1
|
Kaden T, Alonso‐Román R, Stallhofer J, Gresnigt MS, Hube B, Mosig AS. Leveraging Organ-on-Chip Models to Investigate Host-Microbiota Dynamics and Targeted Therapies for Inflammatory Bowel Disease. Adv Healthc Mater 2025; 14:e2402756. [PMID: 39491534 PMCID: PMC12004439 DOI: 10.1002/adhm.202402756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/29/2024] [Indexed: 11/05/2024]
Abstract
Inflammatory bowel disease (IBD) is an idiopathic gastrointestinal disease with drastically increasing incidence rates. Due to its multifactorial etiology, a precise investigation of the pathogenesis is extremely difficult. Although reductionist cell culture models and more complex disease models in animals have clarified the understanding of individual disease mechanisms and contributing factors of IBD in the past, it remains challenging to bridge research and clinical practice. Conventional 2D cell culture models cannot replicate complex host-microbiota interactions and stable long-term microbial culture. Further, extrapolating data from animal models to patients remains challenging due to genetic and environmental diversity leading to differences in immune responses. Human intestine organ-on-chip (OoC) models have emerged as an alternative in vitro model approach to investigate IBD. OoC models not only recapitulate the human intestinal microenvironment more accurately than 2D cultures yet may also be advantageous for the identification of important disease-driving factors and pharmacological interventions targets due to the possibility of emulating different complexities. The predispositions and biological hallmarks of IBD focusing on host-microbiota interactions at the intestinal mucosal barrier are elucidated here. Additionally, the potential of OoCs to explore microbiota-related therapies and personalized medicine for IBD treatment is discussed.
Collapse
Affiliation(s)
- Tim Kaden
- Dynamic42 GmbH07745JenaGermany
- Institute of Biochemistry IICenter for Sepsis Control and CareJena University Hospital07747JenaGermany
| | - Raquel Alonso‐Román
- Department of Microbial Pathogenicity MechanismsLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Junior Research Group Adaptive Pathogenicity StrategiesLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
| | | | - Mark S. Gresnigt
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Junior Research Group Adaptive Pathogenicity StrategiesLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
| | - Bernhard Hube
- Department of Microbial Pathogenicity MechanismsLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Institute of MicrobiologyFaculty of Biological SciencesFriedrich Schiller University07743JenaGermany
| | - Alexander S. Mosig
- Institute of Biochemistry IICenter for Sepsis Control and CareJena University Hospital07747JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
| |
Collapse
|
2
|
Wang M, Wang Z, Li Z, Qu Y, Zhao J, Wang L, Zhou X, Xu Z, Zhang D, Jiang P, Fan B, Liu Y. Targeting programmed cell death in inflammatory bowel disease through natural products: New insights from molecular mechanisms to targeted therapies. Phytother Res 2025; 39:1776-1807. [PMID: 38706097 DOI: 10.1002/ptr.8216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 03/14/2024] [Accepted: 04/11/2024] [Indexed: 05/07/2024]
Abstract
Inflammatory bowel disease (IBD) is an autoimmune disorder primarily characterized by intestinal inflammation and recurrent ulceration, leading to a compromised intestinal barrier and inflammatory infiltration. This disorder's pathogenesis is mainly attributed to extensive damage or death of intestinal epithelial cells, along with abnormal activation or impaired death regulation of immune cells and the release of various inflammatory factors, which contribute to the inflammatory environment in the intestines. Thus, maintaining intestinal homeostasis hinges on balancing the survival and functionality of various cell types. Programmed cell death (PCD) pathways, including apoptosis, pyroptosis, autophagy, ferroptosis, necroptosis, and neutrophil extracellular traps, are integral in the pathogenesis of IBD by mediating the death of intestinal epithelial and immune cells. Natural products derived from plants, fruits, and vegetables have shown potential in regulating PCD, offering preventive and therapeutic avenues for IBD. This article reviews the role of natural products in IBD treatment by focusing on targeting PCD pathways, opening new avenues for clinical IBD management.
Collapse
Affiliation(s)
- Mengjie Wang
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhiyuan Wang
- People's Hospital of Zhengzhou, Zhengzhou, China
| | - Zhichao Li
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuan Qu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiting Zhao
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lei Wang
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinpeng Zhou
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ziqi Xu
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Di Zhang
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ping Jiang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bing Fan
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ying Liu
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
3
|
Wang Z, Yan W, Lin X, Qin G, Li K, Jiang L, Li X, Xiao X, Luo T, Hou Y. Forsythiaside A Alleviates Ulcerative Colitis and Inhibits Neutrophil Extracellular Traps Formation in the Mice. Phytother Res 2025. [PMID: 40099671 DOI: 10.1002/ptr.8440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 12/10/2024] [Accepted: 01/05/2025] [Indexed: 03/20/2025]
Abstract
Forsythiaside A (FA), the primary compound found in Forsythia suspensa (Thunb.) Vahl, has demonstrated various pharmacological effects, but its impact on ulcerative colitis (UC) is underexplored. Our study examined the distribution of FA in different parts of the gastrointestinal tracts and its therapeutic effects on UC, along with the underlying mechanisms. The levels of FA in gastrointestinal tracts and plasma were analyzed by high-performance liquid chromatography; mice were given dextran sulfate sodium in drinking water to develop the UC model. The UC mice were treated with FA (15, 30, and 60 mg/kg) for 10 days. FA showed relatively high concentration retention in the colon within 4 h. The treatment of FA improved body weight loss, diarrhea, rectal bleeding, colon shortening, and histological damage in UC mice. It also increased the expression of the tight junction protein and decreased inflammatory cytokines in the colon. The microbiota analysis using 16S rRNA sequencing revealed that FA could alleviate gut dysbiosis in colitis mice. Of importance, we found FA resulted in a reduction of neutrophil extracellular traps formation (NETosis) and inhibited peptidyl arginine deiminase 4 (PAD4) in colon tissue of colitis mice. In cultured neutrophils, FA pretreatment led to a suppression of PAD4 expression and NETosis induced by PMA. These findings suggest that FA can be retained in the colon and may alleviate UC by inhibiting NETs formation, indicating its potential for preventing or treating UC.
Collapse
Affiliation(s)
- Zhuyun Wang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiyan Yan
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaojing Lin
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guangcheng Qin
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kemeng Li
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lincheng Jiang
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xingwang Li
- Laboratory of Traditional Chinese Medicine, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing, China
| | - Xiaoqiu Xiao
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ting Luo
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Hou
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Laboratory of Traditional Chinese Medicine, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing, China
| |
Collapse
|
4
|
Karsdal M, Cox TR, Parker AL, Willumsen N, Sand JMB, Jenkins G, Hansen HH, Oldenburger A, Geillinger-Kaestle KE, Larsen AT, Black D, Genovese F, Eckersley A, Heinz A, Nyström A, Holm Nielsen S, Bennink L, Johannsson L, Bay-Jensen AC, Orange DE, Friedman S, Røpke M, Fiore V, Schuppan D, Rieder F, Simona B, Borthwick L, Skarsfeldt M, Wennbo H, Thakker P, Stoffel R, Clarke GW, Kalluri R, Ruane D, Zannad F, Mortensen JH, Sinkeviciute D, Sundberg F, Coseno M, Thudium C, Croft AP, Khanna D, Cooreman M, Broermann A, Leeming DJ, Mobasheri A, Ricard-Blum S. Advances in Extracellular Matrix-Associated Diagnostics and Therapeutics. J Clin Med 2025; 14:1856. [PMID: 40142664 PMCID: PMC11943371 DOI: 10.3390/jcm14061856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/28/2025] [Accepted: 02/08/2025] [Indexed: 03/28/2025] Open
Abstract
The extracellular matrix (ECM) is the common denominator of more than 50 chronic diseases. Some of these chronic pathologies lead to enhanced tissue formation and deposition, whereas others are associated with increased tissue degradation, and some exhibit a combination of both, leading to severe tissue alterations. To develop effective therapies for diseases affecting the lung, liver, kidney, skin, intestine, musculoskeletal system, heart, and solid tumors, we need to modulate the ECM's composition to restore its organization and function. Across diverse organ diseases, there are common denominators and distinguishing factors in this fibroinflammatory axis, which may be used to foster new insights into drug development across disease indications. The 2nd Extracellular Matrix Pharmacology Congress took place in Copenhagen, Denmark, from 17 to 19 June 2024 and was hosted by the International Society of Extracellular Matrix Pharmacology. The event was attended by 450 participants from 35 countries, among whom were prominent scientists who brought together state-of-the-art research on organ diseases and asked important questions to facilitate drug development. We highlight key aspects of the ECM in the liver, kidney, skin, intestine, musculoskeletal system, lungs, and solid tumors to advance our understanding of the ECM and its central targets in drug development. We also highlight key advances in the tools and technology that enable this drug development, thereby supporting the ECM.
Collapse
Affiliation(s)
- Morten Karsdal
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Thomas R. Cox
- Garvan Institute of Medical Research, Sydney 2010, Australia; (T.R.C.); (A.L.P.)
- School of Clinical Medicine, St Vincent’s Clinical Campus, UNSW Medicine & Health, UNSW, Sydney 2010, Australia
| | - Amelia L. Parker
- Garvan Institute of Medical Research, Sydney 2010, Australia; (T.R.C.); (A.L.P.)
- School of Clinical Medicine, St Vincent’s Clinical Campus, UNSW Medicine & Health, UNSW, Sydney 2010, Australia
| | - Nicholas Willumsen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Jannie Marie Bülow Sand
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Gisli Jenkins
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, NIHR Imperial Biomedical Research Centre, Imperial College London, London SW7 2AZ, UK;
| | | | | | - Kerstin E. Geillinger-Kaestle
- Department of Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400 Biberach an der Riss, Germany;
| | - Anna Thorsø Larsen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | | | - Federica Genovese
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Alexander Eckersley
- Wellcome Centre for Cell Matrix Research, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, University of Manchester, Manchester M13 9PL, UK;
| | - Andrea Heinz
- LEO Foundation Center for Cutaneous Drug Delivery, Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark;
| | - Alexander Nyström
- Department of Dermatology, Faculty of Medicine, Medical Center—University of Freiburg, 79106 Breisgau, Germany;
| | - Signe Holm Nielsen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | | | | | - Anne-Christine Bay-Jensen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Dana E. Orange
- Hospital for Special Surgery, The Rockefeller University, New York, NY 10065, USA;
| | - Scott Friedman
- Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA;
| | | | - Vincent Fiore
- Boehringer Ingelheim, 55218 Ingelheim am Rhein, Germany;
| | - Detlef Schuppan
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany;
| | - Florian Rieder
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, OH 44195, USA;
| | | | - Lee Borthwick
- FibroFind Ltd., FibroFind Laboratories, Medical School, Newcastle upon Tyne NE2 4HH, UK;
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Mark Skarsfeldt
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Haakan Wennbo
- Takeda, Translational Medicine Biomarkers Gastrointestinal & Global, Boston, MA 02110, USA; (H.W.); (P.T.)
| | - Paresh Thakker
- Takeda, Translational Medicine Biomarkers Gastrointestinal & Global, Boston, MA 02110, USA; (H.W.); (P.T.)
| | - Ruedi Stoffel
- Roche Diagnostics International Ltd., 6343 Rotkreuz, Switzerland;
| | - Graham W. Clarke
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden;
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King’s College, London E1 9RT, UK
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Darren Ruane
- Janssen Immunology, Translational Sciences and Medicine, La Jolla, CA 92037, USA;
| | - Faiez Zannad
- Division of Heart Failure and Hypertension, and of the Inserm CIC, University of Lorraine, 54000 Metz, France;
| | - Joachim Høg Mortensen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Dovile Sinkeviciute
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Fred Sundberg
- Sengenics Corporation LLC, Wilmington, DE 19801, USA; (F.S.); (M.C.)
| | - Molly Coseno
- Sengenics Corporation LLC, Wilmington, DE 19801, USA; (F.S.); (M.C.)
| | - Christian Thudium
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Adam P. Croft
- National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, University of Birmingham, Birmingham B15 2TT, UK;
- Institute of Inflammation and Ageing, Queen Elizabeth Hospital, University of Birmingham, Birmingham B15 2TT, UK
| | - Dinesh Khanna
- Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA;
| | | | - Andre Broermann
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400 Biberach an der Riss, Germany;
| | - Diana Julie Leeming
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Ali Mobasheri
- Faculty of Medicine, University of Oulu, 90570 Oulu, Finland;
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania
- Faculté de Médecine, Université de Liège, 4000 Liège, Belgium
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Sylvie Ricard-Blum
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), UMR 5246 CNRS, ICBMS, University Lyon 1, 69622 Villeurbanne Cedex, France;
| |
Collapse
|
5
|
Li SH, Huang QH, Yang QQ, Huang Q, Wang DX, Yang J, Huang SH, Zhang SY, Wang JM, Xie LS, Yu SG, Wu QF. The shared mechanism of barrier dysfunction in ulcerative colitis and Alzheimer's disease: DDIT4/IL1β neutrophil extracellular traps drive macrophages-mediated phagocytosis. Int Immunopharmacol 2025; 149:114188. [PMID: 39908802 DOI: 10.1016/j.intimp.2025.114188] [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: 11/19/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 02/07/2025]
Abstract
Ulcerative colitis (UC) and Alzheimer's disease (AD) share a common etiology as inflammatory diseases characterized by barrier deterioration. The aim of this study is to elucidate how neutrophil extracellular traps (NETs), serving as a comorbid etiological factor, can trigger the dysfunction in both the intestinal barrier and blood-brain barrier (BBB). Integrated bioinformatics analysis revealed 14 overlapped NETs-related differential expressed genes in UC and AD, which strongly featured barrier dysfunction. The following verification experiments identified enriched NETs, as well as damaged intestinal epithelium and BBB permeability, in the colon and prefrontal cortex of colitis mice and APP/PS1 mice. By employing pharmacological interventions (Cl-amidine and Disulfiram), we disrupted the formation of NETs and discovered significantly restored barrier integrity and attenuated inflammation. Further enrichment and correlation analysis indicated, for the first time, DDIT4/IL-1β NETs might drive macrophage-mediated phagocytosis to induce barrier dysfunction in UC and AD. Our findings originally established the peripheral-central inflammation interactions of UC and AD from the perspective of NETs, highlighting the potential valuable roles in gut-brain interactions and future clinic translational therapeutics.
Collapse
Affiliation(s)
- Si-Hui Li
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China
| | - Qian-Hui Huang
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China
| | - Qing-Qing Yang
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China
| | - Qin Huang
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China
| | - De-Xian Wang
- College of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China
| | - Jiao Yang
- Suining Municipal Hospital of Traditional Chinese Medicine, Suining, Sichuan 629000, China
| | - Si-Han Huang
- Basic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China
| | - Si-Yu Zhang
- Basic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China
| | - Jun-Meng Wang
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China
| | - Lu-Shuang Xie
- Basic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China
| | - Shu-Guang Yu
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China.
| | - Qiao-Feng Wu
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China; Key Laboratory of Acupuncture for Senile Disease (Chengdu University of Traditional Chinese Medicine), Ministry of Education, Chengdu, Sichuan 610075, China; Institute of Acupuncture and Homeostasis Regulation, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China.
| |
Collapse
|
6
|
Shafqat A, Li M, Zakirullah, Liu F, Tong Y, Fan J, Fan H. A comprehensive review of research advances in the study of lactoferrin to treat viral infections. Life Sci 2025; 361:123340. [PMID: 39730037 DOI: 10.1016/j.lfs.2024.123340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Lactoferrin (Lf) is a naturally occurring glycoprotein known for its antiviral and antibacterial properties and is present in various physiological fluids. Numerous studies have demonstrated its antiviral effectiveness against multiple viruses, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), influenza virus (IFV), herpes simplex virus (HSV), hepatitis B virus (HBV), and human immunodeficiency virus (HIV). Lf, a vital component of the mucosal defense system, plays a crucial role in inhibiting viral infection by binding to both host cells and viral particles, such as the Hepatitis C virus (HCV). This interaction enables Lf to keep viral particles away from their target cells, emphasizing its significance as a fundamental element of mucosal defense against viral infections. Additionally, Lf has the ability to modulate cytokine expression and enhance cellular immune responses. In the innate immune system, Lf serves as a unique iron transporter and helps suppress various pathogens like bacteria, fungi, and viruses. This article summarises the potential antiviral properties of Lf against various viruses, along with its other mentioned functions. The advancement of Lf-based therapies supports the homology of food and medicine, providing a promising avenue to address viral infections and other public health challenges.
Collapse
Affiliation(s)
- Amna Shafqat
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Maochen Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Zakirullah
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Feitong Liu
- H&H Group, H&H Research, China Research and Innovation, Guangzhou, China.
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China.
| | - Junfen Fan
- Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.
| | - Huahao Fan
- School of Life Sciences, Tianjin University, Tianjin, China.
| |
Collapse
|
7
|
Kiilerich KF, Andresen T, Darbani B, Gregersen LHK, Liljensøe A, Bennike TB, Holm R, Moeller JB, Andersen V. Advancing Inflammatory Bowel Disease Treatment by Targeting the Innate Immune System and Precision Drug Delivery. Int J Mol Sci 2025; 26:575. [PMID: 39859291 PMCID: PMC11765494 DOI: 10.3390/ijms26020575] [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: 11/13/2024] [Revised: 01/04/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Inflammatory bowel disease (IBD), encompassing Crohn's disease and ulcerative colitis, involves chronic inflammation of the gastrointestinal tract. Current immune-modulating therapies are insufficient for 30-50% of patients or cause significant side effects, emphasizing the need for new treatments. Targeting the innate immune system and enhancing drug delivery to inflamed gut regions are promising strategies. Neutrophils play a central role in IBD by releasing reactive oxygen species (ROS) and neutrophil extracellular traps (NETs) -DNA-based structures with cytotoxic proteins-that contribute to mucosal damage and inflammation. Recent studies linking ROS production, DNA repair, and NET formation have identified NETs as potential therapeutic targets, with preclinical models showing positive outcomes from NET inhibition. Innovative oral drug delivery systems designed to target gut inflammation directly-without systemic absorption-could improve treatment precision and reduce side effects. Advanced formulations utilize properties such as particle size, surface modifications, and ROS-triggered release to selectively target the distal ileum and colon. A dual strategy that combines a deeper understanding of IBD pathophysiology to identify inflammation-related therapeutic targets with advanced drug delivery systems may offer significant promise. For instance, pairing NET inhibition with ROS-responsive nanocarriers could enhance treatment efficacy, though further research is needed. This synergistic approach has the potential to greatly improve outcomes for IBD patients.
Collapse
Affiliation(s)
- Kat F. Kiilerich
- Department of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (K.F.K.); (J.B.M.)
| | - Trine Andresen
- Department of Health Science and Technology, The Faculty of Medicine, Aalborg University, 9220 Aalborg Ø, Denmark; (T.A.); (T.B.B.)
| | - Behrooz Darbani
- Molecular Diagnostic and Clinical Research Unit, University Hospital of Southern Denmark, 6200 Aabenraa, Denmark; (B.D.); (L.H.K.G.); (A.L.)
| | - Laura H. K. Gregersen
- Molecular Diagnostic and Clinical Research Unit, University Hospital of Southern Denmark, 6200 Aabenraa, Denmark; (B.D.); (L.H.K.G.); (A.L.)
- Department of Regional Health Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Anette Liljensøe
- Molecular Diagnostic and Clinical Research Unit, University Hospital of Southern Denmark, 6200 Aabenraa, Denmark; (B.D.); (L.H.K.G.); (A.L.)
| | - Tue B. Bennike
- Department of Health Science and Technology, The Faculty of Medicine, Aalborg University, 9220 Aalborg Ø, Denmark; (T.A.); (T.B.B.)
- Molecular Diagnostic and Clinical Research Unit, University Hospital of Southern Denmark, 6200 Aabenraa, Denmark; (B.D.); (L.H.K.G.); (A.L.)
| | - René Holm
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, 5000 Odense, Denmark;
| | - Jesper B. Moeller
- Department of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (K.F.K.); (J.B.M.)
- Danish Institute for Advanced Study, University of Southern Denmark, 5000 Odense, Denmark
| | - Vibeke Andersen
- Molecular Diagnostic and Clinical Research Unit, University Hospital of Southern Denmark, 6200 Aabenraa, Denmark; (B.D.); (L.H.K.G.); (A.L.)
- Department of Regional Health Research, University of Southern Denmark, 5000 Odense, Denmark
| |
Collapse
|
8
|
Sun T, Wang P, Zhai X, Wang Z, Miao X, Yang Y, Wu J. Neutrophil extracellular traps induce barrier dysfunction in DSS-induced ulcerative colitis via the cGAS-STING pathway. Int Immunopharmacol 2024; 143:113358. [PMID: 39388893 DOI: 10.1016/j.intimp.2024.113358] [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: 07/20/2024] [Revised: 09/20/2024] [Accepted: 10/05/2024] [Indexed: 10/12/2024]
Abstract
Peptidyl arginine deiminase 4 (PAD4)-mediated neutrophil extracellular traps (NETs) play a crucial role in the pathogenesis of ulcerative colitis (UC). The cGAS-STING intracellular DNA-sensing pathway has been recognized as a pivotal mediator of inflammation. This study aimed to explore how NETs contribute to intestinal inflammation and barrier dysfunction in UC, focusing on the cGAS-STING pathway. We observed a significant increase of STING expression in UC mouse colons, which was mitigated by blocking NET formation through PAD4 genetic knockout. Moreover, NETs were discovered to activate the cGAS-STING pathway in MC38 cells in a dose and time-dependent manner, leading to the secretion of inflammatory cytokines and impaired barrier function. Additionally, STING deficiency ameliorated the clinical colitis index, intestinal inflammation, and barrier dysfunction. These findings underscore the involvement of cGAS-STING in regulating NET-mediated intestinal inflammation, suggesting its potential as a novel therapeutic target for UC.
Collapse
Affiliation(s)
- Tao Sun
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Ping Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xinru Zhai
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zhiwei Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xinyu Miao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yang Yang
- Department of Gastroenterology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China.
| | - Jie Wu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
9
|
Shukrun R, Fidel V, Baron S, Unger N, Ben-Shahar Y, Cohen S, Elhasid R, Yerushalmy-Feler A. Neutrophil Extracellular Traps in Pediatric Inflammatory Bowel Disease: A Potential Role in Ulcerative Colitis. Int J Mol Sci 2024; 25:11126. [PMID: 39456908 PMCID: PMC11507660 DOI: 10.3390/ijms252011126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Inflammatory bowel disease (IBD), encompassing Crohn's disease (CD) and ulcerative colitis (UC), is a chronic inflammatory condition of the gut affecting both adults and children. Neutrophil extracellular traps (NETs) are structures released by activated neutrophils, potentially contributing to tissue damage in various diseases. This study aimed to explore the presence and role of NETs in pediatric IBD. We compared intestinal biopsies and peripheral blood from 20 pediatric IBD patients (UC and CD) to controls. Biopsy staining and techniques for neutrophil activation were used to assess neutrophil infiltration and NET formation. We also measured the enzymatic activity of key NET proteins and evaluated NET formation in UC patients in remission. Both UC and CD biopsies showed significantly higher levels of neutrophils and NETs compared to controls (p < 0.01), with UC exhibiting the strongest association. Peripheral blood neutrophils from UC patients at diagnosis displayed increased NET formation compared to controls and CD patients. Interestingly, NET formation normalized in UC patients following remission-inducing treatment. This pilot study suggests a potential role for NETs in pediatric IBD, particularly UC. These findings warrant further investigation into the mechanisms of NET involvement and the potential for targeting NET formation as a therapeutic strategy.
Collapse
Affiliation(s)
- Rachel Shukrun
- Pediatric Hemato-Oncology Research Laboratory, Tel Aviv Medical Center, Tel Aviv 6423906, Israel; (R.S.); (V.F.); (S.B.)
- Department of Pediatric Hemato-Oncology, “Dana-Dwek” Children’s Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (N.U.); (S.C.); (A.Y.-F.)
| | - Victoria Fidel
- Pediatric Hemato-Oncology Research Laboratory, Tel Aviv Medical Center, Tel Aviv 6423906, Israel; (R.S.); (V.F.); (S.B.)
| | - Szilvia Baron
- Pediatric Hemato-Oncology Research Laboratory, Tel Aviv Medical Center, Tel Aviv 6423906, Israel; (R.S.); (V.F.); (S.B.)
| | - Noga Unger
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (N.U.); (S.C.); (A.Y.-F.)
| | - Yoav Ben-Shahar
- Department of Pediatric Surgery, “Dana-Dwek” Children’s Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel;
| | - Shlomi Cohen
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (N.U.); (S.C.); (A.Y.-F.)
- Pediatric Gastroenterology Institute, “Dana-Dwek” Children’s Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Ronit Elhasid
- Department of Pediatric Hemato-Oncology, “Dana-Dwek” Children’s Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Anat Yerushalmy-Feler
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (N.U.); (S.C.); (A.Y.-F.)
- Pediatric Gastroenterology Institute, “Dana-Dwek” Children’s Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| |
Collapse
|
10
|
Long D, Mao C, Xu Y, Zhu Y. The emerging role of neutrophil extracellular traps in ulcerative colitis. Front Immunol 2024; 15:1425251. [PMID: 39170617 PMCID: PMC11335521 DOI: 10.3389/fimmu.2024.1425251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/22/2024] [Indexed: 08/23/2024] Open
Abstract
Ulcerative colitis (UC) is characterized by chronic non-recessive inflammation of the intestinal mucosa involving both innate and adaptive immune responses. Currently, new targeted therapies are urgently needed for UC, and neutrophil extracellular traps (NETs) are new therapeutic options. NETs are DNA-based networks released from neutrophils into the extracellular space after stimulation, in which a variety of granule proteins, proteolytic enzymes, antibacterial peptides, histones, and other network structures are embedded. With the deepening of the studies on NETs, their regulatory role in the development of autoimmune and autoinflammatory diseases has received extensive attention in recent years. Increasing evidence indicates that excess NETs exacerbate the inflammatory response in UC, disrupting the structure and function of the intestinal mucosal barrier and increasing the risk of thrombosis. Although NETs are usually assigned a deleterious role in promoting the pathological process of UC, they also appear to have a protective role in some models. Despite such progress, comprehensive reviews describing the therapeutic promise of NETs in UC remain limited. In this review, we discuss the latest evidence for the formation and degradation of NETs, focusing on their double-edged role in UC. Finally, the potential implications of NETs as therapeutic targets for UC will be discussed. This review aims to provide novel insights into the pathogenesis and therapeutic options for UC.
Collapse
Affiliation(s)
- Dan Long
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chenhan Mao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yin Xu
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ying Zhu
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
11
|
Chen L, Ai F, Wu X, Yu W, Jin X, Ma J, Xiang B, Shen S, Li X. Analysis of neutrophil extracellular trap-related genes in Crohn's disease based on bioinformatics. J Cell Mol Med 2024; 28:e70013. [PMID: 39199011 PMCID: PMC11358036 DOI: 10.1111/jcmm.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 06/28/2024] [Accepted: 07/25/2024] [Indexed: 09/01/2024] Open
Abstract
Crohn's disease (CD) presents with diverse clinical phenotypes due to persistent inflammation of the gastrointestinal tract. Its global incidence is on the rise. Neutrophil extracellular traps (NETs) are networks released by neutrophils that capture microbicidal proteins and oxidases targeting pathogens. Research has shown that NETs are implicated in the pathogenesis of several immune-mediated diseases such as rheumatoid arthritis, systemic lupus erythematosus and inflammatory bowel disease. The goal of this study was to identify a panel of NET-related genes to construct a diagnostic and therapeutic model for CD. Through analysis of the GEO database, we identified 1950 differentially expressed genes (DEGs) associated with CD. Gene enrichment and immune cell infiltration analyses indicate that neutrophil infiltrates and chemokine-related pathways are predominantly involved in CD, with other immune cells such as CD4 and M1 macrophages also playing a role in disease progression. Utilizing weighted gene co-expression network analysis (WGCNA) and protein-protein interaction (PPI) networks, we identified six hub genes (SPP1, SOCS3, TIMP1, IRF1, CXCL2 and CD274). To validate the accuracy of our model, we performed external validation with statistical differences(p < 0.05). Additionally, immunohistochemical experiments demonstrated higher protein expression of the hub genes in colonic tissues from CD patients compared to healthy subjects (p < 0.05). In summary, we identified six effective hub genes associated with NETs as potential diagnostic markers for CD. These markers not only offer targets for future research but also hold promise for the development of novel therapeutic interventions for CD.
Collapse
Affiliation(s)
- Libin Chen
- Department of GastroenterologyThe Third Xiangya Hospital of Central South UniversityChangshaChina
- Hunan Key Laboratory of Nonresolving Inflammation and CancerThe Third Xiangya Hospital of Central South UniversityChangshaChina
| | - Feiyan Ai
- Department of GastroenterologyThe Third Xiangya Hospital of Central South UniversityChangshaChina
- Hunan Key Laboratory of Nonresolving Inflammation and CancerThe Third Xiangya Hospital of Central South UniversityChangshaChina
| | - Xing Wu
- Department of GastroenterologyThe Third Xiangya Hospital of Central South UniversityChangshaChina
- Hunan Key Laboratory of Nonresolving Inflammation and CancerThe Third Xiangya Hospital of Central South UniversityChangshaChina
| | - Wentao Yu
- Department of Pathology, The Third Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xintong Jin
- Department of GastroenterologyThe Third Xiangya Hospital of Central South UniversityChangshaChina
- Hunan Key Laboratory of Nonresolving Inflammation and CancerThe Third Xiangya Hospital of Central South UniversityChangshaChina
| | - Jian Ma
- Hunan Key Laboratory of Nonresolving Inflammation and CancerThe Third Xiangya Hospital of Central South UniversityChangshaChina
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical SciencesCentral South UniversityChangshaChina
| | - Bo Xiang
- Hunan Key Laboratory of Nonresolving Inflammation and CancerThe Third Xiangya Hospital of Central South UniversityChangshaChina
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical SciencesCentral South UniversityChangshaChina
| | - Shourong Shen
- Department of GastroenterologyThe Third Xiangya Hospital of Central South UniversityChangshaChina
- Hunan Key Laboratory of Nonresolving Inflammation and CancerThe Third Xiangya Hospital of Central South UniversityChangshaChina
| | - Xiayu Li
- Department of GastroenterologyThe Third Xiangya Hospital of Central South UniversityChangshaChina
- Hunan Key Laboratory of Nonresolving Inflammation and CancerThe Third Xiangya Hospital of Central South UniversityChangshaChina
| |
Collapse
|
12
|
Tang W, Ma J, Chen K, Wang K, Chen Z, Chen C, Li X, Wang Y, Shu Y, Zhang W, Yuan X, Shi G, Chen T, Wang P, Chen Y. Berbamine ameliorates DSS-induced colitis by inhibiting peptidyl-arginine deiminase 4-dependent neutrophil extracellular traps formation. Eur J Pharmacol 2024; 975:176634. [PMID: 38710356 DOI: 10.1016/j.ejphar.2024.176634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 03/29/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease with immune dysregulation affecting colon inflammatory response. Recent studies have highlighted that neutrophil extracellular traps (NETs) play an important role in the pathogenesis of UC. Berbamine (BBM), one of the bioactive ingredients extracted from Chinese herbal medicine Berberis vulgaris L, has attracted intensive attentions due to its significant anti-inflammatory activity and a marketing drug for treating leukemia in China. However, the exact role and potential molecular mechanism of BBM against UC remains elusive. In the present study, our results showed that BBM could markedly improve the pathological phenotype and the colon inflammation in mice with dextran sulfate sodium (DSS)-induced colitis. Then, comprehensive approaches combining network pharmacology and molecular docking analyses were employed to predict the therapeutic potential of BBM in treating UC by peptidyl-arginine deiminase 4 (PAD4), a crucial molecule involved in NETs formation. The molecular docking results showed BBM had a high affinity for PAD4 with a binding energy of -9.3 kcal/mol Moreover, PAD4 expression and NETs productions, including citrullination of histone H3 (Cit-H3), neutrophil elastase (NE), myeloperoxidase (MPO) in both neutrophils and colonic tissue were reduced after BBM administration. However, in the mice with DSS-induced colitis pretreated with GSK484, a PAD4-specific inhibitor, BBM could not further reduce disease related indexes, expression of PAD4 and NETs productions. Above all, the identification of PAD4 as a potential target for BBM to inhibit NETs formation in colitis provides novel insights into the development of BBM-derived drugs for the clinical management of UC.
Collapse
Affiliation(s)
- Wenwen Tang
- Department of Colorectal Surgery, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China; Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
| | - Jiaze Ma
- Department of Colorectal Surgery, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China; Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
| | - Kaidi Chen
- Department of Colorectal Surgery, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China; Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
| | - Kuiling Wang
- Department of Colorectal Surgery, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China; Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
| | - Zepeng Chen
- Department of Colorectal Surgery, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China; Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
| | - Chen Chen
- Department of Colorectal Surgery, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China; Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
| | - Xun Li
- Institute for Molecular Bioscience, the University of Queensland, Brisbane, 4702, Australia
| | - Yuji Wang
- Department of Colorectal Surgery, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China; Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
| | - Yi Shu
- Department of Colorectal Surgery, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China; Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
| | - Wei Zhang
- Department of Colorectal Surgery, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China; Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
| | - Xiaomin Yuan
- Department of Colorectal Surgery, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China; Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
| | - Guoping Shi
- Department of Colorectal Surgery, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China; Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
| | - Tuo Chen
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China; Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China; Department of Orthopedics, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, PR China; Institute for Molecular Bioscience, the University of Queensland, Brisbane, 4702, Australia.
| | - Peimin Wang
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China; Department of Orthopedics, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, PR China.
| | - Yugen Chen
- Department of Colorectal Surgery, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China; Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China; Jiangsu Collaborative Innovation Center of Chinese Medicine in Prevention and Treatment of Tumor, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China.
| |
Collapse
|
13
|
Zhu J, Wu Y, Ge X, Chen X, Mei Q. Discovery and Validation of Ferroptosis-Associated Genes of Ulcerative Colitis. J Inflamm Res 2024; 17:4467-4482. [PMID: 39006497 PMCID: PMC11246036 DOI: 10.2147/jir.s463042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Background Ulcerative colitis (UC) is a long-lasting idiopathic condition, but its precise mechanisms remain unclear. Meanwhile, evidence has demonstrated that ferroptosis seems to interlock with the progress of UC. This research sought to identify hub genes of UC related to ferroptosis. Methods First, the relevant profiles for this article were obtained from GEO database. From the FerrDb, 479 genes linked to ferroptosis were retrieved. Using analysis of the difference and WGCNA on colonic samples from GSE73661, the remaining six hub genes linked to ferroptosis and UC were discovered. Through logistic regression analyses, the diagnostic model was constructed and was then evaluated by external validation using dataset GSE92415. Afterwards, the correlation between immune cell filtration in UC and hub genes was examined. Finally, a mice model of colitis was established, and the results were verified using qRT-PCR. Results We acquired six hub genes linked to ferroptosis and UC. In order to create a diagnostic model for UC, we used logistic regression analysis to screen three of the six ferroptosis related genes (HIF1A, SLC7A11, and LPIN1). The ROC curve showed that the three hub genes had outstanding potential for disease diagnosis (AUC = 0.976), which was subsequently validated in samples from GSE92415 (AUC = 0.962) and blood samples from GSE3365 (AUC = 0.847) and GSE94648 (AUC = 0.769). These genes might be crucial for UC immunity based upon the results on the immune system. Furthermore, mouse samples examined using qRT-PCR also verified our findings. Conclusion In conclusion, the findings have important implications for ferroptosis and UC, and these hub genes may also offer fresh perspectives on the aetiology and therapeutic approaches of UC.
Collapse
Affiliation(s)
- Jiejie Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, People's Republic of China
- Key Laboratory of Digestive Diseases of Anhui Province, Hefei, People's Republic of China
| | - Yumei Wu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, People's Republic of China
- Key Laboratory of Digestive Diseases of Anhui Province, Hefei, People's Republic of China
| | - Xiaoyuan Ge
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, People's Republic of China
- Key Laboratory of Digestive Diseases of Anhui Province, Hefei, People's Republic of China
| | - Xinwen Chen
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, People's Republic of China
- Key Laboratory of Digestive Diseases of Anhui Province, Hefei, People's Republic of China
| | - Qiao Mei
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, People's Republic of China
- Key Laboratory of Digestive Diseases of Anhui Province, Hefei, People's Republic of China
| |
Collapse
|
14
|
Wang S, Song Y, Wang Z, Chang X, Wu H, Yan Z, Wu J, He Z, Kang L, Hu W, Xia T, Li Z, Ren X, Bai Y. Neutrophil-derived PAD4 induces citrullination of CKMT1 exacerbates mucosal inflammation in inflammatory bowel disease. Cell Mol Immunol 2024; 21:620-633. [PMID: 38720063 PMCID: PMC11143373 DOI: 10.1038/s41423-024-01158-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 03/21/2024] [Indexed: 06/02/2024] Open
Abstract
Peptidyl arginine deiminase 4 (PAD4) plays a pivotal role in infection and inflammatory diseases by facilitating the formation of neutrophil extracellular traps (NETs). However, the substrates of PAD4 and its exact role in inflammatory bowel disease (IBD) remain unclear. In this study, we employed single-cell RNA sequencing (scRNA-seq) and substrate citrullination mapping to decipher the role of PAD4 in intestinal inflammation associated with IBD. Our results demonstrated that PAD4 deficiency alleviated colonic inflammation and restored intestinal barrier function in a dextran sulfate sodium (DSS)-induced colitis mouse model. scRNA-seq analysis revealed significant alterations in intestinal cell populations, with reduced neutrophil numbers and changes in epithelial subsets upon PAD4 deletion. Gene expression analysis highlighted pathways related to inflammation and epithelial cell function. Furthermore, we found that neutrophil-derived extracellular vesicles (EVs) carrying PAD4 were secreted into intestinal epithelial cells (IECs). Within IECs, PAD4 citrullinates mitochondrial creatine kinase 1 (CKMT1) at the R242 site, leading to reduced CKMT1 protein stability via the autophagy pathway. This action compromises mitochondrial homeostasis, impairs intestinal barrier integrity, and induces IECs apoptosis. IEC-specific depletion of CKMT1 exacerbated intestinal inflammation and apoptosis in mice with colitis. Clinical analysis of IBD patients revealed elevated levels of PAD4, increased CKMT1 citrullination, and decreased CKMT1 expression. In summary, our findings highlight the crucial role of PAD4 in IBD, where it modulates IECs plasticity via CKMT1 citrullination, suggesting that PAD4 may be a potential therapeutic target for IBD.
Collapse
Affiliation(s)
- Shuling Wang
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- National Key Laboratory of Immunology and Inflammation, Naval Medical University, Shanghai, 200433, China
- Changhai Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Yihang Song
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- National Key Laboratory of Immunology and Inflammation, Naval Medical University, Shanghai, 200433, China
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Zhijie Wang
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Xin Chang
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- National Key Laboratory of Immunology and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Haicong Wu
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- National Key Laboratory of Immunology and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Ziwei Yan
- National Key Laboratory of Immunology and Inflammation, Naval Medical University, Shanghai, 200433, China
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Jiayi Wu
- National Key Laboratory of Immunology and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Zixuan He
- Changhai Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Le Kang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Wenjun Hu
- National Key Laboratory of Immunology and Inflammation, Naval Medical University, Shanghai, 200433, China
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Tian Xia
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Changhai Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Zhaoshen Li
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
- National Key Laboratory of Immunology and Inflammation, Naval Medical University, Shanghai, 200433, China.
- Changhai Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Xingxing Ren
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Yu Bai
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
- National Key Laboratory of Immunology and Inflammation, Naval Medical University, Shanghai, 200433, China.
- Changhai Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
15
|
Majumder A, Bano S. How the Western Diet Thwarts the Epigenetic Efforts of Gut Microbes in Ulcerative Colitis and Its Association with Colorectal Cancer. Biomolecules 2024; 14:633. [PMID: 38927037 PMCID: PMC11201633 DOI: 10.3390/biom14060633] [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: 05/04/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Ulcerative colitis (UC) is an autoimmune disease in which the immune system attacks the colon, leading to ulcer development, loss of colon function, and bloody diarrhea. The human gut ecosystem consists of almost 2000 different species of bacteria, forming a bioreactor fueled by dietary micronutrients to produce bioreactive compounds, which are absorbed by our body and signal to distant organs. Studies have shown that the Western diet, with fewer short-chain fatty acids (SCFAs), can alter the gut microbiome composition and cause the host's epigenetic reprogramming. Additionally, overproduction of H2S from the gut microbiome due to changes in diet patterns can further activate pro-inflammatory signaling pathways in UC. This review discusses how the Western diet affects the microbiome's function and alters the host's physiological homeostasis and susceptibility to UC. This article also covers the epidemiology, prognosis, pathophysiology, and current treatment strategies for UC, and how they are linked to colorectal cancer.
Collapse
Affiliation(s)
- Avisek Majumder
- Department of Medicine, University of California, San Francisco, CA 94158, USA
| | | |
Collapse
|
16
|
Bai M, Li S, Zhang C, An N, Wang J, Qin J, Jia R, Liu W, Cheng J, Wu X, Xu Q. Suppression of neutrophil extracellular traps is responsible for the amelioration of chemotherapeutic intestinal injury by the natural compound PEITC. Toxicol Appl Pharmacol 2024; 484:116857. [PMID: 38341106 DOI: 10.1016/j.taap.2024.116857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/25/2024] [Accepted: 02/07/2024] [Indexed: 02/12/2024]
Abstract
Intestinal injury is one of the most debilitating side effects of many chemotherapeutic agents, such as irinotecan hydrochloride (CPT-11). Accumulating evidence indicates that neutrophil extracellular traps (NETs) play a critical role in the symptoms of ischemia and inflammation related to chemotherapy. The present study investigated the effects and possible mechanisms of phenethyl isothiocyanate (PEITC) in inhibiting NETs and alleviating chemotherapeutic intestinal injury. CPT-11 induced robust neutrophil activation, as evidenced by increased NETs release, intestinal ischemia, and mRNA expression of inflammatory factors. PEITC prolonged the clotting time of chemotherapeutic mice, improved the intestinal microcirculation, inhibited the expression of inflammatory factors, and protected the tight junctions of the intestinal epithelium. Both in vivo and in vitro experiments revealed that PEITC directly suppresses CPT-11-induced NETs damage to intestinal cells, resulting in significant attenuation of epithelial injury. These results suggest that PEITC may be a novel agent to relieve chemotherapeutic intestinal injury via inhibition of NETs.
Collapse
Affiliation(s)
- Mei Bai
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Shuaifei Li
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Cui Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Ning An
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Jie Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China; School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Jia Qin
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Rumeng Jia
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Wentao Liu
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Jingcai Cheng
- Drug R&D Institute, JC (Wuxi) COMPANY, Inc, Wuxi, China
| | - Xuefeng Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China.
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China.
| |
Collapse
|
17
|
Shao Y, Li L, Yang Y, Ye Y, Guo Z, Liu L, Huang J, Chen Y, Gao X, Sun B. DNase aggravates intestinal microvascular injury in IBD patients by releasing NET-related proteins. FASEB J 2024; 38:e23395. [PMID: 38149880 DOI: 10.1096/fj.202301780r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 12/02/2023] [Accepted: 12/15/2023] [Indexed: 12/28/2023]
Abstract
Neutrophils accumulate in the inflammatory mucosa of patients with inflammatory bowel disease (IBD), and excessive release of NETs (neutrophil extracellular traps may be one of the important factors that cause IBD progression. However, the specific mechanism underlying vascular injury caused by NETs remains unclear. Immunofluorescence, ELISA, and flow cytometry were used in this study to detect the expression of NETs and DNase in the tissue and peripheral blood samples of patients with IBD. DSS mouse model was used to detect colon injury and vascular permeability. We found that NETs and DNase levels increased in the colon of patients with IBD. We found an increase in the activity of NET-related MPO released by DNase. DNase released NET-related proteins and damaged vascular endothelial cells in vitro. In DSS mouse model, the synchronous increase of DNase and NETs in the colon leads to an increase in vascular injury markers (CD44, sTM). DNase aggravated colon injury and increased vascular permeability in vivo, which was inhibited by gentamicin sulfate (GS). GS does not reduce the expression of DNase, but rather reduces the release of NET-related proteins to protect vascular endothelium by inhibiting DNase activity. MPO and histones synergistically damaged the vascular endothelium, and vascular injury can be improved by their active inhibitors. We further found that H2 O2 is an important substrate for MPO induced vascular damage. In conclusion, in IBD, DNase, and NET levels increased synchronously in the lesion area and released NET-related proteins to damage the vascular endothelium. Therefore, targeting DNase may be beneficial for the treatment of IBD.
Collapse
Affiliation(s)
- Yiming Shao
- Department of Burns and Plastic Surgery, Affiliated Hospital of Jining Medical University, Jining, China
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Linbin Li
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Yunxi Yang
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Yulan Ye
- Department of Gastroenterology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Zaiwen Guo
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Lu Liu
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jiamin Huang
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Yi Chen
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Xi Gao
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Bingwei Sun
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| |
Collapse
|
18
|
Küçük İ, Özçelik F, Yazgan Y, Yılmaz İ, Kaplan M, Yıldırım İ. Can Serum Histone H4 Level Be a Biomarker in Ulcerative Colitis? THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2024; 35:4-10. [PMID: 38454272 PMCID: PMC10837591 DOI: 10.5152/tjg.2024.22385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 04/13/2023] [Indexed: 03/09/2024]
Abstract
BACKGROUND/AIMS Histones are a part of neutrophil extracellular trap molecules which were reported to have diagnostic values in some inflammatory diseases. We aimed to evaluate whether serum histone H4 can be a diagnostic and prognostic marker for ulcerative colitis. MATERIALS AND METHODS This case-control study included 58 ulcerative colitis patients (34 males and 24 females) and 45 healthy controls (25 males and 20 females). The Mayo clinical scoring system was used for the clinical and endoscopic features. Truelove-Witt's method was applied to the histology activity index. The human histone H4 kit was used for the enzyme-linked immunosorbent assay of serum histone H4. RESULTS Serum histone H4 was significantly lower in the ulcerative colitis group compared to the control groups [268 (14-1639) vs. 598 (310-2134) ng/L, P < .001, respectively]. Among the ulcerative colitis patients, there was no correlation between serum histone H4 and disease extent, Mayo clinical scoring, Mayo endoscopic activity subscoring, histology activity index, inflammatory markers, d-dimer, and leukocyte and neutrophil counts (r < 0.20, P > .05). Histone H4 levels were not statistically significant between the patients with no medication and those taking 5-aminosalicylate and/or other agents (P > .05). The receiver operating characteristic curve analysis revealed that serum histone H4 concentrations had a 0.782 (95%CI: 0.690-0.857, P < .001) diagnostic accuracy for ulcerative colitis. The specificity and sensitivity for the cutoff level of ≤364 ng/L were 88.9% and 72.4%, respectively. CONCLUSION Decreased serum histone H4 values may be used as an auxiliary marker in the progression and diagnosis of ulcerative colitis. Further studies are needed to delineate this relationship between clinical and laboratory traits of ulcerative colitis and serum histone H4.
Collapse
Affiliation(s)
- İrfan Küçük
- Department of Gastroenterology, University of Health Sciences Sultan 2. Abdulhamid Han Training and Research Hospital, İstanbul, Turkey
| | - Fatih Özçelik
- Department of Medical Biochemistry, University of Health Sciences, Ümraniye Training and Research Hospital, İstanbul, Turkey
| | - Yusuf Yazgan
- Department of Pathology, University of Health Sciences Sultan 2. Abdulhamid Han Training and Research Hospital, İstanbul, Turkey
| | - İsmail Yılmaz
- Department of Pathology, University of Health Sciences Sultan 2. Abdulhamid Han Training and Research Hospital, İstanbul, Turkey
| | - Mustafa Kaplan
- Department of Internal Medicine, University of Health Sciences Sultan 2. Abdulhamid Han Training and Research Hospital, İstanbul, Turkey
| | - İdris Yıldırım
- Department of Pathology, University of Health Sciences Sultan 2. Abdulhamid Han Training and Research Hospital, İstanbul, Turkey
| |
Collapse
|
19
|
Pehrsson M, Domislovic V, Alexdottir MS, Brinar M, Karsdal MA, Barisic A, Krznaric Z, Mortensen JH. Blood-Based Biomarkers Reflecting Protease 3 and MMP-12 Catalyzed Elastin Degradation as Potential Noninvasive Surrogate Markers of Endoscopic and Clinical Disease in Inflammatory Bowel Disease. J Clin Med 2023; 13:21. [PMID: 38202027 PMCID: PMC10779348 DOI: 10.3390/jcm13010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
Chronic inflammation in inflammatory bowel disease (IBD) triggers significant extracellular matrix remodeling, including elastin remodeling, leading to severe clinical complications. Novel methods to assess intestinal tissue destruction may act as surrogate markers of endoscopic disease activity, relieving patients of invasive endoscopy. We explored the noninvasive blood-based biomarkers ELP-3 and ELM-12, measuring elastin degradation in IBD. In a study involving 104 Crohn's disease (CD), 39 ulcerative colitis (UC), and 29 healthy donors, we assessed these biomarkers' association with endoscopic and clinical disease activity using ELISA. Patients were evaluated based on the SES-CD and CDAI for CD patients and modified MES and partial Mayo for UC patients. ELP-3 and ELM-12 were elevated in patients with IBD. Discerning CD patients in endoscopic remission and mild from moderate to severe, ELP-3 provided an AUC of 0.69 and ELM-12 an AUC of 0.73. The ELP-3 biomarker was associated with UC patients and provided the highest diagnostic power of 0.87 for remission vs. active clinical disease. The data suggest an association of ELP-3 with active CD and ELM-12 with endoscopic remission in CD patients. Additionally, ELP-3 could identify UC patients with active clinical disease from patients in remission. The noninvasive biomarkers ELP-3 and ELM-12 could be potential surrogate biomarkers of elastin degradation and endoscopic and clinical disease markers.
Collapse
Affiliation(s)
- Martin Pehrsson
- Biomarkers and Research, Nordic Bioscience A/S, 2730 Herlev, Denmark; (M.S.A.); (M.A.K.); (J.H.M.)
| | - Viktor Domislovic
- Department of Gastroenterology and Hepatology, University Hospital Center Zagreb, 10000 Zagreb, Croatia; (V.D.); (M.B.); (Z.K.)
| | | | - Marko Brinar
- Department of Gastroenterology and Hepatology, University Hospital Center Zagreb, 10000 Zagreb, Croatia; (V.D.); (M.B.); (Z.K.)
| | - Morten Asser Karsdal
- Biomarkers and Research, Nordic Bioscience A/S, 2730 Herlev, Denmark; (M.S.A.); (M.A.K.); (J.H.M.)
| | - Ana Barisic
- Center for Clinical Nutrition, University Hospital Centre Zagreb, 10000 Zagreb, Croatia;
| | - Zeljko Krznaric
- Department of Gastroenterology and Hepatology, University Hospital Center Zagreb, 10000 Zagreb, Croatia; (V.D.); (M.B.); (Z.K.)
| | - Joachim Høg Mortensen
- Biomarkers and Research, Nordic Bioscience A/S, 2730 Herlev, Denmark; (M.S.A.); (M.A.K.); (J.H.M.)
| |
Collapse
|
20
|
Xu C, Ye Z, Jiang W, Wang S, Zhang H. Cyclosporine A alleviates colitis by inhibiting the formation of neutrophil extracellular traps via the regulating pentose phosphate pathway. Mol Med 2023; 29:169. [PMID: 38093197 PMCID: PMC10720086 DOI: 10.1186/s10020-023-00758-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 11/13/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND The aberrant formation of neutrophil extracellular traps (NETs) has been implicated in ulcerative colitis (UC), a chronic recurrent intestinal inflammation. Cyclosporine A (CsA) is now applied as rescue therapy for acute severe UC. In addition, it has been certained that CsA inhibits the formation of NETs in vitro and the mechanism of which was still vague. The study aimed to explore the mechanism CsA inhibits the NETs formation of colitis in vivo and in vitro. METHODS NETs enrichment in clinical samples was analyzed using databases from Gene Expression Omnibus and verified in our center. Dextran sulfate sodium (DSS)-induced acute colitis mice model was used to investigate the effect of CsA on NETs of colonic tissue expression. To clarify the mechanism, intracellular energy metabolites were examined by Liquid Chromatograph Mass Spectrometer, and reactive oxygen species (ROS) levels were examined by fluorescence intensity in neutrophils treated with CsA after LPS stimulation. The transcriptional level and activity of G6PD of neutrophils were also assessed using qRT-PCR and WST-8. RNA Sequencing was used to detect differentially expressed genes of neutrophils stimulated by LPS with or without CsA. The expression levels of related proteins were detected by western blot. RESULTS NETs enrichment was especially elevated in moderate-to-severe UC patients compared to HC. NETs expression in the colon from DSS colitis was decreased after CsA treatment. Compared with neutrophils stimulated by LPS, NETs formation and cellular ROS levels were decreased in LPS + CsA group. Cellular ribulose 5-phosphate and NADPH/NADP + related to the pentose phosphate pathway (PPP) were reduced in LPS + CsA group. In addition, CsA could decrease G6PD activity in neutrophils stimulated with LPS, and the results were further verified by inhibiting G6PD activity. At last, P53 protein was highly expressed in LPS + CsA group compared with the LPS group. Intracellular G6PD activity, ROS level and NETs formation, which were downregulated by CsA, could be reversed by a P53 inhibitor. CONCLUSION Our results indicated CsA could alleviate the severity of colitis by decreasing the formation of NETs in vivo. In vitro, CsA reduced ROS-dependent NETs release via downregulating PPP and cellular ROS levels by decreasing G6PD activity directly by activating the P53 protein.
Collapse
Affiliation(s)
- Chenjing Xu
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ziping Ye
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenyu Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shu Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hongjie Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
21
|
Jiang F, Wu M, Li R. The significance of long non-coding RNAs in the pathogenesis, diagnosis and treatment of inflammatory bowel disease. PRECISION CLINICAL MEDICINE 2023; 6:pbad031. [PMID: 38163004 PMCID: PMC10757071 DOI: 10.1093/pcmedi/pbad031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/06/2023] [Indexed: 01/03/2024] Open
Abstract
Inflammatory bowel diseases (IBD) are a group of chronic relapsing gastrointestinal inflammatory diseases with significant global incidence. Although the pathomechanism of IBD has been extensively investigated, several aspects of its pathogenesis remain unclear. Long non-coding RNAs (lncRNAs) are transcripts with more than 200 nucleotides in length that have potential protein-coding functions. LncRNAs play important roles in biological processes such as epigenetic modification, transcriptional regulation and post-transcriptional regulation. In this review, we summarize recent advances in research on IBD-related lncRNAs from the perspective of the overall intestinal microenvironment, as well as their potential roles as immune regulators, diagnostic biomarkers and therapeutic targets or agents for IBD.
Collapse
Affiliation(s)
- Fei Jiang
- Jiangsu Province Engineering Research Center of Cardiovascular Drugs Targeting Endothelial Cells, School of Life Sciences, Jiangsu Normal University, Xuzhou 221000, China
- Department of Laboratory Medicine, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Min Wu
- Drug Discovery Section, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
- Department of Neurology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Rongpeng Li
- Jiangsu Province Engineering Research Center of Cardiovascular Drugs Targeting Endothelial Cells, School of Life Sciences, Jiangsu Normal University, Xuzhou 221000, China
| |
Collapse
|
22
|
Illes Z, Jørgensen MM, Bæk R, Bente LM, Lauridsen JT, Hyrlov KH, Aboo C, Baumbach J, Kacprowski T, Cotton F, Guttmann CRG, Stensballe A. New Enhancing MRI Lesions Associate with IL-17, Neutrophil Degranulation and Integrin Microparticles: Multi-Omics Combined with Frequent MRI in Multiple Sclerosis. Biomedicines 2023; 11:3170. [PMID: 38137391 PMCID: PMC10740934 DOI: 10.3390/biomedicines11123170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/16/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Blood-barrier (BBB) breakdown and active inflammation are hallmarks of relapsing multiple sclerosis (RMS), but the molecular events contributing to the development of new lesions are not well explored. Leaky endothelial junctions are associated with increased production of endothelial-derived extracellular microvesicles (EVs) and result in the entry of circulating immune cells into the brain. MRI with intravenous gadolinium (Gd) can visualize acute blood-barrier disruption as the initial event of the evolution of new lesions. METHODS Here, weekly MRI with Gd was combined with proteomics, multiplex immunoassay, and endothelial stress-optimized EV array to identify early markers related to BBB disruption. Five patients with RMS with no disease-modifying treatment were monitored weekly using high-resolution 3T MRI scanning with intravenous gadolinium (Gd) for 8 weeks. Patients were then divided into three groups (low, medium, or high MRI activity) defined by the number of new, total, and maximally enhancing Gd-enhancing lesions and the number of new FLAIR lesions. Plasma samples taken at each MRI were analyzed for protein biomarkers of inflammation by quantitative proteomics, and cytokines using multiplex immunoassays. EVs were characterized with an optimized endothelial stress EV array based on exosome surface protein markers for the detection of soluble secreted EVs. RESULTS Proteomics analysis of plasma yielded quantitative information on 208 proteins at each patient time point (n = 40). We observed the highest number of unique dysregulated proteins (DEPs) and the highest functional enrichment in the low vs. high MRI activity comparison. Complement activation and complement/coagulation cascade were also strongly overrepresented in the low vs. high MRI activity comparison. Activation of the alternative complement pathway, pathways of blood coagulation, extracellular matrix organization, and the regulation of TLR and IGF transport were unique for the low vs. high MRI activity comparison as well, with these pathways being overrepresented in the patient with high MRI activity. Principal component analysis indicated the individuality of plasma profiles in patients. IL-17 was upregulated at all time points during 8 weeks in patients with high vs. low MRI activity. Hierarchical clustering of soluble markers in the plasma indicated that all four MRI outcomes clustered together with IL-17, IL-12p70, and IL-1β. MRI outcomes also showed clustering with EV markers CD62E/P, MIC A/B, ICAM-1, and CD42A. The combined cluster of these cytokines, EV markers, and MRI outcomes clustered also with IL-12p40 and IL-7. All four MRI outcomes correlated positively with levels of IL-17 (p < 0.001, respectively), and EV-ICAM-1 (p < 0.0003, respectively). IL-1β levels positively correlated with the number of new Gd-enhancing lesions (p < 0.01), new FLAIR lesions (p < 0.001), and total number of Gd-enhancing lesions (p < 0.05). IL-6 levels positively correlated with the number of new FLAIR lesions (p < 0.05). Random Forests and linear mixed models identified IL-17, CCL17/TARC, CCL3/MIP-1α, and TNF-α as composite biomarkers predicting new lesion evolution. CONCLUSIONS Combination of serial frequent MRI with proteome, neuroinflammation markers, and protein array data of EVs enabled assessment of temporal changes in inflammation and endothelial dysfunction in RMS related to the evolution of new and enhancing lesions. Particularly, the Th17 pathway and IL-1β clustered and correlated with new lesions and Gd enhancement, indicating their importance in BBB disruption and initiating acute brain inflammation in MS. In addition to the Th17 pathway, abundant protein changes between MRI activity groups suggested the role of EVs and the coagulation system along with innate immune responses including acute phase proteins, complement components, and neutrophil degranulation.
Collapse
Affiliation(s)
- Zsolt Illes
- Department of Neurology, Odense University Hospital, 5000 Odense, Denmark
- Department of Clinical Medicine, University of Southern Denmark, 5230 Odense, Denmark
- Institute of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
- Brain Research—Inter Disciplinary Guided Excellence (BRIDGE), University of Southern Denmark, 5230 Odense, Denmark
| | - Malene Møller Jørgensen
- Department of Clinical Immunology, Aalborg University Hospital, 9220 Aalborg, Denmark; (M.M.J.); (R.B.)
| | - Rikke Bæk
- Department of Clinical Immunology, Aalborg University Hospital, 9220 Aalborg, Denmark; (M.M.J.); (R.B.)
| | - Lisa-Marie Bente
- Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics of TU Braunschweig and Hannover Medical School, 38106 Braunschweig, Germany; (L.-M.B.); (T.K.)
- Braunschweig Integrated Centre for Systems Biology (BRICS), TU Braunschweig, 38106 Braunschweig, Germany
| | - Jørgen T. Lauridsen
- Department of Business and Economics, University of Southern Denmark, 5230 Odense, Denmark;
| | - Kirsten H. Hyrlov
- Department of Neurology, Odense University Hospital, 5000 Odense, Denmark
| | - Christopher Aboo
- Department of Health Science and Technology, Aalborg University, 9220 Aalborg, Denmark;
- Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, 101408 Beijing, China
| | - Jan Baumbach
- Department of Mathematics and Computer Science, University of Southern Denmark, 5230 Odense, Denmark;
- Institute for Computational Systems Biology, University of Hamburg, 20148 Hamburg, Germany
| | - Tim Kacprowski
- Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics of TU Braunschweig and Hannover Medical School, 38106 Braunschweig, Germany; (L.-M.B.); (T.K.)
- Braunschweig Integrated Centre for Systems Biology (BRICS), TU Braunschweig, 38106 Braunschweig, Germany
| | - Francois Cotton
- Service de Radiologie, Centre Hospitalier Lyon-Sud, France/CREATIS, Université de Lyon, 69007 Lyon, France;
| | | | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, 9220 Aalborg, Denmark;
- Clinical Cancer Center, Aalborg University Hospital, 9220 Aalborg, Denmark
| |
Collapse
|
23
|
Janker L, Schuster D, Bortel P, Hagn G, Meier-Menches SM, Mohr T, Mader JC, Slany A, Bileck A, Brunmair J, Madl C, Unger L, Hennlich B, Weitmayr B, Del Favero G, Pils D, Pukrop T, Pfisterer N, Feichtenschlager T, Gerner C. Multiomics-empowered Deep Phenotyping of Ulcerative Colitis Identifies Biomarker Signatures Reporting Functional Remission States. J Crohns Colitis 2023; 17:1514-1527. [PMID: 36961872 PMCID: PMC10588787 DOI: 10.1093/ecco-jcc/jjad052] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Indexed: 03/25/2023]
Abstract
INTRODUCTION Ulcerative colitis [UC] is a chronic disease with rising incidence and unclear aetiology. Deep molecular phenotyping by multiomics analyses may provide novel insights into disease processes and characteristic features of remission states. METHODS UC pathomechanisms were assessed by proteome profiling of human tissue specimens, obtained from five distinct colon locations for each of the 12 patients included in the study. Systemic disease-associated alterations were evaluated thanks to a cross-sectional setting of mass spectrometry-based multiomics analyses comprising proteins, metabolites, and eicosanoids of plasma obtained from UC patients during acute episodes and upon remission, in comparison with healthy controls. RESULTS Tissue proteome profiling indicated colitis-associated activation of neutrophils, macrophages, B and T cells, fibroblasts, endothelial cells and platelets, and hypoxic stress, and suggested a general downregulation of mitochondrial proteins accompanying the establishment of apparent wound healing-promoting activities including scar formation. Whereas pro-inflammatory proteins were apparently upregulated by immune cells, the colitis-associated epithelial cells, fibroblasts, endothelial cells, and platelets seemed to predominantly contribute anti-inflammatory and wound healing-promoting proteins. Blood plasma proteomics indicated chronic inflammation and platelet activation, whereas plasma metabolomics identified disease-associated deregulations of gut and gut microbiome-derived metabolites. Upon remission several, but not all, molecular candidate biomarker levels recovered back to normal. CONCLUSION The findings may indicate that microvascular damage and platelet deregulation hardly resolve upon remission, but apparently persist as disease-associated molecular signatures. This study presents local and systemic molecular alterations integrated in a model for UC pathomechanisms, potentially supporting the assessment of disease and remission states in UC patients.
Collapse
Affiliation(s)
- Lukas Janker
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Dina Schuster
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Patricia Bortel
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Gerhard Hagn
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Samuel M Meier-Menches
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Joint Metabolome Facility, University of Vienna, Vienna, Austria
| | - Thomas Mohr
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Joint Metabolome Facility, University of Vienna, Vienna, Austria
| | - Johanna C Mader
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Astrid Slany
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Andrea Bileck
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Joint Metabolome Facility, University of Vienna, Vienna, Austria
| | - Julia Brunmair
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Christian Madl
- Institute of Pathology and Microbiology, Krankenanstalt Rudolfstiftung, Vienna, Austria
| | - Lukas Unger
- Division of General Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Barbara Hennlich
- Institute of Pathology and Microbiology, Krankenanstalt Rudolfstiftung, Vienna, Austria
| | - Barbara Weitmayr
- Institute of Pathology and Microbiology, Krankenanstalt Rudolfstiftung, Vienna, Austria
| | - Giorgia Del Favero
- Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Dietmar Pils
- Department of Obstetrics and Gynaecology, Medical University of Vienna, Vienna, Austria
| | - Tobias Pukrop
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Nikolaus Pfisterer
- Institute of Pathology and Microbiology, Krankenanstalt Rudolfstiftung, Vienna, Austria
| | | | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Joint Metabolome Facility, University of Vienna, Vienna, Austria
| |
Collapse
|
24
|
Zhang D, Duan S, He Z, Zhu Z, Li Z, Yi Q, Cai T, Li J, Chen N, Guo S. Sijunzi Decoction Targets IL1B and TNF to Reduce Neutrophil Extracellular Traps (NETs) in Ulcerative Colitis: Evidence from Silicon Prediction and Experiment Validation. Drug Des Devel Ther 2023; 17:3103-3128. [PMID: 37868820 PMCID: PMC10590142 DOI: 10.2147/dddt.s428814] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/07/2023] [Indexed: 10/24/2023] Open
Abstract
Purpose This study was conducted to explore the mechanism of Sijunzi Decoction (SJZ) in the treatment of ulcerative colitis (UC). Methods The study aimed to investigate the active components and targets of SJZ in the treatment of UC by screening databases such as TCMSP, GeneCards, OMIM, Distinct, TTD, and Drugbank. An online Venn tool, Cytoscape 3.7.2, and Autodock Tools were used to analyze the components and targets. The study also used a mouse model of UC to further investigate the effects of SJZ. HE staining, immunofluorescence, ELISA, qPCR, and Western blot were used to detect various indices. Results Eighty-three active components and 112 action targets were identified from SJZ, including 67 targets for treating UC-related NETs. The five core targets identified were AKT1, JUN, IL1B, PTGS2, and TNF, and molecular docking studies indicated that the five targets were well-docked with ginsenoside Rh2, isoflavones, and formononetin. Animal experiments demonstrated that SJZ could alleviate various parameters such as weight, colon length, spleen index, disease activity index, and intestinal pathology of the UC mice. Immunofluorescence and Western blot showed that SJZ could reduce the expression of IL1B and TNF in intestinal neutrophils while increasing the expression of Occludin. Cellular immunofluorescence suggests that SJZ can reduce the expression of TNF and IL1B in NETs. The qPCR results also suggested that SJZ could inhibit TNF signal. Furthermore, ELISA results suggested that SJZ could inhibit the expression of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6) while promoting the expression of anti-inflammatory cytokines (IL-10, IL-37, TGF-β). Conclusion SJZ treats UC by reducing the content of intestinal NETs, with primary targets on the NETs being IL1B and TNFand suppress TNF signal. The practical components of SJZ may be ginsenoside Rh2, isoflavones, and formononetin.
Collapse
Affiliation(s)
- Dong Zhang
- Gastrointestinal Ward, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
- Gastrointestinal Ward, Shenzhen Hospital of Traditional Chinese Medicine, Shenzhen, People’s Republic of China
| | - Siwei Duan
- Institute of Gastroenterology, Science and Technology Innovation Center of Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Zhangyou He
- Institute of Gastroenterology, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Zeming Zhu
- Institute of Gastroenterology, Science and Technology Innovation Center of Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Zhiping Li
- Institute of Gastroenterology, Science and Technology Innovation Center of Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Qincheng Yi
- Institute of Gastroenterology, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Tiantian Cai
- Gastrointestinal Ward, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
- Gastrointestinal Ward, Shenzhen Hospital of Traditional Chinese Medicine, Shenzhen, People’s Republic of China
| | - Juanjuan Li
- Gastrointestinal Ward, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
- Gastrointestinal Ward, Shenzhen Hospital of Traditional Chinese Medicine, Shenzhen, People’s Republic of China
| | - Nan Chen
- Gastrointestinal Ward, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
- Gastrointestinal Ward, Shenzhen Hospital of Traditional Chinese Medicine, Shenzhen, People’s Republic of China
| | - Shaoju Guo
- Gastrointestinal Ward, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
- Gastrointestinal Ward, Shenzhen Hospital of Traditional Chinese Medicine, Shenzhen, People’s Republic of China
| |
Collapse
|
25
|
Bennike TB. Advances in proteomics: characterization of the innate immune system after birth and during inflammation. Front Immunol 2023; 14:1254948. [PMID: 37868984 PMCID: PMC10587584 DOI: 10.3389/fimmu.2023.1254948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/26/2023] [Indexed: 10/24/2023] Open
Abstract
Proteomics is the characterization of the protein composition, the proteome, of a biological sample. It involves the large-scale identification and quantification of proteins, peptides, and post-translational modifications. This review focuses on recent developments in mass spectrometry-based proteomics and provides an overview of available methods for sample preparation to study the innate immune system. Recent advancements in the proteomics workflows, including sample preparation, have significantly improved the sensitivity and proteome coverage of biological samples including the technically difficult blood plasma. Proteomics is often applied in immunology and has been used to characterize the levels of innate immune system components after perturbations such as birth or during chronic inflammatory diseases like rheumatoid arthritis (RA) and inflammatory bowel disease (IBD). In cancers, the tumor microenvironment may generate chronic inflammation and release cytokines to the circulation. In these situations, the innate immune system undergoes profound and long-lasting changes, the large-scale characterization of which may increase our biological understanding and help identify components with translational potential for guiding diagnosis and treatment decisions. With the ongoing technical development, proteomics will likely continue to provide increasing insights into complex biological processes and their implications for health and disease. Integrating proteomics with other omics data and utilizing multi-omics approaches have been demonstrated to give additional valuable insights into biological systems.
Collapse
Affiliation(s)
- Tue Bjerg Bennike
- Medical Microbiology and Immunology, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| |
Collapse
|
26
|
Jagirdhar GSK, Perez JA, Perez AB, Surani S. Integration and implementation of precision medicine in the multifaceted inflammatory bowel disease. World J Gastroenterol 2023; 29:5211-5225. [PMID: 37901450 PMCID: PMC10600960 DOI: 10.3748/wjg.v29.i36.5211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/31/2023] [Accepted: 09/06/2023] [Indexed: 09/20/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a complex disease with variability in genetic, environmental, and lifestyle factors affecting disease presentation and course. Precision medicine has the potential to play a crucial role in managing IBD by tailoring treatment plans based on the heterogeneity of clinical and temporal variability of patients. Precision medicine is a population-based approach to managing IBD by integrating environmental, genomic, epigenomic, transcriptomic, proteomic, and metabolomic factors. It is a recent and rapidly developing medicine. The widespread adoption of precision medicine worldwide has the potential to result in the early detection of diseases, optimal utilization of healthcare resources, enhanced patient outcomes, and, ultimately, improved quality of life for individuals with IBD. Though precision medicine is promising in terms of better quality of patient care, inadequacies exist in the ongoing research. There is discordance in study conduct, and data collection, utilization, interpretation, and analysis. This review aims to describe the current literature on precision medicine, its multiomics approach, and future directions for its application in IBD.
Collapse
Affiliation(s)
| | - Jose Andres Perez
- Department of Medicine, Saint Francis Health Systems, Tulsa, OK 74133, United States
| | - Andrea Belen Perez
- Department of Research, Columbia University, New York, NY 10027, United States
| | - Salim Surani
- Department of Medicine and Pharmacology, Texas A&M University, College Station, TX 77413, United States
| |
Collapse
|
27
|
Juha M, Molnár A, Jakus Z, Ledó N. NETosis: an emerging therapeutic target in renal diseases. Front Immunol 2023; 14:1253667. [PMID: 37744367 PMCID: PMC10514582 DOI: 10.3389/fimmu.2023.1253667] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 08/15/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction Neutrophil extracellular traps (NETs) are web-like structures composed of nuclear and granular components. The primary role of NETS is to prevent the dissemination of microbes and facilitate their elimination. However, this process is accompanied by collateral proinflammatory adverse effects when the NET release becomes uncontrollable, or clearance is impaired. Although NET-induced organ damage is conducted primarily and indirectly via immune complexes and the subsequent release of cytokines, their direct effects on cells are also remarkable. NETosis plays a critical pathogenic role in several renal disorders, such as the early phase of acute tubular necrosis, anti-neutrophil cytoplasmic antibody-mediated renal vasculitis, lupus nephritis, thrombotic microangiopathies, anti-glomerular basement membrane disease, and diabetic nephropathy. Their substantial contribution in the course of these disorders makes them a desirable target in the therapeutic armamentarium. This article gives an in-depth review of the heterogeneous pathogenesis and physiological regulations of NETosis and its pivotal role in renal diseases. Based on the pathogenesis, the article also outlines the current therapeutic options and possible molecular targets in the treatment of NET-related renal disorders. Methods We carried out thorough literature research published in PubMed and Google Scholar, including a comprehensive review and analysis of the classification, pathomechanisms, and a broad spectrum of NET-related kidney disorders. Conclusions NETosis plays a pivotal role in certain renal diseases. It initiates and maintains inflammatory and autoimmune disorders, thus making it a desirable target for improving patient and renal outcomes. Better understanding and clinical translation of the pathogenesis are crucial aspects to treatment, for improving patient, and renal outcomes.
Collapse
Affiliation(s)
- Márk Juha
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | - Adél Molnár
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | - Zoltán Jakus
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Nóra Ledó
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
28
|
Devarakonda S, Thorsell A, Hedenström P, Rezapour A, Heden L, Banerjee S, Johansson MEV, Birchenough G, Toft Morén A, Gustavsson K, Skokic V, Pettersson VL, Sjöberg F, Kalm M, Al Masri M, Ekh M, Fagman H, Wolving M, Perkins R, Morales RA, Castillo F, Villablanca EJ, Yrlid U, Bergmark K, Steineck G, Bull C. Low-grade intestinal inflammation two decades after pelvic radiotherapy. EBioMedicine 2023; 94:104691. [PMID: 37480626 PMCID: PMC10393618 DOI: 10.1016/j.ebiom.2023.104691] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 07/24/2023] Open
Abstract
BACKGROUND Radiotherapy is effective in the treatment of cancer but also causes damage to non-cancerous tissue. Pelvic radiotherapy may produce chronic and debilitating bowel symptoms, yet the underlying pathophysiology is still undefined. Most notably, although pelvic radiotherapy causes an acute intestinal inflammation there is no consensus on whether the late-phase pathophysiology contains an inflammatory component or not. To address this knowledge gap, we examined the potential presence of a chronic inflammation in mucosal biopsies from irradiated pelvic cancer survivors. METHODS We biopsied 24 cancer survivors two to 20 years after pelvic radiotherapy, and four non-irradiated controls. Using tandem mass tag (TMT) mass spectrometry and mRNA sequencing (mRNA-seq), we charted proteomic and transcriptomic profiles of the mucosal tissue previously exposed to a high or a low/no dose of radiation. Changes in the immune cell populations were determined with flow cytometry. The integrity of the protective mucus layers were determined by permeability analysis and 16S rRNA bacterial detection. FINDINGS 942 proteins were differentially expressed in mucosa previously exposed to a high radiation dose compared to a low radiation dose. The data suggested a chronic low-grade inflammation with neutrophil activity, which was confirmed by mRNA-seq and flow cytometry and further supported by findings of a weakened mucus barrier with bacterial infiltration. INTERPRETATION Our results challenge the idea that pelvic radiotherapy causes an acute intestinal inflammation that either heals or turns fibrotic without progression to chronic inflammation. This provides a rationale for exploring novel strategies to mitigate chronic bowel symptoms in pelvic cancer survivors. FUNDING This study was supported by the King Gustav V Jubilee Clinic Cancer Foundation (CB), The Adlerbertska Research Foundation (CB), The Swedish Cancer Society (GS), The Swedish State under the ALF agreement (GS and CB), Mary von Sydow's foundation (MA and VP).
Collapse
Affiliation(s)
- Sravani Devarakonda
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Annika Thorsell
- Proteomics Core Facility, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Per Hedenström
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden; Department of Gastroenterology and Hepatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Azar Rezapour
- Department of Microbiology and Immunology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lisen Heden
- Pelvic Cancer Rehabilitation, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Sanghita Banerjee
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Malin E V Johansson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - George Birchenough
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Amelie Toft Morén
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karin Gustavsson
- Pelvic Cancer Rehabilitation, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Viktor Skokic
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Molecular Medicine and Surgery and Department of Pelvic Cancer, Karolinska Institute, Stockholm, Sweden
| | - Victor L Pettersson
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fei Sjöberg
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Infectious Diseases, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, 413 90 Gothenburg, Sweden
| | - Marie Kalm
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 413 90 Gothenburg, Sweden
| | - Mohammad Al Masri
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Michaela Ekh
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Fagman
- Department of Laboratory Medicine, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg and Department of Clinical Patology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mats Wolving
- Department of Pathology, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Rosie Perkins
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Rodrigo A Morales
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Francisca Castillo
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Eduardo J Villablanca
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Ulf Yrlid
- Department of Microbiology and Immunology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karin Bergmark
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Gunnar Steineck
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Bull
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
29
|
Stensballe A, Bennike TB, Ravn-Haren G, Mortensen A, Aboo C, Knudsen LA, Rühlemann MC, Birkelund S, Bang C, Franke A, Vogel U, Hansen AK, Andersen V. Impaired Abcb1a function and red meat in a translational colitis mouse model induces inflammation and alters microbiota composition. Front Med (Lausanne) 2023; 10:1200317. [PMID: 37588005 PMCID: PMC10425965 DOI: 10.3389/fmed.2023.1200317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/20/2023] [Indexed: 08/18/2023] Open
Abstract
Inflammatory Bowel Disease (IBD) affects approximately 0.3% of the global population, with incidence rates rising dramatically worldwide. Emerging evidence points to an interplay between exposome factors such as diet and gut microbiota, host genetics, and the immune system as crucial elements in IBD development. ATP-binding cassette (ABC) transporters, including human p-glycoprotein encoded by the Abcb1 gene, influence intestinal inflammation, and their expression may interact with environmental factors such as diet and gut microbes. Our study aimed to examine the impact of protein sources on a genetic colitis mouse model. Methods Abcb1a-deficient colitis mice were fed either casein or red meat-supplemented diets to investigate potential colitis-aggravating components in red meat and their effects on host-microbiota interactions. We conducted deep label free quantitative proteomic inflammation profiling of gastrointestinal tissue (colon, ileum) and urine, and determined the overall microbiome in feces using 16S rRNA gene sequencing. Microbiota shifts by diet and protein transporter impairment were addressed by multivariate statistical analysis. Colon and systemic gut inflammation were validated through histology and immune assays, respectively. Results A quantitative discovery based proteomic analysis of intestinal tissue and urine revealed associations between ileum and urine proteomes in relation to Abcb1a deficiency. The absence of Abcb1a efflux pump function and diet-induced intestinal inflammation impacted multiple systemic immune processes, including extensive neutrophil extracellular trap (NET) components observed in relation to neutrophil degranulation throughout the gastrointestinal tract. The colitis model's microbiome differed significantly from that of wild-type mice, indicating the substantial influence of efflux transporter deficiency on microbiota. Conclusion The proteomic and microbiota analyzes of a well-established murine model enabled the correlation of gastrointestinal interactions not readily identifiable in human cohorts. Insights into dysregulated biological pathways in this disease model might offer translational biomarkers based on NETs and improved understanding of IBD pathogenesis in human patients. Our findings demonstrate that drug transporter deficiency induces substantial changes in the microbiota, leading to increased levels of IBD-associated strains and resulting in intestinal inflammation. GRAPHICAL ABSTRACT.
Collapse
Affiliation(s)
- Allan Stensballe
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Tue Bjerg Bennike
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Gitte Ravn-Haren
- National Food Institute, Technical University of Denmark, Lyngby, Denmark
| | - Alicja Mortensen
- National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Christopher Aboo
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Sino-Danish Center for Research and Education, University of Chinese Academy of Sciences, Beijing, China
| | - Lina Almind Knudsen
- Institute of Regional Health Research-Center Soenderjylland, University of Southern Denmark, Odense, Denmark
| | - Malte C. Rühlemann
- Institute of Clinical Molecular Biology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Svend Birkelund
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Corinne Bang
- Institute of Clinical Molecular Biology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Ulla Vogel
- National Food Institute, Technical University of Denmark, Lyngby, Denmark
- National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Axel Kornerup Hansen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Vibeke Andersen
- Institute of Regional Health Research-Center Soenderjylland, University of Southern Denmark, Odense, Denmark
- Molecular Diagnostic and Clinical Research Unit, University Hospital of Southern Denmark, Aabenraa, Denmark
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
30
|
Mankan AK, Czajka-Francuz P, Prendes M, Ramanan S, Koziej M, Vidal L, Saini KS. Intracellular DNA sensing by neutrophils and amplification of the innate immune response. Front Immunol 2023; 14:1208137. [PMID: 37483598 PMCID: PMC10361817 DOI: 10.3389/fimmu.2023.1208137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/13/2023] [Indexed: 07/25/2023] Open
Abstract
As the first responders, neutrophils lead the innate immune response to infectious pathogens and inflammation inducing agents. The well-established pathogen neutralizing strategies employed by neutrophils are phagocytosis, the action of microbicide granules, the production of ROS, and the secretion of neutrophil extracellular traps (NETs). Only recently, the ability of neutrophils to sense and respond to pathogen-associated molecular patterns is being appreciated. This review brings together the current information about the intracellular recognition of DNA by neutrophils and proposes models of signal amplification in immune response. Finally, the clinical relevance of DNA sensing by neutrophils in infectious and non-infectious diseases including malignancy are also discussed.
Collapse
Affiliation(s)
| | | | - Maria Prendes
- Labcorp Drug Development Inc., Princeton, NJ, United States
| | - Sriram Ramanan
- Labcorp Drug Development Inc., Princeton, NJ, United States
| | | | | | - Kamal S. Saini
- Fortrea, Inc., Durham, NC, United States
- Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| |
Collapse
|
31
|
Kałużna A, Jura-Półtorak A, Derkacz A, Jaruszowiec J, Olczyk K, Komosinska-Vassev K. Circulating Profiles of Serum Proguanylin, S100A12 Protein and Pentraxin 3 as Diagnostic Markers of Ulcerative Colitis. J Clin Med 2023; 12:4339. [PMID: 37445374 DOI: 10.3390/jcm12134339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
The aim of this research was to investigate potential new biomarkers which could be used in the clinical practice of ulcerative colitis (UC). Given the crucial role of intestinal barrier integrity and inflammation in the pathogenesis of UC, the serum profile of proteins linked to intestinal barrier and pro-inflammatory neutrophil products may be useful in diagnosing and monitoring the activity of the disease. We measured serum levels of proguanylin (pro-GN), S100A12, and pentraxin 3 (PTX3) in 31 patients with UC before and after a year of biological treatment, as well as in 20 healthy individuals. Significant differences in the serum profiles of pro-GN (5.27 vs. 11.35, p < 0.001), S100A12 (39.36 vs. 19.74, p < 0.001) and PTX3 (3197.05 vs. 1608.37, p < 0.001) were observed between pre-treatment patients with UC and healthy individuals. Furthermore, in UC patients prior to treatment, the levels of S100A12 (p < 0.0005; r = 0.628) and PTX3 (p < 0.05; r = 0.371) were correlated with disease activity as measured by the Mayo scale. Following a year of biological treatment with adalimumab, the concentration of pro-GN significantly increased (5.27 vs. 6.68, p < 0.005) in the blood of UC patients, while the level of PTX-3 decreased (3197.05 vs. 1946.4, p < 0.0001). Our study demonstrates the usefulness of pro-GN, S100A12, and PTX3 measurements in diagnosing and monitoring the activity of UC.
Collapse
Affiliation(s)
- Aleksandra Kałużna
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland
| | - Agnieszka Jura-Półtorak
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland
| | | | - Julia Jaruszowiec
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland
| | - Krystyna Olczyk
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland
| | - Katarzyna Komosinska-Vassev
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland
| |
Collapse
|
32
|
Fabian O, Bajer L, Drastich P, Harant K, Sticova E, Daskova N, Modos I, Tichanek F, Cahova M. A Current State of Proteomics in Adult and Pediatric Inflammatory Bowel Diseases: A Systematic Search and Review. Int J Mol Sci 2023; 24:ijms24119386. [PMID: 37298338 DOI: 10.3390/ijms24119386] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Inflammatory bowel diseases (IBD) are systemic immune-mediated conditions with predilection for the gastrointestinal tract and include Crohn's disease and ulcerative colitis. Despite the advances in the fields of basic and applied research, the etiopathogenesis remains largely unknown. As a result, only one third of the patients achieve endoscopic remission. A substantial portion of the patients also develop severe clinical complications or neoplasia. The need for novel biomarkers that can enhance diagnostic accuracy, more precisely reflect disease activity, and predict a complicated disease course, thus, remains high. Genomic and transcriptomic studies contributed substantially to our understanding of the immunopathological pathways involved in disease initiation and progression. However, eventual genomic alterations do not necessarily translate into the final clinical picture. Proteomics may represent a missing link between the genome, transcriptome, and phenotypical presentation of the disease. Based on the analysis of a large spectrum of proteins in tissues, it seems to be a promising method for the identification of new biomarkers. This systematic search and review summarize the current state of proteomics in human IBD. It comments on the utility of proteomics in research, describes the basic proteomic techniques, and provides an up-to-date overview of available studies in both adult and pediatric IBD.
Collapse
Affiliation(s)
- Ondrej Fabian
- Clinical and Transplant Pathology Centre, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic
- Department of Pathology and Molecular Medicine, 3rd Faculty of Medicine, Charles University and Thomayer Hospital, 140 59 Prague, Czech Republic
| | - Lukas Bajer
- Department of Gastroenterology and Hepatology, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic
- Institute of Microbiology, Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Pavel Drastich
- Department of Gastroenterology and Hepatology, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic
| | - Karel Harant
- Proteomics Core Facility, Faculty of Science, Charles University, 252 50 Vestec, Czech Republic
| | - Eva Sticova
- Clinical and Transplant Pathology Centre, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic
- Department of Pathology, Royal Vinohrady Teaching Hospital, Srobarova 1150/50, 100 00 Prague, Czech Republic
| | - Nikola Daskova
- Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic
| | - Istvan Modos
- Department of Informatics, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic
| | - Filip Tichanek
- Department of Informatics, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic
| | - Monika Cahova
- Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic
| |
Collapse
|
33
|
Drury B, Chuah CS, Hall R, Hardisty GR, Rossi AG, Ho GT. Neutrophil-dependent Mitochondrial DNA Release Associated With Extracellular Trap Formation in Inflammatory Bowel Disease. GASTRO HEP ADVANCES 2023; 2:788-798. [PMID: 39130118 PMCID: PMC11308081 DOI: 10.1016/j.gastha.2023.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 03/20/2023] [Indexed: 08/13/2024]
Abstract
Background and Aims Inflammatory bowel disease (IBD) is associated with increased circulating damage-associated molecular patterns, in particular, the highly pro-inflammatory mitochondrial DNA (mtDNA). Here, we study the importance of blood neutrophils in mtDNA release via neutrophil extracellular trap (NET) formation and mitochondrial NETosis, where neutrophils specifically expulse mtDNA as potential targetable biological pathways. Methods We investigated the roles of A23187 (a known NET stimulant), granulocyte macrophage stimulating factor, lipopolysaccharide (LPS), and human IBD plasma in their ability to induce NET formation, mitochondrial NETosis, mtDNA, and total DNA release from human blood neutrophils; and the evidence for increased NET formation in IBD. Results We demonstrated that NET formation resulted in significant DNA (P < .0001) and mtDNA release (P < .0001) with long DNA fragments (>1000 base pairs) with NETs containing high levels of mtDNA. Using previously described in vitro conditions for mitochondrial NETosis, granulocyte macrophage stimulating factor + LPS triggered neutrophil mtDNA release at lower levels but not NETosis. LPS alone can trigger neutrophilic DNA release without NET formation. Heterologous coculture with plasma from patients with active IBD (vs remission [n = 6/group]) were not associated with significantly higher levels of NETs and mtDNA release. During coculture with active IBD plasma (vs remission), citrullinated histone 3 (CitH3) (a NETs biomarker) levels were significantly lower (P < .001). Similarly, CitH3 levels were lower in stool supernatants of patients with active IBD vs remission (n = 19/12, P = .0001). Stool CitH3 negatively correlates with stool calprotectin, a biomarker for gut inflammation (r = -0.47, P = .03). Conclusion Hence, although blood neutrophils remain an important source of circulating mtDNA with defined mechanisms for release via NET formation and during neutrophil activation, our data do not support excessive systemic NET formation as a dominant underpinning pathobiological process in IBD.
Collapse
Affiliation(s)
- Broc Drury
- Edinburgh IBD Science Unit, Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Cher S. Chuah
- Edinburgh IBD Science Unit, Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Rebecca Hall
- Edinburgh IBD Science Unit, Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Gareth R. Hardisty
- Edinburgh IBD Science Unit, Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Adriano G. Rossi
- Edinburgh IBD Science Unit, Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Gwo-Tzer Ho
- Edinburgh IBD Science Unit, Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
34
|
Prakash A, García-Seisdedos D, Wang S, Kundu DJ, Collins A, George N, Moreno P, Papatheodorou I, Jones AR, Vizcaíno JA. Integrated View of Baseline Protein Expression in Human Tissues. J Proteome Res 2023; 22:729-742. [PMID: 36577097 PMCID: PMC9990129 DOI: 10.1021/acs.jproteome.2c00406] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The availability of proteomics datasets in the public domain, and in the PRIDE database, in particular, has increased dramatically in recent years. This unprecedented large-scale availability of data provides an opportunity for combined analyses of datasets to get organism-wide protein abundance data in a consistent manner. We have reanalyzed 24 public proteomics datasets from healthy human individuals to assess baseline protein abundance in 31 organs. We defined tissue as a distinct functional or structural region within an organ. Overall, the aggregated dataset contains 67 healthy tissues, corresponding to 3,119 mass spectrometry runs covering 498 samples from 489 individuals. We compared protein abundances between different organs and studied the distribution of proteins across these organs. We also compared the results with data generated in analogous studies. Additionally, we performed gene ontology and pathway-enrichment analyses to identify organ-specific enriched biological processes and pathways. As a key point, we have integrated the protein abundance results into the resource Expression Atlas, where they can be accessed and visualized either individually or together with gene expression data coming from transcriptomics datasets. We believe this is a good mechanism to make proteomics data more accessible for life scientists.
Collapse
Affiliation(s)
- Ananth Prakash
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom.,Open Targets, Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom
| | - David García-Seisdedos
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom
| | - Shengbo Wang
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom
| | - Deepti Jaiswal Kundu
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom
| | - Andrew Collins
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, LiverpoolL69 7ZB, United Kingdom
| | - Nancy George
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom
| | - Pablo Moreno
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom
| | - Irene Papatheodorou
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom.,Open Targets, Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom
| | - Andrew R Jones
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, LiverpoolL69 7ZB, United Kingdom
| | - Juan Antonio Vizcaíno
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom.,Open Targets, Wellcome Genome Campus, Hinxton, CambridgeCB10 1SD, United Kingdom
| |
Collapse
|
35
|
Louis Sam Titus ASC, Vanarsa K, Soomro S, Patel A, Prince J, Kugathasan S, Mohan C. Resistin, Elastase, and Lactoferrin as Potential Plasma Biomarkers of Pediatric Inflammatory Bowel Disease Based on Comprehensive Proteomic Screens. Mol Cell Proteomics 2023; 22:100487. [PMID: 36549591 PMCID: PMC9918796 DOI: 10.1016/j.mcpro.2022.100487] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 11/10/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Inflammatory bowel disease (IBD) is an immune-mediated chronic inflammation of the intestine, which can present in the form of ulcerative colitis (UC) or as Crohn's disease (CD). Biomarkers are needed for reliable diagnosis and disease monitoring in IBD, especially in pediatric patients. Plasma samples from a pediatric IBD cohort were interrogated using an aptamer-based screen of 1322 proteins. The elevated biomarkers identified using the aptamer screen were further validated by ELISA using an independent cohort of 76 pediatric plasma samples, drawn from 30 CD, 30 UC, and 16 healthy controls. Of the 1322 proteins screened in plasma from IBD patients, 129 proteins were significantly elevated when compared with healthy controls. Of these 15 proteins had a fold change greater than 2 and 28 proteins had a fold change >1.5. Neutrophil and extracellular vesicle signatures were detected among the elevated plasma biomarkers. When seven of these proteins were validated by ELISA, resistin was the only protein that was significantly higher in both UC and CD (p < 0.01), with receiver operating characteristic area under the curve value of 0.82 and 0.77, respectively, and the only protein that exhibited high sensitivity and specificity for both CD and UC. The next most discriminatory plasma proteins were elastase and lactoferrin, particularly for UC, with receiver operating characteristic area under the curve values of 0.74 and 0.69, respectively. We have identified circulating resistin, elastase, and lactoferrin as potential plasma biomarkers of IBD in pediatric patients using two independent diagnostic platforms and two independent patient cohorts.
Collapse
Affiliation(s)
| | - Kamala Vanarsa
- Department Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Sanam Soomro
- Department Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Anjali Patel
- Department Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Jarod Prince
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Subra Kugathasan
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA.
| | - Chandra Mohan
- Department Biomedical Engineering, University of Houston, Houston, Texas, USA.
| |
Collapse
|
36
|
Wang H, Wang Y, Wang X, Wei R, Wang X, Gong P, Zhang N, Zhang X, Li X, Li J. Trichomonas gallinae induces heterophil extracellular trap formation in pigeons. Parasitol Res 2023; 122:527-536. [PMID: 36522547 DOI: 10.1007/s00436-022-07755-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
Avian trichomonosis is a worldwide and cross-species epidemic, and the infection in pigeons is particularly severe. Although the disease causes a serious threat to poultry health resulting in significant economic losses, the relationship between Trichomonas gallinae (T. gallinae) and host innate immunity is still not clear. Extracellular traps (ETs) are an innate immunity response to parasitic infections. However, whether host cells can produce ETs after T. gallinae infection has not yet been reported. In the present study, the ability of T. gallinae to induce the production of heterophil extracellular traps (HETs) in pigeons was examined. T. gallinae-induced HETs were observed by scanning electron microscopy (SEM) and the main components of HETs were detected by fluorescence confocal microscopy. Changes in reactive oxygen species (ROS) and lactate dehydrogenase (LDH) were tested during the HETosis. A quantitative analysis of T. gallinae-induced HETs, the role of myeloperoxidase (MPO), store-operated Ca (2+) entry (SOCE), and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase in T. gallinae-induced HET formation were conducted by inhibitor assays. The results showed that T. gallinae induced ET formation in pigeon heterophils. ETs consisted of a DNA skeleton, neutrophil elastase (NE), MPO, and Histone3 (H3). T. gallinae-induced HETs formation in a dose- and time-dependent process. The release of T. gallinae-induced HETs depends on MPO, SOCE, and NADPH oxidase. Furthermore, after T. gallinae stimulated pigeon heterophils, ROS production was significantly increased, while no significant differences in the LDH activity were observed.
Collapse
Affiliation(s)
- Hongyu Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Yuru Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Xuehan Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Ran Wei
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Xiaocen Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Pengtao Gong
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Nan Zhang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Xichen Zhang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Xin Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China.
| | - Jianhua Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China.
| |
Collapse
|
37
|
Saez A, Herrero-Fernandez B, Gomez-Bris R, Sánchez-Martinez H, Gonzalez-Granado JM. Pathophysiology of Inflammatory Bowel Disease: Innate Immune System. Int J Mol Sci 2023; 24:ijms24021526. [PMID: 36675038 PMCID: PMC9863490 DOI: 10.3390/ijms24021526] [Citation(s) in RCA: 191] [Impact Index Per Article: 95.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 12/30/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Inflammatory bowel disease (IBD), comprising Crohn's disease (CD) and ulcerative colitis (UC), is a heterogeneous state of chronic intestinal inflammation with no exact known cause. Intestinal innate immunity is enacted by neutrophils, monocytes, macrophages, and dendritic cells (DCs), and innate lymphoid cells and NK cells, characterized by their capacity to produce a rapid and nonspecific reaction as a first-line response. Innate immune cells (IIC) defend against pathogens and excessive entry of intestinal microorganisms, while preserving immune tolerance to resident intestinal microbiota. Changes to this equilibrium are linked to intestinal inflammation in the gut and IBD. IICs mediate host defense responses, inflammation, and tissue healing by producing cytokines and chemokines, activating the complement cascade and phagocytosis, or presenting antigens to activate the adaptive immune response. IICs exert important functions that promote or ameliorate the cellular and molecular mechanisms that underlie and sustain IBD. A comprehensive understanding of the mechanisms underlying these clinical manifestations will be important for developing therapies targeting the innate immune system in IBD patients. This review examines the complex roles of and interactions among IICs, and their interactions with other immune and non-immune cells in homeostasis and pathological conditions.
Collapse
Affiliation(s)
- Angela Saez
- LamImSys Lab, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria (UFV), 28223 Pozuelo de Alarcón, Spain
| | - Beatriz Herrero-Fernandez
- LamImSys Lab, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
| | - Raquel Gomez-Bris
- LamImSys Lab, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
| | - Hector Sánchez-Martinez
- LamImSys Lab, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Jose M. Gonzalez-Granado
- LamImSys Lab, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-913908766
| |
Collapse
|
38
|
Lai HJ, Doan HT, Lin EY, Chiu YL, Cheng YK, Lin YH, Chiang HS. Histones of Neutrophil Extracellular Traps Directly Disrupt the Permeability and Integrity of the Intestinal Epithelial Barrier. Inflamm Bowel Dis 2023; 29:783-797. [PMID: 36617175 DOI: 10.1093/ibd/izac256] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Indexed: 01/09/2023]
Abstract
BACKGROUND Increased neutrophil extracellular trap (NET) formation and abundant NET-associated proteins are frequently found in the inflamed colon of patients with inflammatory bowel disease. Peptidyl arginine deiminase 4 (PAD4) activation is essential for the generation of NET and NET-mediated pathogenesis. However, the role of PAD4-dependent NET formation in murine inflammatory bowel disease models and the molecular mechanisms responsible for the altered gut barrier function are unknown. METHODS Wild-type and Pad4 knockout (Pad4-/-) mice were administrated 3% dextran sulfate sodium (DSS) in their drinking water. Caco-2 monolayers were used to test the effect of NETs on intestinal barrier function and cytotoxicity. Histones were intrarectally administrated to wild-type mice to determine their effects on intestinal barrier function and cytotoxicity in vivo. RESULTS PAD4 deficiency reduced the severity of DSS-induced colitis with decreased intestinal NET formation and enhanced gut barrier function and integrity in mice. NETs disrupted the barrier function in intestinal epithelial Caco-2 monolayers through their protein, rather than DNA, components. Pretreatment of NETs with histone inhibitors abrogated the effects on epithelial permeability. Consistent with these observations, adding purified histone proteins to Caco-2 monolayers significantly damaged epithelial barrier function, which was associated with the abnormal distribution and integrity of tight junctions as well as with increased cell death. Furthermore, intrarectal administration of histones damaged the intestinal barrier integrity and induced cytotoxicity in the mouse colon epithelium. CONCLUSIONS PAD4-mediated NET formation has a detrimental role in acute colitis. NET-associated histones directly inhibit intestinal barrier function, resulting in cytotoxicity in vitro and in vivo.
Collapse
Affiliation(s)
- Hsuan-Ju Lai
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Ha T Doan
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Elliot Y Lin
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yi-Ling Chiu
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yuan-Kai Cheng
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yi-He Lin
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Hao-Sen Chiang
- Department of Life Science, National Taiwan University, Taipei, Taiwan.,Genome and Systems Biology Degree Program, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
39
|
Wang P, Liu D, Zhou Z, Liu F, Shen Y, You Q, Lu S, Wu J. The role of protein arginine deiminase 4-dependent neutrophil extracellular traps formation in ulcerative colitis. Front Immunol 2023; 14:1144976. [PMID: 37143672 PMCID: PMC10151647 DOI: 10.3389/fimmu.2023.1144976] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/30/2023] [Indexed: 05/06/2023] Open
Abstract
Background Neutrophil extracellular traps (NETs) play an important role in the development and progression of ulcerative colitis (UC). Peptidyl arginine deiminase 4 (PAD4) is essential for the formation of NETs via catalyzing histone citrullination. This study mainly to explore the role of PAD4-mediated NETs in intestinal inflammation of dextran sulfate sodium (DSS)-induced UC. Methods Acute and chronic colitis mouse models were established by supplementing DSS in drinking water. Colon tissues from colitis mice were analyzed for the level of PAD4 expression, citrullinated histone H3(Cit-H3), intestinal histopathology, and inflammatory cytokines secretion. Serum samples were tested for systemic neutrophil activation biomarkers. Colitis mice administered with Cl-amidine, a PAD4 inhibitor, and PAD4 knockout mice were investigated to detect NETs formation, intestinal inflammation, and barrier function. Result We found the formation of NETs significantly increased in DSS-induced colitis mice and was correlated with disease markers. Blocking NETs formation by Cl-amidine or PAD4 genetic knockout could alleviate clinical colitis index, intestinal inflammation, and barrier dysfunction. Conclusion This study provided a research basis for the role of PAD4-mediated NETs formation in the pathogenesis of UC and suggested that inhibition of PAD4 activity and the formation of NETs may be helpful for the prevention and treatment of UC.
Collapse
Affiliation(s)
- Ping Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Dan Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Ziqi Zhou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Fangjun Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yiming Shen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Qi You
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Shiping Lu
- Department of Immunology and Microbiology, Tulane University, New Orleans, LA, United States
- *Correspondence: Jie Wu, ; Shiping Lu,
| | - Jie Wu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- *Correspondence: Jie Wu, ; Shiping Lu,
| |
Collapse
|
40
|
Song YH, Wang ZJ, Kang L, He ZX, Zhao SB, Fang X, Li ZS, Wang SL, Bai Y. PADs and NETs in digestive system: From physiology to pathology. Front Immunol 2023; 14:1077041. [PMID: 36761761 PMCID: PMC9902375 DOI: 10.3389/fimmu.2023.1077041] [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: 10/22/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
Peptidylarginine deiminases (PADs) are the only enzyme class known to deiminate arginine residues into citrulline in proteins, a process known as citrullination. This is an important post-translational modification that functions in several physiological and pathological processes. Neutrophil extracellular traps (NETs) are generated by NETosis, a novel cell death in neutrophils and a double-edged sword in inflammation. Excessive activation of PADs and NETs is critically implicated in their transformation from a physiological to a pathological state. Herein, we review the physiological and pathological functions of PADs and NETs, in particular, the involvement of PAD2 and PAD4 in the digestive system, from inflammatory to oncological diseases, along with related therapeutic prospects.
Collapse
Affiliation(s)
- Yi-Hang Song
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhi-Jie Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Le Kang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zi-Xuan He
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Sheng-Bing Zhao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xue Fang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhao-Shen Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Shu-Ling Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yu Bai
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
41
|
Formyl peptide receptor 2 as a potential therapeutic target for inflammatory bowel disease. Acta Pharmacol Sin 2023; 44:19-31. [PMID: 35840658 DOI: 10.1038/s41401-022-00944-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/15/2022] [Indexed: 01/18/2023]
Abstract
Inflammatory bowel disease (IBD) is a global health burden whose existing treatment is largely dependent on anti-inflammatory agents. Despite showing some therapeutic actions, their clinical efficacy and adverse events are unacceptable. Resolution as an active and orchestrated phase of inflammation involves improper inflammatory response with three key triggers, specialized pro-resolving mediators (SPMs), neutrophils and phagocyte efferocytosis. The formyl peptide receptor 2 (FPR2/ALX) is a human G protein-coupled receptor capable of binding SPMs and participates in the resolution process. This receptor has been implicated in several inflammatory diseases and its association with mouse model of IBD was established in some resolution-related studies. Here, we give an overview of three reported FPR2/ALX agonists highlighting their respective roles in pro-resolving strategies.
Collapse
|
42
|
Porras AM, Zhou H, Shi Q, Xiao X, Longman R, Brito IL. Inflammatory Bowel Disease-Associated Gut Commensals Degrade Components of the Extracellular Matrix. mBio 2022; 13:e0220122. [PMID: 36445085 PMCID: PMC9765649 DOI: 10.1128/mbio.02201-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/02/2022] [Indexed: 12/03/2022] Open
Abstract
Extracellular matrix (ECM) remodeling has emerged as a key feature of inflammatory bowel disease (IBD), and ECM fragments have been proposed as markers of clinical disease severity. Recent studies report increased protease activity in the gut microbiota of IBD patients. Nonetheless, the relationship between gut microbiota and ECM remodeling has remained unexplored. We hypothesized that members of the human gut microbiome could degrade the host ECM and that bacteria-driven remodeling, in turn, could enhance colonic inflammation. Through a variety of in vitro assays, we first confirmed that multiple bacterial species found in the human gut are capable of degrading specific ECM components. Clinical stool samples obtained from ulcerative colitis patients also exhibited higher levels of proteolytic activity in vitro, compared to those of their healthy counterparts. Furthermore, culture supernatants from bacteria species that are capable of degrading human ECM accelerated inflammation in dextran sodium sulfate (DSS)-induced colitis. Finally, we identified several of the bacterial proteases and carbohydrate degrading enzymes (CAZymes) that are potentially responsible for ECM degradation in vitro. Some of these protease families and CAZymes were also found in increased abundance in a metagenomic cohort of IBD. These results demonstrate that some commensal bacteria in the gut are indeed capable of degrading components of human ECM in vitro and suggest that this proteolytic activity may be involved in the progression of IBD. A better understanding of the relationship between nonpathogenic gut microbes, host ECM, and inflammation could be crucial to elucidating some of the mechanisms underlying host-bacteria interactions in IBD and beyond. IMPORTANCE Healthy gut epithelial cells form a barrier that keeps bacteria and other substances from entering the blood or tissues of the body. Those cells sit on scaffolding that maintains the structure of the gut and informs our immune system about the integrity of this barrier. In patients with inflammatory bowel disease (IBD), breaks are formed in this cellular barrier, and bacteria gain access to the underlying tissue and scaffolding. In our study, we discovered that bacteria that normally reside in the gut can modify and disassemble the underlying scaffolding. Additionally, we discovered that changes to this scaffolding affect the onset of IBD in mouse models of colitis as well as the abilities of these mice to recover. We propose that this new information will reveal how breaks in the gut wall lead to IBD and will open up new avenues by which to treat patients with IBD.
Collapse
Affiliation(s)
- Ana Maria Porras
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Hao Zhou
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Qiaojuan Shi
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Xieyue Xiao
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - JRI Live Cell Bank
- Jill Roberts Institute for IBD Research, Weill Cornell Medicine, New York, New York, USA
| | - Randy Longman
- Jill Roberts Institute for IBD Research, Weill Cornell Medicine, New York, New York, USA
| | - Ilana Lauren Brito
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| |
Collapse
|
43
|
Leppkes M, Lindemann A, Gößwein S, Paulus S, Roth D, Hartung A, Liebing E, Zundler S, Gonzalez-Acera M, Patankar JV, Mascia F, Scheibe K, Hoffmann M, Uderhardt S, Schauer C, Foersch S, Neufert C, Vieth M, Schett G, Atreya R, Kühl AA, Bleich A, Becker C, Herrmann M, Neurath MF. Neutrophils prevent rectal bleeding in ulcerative colitis by peptidyl-arginine deiminase-4-dependent immunothrombosis. Gut 2022; 71:2414-2429. [PMID: 34862250 PMCID: PMC9667856 DOI: 10.1136/gutjnl-2021-324725] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 11/02/2021] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Bleeding ulcers and erosions are hallmarks of active ulcerative colitis (UC). However, the mechanisms controlling bleeding and mucosal haemostasis remain elusive. DESIGN We used high-resolution endoscopy and colon tissue samples of active UC (n = 36) as well as experimental models of physical and chemical mucosal damage in mice deficient for peptidyl-arginine deiminase-4 (PAD4), gnotobiotic mice and controls. We employed endoscopy, histochemistry, live-cell microscopy and flow cytometry to study eroded mucosal surfaces during mucosal haemostasis. RESULTS Erosions and ulcerations in UC were covered by fresh blood, haematin or fibrin visible by endoscopy. Fibrin layers rather than fresh blood or haematin on erosions were inversely correlated with rectal bleeding in UC. Fibrin layers contained ample amounts of neutrophils coaggregated with neutrophil extracellular traps (NETs) with detectable activity of PAD. Transcriptome analyses showed significantly elevated PAD4 expression in active UC. In experimentally inflicted wounds, we found that neutrophils underwent NET formation in a PAD4-dependent manner hours after formation of primary blood clots, and remodelled clots to immunothrombi containing citrullinated histones, even in the absence of microbiota. PAD4-deficient mice experienced an exacerbated course of dextrane sodium sulfate-induced colitis with markedly increased rectal bleeding (96 % vs 10 %) as compared with controls. PAD4-deficient mice failed to remodel blood clots on mucosal wounds eliciting impaired healing. Thus, NET-associated immunothrombi are protective in acute colitis, while insufficient immunothrombosis is associated with rectal bleeding. CONCLUSION Our findings uncover that neutrophils induce secondary immunothrombosis by PAD4-dependent mechanisms. Insufficient immunothrombosis may favour rectal bleeding in UC.
Collapse
Affiliation(s)
- Moritz Leppkes
- Medical Clinic 1, University Clinic, Friedrich Alexander University, Erlangen, Germany .,Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Aylin Lindemann
- Medical Clinic 1, University Clinic, Friedrich Alexander University, Erlangen, Germany
| | - Stefanie Gößwein
- Medical Clinic 1, University Clinic, Friedrich Alexander University, Erlangen, Germany
| | - Susanne Paulus
- Medical Clinic 1, University Clinic, Friedrich Alexander University, Erlangen, Germany
| | - Dominik Roth
- Medical Clinic 1, University Clinic, Friedrich Alexander University, Erlangen, Germany
| | - Anne Hartung
- Medical Clinic 1, University Clinic, Friedrich Alexander University, Erlangen, Germany
| | - Eva Liebing
- Medical Clinic 1, University Clinic, Friedrich Alexander University, Erlangen, Germany
| | - Sebastian Zundler
- Medical Clinic 1, University Clinic, Friedrich Alexander University, Erlangen, Germany,Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Miguel Gonzalez-Acera
- Medical Clinic 1, University Clinic, Friedrich Alexander University, Erlangen, Germany
| | - Jay V Patankar
- Medical Clinic 1, University Clinic, Friedrich Alexander University, Erlangen, Germany
| | - Fabrizio Mascia
- Medical Clinic 1, University Clinic, Friedrich Alexander University, Erlangen, Germany
| | - Kristina Scheibe
- Medical Clinic 1, University Clinic, Friedrich Alexander University, Erlangen, Germany
| | - Markus Hoffmann
- Medical Clinic 3, University Clinic, Friedrich Alexander University, Erlangen, Germany
| | - Stefan Uderhardt
- Deutsches Zentrum Immuntherapie, Erlangen, Germany,Medical Clinic 3, University Clinic, Friedrich Alexander University, Erlangen, Germany
| | - Christine Schauer
- Medical Clinic 3, University Clinic, Friedrich Alexander University, Erlangen, Germany
| | | | - Clemens Neufert
- Medical Clinic 1, University Clinic, Friedrich Alexander University, Erlangen, Germany,Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Michael Vieth
- Friedrich Alexander University, Institute of Pathology, Klinikum Bayreuth, Erlangen, Germany
| | - Georg Schett
- Deutsches Zentrum Immuntherapie, Erlangen, Germany,Medical Clinic 3, University Clinic, Friedrich Alexander University, Erlangen, Germany
| | - Raja Atreya
- Medical Clinic 1, University Clinic, Friedrich Alexander University, Erlangen, Germany,Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Anja A Kühl
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andre Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Christoph Becker
- Medical Clinic 1, University Clinic, Friedrich Alexander University, Erlangen, Germany
| | - Martin Herrmann
- Medical Clinic 3, University Clinic, Friedrich Alexander University, Erlangen, Germany
| | - Markus F Neurath
- Medical Clinic 1, University Clinic, Friedrich Alexander University, Erlangen, Germany,Deutsches Zentrum Immuntherapie, Erlangen, Germany
| |
Collapse
|
44
|
Yadav SK, Ito N, Mindur JE, Kumar H, Youssef M, Suresh S, Kulkarni R, Rosario Y, Balashov KE, Dhib-Jalbut S, Ito K. Fecal Lcn-2 level is a sensitive biological indicator for gut dysbiosis and intestinal inflammation in multiple sclerosis. Front Immunol 2022; 13:1015372. [PMID: 36341389 PMCID: PMC9634083 DOI: 10.3389/fimmu.2022.1015372] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/04/2022] [Indexed: 07/30/2023] Open
Abstract
Multiple Sclerosis (MS) has been reported to be associated with intestinal inflammation and gut dysbiosis. To elucidate the underlying biology of MS-linked gut inflammation, we investigated gut infiltration of immune cells during the development of spontaneous experimental autoimmune encephalomyelitis (EAE) in humanized transgenic (Tg) mice expressing HLA-DR2a and human T cell receptor (TCR) specific for myelin basic protein peptide (MBP87-99)/HLA-DR2a complexes. Strikingly, we noted the simultaneous development of EAE and colitis, suggesting a link between autoimmune diseases of the central nervous system (CNS) and intestinal inflammation. Examination of the colon in these mice revealed the infiltration of MBP-specific Th17 cells as well as recruitment of neutrophils. Furthermore, we observed that fecal Lipocalin-2 (Lcn-2), a biomarker of intestinal inflammation, was significantly elevated and predominantly produced by the gut-infiltrating neutrophils. We then extended our findings to MS patients and demonstrate that their fecal Lcn-2 levels are significantly elevated compared to healthy donors (HDs). The elevation of fecal Lcn-2 levels correlated with reduced bacterial diversity and increased levels of other intestinal inflammation markers including neutrophil elastase and calprotectin. Of interest, bacteria thought to be beneficial for inflammatory bowel disease (IBD) such as Anaerobutyricum, Blautia, and Roseburia, were reduced in fecal Lcn-2-high MS patients. We also observed a decreasing trend in serum acetate (a short-chain fatty acid) levels in MS Lcn-2-high patients compared to HDs. Furthermore, a decrease in the relative abundance of Blautia massiliensis was significantly associated with a reduction of acetate in the serum of MS patients. This study suggests that gut infiltration of Th17 cells and recruitment of neutrophils are associated with the development of gut dysbiosis and intestinal inflammation, and that fecal Lcn-2 level is a sensitive biological indicator for gut dysbiosis in multiple sclerosis.
Collapse
Affiliation(s)
- Sudhir K. Yadav
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Naoko Ito
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - John E. Mindur
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Hetal Kumar
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Mysra Youssef
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, United States
- Department of Clinical and Chemical Pathology, National Research Centre, Dokki, Egypt
| | - Shradha Suresh
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Ratuja Kulkarni
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Yaritza Rosario
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Konstantin E. Balashov
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Suhayl Dhib-Jalbut
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, United States
- Department of Neurology, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Kouichi Ito
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, United States
| |
Collapse
|
45
|
Manzanares LD, David J, Ren X, Yalom LK, Piccolo EB, Dehghan Y, David AJ, Hanauer SB, Sumagin R. Atovaquone attenuates experimental colitis by reducing neutrophil infiltration of colonic mucosa. Front Pharmacol 2022; 13:1011115. [PMID: 36313299 PMCID: PMC9614091 DOI: 10.3389/fphar.2022.1011115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/12/2022] [Indexed: 10/08/2023] Open
Abstract
Ulcerative colitis (UC) is a chronic relapsing disease featuring aberrant accumulation of neutrophils in colonic mucosa and the luminal space. Although significant advances in UC therapy have been made with the development of novel biologics and small molecules targeting immune responses, success of most current therapies is still limited, with significant safety concerns. Thus, there is a need to develop additional safe and effective therapies for the treatment of UC. Antimalarial drugs have been safely used for many years to resolve tissue inflammation and the associated pathologies. Atovaquone is a recent FDA-approved antimalarial drug that has shown anti-viral and tumor-suppressive properties in vitro however, its role in mucosal inflammation has not been evaluated. Using pre-clinical murine DSS-induced colitis model combined with complementary in vivo peritonitis and ex vivo human neutrophil activation and chemotaxis assays we investigated functional and mechanistic impacts of atovaquone on disease resolution and neutrophil trafficking. We demonstrate that atovaquone promotes resolution of DSS-induced murine colitis by reducing neutrophil accumulation in the inflamed colonic mucosa. Mechanistically, we show that atovaquone suppressed induction of CD11b expression in neutrophils, reducing their polarization and migratory ability. Thus, our findings identify a new role of atovaquone in promoting resolution of mucosal inflammation, supporting the idea of potential repurposing of this FDA-approved drug as UC therapeutic.
Collapse
Affiliation(s)
- Laura D. Manzanares
- Laboratory 7-065 Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Joseph David
- Department of Medicine, Gastroenterology and Hepatology University of Arizona College of Medicine, Phoenix, AZ, United States
| | - Xingsheng Ren
- Laboratory 7-065 Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Lenore K. Yalom
- Laboratory 7-065 Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Enzo B. Piccolo
- Laboratory 7-065 Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Yalda Dehghan
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Aidan J. David
- College of Arts and Sciences, Case Western Reserve Unviersity, Cleveland, OH, United States
| | - Stephen B. Hanauer
- Department of Medicine, Gastroenterology and Hepatology Northwestern Memorial Hospital, Chicago, IL, United States
| | - Ronen Sumagin
- Laboratory 7-065 Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
46
|
Huang J, Hong W, Wan M, Zheng L. Molecular mechanisms and therapeutic target of NETosis in diseases. MedComm (Beijing) 2022; 3:e162. [PMID: 36000086 PMCID: PMC9390875 DOI: 10.1002/mco2.162] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/03/2022] [Accepted: 07/07/2022] [Indexed: 12/13/2022] Open
Abstract
Evidence shows that neutrophils can protect the host against pathogens in multiple ways, including the formation and release of neutrophil extracellular traps (NETs). NETs are web-like structures composed of fibers, DNA, histones, and various neutrophil granule proteins. NETs can capture and kill pathogens, including bacteria, viruses, fungi, and protozoa. The process of NET formation is called NETosis. According to whether they depend on nicotinamide adenine dinucleotide phosphate (NADPH), NETosis can be divided into two categories: "suicidal" NETosis and "vital" NETosis. However, NET components, including neutrophil elastase, myeloperoxidase, and cell-free DNA, cause a proinflammatory response and potentially severe diseases. Compelling evidence indicates a link between NETs and the pathogenesis of a number of diseases, including sepsis, systemic lupus erythematosus, rheumatoid arthritis, small-vessel vasculitis, inflammatory bowel disease, cancer, COVID-19, and others. Therefore, targeting the process and products of NETosis is critical for treating diseases linked with NETosis. Researchers have discovered that several NET inhibitors, such as toll-like receptor inhibitors and reactive oxygen species scavengers, can prevent uncontrolled NET development. This review summarizes the mechanism of NETosis, the receptors associated with NETosis, the pathology of NETosis-induced diseases, and NETosis-targeted therapy.
Collapse
Affiliation(s)
- Jiayu Huang
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| | - Meihua Wan
- Department of Integrated Traditional Chinese and Western MedicineWest China HospitalSichuan UniversityChengduSichuanChina
| | - Limin Zheng
- Guangdong Province Key Laboratory of Pharmaceutical Functional GenesMOE Key Laboratory of Gene Function and RegulationSchool of Life SciencesSun Yat‐Sen UniversityGuangzhouChina
- State Key Laboratory of Oncology in Southern ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouChina
| |
Collapse
|
47
|
Neutrophil Extracellular Traps and Pancreatic Cancer Development: A Vicious Cycle. Cancers (Basel) 2022; 14:cancers14143339. [PMID: 35884400 PMCID: PMC9318070 DOI: 10.3390/cancers14143339] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/03/2022] [Accepted: 07/07/2022] [Indexed: 12/07/2022] Open
Abstract
Neutrophil extracellular traps (NETs) are a neutrophil-generated extracellular network of chromatin and chromatin-bound molecules with antimicrobial potency. Recent data suggest that NETs are associated with cancer progression and cancer-associated hypercoagulability. Pancreatic adenocarcinoma (PDAC) is a lethal type of cancer in which hypercoagulability and cancer-related thrombosis are among the main complications. In the current report, we summarize the available data on the interplay between NET formation and PDAC development. We conclude that NETs support a dual role during PDAC progression and metastasis. Their formation is on the one hand an important event that shapes the cancer microenvironment to support cancer cell proliferation, invasion and metastasis. On the other hand, NETs may lead to cancer-associated thrombosis. Both mechanisms seem to be dependent on distinct molecular mechanisms that link inflammation to cancer progression. Collectively, NET formation may contribute to the pathogenesis of PDAC, while during cancer development, the proinflammatory environment enables the induction of new NETs and thrombi, forming a vicious cycle. We suggest that targeting NET formation may be an effective mechanism to inhibit both PDAC development and the accompanying hypercoagulability.
Collapse
|
48
|
Chen F, Liu Y, Shi Y, Zhang J, Liu X, Liu Z, Lv J, Leng Y. The emerging role of neutrophilic extracellular traps in intestinal disease. Gut Pathog 2022; 14:27. [PMID: 35733158 PMCID: PMC9214684 DOI: 10.1186/s13099-022-00497-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 05/19/2022] [Indexed: 11/10/2022] Open
Abstract
Neutrophil extracellular traps (NETs) are extracellular reticular fibrillar structures composed of DNA, histones, granulins and cytoplasmic proteins that are delivered externally by neutrophils in response to stimulation with various types of microorganisms, cytokines and host molecules, etc. NET formation has been extensively demonstrated to trap, immobilize, inactivate and kill invading microorganisms and acts as a form of innate response against pathogenic invasion. However, NETs are a double-edged sword. In the event of imbalance between NET formation and clearance, excessive NETs not only directly inflict tissue lesions, but also recruit pro-inflammatory cells or proteins that promote the release of inflammatory factors and magnify the inflammatory response further, driving the progression of many human diseases. The deleterious effects of excessive release of NETs on gut diseases are particularly crucial as NETs are more likely to be disrupted by neutrophils infiltrating the intestinal epithelium during intestinal disorders, leading to intestinal injury, and in addition, NETs and their relevant molecules are capable of directly triggering the death of intestinal epithelial cells. Within this context, a large number of NETs have been reported in several intestinal diseases, including intestinal infections, inflammatory bowel disease, intestinal ischemia–reperfusion injury, sepsis, necrotizing enterocolitis, and colorectal cancer. Therefore, the formation of NET would have to be strictly monitored to prevent their mediated tissue damage. In this review, we summarize the latest knowledge on the formation mechanisms of NETs and their pathophysiological roles in a variety of intestinal diseases, with the aim of providing an essential directional guidance and theoretical basis for clinical interventions in the exploration of mechanisms underlying NETs and targeted therapies.
Collapse
Affiliation(s)
- Feng Chen
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Yongqiang Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.,Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Yajing Shi
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Jianmin Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Xin Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.,Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Zhenzhen Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Jipeng Lv
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Yufang Leng
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China. .,Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.
| |
Collapse
|
49
|
Schroder AL, Chami B, Liu Y, Doyle CM, El Kazzi M, Ahlenstiel G, Ahmad G, Pathma-Nathan N, Collins G, Toh J, Harman A, Byrne S, Ctercteko G, Witting PK. Neutrophil Extracellular Trap Density Increases With Increasing Histopathological Severity of Crohn's Disease. Inflamm Bowel Dis 2022; 28:586-598. [PMID: 34724042 PMCID: PMC9036391 DOI: 10.1093/ibd/izab239] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Intestinal neutrophil recruitment is a characteristic feature of the earliest stages of inflammatory bowel disease (IBD). Neutrophil elastase (NE) and myeloperoxidase (MPO) mediate the formation of neutrophil extracellular traps (NETs); NETs produce the bactericidal oxidant hypochlorous acid (HOCl), causing host tissue damage when unregulated. The project aim was to investigate the relationship between NET formation and clinical IBD in humans. METHODS Human intestinal biopsies were collected from Crohn's disease (CD) patients, endoscopically categorized as unaffected, transitional, or diseased, and assigned a histopathological score. RESULTS A significant linear correlation was identified between pathological score and cell viability (TUNEL+). Immunohistochemical analysis revealed the presence of NET markers NE, MPO, and citrullinated histone (CitH3) that increased significantly with increasing histopathological score. Diseased specimens showed greater MPO+-immunostaining than control (P < .0001) and unaffected CD (P < .0001), with transitional CD specimens also showing greater staining than controls (P < .05) and unaffected CD (P < .05). Similarly, NE+-immunostaining was elevated significantly in diseased CD than controls (P < .0001) and unaffected CD (P < .0001) and was significantly higher in transitional CD than in controls (P < .0001) and unaffected CD (P < .0001). The CitH3+-immunostaining of diseased CD was significantly higher than controls (P < .05), unaffected CD (P < .0001) and transitional CD (P < .05), with transitional CD specimens showing greater staining than unaffected CD (P < .01). Multiplex immunohistochemistry with z-stacking revealed colocalization of NE, MPO, CitH3, and DAPI (cell nuclei), confirming the NET assignment. CONCLUSION These data indicate an association between increased NET formation and CD severity, potentially due to excessive MPO-mediated HOCl production in the extracellular domain, causing host tissue damage that exacerbates CD.
Collapse
Affiliation(s)
- Angie L Schroder
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, NSW, Australia
- Charles Perkins Centre, The University of Sydney, NSW, Australia
| | - Belal Chami
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, NSW, Australia
- Charles Perkins Centre, The University of Sydney, NSW, Australia
| | - Yuyang Liu
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, NSW, Australia
- Charles Perkins Centre, The University of Sydney, NSW, Australia
| | - Chloe M Doyle
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, NSW, Australia
- Westmead Institute for Medical Research, Centre for Immunology and Allergy Research, Westmead, NSW, Australia
| | - Mary El Kazzi
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, NSW, Australia
- Charles Perkins Centre, The University of Sydney, NSW, Australia
| | - Golo Ahlenstiel
- Western Sydney University, Westmead Clinical School and The Westmead Institute for Medical Research, Blacktown Hospital, Blacktown, NSW, Australia
| | - Gulfam Ahmad
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, NSW, Australia
- Charles Perkins Centre, The University of Sydney, NSW, Australia
| | - Nimalan Pathma-Nathan
- Westmead Institute for Medical Research, Centre for Immunology and Allergy Research, Westmead, NSW, Australia
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW,Australia
| | - Geoff Collins
- Westmead Institute for Medical Research, Centre for Immunology and Allergy Research, Westmead, NSW, Australia
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW,Australia
| | - James Toh
- Westmead Institute for Medical Research, Centre for Immunology and Allergy Research, Westmead, NSW, Australia
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW,Australia
- Department of Colorectal Surgery, Westmead Hospital, NSW,Australia
| | - Andrew Harman
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, NSW, Australia
- Westmead Institute for Medical Research, Centre for Immunology and Allergy Research, Westmead, NSW, Australia
| | - Scott Byrne
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, NSW, Australia
- Westmead Institute for Medical Research, Centre for Immunology and Allergy Research, Westmead, NSW, Australia
| | - Grahame Ctercteko
- Westmead Institute for Medical Research, Centre for Immunology and Allergy Research, Westmead, NSW, Australia
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW,Australia
- Department of Colorectal Surgery, Westmead Hospital, NSW,Australia
| | - Paul K Witting
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, NSW, Australia
- Charles Perkins Centre, The University of Sydney, NSW, Australia
| |
Collapse
|
50
|
Dysbiosis in Inflammatory Bowel Disease: Pathogenic Role and Potential Therapeutic Targets. Int J Mol Sci 2022; 23:ijms23073464. [PMID: 35408838 PMCID: PMC8998182 DOI: 10.3390/ijms23073464] [Citation(s) in RCA: 135] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022] Open
Abstract
Microbe-host communication is essential to maintain vital functions of a healthy host, and its disruption has been associated with several diseases, including Crohn's disease and ulcerative colitis, the two major forms of inflammatory bowel disease (IBD). Although individual members of the intestinal microbiota have been associated with experimental IBD, identifying microorganisms that affect disease susceptibility and phenotypes in humans remains a considerable challenge. Currently, the lack of a definition between what is healthy and what is a dysbiotic gut microbiome limits research. Nevertheless, although clear proof-of-concept of causality is still lacking, there is an increasingly evident need to understand the microbial basis of IBD at the microbial strain, genomic, epigenomic, and functional levels and in specific clinical contexts. Recent information on the role of diet and novel environmental risk factors affecting the gut microbiome has direct implications for the immune response that impacts the development of IBD. The complexity of IBD pathogenesis, involving multiple distinct elements, suggests the need for an integrative approach, likely utilizing computational modeling of molecular datasets to identify more specific therapeutic targets.
Collapse
|