, Middle Urumqi Road, Jing'an District, Shanghai, 200000, China.
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China.
- International Network of Scleroderma Clinic and Research, Shandong, China.
- Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China.
| | - Hejian Zou
- Department of Rheumatology, Huashan Hospital, Institute of Rheumatology, Immunology and Allergy, Fudan University, No. 12, Middle Urumqi Road, Jing'an District, Shanghai, 200000, China.
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China.
- International Network of Scleroderma Clinic and Research, Shandong, China.
- Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China.
| | - Yu Xue
- Department of Rheumatology, Huashan Hospital, Institute of Rheumatology, Immunology and Allergy, Fudan University, No. 12, Middle Urumqi Road, Jing'an District, Shanghai, 200000, China.
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China.
- International Network of Scleroderma Clinic and Research, Shandong, China.
- Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Rujirawinitchai P, Foocharoen C, Mahakkanukrauh A, Suwannaroj S, Pongkulkiat P, Onchan T. Correlation Between the Peak of Skin Thickness Progression Rate and Onset of Cardiopulmonary Involvement in Thai Systemic Sclerosis Patients. J Clin Med 2025; 14:2281. [PMID: 40217731 PMCID: PMC11989290 DOI: 10.3390/jcm14072281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: Rapid skin thickness progression assessed using the modified Rodnan skin score (mRSS) is associated with poor outcomes in systemic sclerosis (SSc). However, the correlation between patterns of skin thickness and the onset of internal organ involvement remains unclear. This study aimed to determine the correlation between peak skin thickness progression rate (pSTPR) and the onset of internal organ involvement, particularly interstitial lung disease (ILD) and pulmonary arterial hypertension (PAH) among Thai SSc patients. Method: A prospective cohort study with retrospective analysis was conducted on adult SSc patients who experienced the onset of their first non-Raynaud phenomenon symptoms between January 2013 and December 2020 and had at least a 2-year follow-up. Patients with an overlap syndrome were excluded from this study. The pSTPR was calculated by dividing the peak of the mRSS by the duration of disease at the peak of the mRSS. Result: A total of 509 patients were included in this study. The majority of cases were female (351; 69.0%) and comprised diffuse cutaneous SSc subsets (353 cases; 69.4%). The respective mean age and median pSTPR was 48.2 ± 11.6 years and 1.63 points/year (interquartile range 0.5-4.4). The respective median durations of disease at the onset of significant ILD (>20% extent), ILD, and mean duration of disease at the onset of PAH were 3.4 (Q1-Q3 1.4-7.7), 5.4 (Q1-Q3 2.4-9.2), and 8.0 ± 4.9 years. pSTPR was negatively correlated with disease duration at the onset of significant ILD (Rho -0.509, p < 0.001), ILD (Rho -0.480, p < 0.001), PAH (Rho -0.372, p = 0.03), and disease duration from onset to death (Rho -0.367, p = 0.03) after adjusting for age, sex, and anti-topoisomerase I. Conclusion: SSc patients with a high skin thickness progression rate reaching the maximum point of mRSS were at risk of developing early ILD, PAH, and death. The pSTPR may be used to assess individuals at risk of experiencing early onset cardiopulmonary involvement in SSc.
Collapse
Affiliation(s)
| | | | | | | | | | - Tippawan Onchan
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (P.R.)
| |
Collapse
|
3
|
Odell ID. Cross-tissue organization of myeloid cells in scleroderma and related fibrotic diseases. Curr Opin Rheumatol 2024; 36:379-386. [PMID: 39171604 PMCID: PMC11451931 DOI: 10.1097/bor.0000000000001047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
PURPOSE OF REVIEW Scleroderma and other fibrotic diseases have been investigated using single-cell RNA sequencing (scRNA-Seq), which has demonstrated enrichment in myeloid cell populations in multiple tissues. However, scRNA-Seq studies are inconsistent in their nomenclature of myeloid cell types, including dendritic cells, monocytes, and macrophages. Using cell type-defining gene signatures, I propose a unified nomenclature through analysis of myeloid cell enrichment across fibrotic tissues. RECENT FINDINGS scRNA-Seq of human blood and skin identified a new subset of dendritic cells called DC3. DC3 express similar inflammatory genes to monocytes, including FCN1 , IL1B, VCAN, S100A8, S100A9 , and S100A12 . DC3 can be distinguished from monocytes by expression of EREG and Fc receptor genes such as FCER1A and FCGR2B . scRNA-Seq analyses of scleroderma skin and lung, idiopathic pulmonary fibrosis (IPF), COVID-19 lung fibrosis, myelofibrosis, and liver, kidney, and cardiac fibrosis all showed enrichment in myeloid cell types. Although they were called different names, studies of scleroderma skin and lung as well as liver cirrhosis datasets demonstrated enrichment in DC3. By contrast, lung, heart, and kidney fibrosis were enriched in SPP1 macrophages. High numbers of DC3 in the skin was associated with worse SSc skin and lung fibrosis severity. SUMMARY scRNA-Seq of multiple diseases showed enrichment of DC3 in fibrotic skin, lung, and liver, whereas SPP1 macrophages occurred in fibrotic lung, heart, and kidney. Because DC3 and SPP1 macrophages showed organ-specific enrichment, understanding their signaling mechanisms across tissues will be important for future investigation.
Collapse
Affiliation(s)
- Ian D. Odell
- Department of Dermatology
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
4
|
Matsuda KM, Kotani H, Yamaguchi K, Ono C, Okumura T, Ogawa K, Miya A, Sato A, Uchino R, Yumi M, Matsunaka H, Kono M, Norimatsu Y, Hisamoto T, Kawanabe R, Kuzumi A, Fukasawa T, Yoshizaki-Ogawa A, Okamura T, Shoda H, Fujio K, Matsushita T, Goshima N, Sato S, Yoshizaki A. Autoantibodies to nuclear valosin-containing protein-like protein: systemic sclerosis-specific antibodies revealed by in vitro human proteome. Rheumatology (Oxford) 2024; 63:2865-2873. [PMID: 38290780 DOI: 10.1093/rheumatology/keae063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/29/2023] [Accepted: 01/06/2024] [Indexed: 02/01/2024] Open
Abstract
OBJECTIVES To identify and characterize undescribed systemic sclerosis (SSc)-specific autoantibodies targeting nucleolar antigens and to assess their clinical significance. METHODS We conducted proteome-wide autoantibody screening (PWAS) against serum samples from SSc patients with nucleolar patterned anti-nuclear antibodies (NUC-ANAs) of specific antibodies (Abs) unknown, utilizing wet protein arrays fabricated from in vitro human proteome. Controls included SSc patients with already-known SSc-specific autoantibodies, patients with other connective tissue diseases and healthy subjects. The selection of nucleolar antigens was performed by database search in the Human Protein Atlas. The presence of autoantibodies was certified by immunoblots and immunoprecipitations. Indirect immunofluorescence assays on HEp-2 cells were also conducted. Clinical assessment was conducted by retrospective review of electronic medical records. RESULTS PWAS identified three candidate autoantibodies, including anti-nuclear valosin-containing protein-like (NVL) Ab. Additional measurements in disease controls revealed that only anti-NVL Abs are exclusively detected in SSc. Detection of anti-NVL Abs was reproduced by conventional assays such as immunoblotting and immunoprecipitation. Indirect immunofluorescence assays demonstrated homogeneous nucleolar patterns. Anti-NVL Ab-positive cases were characterized by significantly low prevalence of diffuse skin sclerosis and interstitial lung disease, compared with SSc cases with NUC-ANAs other than anti-NVL Abs, such as anti-U3-RNP and anti-Th/To Abs. CONCLUSION Anti-NVL Ab is an SSc-specific autoantibody associated with a unique combination of clinical features, including limited skin sclerosis and lack of lung involvement.
Collapse
Affiliation(s)
- Kazuki M Matsuda
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Hirohito Kotani
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | - Rikako Uchino
- NOV Academic Research, TOKIWA Pharmaceutical Co., Ltd, Tokyo, Japan
| | - Murakami Yumi
- NOV Academic Research, TOKIWA Pharmaceutical Co., Ltd, Tokyo, Japan
| | | | - Masanori Kono
- Department of Allergy and Rheumatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Yuta Norimatsu
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Teruyoshi Hisamoto
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Ruriko Kawanabe
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Ai Kuzumi
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takemichi Fukasawa
- Department of Clinical Cannabinoid Research, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Asako Yoshizaki-Ogawa
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Tomohisa Okamura
- Department of Allergy and Rheumatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Hirofumi Shoda
- Department of Allergy and Rheumatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takashi Matsushita
- Department of Dermatology, Kanazawa University Graduate School of Medical Sciences, Ishikawa, Japan
| | | | - Shinichi Sato
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Ayumi Yoshizaki
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
- Department of Clinical Cannabinoid Research, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
5
|
Werakiat J, Pussadhamma B, Mahakkanukrauh A, Suwannaroj S, Foocharoen C. Clinical courses and predictors of left ventricular systolic dysfunction in systemic sclerosis: A cohort study. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2024; 5:107-116. [PMID: 39015847 PMCID: PMC11248551 DOI: 10.1515/rir-2024-0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 05/19/2024] [Indexed: 07/18/2024]
Abstract
Background and Objectives Left ventricular systolic dysfunction (LVSD) is a cardiac involvement that is the leading cause of death among patients with systemic sclerosis (SSc). We aimed to define the clinical course and predictors of LVSD among SSc patients. Methods We conducted a cohort study among adult patients with SSc who were followed up from 2013 to 2020. Semiparametric Cox regression analysis with robust clustering by cohort identification number was used to evaluate the predictors of LVSD. Results Among the 3, 987 person-years, LVSD was defined in 35 of 419 SSc patients for an incidence of 0.88 per 100 person-years. The median duration of the disease was 8.5 (interquartile range (IQR) 4.9-12.9) years. Every 1-point increase in the modified Rodnan skin score (mRSS) and salt and pepper skin were strong predictors of LVSD, with a respective adjusted hazard ratio (HR) of 1.05 and 3.17. During follow-up, 26 cases (74.3%) had unimproved LVSD. The strong predictors of the unimprovement of LVSD were every 1-point increase in mRSS (HR 1.05), every 1 mg increase in prednisolone treatment (HR 1.05), and every 1 U/L increase in creatine kinase (CK) (HR 1.001). Mycophenolate treatment was a protective factor against the unimprovement of LVSD in SSc (HR 0.15). Conclusions LVSD was frequently found in patients with diffuse cutaneous SSc, and in most cases, it remained unimproved during follow-up. High mRSS, steroid use, and high CK levels were predictors of unimproved LVSD, whereas mycophenolate treatment might prevent the progression of LVSD. Steroids should be prescribed with caution in patients with longer disease duration.
Collapse
Affiliation(s)
- Jakrapan Werakiat
- Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Burabha Pussadhamma
- Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Ajanee Mahakkanukrauh
- Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Siraphop Suwannaroj
- Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Chingching Foocharoen
- Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
6
|
Matsuda KM, Sugimoto E, Ako Y, Kitamura M, Miyahara M, Kotani H, Norimatsu Y, Hisamoto T, Kuzumi A, Fukasawa T, Sato S, Yoshizaki A. Reliability, validity, and sensitivity of the Japanese version of the University of California Los Angeles scleroderma clinical trial consortium gastrointestinal tract instrument: Application to efficacy assessment of intravenous immunoglobulin administration. J Dermatol 2024; 51:741-751. [PMID: 38558171 PMCID: PMC11483899 DOI: 10.1111/1346-8138.17202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/23/2024] [Accepted: 03/12/2024] [Indexed: 04/04/2024]
Abstract
This study aimed to develop and assess the reliability, validity, and sensitivity of the Japanese version of the University of California Los Angeles Scleroderma Clinical Trial Consortium gastrointestinal tract (GIT) Instrument 2.0 (the GIT score), as an evaluation tool for GIT symptoms in systemic sclerosis (SSc). The Japanese version of the GIT score was constructed using the forward-backward method. The reliability and validity of this instrument were evaluated in a cohort of 38 SSc patients. Correlation analysis was conducted to assess the relationship between the GIT score and existing patient-reported outcome measures. Additionally, the sensitivity of the GIT score was examined by comparing GIT scores before and after intravenous immunoglobulin (IVIG) administration in 10 SSc-myositis overlap patients, as IVIG has recently demonstrated effectiveness in alleviating GIT symptoms of SSc. As a result, the Japanese version of the GIT score exhibited internal consistency and a significant association with the Frequency Scale for the Symptoms of Gastroesophageal Reflux Disease. Furthermore, the total GIT score, as well as the reflux and distention/bloating subscales, displayed moderate correlations with the EuroQol 5 dimensions (EQ-5D) pain/discomfort subscale and the Short Form-36 body pain subscale. Notably, following IVIG treatment, there was a statistically significant reduction in the total GIT score and multiple subscales. We first validated the Japanese version of the GIT score in Japanese SSc patients in real-world clinical settings. This instrument holds promise for application in future clinical trials involving this patient population.
Collapse
Affiliation(s)
- Kazuki M. Matsuda
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Eiki Sugimoto
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Yoshiaki Ako
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Marie Kitamura
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Mai Miyahara
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Hirohito Kotani
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Yuta Norimatsu
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Teruyoshi Hisamoto
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Ai Kuzumi
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Takemichi Fukasawa
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
- Department of Clinical Cannabinoid Research, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Shinichi Sato
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Ayumi Yoshizaki
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
- Department of Clinical Cannabinoid Research, Graduate School of MedicineThe University of TokyoTokyoJapan
| |
Collapse
|
7
|
Hong DR, Huang CY, Xu ZH. Evaluating Skin Involvement in Systemic Sclerosis Using High-Frequency Ultrasound and Virtual Touch Tissue Imaging. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:536-539. [PMID: 38233292 DOI: 10.1016/j.ultrasmedbio.2023.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/12/2023] [Accepted: 12/18/2023] [Indexed: 01/19/2024]
Abstract
OBJECTIVE This study aimed to explore the diagnostic significance of high-frequency ultrasound combined with visual touch tissue imaging quantification (VTIQ) in the diagnosis and management of systemic sclerosis (SSc). METHODS Patients diagnosed with SSc and normal volunteers were recruited and divided into an experimental group and a control group, with 30 cases in each group, respectively. The skin thickness at six sites was assessed using high-frequency ultrasound, and the shear wave velocity (SWV) was determined using the VTIQ method. The differences in skin thickness and SWV between the experimental group and the control group were compared and a receiver operating characteristic (ROC) curve was plotted. The value of high-frequency ultrasound, VTIQ, and high-frequency ultrasound combined with VTIQ for evaluating skin involvement in SSc was determined. RESULTS The difference in SWV sum at six sites and the thickness sum was statistically significant (all p = 0.000 < 0.05) from that of the control group, and there was a strong association between the SWV sum, thickness sum, and Rodnan skin score at the six sites in the experimental group (p = 0.000, r = 0.726; p = 0.000, r = 0.679). Based on the ROC curve, the area under the curve (AUC) for high-frequency ultrasound examination was 0.789. The AUC for VTIQ examination was 0.893, while the AUC for high-frequency ultrasound combined with VTIQ examination was 0.923. The combined examination method showed the highest AUC, indicating the best diagnostic performance. CONCLUSION The integration of high-frequency ultrasound and VTIQ provides a quantitative approach for assessing the extent of skin involvement in SSc patients, offering valuable insights for clinical diagnosis and treatment purposes.
Collapse
Affiliation(s)
- Dao-Rong Hong
- Department of Ultrasonography, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Chun-Yan Huang
- Department of General Practice, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Zhen-Hong Xu
- Department of Ultrasonography, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China.
| |
Collapse
|
8
|
Campochiaro C, Allanore Y, Braun-Moscovici Y, Matucci-Cerinic M, Balbir-Gurman A. Is cyclophosphamide still the gold standard in early severe rapidly progressive systemic sclerosis? Autoimmun Rev 2024; 23:103439. [PMID: 37690478 DOI: 10.1016/j.autrev.2023.103439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 08/28/2023] [Indexed: 09/12/2023]
Abstract
Cyclophosphamide (CYC) has been a gold standard of treatment for severe progressive Systemic Sclerosis (SSc), especially in patients with concomitant interstitial lung disease (ILD). This approach was based on results of several interventional studies, including randomized control trials, which mainly addressed SSc-ILD as a primary end point and skin involvement as a second one. The use of CYC is time-limited due to significant adverse events. More recently, other immunosuppressive and biological agents showed efficacy but better safety profile in patients with SSc and SSc-ILD. With regards to other end-points, post-hoc analyses, systematic reviews and metalysis showed that CYC had limited influence on patients' quality of life, event-free survival and mortality. Comprehensive patient's stratification according to a molecular, cellular and phenotypic pattern may help in choosing of personalized medicine with more ambitious treatment effect and should be the future direction. According to the above available data and even if scientific evidence may be missing, experts' opinion has changed the attitude to CYC as an anchor drug in the management of severe SSc. Indeed, CYC has been pushed to the second and even third treatment option after mycophenolate mofetil, tocilizumab or rituximab. This position became obvious during debate on this topic at CORA meeting 2023.
Collapse
Affiliation(s)
- Corrado Campochiaro
- IRCCS San Raffaele Hospital, Unit of Immunology, Rheumatology, Allergy and Rare Diseases; Vita-Salute San Raffaele University, Milan, Italy
| | - Yannick Allanore
- Service de Rhumatologie, Hôpital Cochin, Université de Paris, Paris, France
| | - Yolanda Braun-Moscovici
- Rheumatology Institute, Rambam Health Care Campus, Haifa, Israel; The Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Rheumatology, Univercity of Florence, Italy; Unit of Immunology, Rheumatology, Allergy and Rare Diseases; Vita-Salute San Raffaele Univercity, Milan, Italy
| | - Alexandra Balbir-Gurman
- Rheumatology Institute, Rambam Health Care Campus, Haifa, Israel; The Rappaport Faculty of Medicine, Technion, Haifa, Israel.
| |
Collapse
|
9
|
Junfei Z, Meihua G, Shuai Z, Xiangting L, Zhidan L, Tianming C, Yajing L, Chu T, Lipu S. Retrospective comparative study of the efficacy of JAK inhibitor (tofacitinib) in the treatment of systemic sclerosis-associated interstitial lung disease. Clin Rheumatol 2023; 42:2823-2832. [PMID: 37335409 DOI: 10.1007/s10067-023-06660-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 04/05/2023] [Accepted: 06/01/2023] [Indexed: 06/21/2023]
Abstract
The oral Janus kinases inhibitor (JAKi) has improved the management of skin manifestations in systemic sclerosis (SSc), and our study aimed to explore the efficacy of non-selective JAKi tofacitinib in ameliorating interstitial lung disease (ILD) in the patients with SSc. The hospitalization data of the SSc-ILD patients from April 2019 to April 2021 were collected, and the changes of pulmonary function and the radiological findings in pulmonary high-resolution CT (HRCT) from the 9 patients who received tofacitinib for at least 6 months and a matched group of 35 SSc-ILD patients treated with conventional immunosuppressants or glucocorticoids, were compared and analyzed. There were no significant differences in demographic data and clinical characteristics between the tofacitinib-treated group (tofa-group) and the matched group. However, in the tofa-group, the changes in serum lactate dehydrogenase (LDH) concentration and serum interleukin-6 levels were significantly lower than those in the matched group. Moreover, the tofa-group showed amelioration in decreased diffusing capacity of the lung for carbon monoxide (DLCO) (62.05 ± 9.47 vs. 66.61 ± 12.39, p = 0.046), reductions in ground-glass attenuation involvement (1.00 ± 0.86 vs. 0.33 ± 0.50, p = 0.024) and irregular pleural thickening (1.33 ± 0.50 vs. 0.67 ± 0.51, p = 0.004) in pulmonary HRCTs, alleviated modified Rodnan skin score (mRSS) of skin sclerosis (9.22 ± 3.81 vs. 7.11 ± 3.92, p = 0.048), and reduced HRCT scores of pulmonary fibrosis (15.00 ± 3.87 vs. 12.66 ± 4.92, p = 0.009). Logistic regression analysis showed that the involvement of ground-glass attenuation (OR 11.43) and the add-on therapy of tofacitinib (OR 9.98) were the relevant factors in the amelioration of HRCT. Our results indicate that the use of JAKi (tofacitinib) may be relevant to significant improvement of the sclerosis and early radiological abnormalities in SSc-ILD patients. Further studies are needed to confirm these findings and to explore its efficacy more precisely. Key Points • The currently available therapies for SSc-ILD have limited therapeutic benefits. • The add-on therapy of the oral JAK inhibitor is available in the real world. • The tofacitinib was promising in the improvement of the sclerosis and early radiological abnormalities in SSc-ILD patients.
Collapse
Affiliation(s)
- Zhou Junfei
- Department of Rheumatology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, No. 7, Weiwu Road, Zhengzhou, 450000, China.
| | - Gao Meihua
- Department of Geriatrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Zhang Shuai
- Department of Dermatology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Lu Xiangting
- Department of Dermatology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Lei Zhidan
- Department of Radiology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Cheng Tianming
- Department of Radiology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Liu Yajing
- Department of Respiratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Tianshu Chu
- Department of Rheumatology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, No. 7, Weiwu Road, Zhengzhou, 450000, China.
| | - Shi Lipu
- Department of Rheumatology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, No. 7, Weiwu Road, Zhengzhou, 450000, China
| |
Collapse
|
10
|
Kwon OC, Park MC, Kim YG. Interleukin-32 as a biomarker in rheumatic diseases: A narrative review. Front Immunol 2023; 14:1140373. [PMID: 36875066 PMCID: PMC9974820 DOI: 10.3389/fimmu.2023.1140373] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Interleukin-32 (IL-32) is an important cytokine involved in the innate and adaptive immune responses. The role of IL-32 has been studied in the context of various diseases. A growing body of research has investigated the role of IL-32 in rheumatic diseases including inflammatory arthritides (rheumatoid arthritis, ankylosing spondylitis, and psoriatic arthritis) and connective tissue diseases (systemic lupus erythematosus, systemic sclerosis, granulomatosis and polyangiitis, and giant cell arteritis). IL-32 has been shown to play different roles according to the type of rheumatic diseases. Hence, the putative role of IL-32 as a biomarker is also different in each rheumatic disease: IL-32 could serve as a biomarker for disease activity in some diseases, whereas in other diseases it could be a biomarker for certain disease manifestations. In this narrative review, we summarize the associations between IL-32 and various rheumatic diseases and discuss the putative role of IL-32 as a biomarker in each disease.
Collapse
Affiliation(s)
- Oh Chan Kwon
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min-Chan Park
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Gil Kim
- Division of Rheumatology, Department of Internal Medicine, University of Ulsan, College of Medicine, Asan Medical Center, Seoul, Republic of Korea
- Convergence Medicine Research Center, Asan Institution for Life Science, Asan Medical Center, Seoul, Republic of Korea
| |
Collapse
|
11
|
Sredojevic SI, Dolijanovic SP, Dozic I, Pficer JK, Aleksic Z, Nikolic-Jakoba NS. Salivary Antioxidant Profile in Patients with Systemic Sclerosis and Periodontitis. Mediators Inflamm 2023; 2023:7886272. [PMID: 37138665 PMCID: PMC10151151 DOI: 10.1155/2023/7886272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/26/2022] [Accepted: 11/24/2022] [Indexed: 05/05/2023] Open
Abstract
Objective The aim of the present study was to compare periodontal status and antioxidant profile in unstimulated saliva of systemic sclerosis (SSc) patients with periodontitis and systemically healthy periodontitis patients. Design Twenty patients with established diagnoses of systemic sclerosis and periodontitis (SSc group) and 20 systemically healthy individuals with periodontitis (P group) were enrolled in the study. Clinical periodontal parameters (clinical attachment level (CAL), gingival recession (GR), periodontal probing depth (PPD), and gingival index (GI)) and concentration of uric acid (UA), superoxide dismutase (SOD), and glutathione peroxidase (GPX) in unstimulated saliva samples were assessed. Results There were significantly higher mean values of CAL (4.8 ± 0.21 mm versus 3.18 ± 0.17 mm; p ≤ 0.001) and GR (1.66 ± 0.90 mm versus 0.46 ± 0.54 mm; p ≤ 0.001) in the SSc group when compared to the P group. Significantly higher level of GPX (p ≤ 0.001) and SOD (p ≤ 0.001) in unstimulated saliva was detected in the SSc group in comparison with the P group. The specific activity of UA did not significantly differ between the two groups (p = 0.083). Conclusion The results may indicate higher periodontal destruction and antioxidant perturbations in unstimulated saliva of SSc patients with periodontitis compared to systemically healthy periodontitis patients.
Collapse
Affiliation(s)
- Stefan I. Sredojevic
- Department of Periodontology, School of Dental Medicine, University of Belgrade, Dr Subotica 4, Belgrade, Serbia
| | | | - Ivan Dozic
- General and Oral Biochemistry, School of Dental Medicine, University of Belgrade, Dr Subotica 4, Belgrade, Serbia
| | - Jovana Kuzmanovic Pficer
- Biomedical Statistics and Informatics, School of Dental Medicine, University of Belgrade, Dr Subotica 4, Belgrade, Serbia
| | - Zoran Aleksic
- Department of Periodontology, School of Dental Medicine, University of Belgrade, Dr Subotica 4, Belgrade, Serbia
| | - Natasa S. Nikolic-Jakoba
- Department of Periodontology, School of Dental Medicine, University of Belgrade, Dr Subotica 4, Belgrade, Serbia
| |
Collapse
|
12
|
Ebata S, Yoshizaki A, Oba K, Kashiwabara K, Ueda K, Uemura Y, Watadani T, Fukasawa T, Miura S, Yoshizaki-Ogawa A, Okiyama N, Kodera M, Hasegawa M, Sato S. Safety and efficacy of rituximab in systemic sclerosis (DESIRES): open-label extension of a double-blind, investigators-initiated, randomised, placebo-controlled trial. THE LANCET. RHEUMATOLOGY 2022; 4:e546-e555. [PMID: 38294008 DOI: 10.1016/s2665-9913(22)00131-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 01/14/2023]
Abstract
BACKGROUND Results from the double-blind phase 2 DESIRES trial showed that rituximab improves skin thickening in systemic sclerosis. Here, we present the findings of a subsequent 24-week open-label extension phase. METHODS Patients with systemic sclerosis aged 20-79 years, who fulfilled the 2013 American College of Rheumatology and European League Against Rheumatism classification criteria, with a baseline modified Rodnan Skin Score (mRSS) of 10 or greater were enrolled into the DESIRES trial, which was an investigator-initiated, phase 2, double-blind, randomised controlled trial of rituximab versus placebo conducted at four sites in Japan. After completion of 24 weeks of treatment with either rituximab or placebo, patients in both groups received a further 24 weeks of rituximab (375 mg/m2 intravenously, once per week for 4 consecutive weeks) in an open-label extension. The primary endpoint of the double-blind trial was mRSS at week 24, which was reassessed at week 48 in the open-label extension. All endpoints were exploratory. Safety analyses included all participants who received at least one dose of study drug; efficacy analyses included those who had received at least one dose and undergone efficacy assessment at 24 weeks in the double-blind phase and at 48 weeks in the extension phase. The DESIRES study is registered with ClinicalTrials.gov, NCT04274257, and UMIN-CTR, UMIN000030139. FINDINGS Between Nov 28, 2017, and Nov 6, 2018, 56 patients were randomly assigned to either rituximab (n=28) or placebo (n=28) in a double-blind study. 26 patients initially assigned to rituximab and 20 assigned to placebo transitioned to the open-label extension and all received at least one dose of rituximab; 24 participants in the rituximab-rituximab group and 19 in the placebo-rituximab group completed the extension phase. In the rituximab-rituximab group, there was an improvement in mRSS from baseline at week 24 (-5·81 [SD 3·16]), with further improvement at week 48 (-8·88 [3·10]). In the placebo-rituximab group, mRSS worsened at week 24 (2·14 [SD 5·51]) but improved at the week 48 assessment (-6·05 [4·43]). One patient each in the rituximab-rituximab and placebo-rituximab groups experienced one serious adverse event during the open-label phase (cholangitis and pneumococcal pneumonia, respectively). There were no deaths during follow-up. INTERPRETATION Two courses of rituximab is a safe treatment that can provide sustained improvement in systemic sclerosis for at least 48 weeks. FUNDING Japan Agency for Medical Research and Development. TRANSLATION For the Japanese translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Satoshi Ebata
- Department of Dermatology, The University of Tokyo, Tokyo, Japan
| | - Ayumi Yoshizaki
- Department of Dermatology, The University of Tokyo, Tokyo, Japan.
| | - Koji Oba
- Department of Biostatistics, School of Public Health, The University of Tokyo, Tokyo, Japan; Graduate School of Medicine, and Interfaculty Initiative in Information Studies, The University of Tokyo, Tokyo, Japan
| | - Kosuke Kashiwabara
- Clinical Research Support Center, Tokyo University Hospital, Tokyo, Japan
| | - Keiko Ueda
- Clinical Research Support Center, Tokyo University Hospital, Tokyo, Japan
| | - Yukari Uemura
- Clinical Research Support Center, Tokyo University Hospital, Tokyo, Japan; Biostatistics Section, Department of Data Science, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Takeyuki Watadani
- Department of Diagnostic Radiology, The University of Tokyo, Tokyo, Japan
| | | | - Shunsuke Miura
- Department of Dermatology, The University of Tokyo, Tokyo, Japan
| | | | - Naoko Okiyama
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Masanari Kodera
- Department of Dermatology, Japan Community Health Care Organization Chukyo Hospital, Aichi, Japan
| | - Minoru Hasegawa
- Department of Dermatology, Division of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Shinichi Sato
- Department of Dermatology, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
13
|
Assessment of disease outcome measures in systemic sclerosis. Nat Rev Rheumatol 2022; 18:527-541. [PMID: 35859133 DOI: 10.1038/s41584-022-00803-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2022] [Indexed: 01/08/2023]
Abstract
The assessment of disease activity in systemic sclerosis (SSc) is challenging owing to its heterogeneous manifestations across multiple organ systems, the variable rate of disease progression and regression, and the relative paucity of patients in early-phase therapeutic trials. Despite some recent successes, most clinical trials have failed to show efficacy, underscoring the need for improved outcome measures linked directly to disease pathogenesis, particularly applicable for biomarker studies focused on skin disease. Current outcome measures in SSc-associated interstitial lung disease and SSc skin disease are largely adequate, although advancing imaging technology and the incorporation of skin mRNA biomarkers might provide opportunities for earlier detection of the therapeutic effect. Biomarkers can further inform pathogenesis, enabling early phase trials to act as reverse translational studies through the incorporation of routine high-throughput sequencing.
Collapse
|
14
|
Matsuda KM, Yoshizaki A, Yamaguchi K, Fukuda E, Okumura T, Ogawa K, Ono C, Norimatsu Y, Kotani H, Hisamoto T, Kawanabe R, Kuzumi A, Fukasawa T, Ebata S, Miyagawa T, Yoshizaki-Ogawa A, Goshima N, Sato S. Autoantibody Landscape Revealed by Wet Protein Array: Sum of Autoantibody Levels Reflects Disease Status. Front Immunol 2022; 13:893086. [PMID: 35603173 PMCID: PMC9114879 DOI: 10.3389/fimmu.2022.893086] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
Autoantibodies are found in various pathological conditions such as autoimmune diseases, infectious diseases, and malignant tumors. However their clinical implications have not yet been fully elucidated. Herein, we conducted proteome-wide autoantibody screening and quantification with wet protein arrays consisting of proteins synthesized from proteome-wide human cDNA library (HuPEX) maintaining their three-dimensional structure. A total of 565 autoantibodies were identified from the sera of three representative inflammatory disorders (systemic sclerosis, psoriasis, and cutaneous arteritis). Each autoantibody level either positively or negatively correlated with serum levels of C-reactive protein, the best-recognized indicator of inflammation. In particular, we discovered total levels of a subset of autoantibodies correlates with the severity of clinical symptoms. From the sera of malignant melanoma, 488 autoantibodies were detected. Notably, patients with metastases had increased overall autoantibody production compared to those with tumors limiting to the primary site. Collectively, proteome-wide screening of autoantibodies using the in vitro proteome can reveal the "autoantibody landscape" of human subjects and may provide novel clinical biomarkers.
Collapse
Affiliation(s)
- Kazuki M Matsuda
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Ayumi Yoshizaki
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Kei Yamaguchi
- Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan.,ProteoBridge Corporation, Tokyo, Japan
| | - Eriko Fukuda
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan
| | - Taishi Okumura
- Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan.,ProteoBridge Corporation, Tokyo, Japan
| | - Koji Ogawa
- Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan.,ProteoBridge Corporation, Tokyo, Japan
| | - Chihiro Ono
- Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan.,ProteoBridge Corporation, Tokyo, Japan
| | - Yuta Norimatsu
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Hirohito Kotani
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Teruyoshi Hisamoto
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Ruriko Kawanabe
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Ai Kuzumi
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takemichi Fukasawa
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Satoshi Ebata
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takuya Miyagawa
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Asako Yoshizaki-Ogawa
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Naoki Goshima
- Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan.,ProteoBridge Corporation, Tokyo, Japan
| | - Shinichi Sato
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
15
|
Kowalska-Kępczyńska A. Systemic Scleroderma-Definition, Clinical Picture and Laboratory Diagnostics. J Clin Med 2022; 11:2299. [PMID: 35566425 PMCID: PMC9100749 DOI: 10.3390/jcm11092299] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/08/2022] [Accepted: 04/18/2022] [Indexed: 02/06/2023] Open
Abstract
(1) Background: Scleroderma (Sc) is a rare connective tissue disease classified as an autoimmune disorder. The pathogenesis of this disease is not fully understood. (2) Methods: This article reviews the literature on systemic scleroderma (SSc). A review of available scientific articles was conducted using the PubMed database with a time range of January 1985 to December 2021. (3) Results and Conclusions: The article is a review of information on epidemiology, criteria for diagnosis, pathogenesis, a variety of clinical pictures and the possibility of laboratory diagnostic in the diagnosis and monitoring of systemic scleroderma.
Collapse
Affiliation(s)
- Anna Kowalska-Kępczyńska
- Department of Biochemical Diagnostics, Chair of Laboratory Diagnostics, Medical University of Lublin, 20-081 Lublin, Poland
| |
Collapse
|
16
|
Gur C, Wang SY, Sheban F, Zada M, Li B, Kharouf F, Peleg H, Aamar S, Yalin A, Kirschenbaum D, Braun-Moscovici Y, Jaitin DA, Meir-Salame T, Hagai E, Kragesteen BK, Avni B, Grisariu S, Bornstein C, Shlomi-Loubaton S, David E, Shreberk-Hassidim R, Molho-Pessach V, Amar D, Tzur T, Kuint R, Gross M, Barboy O, Moshe A, Fellus-Alyagor L, Hirsch D, Addadi Y, Erenfeld S, Biton M, Tzemach T, Elazary A, Naparstek Y, Tzemach R, Weiner A, Giladi A, Balbir-Gurman A, Amit I. LGR5 expressing skin fibroblasts define a major cellular hub perturbed in scleroderma. Cell 2022; 185:1373-1388.e20. [PMID: 35381199 PMCID: PMC7612792 DOI: 10.1016/j.cell.2022.03.011] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 12/26/2021] [Accepted: 03/09/2022] [Indexed: 11/28/2022]
Abstract
Systemic sclerosis (scleroderma, SSc) is an incurable autoimmune disease with high morbidity and mortality rates. Here, we conducted a population-scale single-cell genomic analysis of skin and blood samples of 56 healthy controls and 97 SSc patients at different stages of the disease. We found immune compartment dysfunction only in a specific subtype of diffuse SSc patients but global dysregulation of the stromal compartment, particularly in a previously undefined subset of LGR5+-scleroderma-associated fibroblasts (ScAFs). ScAFs are perturbed morphologically and molecularly in SSc patients. Single-cell multiome profiling of stromal cells revealed ScAF-specific markers, pathways, regulatory elements, and transcription factors underlining disease development. Systematic analysis of these molecular features with clinical metadata associates specific ScAF targets with disease pathogenesis and SSc clinical traits. Our high-resolution atlas of the sclerodermatous skin spectrum will enable a paradigm shift in the understanding of SSc disease and facilitate the development of biomarkers and therapeutic strategies.
Collapse
Affiliation(s)
- Chamutal Gur
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel; Rheumatology Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Shuang-Yin Wang
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel.
| | - Fadi Sheban
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel
| | - Mor Zada
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel
| | - Baoguo Li
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel
| | - Fadi Kharouf
- Rheumatology Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Hagit Peleg
- Rheumatology Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Suhail Aamar
- Rheumatology Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Adam Yalin
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel
| | | | - Yolanda Braun-Moscovici
- Rheumatology Institute, Rambam Health Care Campus, Rappaport Faculty of Medicine, Technion-Israeli Institute of Technology, Haifa, Israel
| | | | - Tomer Meir-Salame
- Flow Cytometry Unit, Department of Biological Services, Weizmann Institute, Rehovot, Israel
| | - Efrat Hagai
- Flow Cytometry Unit, Department of Biological Services, Weizmann Institute, Rehovot, Israel
| | | | - Batia Avni
- Department of Bone Marrow Transplantation, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Sigal Grisariu
- Department of Bone Marrow Transplantation, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | | | | | - Eyal David
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel
| | - Rony Shreberk-Hassidim
- Dermatology Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Vered Molho-Pessach
- Dermatology Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Dalit Amar
- Plastic Surgery Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Tomer Tzur
- Plastic Surgery Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Rottem Kuint
- Institue of Pulmonology Medicine, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Moshe Gross
- Orthopedic Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Oren Barboy
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel
| | - Adi Moshe
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel
| | | | - Dana Hirsch
- The Department of Veterinary Resources, Weizmann Institute, Rehovot, Israel
| | - Yoseph Addadi
- Life Sciences Core Facilities, Weizmann Institute, Rehovot, Israel
| | - Shlomit Erenfeld
- Department of Bone Marrow Transplantation, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Moshe Biton
- Department of Biological Regulation, Weizmann Institute, Rehovot, Israel
| | - Tehila Tzemach
- Rheumatology Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Anat Elazary
- Rheumatology Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Yaakov Naparstek
- Rheumatology Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Reut Tzemach
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel; Rheumatology Institute at the Tel Aviv Sourasky Medical Center, Tel Aviv-Yafo, Israel
| | - Assaf Weiner
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel
| | - Amir Giladi
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel
| | - Alexandra Balbir-Gurman
- Rheumatology Institute, Rambam Health Care Campus, Rappaport Faculty of Medicine, Technion-Israeli Institute of Technology, Haifa, Israel
| | - Ido Amit
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel.
| |
Collapse
|
17
|
Manetti M, Rosa I, Fioretto BS, Matucci-Cerinic M, Romano E. Decreased Serum Levels of SIRT1 and SIRT3 Correlate with Severity of Skin and Lung Fibrosis and Peripheral Microvasculopathy in Systemic Sclerosis. J Clin Med 2022; 11:1362. [PMID: 35268452 PMCID: PMC8910971 DOI: 10.3390/jcm11051362] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/22/2022] [Accepted: 02/28/2022] [Indexed: 02/01/2023] Open
Abstract
Systemic sclerosis (SSc, scleroderma) is a severe autoimmune connective tissue disease characterized by widespread peripheral microvasculopathy, and progressive cutaneous and visceral fibrosis, leading to significant organ dysfunction. Sirtuins (SIRTs) are a family of NAD-dependent protein deacetylases with pleiotropic effects on a variety of biological processes, including metabolism, cell survival, and aging. In the last decades, increasing studies have explored the contribution of SIRTs to the pathogenesis of SSc, highlighting a significant antifibrotic effect of both SIRT1 and SIRT3. On these bases, the aim of this study was to measure circulating SIRT1 and SIRT3 levels by enzyme-linked immune-sorbent assay in a well-characterized cohort of SSc patients (n = 80) and healthy controls (n = 71), focusing on their possible association with disease clinical features, and their potential as biomarkers reflecting SSc activity and severity. Significantly decreased serum levels of both SIRT1 and SIRT3 were found in SSc patients compared to controls. In SSc, the reduction in circulating SIRT1 and SIRT3 associated with a greater extent of cutaneous fibrosis, presence of interstitial lung disease, and worse pulmonary function. Serum SIRT1 and SIRT3 decrease also correlated with the severity of nailfold microvascular damage, with SIRT3 levels being additionally related to the occurrence of digital ulcers. The levels of these two proteins showed a direct correlation with one another in the circulation of SSc patients. Of the two SIRTs, serum SIRT3 was found to better reflect disease activity and severity in a logistic regression analysis model. Our findings suggest that serum SIRT1 and SIRT3 may represent novel potential biomarkers of increased risk for a more severe, life-threatening SSc disease course.
Collapse
Affiliation(s)
- Mirko Manetti
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (I.R.); (B.S.F.)
| | - Irene Rosa
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (I.R.); (B.S.F.)
| | - Bianca Saveria Fioretto
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (I.R.); (B.S.F.)
- Division of Rheumatology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (M.M.-C.); (E.R.)
| | - Marco Matucci-Cerinic
- Division of Rheumatology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (M.M.-C.); (E.R.)
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Hospital, 20132 Milan, Italy
| | - Eloisa Romano
- Division of Rheumatology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (M.M.-C.); (E.R.)
| |
Collapse
|
18
|
OUP accepted manuscript. Rheumatology (Oxford) 2022; 61:4364-4373. [DOI: 10.1093/rheumatology/keac023] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 12/21/2021] [Indexed: 11/13/2022] Open
|
19
|
Günther F, Straub RH, Hartung W, Luchner A, Fleck M, Ehrenstein B. Increased Serum Levels of soluble ST2 as a Predictor of Disease Progression in Systemic Sclerosis. Scand J Rheumatol 2021; 51:315-322. [PMID: 34474647 DOI: 10.1080/03009742.2021.1929457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Interleukin-33 (IL-33) has been investigated as a mediator in the pathogenesis of fibrosis in lung, liver, and heart. There is accumulating evidence for the involvement of the IL-33/IL-33 receptor ST2L signalling pathway in systemic sclerosis (SSc). Little is known about the role of serum sST2 in SSc, which is the subject of the present investigation. METHOD Serum levels of sST2 were measured in 49 patients with SSc, recruited prospectively between November 2017 and March 2019. Patients were divided into those with progressive and those with stable disease. Receiver operating characteristics (ROC) curve analysis was applied to study sST2 as a marker for identifying patients with progressive disease. We used multivariate logistic regression analysis to evaluate the predictive value of sST2 for progressive disease after adjustment for potential confounding factors. RESULTS Serum sST2 levels in patients with progressive disease were significantly elevated compared with patients with stable disease (mean ± sem: 50.4 ± 4.7 ng/mL vs 29.2 ± 2.97 ng/mL, p < 0.001). ROC curve analysis identified an sST2 cut-off value of 37.8 ng/mL as optimal for discriminating patients with progressive disease from those with stable disease (sensitivity 80.0%, specificity 79.3%, area under the curve 0.80). After controlling for potential confounding factors (age, gender, C-reactive protein, pro-brain natriuretic peptide, and sum of internal medicine comorbidities), sST2 remained predictive of progressive disease (odds ratio 1.070, 95% confidence interval 1.017-1.126, p < 0.009). CONCLUSION In the present study, sST2 serum levels were predictive of disease progression in patients with SSc.
Collapse
Affiliation(s)
- F Günther
- Department of Rheumatology and Clinical Immunology, Asklepios Clinic, Bad Abbach, Germany
| | - R H Straub
- Department of Internal Medicine I, University Medical Center, Regensburg, Germany
| | - W Hartung
- Department of Rheumatology and Clinical Immunology, Asklepios Clinic, Bad Abbach, Germany
| | - A Luchner
- Department of Cardiology, Barmherzige Brüder Hospital, Regensburg, Germany
| | - M Fleck
- Department of Rheumatology and Clinical Immunology, Asklepios Clinic, Bad Abbach, Germany.,Department of Internal Medicine I, University Medical Center, Regensburg, Germany
| | - B Ehrenstein
- Department of Rheumatology and Clinical Immunology, Asklepios Clinic, Bad Abbach, Germany.,Department of Internal Medicine I, University Medical Center, Regensburg, Germany
| |
Collapse
|
20
|
Qiu M, Nian X, Pang L, Yu P, Zou S. Prevalence and risk factors of systemic sclerosis-associated interstitial lung disease in East Asia: A systematic review and meta-analysis. Int J Rheum Dis 2021; 24:1449-1459. [PMID: 34418313 PMCID: PMC9292335 DOI: 10.1111/1756-185x.14206] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 08/07/2021] [Accepted: 08/09/2021] [Indexed: 11/29/2022]
Abstract
Objective Interstitial lung disease (ILD) is a common and potentially life‐threatening complication for individuals with systemic sclerosis (SSc). The purpose of this study was to complete a systematic review and meta‐analysis on prevalence and risk factors of SSc‐ILD in East Asia. Methods Medline, EMBASE, and Cochrane Library were searched up to January 22, 2021. The Reporting of Observational Studies in Epidemiology (STROBE) statement was applied to access the methodological quality of the eligible studies. Study characteristics and magnitude of effect sizes were extracted. Then, we calculated the pooled prevalence, weighted mean differences (WMDs), pooled odds ratios (ORs) with corresponding 95% confidence intervals (CIs), and performed subgroup analysis, sensitivity analysis, and publication bias with Egger's test. Results Twenty‐seven of 1584 articles were eligible and a total of 5250 patients with SSc were selected in the meta‐analysis. The pooled prevalence of SSc‐ILD in East Asia was 56% (95% CI 49%‐63%). The SSc‐ILD prevalence was higher in China (72%) than in Japan (46%) and Korea (51%). Longer disease duration (WMD = 1.97, 95% CI 0.55‐3.38), diffuse SSc (OR = 2.84, 95% CI 1.91‐4.21), positive anti‐topoisomerase I antibody (ATA) (OR = 4.92, 95% CI 2.74‐8.84), positive anti‐centromere body antibody (ACA) (OR = 0.14, 95% CI 0.08‐0.25), positive anti‐U3 ribonucleoprotein (RNP) antibody (OR = 0.17, 95% CI 0.04‐0.66), and higher erythrocyte sedimentation rate (ESR) (WMD = 6.62, 95% CI 1.19‐12.05) were associated with SSc‐ILD in East Asia. Conclusion Through this systematic review and meta‐analysis, we found that ILD occurs in up to approximately 56% of patients with SSc in East Asia. Longer disease duration, diffuse SSc, positive ATA, negative ACA, negative anti‐U3 RNP antibody, and higher ESR were risk factors for SSc‐ILD.
Collapse
Affiliation(s)
- Meihua Qiu
- Department of Respiratory and Critical Care Medicine, Yantai Yuhuangding Hospital, Affiliated with the Medical College of Qingdao, Yantai, China
| | - Xueyuan Nian
- Department of Gastroenterology, Yantai Yuhuangding Hospital, Affiliated with the Medical College of Qingdao, Yantai, China
| | - Lingling Pang
- Department of Respiratory and Critical Care Medicine, Yantai Yuhuangding Hospital, Affiliated with the Medical College of Qingdao, Yantai, China
| | - Pengfei Yu
- Department of Respiratory and Critical Care Medicine, Yantai Yuhuangding Hospital, Affiliated with the Medical College of Qingdao, Yantai, China
| | - Shenchun Zou
- Department of Respiratory and Critical Care Medicine, Yantai Yuhuangding Hospital, Affiliated with the Medical College of Qingdao, Yantai, China
| |
Collapse
|
21
|
Cozzani E, Muracchioli A, Murdaca G, Beccalli M, Caprioli S, Zentilin P, Ameri P, Grosso M, Russo R, Carmisciano L, Parodi A. Correlation Between Skin and Affected Organs in 52 Sclerodermic Patients Followed in a Diseases Management Team: Development of a Risk Prediction Model of Organ-Specific Complications. Front Immunol 2021; 12:588753. [PMID: 34149680 PMCID: PMC8207468 DOI: 10.3389/fimmu.2021.588753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 05/10/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To identify the existence of a correlation among the various organs affected, focusing primarily on immuno-dermatological aspects, and to create a risk prediction model of organ-specific complications. MATERIAL AND METHODS Fifty-two patients with stable scleroderma, followed between 2015 and 2019, were investigated through an extensive multidisciplinary evaluation in the last year. RESULTS Patients with lung involvement presented a worse degree of skin fibrosis than patients without it (p <0.001). No relationship was observed for the heart, kidney, and esophagus. Patients with pulmonary involvement had a lower pressure of the low esophagus sphincter and a higher Warrick score than patients without it (p <0.05). Age was significantly higher in patients with kidney involvement. Diffuse scleroderma patients had a worse pulmonary impairment than limited scleroderma patients (p <0.05). The manometric "sclerodermic" pattern was observed to be the most frequent (55.6%, p <0.05) in dcSSc patients while the sclerodermic and normal pattern were equally represented (41.2 and 32.4% respectively, p <0.05) in lcSSc patients. When compared to the negative serological groups, anti-Scl-70 positive patients presented a worse lung involvement while anti-centromere patients presented a better lung outcome (p <0.05). PM-Scl 100/75 positive patients presented mostly a pulmonary fibrotic pattern (p <0.05) and, also, heart complications were more likely associated with anti PM-Scl 100/75 positivity (p <0.05). The risk prediction model for organ-specific complications had an accuracy of 84.4% (95%CI 78, 89) in complication-site prediction, AUC of 0.871, 86% of sensitivity, and 83% of specificity, Cohen's Kappa (k) of 0.68. CONCLUSIONS Out of all the organs studied, the skin is the one that correlates with the lung. Patients with a diffuse form of disease presented more frequently the anti Scl-70 antibody and had a worse lung and esophageal involvement (scleroderma pattern) than the negative group. Conversely, patients with limited disease presented all positive for the anti-centromere antibody with a better lung involvement than the negative group, without any difference among the esophageal manometric pattern. Anti PM-Scl 100/75 antibody patients were associated with pulmonary fibrosis and presented cardiac involvement. The model created has demonstrated excellent values of sensitivity, specificity, and accuracy, but further studies are needed for validation.
Collapse
Affiliation(s)
- Emanuele Cozzani
- Dermatologic Unit, University of Genoa, DiSSal, Ospedale-Policlinico San Martino, IRCCS, Genova, Italy
| | - Andrea Muracchioli
- Dermatologic Unit, University of Genoa, DiSSal, Ospedale-Policlinico San Martino, IRCCS, Genova, Italy
| | - Giuseppe Murdaca
- Departments of Internal Medicine, Scleroderma Unit, Clinical Immunology Unit, University of Genova, Ospedale-Policlinico San Martino, IRCCS, Genova, Italy
| | - Mirko Beccalli
- Departments of Internal Medicine, Scleroderma Unit, Clinical Immunology Unit, University of Genova, Ospedale-Policlinico San Martino, IRCCS, Genova, Italy
| | - Simone Caprioli
- Department of Healt Sciences DiSSal, University of Genova, Ospedale-Policlinico San Martino, IRCCS, Genova, Italy
| | - Patrizia Zentilin
- Division of Gastroenterology, Department of Internal Medicine, University of Genova, Ospedale-Policlinico San Martino, IRCCS, Genova, Italy
| | - Pietro Ameri
- Cardiovascular Disease Unit, IRCCS, Ospedale-Policlinico San Martino; Department of Internal Medicine, University of Genova, Genova, Italy
| | - Marco Grosso
- Interventional Pneumology Unit, Ospedale-Policlinico San Martino, IRCCS, Genova, Italy
| | - Rodolfo Russo
- Nephrology, Dialysis, and Transplantation, Ospedale-Policlinico San Martino, IRCCS, Genova, Italy
| | - Luca Carmisciano
- Department of Healt Sciences DiSSal, University of Genova, Ospedale-Policlinico San Martino, IRCCS, Genova, Italy
| | - Aurora Parodi
- Dermatologic Unit, University of Genoa, DiSSal, Ospedale-Policlinico San Martino, IRCCS, Genova, Italy
| |
Collapse
|
22
|
Zhang MY, Fang S, Gao H, Zhang X, Gu D, Liu Y, Wan J, Xie J. A critical role of AREG for bleomycin-induced skin fibrosis. Cell Biosci 2021; 11:40. [PMID: 33622407 PMCID: PMC7903615 DOI: 10.1186/s13578-021-00553-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 02/10/2021] [Indexed: 12/16/2022] Open
Abstract
We report our discovery of an important player in the development of skin fibrosis, a hallmark of scleroderma. Scleroderma is a fibrotic disease, affecting 70,000 to 150,000 Americans. Fibrosis is a pathological wound healing process that produces an excessive extracellular matrix to interfere with normal organ function. Fibrosis contributes to nearly half of human mortality. Scleroderma has heterogeneous phenotypes, unpredictable outcomes, no validated biomarkers, and no effective treatment. Thus, strategies to slow down scleroderma progression represent an urgent medical need. While a pathological wound healing process like fibrosis leaves scars and weakens organ function, oral mucosa wound healing is a scarless process. After re-analyses of gene expression datasets from oral mucosa wound healing and skin fibrosis, we discovered that several pathways constitutively activated in skin fibrosis are transiently induced during oral mucosa wound healing process, particularly the amphiregulin (Areg) gene. Areg expression is upregulated ~ 10 folds 24hrs after oral mucosa wound but reduced to the basal level 3 days later. During bleomycin-induced skin fibrosis, a commonly used mouse model for skin fibrosis, Areg is up-regulated throughout the fibrogenesis and is associated with elevated cell proliferation in the dermis. To demonstrate the role of Areg for skin fibrosis, we used mice with Areg knockout, and found that Areg deficiency essentially prevents bleomycin-induced skin fibrosis. We further determined that bleomycin-induced cell proliferation in the dermis was not observed in the Areg null mice. Furthermore, we found that inhibiting MEK, a downstream signaling effector of Areg, by selumetinib also effectively blocked bleomycin-based skin fibrosis model. Based on these results, we concluded that the Areg-EGFR-MEK signaling axis is critical for skin fibrosis development. Blocking this signaling axis may be effective in treating scleroderma.
Collapse
Affiliation(s)
- Mary Yinghua Zhang
- Department of Pediatrics, The Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shuyi Fang
- Department of BioHealth Informatics, School of Informatics and Computing At IUPUI, Indiana University, Indianapolis, IN, USA
| | - Hongyu Gao
- The IU Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, USA
| | - Xiaoli Zhang
- Department of Pediatrics, The Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Dongsheng Gu
- Department of Pediatrics, The Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yunlong Liu
- Department of BioHealth Informatics, School of Informatics and Computing At IUPUI, Indiana University, Indianapolis, IN, USA
- The IU Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, USA
- The Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jun Wan
- Department of BioHealth Informatics, School of Informatics and Computing At IUPUI, Indiana University, Indianapolis, IN, USA
- The IU Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, USA
- The Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jingwu Xie
- Department of Pediatrics, The Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
- The IU Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
23
|
Matsuda KM, Yoshizaki A, Kotani H, Norimatsu Y, Kuzumi A, Fukayama M, Fukasawa T, Ebata S, Yoshizaki-Ogawa A, Asano Y, Oba K, Sato S. Serum heat shock protein 27 levels in patients with systemic sclerosis: a possible biomarker of skin sclerosis. J Eur Acad Dermatol Venereol 2020; 35:e157-e159. [PMID: 32805744 DOI: 10.1111/jdv.16885] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 07/28/2020] [Accepted: 08/06/2020] [Indexed: 12/14/2022]
Affiliation(s)
- K M Matsuda
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - A Yoshizaki
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - H Kotani
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Y Norimatsu
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - A Kuzumi
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - M Fukayama
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - T Fukasawa
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - S Ebata
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - A Yoshizaki-Ogawa
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Y Asano
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - K Oba
- Department of Biostatistics, School of Public Health, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - S Sato
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
24
|
Mostmans Y, Richert B, Badot V, Nagant C, Smith V, Michel O. The importance of skin manifestations, serology and nailfold (video)capillaroscopy in morphea and systemic sclerosis: current understanding and new insights. J Eur Acad Dermatol Venereol 2020; 35:597-606. [PMID: 32656859 DOI: 10.1111/jdv.16813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 06/24/2020] [Indexed: 11/29/2022]
Abstract
Since the field around morphea and systemic sclerosis (SSc) is evolving rapidly, this review approaches conventional as well as more recent clinical developments from a dermatological point of view. Skin manifestations are critical in sub-classifying these diseases ensuring a correct prognosis for these patients. They can be discretely present, and therefore, diagnosis can be challenging sometimes, implicating a thorough dermatological examination is mandatory. Furthermore, a growing amount of dermatologists perform nailfold videocapillaroscopy (NVC), a more recent reliable non-invasive imaging technique used for in vivo assessment of the microcirculation at the nailfold. After all, specific NVC-changes are present in a majority of patients with SSc. This way, dermatologists not only take part in the diagnosis process through clinical investigation but also through the use of a modern state of the art imaging technique that is becoming the golden standard in SSc multidisciplinary workup. In this review, current understandings for NVC in morphea and SSc are revised. So far, the role of NVC in the diagnosis/prognosis/classification of morphea patients has not been thoroughly investigated to make proper conclusions. As for SSc, it is well known that NVC contributes to the diagnosis and can make a fundamental difference especially when obvious clinical SSc signs are absent. This review emphasizes the (somewhat underestimated) role of dermatologists in the process of diagnosis and follow-up, and thus, the difference we can make for our patients and fellow colleagues in the multidisciplinary workup of SSc and morphea.
Collapse
Affiliation(s)
- Y Mostmans
- Department of Immunology-Allergology, CHU Brugmann, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Department of Dermatology, CHU Brugmann, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - B Richert
- Department of Dermatology, CHU Brugmann, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - V Badot
- Department of Immunology-Allergology, CHU Brugmann, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Department of Rheumatology, CHU Brugmann, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - C Nagant
- Department of Immunology IRIS Laboratory, CHU Brugmann, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - V Smith
- Department of Rheumatology, Ghent University Hospital, University Hospital (UZ) Ghent, Ghent, Belgium
| | - O Michel
- Department of Immunology-Allergology, CHU Brugmann, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
25
|
Di Benedetto P, Guggino G, Manzi G, Ruscitti P, Berardicurti O, Panzera N, Grazia N, Badagliacca R, Riccieri V, Vizza CD, Radchenko G, Liakouli V, Ciccia F, Cipriani P, Giacomelli R. Interleukin-32 in systemic sclerosis, a potential new biomarker for pulmonary arterial hypertension. Arthritis Res Ther 2020; 22:127. [PMID: 32487240 PMCID: PMC7268373 DOI: 10.1186/s13075-020-02218-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/13/2020] [Indexed: 02/07/2023] Open
Abstract
Background Pulmonary arterial hypertension (PAH) is a severe complication of systemic sclerosis (SSc), associated with a progressive elevation in pulmonary vascular resistance and subsequent right heart failure and death. Due to unspecific symptoms, the diagnosis of PAH is often delayed. On this basis, it is of great value to improve current diagnostic methods and develop new strategies for evaluating patients with suspected PAH. Interleukin-32 (IL-32) is a proinflammatory cytokine expressed in damaged vascular cells, and the present study aimed to assess if this cytokine could be a new biomarker of PAH during SSc. Methods The IL-32 expression was evaluated in the sera and skin samples of 18 SSc-PAH patients, 21 SSc patients without PAH, 15 patients with idiopathic PAH (iPAH) and 14 healthy controls (HCs), by enzyme-linked immunosorbent assay (ELISA) and immunohistochemistry (IHC). Receiver-operating characteristic (ROC) curves were performed to evaluate the cut-off of IL-32 in identifying patients with PAH. Furthermore, in SSc patients, correlation analyses were performed between IL-32 sera levels and mean pulmonary artery pressure (mPAP) evaluated by right heart catheterization (RHC) and systolic pulmonary artery pressure (sPAP), obtained by echocardiography. Additionally, the number of skin IL-32+ cells was correlated with modified Rodnan skin score (mRSS). Results In SSc-PAH patients, IL-32 sera levels were significantly higher when compared with SSc patients without PAH and patients affected by iPAH. The analysis of ROC curve showed that IL-32 sera levels above 11.12 pg/ml were able to predict patients with PAH (sensitivity = 90%, specificity = 100%). Furthermore, the IL-32 sera levels of patients with SSc correlated with both mPAP and sPAP. In the skin derived from SSc-PAH patients, the number of IL-32+ cells was significantly increased when compared with the skin derived from SSc patients without PAH, correlating with the mRSS. Conclusion Our study suggested that sera determination of IL-32 may be a promising approach to evaluate the presence of PAH in SSc patients and together with longitudinal future studies could help to increase the understanding how these biomarkers mirror the vascular changes and the inflammatory process during SSc.
Collapse
Affiliation(s)
- Paola Di Benedetto
- Clinical Pathology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Giuliana Guggino
- Rheumatology Section, Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - Giovanna Manzi
- Department of Cardiovascular and Respiratory Sciences, Sapienza University of Rome, Rome, Italy
| | - 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
| | - Noemi Panzera
- Clinical Pathology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Nicolò Grazia
- Clinical Pathology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Roberto Badagliacca
- Department of Cardiovascular and Respiratory Sciences, Sapienza University of Rome, Rome, Italy
| | - Valeria Riccieri
- Department of Internal Medicine and Medical Specialities, Sapienza University of Rome, Rome, Italy
| | - Carmine Dario Vizza
- Department of Cardiovascular and Respiratory Sciences, Sapienza University of Rome, Rome, Italy
| | - Ganna Radchenko
- Secondary Hypertension Department with Pulmonary Hypertension Center, State Institute National Scientific Center "MD Strazhesko Institute of Cardiology" of Ukrainian National Academy of Medical Science, Kyiv, Ukraine
| | - Vasiliki Liakouli
- Rheumatology Section, Department of Clinical and Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Francesco Ciccia
- Rheumatology Section, Department of Clinical and Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Paola Cipriani
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Roberto Giacomelli
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| |
Collapse
|
26
|
Alves F, Gonçalo M. Suspected inflammatory rheumatic diseases in patients presenting with skin rashes. Best Pract Res Clin Rheumatol 2019; 33:101440. [PMID: 31585842 DOI: 10.1016/j.berh.2019.101440] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Skin lesions occur, often at very early stages, in many of the most frequent inflammatory rheumatic diseases such as in systemic lupus erythematosus (SLE), dermatomyositis (DM), systemic sclerosis (SSc), Sjögren's syndrome, rheumatoid arthritis (RA), and psoriatic arthritis. It is important to recognize the different specific cutaneous lesions in SLE (e.g., "butterfly" rash in acute, annular or psoriasiform photosensitive lesions in the subacute form, and discoid lesions in the chronic form) for an early diagnosis and to estimate the associated risks of internal disease, whereas nonspecific lesions (exanthema, vasculitis, and alopecia) can be part of SLE flares. Cutaneous lesions in DM (Gottron's papules and sign, heliotrope rash, dystrophic cuticles, and nailfold capillary abnormalities) may occur before any clinically evident muscular or systemic organ involvement and are of utmost importance for early diagnosis. The pattern of cutaneous lesions and associated autoantibodies also allow the distinction of different phenotypes, either more prone to life-threatening interstitial lung disease (MDA-5) or with higher risk for neoplasia (TIF1-γ). Many other skin lesions, although not specific, require further investigation to look for a possible underlying inflammatory rheumatic disease: non-pruritic urticarial lesions in anti-C1q-associated urticarial vasculitis, Still's disease or hereditary auto-inflammatory syndromes, transient macular purpura of vasculitis in Sjögren's syndrome, Behçet's disease, or RA, Raynaud's phenomenon in SSc and mixed connective tissue disease, erythema nodosum or other panniculitis in RA, Behçet's disease and SLE, pustular eruptions in Behçet's disease, psoriasis, and hereditary auto-inflammatory syndromes. After reviewing in detail the cutaneous manifestations of the most frequent inflammatory rheumatic diseases, we describe a topographic and morphological approach to skin rashes, calling attention to facial rashes, hand involvement, scalp, nail, or leg lesions or to some morphological aspects of skin lesions (annular, pustular, urticarial, or exanthematous) that may be the initial manifestations of inflammatory rheumatic diseases. The importance of skin lesions is confirmed by their presence as part of the classification criteria of many inflammatory rheumatic diseases. They also contribute to early diagnosis, to characterize disease phenotypes, to aid in effective patient management, and, ultimately, to impact on disease prognosis.
Collapse
Affiliation(s)
- Francisca Alves
- Clinic of Dermatology, University Hospital of Coimbra, Portugal
| | - Margarida Gonçalo
- Clinic of Dermatology, University Hospital of Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
27
|
Corallo C, Fioravanti A, Tenti S, Pecetti G, Nuti R, Giordano N. Sarcopenia in systemic sclerosis: the impact of nutritional, clinical, and laboratory features. Rheumatol Int 2019; 39:1767-1775. [PMID: 31372720 DOI: 10.1007/s00296-019-04401-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/26/2019] [Indexed: 12/22/2022]
Abstract
We evaluated the presence of sarcopenia in a population of systemic sclerosis (SSc) patients, with respect to nutritional, clinical, and laboratory features. A total of 62 patients who met the ACR/EULAR 2013 classification criteria were enrolled. Sarcopenia was defined according to the Relative Skeletal Mass Index (RSMI) and hand grip strength (HGS). Body composition was assessed with the calculation of the Body Mass Index (BMI), lean body mass (LBM) and fat mass (FM). Malnutrition was evaluated according to the ESPEN criteria. Clinical evaluation included nailfold capillaroscopy and skin evaluation by modified Rodnan Skin Score (mRSS), pulmonary function tests (PFT) with diffusing capacity for carbon monoxide adjusted for hemoglobin (DLCO), high-resolution computed tomography (HR-CT) of the lungs, echocardiography and high-resolution manometry (HRM) for esophageal involvement. Laboratory evaluation included erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), hemoglobin, creatinine, creatine kinase (CK), transaminases, lipid profile, glycemia, albumin, and vitamin-D. Antinuclear antibodies (ANA) and extractable nuclear antigens (ENA) were also assessed. Considering RSMI, the prevalence of sarcopenia is 42%. In this case, age, malnutrition, disease duration, mRSS, capillaroscopy score, esophageal involvement, ESR, and ANA titer are higher in the sarcopenic group, while DLCO and LBM are lower. Considering HGS, the prevalence of sarcopenia is 55%. Age, disease duration, malnutrition, FM, mRSS, capillaroscopy score, esophageal involvement, ESR, and ENA positivity are higher in the sarcopenic group, while DLCO is lower. By using both RSMI and HGS to assess sarcopenia in SSc, the results of this study demonstrated that this condition correlates with different nutritional, clinical, and biochemical parameters associated with the worsening of the disease.
Collapse
Affiliation(s)
- Claudio Corallo
- Scleroderma Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy.
| | - Antonella Fioravanti
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Sara Tenti
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Gianluca Pecetti
- Medical and Scientific Direction, Actelion Pharmaceuticals Italia, Imola, Italy
| | - Ranuccio Nuti
- Scleroderma Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Nicola Giordano
- Scleroderma Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| |
Collapse
|