1
|
Xu X, Wang M, Geng Z, Jin Y, Bai G, Dawn B, Gong F, Zhao L. Identification of m 6A-related biomarkers in Kawasaki disease. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167744. [PMID: 39988181 DOI: 10.1016/j.bbadis.2025.167744] [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: 10/04/2024] [Revised: 11/26/2024] [Accepted: 02/15/2025] [Indexed: 02/25/2025]
Abstract
Kawasaki disease (KD) is a widely prevalent acute vasculitis in children that often leads to cardiovascular complications. Although m6A modification plays a crucial role in various cardiovascular diseases, m6A-related biomarkers for KD remain unknown. We utilized GEO datasets to perform WGCNA to identify m6A-related differentially expressed genes in KD. Feature genes associated with m6A and key immune cells were identified using RF and SVM-RFE algorithms, and CIBERSORT, and the correlation was evaluated using CytoHubba and ROC analysis. The expression of hub genes was assessed in blood from patients with KD and in mice with CAWS-induced vasculitis. Our analysis identified four m6A-related hub genes: SNRK, PCCB, PIGP, and PRPS1, which exhibited significant negative correlation with M2 macrophages. A total of 477 microRNAs, 22 lncRNAs, and 3 snRNAs were identified as potential regulators of these hub genes. The ROC analysis demonstrated a robust diagnostic accuracy of these hub genes for KD. The expression of these hub genes was reduced in blood from patients with KD and in mice with vasculitis. In conclusion, SNRK, PCCB, PIGP, and PRPS1 demonstrate significant diagnostic value for KD and may also be considered as potential therapeutic targets.
Collapse
Affiliation(s)
- Xiao Xu
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China
| | - Min Wang
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China
| | - Zhimin Geng
- National Clinical Research Center for Child Health, Children's Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Yihua Jin
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China
| | - Guannan Bai
- National Clinical Research Center for Child Health, Children's Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Buddhadeb Dawn
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at the University of Nevada, Las Vegas, NV, USA
| | - Fangqi Gong
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China.
| | - Lin Zhao
- National Clinical Research Center for Child Health, Children's Hospital, Zhejiang University School of Medicine, Zhejiang, China.
| |
Collapse
|
2
|
Jena PK, Wakita D, Gomez AC, Carvalho TT, Atici AE, Aubuchon E, Narayanan M, Lee Y, Fishbein MC, Takasato Y, Kurashima Y, Kiyono H, Cani PD, de Vos WM, Underhill DM, Devkota S, Chen S, Shimada K, Crother TR, Arditi M, Rivas MN. Intestinal Microbiota Contributes to the Development of Cardiovascular Inflammation and Vasculitis in Mice. Circ Res 2025; 136:e53-e72. [PMID: 40026151 PMCID: PMC11985309 DOI: 10.1161/circresaha.124.325079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Alterations in the intestinal microbiota contribute to the pathogenesis of various cardiovascular disorders, but how they affect the development of Kawasaki disease (KD) an acute pediatric vasculitis, remains unclear. METHODS We used the Lactobacillus casei cell wall extract (LCWE) murine model of KD vasculitis to assess the contribution of the intestinal microbiota to the development of vascular inflammation. We evaluated the severity of vasculitis in microbiota-depleted mice. 16S rRNA gene sequencing was used to characterize the fecal microbiome composition of LCWE-injected mice. Some groups of mice were orally treated with selected live or pasteurized bacteria, short-chain fatty acids, or Amuc_1100, the Toll-like receptor 2 signaling outer membrane protein from Akkermansia muciniphila, and their impact on vasculitis development was assessed. RESULTS We report that depleting the gut microbiota reduces the development of cardiovascular inflammation in a murine model mimicking KD vasculitis. The development of cardiovascular lesions was associated with alterations in the intestinal microbiota composition and, notably, a decreased abundance of Akkermansia muciniphila and Faecalibacterium prausnitzii. Oral supplementation with either of these live or pasteurized individual bacteria or with short-chain fatty acids produced by them attenuated cardiovascular inflammation, as reflected by decreased local immune cell infiltrations. Treatment with Amuc_1100 also reduced the severity of vascular inflammation. CONCLUSIONS This study reveals an underappreciated gut microbiota-cardiovascular inflammation axis in KD vasculitis pathogenesis and identifies specific intestinal commensals that regulate vasculitis in mice by producing metabolites or via extracellular proteins capable of enhancing and supporting gut barrier function.
Collapse
Affiliation(s)
- Prasant K. Jena
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Daiko Wakita
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Angela C. Gomez
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Thacyana T. Carvalho
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Asli E. Atici
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Emily Aubuchon
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Meena Narayanan
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Youngho Lee
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael C. Fishbein
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, CA, USA
| | - Yoshihiro Takasato
- Department of Allergy, Allergy and Immunology Center, Aichi Children’s Health and Medical Center, Obu, Japan
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yosuke Kurashima
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Japan
| | - Hiroshi Kiyono
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Japan
| | - Patrice D. Cani
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- WELBIO-Walloon Excellence in Life Sciences and BIOtechnology, WELBIO department, WEL Research Institute, Wavre, Belgium
- Institute of Experimental and Clinical Research (IREC), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Willem M. de Vos
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - David M. Underhill
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- F. Widjaja Inflammatory Bowel Diseases Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Suzanne Devkota
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- F. Widjaja Inflammatory Bowel Diseases Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Human Microbiome Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Shuang Chen
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kenichi Shimada
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Timothy R. Crother
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Moshe Arditi
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Smidt Heart Research Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
3
|
Dalman JM, Blaustein ER, van Solingen C. Gut Instincts: The Gut Microbiome-Cardiovascular Inflammation Axis. Circ Res 2025; 136:806-808. [PMID: 40208926 PMCID: PMC11990081 DOI: 10.1161/circresaha.125.326363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Affiliation(s)
- Jessie M. Dalman
- Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY 10016, USA
- University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Emma R. Blaustein
- Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Coen van Solingen
- Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
4
|
Jena PK, Arditi M, Rivas MN. Gut Microbiota Alterations in Patients With Kawasaki Disease. Arterioscler Thromb Vasc Biol 2025; 45:345-358. [PMID: 39846163 PMCID: PMC11998981 DOI: 10.1161/atvbaha.124.321201] [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: 10/14/2024] [Revised: 12/09/2024] [Accepted: 01/09/2025] [Indexed: 01/24/2025]
Abstract
The intestinal microbiota influences many host biological processes, including metabolism, intestinal barrier functions, and immune responses in the gut and distant organs. Alterations in its composition have been associated with the development of inflammatory disorders and cardiovascular diseases, including Kawasaki disease (KD). KD is an acute pediatric vasculitis of unknown etiology and the leading cause of acquired heart disease in children in the United States. The presence of gastrointestinal symptoms in the acute phase of KD has been associated with an increased risk of treatment resistance and the development of coronary artery aneurysms. Studies report alterations in fecal bacterial communities of patients with KD, characterized by the blooming of pathogenic bacteria and decreased relative abundance of short-chain fatty acid-producing bacteria. However, causality and functionality cannot be established from these observational patient cohorts of KD. This highlights the need for more advanced and rigorous studies to establish causality and functionality in both experimental models of KD vasculitis and patient cohorts. Here, we review the evidence linking an altered gut microbiota composition to the development of KD, assess the potential mechanisms involved in this process, and discuss the potential therapeutic value of these observations.
Collapse
Affiliation(s)
- Prasant K. Jena
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children’s, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Moshe Arditi
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children’s, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children’s, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
5
|
Zhou L, Zhu X, Lei S, Wang Y, Xia Z. The role of the ER stress sensor IRE1 in cardiovascular diseases. Mol Cell Biochem 2025; 480:683-691. [PMID: 38717685 DOI: 10.1007/s11010-024-05014-z] [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: 02/22/2024] [Accepted: 04/18/2024] [Indexed: 02/19/2025]
Abstract
Despite enormous advances in the treatment of cardiovascular diseases, including I/R injury and heart failure, heart diseases remain a leading cause of mortality worldwide. Inositol-requiring enzyme 1 (IRE1) is an evolutionarily conserved sensor endoplasmic reticulum (ER) transmembrane protein that senses ER stress. It manages ER stress induced by the accumulation of unfolded/misfolded proteins via the unfolded protein response (UPR). However, if the stress still persists, the UPR pathways are activated and induce cell death. Emerging evidence shows that, beyond the UPR, IRE1 participates in the progression of cardiovascular diseases by regulating inflammation levels, immunity, and lipid metabolism. Here, we summarize the recent findings and discuss the potential therapeutic effects of IRE1 in the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Lu Zhou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xizi Zhu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shaoqing Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yafeng Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan, China.
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
6
|
Qian G, Wang Y, Yao H, Zhang Z, Wang W, Xu L, Li W, Huang L, Li X, Gao Y, Wang N, Wang S, Pan J, Lv H. Involvement of USP7 in aggravating Kawasaki disease by promoting TGFβ2 signaling mediated endothelial-mesenchymal transition and coronary artery remodeling. Int Immunopharmacol 2025; 146:113823. [PMID: 39674001 PMCID: PMC11799894 DOI: 10.1016/j.intimp.2024.113823] [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: 11/05/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/16/2024]
Abstract
Kawasaki disease (KD), characterized by systematic vasculitis, is a leading cause of pediatric heart disease. Although recent studies have highlighted the critical role of deubiquitinases in vascular pathophysiology, their specific contribution to KD remains largely unknown. Herein, we investigated the function of the deubiquitinase USP7 in both KD patients and a CAWS-induced KD murine model. USP7 expression level is increased both in HCAECs induced by KD sera and cardiac CD31+ endothelial cells of KD mice. Whereas knockout of USP7 increases the cellular proportion of endothelial cells and potentially attenuates the elevated EndoMT, fibrosis, and inflammation in cardiac tissue of KD mice, consistently with the in vitro experiment observed in HCAECs induced by TGF-β2. Mechanistically, USP7 interacts with SMAD2/3, enhancing their protein stability by removing the K48 ubiquitin chain from both proteins and preventing their proteasome degradation, thus increasing the p-SMAD2 levels and nuclear entry. Importantly, intraperitoneal injection of USP7 inhibitor, P22077 elicited a robust anti-EndoMT and anti-vascular inflammation effect in KD model mice. Therefore, our study uncovered a previously unrecognized function of increased USP7 in KD by augmenting TGFβ2/SMAD2/SMAD3 signaling, thus facilitating the transcription of genes implicated in the EndoMT, cardiac fibrosis, and vascular remodeling. Our finding suggests that USP7 could serve as a potential therapeutic target for the prevention and treatment of coronary artery lesions in KD and related vascular diseases.
Collapse
Affiliation(s)
- Guanghui Qian
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China.
| | - Yan Wang
- Department of Cardiology, The Affiliated Xuzhou Children's Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province 221000, China.
| | - Hongwei Yao
- Providence VA Medical Center, Providence, RI, USA; Departments of Medicine, Molecular Biology, Cell Biology & Biochemistry, Brown University, Providence, RI, USA.
| | - Zimu Zhang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China.
| | - Wang Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China
| | - Lei Xu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China.
| | - Wenjie Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China.
| | - Li Huang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China.
| | - Xuan Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China.
| | - Yang Gao
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China
| | - Nana Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China.
| | - Shuhui Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China.
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China.
| | - Haitao Lv
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China.
| |
Collapse
|
7
|
Sharma S, Goel S, Goyal T, Pilania RK, Aggarwal R, Kaur T, Dhaliwal M, Rawat A, Singh S. Single-cell RNA sequencing: an emerging tool revealing dysregulated innate and adaptive immune response at single cell level in Kawasaki disease. Expert Rev Clin Immunol 2025; 21:83-92. [PMID: 39230194 DOI: 10.1080/1744666x.2024.2401105] [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: 02/09/2024] [Revised: 08/07/2024] [Accepted: 09/02/2024] [Indexed: 09/05/2024]
Abstract
INTRODUCTION Kawasaki disease [KD] is a systemic disorder characterized by acute febrile illness due to widespread medium-vessel vasculitis, mainly affecting children. Despite the ongoing advanced research into the disease pathophysiology and molecular mechanisms, the exact etiopathogenesis of KD is still an enigma. Recently, single-cell RNA sequencing [scRNA-seq], has been utilized to elucidate the pathophysiology of KD at a resolution higher than that of previous methods. AREA COVERED In the present article, we re-emphasize the pivotal role of this high-resolution technique, scRNA-seq, in the characterization of immune cell transcriptomic profile and signaling/response pathways in KD and explore the diagnostic, prognostic, and therapeutic potential of this new technique in KD. Using combinations of the search phrases 'KD, scRNA-seq, CAA, childhood vasculitis' a literature search was carried out on Scopus, Google Scholar, and PubMed until the beginning of 2024. EXPERT OPINION scRNA-seq presents a transformative tool for dissecting KD at the cellular level. By revealing rare cell populations, gene expression alterations, and disease-specific pathways, scRNA-seq aids in understanding the intricacies of KD pathogenesis. This review will provide new insights into pathogenesis of KD and the field of applications of scRNA-seq in personalized therapeutics for KD in the future.
Collapse
Affiliation(s)
- Saniya Sharma
- Department of Pediatrics, Allergy Immunology Unit, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sumit Goel
- Department of Pediatrics, Allergy Immunology Unit, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Taru Goyal
- Department of Pediatrics, Allergy Immunology Unit, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rakesh Kumar Pilania
- Department of Pediatrics, Allergy Immunology Unit, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ridhima Aggarwal
- Department of Pediatrics, Allergy Immunology Unit, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Taranpreet Kaur
- Department of Pediatrics, Allergy Immunology Unit, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Manpreet Dhaliwal
- Department of Pediatrics, Allergy Immunology Unit, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amit Rawat
- Department of Pediatrics, Allergy Immunology Unit, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Surjit Singh
- Department of Pediatrics, Allergy Immunology Unit, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
8
|
Atici AE, Noval Rivas M, Arditi M. The Central Role of Interleukin-1 Signalling in the Pathogenesis of Kawasaki Disease Vasculitis: Path to Translation. Can J Cardiol 2024; 40:2305-2320. [PMID: 39084253 PMCID: PMC11646188 DOI: 10.1016/j.cjca.2024.07.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/16/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024] Open
Abstract
Kawasaki disease (KD) manifests as an acute febrile condition and systemic vasculitis, the etiology of which remains elusive. Primarily affecting children under 5 years of age, if untreated KD can lead to a significant risk of coronary artery aneurysms and subsequent long-term cardiovascular sequelae, including myocardial ischemia and myocardial infarction. Intravenous immunoglobulin therapy mitigates the risk of aneurysm formation, but a subset of patients exhibit resistance to this treatment, increasing the susceptibility of coronary artery lesions. Furthermore, the absence of a KD-specific diagnostic test or biomarkers complicates early detection and appropriate treatment. Experimental murine models of KD vasculitis have substantially improved our understanding of the disease pathophysiology, revealing the key roles of the NLRP3 inflammasome and interleukin-1 (IL-1) signalling pathway. This review aims to delineate the pathophysiologic findings of KD while summarising the findings for the emerging key role of IL-1β in its pathogenesis, derived from both human data and experimental murine models, and the translational potential of these findings for anti-IL-1 therapies for children with KD.
Collapse
Affiliation(s)
- Asli Ekin Atici
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Guerin Children's at Cedars-Sinai Medical Center, Los Angeles, California, USA; Infectious and Immunologic Diseases Research Center, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Magali Noval Rivas
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Guerin Children's at Cedars-Sinai Medical Center, Los Angeles, California, USA; Infectious and Immunologic Diseases Research Center, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Moshe Arditi
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Guerin Children's at Cedars-Sinai Medical Center, Los Angeles, California, USA; Infectious and Immunologic Diseases Research Center, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA.
| |
Collapse
|
9
|
Domiciano TP, Lee Y, Carvalho TT, Wakita D, Martinon D, Jena PK, Fert-Bober J, Borges V, Crother TR, Chen S, Moreira D, Van Eyk JE, Noval Rivas M, Arditi M, Shimada K. Redundant role of PAD2 and PAD4 in the development of cardiovascular lesions in a mouse model of Kawasaki disease vasculitis. Clin Exp Immunol 2024; 218:314-328. [PMID: 39250707 PMCID: PMC11557146 DOI: 10.1093/cei/uxae080] [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: 02/08/2024] [Revised: 07/21/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024] Open
Abstract
Kawasaki disease (KD) is the leading cause of acquired heart disease in children. While circulating neutrophils are increased and activated during acute KD, it is unclear whether neutrophils and neutrophil extracellular traps (NETs) contribute to the pathogenesis of KD. Peptidylarginine deiminase 4 (PAD4), an enzyme involved in protein citrullination and essential for NETs formation, is implicated in the pathogenesis of various diseases. Here, we used the Lactobacillus casei cell wall extract (LCWE)-induced mouse model of KD vasculitis to determine the contribution of PAD4 in KD vasculitis. We found that the pan-PADs inhibitor, Cl-amidine, significantly reduced LCWE-induced cardiovascular lesions, but neutrophil-specific Padi4 KO mice did not impact the development of KD vasculitis. While in vitro treatment of macrophages, which highly express Padi4, with Cl-amidine inhibited IL-1β secretion, macrophage-specific Padi4 KO mice did not reduce the lesions. Padi4-/- mice also developed KD vasculitis, AFM30a, a PAD2 inhibitor, significantly reduced KD vasculitis in Padi4-/- mice, indicating a compensatory role of PAD2 in PAD4 deficiency. We also identified several citrullinated proteins in macrophages with constitutively active NLRP3 inflammasome that were inhibited by Cl-amidine treatment, suggesting that protein citrullination participates in NLRP3 inflammasome activation. These data indicate a dispensable role for PAD4-dependent NETs formation, and a redundant role of PAD2 and PAD4 in this murine KD vasculitis. The cardioprotective effects of Cl-amidine to reduce the severity of murine KD vasculitis are not limited to PAD4 inhibition and may include decreased citrullination in the inflammasome pathway.
Collapse
Affiliation(s)
- Talita P Domiciano
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Youngho Lee
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Thacyana T Carvalho
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Daiko Wakita
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Daisy Martinon
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Prasant K Jena
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Justyna Fert-Bober
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Vanessa Borges
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Timothy R Crother
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Shuang Chen
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Debbie Moreira
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jennifer E Van Eyk
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Moshe Arditi
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kenichi Shimada
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
10
|
Day-Lewis M, Son MBF, Lo MS. Kawasaki disease: contemporary perspectives. THE LANCET. CHILD & ADOLESCENT HEALTH 2024; 8:781-792. [PMID: 39299749 DOI: 10.1016/s2352-4642(24)00169-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/25/2024] [Accepted: 06/28/2024] [Indexed: 09/22/2024]
Abstract
Kawasaki disease is a paediatric vasculitis that presents with fever, rash, conjunctivitis, mucositis, lymphadenopathy, and extremity changes, and primarily affects children younger than 5 years. Coronary artery aneurysms are observed in approximately 20% of patients without treatment. Giant coronary artery aneurysms are rare but can result in substantial morbidity and mortality due to the risk of thrombosis, stenosis, and myocardial infarction. Infants younger than 6 months and children with coronary artery abnormalities are at highest risk for the development of large or giant coronary artery aneurysms, necessitating swift identification and aggressive treatment. The children at high risk for coronary artery aneurysms warrant primary intensification therapy; however, what the most optimal adjunct therapy might be to reduce their risk is unclear and large-scale international trials are needed. Kawasaki disease is a clinical diagnosis that shares many features with other common febrile illnesses, including multisystem inflammatory syndrome in children. Identifying biomarkers that can distinguish Kawasaki disease from similar conditions and predict coronary artery aneurysm risk are needed to aid timely diagnosis, guide management, and improve patient outcomes.
Collapse
Affiliation(s)
- Megan Day-Lewis
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Mary Beth F Son
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| | - Mindy S Lo
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
Zhou QY, Liu W, Gong SX, Tian Y, Ma XF, Wang AP. Pulmonary artery smooth muscle cell pyroptosis promotes the proliferation of PASMCs by paracrine IL‑1β and IL‑18 in monocrotaline‑induced pulmonary arterial hypertensive rats. Exp Ther Med 2024; 28:394. [PMID: 39171148 PMCID: PMC11336803 DOI: 10.3892/etm.2024.12683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 07/12/2024] [Indexed: 08/23/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a common vascular disease, and pulmonary vascular remodeling is a pivotal pathophysiological mechanism of PAH. Major pathological changes of pulmonary arterial remodeling, including proliferation, hypertrophy and enhanced secretory activity, can occur in pulmonary artery smooth muscle cells (PASMCs). Multiple active factors and cytokines play important roles in PAH. However, the regulatory mechanisms of the active factors and cytokines in PAH remain unclear. The present study aimed to reveal the crucial role of PASMC pyroptosis in PAH and to elucidate the intrinsic mechanisms. To establish the PAH rat models, Sprague-Dawley rats were injected intraperitoneally with monocrotaline (MCT) at a dose of 60 mg/kg. The expression of proteins and interleukins were detected by western blotting and ELISA assay. The results indicated that the pyroptosis of PASMCs is significantly increased in MCT-induced PAH rats. Notably, pyroptotic PASMCs can secret IL-1β and IL-18 to promote the proliferation of PASMCs. On this basis, inhibiting the secretion of IL-1β and IL-18 can markedly inhibit PASMC proliferation. Collectively, the findings of the present study indicate a critical role for PASMC pyroptosis in MCT-induced PAH rats, prompting a new preventive and therapeutic strategy for PAH.
Collapse
Affiliation(s)
- Qin-Yi Zhou
- Department of Cardiology, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421002, P.R. China
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Wang Liu
- Department of Cardiology, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421002, P.R. China
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Shao-Xin Gong
- Department of Pathology, First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Ying Tian
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xiao-Feng Ma
- Department of Cardiology, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421002, P.R. China
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Ai-Ping Wang
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
12
|
Stock AT, Parsons S, Hansen JA, D'Silva DB, Starkey G, Fayed A, Lim XY, D'Costa R, Gordon CL, Wicks IP. mTOR signalling controls the formation of smooth muscle cell-derived luminal myofibroblasts during vasculitis. EMBO Rep 2024; 25:4570-4593. [PMID: 39271773 PMCID: PMC11467406 DOI: 10.1038/s44319-024-00251-1] [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: 12/19/2023] [Revised: 08/02/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
The accumulation of myofibroblasts within the intimal layer of inflamed blood vessels is a potentially catastrophic complication of vasculitis, which can lead to arterial stenosis and ischaemia. In this study, we have investigated how these luminal myofibroblasts develop during Kawasaki disease (KD), a paediatric vasculitis typically involving the coronary arteries. By performing lineage tracing studies in a murine model of KD, we reveal that luminal myofibroblasts develop independently of adventitial fibroblasts and endothelial cells, and instead derive from smooth muscle cells (SMCs). Notably, the emergence of SMC-derived luminal myofibroblasts-in both mice and patients with KD, Takayasu's arteritis and Giant Cell arteritis-coincided with activation of the mechanistic target of rapamycin (mTOR) signalling pathway. Moreover, SMC-specific deletion of mTOR signalling, or pharmacological inhibition, abrogated the emergence of luminal myofibroblasts. Thus, mTOR is an intrinsic and essential regulator of luminal myofibroblast formation that is activated in vasculitis patients and therapeutically tractable. These findings provide molecular insight into the pathogenesis of coronary artery stenosis and identify mTOR as a therapeutic target in vasculitis.
Collapse
Affiliation(s)
| | - Sarah Parsons
- Department of Forensic Medicine, Monash University, Melbourne, VIC, 3006, Australia
- Victorian Institute of Forensic Medicine, Melbourne, VIC, 3006, Australia
| | | | | | - Graham Starkey
- Liver & Intestinal Transplant Unit, Austin Health, Melbourne, VIC, 3084, Australia
- Department of Surgery, The University of Melbourne, Austin Health, Melbourne, VIC, 3084, Australia
| | - Aly Fayed
- Department of Surgery, Austin Health, Melbourne, VIC, 3084, Australia
| | - Xin Yi Lim
- Department of Infectious Diseases, Austin Health, Melbourne, VIC, 3084, Australia
| | - Rohit D'Costa
- DonateLife Victoria, Carlton, VIC, 3053, Australia
- Department of Intensive Care Medicine, Melbourne Health, Melbourne, VIC, 3084, Australia
| | - Claire L Gordon
- Department of Infectious Diseases, Austin Health, Melbourne, VIC, 3084, Australia
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3052, Australia
- North Eastern Public Health Unit, Austin Health, Melbourne, VIC, 3084, Australia
| | - Ian P Wicks
- WEHI, Melbourne, VIC, 3052, Australia.
- Rheumatology Unit, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia.
- University of Melbourne, Department of Medical Biology, Melbourne, VIC, 3052, Australia.
| |
Collapse
|
13
|
Gu W, Mirsaidi-Madjdabadi S, Ramirez F, Simonson TS, Makino A. Transcriptome meta-analysis of Kawasaki disease in humans and mice. Front Pediatr 2024; 12:1423958. [PMID: 39350793 PMCID: PMC11440715 DOI: 10.3389/fped.2024.1423958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024] Open
Abstract
Kawasaki Disease (KD) affects young children less than five years old with severe blood vessel inflammation. Despite being treatable, the causes and mechanisms remain elusive. This study conducted a meta-analysis of RNA sequencing (RNA-seq) data from human and animal models to explore KD's transcriptomic profile and evaluate animal models. We retrieved bulk and single-cell RNA-seq data from Gene Expression Omnibus, with blood and coronary artery samples from KD patients, aorta samples from KD mouse models (Lactobacillus casei cell wall extract-injected mice), and their controls. Upon consistent quality control, we applied Fisher's exact test to assess differential gene expression, followed by an enrichment analysis of overlapping genes. These studies identified 400 differentially expressed genes in blood samples of KD patients compared to controls and 413 genes in coronary artery samples. The data from KD blood and KD coronary artery samples shared only 16 differentially expressed genes. Eighty-one genes overlapped between KD human coronary arteries and KD mouse aortas, and 67 of these 81 genes were regulated in parallel in both humans and mice: 30 genes were up-regulated, and 37 were down-regulated. These included previously identified KD-upregulated genes: CD74, SFRP4, ITGA4, and IKZF1. Gene enrichment analysis revealed significant alterations in the cardiomyopathy pathway. Single-cell RNAseq showed a few significant markers, with known KD markers like S100A9, S100A8, CD74, CD14, IFITM2, and IFITM3, being overexpressed in KD cohorts. Gene profiles obtained from KD human coronary artery are more compatible with data from aorta samples of KD mice than blood samples of KD humans, validating KD animal models for identifying therapeutic targets. Although blood samples can be utilized to discover novel biomarkers, more comprehensive single-cell sequencing is required to detail gene expression in different blood cell populations. This study identifies critical genes from human and mouse tissues to help develop new treatment strategies for KD.
Collapse
Affiliation(s)
- Wanjun Gu
- Department of Medicine, University of California, San Diego, CA, United States
| | | | - Francisco Ramirez
- Department of Medicine, University of California, San Diego, CA, United States
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim University of Florida Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, United States
| | - Tatum S. Simonson
- Department of Medicine, University of California, San Diego, CA, United States
| | - Ayako Makino
- Department of Medicine, University of California, San Diego, CA, United States
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim University of Florida Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, United States
| |
Collapse
|
14
|
陈 露, 杨 轶, 赵 苗, 李 翰, 孙 文, 石 曌. [Mechanism of tetramethylpyrazine attenuates inflammatory injury in endothelial cells by activating the SIRT1 signaling pathway]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:967-973. [PMID: 39267513 PMCID: PMC11404461 DOI: 10.7499/j.issn.1008-8830.2405084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 07/24/2024] [Indexed: 09/17/2024]
Abstract
OBJECTIVES To study the effects and mechanisms of tetramethylpyrazine (TMP) on tumor necrosis factor-α (TNF-α)-induced inflammatory injury in human coronary artery endothelial cells (HCAEC). METHODS HCAEC were randomly divided into four groups: the control group (no treatment), the model group (treated with TNF-α, 50 ng/mL for 24 hours), the TMP group (pre-treated with TMP, 80 μg/mL for 12 hours followed by TNF-α treatment for 24 hours), and the SIRT1 inhibitor group (pre-treated with TMP and the specific SIRT1 inhibitor EX527 for 12 hours followed by TNF-α treatment for 24 hours). Cell viability was assessed using the CCK-8 method, lactate dehydrogenase (LDH) activity was measured using an LDH assay kit, reactive oxygen species (ROS) levels were observed using DCFH-DA staining, expression of pyroptosis-related proteins was detected by Western blot, and SIRT1 expression was analyzed using immunofluorescence staining. RESULTS Compared to the control group, the model group showed decreased cell viability, increased LDH activity, ROS level and expression of pyroptosis-related proteins, and decreased SIRT1 expression (P<0.05). Compared to the model group, the TMP group exhibited increased cell viability, decreased LDH activity, ROS level and expression of pyroptosis-related proteins, and increased SIRT1 expression (P<0.05). In comparison to the TMP group, the SIRT1 inhibitor group showed decreased cell viability, increased LDH activity, ROS level and expression of pyroptosis-related proteins, and decreased SIRT1 expression (P<0.05). CONCLUSIONS TMP may attenuate TNF-α-induced inflammatory injury in HCAEC, which is associated with the inhibition of pyroptosis and activation of the SIRT1 signaling pathway.
Collapse
|
15
|
Wang Y, Liu Y, Wang N, Liu Z, Qian G, Li X, Huang H, Zhuo W, Xu L, Zhang J, Lv H, Gao Y. Identification of novel mitophagy-related biomarkers for Kawasaki disease by integrated bioinformatics and machine-learning algorithms. Transl Pediatr 2024; 13:1439-1456. [PMID: 39263286 PMCID: PMC11384439 DOI: 10.21037/tp-24-230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/06/2024] [Indexed: 09/13/2024] Open
Abstract
Background Kawasaki disease (KD) is a systemic vasculitis primarily affecting the coronary arteries in children. Despite growing attention to its symptoms and pathogenesis, the exact mechanisms of KD remain unclear. Mitophagy plays a critical role in inflammation regulation, however, its significance in KD has only been minimally explored. This study sought to identify crucial mitophagy-related biomarkers and their mechanisms in KD, focusing on their association with immune cells in peripheral blood. Methods This research used four datasets from the Gene Expression Omnibus (GEO) database that were categorized as the merged and validation datasets. Screening for differentially expressed mitophagy-related genes (DE-MRGs) was conducted, followed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. A weighted gene co-expression network analysis (WGCNA) identified the hub module, while machine-learning algorithms [random forest-recursive feature elimination (RF-RFE) and support vector machine-recursive feature elimination (SVM-RFE)] pinpointed the hub genes. Receiver operating characteristic (ROC) curves were generated for these genes. Additionally, the CIBERSORT algorithm was used to assess the infiltration of 22 immune cell types to explore their correlations with hub genes. Interactions between transcription factors (TFs), genes, and Gene-microRNAs (miRNAs) of hub genes were mapped using the NetworkAnalyst platform. The expression difference of the hub genes was validated using quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). Results Initially, 306 DE-MRGs were identified between the KD patients and healthy controls. The enrichment analysis linked these MRGs to autophagy, mitochondrial function, and inflammation. The WGCNA revealed a hub module of 47 KD-associated DE-MRGs. The machine-learning algorithms identified cytoskeleton-associated protein 4 (CKAP4) and serine-arginine protein kinase 1 (SRPK1) as critical hub genes. In the merged dataset, the area under the curve (AUC) values for CKAP4 and SRPK1 were 0.933 [95% confidence interval (CI): 0.901 to 0.964] and 0.936 (95% CI: 0.906 to 0.966), respectively, indicating high diagnostic potential. The validation dataset results corroborated these findings with AUC values of 0.872 (95% CI: 0.741 to 1.000) for CKAP4 and 0.878 (95% CI: 0.750 to 1.000) for SRPK1. The CIBERSORT analysis connected CKAP4 and SRPK1 with specific immune cells, including activated cluster of differentiation 4 (CD4) memory T cells. TFs such as MAZ, SAP30, PHF8, KDM5B, miRNAs like hsa-mir-7-5p play essential roles in regulating these hub genes. The qRT-PCR results confirmed the differential expression of these genes between the KD patients and healthy controls. Conclusions CKAP4 and SRPK1 emerged as promising diagnostic biomarkers for KD. These genes potentially influence the progression of KD through mitophagy regulation.
Collapse
Affiliation(s)
- Yan Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
- Department of Cardiology, The Affiliated Xuzhou Children's Hospital of Xuzhou Medical University, Xuzhou, China
| | - Ying Liu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Nana Wang
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, China
| | - Zhiheng Liu
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, China
| | - Guanghui Qian
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Xuan Li
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, China
| | - Hongbiao Huang
- Department of Pediatrics, Fujian Provincial Hospital, Fujian Provincial Clinical College of Fujian Medical University, Fuzhou, China
| | - Wenyu Zhuo
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Lei Xu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Jiaying Zhang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Haitao Lv
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, China
| | - Yang Gao
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
- Department of Pediatrics, The First People's Hospital of Lianyungang, Xuzhou Medical University Affiliated Hospital of Lianyungang (Lianyungang Clinical College of Nanjing Medical University), Lianyungang, China
| |
Collapse
|
16
|
Noval Rivas M, Kocatürk B, Franklin BS, Arditi M. Platelets in Kawasaki disease: mediators of vascular inflammation. Nat Rev Rheumatol 2024; 20:459-472. [PMID: 38886559 DOI: 10.1038/s41584-024-01119-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2024] [Indexed: 06/20/2024]
Abstract
Kawasaki disease, a systemic vasculitis that affects young children and can result in coronary artery aneurysms, is the leading cause of acquired heart disease among children. A hallmark of Kawasaki disease is increased blood platelet counts and platelet activation, which is associated with an increased risk of developing resistance to intravenous immunoglobulin and coronary artery aneurysms. Platelets and their releasate, including granules, microparticles, microRNAs and transcription factors, can influence innate immunity, enhance inflammation and contribute to vascular remodelling. Growing evidence indicates that platelets also interact with immune and non-immune cells to regulate inflammation. Platelets boost NLRP3 inflammasome activation and IL-1β production by human immune cells by releasing soluble mediators. Activated platelets form aggregates with leukocytes, such as monocytes and neutrophils, enhancing numerous functions of these cells and promoting thrombosis and inflammation. Leukocyte-platelet aggregates are increased in children with Kawasaki disease during the acute phase of the disease and can be used as biomarkers for disease severity. Here we review the role of platelets in Kawasaki disease and discuss progress in understanding the immune-effector role of platelets in amplifying inflammation related to Kawasaki disease vasculitis and therapeutic strategies targeting platelets or platelet-derived molecules.
Collapse
Affiliation(s)
- Magali Noval Rivas
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children's, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Begüm Kocatürk
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Bernardo S Franklin
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Moshe Arditi
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children's, Cedars Sinai Medical Center, Los Angeles, CA, USA.
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
17
|
Lombardi Pereira AP, Aubuchon E, Moreira DP, Lane M, Carvalho TT, Mesquita TRR, Lee Y, Crother TR, Porritt RA, Verri WA, Noval Rivas M, Arditi M. Long-term cardiovascular inflammation and fibrosis in a murine model of vasculitis induced by Lactobacillus casei cell wall extract. Front Immunol 2024; 15:1411979. [PMID: 38989288 PMCID: PMC11234797 DOI: 10.3389/fimmu.2024.1411979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/11/2024] [Indexed: 07/12/2024] Open
Abstract
Background Kawasaki disease (KD), an acute febrile illness and systemic vasculitis, is the leading cause of acquired heart disease in children in industrialized countries. KD leads to the development of coronary artery aneurysms (CAA) in affected children, which may persist for months and even years after the acute phase of the disease. There is an unmet need to characterize the immune and pathological mechanisms of the long-term complications of KD. Methods We examined cardiovascular complications in the Lactobacillus casei cell wall extract (LCWE) mouse model of KD-like vasculitis over 4 months. The long-term immune, pathological, and functional changes occurring in cardiovascular lesions were characterized by histological examination, flow cytometric analysis, immunofluorescent staining of cardiovascular tissues, and transthoracic echocardiogram. Results CAA and abdominal aorta dilations were detected up to 16 weeks following LCWE injection and initiation of acute vasculitis. We observed alterations in the composition of circulating immune cell profiles, such as increased monocyte frequencies in the acute phase of the disease and higher counts of neutrophils. We determined a positive correlation between circulating neutrophil and inflammatory monocyte counts and the severity of cardiovascular lesions early after LCWE injection. LCWE-induced KD-like vasculitis was associated with myocarditis and myocardial dysfunction, characterized by diminished ejection fraction and left ventricular remodeling, which worsened over time. We observed extensive fibrosis within the inflamed cardiac tissue early in the disease and myocardial fibrosis in later stages. Conclusion Our findings indicate that increased circulating neutrophil counts in the acute phase are a reliable predictor of cardiovascular inflammation severity in LCWE-injected mice. Furthermore, long-term cardiac complications stemming from inflammatory cell infiltrations in the aortic root and coronary arteries, myocardial dysfunction, and myocardial fibrosis persist over long periods and are still detected up to 16 weeks after LCWE injection.
Collapse
Affiliation(s)
- Ana Paula Lombardi Pereira
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, Brazil
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Emily Aubuchon
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Debbie P. Moreira
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Malcolm Lane
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Thacyana T. Carvalho
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | | | - Youngho Lee
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Timothy R. Crother
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Rebecca A. Porritt
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Waldiceu A. Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, Brazil
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Moshe Arditi
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
18
|
Jena PK, Wakita D, Gomez AC, Carvalho TT, Atici AE, Narayanan M, Lee Y, Fishbein MC, Cani PD, de Vos WM, Underhill DM, Devkota S, Chen S, Shimada K, Crother TR, Arditi M, Rivas MN. The intestinal microbiota contributes to the development of immune-mediated cardiovascular inflammation and vasculitis in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.28.596258. [PMID: 38853964 PMCID: PMC11160596 DOI: 10.1101/2024.05.28.596258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Alterations in the intestinal microbiota contribute to the pathogenesis of various cardiovascular disorders, but how they affect the development of Kawasaki disease (KD), an acute pediatric vasculitis, remains unclear. We report that depleting the gut microbiota reduces the development of cardiovascular inflammation in a murine model mimicking KD vasculitis. The development of cardiovascular lesions was associated with alterations in the intestinal microbiota composition and, notably, a decreased abundance of Akkermansia muciniphila and Faecalibacterium prausnitzii. Oral supplementation with either of these live or pasteurized individual bacteria, or with short-chain fatty acids (SCFAs) produced by them, attenuated cardiovascular inflammation. Treatment with Amuc_1100, the TLR-2 signaling outer membrane protein from A. muciniphila , also decreased the severity of vascular inflammation. This study reveals an underappreciated gut microbiota-cardiovascular inflammation axis in KD vasculitis pathogenesis and identifies specific intestinal commensals that regulate vasculitis in mice by producing metabolites or via extracellular proteins acting on gut barrier function. IN BRIEF It remains unclear whether changes in the intestinal microbiota composition are involved in the development of cardiovascular lesions associated with Kawasaki disease (KD), an immune-mediated vasculitis. Jena et al. observe alterations in the intestinal microbiota composition of mice developing vasculitis, characterized by reduced A. muciniphila and F. prausnitzii . Oral supplementation with either of these bacteria, live or pasteurized, or with bacteria-produced short-chain fatty acids (SCFAs) or Amuc_1100, the TLR-2 signaling outer membrane protein of A. muciniphila , was sufficient to alleviate the development of cardiovascular lesions in mice by promoting intestinal barrier function. HIGHLIGHTS Absence or depletion of the microbiota decreases the severity of vasculitis in a murine model mimicking KD vasculitis. Supplementation of B. wadsworthia and B. fragilis promotes murine KD vasculitis. Decreased abundances of F. prausnitzii and A. muciniphila are associated with the development of cardiovascular lesions in mice. Supplementation with either live or pasteurized A. muciniphila and F. prausnitzii, or the TLR-2 signaling Amuc_1100, reduces the severity of vasculitis by promoting gut barrier function.
Collapse
|
19
|
Zhang Z, Kean IRL, Dratva LM, Clark JA, Syrimi E, Khan N, Daubney E, White D, O'Neill L, Chisholm C, Payne C, Benkenstein S, Kupiec K, Galassini R, Wright V, Winmill H, Robbins C, Brown K, Ramnarayan P, Scholefield B, Peters M, Klein N, Montgomery H, Meyer KB, Teichmann SA, Bryant C, Taylor G, Pathan N. Enhanced CD95 and interleukin 18 signalling accompany T cell receptor Vβ21.3+ activation in multi-inflammatory syndrome in children. Nat Commun 2024; 15:4227. [PMID: 38762592 PMCID: PMC11102542 DOI: 10.1038/s41467-024-48699-y] [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: 10/31/2022] [Accepted: 05/10/2024] [Indexed: 05/20/2024] Open
Abstract
Multisystem inflammatory syndrome in children is a post-infectious presentation SARS-CoV-2 associated with expansion of the T cell receptor Vβ21.3+ T-cell subgroup. Here we apply muti-single cell omics to compare the inflammatory process in children with acute respiratory COVID-19 and those presenting with non SARS-CoV-2 infections in children. Here we show that in Multi-Inflammatory Syndrome in Children (MIS-C), the natural killer cell and monocyte population demonstrate heightened CD95 (Fas) and Interleuking 18 receptor expression. Additionally, TCR Vβ21.3+ CD4+ T-cells exhibit skewed differentiation towards T helper 1, 17 and regulatory T cells, with increased expression of the co-stimulation receptors ICOS, CD28 and interleukin 18 receptor. We observe no functional evidence for NLRP3 inflammasome pathway overactivation, though MIS-C monocytes show elevated active caspase 8. This, coupled with raised IL18 mRNA expression in CD16- NK cells on single cell RNA sequencing analysis, suggests interleukin 18 and CD95 signalling may trigger activation of TCR Vβ21.3+ T-cells in MIS-C, driven by increased IL-18 production from activated monocytes and CD16- Natural Killer cells.
Collapse
MESH Headings
- Humans
- Interleukin-18/metabolism
- Child
- Signal Transduction
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- fas Receptor/metabolism
- fas Receptor/genetics
- Monocytes/immunology
- Monocytes/metabolism
- Systemic Inflammatory Response Syndrome/immunology
- Systemic Inflammatory Response Syndrome/metabolism
- COVID-19/immunology
- COVID-19/virology
- COVID-19/metabolism
- COVID-19/complications
- Inflammasomes/metabolism
- Inflammasomes/immunology
- SARS-CoV-2/immunology
- Adolescent
- Male
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Female
- Child, Preschool
- Single-Cell Analysis
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD28 Antigens/metabolism
- Lymphocyte Activation/immunology
- Receptors, Interleukin-18/metabolism
- Receptors, Interleukin-18/genetics
- Receptors, Interleukin-18/immunology
Collapse
Affiliation(s)
- Zhenguang Zhang
- Departments of Paediatrics, University of Cambridge, Cambridge, UK
| | - Iain R L Kean
- Departments of Paediatrics, University of Cambridge, Cambridge, UK
| | - Lisa M Dratva
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - John A Clark
- Departments of Paediatrics, University of Cambridge, Cambridge, UK
| | - Eleni Syrimi
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Naeem Khan
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Esther Daubney
- Paediatric Intensive Care Unit, Addenbrookes Hospital, Cambridge, UK
| | - Deborah White
- Paediatric Intensive Care Unit, Addenbrookes Hospital, Cambridge, UK
| | - Lauran O'Neill
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
| | - Catherine Chisholm
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
| | - Caroline Payne
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
| | - Sarah Benkenstein
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
| | - Klaudia Kupiec
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
| | | | - Victoria Wright
- Department of Paediatrics, Imperial College London, London, UK
| | - Helen Winmill
- Paediatric Intensive Care Unit, Birmingham Children's Hospital, Birmingham, UK
| | - Ceri Robbins
- Paediatric Intensive Care Unit, Birmingham Children's Hospital, Birmingham, UK
| | - Katherine Brown
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
| | | | - Barnaby Scholefield
- Paediatric Intensive Care Unit, Birmingham Children's Hospital, Birmingham, UK
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Mark Peters
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
- Departments of Paediatrics, University College London, London, UK
| | - Nigel Klein
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
- Departments of Paediatrics, University College London, London, UK
| | | | - Kerstin B Meyer
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Department of Theory of Condensed Matter, Cavendish Laboratory, Department of Physics University of Cambridge, Cambridge, UK
| | - Clare Bryant
- Department of Medicine, University of Cambridge, Cambridge, UK.
| | - Graham Taylor
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
| | - Nazima Pathan
- Departments of Paediatrics, University of Cambridge, Cambridge, UK.
- Paediatric Intensive Care Unit, Addenbrookes Hospital, Cambridge, UK.
| |
Collapse
|
20
|
Cao J, Li C, Cui Z, Deng S, Lei T, Liu W, Yang H, Chen P. Spatial Transcriptomics: A Powerful Tool in Disease Understanding and Drug Discovery. Theranostics 2024; 14:2946-2968. [PMID: 38773973 PMCID: PMC11103497 DOI: 10.7150/thno.95908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 04/25/2024] [Indexed: 05/24/2024] Open
Abstract
Recent advancements in modern science have provided robust tools for drug discovery. The rapid development of transcriptome sequencing technologies has given rise to single-cell transcriptomics and single-nucleus transcriptomics, increasing the accuracy of sequencing and accelerating the drug discovery process. With the evolution of single-cell transcriptomics, spatial transcriptomics (ST) technology has emerged as a derivative approach. Spatial transcriptomics has emerged as a hot topic in the field of omics research in recent years; it not only provides information on gene expression levels but also offers spatial information on gene expression. This technology has shown tremendous potential in research on disease understanding and drug discovery. In this article, we introduce the analytical strategies of spatial transcriptomics and review its applications in novel target discovery and drug mechanism unravelling. Moreover, we discuss the current challenges and issues in this research field that need to be addressed. In conclusion, spatial transcriptomics offers a new perspective for drug discovery.
Collapse
Affiliation(s)
- Junxian Cao
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Analysis of Complex Effects of Proprietary Chinese Medicine, Hunan Provincial Key Laboratory, Yongzhou City, Hunan Province, China
| | - Caifeng Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Zhao Cui
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Shiwen Deng
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Analysis of Complex Effects of Proprietary Chinese Medicine, Hunan Provincial Key Laboratory, Yongzhou City, Hunan Province, China
| | - Tong Lei
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Wei Liu
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Hongjun Yang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Analysis of Complex Effects of Proprietary Chinese Medicine, Hunan Provincial Key Laboratory, Yongzhou City, Hunan Province, China
| | - Peng Chen
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Analysis of Complex Effects of Proprietary Chinese Medicine, Hunan Provincial Key Laboratory, Yongzhou City, Hunan Province, China
| |
Collapse
|
21
|
Huang J, Zhao C, Zhang S. Semaphorin 7A promotes endothelial permeability and inflammation via plexin C1 and integrin β1 in Kawasaki disease. BMC Pediatr 2024; 24:285. [PMID: 38678170 PMCID: PMC11055240 DOI: 10.1186/s12887-024-04766-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 04/16/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Kawasaki disease (KD) is a pediatric systemic vasculitis characterized by endothelial cell dysfunction. Semaphorin 7A (Sema7A) has been reported to regulate endothelial phenotypes associated with cardiovascular diseases, while its role in KD remains unknown. This study aims to investigate the effect of Sema7A on endothelial permeability and inflammatory response in KD conditions. METHODS Blood samples were collected from 68 KD patients and 25 healthy children (HC). The levels of Sema7A and A Disintegrin and Metalloprotease 17 (ADAM17) in serum were measured by enzyme-linked immunosorbent assay (ELISA), and Sema7A expression in blood cells was analyzed by flow cytometry. Ex vivo monocytes were used for Sema7A shedding assays. In vitro human coronary artery endothelial cells (HCAECs) were cultured in KD sera and stimulated with Sema7A, and TNF-α, IL-1β, IL-6, and IL-18 of HCAECs were measured by ELISA and qRT-PCR. HCAECs monolayer permeability was measured by FITC-dextran. RESULTS The serum level of Sema7A was significantly higher in KD patients than in HC and correlated with disease severity. Monocytes were identified as one of the source of elevated serum Sema7A, which implicates a process of ADAM17-dependent shedding. Sera from KD patients induced upregulation of plexin C1 and integrin β1 in HCAECs compared to sera from HC. Sema7A mediated the proinflammatory cytokine production of HCAECs in an integrin β1-dependent manner, while both plexin C1 and integrin β1 contributed to Sema7A-induced HCAEC hyperpermeability. CONCLUSIONS Sema7A is involved in the progression of KD vasculitis by promoting endothelial permeability and inflammation through a plexin C1 and integrin β1-dependent pathway. Sema7A may serve as a potential biomarker and therapeutic target in the prognosis and treatment of KD.
Collapse
Affiliation(s)
- Junhua Huang
- School of Medical Technology, Xi'an Medical University, Xi'an, 710021, Shaanxi Province, China
| | - Chuanmei Zhao
- Department of Clinical Laboratory, Xi'an Children's Hospital, Xi'an, 710003, Shaanxi Province, China
| | - Shuwan Zhang
- Department of Clinical Laboratory, Xi'an Children's Hospital, Xi'an, 710003, Shaanxi Province, China.
| |
Collapse
|
22
|
Xu T, Chen T, Fang H, Shen X, Shen X, Tang Z, Zhao J. Human Umbilical Cord Mesenchymal Stem Cells Repair Endothelial Injury and Dysfunction by Regulating NLRP3 to Inhibit Endothelial Cell Pyroptosis in Kawasaki Disease. Inflammation 2024; 47:483-502. [PMID: 37948033 DOI: 10.1007/s10753-023-01921-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/26/2023] [Accepted: 10/17/2023] [Indexed: 11/12/2023]
Abstract
Vascular endothelial inflammation and endothelial dysfunction are the main causes of endothelial injury in Kawasaki disease (KD). Human umbilical cord-derived mesenchymal stem cells (Huc-MSCs) have multiple functions in immune regulation. This study examined whether Huc-MSCs inhibited endothelial inflammation and improved endothelial function in KD through constructing cell and in vivo animal KD vasculitis models. The pyroptosis factor NOD-like receptor protein 3 (NLRP3) was involved in the inflammatory process in the acute phase of KD. After tail vein injection of Huc-MSCs, inflammatory cell infiltration and the expression of pyroptosis-related proteins in the LCWE-induced KD mouse vasculitis model were significantly reduced. In vitro, NLRP3-dependent pyroptosis successfully induced human umbilical vein endothelial cell (HUVEC) damage. Huc-MSCs effectively increased the abilities of impaired HUVECs to proliferate, migrate, invade, and form vessel-like tubes, while inhibiting their apoptosis, suggesting that Huc-MSCs can reduce inflammation and improve vascular endothelial function by inhibiting the NLRP3-dependent pyroptosis pathway in KD, providing a possibility and novel target for KD endothelial injury and dysfunction.
Collapse
Affiliation(s)
- Ting Xu
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China
- Research Institute of Comparative Medicine, Nantong University, Nantong Jiangsu Province, 226001, China
| | - Tao Chen
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China
| | - Hao Fang
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China
- Research Institute of Comparative Medicine, Nantong University, Nantong Jiangsu Province, 226001, China
| | - Xiwei Shen
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China
- Research Institute of Comparative Medicine, Nantong University, Nantong Jiangsu Province, 226001, China
| | - Xianjuan Shen
- Department of Clinical Laboratory, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China
| | - Zhiyuan Tang
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| | - Jianmei Zhao
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China.
| |
Collapse
|
23
|
Mesquita T, Lin YN, Chen S, Lee Y, Miguel-dos-Santos R, Atici AE, Fishbein MC, Rivas MN, Arditi M, Cingolani E. Inhibition of IL-1 Ameliorates Cardiac Dysfunction and Arrhythmias in a Murine Model of Kawasaki Disease. Arterioscler Thromb Vasc Biol 2024; 44:e117-e130. [PMID: 38385289 PMCID: PMC10978283 DOI: 10.1161/atvbaha.123.320382] [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: 11/03/2023] [Accepted: 02/09/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND Kawasaki disease (KD) is an acute febrile illness and systemic vasculitis often associated with cardiac sequelae, including arrhythmias. Abundant evidence indicates a central role for IL (interleukin)-1 and TNFα (tumor necrosis factor-alpha) signaling in the formation of arterial lesions in KD. We aimed to investigate the mechanisms underlying the development of electrophysiological abnormalities in a murine model of KD vasculitis. METHODS Lactobacillus casei cell wall extract-induced KD vasculitis model was used to investigate the therapeutic efficacy of clinically relevant IL-1Ra (IL-1 receptor antagonist) and TNFα neutralization. Echocardiography, in vivo electrophysiology, whole-heart optical mapping, and imaging were performed. RESULTS KD vasculitis was associated with impaired ejection fraction, increased ventricular tachycardia, prolonged repolarization, and slowed conduction velocity. Since our transcriptomic analysis of human patients showed elevated levels of both IL-1β and TNFα, we asked whether either cytokine was linked to the development of myocardial dysfunction. Remarkably, only inhibition of IL-1 signaling by IL-1Ra but not TNFα neutralization was able to prevent changes in ejection fraction and arrhythmias, whereas both IL-1Ra and TNFα neutralization significantly improved vasculitis and heart vessel inflammation. The treatment of L casei cell wall extract-injected mice with IL-1Ra also restored conduction velocity and improved the organization of Cx43 (connexin 43) at the intercalated disk. In contrast, in mice with gain of function of the IL-1 signaling pathway, L casei cell wall extract induced spontaneous ventricular tachycardia and premature deaths. CONCLUSIONS Our results characterize the electrophysiological abnormalities associated with L casei cell wall extract-induced KD and show that IL-1Ra is more effective in preventing KD-induced myocardial dysfunction and arrhythmias than anti-TNFα therapy. These findings support the advancement of clinical trials using IL-1Ra in patients with KD.
Collapse
Affiliation(s)
- Thassio Mesquita
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yen-Nien Lin
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Shuang Chen
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, California, USA
- Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Youngho Lee
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, California, USA
- Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | | | - Asli E. Atici
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, California, USA
- Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Michael C. Fishbein
- Department of Pathology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, California, USA
- Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Moshe Arditi
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, California, USA
- Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Eugenio Cingolani
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Vasculitis are a group of heterogeneous conditions characterized by chronic inflammation of blood vessels, leading to tissue destruction and organ failure. Vasculitis is an inflammatory process in which immune effector cells infiltrate blood vessels and surrounding tissues. The involvement of inflammasomes seems to occur during inflammatory processes. RECENT FINDINGS Studies have emphasized that genetic susceptibility is an important aspect of the pathogenesis of vasculitis. The innate immune system is a major contributor to these inflammatory diseases, suggesting that the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome plays a key role. NLRP3 activation causes the assembly of a large multiprotein and leads to the secretion of bioactive interleukin (IL)-1β and IL-18 as well as the induction of inflammatory cell death, termed pyroptosis. Accumulating evidence confirms the involvement of this cascade in sterile inflammatory diseases and other vascular diseases. SUMMARY In this review, we will summarize the current state of knowledge regarding the role of NLRP3 inflammasome in vascular diseases, and discuss the potential of the NLRP3 inflammasome as a therapeutic target.
Collapse
Affiliation(s)
- Kamel Hamzaoui
- Laboratory Research 19SP02 'Chronic Pathologies: From Genome to Management', Department of Respiratory Diseases, Tunis El Manar University
| | - Agnès Hamzaoui
- Laboratory Research 19SP02 'Chronic Pathologies: From Genome to Management', Department of Respiratory Diseases, Tunis El Manar University
- Department of Respiratory Diseases, Pavillon B. Abderrahman Mami Hospital, Tunis, Tunisia
| |
Collapse
|
25
|
Tsoukas P, Yeung RSM. Kawasaki Disease-Associated Cytokine Storm Syndrome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1448:365-383. [PMID: 39117827 DOI: 10.1007/978-3-031-59815-9_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Kawasaki disease (KD) is a hyperinflammatory syndrome manifesting as an acute systemic vasculitis characterized by fever, nonsuppurative conjunctival injection, rash, oral mucositis, extremity changes, and cervical lymphadenopathy. KD predominantly affects young children and shares clinical features and immunobiology with other hyperinflammation syndromes including systemic juvenile idiopathic arthritis (sJIA) and multisystem inflammatory syndrome in children (MIS-C). Cytokine storm syndrome (CSS) is an acute complication in ~2% of KD patients; however, the incidence is likely underestimated as many clinical and laboratory features of both diseases overlap. CSS should be entertained when a child with KD is unresponsive to IVIG therapy with recalcitrant fever. Early recognition and prompt institution of immunomodulatory treatment can substantially reduce the mortality and morbidity of CSS in KD. Given the known pathogenetic role of IL-1β in both syndromes, the early use of IL-1 blockers in refractory KD with CSS deserves consideration.
Collapse
Affiliation(s)
- Paul Tsoukas
- Division of Rheumatology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Paediatrics, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Rae S M Yeung
- Division of Rheumatology, The Hospital for Sick Children, Toronto, ON, Canada.
- Department of Paediatrics, Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
- Department of Immunology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
26
|
Atici AE, Crother TR, Noval Rivas M. Mitochondrial quality control in health and cardiovascular diseases. Front Cell Dev Biol 2023; 11:1290046. [PMID: 38020895 PMCID: PMC10657886 DOI: 10.3389/fcell.2023.1290046] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Cardiovascular diseases (CVDs) are one of the primary causes of mortality worldwide. An optimal mitochondrial function is central to supplying tissues with high energy demand, such as the cardiovascular system. In addition to producing ATP as a power source, mitochondria are also heavily involved in adaptation to environmental stress and fine-tuning tissue functions. Mitochondrial quality control (MQC) through fission, fusion, mitophagy, and biogenesis ensures the clearance of dysfunctional mitochondria and preserves mitochondrial homeostasis in cardiovascular tissues. Furthermore, mitochondria generate reactive oxygen species (ROS), which trigger the production of pro-inflammatory cytokines and regulate cell survival. Mitochondrial dysfunction has been implicated in multiple CVDs, including ischemia-reperfusion (I/R), atherosclerosis, heart failure, cardiac hypertrophy, hypertension, diabetic and genetic cardiomyopathies, and Kawasaki Disease (KD). Thus, MQC is pivotal in promoting cardiovascular health. Here, we outline the mechanisms of MQC and discuss the current literature on mitochondrial adaptation in CVDs.
Collapse
Affiliation(s)
- Asli E. Atici
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Timothy R. Crother
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
27
|
Xu Y, Jiang K, Su F, Deng R, Cheng Z, Wang D, Yu Y, Xiang Y. A transient wave of Bhlhe41 + resident macrophages enables remodeling of the developing infarcted myocardium. Cell Rep 2023; 42:113174. [PMID: 37751357 DOI: 10.1016/j.celrep.2023.113174] [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: 04/13/2023] [Revised: 08/03/2023] [Accepted: 09/06/2023] [Indexed: 09/28/2023] Open
Abstract
The immune system plays a critical role during myocardial injury, contributing to repair and remodeling post myocardial infarction (MI). The myocardial infarct and border zone exhibit high heterogeneity, in turn leading to reconstructing macrophage subsets and specific functions. Here we use a combination of single-cell RNA sequencing, spatial transcriptomes, and reporter mice to characterize temporal-spatial dynamics of cardiac macrophage subtype in response to MI. We identify that transient appearance of monocyte-derived Bhlhe41+ Mφs in the "developing" infarct zone peaked at day 7, while other monocyte-derived macrophages are identified in "old" infarct zone. Functional characterization by co-culture of Bhlhe41+ Mφs with cardiomyocytes and fibroblasts or depletion of Bhlhe41+ Mφs unveils a crucial contribution of Bhlhe41+ Mφs in suppression of myofibroblast activation. This work highlights the importance of Bhlhe41+ Mφ phenotype and plasticity in preventing excessive fibrosis and limiting the expansion of developing infarct area.
Collapse
Affiliation(s)
- Yue Xu
- Shanghai East Hospital, Key Laboratory of Arrhythmias of the Ministry of Education of China, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Kai Jiang
- Shanghai East Hospital, Key Laboratory of Arrhythmias of the Ministry of Education of China, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Fanghua Su
- Shanghai East Hospital, Key Laboratory of Arrhythmias of the Ministry of Education of China, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Ruhua Deng
- Shanghai East Hospital, Key Laboratory of Arrhythmias of the Ministry of Education of China, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Zhiyang Cheng
- Shanghai East Hospital, Key Laboratory of Arrhythmias of the Ministry of Education of China, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Dandan Wang
- Shanghai East Hospital, Key Laboratory of Arrhythmias of the Ministry of Education of China, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yong Yu
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yaozu Xiang
- Shanghai East Hospital, Key Laboratory of Arrhythmias of the Ministry of Education of China, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| |
Collapse
|
28
|
Kasmani MY, Topchyan P, Brown AK, Brown RJ, Wu X, Chen Y, Khatun A, Alson D, Wu Y, Burns R, Lin CW, Kudek MR, Sun J, Cui W. A spatial sequencing atlas of age-induced changes in the lung during influenza infection. Nat Commun 2023; 14:6597. [PMID: 37852965 PMCID: PMC10584893 DOI: 10.1038/s41467-023-42021-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 09/26/2023] [Indexed: 10/20/2023] Open
Abstract
Influenza virus infection causes increased morbidity and mortality in the elderly. Aging impairs the immune response to influenza, both intrinsically and because of altered interactions with endothelial and pulmonary epithelial cells. To characterize these changes, we performed single-cell RNA sequencing (scRNA-seq), spatial transcriptomics, and bulk RNA sequencing (bulk RNA-seq) on lung tissue from young and aged female mice at days 0, 3, and 9 post-influenza infection. Our analyses identified dozens of key genes differentially expressed in kinetic, age-dependent, and cell type-specific manners. Aged immune cells exhibited altered inflammatory, memory, and chemotactic profiles. Aged endothelial cells demonstrated characteristics of reduced vascular wound healing and a prothrombotic state. Spatial transcriptomics identified novel profibrotic and antifibrotic markers expressed by epithelial and non-epithelial cells, highlighting the complex networks that promote fibrosis in aged lungs. Bulk RNA-seq generated a timeline of global transcriptional activity, showing increased expression of genes involved in inflammation and coagulation in aged lungs. Our work provides an atlas of high-throughput sequencing methodologies that can be used to investigate age-related changes in the response to influenza virus, identify novel cell-cell interactions for further study, and ultimately uncover potential therapeutic targets to improve health outcomes in the elderly following influenza infection.
Collapse
Affiliation(s)
- Moujtaba Y Kasmani
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, 53226, USA
| | - Paytsar Topchyan
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, 53226, USA
| | - Ashley K Brown
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, 53226, USA
| | - Ryan J Brown
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, 53226, USA
| | - Xiaopeng Wu
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, 53226, USA
| | - Yao Chen
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, 53226, USA
| | - Achia Khatun
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, 53226, USA
| | - Donia Alson
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Yue Wu
- Carter Immunology Center, University of Virginia, Charlottesville, VA, 22908, USA
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, 22908, USA
| | - Robert Burns
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, 53226, USA
| | - Chien-Wei Lin
- Department of Biostatistics, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Matthew R Kudek
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Jie Sun
- Carter Immunology Center, University of Virginia, Charlottesville, VA, 22908, USA
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, 22908, USA
| | - Weiguo Cui
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, 53226, USA.
- Department of Pathology, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
29
|
Zhou R, Yang G, Zhang Y, Wang Y. Spatial transcriptomics in development and disease. MOLECULAR BIOMEDICINE 2023; 4:32. [PMID: 37806992 PMCID: PMC10560656 DOI: 10.1186/s43556-023-00144-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/29/2023] [Indexed: 10/10/2023] Open
Abstract
The proper functioning of diverse biological systems depends on the spatial organization of their cells, a critical factor for biological processes like shaping intricate tissue functions and precisely determining cell fate. Nonetheless, conventional bulk or single-cell RNA sequencing methods were incapable of simultaneously capturing both gene expression profiles and the spatial locations of cells. Hence, a multitude of spatially resolved technologies have emerged, offering a novel dimension for investigating regional gene expression, spatial domains, and interactions between cells. Spatial transcriptomics (ST) is a method that maps gene expression in tissue while preserving spatial information. It can reveal cellular heterogeneity, spatial organization and functional interactions in complex biological systems. ST can also complement and integrate with other omics methods to provide a more comprehensive and holistic view of biological systems at multiple levels of resolution. Since the advent of ST, new methods offering higher throughput and resolution have become available, holding significant potential to expedite fresh insights into comprehending biological complexity. Consequently, a rapid increase in associated research has occurred, using these technologies to unravel the spatial complexity during developmental processes or disease conditions. In this review, we summarize the recent advancement of ST in historical, technical, and application contexts. We compare different types of ST methods based on their principles and workflows, and present the bioinformatics tools for analyzing and integrating ST data with other modalities. We also highlight the applications of ST in various domains of biomedical research, especially development and diseases. Finally, we discuss the current limitations and challenges in the field, and propose the future directions of ST.
Collapse
Affiliation(s)
- Ran Zhou
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gaoxia Yang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yan Zhang
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Yuan Wang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
30
|
Lin PK, Davis GE. Extracellular Matrix Remodeling in Vascular Disease: Defining Its Regulators and Pathological Influence. Arterioscler Thromb Vasc Biol 2023; 43:1599-1616. [PMID: 37409533 PMCID: PMC10527588 DOI: 10.1161/atvbaha.123.318237] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/23/2023] [Indexed: 07/07/2023]
Abstract
Because of structural and cellular differences (ie, degrees of matrix abundance and cross-linking, mural cell density, and adventitia), large and medium-sized vessels, in comparison to capillaries, react in a unique manner to stimuli that induce vascular disease. A stereotypical vascular injury response is ECM (extracellular matrix) remodeling that occurs particularly in larger vessels in response to injurious stimuli, such as elevated angiotensin II, hyperlipidemia, hyperglycemia, genetic deficiencies, inflammatory cell infiltration, or exposure to proinflammatory mediators. Even with substantial and prolonged vascular damage, large- and medium-sized arteries, persist, but become modified by (1) changes in vascular wall cellularity; (2) modifications in the differentiation status of endothelial cells, vascular smooth muscle cells, or adventitial stem cells (each can become activated); (3) infiltration of the vascular wall by various leukocyte types; (4) increased exposure to critical growth factors and proinflammatory mediators; and (5) marked changes in the vascular ECM, that remodels from a homeostatic, prodifferentiation ECM environment to matrices that instead promote tissue reparative responses. This latter ECM presents previously hidden matricryptic sites that bind integrins to signal vascular cells and infiltrating leukocytes (in coordination with other mediators) to proliferate, invade, secrete ECM-degrading proteinases, and deposit injury-induced matrices (predisposing to vessel wall fibrosis). In contrast, in response to similar stimuli, capillaries can undergo regression responses (rarefaction). In summary, we have described the molecular events controlling ECM remodeling in major vascular diseases as well as the differential responses of arteries versus capillaries to key mediators inducing vascular injury.
Collapse
Affiliation(s)
- Prisca K. Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL 33612
| | - George E. Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL 33612
| |
Collapse
|
31
|
Kocatürk B, Lee Y, Nosaka N, Abe M, Martinon D, Lane ME, Moreira D, Chen S, Fishbein MC, Porritt RA, Franklin BS, Noval Rivas M, Arditi M. Platelets exacerbate cardiovascular inflammation in a murine model of Kawasaki disease vasculitis. JCI Insight 2023; 8:e169855. [PMID: 37279077 PMCID: PMC10443810 DOI: 10.1172/jci.insight.169855] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/31/2023] [Indexed: 06/07/2023] Open
Abstract
Kawasaki disease (KD) is the leading cause of acquired heart disease among children. Increased platelet counts and activation are observed during the course of KD, and elevated platelet counts are associated with higher risks of developing intravenous immunoglobulin resistance and coronary artery aneurysms. However, the role of platelets in KD pathogenesis remains unclear. Here, we analyzed transcriptomics data generated from the whole blood of patients with KD and discovered changes in the expression of platelet-related genes during acute KD. In the Lactobacillus casei cell wall extract (LCWE) murine model of KD vasculitis, LCWE injection increased platelet counts and the formation of monocyte-platelet aggregates (MPAs), upregulated the concentration of soluble P-selectin, and increased circulating thrombopoietin and interleukin 6 (IL-6). Furthermore, platelet counts correlated with the severity of cardiovascular inflammation. Genetic depletion of platelets (Mpl-/- mice) or treatment with an anti-CD42b antibody significantly reduced LCWE-induced cardiovascular lesions. Furthermore, in the mouse model, platelets promoted vascular inflammation via the formation of MPAs, which likely amplified IL-1B production. Altogether, our results indicate that platelet activation exacerbates the development of cardiovascular lesions in a murine model of KD vasculitis. These findings enhance our understanding of KD vasculitis pathogenesis and highlight MPAs, which are known to enhance IL-1B production, as a potential therapeutic target for this disorder.
Collapse
Affiliation(s)
- Begüm Kocatürk
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, California, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Youngho Lee
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, California, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Nobuyuki Nosaka
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, California, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Masanori Abe
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, California, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Daisy Martinon
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, California, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Malcolm E. Lane
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, California, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Debbie Moreira
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, California, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Shuang Chen
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, California, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Michael C. Fishbein
- Department of Pathology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Rebecca A. Porritt
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, California, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Bernardo S. Franklin
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, California, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Moshe Arditi
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, California, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
32
|
Yuan W, Zhang X, Fan X. The Role of the Piezo1 Mechanosensitive Channel in Heart Failure. Curr Issues Mol Biol 2023; 45:5830-5848. [PMID: 37504285 PMCID: PMC10378680 DOI: 10.3390/cimb45070369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/02/2023] [Accepted: 07/08/2023] [Indexed: 07/29/2023] Open
Abstract
Mechanotransduction (MT) is inseparable from the pathobiology of heart failure (HF). However, the effects of mechanical forces on HF remain unclear. This review briefly describes how Piezo1 functions in HF-affected cells, including endothelial cells (ECs), cardiac fibroblasts (CFs), cardiomyocytes (CMs), and immune cells. Piezo1 is a mechanosensitive ion channel that has been extensively studied in recent years. Piezo1 responds to different mechanical forces and converts them into intracellular signals. The pathways that modulate the Piezo1 switch have also been briefly described. Experimental drugs that specifically activate Piezo1-like proteins, such as Yoda1, Jedi1, and Jedi2, are available for clinical studies to treat Piezo1-related diseases. The only mechanosensitive ion-channel-specific inhibitor available is GsMTx4, which can turn off Piezo1 by modulating the local membrane tension. Ultrasound waves can modulate Piezo1 switching in vitro with the assistance of microbubbles. This review provides new possible targets for heart failure therapy by exploring the cellular functions of Piezo1 that are involved in the progression of the disease. Modulation of Piezo1 activity may, therefore, effectively delay the progression of heart failure.
Collapse
Affiliation(s)
- Weihua Yuan
- National Clinical Research Center for Child Health, Children's Hospital, Zhejiang University School of Medicine, 3333 Binsheng Rd, Hangzhou 310052, China
| | - Xicheng Zhang
- National Clinical Research Center for Child Health, Department of Cardiac Surgery, Children's Hospital, Zhejiang University School of Medicine, 3333 Binsheng Rd, Hangzhou 310052, China
| | - Xiangming Fan
- National Clinical Research Center for Child Health, Department of Cardiac Surgery, Children's Hospital, Zhejiang University School of Medicine, 3333 Binsheng Rd, Hangzhou 310052, China
| |
Collapse
|
33
|
Shi J, Pan Y, Liu X, Cao W, Mu Y, Zhu Q. Spatial Omics Sequencing Based on Microfluidic Array Chips. BIOSENSORS 2023; 13:712. [PMID: 37504111 PMCID: PMC10377411 DOI: 10.3390/bios13070712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/29/2023]
Abstract
Spatial profiling technologies fill the gap left by the loss of spatial information in traditional single-cell sequencing, showing great application prospects. After just a few years of quick development, spatial profiling technologies have made great progress in resolution and simplicity. This review introduces the development of spatial omics sequencing based on microfluidic array chips and describes barcoding strategies using various microfluidic designs with simplicity and efficiency. At the same time, the pros and cons of each strategy are compared. Moreover, commercialized solutions for spatial profiling are also introduced. In the end, the future perspective of spatial omics sequencing and research directions are discussed.
Collapse
Affiliation(s)
- Jianyu Shi
- State Key Laboratory of Industrial Control Technology, Research Center for Analytical Instrumentation, Institute of Cyber-Systems and Control, College of Control Science and Engineering, Zhejiang University, Hangzhou 310000, China
| | - Yating Pan
- State Key Laboratory of Industrial Control Technology, Research Center for Analytical Instrumentation, Institute of Cyber-Systems and Control, College of Control Science and Engineering, Zhejiang University, Hangzhou 310000, China
| | - Xudong Liu
- State Key Laboratory of Industrial Control Technology, Research Center for Analytical Instrumentation, Institute of Cyber-Systems and Control, College of Control Science and Engineering, Zhejiang University, Hangzhou 310000, China
| | - Wenjian Cao
- State Key Laboratory of Industrial Control Technology, Research Center for Analytical Instrumentation, Institute of Cyber-Systems and Control, College of Control Science and Engineering, Zhejiang University, Hangzhou 310000, China
| | - Ying Mu
- State Key Laboratory of Industrial Control Technology, Research Center for Analytical Instrumentation, Institute of Cyber-Systems and Control, College of Control Science and Engineering, Zhejiang University, Hangzhou 310000, China
| | - Qiangyuan Zhu
- State Key Laboratory of Industrial Control Technology, Research Center for Analytical Instrumentation, Institute of Cyber-Systems and Control, College of Control Science and Engineering, Zhejiang University, Hangzhou 310000, China
| |
Collapse
|
34
|
Li JSY, Raghubar AM, Matigian NA, Ng MSY, Rogers NM, Mallett AJ. The Utility of Spatial Transcriptomics for Solid Organ Transplantation. Transplantation 2023; 107:1463-1471. [PMID: 36584371 DOI: 10.1097/tp.0000000000004466] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Spatial transcriptomics (ST) measures and maps transcripts within intact tissue sections, allowing the visualization of gene activity within the spatial organization of complex biological systems. This review outlines advances in genomic sequencing technologies focusing on in situ sequencing-based ST, including applications in transplant and relevant nontransplant settings. We describe the experimental and analytical pipelines that underpin the current generation of spatial technologies. This context is important for understanding the potential role ST may play in expanding our knowledge, including in organ transplantation, and the important caveats/limitations when interpreting the vast data output generated by such methodological platforms.
Collapse
Affiliation(s)
- Jennifer S Y Li
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Arti M Raghubar
- Kidney Health Service, Royal Brisbane and Women's Hospital, QLD, Australia
- Conjoint Internal Medicine Laboratory, Pathology Queensland, Health Support Queensland, QLD, Australia
- Department of Anatomical Pathology, Pathology Queensland, Health Support Queensland, QLD, Australia
- Faculty of Medicine, University of Queensland, QLD, Australia
- Institute for Molecular Bioscience, University of Queensland, QLD, Australia
| | - Nicholas A Matigian
- QCIF Facility for Advanced Bioinformatics, The University of Queensland, QLD, Australia
| | - Monica S Y Ng
- Kidney Health Service, Royal Brisbane and Women's Hospital, QLD, Australia
- Conjoint Internal Medicine Laboratory, Pathology Queensland, Health Support Queensland, QLD, Australia
- Faculty of Medicine, University of Queensland, QLD, Australia
- Institute for Molecular Bioscience, University of Queensland, QLD, Australia
- Nephrology Department, Princess Alexandra Hospital, QLD, Australia
| | - Natasha M Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Department of Renal Medicine, Westmead Hospital, Westmead, NSW, Australia
| | - Andrew J Mallett
- Faculty of Medicine, University of Queensland, QLD, Australia
- Institute for Molecular Bioscience, University of Queensland, QLD, Australia
- College of Medicine and Dentistry, James Cook University, QLD, Australia
- Department of Renal Medicine, Townsville University Hospital, QLD, Australia
| |
Collapse
|
35
|
Ahlmann-Eltze C, Huber W. Comparison of transformations for single-cell RNA-seq data. Nat Methods 2023; 20:665-672. [PMID: 37037999 PMCID: PMC10172138 DOI: 10.1038/s41592-023-01814-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 02/11/2023] [Indexed: 04/12/2023]
Abstract
The count table, a numeric matrix of genes × cells, is the basic input data structure in the analysis of single-cell RNA-sequencing data. A common preprocessing step is to adjust the counts for variable sampling efficiency and to transform them so that the variance is similar across the dynamic range. These steps are intended to make subsequent application of generic statistical methods more palatable. Here, we describe four transformation approaches based on the delta method, model residuals, inferred latent expression state and factor analysis. We compare their strengths and weaknesses and find that the latter three have appealing theoretical properties; however, in benchmarks using simulated and real-world data, it turns out that a rather simple approach, namely, the logarithm with a pseudo-count followed by principal-component analysis, performs as well or better than the more sophisticated alternatives. This result highlights limitations of current theoretical analysis as assessed by bottom-line performance benchmarks.
Collapse
Affiliation(s)
- Constantin Ahlmann-Eltze
- Genome Biology Unit, EMBL, Heidelberg, Germany.
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany.
| | | |
Collapse
|
36
|
Long X, Yuan X, Du J. Single-cell and spatial transcriptomics: Advances in heart development and disease applications. Comput Struct Biotechnol J 2023; 21:2717-2731. [PMID: 37181659 PMCID: PMC10173363 DOI: 10.1016/j.csbj.2023.04.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/11/2023] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
Current transcriptomics technologies, including bulk RNA-seq, single-cell RNA sequencing (scRNA-seq), single-nucleus RNA-sequencing (snRNA-seq), and spatial transcriptomics (ST), provide novel insights into the spatial and temporal dynamics of gene expression during cardiac development and disease processes. Cardiac development is a highly sophisticated process involving the regulation of numerous key genes and signaling pathways at specific anatomical sites and developmental stages. Exploring the cell biological mechanisms involved in cardiogenesis also contributes to congenital heart disease research. Meanwhile, the severity of distinct heart diseases, such as coronary heart disease, valvular disease, cardiomyopathy, and heart failure, is associated with cellular transcriptional heterogeneity and phenotypic alteration. Integrating transcriptomic technologies in the clinical diagnosis and treatment of heart diseases will aid in advancing precision medicine. In this review, we summarize applications of scRNA-seq and ST in the cardiac field, including organogenesis and clinical diseases, and provide insights into the promise of single-cell and spatial transcriptomics in translational research and precision medicine.
Collapse
Affiliation(s)
- Xianglin Long
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xin Yuan
- Department of Nephrology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jianlin Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
37
|
Banos A, Thomas K, Garantziotis P, Filia A, Malissovas N, Pieta A, Nikolakis D, Panagiotopoulos AG, Chalkia A, Petras D, Bertsias G, Boumpas DT, Vassilopoulos D. The genomic landscape of ANCA-associated vasculitis: Distinct transcriptional signatures, molecular endotypes and comparison with systemic lupus erythematosus. Front Immunol 2023; 14:1072598. [PMID: 37051253 PMCID: PMC10083368 DOI: 10.3389/fimmu.2023.1072598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 03/13/2023] [Indexed: 03/29/2023] Open
Abstract
IntroductionAnti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAVs) present with a complex phenotype and are associated with high mortality and multi-organ involvement. We sought to define the transcriptional landscape and molecular endotypes of AAVs and compare it to systemic lupus erythematosus (SLE).MethodsWe performed whole blood mRNA sequencing from 30 patients with AAV (granulomatosis with polyangiitis/GPA and microscopic polyangiitis/MPA) combined with functional enrichment and network analysis for aberrant pathways. Key genes and pathways were validated in an independent cohort of 18 AAV patients. Co-expression network and hierarchical clustering analysis, identified molecular endotypes. Multi-level transcriptional overlap analysis to SLE was based on our published data from 142 patients.ResultsWe report here that “Pan-vasculitis” signature contained 1,982 differentially expressed genes, enriched in leukocyte differentiation, cytokine signaling, type I and type II IFN signaling and aberrant B-T cell immunity. Active disease was characterized by signatures linked to cell cycle checkpoints and metabolism pathways, whereas ANCA-positive patients exhibited a humoral immunity transcriptional fingerprint. Differential expression analysis of GPA and MPA yielded an IFN-g pathway (in addition to a type I IFN) in the former and aberrant expression of genes related to autophagy and mRNA splicing in the latter. Unsupervised molecular taxonomy analysis revealed four endotypes with neutrophil degranulation, aberrant metabolism and B-cell responses as potential mechanistic drivers. Transcriptional perturbations and molecular heterogeneity were more pronounced in SLE. Molecular analysis and data-driven clustering of AAV uncovered distinct transcriptional pathways that could be exploited for targeted therapy.DiscussionWe conclude that transcriptomic analysis of AAV reveals distinct endotypes and molecular pathways that could be targeted for therapy. The AAV transcriptome is more homogenous and less fragmented compared to the SLE which may account for its superior rates of response to therapy.
Collapse
Affiliation(s)
- Aggelos Banos
- Laboratory of Autoimmunity and Inflammation, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Konstantinos Thomas
- Clinical Immunology- Rheumatology Unit, 2nd Department of Medicine and Laboratory, General Hospital of Athens Ippokrateio, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Garantziotis
- Laboratory of Autoimmunity and Inflammation, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Department Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Anastasia Filia
- Laboratory of Autoimmunity and Inflammation, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Nikolaos Malissovas
- Laboratory of Autoimmunity and Inflammation, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Antigone Pieta
- Laboratory of Autoimmunity and Inflammation, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Rheumatology and Clinical Immunology Unit, 4th Department of Internal Medicine, Attikon University Hospital, Athens, Greece
| | - Dimitrios Nikolakis
- Laboratory of Autoimmunity and Inflammation, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Amsterdam Institute for Gastroenterology Endocrinology and Metabolism, Department of Gastroenterology, Academic Medical Center, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam, Netherlands
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection & Immunity, Department of Experimental Immunology, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Alexandros G. Panagiotopoulos
- Clinical Immunology- Rheumatology Unit, 2nd Department of Medicine and Laboratory, General Hospital of Athens Ippokrateio, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Aglaia Chalkia
- Nephrology Department, General Hospital of Athens Ippokrateio, Athens, Greece
| | - Dimitrios Petras
- Nephrology Department, General Hospital of Athens Ippokrateio, Athens, Greece
| | - George Bertsias
- Department of Rheumatology and Clinical Immunology, University Hospital of Heraklion, Medical School, University of Crete, Heraklion, Greece
- Department of Immunity, Institute of Molecular Biology and Biotechnology-Foundation of Research and Technology-Hellas (FORTH), Heraklion, Greece
| | - Dimitrios T. Boumpas
- Laboratory of Autoimmunity and Inflammation, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Rheumatology and Clinical Immunology Unit, 4th Department of Internal Medicine, Attikon University Hospital, Athens, Greece
- Joint Academic Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Vassilopoulos
- Clinical Immunology- Rheumatology Unit, 2nd Department of Medicine and Laboratory, General Hospital of Athens Ippokrateio, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Joint Academic Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- *Correspondence: Dimitrios Vassilopoulos,
| |
Collapse
|
38
|
Shahi A, Afzali S, Firoozi Z, Mohaghegh P, Moravej A, Hosseinipour A, Bahmanyar M, Mansoori Y. Potential roles of NLRP3 inflammasome in the pathogenesis of Kawasaki disease. J Cell Physiol 2023; 238:513-532. [PMID: 36649375 DOI: 10.1002/jcp.30948] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/12/2022] [Accepted: 01/05/2023] [Indexed: 01/19/2023]
Abstract
There is a heterogeneous group of rare illnesses that fall into the vasculitis category and are characterized mostly by blood vessel inflammation. Ischemia and disrupted blood flow will cause harm to the organs whose blood arteries become inflamed. Kawasaki disease (KD) is the most prevalent kind of vasculitis in children aged 5 years or younger. Because KD's cardiovascular problems might persist into adulthood, it is no longer thought of as a self-limiting disease. KD is a systemic vasculitis with unknown initiating factors. Numerous factors, such as genetic predisposition and infectious pathogens, are implicated in the etiology of KD. As endothelial cell damage and inflammation can lead to coronary endothelial dysfunction in KD, some studies hypothesized the crucial role of pyroptosis in the pathogenesis of KD. Additionally, pyroptosis-related proteins like caspase-1, apoptosis-associated speck-like protein containing a CARD (ASC), proinflammatory cytokines like IL-1 and IL-18, lactic dehydrogenase, and Gasdermin D (GSDMD) have been found to be overexpressed in KD patients when compared to healthy controls. These occurrences may point to an involvement of inflammasomes and pyroptotic cell death in the etiology of KD and suggest potential treatment targets. Based on these shreds of evidence, in this review, we aim to focus on one of the well-defined inflammasomes, NLRP3, and its role in the pathophysiology of KD.
Collapse
Affiliation(s)
- Abbas Shahi
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shima Afzali
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Firoozi
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Medical Genetics, Fasa University of Medical Sciences, Fasa, Iran
| | - Poopak Mohaghegh
- Pediatrics Department, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Ali Moravej
- Department of Immunology, Fasa University of Medical Sciences, Fasa, Iran
| | - Ali Hosseinipour
- Department of Internal Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Maryam Bahmanyar
- Pediatrics Department, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Yaser Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Medical Genetics, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
39
|
Huang H, Dong J, Jiang J, Yang F, Zheng Y, Wang S, Wang N, Ma J, Hou M, Ding Y, Meng L, Zhuo W, Yang D, Qian W, Chen Q, You G, Qian G, Gu L, Lv H. The role of FOXO4/NFAT2 signaling pathway in dysfunction of human coronary endothelial cells and inflammatory infiltration of vasculitis in Kawasaki disease. Front Immunol 2023; 13:1090056. [PMID: 36700213 PMCID: PMC9869249 DOI: 10.3389/fimmu.2022.1090056] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Aims The Ca+/NFAT (Nuclear factor of activated T cells) signaling pathway activation is implicated in the pathogenesis of Kawasaki disease (KD); however, we lack detailed information regarding the regulatory network involved in the human coronary endothelial cell dysfunction and cardiovascular lesion development. Herein, we aimed to use mouse and endothelial cell models of KD vasculitis in vivo and in vitro to characterize the regulatory network of NFAT pathway in KD. Methods and Results Among the NFAT gene family, NFAT2 showed the strongest transcriptional activity in peripheral blood mononuclear cells (PBMCs) from patients with KD. Then, NFAT2 overexpression and knockdown experiments in Human coronary artery endothelial cells (HCAECs) indicated that NFAT2 overexpression disrupted endothelial cell homeostasis by regulation of adherens junctions, whereas its knockdown protected HCAECs from such dysfunction. Combined analysis using RNA-sequencing and transcription factor (TF) binding site analysis in the NFAT2 promoter region predicted regulation by Forkhead box O4 (FOXO4). Western blotting, chromatin immunoprecipitation, and luciferase assays validated that FOXO4 binds to the promoter and transcriptionally represses NFAT2. Moreover, Foxo4 knockout increased the extent of inflamed vascular tissues in a mouse model of KD vasculitis. Functional experiments showed that inhibition NFAT2 relieved Foxo4 knockout exaggerated vasculitis in vivo. Conclusions Our findings revealed the FOXO4/NFAT2 axis as a vital pathway in the progression of KD that is associated with endothelial cell homeostasis and cardiovascular inflammation development.
Collapse
Affiliation(s)
- Hongbiao Huang
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China,Department of Pediatrics, Fujian Provincial Hospital, Fujian Provincial Clinical College of Fujian Medical University, Fuzhou, Fujian, China,Epigenetics Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Jinfeng Dong
- Department of Hematology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Jiaqi Jiang
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fang Yang
- Department of Pediatrics, Fujian Provincial Hospital, Fujian Provincial Clinical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Yiming Zheng
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Shuhui Wang
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Nana Wang
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jin Ma
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Miao Hou
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yueyue Ding
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lijun Meng
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Wenyu Zhuo
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Daoping Yang
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Weiguo Qian
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qiaobin Chen
- Department of Pediatrics, Fujian Provincial Hospital, Fujian Provincial Clinical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Guoping You
- Department of Emergency, Fujian Provincial Hospital, Fujian Provincial Clinical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Guanghui Qian
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lei Gu
- Epigenetics Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany,Cardiopulmonary Institute (CPI), Bad Nauheim, Germany,*Correspondence: Haitao Lv, ; Lei Gu,
| | - Haitao Lv
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China,*Correspondence: Haitao Lv, ; Lei Gu,
| |
Collapse
|
40
|
Kobayashi H, Kimura MY, Hasegawa I, Suganuma E, Ikehara Y, Azuma K, Ito T, Ebata R, Kurashima Y, Kawasaki Y, Shiko Y, Saito N, Iwase H, Lee Y, Noval Rivas M, Arditi M, Zuka M, Hamada H, Nakayama T. Increased Myosin light chain 9 expression during Kawasaki disease vasculitis. Front Immunol 2023; 13:1036672. [PMID: 36685558 PMCID: PMC9853906 DOI: 10.3389/fimmu.2022.1036672] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 12/05/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction Kawasaki disease (KD) is an acute systemic vasculitis that predominantly afflicts children. KD development is known to be associated with an aberrant immune response and abnormal platelet activation, however its etiology is still largely unknown. Myosin light chain 9 (Myl9) is known to regulate cellular contractility of both non-muscle and smooth muscle cells, and can be released from platelets, whereas any relations of Myl9 expression to KD vasculitis have not been examined. Methods Plasma Myl9 concentrations in KD patients and children with febrile illness were measured and associated with KD clinical course and prognosis. Myl9 release from platelets in KD patients was also evaluated in vitro. Myl9 expression was determined in coronary arteries from Lactobacillus casei cell wall extract (LCWE)-injected mice that develop experimental KD vasculitis, as well as in cardiac tissues obtained at autopsy from KD patients. Results and discussion Plasma Myl9 levels were significantly higher in KD patients during the acute phase compared with healthy controls or patients with other febrile illnesses, declined following IVIG therapy in IVIG-responders but not in non-responders. In vitro, platelets from KD patients released Myl9 independently of thrombin stimulation. In the LCWE-injected mice, Myl9 was detected in cardiac tissue at an early stage before inflammatory cell infiltration was observed. In tissues obtained at autopsy from KD patients, the highest Myl9 expression was observed in thrombi during the acute phase and in the intima and adventitia of coronary arteries during the chronic phase. Thus, our studies show that Myl9 expression is significantly increased during KD vasculitis and that Myl9 levels may be a useful biomarker to estimate inflammation and IVIG responsiveness to KD.
Collapse
Affiliation(s)
- Hironobu Kobayashi
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Experimental Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Motoko Y. Kimura
- Department of Experimental Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Chiba University “Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Japan Initiative for World-leading Vaccine Research and Development Centers, Japan Agency for Medical Research and Development (AMED), Chiba, Japan, Chiba, Japan
| | - Ichita Hasegawa
- Department of Experimental Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Eisuke Suganuma
- Division of Infectious Diseases and Immunology, Allergy, Saitama Children’s Medical Center, Saitama, Japan
| | - Yuzuru Ikehara
- Department of Molecular and Tumor Pathology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kazuhiko Azuma
- Department of Molecular and Tumor Pathology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshihiro Ito
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ryota Ebata
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yosuke Kurashima
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yohei Kawasaki
- Clinical Research Center, Chiba University Hospital, Chiba, Japan
| | - Yuki Shiko
- Clinical Research Center, Chiba University Hospital, Chiba, Japan
| | - Naoki Saito
- Department of Legal Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hirotaro Iwase
- Department of Legal Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Youngho Lee
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Moshe Arditi
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Masahiko Zuka
- Department of Forensic Medicine and Pathology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Hiromichi Hamada
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Japan Agency for Medical Research and Development (AMED)-Core Research for Evolutional Science and Technology (CREST), AMED, Chiba, Japan
| |
Collapse
|
41
|
Hu S, Wang L, Xu Y, Li F, Wang T. Disulfiram attenuates hypoxia-induced pulmonary hypertension by inhibiting GSDMD cleavage and pyroptosis in HPASMCs. Respir Res 2022; 23:353. [PMID: 36527086 PMCID: PMC9756478 DOI: 10.1186/s12931-022-02279-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Pulmonary hypertension (PH) is characterized by progressive pulmonary arterial remodelling, associated with different severities of inflammation and altered immune processes. Disulfiram eliminates the formation of N-gasdermin D (GSDMD) plasma membrane pores to prevent pyroptosis. Pyroptosis is a form of lytic cell death characterized by inflammasome activation and proinflammatory cytokine release that acts in the development of PH. We sought to investigate whether disulfiram could alleviate hypoxia-induced PH by inhibiting pyroptosis. METHODS To investigate whether disulfiram alleviates the progression of pulmonary hypertension, rodents were exposed to chronic hypoxia (10% oxygen, 4 weeks) to induce PH. The severity of PH was assessed by measuring right ventricular systolic pressure, mean pulmonary artery pressure, and the degree of right ventricular hypertrophy. Western blotting was used to measure proteins associated with the pyroptosis pathway, and ELISA was performed to measure the secretion of IL-18 and IL-1β, both of which are the primary methods for assessing pyroptosis. RESULTS IL-18 and IL-1β concentrations were higher in patients with PH than in normal controls. Disulfiram suppressed the progression of PH in mice and rats through the alleviation of pulmonary arterial remodelling. Pyroptosis-related proteins and the inflammasome were activated in rodent models of PH. Disulfiram inhibited the processing of GSDMD into N-GSDMD and attenuated the secretion of IL-1β and IL18. In vivo experiments showed that disulfiram also inhibited lytic death in HPASMCs. CONCLUSIONS Disulfiram treatment reduces PH progression through suppressing vascular remodelling by inhibiting GSDMD cleavage and pyroptosis. It might become a novel therapeutic option for the treatment of PH.
Collapse
Affiliation(s)
- Shunlian Hu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- The Center for Biomedical Research, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Lu Wang
- Department of Respiratory and Critical Care Medicine, Miyun Teaching Hospital of Capital Medical University, Beijing, People's Republic of China
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Miyun District, Beijing, People's Republic of China
- Department of Respiratory and Critical Care Medicine, Beijing Miyun Hospital, Beijing, People's Republic of China
| | - Yahan Xu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- The Center for Biomedical Research, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Fajiu Li
- The Sixth Hospital of Wuhan City, Affiliated Hospital of Jianghan University, Beijing, People's Republic of China
| | - Tao Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
- The Center for Biomedical Research, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
42
|
Ruoss S, Esparza MC, Vasquez-Bolanos LS, Nasamran CA, Fisch KM, Engler AJ, Ward SR. Spatial transcriptomics tools allow for regional exploration of heterogeneous muscle pathology in the pre-clinical rabbit model of rotator cuff tear. J Orthop Surg Res 2022; 17:440. [PMID: 36195913 PMCID: PMC9531386 DOI: 10.1186/s13018-022-03326-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/18/2022] [Indexed: 11/21/2022] Open
Abstract
Background Conditions affecting skeletal muscle, such as chronic rotator cuff tears, low back pain, dystrophies, and many others, often share changes in muscle phenotype: intramuscular adipose and fibrotic tissue increase while contractile tissue is lost. The underlying changes in cell populations and cell ratios observed with these phenotypic changes complicate the interpretation of tissue-level transcriptional data. Novel single-cell transcriptomics has limited capacity to address this problem because muscle fibers are too long to be engulfed in single-cell droplets and single nuclei transcriptomics are complicated by muscle fibers’ multinucleation. Therefore, the goal of this project was to evaluate the potential and challenges of a spatial transcriptomics technology to add dimensionality to transcriptional data in an attempt to better understand regional cellular activity in heterogeneous skeletal muscle tissue. Methods The 3′ Visium spatial transcriptomics technology was applied to muscle tissue of a rabbit model of rotator cuff tear. Healthy control and tissue collected at 2 and 16 weeks after tenotomy was utilized and freshly snap frozen tissue was compared with tissue stored for over 6 years to evaluate whether this technology is retrospectively useful in previously acquired tissues. Transcriptional information was overlayed with standard hematoxylin and eosin (H&E) stains of the exact same histological sections. Results Sequencing saturation and number of genes detected was not affected by sample storage duration. Unbiased clustering matched the underlying tissue type-based on H&E assessment. Connective-tissue-rich areas presented with lower unique molecular identifier counts are compared with muscle fibers even though tissue permeabilization was standardized across the section. A qualitative analysis of resulting datasets revealed heterogeneous fiber degeneration–regeneration after tenotomy based on (neonatal) myosin heavy chain 8 detection and associated differentially expressed gene analysis. Conclusions This protocol can be used in skeletal muscle to explore spatial transcriptional patterns and confidently relate them to the underlying histology, even for tissues that have been stored for up to 6 years. Using this protocol, there is potential for novel transcriptional pathway discovery in longitudinal studies since the transcriptional information is unbiased by muscle composition and cell type changes.
Collapse
Affiliation(s)
- Severin Ruoss
- Department of Orthopaedic Surgery, UC San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0863, USA
| | - Mary C Esparza
- Department of Orthopaedic Surgery, UC San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0863, USA
| | - Laura S Vasquez-Bolanos
- Department of Orthopaedic Surgery, UC San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0863, USA.,Department of Bioengineering, UC San Diego, La Jolla, CA, USA
| | - Chanond A Nasamran
- Center for Computational Biology and Bioinformatics, UC San Diego, La Jolla, CA, USA
| | - Kathleen M Fisch
- Center for Computational Biology and Bioinformatics, UC San Diego, La Jolla, CA, USA.,Department of Obstetrics, Gynecology and Reproductive Sciences, UC San Diego, La Jolla, CA, USA
| | - Adam J Engler
- Department of Bioengineering, UC San Diego, La Jolla, CA, USA
| | - Samuel R Ward
- Department of Orthopaedic Surgery, UC San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0863, USA. .,Department of Bioengineering, UC San Diego, La Jolla, CA, USA. .,Department of Radiology, UC San Diego, La Jolla, CA, USA.
| |
Collapse
|
43
|
Koca-Ünsal RB, Şehirli AÖ, Sayıner S, Aksoy U. Relationship of NLRP3 inflammasome with periodontal, endodontic and related systemic diseases. Mol Biol Rep 2022; 49:11123-11132. [DOI: 10.1007/s11033-022-07894-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 11/28/2022]
|
44
|
Stock AT, Parsons S, Sharma VJ, James F, Starkey G, D'Costa R, Gordon CL, Wicks IP. Intimal macrophages develop from circulating monocytes during vasculitis. Clin Transl Immunology 2022; 11:e1412. [PMID: 35991774 PMCID: PMC9375838 DOI: 10.1002/cti2.1412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/12/2022] [Accepted: 07/29/2022] [Indexed: 11/26/2022] Open
Abstract
Objective Vasculitis is characterised by inflammation of the blood vessels. While all layers of the vessel can be affected, inflammation within the intimal layer can trigger thrombosis and arterial occlusion and is therefore of particular clinical concern. Given this pathological role, we have examined how intimal inflammation develops by exploring which (and how) macrophages come to populate this normally immune‐privileged site during vasculitis. Methods We have addressed this question for Kawasaki disease (KD), which is a type of vasculitis in children that typically involves the coronary arteries. We used confocal microscopy and flow cytometry to characterise the macrophages that populate the coronary artery intima in KD patient samples and in a mouse model of KD, and furthermore, have applied an adoptive transfer system to trace how these intimal macrophages develop. Results In KD patients, intimal hyperplasia coincided with marked macrophage infiltration of the coronary artery intima. Phenotypic analysis revealed that these ‘intimal macrophages’ did not express markers of resident cardiac macrophages, such as Lyve‐1, and instead, were uniformly positive for the chemokine receptor Ccr2, suggesting a monocytic lineage. In support of this origin, we show that circulating monocytes directly invade the intima via transluminal migration during established disease, coinciding with the activation of endothelial cells lining the coronary arteries. Conclusions During KD, intimal macrophages develop from circulating monocytes that infiltrate the inflamed coronary artery intima by transluminal migration.
Collapse
Affiliation(s)
- Angus T Stock
- Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
| | - Sarah Parsons
- Department of Forensic Medicine Monash University Melbourne VIC Australia.,Victorian Institute of Forensic Medicine Melbourne VIC Australia
| | - Varun J Sharma
- Liver & Intestinal Transplant Unit Austin Health Melbourne VIC Australia.,Department of Surgery The University of Melbourne, Austin Health Melbourne VIC Australia.,Department of Cardiac Surgery Austin Health Melbourne VIC Australia
| | - Fiona James
- Department of Infectious Diseases Austin Health Melbourne VIC Australia
| | - Graham Starkey
- Liver & Intestinal Transplant Unit Austin Health Melbourne VIC Australia.,Department of Surgery The University of Melbourne, Austin Health Melbourne VIC Australia
| | - Rohit D'Costa
- DonateLife Victoria Carlton VIC Australia.,Department of Intensive Care Medicine Melbourne Health Melbourne VIC Australia
| | - Claire L Gordon
- Department of Infectious Diseases Austin Health Melbourne VIC Australia.,Department of Microbiology and Immunology The Peter Doherty Institute for Infection and Immunity, The University of Melbourne Melbourne VIC Australia.,North Eastern Public Health Unit Austin Health Melbourne VIC Australia
| | - Ian P Wicks
- Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia.,Rheumatology Unit The Royal Melbourne Hospital Melbourne VIC Australia.,Department of Medical Biology University of Melbourne Melbourne VIC Australia
| |
Collapse
|
45
|
Elishaev M, Hodonsky CJ, Ghosh SKB, Finn AV, von Scheidt M, Wang Y. Opportunities and Challenges in Understanding Atherosclerosis by Human Biospecimen Studies. Front Cardiovasc Med 2022; 9:948492. [PMID: 35872917 PMCID: PMC9300954 DOI: 10.3389/fcvm.2022.948492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Over the last few years, new high-throughput biotechnologies and bioinformatic methods are revolutionizing our way of deep profiling tissue specimens at the molecular levels. These recent innovations provide opportunities to advance our understanding of atherosclerosis using human lesions aborted during autopsies and cardiac surgeries. Studies on human lesions have been focusing on understanding the relationship between molecules in the lesions with tissue morphology, genetic risk of atherosclerosis, and future adverse cardiovascular events. This review will highlight ways to utilize human atherosclerotic lesions in translational research by work from large cardiovascular biobanks to tissue registries. We will also discuss the opportunities and challenges of working with human atherosclerotic lesions in the era of next-generation sequencing.
Collapse
Affiliation(s)
- Maria Elishaev
- Department of Pathology and Laboratory Medicine, Center for Heart Lung Innovation, University of British Columbia, Vancouver, BC, Canada
| | - Chani J. Hodonsky
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA, United States
| | | | - Aloke V. Finn
- Cardiovascular Pathology Institute, Gaithersburg, MD, United States
| | - Moritz von Scheidt
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung, Partner Site Munich Heart Alliance, Munich, Germany
| | - Ying Wang
- Department of Pathology and Laboratory Medicine, Center for Heart Lung Innovation, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
46
|
Marek-Iannucci S, Yildirim AD, Hamid SM, Ozdemir AB, Gomez AC, Kocatürk B, Porritt RA, Fishbein MC, Iwawaki T, Noval Rivas M, Erbay E, Arditi M. Targeting IRE1 endoribonuclease activity alleviates cardiovascular lesions in a murine model of Kawasaki disease vasculitis. JCI Insight 2022; 7:157203. [PMID: 35167493 PMCID: PMC8986066 DOI: 10.1172/jci.insight.157203] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/09/2022] [Indexed: 11/17/2022] Open
Abstract
Kawasaki disease (KD) is the leading cause of non-congenital heart disease in children. Studies in mice and humans propound the NLRP3-IL-1β pathway as the principal driver of KD pathophysiology. Endoplasmic reticulum (ER) stress can activate the NLRP3 inflammasome, but the potential implication of ER stress in KD pathophysiology has not been investigated. We used human patient data and the Lactobacillus casei cell wall extract (LCWE) murine model of KD vasculitis to characterize the impact of ER stress on the development of cardiovascular lesions. KD patient transcriptomics and single-cell RNA sequencing of the abdominal aorta from LCWE-injected mice revealed changes in the expression of ER stress genes. Alleviating ER stress genetically, by conditional deletion of Inositol Requiring Enzyme-1 (IRE1) in myeloid cells, or pharmacologically, by inhibition of IRE1 endoribonuclease (RNase) activity, led to significant reduction of LCWE-induced cardiovascular lesion formation as well as reduced caspase-1 activity and IL-1β secretion. These results demonstrate the causal relationship of ER stress to KD pathogenesis, and highlight IRE1 RNase activity as a potential new therapeutic target.
Collapse
Affiliation(s)
- Stefanie Marek-Iannucci
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Asli D Yildirim
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Syed M Hamid
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Asli B Ozdemir
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Angela C Gomez
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Begüm Kocatürk
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Rebecca A Porritt
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | | | - Takao Iwawaki
- Department of Life Science, Medical Research Institute, Kanazawa Medical University, Kahoku, Japan
| | - Magali Noval Rivas
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Ebru Erbay
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Moshe Arditi
- Cedars-Sinai Medical Center, Los Angeles, United States of America
| |
Collapse
|
47
|
Liu H, Hu J, Zheng Q, Feng X, Zhan F, Wang X, Xu G, Hua F. Piezo1 Channels as Force Sensors in Mechanical Force-Related Chronic Inflammation. Front Immunol 2022; 13:816149. [PMID: 35154133 PMCID: PMC8826255 DOI: 10.3389/fimmu.2022.816149] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/03/2022] [Indexed: 12/14/2022] Open
Abstract
Mechanical damage is one of the predisposing factors of inflammation, and it runs through the entire inflammatory pathological process. Repeated or persistent damaging mechanical irritation leads to chronic inflammatory diseases. The mechanism of how mechanical forces induce inflammation is not fully understood. Piezo1 is a newly discovered mechanically sensitive ion channel. The Piezo1 channel opens in response to mechanical stimuli, transducing mechanical signals into an inflammatory cascade in the cell leading to tissue inflammation. A large amount of evidence shows that Piezo1 plays a vital role in the occurrence and progression of chronic inflammatory diseases. This mini-review briefly presents new evidence that Piezo1 responds to different mechanical stresses to trigger inflammation in various tissues. The discovery of Piezo1 provides new insights for the treatment of chronic inflammatory diseases related to mechanical stress. Inhibiting the transduction of damaging mechanical signals into inflammatory signals can inhibit inflammation and improve the outcome of inflammation at an early stage. The pharmacology of Piezo1 has shown bright prospects. The development of tissue-specific Piezo1 drugs for clinical use may be a new target for treating chronic inflammation.
Collapse
Affiliation(s)
- Hailin Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jialing Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qingcui Zheng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaojin Feng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fenfang Zhan
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xifeng Wang
- Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Guohai Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
48
|
Marek-Iannucci S, Ozdemir AB, Moreira D, Gomez AC, Lane M, Porritt RA, Lee Y, Shimada K, Abe M, Stotland A, Zemmour D, Parker S, Sanchez-Lopez E, Van Eyk J, Gottlieb RA, Fishbein M, Karin M, Crother TR, Noval Rivas M, Arditi M. Autophagy-mitophagy induction attenuates cardiovascular inflammation in a murine model of Kawasaki disease vasculitis. JCI Insight 2021; 6:e151981. [PMID: 34403365 PMCID: PMC8492304 DOI: 10.1172/jci.insight.151981] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/11/2021] [Indexed: 01/18/2023] Open
Abstract
Kawasaki disease (KD) is the leading cause of acquired heart disease among children. Murine and human data suggest that the NLRP3-IL-1β pathway is the main driver of KD pathophysiology. NLRP3 can be activated during defective autophagy/mitophagy. We used the Lactobacillus casei cell wall extract (LCWE) murine model of KD vasculitis, to examine the role of autophagy/mitophagy on cardiovascular lesion development. LCWE-injected mice had impaired autophagy/mitophagy and increased levels of ROS in cardiovascular lesions, together with increased systemic 8-OHdG release. Enhanced autophagic flux significantly reduced cardiovascular lesions in LCWE-injected mice, whereas autophagy blockade increased inflammation. Vascular smooth muscle cell specific deletion of Atg16l1 and global Parkin-/- significantly increased disease formation, supporting the importance of autophagy/mitophagy in this model. Ogg1-/- mice had significantly increased lesions with increased NLRP3 activity, whereas treatment with MitoQ, reduced vascular tissue inflammation, ROS production and systemic 8-OHdG release. Treatment with MN58b or Metformin (increasing AMPK and reducing ROS), resulted in decreased disease formation. Our results demonstrate that impaired autophagy/mitophagy and ROS-dependent damage exacerbate the development of murine KD vasculitis. This pathway can be efficiently targeted to reduce disease severity. These findings enhance our understanding of KD pathogenesis and identify novel therapeutic avenues for KD treatment.
Collapse
Affiliation(s)
- Stefanie Marek-Iannucci
- Graduate School of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - A Beyza Ozdemir
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Debbie Moreira
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Angela C Gomez
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Malcolm Lane
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Rebecca A Porritt
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Youngho Lee
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Kenichi Shimada
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Masanori Abe
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Aleksandr Stotland
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - David Zemmour
- Department of Pathology, Harvard Medical School and Brigham and Women's Hospital, Boston, United States of America
| | - Sarah Parker
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | | | - Jennifer Van Eyk
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Roberta A Gottlieb
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Michael Fishbein
- Department of Pathology, UCLA, Los Angeles, United States of America
| | - Michael Karin
- Department of Pathology, UCSD, San Diego, United States of America
| | - Timothy R Crother
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Magali Noval Rivas
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Moshe Arditi
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| |
Collapse
|