1
|
Cai XX, Huang YH, Lin YCD, Huang HY, Chen YG, Zhang DP, Zhang T, Liu Y, Zuo HL, Huang HD. A comprehensive review of small molecules, targets, and pathways in ulcerative colitis treatment. Eur J Med Chem 2025; 291:117645. [PMID: 40279769 DOI: 10.1016/j.ejmech.2025.117645] [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: 02/11/2025] [Revised: 04/06/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025]
Abstract
Ulcerative colitis (UC), a chronic inflammatory bowel disease (IBD), poses significant clinical challenges because of its complex pathophysiology, long-term nature, and the limited efficacy of existing treatments. Small-molecule compounds, particularly those that are able to modulate inflammation-related signaling pathways and, in many cases, occur in nature, offer a promising alternative or supplement to conventional therapies. Studies on molecules for UC therapeutics reported in 1394 publications over the past 30 years were collected from the Web of Science (WOS) database. Only studies that verified therapeutic efficacy through animal experiments were included. Through an analysis of the molecular classes, structures, common targets, and pathways using network pharmacology, we identified 14 classes of compounds, 5 direct-target modules, and 3 crucial downstream pathways. Alkaloids, phenylpropanoids, flavonoids, and terpenes (and their derivatives) appeared most frequently and mainly targeted lipid metabolism, oxidative stress, immune regulation, signaling transduction, and cancer-related pathways. Notably, there has been an increasing trend of applying naturally sourced compounds in both preclinical and clinical trials, especially flavonoids, over the last five years. Although progress in UC research has been made, the majority of studies have focused on the overall therapeutic effects and biomarker alterations, with limited emphasis on the direct targets and underlying mechanisms. These findings highlight the need to explore novel small-molecule therapeutic strategies for UC, focusing on clearly defined targets and precise modes of action.
Collapse
Affiliation(s)
- Xiao-Xuan Cai
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China
| | - Yi-Han Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China
| | - Yang-Chi-Dung Lin
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China
| | - Hsi-Yuan Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China
| | - Yi-Gang Chen
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China
| | - Da-Peng Zhang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China
| | - Tao Zhang
- R&D center, Better Way (Shanghai) Cosmetics Co., Ltd., Shanghai, 201103, PR China
| | - Yue Liu
- R&D center, Better Way (Shanghai) Cosmetics Co., Ltd., Shanghai, 201103, PR China
| | - Hua-Li Zuo
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China.
| | - Hsien-Da Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Department of Endocrinology, Key Laboratory of Endocrinology of National Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, PR China.
| |
Collapse
|
2
|
Eladham MW, Sharif-Askari NS, Sekar P, Mdkhana B, Selvakumar B, Al-Sheakly BKS, Sharif-Askari FS, Hachim I, Halwani R. The role of gut leakage and immune cell miss-homing on gut dysbiosis-induced lung inflammation in a DSS mice model. PLoS One 2025; 20:e0324230. [PMID: 40435188 PMCID: PMC12118880 DOI: 10.1371/journal.pone.0324230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/22/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND Inflammatory Bowel Disease (IBD), encompassing Crohn's disease and ulcerative colitis, affects millions globally, with extraintestinal manifestations (EIMs) occurring in 25-40% of patients. Among these, respiratory complications are of particular concern, yet the immunologic and physiologic mechanisms underlying gut-lung interactions remain poorly understood. The gut-lung axis (GLA) describes bi-directional communication between the gut and lungs, where microbial dysbiosis in the gut can drive lung inflammation and immune dysregulation. METHODS Mice were treated with 4% DSS for 7 days to induce colitis. Gut permeability, tight junction protein expression, lung inflammation, immune cell trafficking, and microbial translocation were assessed through histology, qPCR, flow cytometry, and GFP-tagged fecal microbiome experiments. RESULTS DSS treatment led to significant disruption of the gut barrier, with upregulation of gut leakage markers and downregulation of tight junction proteins. Lung inflammation was characterized by elevated IL-17, neutrophil infiltration, and airway hyperresponsiveness. Flow cytometry revealed mis-homing of gut-primed immune cells (α4β7+ and CCR9 + CD4+) to the lungs and tracking bacteria via GFP- tagged fecal microbiome confirmed microbial translocation from the gut to the lungs which may contribute to lung inflammation. CONCLUSION Disrupted gut integrity facilitates microbial translocation and immune cell mis-homing, contributing to lung inflammation. These results provide new insights into how gut dysbiosis influences respiratory inflammation.
Collapse
Affiliation(s)
- Mariam Wed Eladham
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Narjes Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Priyadharshini Sekar
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Bushra Mdkhana
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Balachandar Selvakumar
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | | | - Fatemeh Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Ibrahim Hachim
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Rabih Halwani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Prince Abdullah Ben Khaled Celiac Disease Chair, Department of Pediatrics, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
3
|
Kim SH, White Z, Gainullina A, Kang S, Kim J, Dominguez JR, Choi Y, Cabrera I, Plaster M, Takahama M, Czepielewski RS, Yeom J, Gunzer M, Hay N, David O, Chevrier N, Sano T, Kim KW. IL-10 sensing by lung interstitial macrophages prevents bacterial dysbiosis-driven pulmonary inflammation and maintains immune homeostasis. Immunity 2025; 58:1306-1326.e7. [PMID: 40306274 DOI: 10.1016/j.immuni.2025.04.004] [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: 02/02/2024] [Revised: 10/02/2024] [Accepted: 04/03/2025] [Indexed: 05/02/2025]
Abstract
Crosstalk between the immune system and the microbiome is critical for maintaining immune homeostasis. Here, we examined this communication and the impact of immune-suppressive IL-10 signaling on pulmonary homeostasis. We found that IL-10 sensing by interstitial macrophages (IMs) is required to prevent spontaneous lung inflammation. Loss of IL-10 signaling in IMs initiated an inflammatory cascade through the activation of classical monocytes and CD4+ T cell subsets, leading to chronic lung inflammation with age. Analyses of antibiotic-treated and germ-free mice established that lung inflammation in the animals lacking IL-10 signaling was triggered by commensal bacteria. 16S rRNA sequencing revealed Delftia acidovorans and Rhodococcus erythropolis as potential drivers of lung inflammation. Intranasal administration of these bacteria or transplantation of human fecal microbiota elicited lung inflammation in gnotobiotic Il10-deficient mice. These findings highlight that IL-10 sensing by IMs contributes to pulmonary homeostasis by preventing lung inflammation caused by commensal dysbiosis.
Collapse
Affiliation(s)
- Seung Hyeon Kim
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Zachary White
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL, USA
| | | | - Soeun Kang
- Department of Biochemistry and Genetics, University of Illinois College of Medicine, Chicago, IL, USA
| | - Jiseon Kim
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Joseph R Dominguez
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Yeonwoo Choi
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Ivan Cabrera
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Madison Plaster
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Michihiro Takahama
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Rafael S Czepielewski
- Immunology Center of Georgia, Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA; Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Jinki Yeom
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Nissim Hay
- Department of Biochemistry and Genetics, University of Illinois College of Medicine, Chicago, IL, USA; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Odile David
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA; Department of Pathology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Nicolas Chevrier
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Teruyuki Sano
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL, USA; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA.
| | - Ki-Wook Kim
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
4
|
Tan J, Han X, Li S, Wang Q, Zhao L, Li Y, Duan S, Zhang L. Platelet-Activating Factor Disrupts the Nasal Epithelial Barrier Independently of the Platelet-Activating Factor Receptor Pathway. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2025; 17:212-225. [PMID: 40204506 PMCID: PMC11982642 DOI: 10.4168/aair.2025.17.2.212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 04/11/2025]
Abstract
PURPOSE Platelet-activating factor (PAF) mediates nasal congestion and rhinorrhea by affecting vascular permeability, but the underlying mechanisms remain unclear. Here, we sought to explore the effect of PAF on the nasal epithelial barrier in chronic rhinosinusitis with nasal polyps (CRSwNP). METHODS Human nasal epithelial cells (hNECs) were pre-treated with Apafant, a PAF receptor (PAFR) inhibitor, or MCC950, an NOD-like receptor protein 3 (NLRP3) inflammasome inhibitor, before PAF stimulation. The nasal epithelial barrier function was assessed by measuring the transepithelial electrical resistance (TER) and sodium fluorescein flux. Additionally, the expression of mRNAs and proteins of tight junctions were assessed. RESULTS PAF significantly decreased TER and enhanced the fluorescein flux permeability in air-liquid interface cultures of hNECs, while also downregulating the expression of ZO-1, occludin, claudin-1, and claudin-4. However, the disruptive effect of PAF on the nasal epithelial barrier was attenuated by MCC950, but not by Apafant. Furthermore, MCC950 inhibited PAF-induced NLRP3 activation and its downstream molecules, including caspase-1, ASC, interleukin (IL)-1β, and IL-18. CONCLUSIONS Our findings indicate that PAF has the potential to disrupt the nasal epithelial barrier in CRSwNP and may be linked to NLRP3 activation, while PAFR is not essential for this process. This discovery helps to explain why PAFR antagonists are ineffective in blocking PAF-mediated inflammation in clinical settings.
Collapse
Affiliation(s)
- Juan Tan
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Xinling Han
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Shenting Li
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Qiqi Wang
- Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Limin Zhao
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Ying Li
- Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Su Duan
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China.
| | - Luo Zhang
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
- Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Research Unit of Diagnosis and Treatment of Chronic Nasal Disease, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
5
|
Fan W, Xu Y, He X, Luo P, Zhu J, Li J, Wang R, Yuan Q, Wu K, Hu W, Zhao Y, Xu S, Cheng X, Wang Y, Xu HE, Zhuang Y. Molecular basis for the activation of PAF receptor by PAF. Cell Rep 2024; 43:114422. [PMID: 38943642 DOI: 10.1016/j.celrep.2024.114422] [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: 03/14/2024] [Revised: 05/07/2024] [Accepted: 06/14/2024] [Indexed: 07/01/2024] Open
Abstract
Platelet-activating factor (PAF) is a potent phospholipid mediator crucial in multiple inflammatory and immune responses through binding and activating the PAF receptor (PAFR). However, drug development targeting the PAFR has been limited, partly due to an incomplete understanding of its activation mechanism. Here, we present a 2.9-Å structure of the PAF-bound PAFR-Gi complex. Structural and mutagenesis analyses unveil a specific binding mode of PAF, with the choline head forming cation-π interactions within PAFR hydrophobic pocket, while the alkyl tail penetrates deeply into an aromatic cleft between TM4 and TM5. Binding of PAF modulates conformational changes in key motifs of PAFR, triggering the outward movement of TM6, TM7, and helix 8 for G protein coupling. Molecular dynamics simulation suggests a membrane-side pathway for PAF entry into PAFR via the TM4-TM5 cavity. By providing molecular insights into PAFR signaling, this work contributes a foundation for developing therapeutic interventions targeting PAF signal axis.
Collapse
Affiliation(s)
- Wenjia Fan
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210046, China; The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Youwei Xu
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xinheng He
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ping Luo
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jingpeng Zhu
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Junrui Li
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ruolan Wang
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qingning Yuan
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; The Shanghai Advanced Electron Microscope Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Kai Wu
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; The Shanghai Advanced Electron Microscope Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wen Hu
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; The Shanghai Advanced Electron Microscope Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuxi Zhao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210046, China; The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Shiqi Xu
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xi Cheng
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yue Wang
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - H Eric Xu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210046, China; The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Youwen Zhuang
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Medicinal Bioinformatics Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China.
| |
Collapse
|
6
|
Silva IS, Almeida AD, Lima Filho ACM, Fernandes-Braga W, Barra A, Oliveira HMC, Cassali GD, Capettini LSA, Menezes GB, Alvarez-Leite JI, Leite MF, Klein A. Platelet-activating factor and protease-activated receptor 2 cooperate to promote neutrophil recruitment and lung inflammation through nuclear factor-kappa B transactivation. Sci Rep 2023; 13:21637. [PMID: 38062077 PMCID: PMC10703791 DOI: 10.1038/s41598-023-48365-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
Although it is well established that platelet-activated receptor (PAF) and protease-activated receptor 2 (PAR2) play a pivotal role in the pathophysiology of lung and airway inflammatory diseases, a role for a PAR2-PAFR cooperation in lung inflammation has not been investigated. Here, we investigated the role of PAR2 in PAF-induced lung inflammation and neutrophil recruitment in lungs of BALB/c mice. Mice were pretreated with the PAR2 antagonist ENMD1068, PAF receptor (PAFR) antagonist WEB2086, or aprotinin prior to intranasal instillation of carbamyl-PAF (C-PAF) or the PAR2 agonist peptide SLIGRL-NH2 (PAR2-AP). Leukocyte infiltration in bronchoalveolar lavage fluid (BALF), C-X-C motif ligand 1 (CXCL)1 and CXCL2 chemokines, myeloperoxidase (MPO), and N-acetyl-glycosaminidase (NAG) levels in BALF, or lung inflammation were evaluated. Intracellular calcium signaling, PAFR/PAR2 physical interaction, and the expression of PAR2 and nuclear factor-kappa B (NF-КB, p65) transcription factor were investigated in RAW 264.7 cells stimulated with C-PAF in the presence or absence of ENMD1068. C-PAF- or PAR2-AP-induced neutrophil recruitment into lungs was inhibited in mice pretreated with ENMD1068 and aprotinin or WEB2086, respectively. PAR2 blockade impaired C-PAF-induced neutrophil rolling and adhesion, lung inflammation, and production of MPO, NAG, CXCL1, and CXCL2 production in lungs of mice. PAFR activation reduced PAR2 expression and physical interaction of PAR2 and PAFR; co-activation is required for PAFR/PAR2 physical interaction. PAR2 blockade impaired C-PAF-induced calcium signal and NF-κB p65 translocation in RAW 264.7 murine macrophages. This study provides the first evidence for a cooperation between PAFR and PAR2 mediating neutrophil recruitment, lung inflammation, and macrophage activation.
Collapse
Affiliation(s)
- Irismara Sousa Silva
- Laboratory of Inflammation and Proteases, Department of Pharmacology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Aline D Almeida
- Laboratory of Inflammation and Proteases, Department of Pharmacology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | | | - Weslley Fernandes-Braga
- Laboratory of Atherosclerosis and Nutritional Biochemistry (LABIN-UFMG), Department of Biochemistry and Immunology, ICB/UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Ayslan Barra
- Laboratory of Inflammation and Proteases, Department of Pharmacology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | | | | | - Luciano S A Capettini
- Laboratory of Vascular Biology, Department of Pharmacology, ICB/UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Gustavo B Menezes
- Department of Morphology, ICB/UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Jacqueline I Alvarez-Leite
- Laboratory of Atherosclerosis and Nutritional Biochemistry (LABIN-UFMG), Department of Biochemistry and Immunology, ICB/UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Maria F Leite
- Department of Physiology and Biophysics, ICB/UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - André Klein
- Laboratory of Inflammation and Proteases, Department of Pharmacology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
7
|
Liu G, Haw TJ, Starkey MR, Philp AM, Pavlidis S, Nalkurthi C, Nair PM, Gomez HM, Hanish I, Hsu AC, Hortle E, Pickles S, Rojas-Quintero J, Estepar RSJ, Marshall JE, Kim RY, Collison AM, Mattes J, Idrees S, Faiz A, Hansbro NG, Fukui R, Murakami Y, Cheng HS, Tan NS, Chotirmall SH, Horvat JC, Foster PS, Oliver BG, Polverino F, Ieni A, Monaco F, Caramori G, Sohal SS, Bracke KR, Wark PA, Adcock IM, Miyake K, Sin DD, Hansbro PM. TLR7 promotes smoke-induced experimental lung damage through the activity of mast cell tryptase. Nat Commun 2023; 14:7349. [PMID: 37963864 PMCID: PMC10646046 DOI: 10.1038/s41467-023-42913-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/25/2023] [Indexed: 11/16/2023] Open
Abstract
Toll-like receptor 7 (TLR7) is known for eliciting immunity against single-stranded RNA viruses, and is increased in both human and cigarette smoke (CS)-induced, experimental chronic obstructive pulmonary disease (COPD). Here we show that the severity of CS-induced emphysema and COPD is reduced in TLR7-deficient mice, while inhalation of imiquimod, a TLR7-agonist, induces emphysema without CS exposure. This imiquimod-induced emphysema is reduced in mice deficient in mast cell protease-6, or when wild-type mice are treated with the mast cell stabilizer, cromolyn. Furthermore, therapeutic treatment with anti-TLR7 monoclonal antibody suppresses CS-induced emphysema, experimental COPD and accumulation of pulmonary mast cells in mice. Lastly, TLR7 mRNA is increased in pre-existing datasets from patients with COPD, while TLR7+ mast cells are increased in COPD lungs and associated with severity of COPD. Our results thus support roles for TLR7 in mediating emphysema and COPD through mast cell activity, and may implicate TLR7 as a potential therapeutic target.
Collapse
Affiliation(s)
- Gang Liu
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Tatt Jhong Haw
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Malcolm R Starkey
- Depatrment of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Ashleigh M Philp
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Healthcare clinical campus, UNSW, Sydney, Australia
| | - Stelios Pavlidis
- The Airways Disease Section, National Heart & Lung Institute, Imperial College London, London, UK
| | - Christina Nalkurthi
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Prema M Nair
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Henry M Gomez
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Irwan Hanish
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Alan Cy Hsu
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Elinor Hortle
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Sophie Pickles
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | | | - Raul San Jose Estepar
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Jacqueline E Marshall
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Richard Y Kim
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, Australia
| | - Adam M Collison
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Joerg Mattes
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Sobia Idrees
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Alen Faiz
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Nicole G Hansbro
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Ryutaro Fukui
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minatoku, Tokyo, Japan
| | - Yusuke Murakami
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Musashino University, Nishitokyo-shi, Tokyo, Japan
| | - Hong Sheng Cheng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Sanjay H Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - Jay C Horvat
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Paul S Foster
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Brian Gg Oliver
- Woolcock Institute of Medical Research, University of Sydney & School of Life Sciences, University of Technology, Sydney, Australia
| | | | - Antonio Ieni
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Section of Anatomic Pathology, Università di Messina, Messina, Italy
| | - Francesco Monaco
- Thoracic Surgery, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Gaetano Caramori
- Pneumologia, Dipartimento BIOMORF and Dipartimento di Medicina e Chirurgia, Universities of Messina and Parma, Messina, Italy
| | - Sukhwinder S Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, Australia
| | - Ken R Bracke
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Peter A Wark
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Ian M Adcock
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Healthcare clinical campus, UNSW, Sydney, Australia
| | - Kensuke Miyake
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minatoku, Tokyo, Japan
| | - Don D Sin
- The University of British Columbia Centre for Heart Lung Innovation, St Paul's Hospital & Respiratory Division, Dept of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia.
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia.
| |
Collapse
|
8
|
Hu M, Zhao X, Liu Y, Zhou H, You Y, Xue Z. Complex interplay of gut microbiota between obesity and asthma in children. Front Microbiol 2023; 14:1264356. [PMID: 38029078 PMCID: PMC10655108 DOI: 10.3389/fmicb.2023.1264356] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Obesity is an important risk factor and common comorbidity of childhood asthma. Simultaneously, obesity-related asthma, a distinct asthma phenotype, has attracted significant attention owing to its association with more severe clinical manifestations, poorer disease control, and reduced quality of life. The establishment of the gut microbiota during early life is essential for maintaining metabolic balance and fostering the development of the immune system in children. Microbial dysbiosis influences host lipid metabolism, triggers chronic low-grade inflammation, and affects immune responses. It is intimately linked to the susceptibility to childhood obesity and asthma and plays a potentially crucial transitional role in the progression of obesity-related asthma. This review article summarizes the latest research on the interplay between asthma and obesity, with a particular focus on the mediating role of gut microbiota in the pathogenesis of obesity-related asthma. This study aims to provide valuable insight to enhance our understanding of this condition and offer preliminary evidence to support the development of therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | | | - Yannan You
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zheng Xue
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
9
|
Lam GA, Albarrak H, McColl CJ, Pizarro A, Sanaka H, Gomez-Nguyen A, Cominelli F, Paes Batista da Silva A. The Oral-Gut Axis: Periodontal Diseases and Gastrointestinal Disorders. Inflamm Bowel Dis 2023; 29:1153-1164. [PMID: 36527679 PMCID: PMC10320234 DOI: 10.1093/ibd/izac241] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Indexed: 06/17/2023]
Abstract
One of the prospective sequelae of periodontal disease (PD), chronic inflammation of the oral mucosa, is the development of inflammatory gastrointestinal (GI) disorders due to the amplification and expansion of the oral pathobionts. In addition, chronic inflammatory diseases related to the GI tract, which include inflammatory bowel disease (IBD), can lead to malignancy susceptibility in the colon of both animals and humans. Recent studies suggest that dysbiosis of the oral microbiota can alter the microbial composition in relative abundance or diversity of the distal gut, leading to the progression of digestive carcinogenesis. The link between PD and specific GI disorders is also closely associated with the migration and colonization of periodontal pathogens and the subsequent microbe-reactive T cell induction within the intestines. In this review, an in-depth examination of this relationship and the accessibility of different mouse models of IBD and PD may shed light on the current dogma. As such, oral microbiota dysbiosis involving specific bacteria, including Fusobacterium nucleatum and Porphyromonas gingivalis, can ultimately lead to gut malignancies. Further understanding the precise mechanism(s) of the oral-gut microbial axis in PD, IBD, and colorectal cancer pathogenesis will be pivotal in diagnosis, prognosis, and future treatment.
Collapse
Affiliation(s)
- Gretchen A Lam
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA
- School of Dental Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Hala Albarrak
- School of Dental Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Periodontics, Case Western Reserve University, Cleveland, Ohio, USA
| | | | | | | | - Adrian Gomez-Nguyen
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Fabio Cominelli
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Andre Paes Batista da Silva
- School of Dental Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Periodontics, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
10
|
Filler R, Yeganeh M, Li B, Lee C, Alganabi M, Hock A, Biouss G, Balsamo F, Lee D, Miyake H, Pierro A. Bovine milk-derived exosomes attenuate NLRP3 inflammasome and NF-κB signaling in the lung during neonatal necrotizing enterocolitis. Pediatr Surg Int 2023; 39:211. [PMID: 37268798 DOI: 10.1007/s00383-023-05490-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/13/2023] [Indexed: 06/04/2023]
Abstract
PURPOSE Necrotizing enterocolitis (NEC), an inflammatory intestinal disease common in premature infants, has been associated with the development of lung damage. Toll-like receptor 4 has been shown to regulate inflammation in the NEC lungs, however, other important inflammatory mechanisms have not been thoroughly investigated. In addition, we reported that milk-derived exosomes were able to attenuate intestinal injury and inflammation in experimental NEC. This study aims to (i) investigate the role of the NLRP3 inflammasome and NF-κB pathway in regulating lung damage during experimental NEC; and (ii) evaluate the therapeutic potential of bovine milk exosomes in reducing lung inflammation and injury during NEC. METHODS NEC was induced by gavage feeding of hyperosmolar formula, hypoxia, and lipopolysaccharide administration in neonatal mice from postnatal days 5-9. Exosomes were obtained by ultracentrifugation of bovine milk and administered during each formula feed. RESULTS The lung of NEC pups showed increased inflammation, tissue damage, NLRP3 inflammasome expression, and NF-κB pathway activation, which were attenuated upon exosome administration. CONCLUSION Our findings suggest that the lung undergoes significant inflammation and injury following experimental NEC which are attenuated by bovine milk-derived exosomes. This emphasizes the therapeutic potential of exosomes not just on the intestine but also on the lung.
Collapse
Affiliation(s)
- Rachel Filler
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Mina Yeganeh
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Bo Li
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Carol Lee
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Mashriq Alganabi
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Alison Hock
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - George Biouss
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Felicia Balsamo
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Dorothy Lee
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Hiromu Miyake
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Agostino Pierro
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
11
|
Tian Z, Khan AI, Rehman AU, Deng T, Ma C, Wang L. Virulence factors and mechanisms of paediatric pneumonia caused by Enterococcus faecalis. Gut Pathog 2023; 15:2. [PMID: 36624474 PMCID: PMC9830894 DOI: 10.1186/s13099-022-00522-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 12/09/2022] [Indexed: 01/11/2023] Open
Abstract
Paediatric pneumonia is a respiratory infection that affects infants and young children under the age of 3. This disease is the leading cause of infant and child mortality in developing countries because of the weak immune system of young children. The difficulty and length of time required to identify the pathogen and causative agent are the main reasons for this high mortality rate. In addition, the identification of certain causative agents is particularly important for the treatment of paediatric pneumonia. In this study, we explored the possible mechanisms by which pathogenic Enterococcus faecalis induced pneumonia in vivo. The potential virulence factors of bacteria isolated from the intestines of paediatric pneumonia patients were determined. Taken together, the results suggested that lysophosphatidic acid (LTA) from pathogenic E. faecalis decreases the expression of platelet-activating factor receptor (PAFR), which in turn disrupts the function of intestinal tight junctions (Occ and Ccldn1), leading to the entry of LE-LTA into the bloodstream because of the disruption of the intestinal barrier. Although LTA can enter circulation, it cannot directly infiltrate the lungs, which indicates that lung inflammation in mice is not caused by the direct entry of LE-LTA into the lungs. We further found that LTA activates immune cells, such as CD8 + T cells and type 2 innate lymphocytes, in vivo. Interleukin-6 and interleukin-17 can produce large amounts of inflammatory factors and thus promote the development of pneumonia. In conclusion, our findings demonstrate that the LTA of pathogenic E. faecalis in the intestine is a virulence factor that can cause paediatric pneumonia. This study found that intestinal bacterial virulence factors can induce immune responses in the lungs and blood. These findings could provide further insight into the mechanism of infectious diseases in the lung that are caused by bacteria in the intestine.
Collapse
Affiliation(s)
- Zhiying Tian
- Laboratory of Biochemistry and Molecular Biology, Department of Biotechnology, College of Basic Medicine, Dalian Medical University, Dalian, China
| | - Asif Iqbal Khan
- Laboratory of Biochemistry and Molecular Biology, Department of Biotechnology, College of Basic Medicine, Dalian Medical University, Dalian, China
| | - Ata Ur Rehman
- Laboratory of Biochemistry and Molecular Biology, Department of Biotechnology, College of Basic Medicine, Dalian Medical University, Dalian, China
| | - Ting Deng
- Laboratory of Biochemistry and Molecular Biology, Department of Biotechnology, College of Basic Medicine, Dalian Medical University, Dalian, China
| | - Chao Ma
- Laboratory of Biochemistry and Molecular Biology, Department of Biotechnology, College of Basic Medicine, Dalian Medical University, Dalian, China
| | - Liang Wang
- National Joint Engineering Laboratory, Regenerative Medicine Centre, Stem Cell Clinical Research Centre, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
12
|
Liu G, Xu X. Editorial of the Special Issue "Dietary Fiber and Inflammatory Bowel Disease". Nutrients 2022; 14:nu14224861. [PMID: 36432553 PMCID: PMC9698537 DOI: 10.3390/nu14224861] [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: 10/26/2022] [Accepted: 11/09/2022] [Indexed: 11/19/2022] Open
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a chronic disease of the gastrointestinal (GI) tract; its burden has significantly increased in recent decades, with 6.8 million cases of IBD reported in 2017 according to the Global Burden of Disease study [...].
Collapse
Affiliation(s)
- Gang Liu
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia
- Centre for Inflammation, Centenary Institute, Camperdown, NSW 2050, Australia
- Correspondence:
| | - Xiaoyue Xu
- School of Population Health, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
13
|
Shen S, Gong M, Wang G, Dua K, Xu J, Xu X, Liu G. COVID-19 and Gut Injury. Nutrients 2022; 14:nu14204409. [PMID: 36297092 PMCID: PMC9608818 DOI: 10.3390/nu14204409] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/11/2022] [Accepted: 10/18/2022] [Indexed: 01/28/2023] Open
Abstract
COVID-19 induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is currently a pandemic and it has led to more than 620 million patients with 6.56 million deaths globally. Males are more susceptible to COVID-19 infection and associated with a higher chance to develop severe COVID-19 than females. Aged people are at a high risk of COVID-19 infection, while young children have also increased cases. COVID-19 patients typically develop respiratory system pathologies, however symptoms in the gastrointestinal (GI) tract are also very common. Inflammatory cell recruitments and their secreted cytokines are found in the GI tract in COVID-19 patients. Microbiota changes are the key feature in COVID-19 patients with gut injury. Here, we review all current known mechanisms of COVID-19-induced gut injury, and the most acceptable one is that SARS-CoV-2 binds to angiotensin-converting enzyme 2 (ACE2) receptor on host cells in the GI tract. Interestingly, inflammatory bowel disease (IBD) is an inflammatory disorder, but the patients with IBD do not have the increased risk to develop COVID-19. There is currently no cure for COVID-19, but anti-viruses and monoclonal antibodies reduce viral load and shorten the recovery time of the disease. We summarize current therapeutics that target symptoms in the GI tract, including probiotics, ACE2 inhibitors and nutrients. These are promising therapeutic options for COVID-19-induced gut injury.
Collapse
Affiliation(s)
- Sj Shen
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campus, University of New South Wales, Sydney, NSW 2217, Australia
| | - Muxue Gong
- School of Clinical Medicine, Bengbu Medicine College, Bengbu 233030, China
| | - Gang Wang
- Department of Respiratory and Critical Care Medicine, Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Jincheng Xu
- Stomatology Department, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
- School of Dental Medicine, Bengbu Medical College, Bengbu 233030, China
| | - Xiaoyue Xu
- School of Population Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Gang Liu
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia
- Centre for Inflammation, Centenary Institute, Camperdown, NSW 2050, Australia
- Correspondence:
| |
Collapse
|
14
|
Sazonovs A, Stevens CR, Venkataraman GR, Yuan K, Avila B, Abreu MT, Ahmad T, Allez M, Ananthakrishnan AN, Atzmon G, Baras A, Barrett JC, Barzilai N, Beaugerie L, Beecham A, Bernstein CN, Bitton A, Bokemeyer B, Chan A, Chung D, Cleynen I, Cosnes J, Cutler DJ, Daly A, Damas OM, Datta LW, Dawany N, Devoto M, Dodge S, Ellinghaus E, Fachal L, Farkkila M, Faubion W, Ferreira M, Franchimont D, Gabriel SB, Ge T, Georges M, Gettler K, Giri M, Glaser B, Goerg S, Goyette P, Graham D, Hämäläinen E, Haritunians T, Heap GA, Hiltunen M, Hoeppner M, Horowitz JE, Irving P, Iyer V, Jalas C, Kelsen J, Khalili H, Kirschner BS, Kontula K, Koskela JT, Kugathasan S, Kupcinskas J, Lamb CA, Laudes M, Lévesque C, Levine AP, Lewis JD, Liefferinckx C, Loescher BS, Louis E, Mansfield J, May S, McCauley JL, Mengesha E, Mni M, Moayyedi P, Moran CJ, Newberry RD, O'Charoen S, Okou DT, Oldenburg B, Ostrer H, Palotie A, Paquette J, Pekow J, Peter I, Pierik MJ, Ponsioen CY, Pontikos N, Prescott N, Pulver AE, Rahmouni S, Rice DL, Saavalainen P, Sands B, Sartor RB, Schiff ER, Schreiber S, Schumm LP, Segal AW, Seksik P, Shawky R, et alSazonovs A, Stevens CR, Venkataraman GR, Yuan K, Avila B, Abreu MT, Ahmad T, Allez M, Ananthakrishnan AN, Atzmon G, Baras A, Barrett JC, Barzilai N, Beaugerie L, Beecham A, Bernstein CN, Bitton A, Bokemeyer B, Chan A, Chung D, Cleynen I, Cosnes J, Cutler DJ, Daly A, Damas OM, Datta LW, Dawany N, Devoto M, Dodge S, Ellinghaus E, Fachal L, Farkkila M, Faubion W, Ferreira M, Franchimont D, Gabriel SB, Ge T, Georges M, Gettler K, Giri M, Glaser B, Goerg S, Goyette P, Graham D, Hämäläinen E, Haritunians T, Heap GA, Hiltunen M, Hoeppner M, Horowitz JE, Irving P, Iyer V, Jalas C, Kelsen J, Khalili H, Kirschner BS, Kontula K, Koskela JT, Kugathasan S, Kupcinskas J, Lamb CA, Laudes M, Lévesque C, Levine AP, Lewis JD, Liefferinckx C, Loescher BS, Louis E, Mansfield J, May S, McCauley JL, Mengesha E, Mni M, Moayyedi P, Moran CJ, Newberry RD, O'Charoen S, Okou DT, Oldenburg B, Ostrer H, Palotie A, Paquette J, Pekow J, Peter I, Pierik MJ, Ponsioen CY, Pontikos N, Prescott N, Pulver AE, Rahmouni S, Rice DL, Saavalainen P, Sands B, Sartor RB, Schiff ER, Schreiber S, Schumm LP, Segal AW, Seksik P, Shawky R, Sheikh SZ, Silverberg MS, Simmons A, Skeiceviciene J, Sokol H, Solomonson M, Somineni H, Sun D, Targan S, Turner D, Uhlig HH, van der Meulen AE, Vermeire S, Verstockt S, Voskuil MD, Winter HS, Young J, Duerr RH, Franke A, Brant SR, Cho J, Weersma RK, Parkes M, Xavier RJ, Rivas MA, Rioux JD, McGovern DPB, Huang H, Anderson CA, Daly MJ. Large-scale sequencing identifies multiple genes and rare variants associated with Crohn's disease susceptibility. Nat Genet 2022; 54:1275-1283. [PMID: 36038634 PMCID: PMC9700438 DOI: 10.1038/s41588-022-01156-2] [Show More Authors] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 07/12/2022] [Indexed: 01/18/2023]
Abstract
Genome-wide association studies (GWASs) have identified hundreds of loci associated with Crohn's disease (CD). However, as with all complex diseases, robust identification of the genes dysregulated by noncoding variants typically driving GWAS discoveries has been challenging. Here, to complement GWASs and better define actionable biological targets, we analyzed sequence data from more than 30,000 patients with CD and 80,000 population controls. We directly implicate ten genes in general onset CD for the first time to our knowledge via association to coding variation, four of which lie within established CD GWAS loci. In nine instances, a single coding variant is significantly associated, and in the tenth, ATG4C, we see additionally a significantly increased burden of very rare coding variants in CD cases. In addition to reiterating the central role of innate and adaptive immune cells as well as autophagy in CD pathogenesis, these newly associated genes highlight the emerging role of mesenchymal cells in the development and maintenance of intestinal inflammation.
Collapse
Affiliation(s)
- Aleksejs Sazonovs
- Genomics of Inflammation and Immunity Group, Human Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Christine R Stevens
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - Kai Yuan
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Brandon Avila
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Maria T Abreu
- Crohn's and Colitis Center, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | | | - Matthieu Allez
- Hopital Saint-Louis, APHP, Universite de Paris, INSERM U1160, Paris, France
| | - Ashwin N Ananthakrishnan
- Division of Gastroenterology, Crohn's and Colitis Center, Massachusetts General Hospital, Boston, MA, USA
| | - Gil Atzmon
- Department for Human Biology, University of Haifa, Haifa, Israel
- Departments of Medicine and Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aris Baras
- Regeneron Genetics Center, Tarrytown, NY, USA
| | - Jeffrey C Barrett
- Human Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Nir Barzilai
- Departments of Medicine and Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- The Institute for Aging Research, The Nathan Shock Center of Excellence in the Basic Biology of Aging and the Paul F. Glenn Center for the Biology of Human Aging Research at Albert Einstein College of Medicine of Yeshiva University, Bronx, NY, USA
| | - Laurent Beaugerie
- Gastroenterology Department, Sorbonne Universite, Saint Antoine Hospital, Paris, France
| | - Ashley Beecham
- John P. Hussman Institute for Human Genomics, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
- The Dr. John T. Macdonald Foundation Department of Human Genetics, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | | | - Alain Bitton
- McGill University and McGill University Health Centre, Montreal, Quebec, Canada
| | - Bernd Bokemeyer
- Department of Internal Medicine, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Andrew Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Womens Hospital, Boston, MA, USA
| | | | | | - Jacques Cosnes
- Professeur Chef de Service chez APHP and Universite Paris-6, Paris, France
| | - David J Cutler
- Department of Human Genetics, Emory University, Atlanta, GA, USA
- Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Allan Daly
- Human Genetics Informatics, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Lisa W Datta
- Meyerhoff Inflammatory Bowel Disease Center, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Noor Dawany
- Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Marcella Devoto
- Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
- University of Rome Sapienza, Rome, Italy
- IRGB - CNR, Cagliari, Italy
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Sheila Dodge
- Genomics Platform, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Eva Ellinghaus
- Christian-Albrechts-University of Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Laura Fachal
- Genomics of Inflammation and Immunity Group, Human Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | | | | | | | - Stacey B Gabriel
- Genomics Platform, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Tian Ge
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Precision Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | | | - Kyle Gettler
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mamta Giri
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin Glaser
- Department of Endocrinology and Metabolism, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Philippe Goyette
- Research Center Montreal Heart Institute, Montreal, Quebec, Canada
| | - Daniel Graham
- Infectious Disease and Microbiome Program, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Eija Hämäläinen
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Talin Haritunians
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | | | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Marc Hoeppner
- Christian-Albrechts-University of Kiel, Kiel, Germany
| | | | - Peter Irving
- Department of Gastroenterology, Guys and Saint Thomas Hospital, London, UK
- School of Immunology and Microbial Sciences, Kings College London, London, UK
| | - Vivek Iyer
- Human Genetics Informatics, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Chaim Jalas
- Director of Genetic Resources and Services, Center for Rare Jewish Genetic Disorders, Bonei Olam, Brooklyn, NY, USA
| | - Judith Kelsen
- Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Hamed Khalili
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Barbara S Kirschner
- Department of Gastroenterology, University of Chicago Medicine, Chicago, IL, USA
| | - Kimmo Kontula
- Department of Medicine, Helsinki University Hospital, and Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland
| | - Jukka T Koskela
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Subra Kugathasan
- Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Juozas Kupcinskas
- Department of Gastroenterology and Institute for Digestive Research, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Christopher A Lamb
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Gastroenterology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | - Chloé Lévesque
- Research Center Montreal Heart Institute, Montreal, Quebec, Canada
| | | | - James D Lewis
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
- Crohn's and Colitis Foundation, New York, NY, USA
| | | | - Britt-Sabina Loescher
- Christian-Albrechts-University of Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | | | - John Mansfield
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Gastroenterology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Sandra May
- Christian-Albrechts-University of Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Jacob L McCauley
- John P. Hussman Institute for Human Genomics, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
- The Dr. John T. Macdonald Foundation Department of Human Genetics, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Emebet Mengesha
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Myriam Mni
- University of Liège, ULG, Liège, Belgium
| | | | | | | | | | - David T Okou
- Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
- Institut National de Sante Publique (INSP), Abidjan, Côte d'Ivoire
| | - Bas Oldenburg
- Department of Gastroenterology and Hepatology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Harry Ostrer
- Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aarno Palotie
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Jean Paquette
- Research Center Montreal Heart Institute, Montreal, Quebec, Canada
| | - Joel Pekow
- Department of Gastroenterology, University of Chicago Medicine, Chicago, IL, USA
| | - Inga Peter
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marieke J Pierik
- Department of Gastroenterology and Hepatology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Cyriel Y Ponsioen
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | | | - Natalie Prescott
- Department of Medical and Molecular Genetics, Kings College London, London, UK
| | - Ann E Pulver
- School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | | | - Daniel L Rice
- Genomics of Inflammation and Immunity Group, Human Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Päivi Saavalainen
- Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland
| | - Bruce Sands
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - R Balfour Sartor
- Center for Gastrointestinal Biology and Disease, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | | | - Stefan Schreiber
- Christian-Albrechts-University of Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - L Philip Schumm
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | | | - Philippe Seksik
- Gastroenterology Department, Sorbonne Universite, Saint Antoine Hospital, Paris, France
| | - Rasha Shawky
- IBD BioResource, NIHR BioResource, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Shehzad Z Sheikh
- Center for Gastrointestinal Biology and Disease, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | | | - Alison Simmons
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Jurgita Skeiceviciene
- Department of Gastroenterology and Institute for Digestive Research, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Harry Sokol
- Gastroenterology Department, Sorbonne Universite, Saint Antoine Hospital, Paris, France
| | - Matthew Solomonson
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hari Somineni
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Dylan Sun
- Regeneron Genetics Center, Tarrytown, NY, USA
| | - Stephan Targan
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Dan Turner
- Shaare Zedek Medical Center, Jerusalem, Israel
| | - Holm H Uhlig
- Translational Gastroenterology Unit and Biomedical Research Centre, Nuffield Department of Clinical Medicine, Experimental Medicine Division, University of Oxford, Oxford, UK
- Department of Pediatrics, John Radcliffe Hospital, Oxford, UK
| | - Andrea E van der Meulen
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Séverine Vermeire
- University Hospitals Leuven, Leuven, Belgium
- Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Sare Verstockt
- Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Michiel D Voskuil
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | | | | | | | - Andre Franke
- Christian-Albrechts-University of Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Steven R Brant
- Meyerhoff Inflammatory Bowel Disease Center, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Crohn's Colitis Center of New Jersey, Department of Medicine, Rutgers Robert Wood Johnson Medical School and Department of Genetics and the Human Genetics Institute of New Jersey, Rutgers University, New Brunswick and Piscataway, NJ, USA
| | - Judy Cho
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Miles Parkes
- Department of Gastroenterology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Ramnik J Xavier
- Infectious Disease and Microbiome Program, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Kurt Isselbacher Professor of Medicine at Harvard Medical School, Cambridge, MA, USA
- Core Institute Member, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Klarman Cell Observatory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Immunology Program, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Microbiome Informatics and Therapeutics at MIT, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Manuel A Rivas
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - John D Rioux
- Research Center Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Dermot P B McGovern
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Hailiang Huang
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
| | - Carl A Anderson
- Genomics of Inflammation and Immunity Group, Human Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
| | - Mark J Daly
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
15
|
Schmid F, Chao CM, Däbritz J. Pathophysiological Concepts and Management of Pulmonary Manifestation of Pediatric Inflammatory Bowel Disease. Int J Mol Sci 2022; 23:7287. [PMID: 35806292 PMCID: PMC9266732 DOI: 10.3390/ijms23137287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 02/01/2023] Open
Abstract
Pulmonary manifestation (PM) of inflammatory bowel disease (IBD) in children is a rare condition. The exact pathogenesis is still unclear, but several explanatory concepts were postulated and several case reports in children were published. We performed a systematic Medline search between April 1976 and April 2022. Different pathophysiological concepts were identified, including the shared embryological origin, "miss-homing" of intestinal based neutrophils and T lymphocytes, inflammatory triggering via certain molecules (tripeptide proline-glycine-proline, interleukin 25), genetic factors and alterations in the microbiome. Most pediatric IBD patients with PM are asymptomatic, but can show alterations in pulmonary function tests and breathing tests. In children, the pulmonary parenchyma is more affected than the airways, leading histologically mainly to organizing pneumonia. Medication-associated lung injury has to be considered in pulmonary symptomatic pediatric IBD patients treated with certain agents (i.e., mesalamine, sulfasalazine or infliximab). Furthermore, the risk of pulmonary embolism is generally increased in pediatric IBD patients. The initial treatment of PM is based on corticosteroids, either inhaled for the larger airways or systemic for smaller airways and parenchymal disease. In summary, this review article summarizes the current knowledge about PM in pediatric IBD patients, focusing on pathophysiological and clinical aspects.
Collapse
Affiliation(s)
- Florian Schmid
- Catholic Children’s Hospital Wilhelmstift, 22149 Hamburg, Germany;
| | - Cho-Ming Chao
- Department of Pediatrics, University Medical Center Rostock, 18057 Rostock, Germany;
- Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center of Lung Research (DZL), Justus-Liebig-University, 35398 Giessen, Germany
| | - Jan Däbritz
- Department of Pediatrics, University Medical Center Greifswald, 17475 Greifswald, Germany
| |
Collapse
|
16
|
Melo-González F, Sepúlveda-Alfaro J, Schultz BM, Suazo ID, Boone DL, Kalergis AM, Bueno SM. Distal Consequences of Mucosal Infections in Intestinal and Lung Inflammation. Front Immunol 2022; 13:877533. [PMID: 35572549 PMCID: PMC9095905 DOI: 10.3389/fimmu.2022.877533] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 03/28/2022] [Indexed: 11/17/2022] Open
Abstract
Infectious diseases are one of the leading causes of morbidity and mortality worldwide, affecting high-risk populations such as children and the elderly. Pathogens usually activate local immune responses at the site of infection, resulting in both protective and inflammatory responses, which may lead to local changes in the microbiota, metabolites, and the cytokine environment. Although some pathogens can disseminate and cause systemic disease, increasing evidence suggests that local infections can affect tissues not directly invaded. In particular, diseases occurring at distal mucosal barriers such as the lung and the intestine seem to be linked, as shown by epidemiological studies in humans. These mucosal barriers have bidirectional interactions based mainly on multiple signals derived from the microbiota, which has been termed as the gut-lung axis. However, the effects observed in such distal places are still incompletely understood. Most of the current research focuses on the systemic impact of changes in microbiota and bacterial metabolites during infection, which could further modulate immune responses at distal tissue sites. Here, we describe how the gut microbiota and associated metabolites play key roles in maintaining local homeostasis and preventing enteric infection by direct and indirect mechanisms. Subsequently, we discuss recent murine and human studies linking infectious diseases with changes occurring at distal mucosal barriers, with particular emphasis on bacterial and viral infections affecting the lung and the gastrointestinal tract. Further, we discuss the potential mechanisms by which pathogens may cause such effects, promoting either protection or susceptibility to secondary infection.
Collapse
Affiliation(s)
- Felipe Melo-González
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Javiera Sepúlveda-Alfaro
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bárbara M. Schultz
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Isidora D. Suazo
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - David L. Boone
- Department of Microbiology and Immunology, Indiana University School of Medicine-South Bend, South Bend, IN, United States
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
17
|
Lu Q, Yang MF, Liang YJ, Xu J, Xu HM, Nie YQ, Wang LS, Yao J, Li DF. Immunology of Inflammatory Bowel Disease: Molecular Mechanisms and Therapeutics. J Inflamm Res 2022; 15:1825-1844. [PMID: 35310454 PMCID: PMC8928114 DOI: 10.2147/jir.s353038] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/26/2022] [Indexed: 12/12/2022] Open
Abstract
As a main digestive organ and an important immune organ, the intestine plays a vital role in resisting the invasion of potential pathogens into the body. Intestinal immune dysfunction remains important pathogenesis of inflammatory bowel disease (IBD). In this review, we explained the interactions among symbiotic flora, intestinal epithelial cells, and the immune system, clarified the operating mechanism of the intestinal immune system, and highlighted the immunological pathogenesis of IBD, with a focus on the development of immunotherapy for IBD. In addition, intestinal fibrosis is a significant complication in patients with long-term IBD, and we reviewed the immunological pathogenesis involved in the development of intestinal fibrogenesis and provided novel antifibrotic immunotherapies for IBD.
Collapse
Affiliation(s)
- Quan Lu
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
- Department of Gastroenterology, Shenzhen People’s Hospital (The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, People’s Republic of China
| | - Mei-feng Yang
- Department of Hematology, Yantian District People’s Hospital, Shenzhen, Guangdong, People’s Republic of China
| | - Yu-jie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen, Guangdong, People’s Republic of China
| | - Jing Xu
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital (School of Medicine of South China University of Technology), Guangzhou, Guangdong, People’s Republic of China
| | - Hao-ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital (School of Medicine of South China University of Technology), Guangzhou, Guangdong, People’s Republic of China
| | - Yu-qiang Nie
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital (School of Medicine of South China University of Technology), Guangzhou, Guangdong, People’s Republic of China
| | - Li-sheng Wang
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
- Department of Gastroenterology, Shenzhen People’s Hospital (The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, People’s Republic of China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
- Department of Gastroenterology, Shenzhen People’s Hospital (The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, People’s Republic of China
| | - De-feng Li
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
- Department of Gastroenterology, Shenzhen People’s Hospital (The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
18
|
Li Y, Li N, Liu J, Wang T, Dong R, Ge D, Peng G. Gegen Qinlian Decoction Alleviates Experimental Colitis and Concurrent Lung Inflammation by Inhibiting the Recruitment of Inflammatory Myeloid Cells and Restoring Microbial Balance. J Inflamm Res 2022; 15:1273-1291. [PMID: 35237061 PMCID: PMC8884713 DOI: 10.2147/jir.s352706] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/04/2022] [Indexed: 12/12/2022] Open
Abstract
Objective Ulcerative colitis (UC) as one of the intractable diseases in gastroenterology seriously threatens human health. Respiratory pathology is a representative extraintestinal manifestation of UC affecting the quality of life of patients. Gegen Qinlian Decoction (GQD) is a classical traditional Chinese medicine prescription for UC or acute lung injury. This study was aimed to reveal the therapeutic effect of GQD on UC and its pulmonary complications and uncover its molecular mechanism mediated by myeloid cells and microbiota. Methods Mice with DSS-induced colitis were orally administrated with GQD. Overall vital signs were assessed by body weight loss and disease activity index (DAI). Pulmonary general signs were evaluated by pulmonary pathology and lung function. The mechanism of GQD relieving UC was characterized by detecting myeloid cells (neutrophils, macrophages, inflammatory monocytes, and resident monocytes) in colonic and lung tissues, related inflammatory cytokines, as well as the microbiota in bronchoalveolar lavage fluid (BALF) and feces. Results GQD significantly reduced weight loss, DAI scores, and lung injury but improved the lung function of colitis mice. The DSS-induced colonic and concurrent pulmonary inflammation were also alleviated by GQD, as indicated by the down-regulated expressions of inflammatory cytokines (TNF-α, IL-1β, IL-6, CCR2, and CCL2) and the suppressed recruitment of neutrophils and inflammatory monocytes. Meanwhile, GQD greatly improved intestinal microbiota imbalance by enriching Ruminococcaceae UCG-013 while decreasing Parabacteroides, [Eubacterium]_fissicatena_group, and Akkermansia in the feces of colitis mice. Expectantly, GQD also restored lung microbiota imbalance by clearing excessive Coprococcus 2 and Ochrobactrum in the BALF of colitis mice. Finally, significant correlations appeared between GQD-mediated specific bacteria and inflammatory cytokines or immune cells. Conclusion GQD could alleviate UC by decreasing excessive inflammatory myeloid cells and cytokines, and reshaping the microbiota between the colon and lung, which contributes to clarifying the mechanism by which GQD ameliorates colitis-associated pulmonary inflammation.
Collapse
Affiliation(s)
- Yalan Li
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Na Li
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Jiajing Liu
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Tieshan Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Ruijuan Dong
- Experimental Teaching Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Dongyu Ge
- Experimental Teaching Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Guiying Peng
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
- Correspondence: Guiying Peng, Email
| |
Collapse
|
19
|
Zhang Y, Yang W, Li W, Zhao Y. NLRP3 Inflammasome: Checkpoint Connecting Innate and Adaptive Immunity in Autoimmune Diseases. Front Immunol 2021; 12:732933. [PMID: 34707607 PMCID: PMC8542789 DOI: 10.3389/fimmu.2021.732933] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/20/2021] [Indexed: 12/12/2022] Open
Abstract
Autoimmune diseases are a broad spectrum of human diseases that are characterized by the breakdown of immune tolerance and the production of autoantibodies. Recently, dysfunction of innate and adaptive immunity is considered to be a key step in the initiation and maintenance of autoimmune diseases. NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome is a multimeric protein complex, which can detect exogenous pathogen irritants and endogenous danger signals. The main function of NLRP3 inflammasome is to promote secretion of interleukin (IL)-1β and IL-18, and pyroptosis mediated by caspase-1. Served as a checkpoint in innate and adaptive immunity, aberrant activation and regulation of NLRP3 inflammasome plays an important role in the pathogenesis of autoimmune diseases. This paper reviewed the roles of NLRP3 inflammasome in autoimmune diseases, which shows NLRP3 inflammasome may be a potential target for autoimmune diseases deserved further study.
Collapse
Affiliation(s)
- Yiwen Zhang
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenlin Yang
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wangen Li
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yunjuan Zhao
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
20
|
PAF Receptor Inhibition Attenuates Neuronal Pyroptosis in Cerebral Ischemia/Reperfusion Injury. Mol Neurobiol 2021; 58:6520-6539. [PMID: 34562185 DOI: 10.1007/s12035-021-02537-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023]
Abstract
Ischemic stroke is an inflammation-related disease, during which process activation of NLRP3 inflammasome and subsequent pyroptosis play crucial roles. Platelet-activating factor (PAF) is a potent phospholipid regulator of inflammation which exerts its effect via binding specific PAF receptor (PAFR). However, whether PAFR contributes to pyroptosis during ischemia/reperfusion (I/R) injury remains to be elucidated. To explore the underlying effect of PAFR on ischemic stroke from the perspective of pyroptosis, mice were subjected to middle cerebral artery occlusion/reperfusion (MCAO/R) injury and primary cultures of mice cerebral cortical neurons were exposed to oxygen-glucose deprivation/reoxygenation (OGD/R) injury to mimic I/R in vivo and in vitro, after which indexes associated with pyroptosis were analyzed. Intriguingly, our results indicated that inhibition of PAFR with its inhibitor XQ-1H or PAFR siRNA exerted a neuroprotective effect against I/R injury both in vivo and in vitro. Furthermore, inflammasome activation and pyroptosis after ischemic challenge were attenuated by XQ-1H or PAFR siRNA. Besides, the protection of XQ-1H was abolished by PAF stimulaiton to some extent. Moreover, XQ-1H or PAFR siRNA alleviated the neuronal pyroptosis induced by LPS and nigericin (an NLRP3 activator) in cortical neurons. Taken together, this study firstly demonstrates that PAFR is involved in neuronal pyroptosis after I/R injury, and XQ-1H, a specific PAFR inhibitor, has a promising prospect in attenuating I/R injury from the perspective of anti-pyroptosis.
Collapse
|
21
|
Wen Y, Xiao H, Liu Y, Yang Y, Wang Y, Xu S, Huang S, Hou S, Liang J. Polysaccharides from Dendrobium officinale ameliorate colitis-induced lung injury via inhibiting inflammation and oxidative stress. Chem Biol Interact 2021; 347:109615. [PMID: 34363819 DOI: 10.1016/j.cbi.2021.109615] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/20/2021] [Accepted: 08/04/2021] [Indexed: 01/13/2023]
Abstract
It has been reported that Dendrobium officinale polysaccharides (DOPS) could alleviate colitis in animal model and suppress the activation of NLRP3 inflammasome and β-arrestin1 in vitro. However, it remains unclear whether DOPS has effect on protecting against colitis-induced pulmonary injury. The purpose of this study was to explore the protective effect and mechanism of DOPS on colitis-induced lung injury. A dextran sodium sulfate (DSS)-induced mice colitis model and lipopolysaccharide (LPS)-stimulated BEAS-2B cells model were applied in this study. The results showed that DOPS treatment restored histopathological changes, reduced inflammatory cells infiltration, pro-inflammatory cytokines levels, reactive oxygen species (ROS) formation and MDA generation, and increased anti-oxidative enzymes activities including SOD and GSH-Px in colitis mice. Further investigation showed that DOPS significantly inhibited the protein expression of TLR4, and apparently up-regulated proteins expressions of nuclear-Nrf2, HO-1 and NQO-1 in lung tissues of colitis mice and in BEAS-2B cells. These results indicated that DOPS significantly inhibited inflammation and oxidative stress to alleviate colitis-induced secondary lung injury, and its mechanisms are closely related to the inhibition of TLR4 signaling pathway and the activation of Nrf2 signaling pathway. DOPS may be a promising drug for alleviating colitis-induced lung injury.
Collapse
Affiliation(s)
- Yifan Wen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China
| | - Hongyu Xiao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China
| | - Ying Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China
| | - Yiqi Yang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, PR China
| | - Yumin Wang
- Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, School of Chemistry, South China Normal University, Guangzhou, Guangdong, 510006, PR China
| | - Shijie Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China
| | - Song Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China.
| | - Shaozhen Hou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China.
| | - Jian Liang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China.
| |
Collapse
|
22
|
Shang Q, Zhou X, Yang MR, Lu JG, Pan Y, Zhu GY, Jiang ZH. Amide Derivatives of Ginkgolide B and Their Inhibitory Effects on PAF-Induced Platelet Aggregation. ACS OMEGA 2021; 6:22497-22503. [PMID: 34514222 PMCID: PMC8427636 DOI: 10.1021/acsomega.1c01682] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 08/10/2021] [Indexed: 06/13/2023]
Abstract
Ginkgolides are the most important components of Ginkgo biloba extracts, whose lactone can be hydrolyzed in the aqueous environment. Although the hydrolyzed products have complex structures and their functions are not well-understood, opening the lactone ring is an important strategy in producing novel derivatives of ginkgolide. The preparation of a single pure aminolyzed ginkgolide for the study of its bioactivity and understanding of the process of aminolysis are challenging. To obtain stable aminolyzed products, four amide derivatives (2-5) of ginkgolide B (GB, 1) were prepared via the ring-opening reaction of its lactone with propylamine. These products were purified and fully identified by high-resolution mass spectrometry (HRMS) and nuclear magnetic resonance (NMR) spectroscopy and were further evaluated for their ability to inhibit the PAF-induced platelet aggregation of rabbit platelets in vitro. Compound 2, which was obtained by selective aminolysis of the lactone ring C of GB, showed a much better inhibitory activity of platelet aggregation (IC50, 15 nM) than the parent compound GB (IC50, 442 nM). The other three products (3-5), which were obtained by the aminolysis of lactone rings C and F of GB, did not show platelet aggregation inhibitory activity. The results greatly extended our understanding of the chemistry of GB and provided important structural information for the exploration and development of new drugs based on ginkgolides in G. biloba.
Collapse
Affiliation(s)
- Qiang Shang
- State
Key Laboratory of Quality Research in Chinese Medicine, Guangdong−Hong
Kong−Macao Joint Laboratory of Respiratory Infectious Disease,
Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau 999078, People’s Republic of China
- Livzon
Pharmaceutical Group Inc., Zhuhai 519000, People’s Republic
of China
| | - Xiaobo Zhou
- State
Key Laboratory of Quality Research in Chinese Medicine, Guangdong−Hong
Kong−Macao Joint Laboratory of Respiratory Infectious Disease,
Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau 999078, People’s Republic of China
| | - Ming-Rong Yang
- State
Key Laboratory of Quality Research in Chinese Medicine, Guangdong−Hong
Kong−Macao Joint Laboratory of Respiratory Infectious Disease,
Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau 999078, People’s Republic of China
| | - Jing-Guang Lu
- State
Key Laboratory of Quality Research in Chinese Medicine, Guangdong−Hong
Kong−Macao Joint Laboratory of Respiratory Infectious Disease,
Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau 999078, People’s Republic of China
| | - Yu Pan
- State
Key Laboratory of Quality Research in Chinese Medicine, Guangdong−Hong
Kong−Macao Joint Laboratory of Respiratory Infectious Disease,
Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau 999078, People’s Republic of China
| | - Guo-Yuan Zhu
- State
Key Laboratory of Quality Research in Chinese Medicine, Guangdong−Hong
Kong−Macao Joint Laboratory of Respiratory Infectious Disease,
Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau 999078, People’s Republic of China
| | - Zhi-Hong Jiang
- State
Key Laboratory of Quality Research in Chinese Medicine, Guangdong−Hong
Kong−Macao Joint Laboratory of Respiratory Infectious Disease,
Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau 999078, People’s Republic of China
| |
Collapse
|
23
|
Xue Q, Wen D, Ji MH, Tong J, Yang JJ, Zhou CM. Developing Machine Learning Algorithms to Predict Pulmonary Complications After Emergency Gastrointestinal Surgery. Front Med (Lausanne) 2021; 8:655686. [PMID: 34409047 PMCID: PMC8365303 DOI: 10.3389/fmed.2021.655686] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022] Open
Abstract
Objective: Investigate whether machine learning can predict pulmonary complications (PPCs) after emergency gastrointestinal surgery in patients with acute diffuse peritonitis. Methods: This is a secondary data analysis study. We use five machine learning algorithms (Logistic regression, DecisionTree, GradientBoosting, Xgbc, and gbm) to predict postoperative pulmonary complications. Results: Nine hundred and twenty-six cases were included in this study; 187 cases (20.19%) had PPCs. The five most important variables for the postoperative weight were preoperative albumin, cholesterol on the 3rd day after surgery, albumin on the day of surgery, platelet count on the 1st day after surgery and cholesterol count on the 1st day after surgery for pulmonary complications. In the test group: the logistic regression model shows AUC = 0.808, accuracy = 0.824 and precision = 0.621; Decision tree shows AUC = 0.702, accuracy = 0.795 and precision = 0.486; The GradientBoosting model shows AUC = 0.788, accuracy = 0.827 and precision = 1.000; The Xgbc model shows AUC = 0.784, accuracy = 0.806 and precision = 0.583. The Gbm model shows AUC = 0.814, accuracy = 0.806 and precision = 0.750. Conclusion: Machine learning algorithms can predict patients' PPCs with acute diffuse peritonitis. Moreover, the results of the importance matrix for the Gbdt algorithm model show that albumin, cholesterol, age, and platelets are the main variables that account for the highest pulmonary complication weights.
Collapse
Affiliation(s)
- Qiong Xue
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Duan Wen
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mu-Huo Ji
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Anesthesiology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Jianhua Tong
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Cheng-Mao Zhou
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
24
|
Walters EH, Shukla SD, Mahmood MQ, Ward C. Fully integrating pathophysiological insights in COPD: an updated working disease model to broaden therapeutic vision. Eur Respir Rev 2021; 30:200364. [PMID: 34039673 PMCID: PMC9488955 DOI: 10.1183/16000617.0364-2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/18/2021] [Indexed: 12/17/2022] Open
Abstract
Our starting point is that relatively new findings into the pathogenesis and pathophysiology of airway disease in smokers that lead to chronic obstructive pulmonary disease (COPD) need to be reassessed as a whole and integrated into "mainstream" thinking along with traditional concepts which have stood the test of time. Such a refining of the accepted disease paradigm is urgently needed as thinking on therapeutic targets is currently under active reconsideration. We feel that generalised airway wall "inflammation" is unduly over-emphasised, and highlight the patchy and variable nature of the pathology (with the core being airway remodelling). In addition, we present evidence for airway wall disease in smokers/COPD as including a hypocellular, hypovascular, destructive, fibrotic pathology, with a likely spectrum of epithelial-mesenchymal transition states as significant drivers of this remodelling. Furthermore, we present data from a number of research modalities and integrate this with the aetiology of lung cancer, the role of chronic airway luminal colonisation/infection by a specific group of "respiratory" bacteria in smokers (which results in luminal inflammation) and the central role for oxidative stress on the epithelium. We suggest translation of these insights into more focus on asymptomatic smokers and early COPD, with the potential for fresh preventive and therapeutic approaches.
Collapse
Affiliation(s)
- E Haydn Walters
- School of Medicine and Menzies Institute, University of Tasmania, Hobart, Australia
| | - Shakti D Shukla
- Priority Research Centre for Healthy Lungs and School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, Australia
- Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Malik Q Mahmood
- School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Australia
| | - Chris Ward
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University Medical School, Newcastle University, UK
| |
Collapse
|
25
|
Zhang G, Chen H, Guo Y, Zhang W, Jiang Q, Zhang S, Han L, Chen S, Xue R. Activation of Platelet NLRP3 Inflammasome in Crohn's Disease. Front Pharmacol 2021; 12:705325. [PMID: 34262463 PMCID: PMC8273542 DOI: 10.3389/fphar.2021.705325] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/11/2021] [Indexed: 01/04/2023] Open
Abstract
Patients with Crohn's disease (CD) are inclined to have platelet hyperactivity and an increased risk of intestinal micro-thrombosis. However, the mechanisms underlying platelet hyperactivity in CD are not well understood. We investigated the assembly of platelet NLRP3 inflammasome in patients with active CD and its correlation with platelet hyperactivity. In this study, Real-time PCR and western blotting analyses uncovered that ASC, NLRP3, and active caspase-1 were significantly upregulated in platelets from patients with active CD compared with healthy subjects. As revealed by flow cytometry (FCM) and ELISA analyses, the levels of interleukin-1β in both serum and isolated platelets were elevated in patients with active CD. Co-immunoprecipitation and immunofluorescence experiments revealed an increased assembly of NLRP3 inflammasome in platelets from patients with active CD. In addition, higher levels of intracellular reactive oxygen species (ROS) were observed in these platelets by FCM. Furthermore, elevated levels of platelet P-selectin exposure and fibrinogen binding were demonstrated in patients with active CD by FCM. They were positively correlated with the protein levels of NLRP3 inflammasome components. Collectively, our results indicate that the ROS-NLRP3 inflammasome-interleukin-1β axis may contribute to platelet hyperactivity in active CD.
Collapse
Affiliation(s)
- Ge Zhang
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - He Chen
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yifan Guo
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Zhang
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qiuyu Jiang
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Si Zhang
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Liping Han
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - She Chen
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ruyi Xue
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Wilson NE, Reaves BJ, Wolstenholme AJ. Lack of detectable short-term effects of a single dose of ivermectin on the human immune system. Parasit Vectors 2021; 14:304. [PMID: 34090504 PMCID: PMC8179708 DOI: 10.1186/s13071-021-04810-6] [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: 04/08/2021] [Accepted: 05/25/2021] [Indexed: 12/03/2022] Open
Abstract
Background Ivermectin is widely used in human and animal medicine to treat and prevent parasite nematode infections. It has been suggested that its mode of action requires the host immune system, as it is difficult to reproduce its clinical efficacy in vitro. We therefore studied the effects of a single dose of ivermectin (Stromectol®—0.15 mg/kg) on cytokine levels and immune cell gene expression in human volunteers. This dose reduces bloodstream microfilariae rapidly and for several months when given in mass drug administration programmes. Methods Healthy volunteers with no travel history to endemic regions were given 3–4 tablets, depending on their weight, of either ivermectin or a placebo. Blood samples were drawn immediately prior to administration, 4 h and 24 h afterwards, and complete blood counts performed. Serum levels of 41 cytokines and chemokines were measured using Luminex® and expression levels of 770 myeloid-cell-related genes determined using the NanoString nCounter®. Cytokine levels at 4 h and 24 h post-treatment were compared to the levels pre-treatment using simple t tests to determine if any individual results required further investigation, taking p = < 0.05 as the level of significance. NanoString data were analysed on the proprietary software, nSolver™. Results No significant differences were observed in complete blood counts or cytokine levels at either time point between people given ivermectin versus placebo. Only three genes showed a significant change in expression in peripheral blood mononuclear cells 4 h after ivermectin was given; there were no significant changes 24 h after drug administration or in polymorphonuclear cells at either time point. Leukocytes isolated from those participants given ivermectin showed no difference in their ability to kill Brugia malayi microfilariae in vitro. Conclusions Overall, our data do not support a direct effect of ivermectin, when given at the dose used in current filarial elimination programmes, on the human immune system. Trial registration ClinicalTrials.gov NCT03459794 Registered 9th March 2018, Retrospectively registered https://clinicaltrials.gov/ct2/show/NCT03459794?term=NCT03459794&draw=2&rank=1. Graphic abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13071-021-04810-6.
Collapse
Affiliation(s)
- Natalie E Wilson
- Department of Infectious Diseases, University of Georgia, Athens, GA, 30602, USA.,Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, 30602, USA
| | - Barbara J Reaves
- Department of Infectious Diseases, University of Georgia, Athens, GA, 30602, USA.,Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, 30602, USA
| | - Adrian J Wolstenholme
- Department of Infectious Diseases, University of Georgia, Athens, GA, 30602, USA. .,Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, 30602, USA. .,INRAE Centre Val du Loire, 37380, Nouzilly, France.
| |
Collapse
|
27
|
Biological functions of NLRP3 inflammasome: A therapeutic target in inflammatory bowel disease. Cytokine Growth Factor Rev 2021; 60:61-75. [PMID: 33773897 DOI: 10.1016/j.cytogfr.2021.03.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 03/10/2021] [Indexed: 12/15/2022]
Abstract
Cases of inflammatory bowel disease (IBD), a debilitating intestinal disorder with complex pathological mechanisms, have been increasing in recent years, straining the capacity of healthcare systems. Thus, novel therapeutic targets and innovative agents must be developed. Notably, the NLRP3 inflammasome is upregulated in patients with IBD and/or in animal experimental models. As an innate immune supramolecular assembly, the NLRP3 inflammasome is persistently activated during the pathogenesis of IBD by multiple stimuli. Moreover, this protein complex regulates pro-inflammatory cytokines. Thus, targeting this multiprotein oligomer may offer a feasible way to relieve IBD symptoms and improve clinical outcomes. The mechanisms by which the NLRP3 inflammasome is activated, its role in IBD pathogenesis, and the drugs administered to target this protein complex are reviewed herein. This review establishes that the use of inflammasome-targeting drugs are effective for IBD treatment. Moreover, this review suggests that the value and potential of naturally sourced or derived medicines for IBD treatment must be recognized and appreciated.
Collapse
|
28
|
Lannes-Costa PS, de Oliveira JSS, da Silva Santos G, Nagao PE. A current review of pathogenicity determinants of Streptococcus sp. J Appl Microbiol 2021; 131:1600-1620. [PMID: 33772968 DOI: 10.1111/jam.15090] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/16/2021] [Accepted: 03/24/2021] [Indexed: 12/16/2022]
Abstract
The genus Streptococcus comprises important pathogens, many of them are part of the human or animal microbiota. Advances in molecular genetics, taxonomic approaches and phylogenomic studies have led to the establishment of at least 100 species that have a severe impact on human health and are responsible for substantial economic losses to agriculture. The infectivity of the pathogens is linked to cell-surface components and/or secreted virulence factors. Bacteria have evolved sophisticated and multifaceted adaptation strategies to the host environment, including biofilm formation, survival within professional phagocytes, escape the host immune response, amongst others. This review focuses on virulence mechanism and zoonotic potential of Streptococcus species from pyogenic (S. agalactiae, S. pyogenes) and mitis groups (S. pneumoniae).
Collapse
Affiliation(s)
- P S Lannes-Costa
- Laboratory of Molecular Biology and Physiology of Streptococci, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University (UERJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - J S S de Oliveira
- Laboratory of Molecular Biology and Physiology of Streptococci, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University (UERJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - G da Silva Santos
- Laboratory of Molecular Biology and Physiology of Streptococci, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University (UERJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - P E Nagao
- Laboratory of Molecular Biology and Physiology of Streptococci, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University (UERJ), Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
29
|
Liu G, Philp AM, Corte T, Travis MA, Schilter H, Hansbro NG, Burns CJ, Eapen MS, Sohal SS, Burgess JK, Hansbro PM. Therapeutic targets in lung tissue remodelling and fibrosis. Pharmacol Ther 2021; 225:107839. [PMID: 33774068 DOI: 10.1016/j.pharmthera.2021.107839] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/03/2021] [Indexed: 02/07/2023]
Abstract
Structural changes involving tissue remodelling and fibrosis are major features of many pulmonary diseases, including asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). Abnormal deposition of extracellular matrix (ECM) proteins is a key factor in the development of tissue remodelling that results in symptoms and impaired lung function in these diseases. Tissue remodelling in the lungs is complex and differs between compartments. Some pathways are common but tissue remodelling around the airways and in the parenchyma have different morphologies. Hence it is critical to evaluate both common fibrotic pathways and those that are specific to different compartments; thereby expanding the understanding of the pathogenesis of fibrosis and remodelling in the airways and parenchyma in asthma, COPD and IPF with a view to developing therapeutic strategies for each. Here we review the current understanding of remodelling features and underlying mechanisms in these major respiratory diseases. The differences and similarities of remodelling are used to highlight potential common therapeutic targets and strategies. One central pathway in remodelling processes involves transforming growth factor (TGF)-β induced fibroblast activation and myofibroblast differentiation that increases ECM production. The current treatments and clinical trials targeting remodelling are described, as well as potential future directions. These endeavours are indicative of the renewed effort and optimism for drug discovery targeting tissue remodelling and fibrosis.
Collapse
Affiliation(s)
- Gang Liu
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Ashleigh M Philp
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia; St Vincent's Medical School, UNSW Medicine, UNSW, Sydney, NSW, Australia
| | - Tamera Corte
- Royal Prince Alfred Hospital, Camperdown, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Mark A Travis
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre and Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom
| | - Heidi Schilter
- Pharmaxis Ltd, 20 Rodborough Road, Frenchs Forest, Sydney, NSW, Australia
| | - Nicole G Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Chris J Burns
- Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Mathew S Eapen
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Sukhwinder S Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Department of Pathology and Medical Biology, Groningen, The Netherlands; Woolcock Institute of Medical Research, Discipline of Pharmacology, The University of Sydney, Sydney, NSW, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia.
| |
Collapse
|
30
|
Alemao CA, Budden KF, Gomez HM, Rehman SF, Marshall JE, Shukla SD, Donovan C, Forster SC, Yang IA, Keely S, Mann ER, El Omar EM, Belz GT, Hansbro PM. Impact of diet and the bacterial microbiome on the mucous barrier and immune disorders. Allergy 2021; 76:714-734. [PMID: 32762040 DOI: 10.1111/all.14548] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/10/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022]
Abstract
The prevalence of chronic immune and metabolic disorders is increasing rapidly. In particular, inflammatory bowel diseases, obesity, diabetes, asthma and chronic obstructive pulmonary disease have become major healthcare and economic burdens worldwide. Recent advances in microbiome research have led to significant discoveries of associative links between alterations in the microbiome and health, as well as these chronic supposedly noncommunicable, immune/metabolic disorders. Importantly, the interplay between diet, microbiome and the mucous barrier in these diseases has gained significant attention. Diet modulates the mucous barrier via alterations in gut microbiota, resulting in either disease onset/exacerbation due to a "poor" diet or protection against disease with a "healthy" diet. In addition, many mucosa-associated disorders possess a specific gut microbiome fingerprint associated with the composition of the mucous barrier, which is further influenced by host-microbiome and inter-microbial interactions, dietary choices, microbe immigration and antimicrobials. Our review focuses on the interactions of diet (macronutrients and micronutrients), gut microbiota and mucous barriers (gastrointestinal and respiratory tract) and their importance in the onset and/or progression of major immune/metabolic disorders. We also highlight the key mechanisms that could be targeted therapeutically to prevent and/or treat these disorders.
Collapse
Affiliation(s)
- Charlotte A. Alemao
- Priority Research Centre for Healthy Lungs Hunter Medical Research Institute New Lambton, Newcastle NSW Australia
- The University of Newcastle Newcastle NSW Australia
| | - Kurtis F. Budden
- Priority Research Centre for Healthy Lungs Hunter Medical Research Institute New Lambton, Newcastle NSW Australia
- The University of Newcastle Newcastle NSW Australia
| | - Henry M. Gomez
- Priority Research Centre for Healthy Lungs Hunter Medical Research Institute New Lambton, Newcastle NSW Australia
- The University of Newcastle Newcastle NSW Australia
| | - Saima F. Rehman
- Priority Research Centre for Healthy Lungs Hunter Medical Research Institute New Lambton, Newcastle NSW Australia
- The University of Newcastle Newcastle NSW Australia
| | - Jacqueline E. Marshall
- Faculty of Science Centre for Inflammation Centenary Institute University of Technology Sydney Sydney NSW Australia
| | - Shakti D. Shukla
- Priority Research Centre for Healthy Lungs Hunter Medical Research Institute New Lambton, Newcastle NSW Australia
- The University of Newcastle Newcastle NSW Australia
| | - Chantal Donovan
- Faculty of Science Centre for Inflammation Centenary Institute University of Technology Sydney Sydney NSW Australia
| | - Samuel C. Forster
- Department of Molecular and Translational Sciences Hudson Institute of Medical Research Centre for Innate Immunity and Infectious Diseases Monash University Clayton VIC Australia
| | - Ian A. Yang
- Thoracic Program The Prince Charles Hospital Metro North Hospital and Health Service Brisbane QLD Australia
- Faculty of Medicine UQ Thoracic Research Centre The University of Queensland Brisbane QLD Australia
| | - Simon Keely
- Hunter Medical Research Institute Priority Research Centre for Digestive Health and Neurogastroenterology University of Newcastle New Lambton Heights NSW Australia
| | - Elizabeth R. Mann
- Lydia Becker Institute of Immunology and Inflammation University of Manchester Manchester UK
- Faculty of Biology Medicine and Health Manchester Collaborative Centre for Inflammation Research Manchester Academic Health Science Centre University of Manchester Manchester UK
| | - Emad M. El Omar
- St George & Sutherland Clinical School Microbiome Research Centre University of New South Wales Sydney NSW Australia
| | - Gabrielle T. Belz
- Diamantina Institute University of Queensland Woolloongabba QLD Australia
- Department of Medical Biology Walter and Eliza Hall Institute of Medical Research University of Melbourne Parkville VIC Australia
| | - Philip M. Hansbro
- Priority Research Centre for Healthy Lungs Hunter Medical Research Institute New Lambton, Newcastle NSW Australia
- The University of Newcastle Newcastle NSW Australia
- Faculty of Science Centre for Inflammation Centenary Institute University of Technology Sydney Sydney NSW Australia
| |
Collapse
|
31
|
Ma Y, Yang X, Chatterjee V, Wu MH, Yuan SY. The Gut-Lung Axis in Systemic Inflammation. Role of Mesenteric Lymph as a Conduit. Am J Respir Cell Mol Biol 2021; 64:19-28. [PMID: 32877613 DOI: 10.1165/rcmb.2020-0196tr] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Emerging evidence shows that after injury or infection, the mesenteric lymph acts as a conduit for gut-derived toxic factors to enter the blood circulation, causing systemic inflammation and acute lung injury. Neither the cellular and molecular identity of lymph factors nor their mechanisms of action have been well understood and thus have become a timely topic of investigation. This review will first provide a summary of background knowledge on gut barrier and mesenteric lymphatics, followed by a discussion focusing on the current understanding of potential injurious factors in the lymph and their mechanistic contributions to lung injury. We also examine lymph factors with antiinflammatory properties as well as the bidirectional nature of the gut-lung axis in inflammation.
Collapse
Affiliation(s)
- Yonggang Ma
- Department of Molecular Pharmacology and Physiology, and
| | - Xiaoyuan Yang
- Department of Molecular Pharmacology and Physiology, and
| | | | - Mack H Wu
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Sarah Y Yuan
- Department of Molecular Pharmacology and Physiology, and.,Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, Florida
| |
Collapse
|
32
|
Anka Idrissi D, Senhaji N, Aouiss A, Khalki L, Tijani Y, Zaid N, Marhoume FZ, Naya A, Oudghiri M, Kabine M, Zaid Y. IL-1 and CD40/CD40L platelet complex: elements of induction of Crohn's disease and new therapeutic targets. Arch Pharm Res 2021; 44:117-132. [PMID: 33394309 DOI: 10.1007/s12272-020-01296-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 11/21/2020] [Indexed: 12/15/2022]
Abstract
Ulcerative colitis (UC) and Crohn's disease (CD) are chronic and multifactorial diseases that affect the intestinal tract, both characterized by recurrent inflammation of the intestinal mucosa, resulting in abdominal pain, diarrhea, vomiting and, rectal bleeding. Inflammatory bowel diseases (IBD) regroup these two disorders. The exact pathological mechanism of IBD remains ambiguous and poorly known. In genetically predisposed patients, defects in intestinal mucosal barrier are due to an uncontrolled inflammatory response to normal flora. In addition to the genetic predisposition, these defects could be triggered by environmental factors or by a specific lifestyle which is widely accepted as etiological hypothesis. The involvement of the CD40/CD40L platelet complex in the development of IBD has been overwhelmingly demonstrated. CD40L is climacteric in cell signalling in innate and adaptive immunity, the CD40L expression on the platelet cell surface gives them an immunological competence. The IL-1, a major inflammation mediator could be involved in different ways in the development of IBD. Here, we provide a comprehensive review regarding the role of platelet CD40/CD40L in the pathophysiological effect of IL-1 in the development of Crohn's disease (CD). This review could potentially help future approaches aiming to target these two pathways for therapeutic purposes and elucidate the immunological mechanisms driving gut inflammation.
Collapse
Affiliation(s)
- Doha Anka Idrissi
- Department of Biology, Faculty of Sciences, Hassan II University, Casablanca, Morocco
| | - Nezha Senhaji
- Laboratory of Genetic and Molecular Pathology, Faculty of Medicine, Hassan II University, Casablanca, Morocco
| | - Asmae Aouiss
- Department of Biology, Faculty of Sciences, Hassan II University, Casablanca, Morocco
| | - Loubna Khalki
- Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Youssef Tijani
- Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Nabil Zaid
- Faculty of Sciences, Department of Biology, Mohammed V University, Rabat, Morocco
| | - Fatima Zahra Marhoume
- Faculty of Sciences and Technology, Laboratory of Biochemistry and Neuroscience, Integrative and Computational Neuroscience Team, Hassan First University, Settat, Morocco
| | - Abdallah Naya
- Department of Biology, Faculty of Sciences, Hassan II University, Casablanca, Morocco
| | - Mounia Oudghiri
- Department of Biology, Faculty of Sciences, Hassan II University, Casablanca, Morocco
| | - Mostafa Kabine
- Department of Biology, Faculty of Sciences, Hassan II University, Casablanca, Morocco
| | - Younes Zaid
- Faculty of Sciences, Department of Biology, Mohammed V University, Rabat, Morocco. .,Research Center of Abulcasis, University of Health Sciences, Rabat, Morocco.
| |
Collapse
|
33
|
Wang J, Fan K, He C, Wang Q, Zhang Q, Huang W. A novel Danshensu/tetramethylpyrazine protects against Myocardial Ischemia Reperfusion Injury in rats. Int J Med Sci 2021; 18:2716-2724. [PMID: 34104104 PMCID: PMC8176181 DOI: 10.7150/ijms.59411] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/04/2021] [Indexed: 12/21/2022] Open
Abstract
A new Danshensu/tetramethylpyrazine derivative (ADTM) with cardio-protection effects such as antioxidant, arterial relaxation, pro-angiogenesis and antiplatelet activities. Platelet activating factor receptor (PAFR) plays a key role in myocardial ischemia reperfusion (MIR) injury. This study aims to investigate the protective role of ADTM in MIR injury and clarify the potential role of PAFR. We measured the effects of ADTM on MIR injury in rats in vivo and hypoxia re-oxygenation (HR) injury in neonatal rat ventricular myocytes (NRVMs) in vitro. The results show that ADTM can significantly improve the IR-induced decline in heart function as increasing EF and FS, and restore the decreased cardiac hemodynamic parameters (LVSP, ± dp/dt max) and increased the level of LVEDP, decrease the infarct size of damaged myocardium and lactate dehydrogenase (LDH) activity in serum. Additionally, ADTM inhibits cardiomyocytes apoptosis, caspase-3 activity, and inflammatory response as well as down-regulates the MIR-induced IL-1β and TNFα production. Next, PAFR expression was significantly down-regulated in cardiomyocytes of MIR model in vivo and in vitro after treated with ADTM compare to IR group. At the same time, ADTM and PAFR small interfering RNA (siRNA) could inhibit cardiomyocytes apoptosis and inflammation during HR, while PAF presents the opposite effect. Furthermore, the above effects of PAF in HR induced cardiomyocytes were reversed by co-treatment of ADTM. Our findings demonstrate for the first time that ADTM protects against MIR injury through inhibition of PAFR signaling, which provides a new treatment for MIR.
Collapse
Affiliation(s)
- Jinghao Wang
- Department of Pharmacy, the First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Kai Fan
- Department of Pathophysiology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Cong He
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Qingyang Wang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Qianhui Zhang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Wei Huang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| |
Collapse
|
34
|
Wagatsuma K, Nakase H. Contradictory Effects of NLRP3 Inflammasome Regulatory Mechanisms in Colitis. Int J Mol Sci 2020; 21:ijms21218145. [PMID: 33143375 PMCID: PMC7662299 DOI: 10.3390/ijms21218145] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 02/08/2023] Open
Abstract
The inflammasome is an intracellular molecular complex, which is mainly involved in innate immunity. Inflammasomes are formed in response to danger signals, associated with infection and injury, and mainly regulate the secretion of interleukin-1β and interleukin-18. Inflammasome dysregulation is known to be associated with various diseases and conditions, and its regulatory mechanisms have become of great interest in recent years. In the colon, inflammasomes have been reported to be associated with autophagy and the microbiota, and their dysregulation contributes to colitis and. However, the detailed role of inflammasomes in inflammatory bowel disease is still under debate because the mechanisms that regulate the inflammasome are complex and the inflammasome components and cytokines show seemingly contradictory multiple effects. Herein, we comprehensively review the literature on inflammasome functioning in the colon and describe the complex interactions of the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome components with inflammatory cytokines, autophagy, and the microbiota in experimental colitis models and patients with inflammatory bowel disease.
Collapse
|
35
|
Donovan C, Liu G, Shen S, Marshall JE, Kim RY, Alemao CA, Budden KF, Choi JP, Kohonen-Corish M, El-Omar EM, Yang IA, Hansbro PM. The role of the microbiome and the NLRP3 inflammasome in the gut and lung. J Leukoc Biol 2020; 108:925-935. [PMID: 33405294 DOI: 10.1002/jlb.3mr0720-472rr] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/15/2022] Open
Abstract
The nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family, pyrin domain-containing protein 3 (NLRP3) inflammasome, is one of the most well-characterized inflammasomes, activated by pathogen-associated molecular patterns and damage-associated molecular patterns, including from commensal or pathogenic bacterial and viral infections. The NLRP3 inflammasome promotes inflammatory cell recruitment and regulates immune responses in tissues such as the gastrointestinal tract and the lung, and is involved in many diseases that affect the gut and lung. Recently, the microbiome in the gut and the lung, and the crosstalk between these organs (gut-lung axis), has been identified as a potential mechanism that may influence disease in a bidirectional manner. In this review, we focus on themes presented in this area at the 2019 World Congress on Inflammation. We discuss recent evidence on how the microbiome can affect NLRP3 inflammasome responses in the gut and lung, the role of this inflammasome in regulating gut and lung inflammation in disease, and its potential role in the gut-lung axis. We highlight the exponential increase in our understanding of the NLRP3 inflammasome due to the synthesis of the NLRP3 inflammasome inhibitor, MCC950, and propose future studies that may further elucidate the roles of the NLRP3 inflammasome in gut and lung diseases.
Collapse
Affiliation(s)
- Chantal Donovan
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, New South Wales, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Gang Liu
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, New South Wales, Australia
| | - Sj Shen
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, New South Wales, Australia
| | - Jacqueline E Marshall
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, New South Wales, Australia
| | - Richard Y Kim
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, New South Wales, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Charlotte A Alemao
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Kurtis F Budden
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Jaesung P Choi
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, New South Wales, Australia
| | - Maija Kohonen-Corish
- Woolcock Institute of Medical Research and Faculty of Science, University of Technology Sydney, Garvan Institute of Medical Research and St George and Sutherland Clinical School, University of New South Wales, Kogarah, New South Wales, Australia
| | - Emad M El-Omar
- Microbiome Research Centre, St George and Sutherland Clinical School, University of New South Wales, Kogarah, New South Wales, Australia
| | - Ian A Yang
- The Prince Charles Hospital and The University of Queensland, Brisbane, Queensland, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, New South Wales, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
36
|
Xu J, Xu X, Jiang L, Dua K, Hansbro PM, Liu G. SARS-CoV-2 induces transcriptional signatures in human lung epithelial cells that promote lung fibrosis. Respir Res 2020; 21:182. [PMID: 32664949 PMCID: PMC7359430 DOI: 10.1186/s12931-020-01445-6] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/06/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Severe acute respiratory syndrome (SARS)-CoV-2-induced coronavirus disease-2019 (COVID-19) is a pandemic disease that affects > 2.8 million people worldwide, with numbers increasing dramatically daily. However, there is no specific treatment for COVID-19 and much remains unknown about this disease. Angiotensin-converting enzyme (ACE)2 is a cellular receptor of SARS-CoV-2. It is cleaved by type II transmembrane serine protease (TMPRSS)2 and disintegrin and metallopeptidase domain (ADAM)17 to assist viral entry into host cells. Clinically, SARS-CoV-2 infection may result in acute lung injury and lung fibrosis, but the underlying mechanisms of COVID-19 induced lung fibrosis are not fully understood. METHODS The networks of ACE2 and its interacting molecules were identified using bioinformatic methods. Their gene and protein expressions were measured in human epithelial cells after 24 h SARS-CoV-2 infection, or in existing datasets of lung fibrosis patients. RESULTS We confirmed the binding of SARS-CoV-2 and ACE2 by bioinformatic analysis. TMPRSS2, ADAM17, tissue inhibitor of metalloproteinase (TIMP)3, angiotensinogen (AGT), transformation growth factor beta (TGFB1), connective tissue growth factor (CTGF), vascular endothelial growth factor (VEGF) A and fibronectin (FN) were interacted with ACE2, and the mRNA and protein of these molecules were expressed in lung epithelial cells. SARS-CoV-2 infection increased ACE2, TGFB1, CTGF and FN1 mRNA that were drivers of lung fibrosis. These changes were also found in lung tissues from lung fibrosis patients. CONCLUSIONS Therefore, SARS-CoV-2 binds with ACE2 and activates fibrosis-related genes and processes to induce lung fibrosis.
Collapse
Affiliation(s)
- Jincheng Xu
- School of Stomatology, Bengbu Medical College, Bengbu, 2033, Anhui, China
| | - Xiaoyue Xu
- School of Public Health and Community Medicine, Faculty of Medicine, University of New South Wales, Kensington, 233000, NSW, Australia
- Faculty of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Lina Jiang
- School of Stomatology, Bengbu Medical College, Bengbu, 2033, Anhui, China
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia
- Centre for Inflammation, Centenary Institute, Camperdown, NSW, 2050, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, Camperdown, NSW, 2050, Australia
- School of Life Science, Faculty of Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Gang Liu
- Centre for Inflammation, Centenary Institute, Camperdown, NSW, 2050, Australia.
- School of Life Science, Faculty of Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
37
|
Shinde T, Hansbro PM, Sohal SS, Dingle P, Eri R, Stanley R. Microbiota Modulating Nutritional Approaches to Countering the Effects of Viral Respiratory Infections Including SARS-CoV-2 through Promoting Metabolic and Immune Fitness with Probiotics and Plant Bioactives. Microorganisms 2020; 8:E921. [PMID: 32570850 PMCID: PMC7355654 DOI: 10.3390/microorganisms8060921] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/16/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Viral respiratory infections (VRIs) can spread quickly and cause enormous morbidity and mortality worldwide. These events pose serious threats to public health due to time lags in developing vaccines to activate the acquired immune system. The high variability of people's symptomatic responses to viral infections, as illustrated in the current COVID-19 pandemic, indicates the potential to moderate the severity of morbidity from VRIs. Growing evidence supports roles for probiotic bacteria (PB) and prebiotic dietary fiber (DF) and other plant nutritional bioactives in modulating immune functions. While human studies help to understand the epidemiology and immunopathology of VRIs, the chaotic nature of viral transmissions makes it difficult to undertake mechanistic study where the pre-conditioning of the metabolic and immune system could be beneficial. However, recent experimental studies have significantly enhanced our understanding of how PB and DF, along with plant bioactives, can significantly modulate innate and acquired immunity responses to VRIs. Synbiotic combinations of PB and DF potentiate increased benefits primarily through augmenting the production of short-chain fatty acids (SCFAs) such as butyrate. These and specific plant polyphenolics help to regulate immune responses to both restrain VRIs and temper the neutrophil response that can lead to acute respiratory distress syndrome (ARDS). This review highlights the current understanding of the potential impact of targeted nutritional strategies in setting a balanced immune tone for viral clearance and reinforcing homeostasis. This knowledge may guide the development of public health tactics and the application of functional foods with PB and DF components as a nutritional approach to support countering VRI morbidity.
Collapse
Affiliation(s)
- Tanvi Shinde
- Centre for Food Innovation, Tasmanian Institute of Agriculture, University of Tasmania, Launceston, TAS 7250, Australia
- Gut Health Research Group, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS 7250, Australia;
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, Sydney, NSW 2050, and University of Technology Sydney, Faculty of Science, Ultimo, NSW 2007, Australia;
| | - Sukhwinder Singh Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS 7248, Australia;
| | - Peter Dingle
- Dingle Wellness, South Fremantle, WA 6162, Australia;
| | - Rajaraman Eri
- Gut Health Research Group, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS 7250, Australia;
| | - Roger Stanley
- Centre for Food Innovation, Tasmanian Institute of Agriculture, University of Tasmania, Launceston, TAS 7250, Australia
| |
Collapse
|
38
|
Zhang X, Wei D, Zhao Y, Zhong Z, Wang Y, Song Y, Cai M, Zhang W, Zhao J, Lv C, Zhu H. Immunization With a Secreted Esterase Protects Mice Against Multiple Serotypes (M1, M3, and M28) of Group A Streptococcus. Front Microbiol 2020; 11:565. [PMID: 32308652 PMCID: PMC7145942 DOI: 10.3389/fmicb.2020.00565] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 03/16/2020] [Indexed: 11/13/2022] Open
Abstract
Streptococcal secreted esterase (Sse) is a platelet-activating factor acetylhydrolase that is critical for Group A Streptococcus (GAS) skin invasion and innate immune evasion. There are two Sse variant complexes that share >98% identity within each complex but display about 37% variation between the complexes in amino acid sequences. Sse immunization protects mice against lethal infection and skin invasion in subcutaneous infection with the hypervirulent CovRS mutant strain, MGAS5005. However, it is not known whether Sse immunization provides significant protection against infection of GAS with functional CovRS and whether immunization with Sse of one variant complex provides protection against infection of GAS that produces Sse of another variant complex. This study was designed to address these questions. Mice were immunized with recombinant Sse of M1 GAS (SseM1) and challenged with MGAS5005 (serotype M1, CovS mutant, and Sse of variant complex I), MGAS315 (M3, CovS mutant, and Sse of variant complex I), MGAS2221 (M1, wild-type CovRS, and Sse of variant complex I), and MGAS6180 (M28, wild-type CovRS, and Sse of variant complex II). SseM1 immunization significantly increased survival rates of mice in subcutaneous MGAS5005 and intraperitoneal MGAS6180 challenges and showed consistently higher or longer survival in the other challenges. Immunized mice had smaller skin lesion and higher neutrophil responses in subcutaneous infections and lower GAS burdens in spleen, liver, and kidney in most of the challenge experiments than control mice. SseM1 immunization enhanced proinflammatory responses. These data suggest that Sse immunization has a broad benefit against GAS infections that can vary in extent from strain to strain and that the benefit may be due to the immunization-enhanced proinflammatory responses. In particular, immunization with SseM1 can provide protection against M28 GAS infection even though its Sse and SseM1 have significant variations.
Collapse
Affiliation(s)
- Xiaolan Zhang
- College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Deqin Wei
- College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Yuan Zhao
- College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Zhaohua Zhong
- College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Yue Wang
- College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Yingli Song
- College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Minghui Cai
- College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Wenli Zhang
- College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Jizi Zhao
- College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Chunmei Lv
- College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Hui Zhu
- College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| |
Collapse
|
39
|
Liu G, Baird AW, Parsons MJ, Fan K, Skerrett-Byrne DA, Nair PM, Makanyengo S, Chen J, Neal R, Goggins BJ, Tay H, Mathe A, Soh WS, Minahan K, Hansbro PM, Nixon B, McCaughan GW, Holtmann G, Colgan SP, Keely S. Platelet activating factor receptor acts to limit colitis-induced liver inflammation. FASEB J 2020; 34:7718-7732. [PMID: 32293760 DOI: 10.1096/fj.201901779r] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 03/25/2020] [Accepted: 03/25/2020] [Indexed: 12/31/2022]
Abstract
Liver inflammation is a common extraintestinal manifestation in inflammatory bowel disease (IBD), yet, the mechanisms driving gut-liver axis inflammation remain poorly understood. IBD leads to a breakdown in the integrity of the intestinal barrier causing an increase in portal and systemic gut-derived antigens, which challenge the liver. Here, we examined the role of platelet activating factor receptor (PAFR) in colitis-associated liver damage using dextran sulfate sodium (DSS) and anti-CD40-induced colitis models. Both DSS and anti-CD40 models exhibited liver inflammation associated with colitis. Colitis reduced global PAFR protein expression in mouse livers causing an exclusive re-localization of PAFR to the portal triad. The global decrease in liver PAFR was associated with increased sirtuin 1 while relocalized PAFR expression was limited to Kupffer cells (KCs) and co-localized with toll-like receptor 4. DSS activated the NLRP3-inflammasome and increased interleukin (IL)-1β in the liver. Antagonism of PAFR amplified the inflammasome response by increasing NLRP3, caspase-1, and IL-1β protein levels in the liver. LPS also increased NLRP3 response in human hepatocytes, however, overexpression of PAFR restored the levels of NLPR3 and caspase-1 proteins. Interestingly, KCs depletion also increased IL-1β protein in mouse liver after DSS challenge. These data suggest a protective role for PAFR-expressing KCs during colitis and that regulation of PAFR is important for gut-liver axis homeostasis.
Collapse
Affiliation(s)
- Gang Liu
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia.,School of Life Science, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia.,Centre for Inflammation, Centenary Institute, Camperdown, NSW, Australia
| | - Alan W Baird
- UCD School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Marie J Parsons
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Kening Fan
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - David A Skerrett-Byrne
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Environmental and Life Sciences, Priority Research Centre for Reproductive Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Prema M Nair
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Samwel Makanyengo
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Jinbiao Chen
- Liver Injury and Cancer Program, Centenary Research Institute, Camperdown, NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Rachel Neal
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Bridie J Goggins
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Hock Tay
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia.,Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia
| | - Andrea Mathe
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Wai S Soh
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Kyra Minahan
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Phil M Hansbro
- School of Life Science, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia.,Centre for Inflammation, Centenary Institute, Camperdown, NSW, Australia
| | - Brett Nixon
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Environmental and Life Sciences, Priority Research Centre for Reproductive Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Geoffrey W McCaughan
- Liver Injury and Cancer Program, Centenary Research Institute, Camperdown, NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Gerald Holtmann
- Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Woolloongabba, QLD, Australia.,Faculty of Medicine, The University of Queensland, Woolloongabba, QLD, Australia
| | - Sean P Colgan
- University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Simon Keely
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
40
|
Ameliorative effect of selective NLRP3 inflammasome inhibitor MCC950 in an ovalbumin-induced allergic rhinitis murine model. Int Immunopharmacol 2020; 83:106394. [PMID: 32193102 DOI: 10.1016/j.intimp.2020.106394] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 03/09/2020] [Accepted: 03/09/2020] [Indexed: 12/14/2022]
Abstract
Allergic rhinitis (AR) is a complex IgE-mediated nasal allergic and inflammatory disease. Nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3) is essential in the process of allergic and inflammatory responses. MCC950 is a selective NLRP3 inhibitor. However, its role and mechanism in AR remains undetermined. The present study aimed to explore the effect and mechanism of MCC950 on an ovalbumin (OVA) induced mouse model of AR. The AR BALB/c mice were constructed using OVA and administrated intranasally with MCC950. Concentrations of OVA-specific IgE, histamines and leukotrienes C4 (LTC4) in serum, and OVA-specific IgE, ECP, IFN-γ, IL-4, IL-5, IL-13, IL-1β and IL-18 in nasal lavage fluid (NLF) were assayed by enzyme-linked immunosorbent assay (ELISA). Inflammatory cells were counted in NLF. HE and PAS staing were used for evaluating eosinophils and goblet cells. Immunohistochemistry (IHC) staining were employed to evaluate immunolabeling of NLRP3, Caspase-1, ASC, IL-1β and IL-18 in nasal mucosas of mice. Real-time PCR was conducted to assay NLRP3, Caspase-1, ASC, IL-1β and IL-18 mRNA levels. In vitro studies, western blotting, real-time PCR and ELISA were performed to evaluate the effects and mechanisms of OVA and NLRP3 inhibitor MCC950 on spleen mononuclear cells. We found significant downregulation of sneezing, nasal rubbing, inflammatory cytokines, inflammatory cells and NLRP3, Caspase-1, ASC, IL-1β and IL-18 expression in MCC950 treated mice compared with untreated AR mice. In spleen mononuclear cells culture and stimulation experiment, NLRP3, Caspase-1, ASC, IL-1β and IL-18 levels were upregulated by OVA but inhibited by MCC950. In conclusion, MCC950 could effectively exert its ameliorative effect in murine AR by inhibiting NLRP3 and leads to reduction of Caspase-1, ASC, IL-1β and IL-18, resulting in the attenuation of the allergic and inflammatory responses.
Collapse
|
41
|
Li J, Xu X, Jiang Y, Hansbro NG, Hansbro PM, Xu J, Liu G. Elastin is a key factor of tumor development in colorectal cancer. BMC Cancer 2020; 20:217. [PMID: 32171282 PMCID: PMC7071655 DOI: 10.1186/s12885-020-6686-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 02/26/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the most common cancer and a leading cause of death worldwide. Extracellular matrix (ECM) proteins regulate tumor growth and development in CRC. Elastin (ELN) is a component of ECM proteins involved in the tumor microenvironment. However, the role of ELN in CRC remains unclear. METHODS In this study, we analyzed ELN gene expression in tumors from CRC patients and adjacent non-tumor colon tissues and healthy controls from two existing microarray datasets. ELN protein was measured in human normal colon cells and colon cancer epithelial cells and tumor development was assessed in colon epithelial cells cultured in medium with or without ELN peptide on plates coated with ELN recombinant protein. Control plates were coated with PBS only. RESULTS We found ELN gene expression was increased in tumors from CRC patients compared to adjacent non-tumor tissues and healthy controls. ELN protein was increased in cancer cells compared to normal colon epithelial cells. Transforming growth factor beta (TGF-β) was a key cytokine to induce production of ECM proteins, but it did not induce ELN expression in colon cancer cells. Matrix metalloproteinase 9 (MMP9) gene expression was increased, but that of MMP12 (elastase) did not change between CRC patients and control. Tissue inhibitor of metalloproteinases 3 (TIMP3) gene expression was decreased in colon tissues from CRC patients compared to healthy controls. However, MMP9, MMP12 and TIMP3 proteins were increased in colon cancer cells. ELN recombinant protein increased proliferation and wound healing in colon cancer epithelial cells. This had further increased in cancer cells incubated in plates coated with recombinant ELN coated plate and in culture media containing ELN peptide. A potential mechanism was that ELN induced epithelial mesenchymal transition with increased alpha-smooth muscle actin and vimentin proteins but decreased E-cadherin protein. Tumor necrosis factor alpha (TNF) mRNA was also increased in CRC patients compared to controls. ELN recombinant protein induced further increases in TNF protein in mouse bone marrow derived macrophages after lipopolysaccharide stimulation. CONCLUSIONS These data suggest ELN regulates tumor development and the microenvironment in CRC.
Collapse
Affiliation(s)
- Jinzhi Li
- School of Nursing, Bengbu Medical College, Bengbu, Anhui, China
| | - Xiaoyue Xu
- Faculty of Health, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Yanyan Jiang
- School of Anatomy, Bengbu Medical College, Bengbu, Anhui, China
| | - Nicole G Hansbro
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, New South Wales, Australia.,Centre for Inflammation, Centenary Institute, Camperdown, New South Wales, Australia.,Priority Research Centre for Health Lungs, Hunter Medical Research Institute, The University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Philip M Hansbro
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, New South Wales, Australia.,Centre for Inflammation, Centenary Institute, Camperdown, New South Wales, Australia.,Priority Research Centre for Health Lungs, Hunter Medical Research Institute, The University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Jincheng Xu
- Stomatology Department, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China. .,School of Dental Medicine, Bengbu Medical College, Bengbu, Anhui, China.
| | - Gang Liu
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, New South Wales, Australia. .,Centre for Inflammation, Centenary Institute, Camperdown, New South Wales, Australia.
| |
Collapse
|
42
|
Forty Years Since the Structural Elucidation of Platelet-Activating Factor (PAF): Historical, Current, and Future Research Perspectives. Molecules 2019; 24:molecules24234414. [PMID: 31816871 PMCID: PMC6930554 DOI: 10.3390/molecules24234414] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 12/11/2022] Open
Abstract
In the late 1960s, Barbaro and Zvaifler described a substance that caused antigen induced histamine release from rabbit platelets producing antibodies in passive cutaneous anaphylaxis. Henson described a ‘soluble factor’ released from leukocytes that induced vasoactive amine release in platelets. Later observations by Siraganuan and Osler observed the existence of a diluted substance that had the capacity to cause platelet activation. In 1972, the term platelet-activating factor (PAF) was coined by Benveniste, Henson, and Cochrane. The structure of PAF was later elucidated by Demopoulos, Pinckard, and Hanahan in 1979. These studies introduced the research world to PAF, which is now recognised as a potent phospholipid mediator. Since its introduction to the literature, research on PAF has grown due to interest in its vital cell signalling functions and more sinisterly its role as a pro-inflammatory molecule in several chronic diseases including cardiovascular disease and cancer. As it is forty years since the structural elucidation of PAF, the aim of this review is to provide a historical account of the discovery of PAF and to provide a general overview of current and future perspectives on PAF research in physiology and pathophysiology.
Collapse
|
43
|
Vaughan A, Frazer ZA, Hansbro PM, Yang IA. COPD and the gut-lung axis: the therapeutic potential of fibre. J Thorac Dis 2019; 11:S2173-S2180. [PMID: 31737344 DOI: 10.21037/jtd.2019.10.40] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Current management strategies for chronic obstructive pulmonary disease (COPD) incorporate a step-wise, multidisciplinary approach to effectively manage patient symptoms and prevent disease progression. However, there has been limited advancement in therapies to address the underlying cause of COPD pathogenesis. Recent research has established the link between the lungs and the gut-the gut-lung axis -and the gut microbiome is a major component. The gut microbiome is likely perturbed in COPD, contributing to chronic inflammation. Diet is a readily modifiable factor and the diet of COPD patients is often deficient in nutrients such as fibre. The metabolism of dietary fibre by gut microbiomes produces anti-inflammatory short chain fatty acid (SCFAs), which could protect against inflammation in the lungs. By addressing the 'fibre gap' in the diet of COPD patients, this targeted dietary intervention may reduce inflammation, both systemically and in the airways, and value-add to the paradigm shift in respiratory medicine, from reactive to personalised and participatory medicine.
Collapse
Affiliation(s)
- Annalicia Vaughan
- UQ Thoracic Research Centre, Faculty of Medicine, The University of Queensland, Chermside, Brisbane, Australia.,Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, Australia
| | - Zoe A Frazer
- UQ Thoracic Research Centre, Faculty of Medicine, The University of Queensland, Chermside, Brisbane, Australia.,Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, University of Technology Sydney, Faculty of Science, Camperdown, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Callaghan, Australia
| | - Ian A Yang
- UQ Thoracic Research Centre, Faculty of Medicine, The University of Queensland, Chermside, Brisbane, Australia.,Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, Australia
| |
Collapse
|
44
|
Shukla SD, Shastri MD, Chong WC, Dua K, Budden KF, Mahmood MQ, Hansbro NG, Keely S, Eri R, Patel RP, Peterson GM, Hansbro PM. Microbiome-focused asthma management strategies. Curr Opin Pharmacol 2019; 46:143-149. [PMID: 31357048 DOI: 10.1016/j.coph.2019.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/24/2019] [Accepted: 06/24/2019] [Indexed: 12/11/2022]
Abstract
Asthma is a common, heterogeneous and serious disease with high prevalence globally. Poorly controlled, steroid-resistant asthma is particularly important as there are no effective therapies and it exerts substantial healthcare and societal burden. The role of microbiomes, particularly in chronic diseases has generated considerable interest in recent times. Existing evidence clearly demonstrates an association between asthma initiation and the microbiome, both respiratory and gastro-intestinal, although its' roles are poorly understood when assessing the asthma progression or heterogeneity (i.e. phenotypes/endotypes) across different geographical locations. Moreover, modulating microbiomes could be preventive and/or therapeutic in patients with asthma warrants urgent attention. Here, we review recent advances in assessing the role of microbiomes in asthma and present the challenges associated with the potential therapeutic utility of modifying microbiomes in management.
Collapse
Affiliation(s)
- Shakti D Shukla
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia
| | - Madhur D Shastri
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Australia
| | - Wai Chin Chong
- Department of Molecular and Translational Science, Monash University, Clayton, Australia; Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Australia
| | - Kamal Dua
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, Australia
| | - Kurtis F Budden
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia
| | - Malik Quasir Mahmood
- Medicine, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Nicole G Hansbro
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia; Centre for inflammation, Centenary Institute, Sydney, and School of Life Sciences, University of Technology, Ultimo, NSW, Australia
| | - Simon Keely
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia
| | - Rajaraman Eri
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Australia
| | - Rahul P Patel
- Pharmacy, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Gregory M Peterson
- Pharmacy, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia; Centre for inflammation, Centenary Institute, Sydney, and School of Life Sciences, University of Technology, Ultimo, NSW, Australia.
| |
Collapse
|
45
|
Liu G, Cooley MA, Jarnicki AG, Borghuis T, Nair PM, Tjin G, Hsu AC, Haw TJ, Fricker M, Harrison CL, Jones B, Hansbro NG, Wark PA, Horvat JC, Argraves WS, Oliver BG, Knight DA, Burgess JK, Hansbro PM. Fibulin-1c regulates transforming growth factor-β activation in pulmonary tissue fibrosis. JCI Insight 2019; 5:124529. [PMID: 31343988 DOI: 10.1172/jci.insight.124529] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Tissue remodeling/fibrosis is a major feature of all fibrotic diseases, including idiopathic pulmonary fibrosis (IPF). It is underpinned by accumulating extracellular matrix (ECM) proteins. Fibulin-1c (Fbln1c) is a matricellular ECM protein associated with lung fibrosis in both humans and mice, and stabilizes collagen formation. Here we discovered that Fbln1c was increased in the lung tissues of IPF patients and experimental bleomycin-induced pulmonary fibrosis. Fbln1c-deficient (-/-) mice had reduced pulmonary remodeling/fibrosis and improved lung function after bleomycin challenge. Fbln1c interacted with fibronectin, periostin and tenascin-c in collagen deposits following bleomycin challenge. In a novel mechanism of fibrosis Fbln1c bound to latent transforming growth factor (TGF)-β binding protein-1 (LTBP1) to induce TGF-β activation, and mediated downstream Smad3 phosphorylation/signaling. This process increased myofibroblast numbers and collagen deposition. Fbln1 and LTBP1 co-localized in lung tissues from IPF patients. Thus, Fbln1c may be a novel driver of TGF-β-induced fibrosis involving LTBP1 and may be an upstream therapeutic target.
Collapse
Affiliation(s)
- Gang Liu
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and the University of Newcastle, Newcastle, New South Wales, Australia.,School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia.,Centenary Institute, Sydney, New South Wales, Australia
| | - Marion A Cooley
- Department of Oral Biology and Diagnostic Sciences, Augusta University, Augusta, Georgia, USA
| | - Andrew G Jarnicki
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and the University of Newcastle, Newcastle, New South Wales, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Theo Borghuis
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Department of Pathology and Medical Biology, Groningen, Netherlands
| | - Prema M Nair
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and the University of Newcastle, Newcastle, New South Wales, Australia
| | - Gavin Tjin
- Woolcock Institute of Medical Research, Discipline of Pharmacology, the University of Sydney, Sydney, New South Wales, Australia
| | - Alan C Hsu
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and the University of Newcastle, Newcastle, New South Wales, Australia
| | - Tatt Jhong Haw
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and the University of Newcastle, Newcastle, New South Wales, Australia
| | - Michael Fricker
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and the University of Newcastle, Newcastle, New South Wales, Australia
| | - Celeste L Harrison
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and the University of Newcastle, Newcastle, New South Wales, Australia
| | - Bernadette Jones
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and the University of Newcastle, Newcastle, New South Wales, Australia
| | - Nicole G Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and the University of Newcastle, Newcastle, New South Wales, Australia.,School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia.,Centenary Institute, Sydney, New South Wales, Australia
| | - Peter A Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and the University of Newcastle, Newcastle, New South Wales, Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and the University of Newcastle, Newcastle, New South Wales, Australia
| | - W Scott Argraves
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Brian G Oliver
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia.,Woolcock Institute of Medical Research, Discipline of Pharmacology, the University of Sydney, Sydney, New South Wales, Australia
| | - Darryl A Knight
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and the University of Newcastle, Newcastle, New South Wales, Australia
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Department of Pathology and Medical Biology, Groningen, Netherlands.,Woolcock Institute of Medical Research, Discipline of Pharmacology, the University of Sydney, Sydney, New South Wales, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and the University of Newcastle, Newcastle, New South Wales, Australia.,School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia.,Centenary Institute, Sydney, New South Wales, Australia
| |
Collapse
|