1
|
Jiang Y, Wang Y, Guo J, Wang Z, Wang X, Yao X, Yang H, Zou Y. Exploring potential therapeutic targets for asthma: a proteome-wide Mendelian randomization analysis. J Transl Med 2024; 22:978. [PMID: 39472987 PMCID: PMC11520847 DOI: 10.1186/s12967-024-05782-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/18/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Asthma poses a significant global health challenge, characterized by high rates of morbidity and mortality. Despite available treatments, many severe asthma patients remain poorly managed, highlighting the need for novel therapeutic strategies. This study aims to identify potential drug targets for asthma by examining the influence of circulating plasma proteins on asthma risk. METHODS This study employs summary-data-based Mendelian randomization (MR) and two-sample MR methods to investigate the association between 2940 plasma proteins from the UK Biobank study and asthma. The analysis includes discovery (FinnGen cohort) and replication (GERA cohort) phases, with Bayesian colocalization used to validate the relationships between proteins and asthma. Furthermore, protein-protein interaction and druggability assessments were conducted on high-evidence strength protein biomarkers, and candidate drug prediction and molecular docking were performed for proteins without targeted drugs. Given the complexity of asthma pathogenesis, the study also explores the relationships between plasma proteins and asthma-related endpoints (e.g., obesity-related asthma, infection-related asthma, childhood asthma) to identify potential therapeutic targets for different subtypes. RESULTS In the discovery cohort, 75 plasma proteins were associated with asthma, including IL1RAP, IL1RL1, IL6, CXCL5, and CXCL8. Additionally, 6 proteins (IL4R, LTB, CASP8, MAX, PCDH12, and SCLY) were validated through co-localization analysis and validation cohort. The assessment of drug targetability revealed potential drug targets for IL4R, CASP8, and SCLY, while candidate drugs were predicted for LTB and MAX proteins. MAX exhibited strong binding affinity with multiple small molecules indicating a highly stable interaction and significant druggability potential. Analysis of the 75 proteins with 9 asthma-related endpoints highlighted promising targets such as DOK2, ITGAM, CA1, BTN2A1, and GZMB. CONCLUSION These findings elucidate the link between asthma, its related endpoints, and plasma proteins, advancing our understanding of molecular pathogenesis and treatment strategies. The discovery of potential therapeutic targets offers new insights into asthma drug target research.
Collapse
Affiliation(s)
- Yuhan Jiang
- Clinical School of Pediatrics, Tianjin Medical University, Tianjin, China
- Department of Pulmonology, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Machang Compus, 225 Machang Road, Hexi District, Tianjin, 300074, China
| | - Yifan Wang
- Clinical School of Pediatrics, Tianjin Medical University, Tianjin, China
- Department of Pulmonology, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Machang Compus, 225 Machang Road, Hexi District, Tianjin, 300074, China
| | - Ju Guo
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Zixuan Wang
- Clinical School of Pediatrics, Tianjin Medical University, Tianjin, China
| | - Xuelin Wang
- Department of Pulmonology, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Machang Compus, 225 Machang Road, Hexi District, Tianjin, 300074, China
| | - Xueming Yao
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Hongxi Yang
- Department of Bioinformatics, School of Basic Medical Science, Tianjin Medical University, Tianjin, 300070, China.
| | - Yingxue Zou
- Clinical School of Pediatrics, Tianjin Medical University, Tianjin, China.
- Department of Pulmonology, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Machang Compus, 225 Machang Road, Hexi District, Tianjin, 300074, China.
| |
Collapse
|
2
|
Chen S, Gao J, Zhang T. From mesenchymal stem cells to their extracellular vesicles: Progress and prospects for asthma therapy. Asian J Pharm Sci 2024; 19:100942. [PMID: 39253613 PMCID: PMC11382190 DOI: 10.1016/j.ajps.2024.100942] [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: 04/22/2023] [Revised: 12/18/2023] [Accepted: 05/20/2024] [Indexed: 09/11/2024] Open
Abstract
Asthma is a widespread public health concern, with an increasing incidence. Despite the implementation of current treatment strategies, asthma control, particularly for severe cases, remains suboptimal. Recent research has revealed the encouraging prospects of extracellular vesicles (EVs) secreted by mesenchymal stem cells (MSCs) as a viable therapeutic option for alleviating asthma symptoms. Therefore, the present review aims to provide an overview of the current progress and the therapeutic mechanisms of using MSC-derived EVs (MSC-EVs) for asthma treatment. Additionally, different administration approaches for EVs and their impacts on biodistribution and the curative outcomes of EVs are summarized. Notably, the potential benefits of nebulized inhalation of MSC-EVs are addressed. Also, the possibilities and challenges of using MSC-EVs for asthma treatment in clinics are highlighted. Overall, this review is intended to give new insight into the utilization of MSC-EVs as a potential biological drug for asthma treatment.
Collapse
Affiliation(s)
- Shihan Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Tianyuan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
3
|
Liu T, Woodruff PG, Zhou X. Advances in non-type 2 severe asthma: from molecular insights to novel treatment strategies. Eur Respir J 2024; 64:2300826. [PMID: 38697650 PMCID: PMC11325267 DOI: 10.1183/13993003.00826-2023] [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: 05/17/2023] [Accepted: 04/18/2024] [Indexed: 05/05/2024]
Abstract
Asthma is a prevalent pulmonary disease that affects more than 300 million people worldwide and imposes a substantial economic burden. While medication can effectively control symptoms in some patients, severe asthma attacks, driven by airway inflammation induced by environmental and infectious exposures, continue to be a major cause of asthma-related mortality. Heterogeneous phenotypes of asthma include type 2 (T2) and non-T2 asthma. Non-T2 asthma is often observed in patients with severe and/or steroid-resistant asthma. This review covers the molecular mechanisms, clinical phenotypes, causes and promising treatments of non-T2 severe asthma. Specifically, we discuss the signalling pathways for non-T2 asthma including the activation of inflammasomes, interferon responses and interleukin-17 pathways, and their contributions to the subtypes, progression and severity of non-T2 asthma. Understanding the molecular mechanisms and genetic determinants underlying non-T2 asthma could form the basis for precision medicine in severe asthma treatment.
Collapse
Affiliation(s)
- Tao Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Nanjing, China
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Prescott G Woodruff
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine and Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Li Q, Jiang G, Lv Y. Inhibition of phosphoinositide 3-kinase activity attenuates neutrophilic airway inflammation and inhibits pyrin domain-containing 3 inflammasome activation in an ovalbumin-lipopolysaccharide-induced asthma murine model. Mol Biol Rep 2024; 51:698. [PMID: 38811424 PMCID: PMC11136729 DOI: 10.1007/s11033-024-09360-5] [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: 01/24/2024] [Accepted: 02/16/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND Existing investigations suggest that the blockade of phosphoinositide 3-kinase (PI3K) activity contributes to inflammatory solution in allergic asthma, but whether this inhibition directly attenuates neutrophilic airway inflammation in vivo is still unclear. We explored the pharmacological effects of LY294002, a specific inhibitor of PI3K on the progression of neutrophilic airway inflammation and investigated the underlying mechanism. METHODS AND RESULTS Female C57BL/6 mice were intranasally sensitized with ovalbumin (OVA) together with lipopolysaccharide (LPS) on days 0 and 6, and challenged with OVA on days 14-17 to establish a neutrophilic airway disease model. In the challenge phase, a subset of mice was treated intratracheally with LY294002. We found that treatment of LY294002 attenuates clinic symptoms of inflammatory mice. Histological studies showed that LY294002 significantly inhibited inflammatory cell infiltration and mucus production. The treatment also significantly inhibited OVA-LPS induced increases in inflammatory cell counts, especially neutrophil counts, and IL-17 levels in bronchoalveolar lavage fluid (BALF). LY294002 treated mice exhibited significantly increased IL-10 levels in BALF compared to the untreated mice. In addition, LY294002 reduced the plasma concentrations of IL-6 and IL-17. The anti-inflammatory effects of LY29402 were correlated with the downregulation of NLRP3 inflammasome. CONCLUSIONS Our findings suggested that LY294002 as a potential pharmacological target for neutrophilic airway inflammation.
Collapse
Affiliation(s)
- Qun Li
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Bengbu Medical University, Anhui, China
| | - Guiyun Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, China
| | - Yunxiang Lv
- Department of Pulmonary and Critical Care MedicineAnhui Clinical and Preclinical Key Laboratory of Respiratory DiseaseMolecular Diagnosis Center, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, China.
| |
Collapse
|
5
|
Quoc QL, Choi Y, Hur GY, Park HS. New targets for type 2-low asthma. Korean J Intern Med 2024; 39:215-227. [PMID: 38317271 PMCID: PMC10918384 DOI: 10.3904/kjim.2023.299] [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: 07/14/2023] [Revised: 10/20/2023] [Accepted: 10/30/2023] [Indexed: 02/07/2024] Open
Abstract
Asthma is characterized by airway obstruction and inflammation, and presents significant diagnostic and treatment challenges. The concept of endotypes has improved understanding of the mechanisms of asthma and has stimulated the development of effective treatment strategies. Sputum profiles may be used to classify asthma into two major inflammatory types: type 2-high (T2H) and type 2-low (T2L) asthma. T2H, characterized by elevated type 2 inflammation, has been extensively studied and several effective biologic treatments have been developed. However, managing T2L is more difficult due to the lack of reliable biomarkers for accurate diagnosis and classification. Additionally, conventional anti-inflammatory therapy does not completely control the symptoms of T2L; therefore, further research is needed to identify effective biologic treatments. This review provides new insights into the clinical characteristics and underlying mechanisms of severe T2L and investigates potential therapeutic approaches to control the disease.
Collapse
Affiliation(s)
- Quang Luu Quoc
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon,
Korea
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon,
Korea
| | - Youngwoo Choi
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang,
Korea
| | - Gyu-Young Hur
- Department of Internal Medicine, Korea University College of Medicine, Seoul,
Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon,
Korea
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon,
Korea
| |
Collapse
|
6
|
Salcman B, Bahri R, West PW, Tontini C, Affleck K, Bulfone-Paus S. P2X7 Receptor-Induced Human Mast Cell Degranulation Is Enhanced by Interleukin 33. Int J Mol Sci 2024; 25:1730. [PMID: 38339008 PMCID: PMC10855801 DOI: 10.3390/ijms25031730] [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: 01/18/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
MCs are tissue-resident immune cells that strategically reside in barrier organs and respond effectively to a wide range of stimuli, such as IL-33, a mediator released upon epithelial damage. Adenosine triphosphate (ATP) accumulates at sites of tissue injury and is known to modulate MC activities. This study investigated how an inflammatory tissue environment rich in IL-33 modulates the ATP-mediated activation of MCs. Human primary MCs primed with IL-33 displayed a strongly increased response to ATP but not ADP. This resulted in increased degranulation, IL-8 release, and pERK1/2 signalling. Such effects are unique to IL-33 stimulation and not shared by the epithelial alarmin, TSLP. MC exposure to IL-33 also increased membrane expression of purinergic and ATP-binding P2X receptors. The use of selective P2X receptor inhibitors identified P2X7 receptor as the key mediator of the enhanced ATP-induced ERK1/2 signalling and degranulation in IL-33-primed MCs. Whilst the inhibition of P2X1 and P2X4 receptors had no effect on MC degranulation, inhibiting these receptors together with P2X7 resulted in further decreased MC-mediated degranulation. These data therefore point toward the potential mechanisms by which IL-33 contributes to the modulation of ATP-mediated activation in human MCs.
Collapse
Affiliation(s)
- Barbora Salcman
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9NT, UK; (B.S.); (R.B.); (P.W.W.); (C.T.)
| | - Rajia Bahri
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9NT, UK; (B.S.); (R.B.); (P.W.W.); (C.T.)
| | - Peter W. West
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9NT, UK; (B.S.); (R.B.); (P.W.W.); (C.T.)
| | - Chiara Tontini
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9NT, UK; (B.S.); (R.B.); (P.W.W.); (C.T.)
| | | | - Silvia Bulfone-Paus
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9NT, UK; (B.S.); (R.B.); (P.W.W.); (C.T.)
| |
Collapse
|
7
|
Guo Y, Zhou J, Wang Y, Wu X, Mou Y, Song X. Cell type-specific molecular mechanisms and implications of necroptosis in inflammatory respiratory diseases. Immunol Rev 2024; 321:52-70. [PMID: 37897080 DOI: 10.1111/imr.13282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Abstract
Necroptosis is generally considered as an inflammatory cell death form. The core regulators of necroptotic signaling are receptor-interacting serine-threonine protein kinases 1 (RIPK1) and RIPK3, and the executioner, mixed lineage kinase domain-like pseudokinase (MLKL). Evidence demonstrates that necroptosis contributes profoundly to inflammatory respiratory diseases that are common public health problem. Necroptosis occurs in nearly all pulmonary cell types in the settings of inflammatory respiratory diseases. The influence of necroptosis on cells varies depending upon the type of cells, tissues, organs, etc., which is an important factor to consider. Thus, in this review, we briefly summarize the current state of knowledge regarding the biology of necroptosis, and focus on the key molecular mechanisms that define the necroptosis status of specific cell types in inflammatory respiratory diseases. We also discuss the clinical potential of small molecular inhibitors of necroptosis in treating inflammatory respiratory diseases, and describe the pathological processes that engage cross talk between necroptosis and other cell death pathways in the context of respiratory inflammation. The rapid advancement of single-cell technologies will help understand the key mechanisms underlying cell type-specific necroptosis that are critical to effectively treat pathogenic lung infections and inflammatory respiratory diseases.
Collapse
Affiliation(s)
- Ying Guo
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Jin Zhou
- Key Laboratory of Spatiotemporal Single-Cell Technologies and Translational Medicine, Yantai, Shandong, China
- Department of Endocrinology, Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Yaqi Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Xueliang Wu
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
- Tumor Research Institute, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Yakui Mou
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Xicheng Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
- Key Laboratory of Spatiotemporal Single-Cell Technologies and Translational Medicine, Yantai, Shandong, China
| |
Collapse
|
8
|
Zheng R, Wu X, Li S, Chen X, Yan D, He J. Mechanism Exploration on the Immunoregulation of Allogeneic Heart Transplantation Rejection in Rats With Exosome miRNA and Proteins From Overexpressed IDO1 BMSCs. Cell Transplant 2024; 33:9636897241245796. [PMID: 38629748 PMCID: PMC11025427 DOI: 10.1177/09636897241245796] [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: 12/03/2023] [Revised: 01/29/2024] [Accepted: 03/21/2024] [Indexed: 04/19/2024] Open
Abstract
Immunoregulation and indoleamine 2,3-dioxygenase 1 (IDO1) play pivotal roles in the rejection of allogeneic organ transplantation. This study aims to elucidate the immune-related functional mechanisms of exosomes (Exos) derived from bone marrow-derived mesenchymal stem cells (BMSCs) overexpressing IDO1 in the context of allogeneic heart transplantation (HTx) rejection. A rat model of allogeneic HTx was established. Exos were extracted after transfection with oe-IDO1 and oe-NC from rat BMSCs. Exos were administered via the caudal vein for treatment. The survival of rats was analyzed, and reverse transcription qualitative PCR (RT-qPCR) and immunohistochemistry (IHC) were employed to detect the expression of related genes. Histopathological examination was conducted using hematoxylin and eosin (HE) staining, and flow cytometry was utilized to analyze T-cell apoptosis. Proteomics and RNA-seq analyses were performed on Exos. The data were subjected to functional enrichment analysis using the R language. A protein interaction network was constructed using the STRING database, and miRWalk, TargetScan, and miRDB databases predicted the target genes, differentially expressed miRNAs, and transcription factors (TFs). Exos from BMSCs overexpressing IDO1 prolonged the survival time of rats undergoing allogeneic HTx. These Exos reduced inflammatory cell infiltration, mitigated myocardial damage, induced CD4 T-cell apoptosis, and alleviated transplantation rejection. The correlation between Exos from BMSCs overexpressing IDO1 and immune regulation was profound. Notably, 13 immune-related differential proteins (Anxa1, Anxa2, C3, Ctsb, Hp, Il1rap, Ntn1, Ptx3, Thbs1, Hspa1b, Vegfc, Dcn, and Ptpn11) and 10 significantly different miRNAs were identified. Finally, six key immune proteins related to IDO1 were identified through common enrichment pathways, including Thbs1, Dcn, Ptpn11, Hspa1b, Il1rap, and Vegfc. Thirteen TFs of IDO1-related key miRNAs were obtained, and a TF-miRNA-mRNA-proteins regulatory network was constructed. Exosome miRNA derived from BMSCs overexpressing IDO1 may influence T-cell activation and regulate HTx rejection by interacting with mRNA.
Collapse
Affiliation(s)
- Rui Zheng
- Department of Laboratory, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Xinxin Wu
- Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Si Li
- Department of Cardiovascular Surgery, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Xinhao Chen
- Department of Cardiovascular Surgery, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Dan Yan
- Department of MICU, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Jigang He
- Department of Cardiovascular Surgery, The First People’s Hospital of Yunnan Province, Kunming, China
| |
Collapse
|
9
|
Zhou Y, Xu Z, Liu Z. Role of IL-33-ST2 pathway in regulating inflammation: current evidence and future perspectives. J Transl Med 2023; 21:902. [PMID: 38082335 PMCID: PMC10714644 DOI: 10.1186/s12967-023-04782-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023] Open
Abstract
Interleukin (IL)-33 is an alarmin of the IL-1 superfamily localized to the nucleus of expressing cells, such as endothelial cells, epithelial cells, and fibroblasts. In response to cellular damage or stress, IL-33 is released and activates innate immune responses in some immune and structural cells via its receptor interleukin-1 receptor like-1 (IL-1RL1 or ST2). Recently, IL-33 has become a hot topic of research because of its role in pulmonary inflammation. The IL-33-ST2 signaling pathway plays a pro-inflammatory role by activating the type 2 inflammatory response, producing type 2 cytokines and chemokines. Elevated levels of IL-33 and ST2 have been observed in chronic pulmonary obstructive disease (COPD). Notably, IL-33 is present in COPD induced by cigarette smoke or acute inflammations. The role of IL-33 in sepsis is becoming increasingly prominent, and understanding its significance in the treatment of sepsis associated with high mortality is critical. In addition to its pro-inflammatory effects, the IL-33-ST2 axis appears to play a role in bacterial clearance and tissue repair. In this review, we focused on the role of the IL-33-ST2 axis in sepsis, asthma, and COPD and summarized the therapeutic targets associated with this axis, providing a basis for future treatment.
Collapse
Affiliation(s)
- Yilu Zhou
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhendong Xu
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Zhiqiang Liu
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
10
|
Bailey TW, do Nascimento NC, Santos AP, Cox A, Sivasankar MP. Impact of Rehydration Following Systemic Dehydration on Vocal Fold Gene Expression. Laryngoscope 2023; 133:3499-3505. [PMID: 37345579 PMCID: PMC10739564 DOI: 10.1002/lary.30840] [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: 02/13/2023] [Revised: 05/17/2023] [Accepted: 06/09/2023] [Indexed: 06/23/2023]
Abstract
OBJECTIVE Biological data on the beneficial effects of vocal fold rehydration are lacking. This study aimed to examine the effects of acute systemic dehydration on vocal fold gene expression and determine whether rehydration would reverse these changes. METHODS Male New Zealand White rabbits (N = 24, n = 8/group) provided the animal model. Systemic dehydration was induced by 5 days of water volume restriction. Rehydration was provided by ad-lib water for 3 days following dehydration. Euhydrated rabbits were used as the control group. Vocal fold tissue was dissected. Seventeen genes were selected based on physiological function and role in supporting vocal fold structure, oxidative stress, hemodynamics, and extracellular matrix turnover. Relative gene expression was assessed by RT-qPCR. RESULTS Rehydration following systemic dehydration can modulate gene expression, with expression patterns suggesting that rehydration reverses dehydration-induced changes in over half of the tested genes. CLIC5 (chloride intracellular channel 5) and EFEMP1 (EGF containing fibulin extracellular matrix protein 1) genes were significantly upregulated in the dehydration group compared with the euhydrated control. A1BG (alpha-1B-glycoprotein) and IL1RAP (interleukin 1 receptor accessory protein) were downregulated by rehydration compared with the dehydration group. CONCLUSION This study provides molecular evidence for a transcriptional response to rehydration following acute systemic dehydration in the vocal folds. These data are the first to study gene expression following realistic dehydration and rehydration paradigms and provide biological data to support clinical recommendations to increase water intake after acute dehydration. LEVEL OF EVIDENCE NA Laryngoscope, 133:3499-3505, 2023.
Collapse
Affiliation(s)
- Taylor W Bailey
- Department of Speech, Language, and Hearing Sciences, Purdue University, West Lafayette, Indiana, USA
| | | | - Andrea P Santos
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, USA
| | - Abigail Cox
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, USA
| | - M Preeti Sivasankar
- Department of Speech, Language, and Hearing Sciences, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
11
|
Faiz A, Mahbub RM, Boedijono FS, Tomassen MI, Kooistra W, Timens W, Nawijn M, Hansbro PM, Johansen MD, Pouwels SD, Heijink IH, Massip F, de Biase MS, Schwarz RF, Adcock IM, Chung KF, van der Does A, Hiemstra PS, Goulaouic H, Xing H, Abdulai R, de Rinaldis E, Cunoosamy D, Harel S, Lederer D, Nivens MC, Wark PA, Kerstjens HAM, Hylkema MN, Brandsma CA, van den Berge M. IL-33 Expression Is Lower in Current Smokers at both Transcriptomic and Protein Levels. Am J Respir Crit Care Med 2023; 208:1075-1087. [PMID: 37708400 PMCID: PMC10867944 DOI: 10.1164/rccm.202210-1881oc] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 09/14/2023] [Indexed: 09/16/2023] Open
Abstract
Rationale: IL-33 is a proinflammatory cytokine thought to play a role in the pathogenesis of asthma and chronic obstructive pulmonary disease (COPD). A recent clinical trial using an anti-IL-33 antibody showed a reduction in exacerbation and improved lung function in ex-smokers but not current smokers with COPD. Objectives: This study aimed to understand the effects of smoking status on IL-33. Methods: We investigated the association of smoking status with the level of gene expression of IL-33 in the airways in eight independent transcriptomic studies of lung airways. Additionally, we performed Western blot analysis and immunohistochemistry for IL-33 in lung tissue to assess protein levels. Measurements and Main Results: Across the bulk RNA-sequencing datasets, IL-33 gene expression and its signaling pathway were significantly lower in current versus former or never-smokers and increased upon smoking cessation (P < 0.05). Single-cell sequencing showed that IL-33 is predominantly expressed in resting basal epithelial cells and decreases during the differentiation process triggered by smoke exposure. We also found a higher transitioning of this cellular subpopulation into a more differentiated cell type during chronic smoking, potentially driving the reduction of IL-33. Protein analysis demonstrated lower IL-33 levels in lung tissue from current versus former smokers with COPD and a lower proportion of IL-33-positive basal cells in current versus ex-smoking controls. Conclusions: We provide strong evidence that cigarette smoke leads to an overall reduction in IL-33 expression in transcriptomic and protein level, and this may be due to the decrease in resting basal cells. Together, these findings may explain the clinical observation that a recent antibody-based anti-IL-33 treatment is more effective in former than current smokers with COPD.
Collapse
Affiliation(s)
- Alen Faiz
- Respiratory Bioinformatics and Molecular Biology, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
- Groningen Research Institute for Asthma and COPD
- Department of Pulmonary Diseases, and
| | - Rashad M. Mahbub
- Respiratory Bioinformatics and Molecular Biology, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Fia Sabrina Boedijono
- Respiratory Bioinformatics and Molecular Biology, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
- Centre for Inflammation, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, New South Wales, Australia
| | - Milan I. Tomassen
- Groningen Research Institute for Asthma and COPD
- Department of Pathology & Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Wierd Kooistra
- Groningen Research Institute for Asthma and COPD
- Department of Pathology & Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Wim Timens
- Groningen Research Institute for Asthma and COPD
- Department of Pathology & Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Martijn Nawijn
- Groningen Research Institute for Asthma and COPD
- Department of Pathology & Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Philip M. Hansbro
- Centre for Inflammation, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, New South Wales, Australia
| | - Matt D. Johansen
- Centre for Inflammation, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, New South Wales, Australia
| | - Simon D. Pouwels
- Groningen Research Institute for Asthma and COPD
- Department of Pulmonary Diseases, and
| | - Irene H. Heijink
- Groningen Research Institute for Asthma and COPD
- Department of Pulmonary Diseases, and
- Department of Pathology & Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Florian Massip
- Centre for Computational Biology, Mines ParisTech, Paris Sciences et Lettres Research University, Paris, France
- Cancer and Genome: Bioinformatics, Biostatistics and Epidemiology of Complex Systems Institut Curie, Paris, France
- Institut Nationale de la Santé et de la Recherche Médicale U900, Paris, France
| | - Maria Stella de Biase
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Roland F. Schwarz
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Institute for Computational Cancer Biology, Center for Integrated Oncology, Cancer Research Center Cologne Essen, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
- Berlin Institute for the Foundations of Learning and Data, Berlin, Germany
| | - Ian M. Adcock
- National Heart & Lung Institute, Imperial College London, London, United Kingdom
| | - Kian F. Chung
- National Heart & Lung Institute, Imperial College London, London, United Kingdom
| | - Anne van der Does
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Pieter S. Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | - Sivan Harel
- Regeneron Pharmaceuticals, Tarrytown, New York
| | | | | | - Peter A. Wark
- Centre for Asthma & Respiratory Disease, The University of Newcastle, Newcastle, New South Wales, Australia; and
- Hunter Medical Research Institute, Vaccines, Infection, Viruses & Asthma Newcastle, New South Wales, Australia
| | - Huib A. M. Kerstjens
- Groningen Research Institute for Asthma and COPD
- Department of Pulmonary Diseases, and
| | - Machteld N. Hylkema
- Groningen Research Institute for Asthma and COPD
- Department of Pathology & Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Corry-Anke Brandsma
- Groningen Research Institute for Asthma and COPD
- Department of Pathology & Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Maarten van den Berge
- Groningen Research Institute for Asthma and COPD
- Department of Pulmonary Diseases, and
| | - the Cambridge Lung Cancer Early Detection Programme
- Respiratory Bioinformatics and Molecular Biology, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
- Groningen Research Institute for Asthma and COPD
- Department of Pulmonary Diseases, and
- Department of Pathology & Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- Centre for Inflammation, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, New South Wales, Australia
- Centre for Computational Biology, Mines ParisTech, Paris Sciences et Lettres Research University, Paris, France
- Cancer and Genome: Bioinformatics, Biostatistics and Epidemiology of Complex Systems Institut Curie, Paris, France
- Institut Nationale de la Santé et de la Recherche Médicale U900, Paris, France
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Institute for Computational Cancer Biology, Center for Integrated Oncology, Cancer Research Center Cologne Essen, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
- Berlin Institute for the Foundations of Learning and Data, Berlin, Germany
- National Heart & Lung Institute, Imperial College London, London, United Kingdom
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
- Sanofi, Chilly-Mazarin, France
- Sanofi, Cambridge, Massachusetts
- Regeneron Pharmaceuticals, Tarrytown, New York
- Centre for Asthma & Respiratory Disease, The University of Newcastle, Newcastle, New South Wales, Australia; and
- Hunter Medical Research Institute, Vaccines, Infection, Viruses & Asthma Newcastle, New South Wales, Australia
| |
Collapse
|
12
|
Cruz T, Mendoza N, Casas-Recasens S, Noell G, Hernandez-Gonzalez F, Frino-Garcia A, Alsina-Restoy X, Molina M, Rojas M, Agustí A, Sellares J, Faner R. Lung immune signatures define two groups of end-stage IPF patients. Respir Res 2023; 24:236. [PMID: 37770891 PMCID: PMC10540496 DOI: 10.1186/s12931-023-02546-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 09/21/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND The role of the immune system in the pathobiology of Idiopathic Pulmonary Fibrosis (IPF) is controversial. METHODS To investigate it, we calculated immune signatures with Gene Set Variation Analysis (GSVA) and applied them to the lung transcriptome followed by unbiased cluster analysis of GSVA immune-enrichment scores, in 109 IPF patients from the Lung Tissue Research Consortium (LTRC). Results were validated experimentally using cell-based methods (flow cytometry) in lung tissue of IPF patients from the University of Pittsburgh (n = 26). Finally, differential gene expression and hypergeometric test were used to explore non-immune differences between clusters. RESULTS We identified two clusters (C#1 and C#2) of IPF patients of similar size in the LTRC dataset. C#1 included 58 patients (53%) with enrichment in GSVA immune signatures, particularly cytotoxic and memory T cells signatures, whereas C#2 included 51 patients (47%) with an overall lower expression of GSVA immune signatures (results were validated by flow cytometry with similar unbiased clustering generation). Differential gene expression between clusters identified differences in cilium, epithelial and secretory cell genes, all of them showing an inverse correlation with the immune response signatures. Notably, both clusters showed distinct features despite clinical similarities. CONCLUSIONS In end-stage IPF lung tissue, we identified two clusters of patients with very different levels of immune signatures and gene expression but with similar clinical characteristics. Weather these immune clusters differentiate diverse disease trajectories remains unexplored.
Collapse
Affiliation(s)
- Tamara Cruz
- Centro Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), Barcelona, Spain
- Fundació Clínic Per a La Recerca Biomèdica - IDIBAPS (FCRB-IDIBAPS), C/Casanova 143, Cellex, P2A, 08036, Barcelona, Spain
| | - Núria Mendoza
- Centro Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), Barcelona, Spain
- Fundació Clínic Per a La Recerca Biomèdica - IDIBAPS (FCRB-IDIBAPS), C/Casanova 143, Cellex, P2A, 08036, Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Sandra Casas-Recasens
- Centro Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), Barcelona, Spain
| | - Guillaume Noell
- Centro Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), Barcelona, Spain
- Fundació Clínic Per a La Recerca Biomèdica - IDIBAPS (FCRB-IDIBAPS), C/Casanova 143, Cellex, P2A, 08036, Barcelona, Spain
| | - Fernanda Hernandez-Gonzalez
- Fundació Clínic Per a La Recerca Biomèdica - IDIBAPS (FCRB-IDIBAPS), C/Casanova 143, Cellex, P2A, 08036, Barcelona, Spain
- Department of Pulmonology, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Alejandro Frino-Garcia
- Fundació Clínic Per a La Recerca Biomèdica - IDIBAPS (FCRB-IDIBAPS), C/Casanova 143, Cellex, P2A, 08036, Barcelona, Spain
- Department of Pulmonology, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Xavi Alsina-Restoy
- Fundació Clínic Per a La Recerca Biomèdica - IDIBAPS (FCRB-IDIBAPS), C/Casanova 143, Cellex, P2A, 08036, Barcelona, Spain
- Department of Pulmonology, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - María Molina
- Interstitial Lung Disease Unit, Respiratory Department, University Hospital of Bellvitge, IDIBELL, Hospitalet de Llobregat (Barcelona), CIBERES, Barcelona, Spain
| | - Mauricio Rojas
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Alvar Agustí
- Centro Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), Barcelona, Spain
- Fundació Clínic Per a La Recerca Biomèdica - IDIBAPS (FCRB-IDIBAPS), C/Casanova 143, Cellex, P2A, 08036, Barcelona, Spain
- Department of Pulmonology, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Jacobo Sellares
- Centro Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), Barcelona, Spain
- Fundació Clínic Per a La Recerca Biomèdica - IDIBAPS (FCRB-IDIBAPS), C/Casanova 143, Cellex, P2A, 08036, Barcelona, Spain
- Department of Pulmonology, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Rosa Faner
- Centro Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), Barcelona, Spain.
- Fundació Clínic Per a La Recerca Biomèdica - IDIBAPS (FCRB-IDIBAPS), C/Casanova 143, Cellex, P2A, 08036, Barcelona, Spain.
- Biomedicine Department, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
13
|
Chung KF. Type-2-low severe asthma endotypes for new treatments: the new asthma frontier. Curr Opin Allergy Clin Immunol 2023; 23:199-204. [PMID: 37185823 DOI: 10.1097/aci.0000000000000899] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
PURPOSE OF REVIEW Type-2 (T2)-high asthma represents a well defined group of severe eosinophilic asthma for which there are now effective biologic therapies targetting the interleukins (ILs) 4, 5 and 13, and Immunoglobulin E. T2-low asthma detected in the clinic by a low blood eosinophil count remains ill-defined and is the focus of this review. RECENT FINDINGS By analysing transcriptomic and proteomic expression in sputum samples in U-BIOPRED cohort, both T2-high and -low molecular phenotypes have been described. Using clustering approaches, a neutrophilic-predominant cluster associated with activation markers of neutrophilic and inflammasome activation with interferon and tumour necrosis factor expression, together with a cluster of paucigranulocytic inflammation linked to oxidative phosphorylation and senescence pathways have been described. Using gene set variation analysis, specific molecular phenotypes driven by IL-6 trans-signalling pathway, or those by IL-6, IL-17 and IL-22 pathways were identified linked to a mixed granulocytic or neutrophilic inflammation. SUMMARY Previous trials of antineutrophilic agents in asthma have failed because enrolled patients were not specifically chosen for these targeted treatments. Although the T2-low molecular pathways should be validated in other cohorts, the availability of targeted therapies indicated for other autoimmune conditions should encourage a trial of these respective biological therapies for these specific molecular phenotypes.
Collapse
Affiliation(s)
- Kian Fan Chung
- National Heart & Lung Institute, Imperial College London; Royal Brompton and Harefield Hospitals, London, UK
| |
Collapse
|
14
|
Beetler DJ, Bruno KA, Di Florio DN, Douglass EJ, Shrestha S, Tschöpe C, Cunningham MW, Krejčí J, Bienertová-Vašků J, Pankuweit S, McNamara DM, Jeon ES, van Linthout S, Blauwet LA, Cooper LT, Fairweather D. Sex and age differences in sST2 in cardiovascular disease. Front Cardiovasc Med 2023; 9:1073814. [PMID: 36741845 PMCID: PMC9889877 DOI: 10.3389/fcvm.2022.1073814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/23/2022] [Indexed: 01/19/2023] Open
Abstract
Aims The goal of this study was to determine whether sex and age differences exist for soluble ST2 (sST2) for several cardiovascular diseases (CVDs). Methods We examined sST2 levels using an ELISA kit for myocarditis (n = 303), cardiomyopathy (n = 293), coronary artery disease (CAD) (n = 239), myocardial infarct (MI) (n = 159), and congestive heart failure (CHF) (n = 286) and compared them to controls that did not have CVDs (n = 234). Results Myocarditis occurred in this study in relatively young patients around age 40 while the other CVDs occurred more often in older individuals around age 60. We observed a sex difference in sST2 by age only in myocarditis patients (men aged 38, women 46, p = 0.0002), but not for other CVDs. Sera sST2 levels were significantly elevated compared to age-matched controls for all CVDs: myocarditis (p ≤ 0.0001), cardiomyopathy (p = 0.0009), CAD (p = 0.03), MI (p = 0.034), and CHF (p < 0.0001) driven by elevated sST2 levels in females for all CVDs except myocarditis, which was elevated in both females (p = 0.002) and males (p ≤ 0.0001). Sex differences in sST2 levels were found for myocarditis and cardiomyopathy but no other CVDs and were higher in males (myocarditis p = 0.0035; cardiomyopathy p = 0.0047). sST2 levels were higher in women with myocarditis over 50 years of age compared to men (p = 0.0004) or women under 50 years of age (p = 0.015). In cardiomyopathy and MI patients, men over 50 had significantly higher levels of sST2 than women (p = 0.012 and p = 0.043, respectively) but sex and age differences were not detected in other CVDs. However, women with cardiomyopathy that experienced early menopause had higher sST2 levels than those who underwent menopause at a natural age range (p = 0.02). Conclusion We found that sex and age differences in sera sST2 exist for myocarditis, cardiomyopathy, and MI, but were not observed in other CVDs including CAD and CHF. These initial findings in patients with self-reported CVDs indicate that more research is needed into sex and age differences in sST2 levels in individual CVDs.
Collapse
Affiliation(s)
- Danielle J. Beetler
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, United States
| | - Katelyn A. Bruno
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida, Gainesville, FL, United States
| | - Damian N. Di Florio
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, United States
| | - Erika J. Douglass
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Swikriti Shrestha
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, United States
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Carsten Tschöpe
- Berlin Institute of Health (BIH) at Charité – Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- German Centre for Cardiovascular Research, Berlin, Germany
- Department of Cardiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Madeleine W. Cunningham
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Jan Krejčí
- First Department of Internal Medicine and Cardioangiology, St. Anne’s University Hospital, Brno, Czechia
- Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Julie Bienertová-Vašků
- Incubator of Kinanthropology Research, Faculty of Sports, Masaryk University, Brno, Czechia
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Sabine Pankuweit
- Department Internal Medicine-Cardiology, Philipps-University of Marburg, Marburg, Germany
| | - Dennis M. McNamara
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- University of Pittsburgh Medical Center (UPMC), Heart and Vascular Institute, Pittsburgh, PA, United States
| | - Eun-Seok Jeon
- Division of Cardiology, Department of Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sophie van Linthout
- Berlin Institute of Health (BIH) at Charité – Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- German Centre for Cardiovascular Research, Berlin, Germany
| | | | - Leslie T. Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
- Department of Environmental Health Sciences and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
15
|
Li Y, Liu Y, Duo M, Wu R, Jiang T, Li P, Wang Y, Cheng Z. Bioinformatic analysis and preliminary validation of potential therapeutic targets for COVID-19 infection in asthma patients. Cell Commun Signal 2022; 20:201. [PMID: 36575422 PMCID: PMC9793391 DOI: 10.1186/s12964-022-01010-2] [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: 10/20/2022] [Accepted: 12/03/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 causes coronavirus disease 19 (COVID-19). The number of confirmed cases of COVID-19 is also rapidly increasing worldwide, posing a significant challenge to human safety. Asthma is a risk factor for COVID-19, but the underlying molecular mechanisms of the asthma-COVID-19 interaction remain unclear. METHODS We used transcriptome analysis to discover molecular biomarkers common to asthma and COVID-19. Gene Expression Omnibus database RNA-seq datasets (GSE195599 and GSE196822) were used to identify differentially expressed genes (DEGs) in asthma and COVID-19 patients. After intersecting the differentially expressed mRNAs, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed to identify the common pathogenic molecular mechanism. Bioinformatic methods were used to construct protein-protein interaction (PPI) networks and identify key genes from the networks. An online database was used to predict interactions between transcription factors and key genes. The differentially expressed long noncoding RNAs (lncRNAs) in the GSE195599 and GSE196822 datasets were intersected to construct a competing endogenous RNA (ceRNA) regulatory network. Interaction networks were constructed for key genes with RNA-binding proteins (RBPs) and oxidative stress-related proteins. The diagnostic efficacy of key genes in COVID-19 was verified with the GSE171110 dataset. The differential expression of key genes in asthma was verified with the GSE69683 dataset. An asthma cell model was established with interleukins (IL-4, IL-13 and IL-17A) and transfected with siRNA-CXCR1. The role of CXCR1 in asthma development was preliminarily confirmed. RESULTS By intersecting the differentially expressed genes for COVID-19 and asthma, 393 common DEGs were obtained. GO and KEGG enrichment analyses of the DEGs showed that they mainly affected inflammation-, cytokine- and immune-related functions and inflammation-related signaling pathways. By analyzing the PPI network, we obtained 10 key genes: TLR4, TLR2, MMP9, EGF, HCK, FCGR2A, SELP, NFKBIA, CXCR1, and SELL. By intersecting the differentially expressed lncRNAs for COVID-19 and asthma, 13 common differentially expressed lncRNAs were obtained. LncRNAs that regulated microRNAs (miRNAs) were mainly concentrated in intercellular signal transduction, apoptosis, immunity and other related functional pathways. The ceRNA network suggested that there were a variety of regulatory miRNAs and lncRNAs upstream of the key genes. The key genes could also bind a variety of RBPs and oxidative stress-related genes. The key genes also had good diagnostic value in the verification set. In the validation set, the expression of key genes was statistically significant in both the COVID-19 group and the asthma group compared with the healthy control group. CXCR1 expression was upregulated in asthma cell models, and interference with CXCR1 expression significantly reduced cell viability. CONCLUSIONS Key genes may become diagnostic and predictive biomarkers of outcomes in COVID-19 and asthma. Video Abstract.
Collapse
Affiliation(s)
- Yue Li
- grid.412633.10000 0004 1799 0733Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Ye Liu
- grid.412633.10000 0004 1799 0733Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Mengjie Duo
- grid.412633.10000 0004 1799 0733Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Ruhao Wu
- grid.412633.10000 0004 1799 0733Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Tianci Jiang
- grid.412633.10000 0004 1799 0733Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Pengfei Li
- grid.412633.10000 0004 1799 0733Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Yu Wang
- grid.412633.10000 0004 1799 0733Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Zhe Cheng
- grid.412633.10000 0004 1799 0733Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| |
Collapse
|