1
|
Ma Z, Wen X, Zhang Y, Ai Z, Zhao X, Dong N, Dou X, Shan A. Thymol Alleviates Colitis by Modulating Intestinal Barrier Damage, Gut Microbiota, and Amino Acid Metabolic Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:7211-7227. [PMID: 40077957 DOI: 10.1021/acs.jafc.4c10406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Thymol (THY) is a phenolic monoterpene compound that has garnered attention due to its various biological properties, including antioxidant, anti-inflammatory, and immune-regulatory effects. The purpose of this study was to determine the therapeutic and protective effects of THY in colitic mice, with a particular focus on the mechanisms involving gut microbiota. The results showed that early intervention with THY (40 and 80 mg/kg) not only alleviated the clinical symptoms and colonic damage in mice with dextran sodium sulfate (DSS)-induced colitis but also suppressed the colonic production of inflammatory cytokines (IL-1β, IL-6, and IL-18) and enhanced the expression of mucins (MUC1 and MUC2) and trefoil factor family 3 (TFF3), thereby improving the integrity of the intestinal epithelial barrier. In addition, THY altered the composition of the gut microbiota in colitis mice by increasing the abundance of Bacteroides and reducing the abundance of Proteobacteria. Fecal microbial transplantation (FMT) results demonstrated that FM from THY donor mice significantly improved symptoms of inflammatory bowel disease (IBD), confirming the crucial role of the gut microbiota. Metagenomic and untargeted metabolomic studies found that the characteristic microbiota of THY is Prevotellaceae, and THY significantly upregulated the amino acid metabolic pathways related to arginine and proline metabolism, arginine biosynthesis, and glycerophospholipid metabolism. In summary, THY holds significant potential as a functional additive to enhance host intestinal activity.
Collapse
Affiliation(s)
- Ziwen Ma
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Xin Wen
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Yahan Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Zichun Ai
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Xinyi Zhao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Na Dong
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Xiujing Dou
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Anshan Shan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| |
Collapse
|
2
|
Parsad R, Ahlawat S, Bagiyal M, Gera R, Chhabra P, Sharma U, Arora R, Sharma R. Cathelicidins in farm animals: Structural diversity, mechanisms of action, and therapeutic potential in the face of antimicrobial resistance. Vet Immunol Immunopathol 2025; 279:110866. [PMID: 39708585 DOI: 10.1016/j.vetimm.2024.110866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/12/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Cathelicidins are a diverse family of antimicrobial peptides found across many vertebrate species, playing a pivotal role in the innate immune system. These peptides exhibit a variety of structural motifs, including α-helices, β-hairpins, and random coils, contributing to their broad-spectrum antimicrobial activity. The structural diversity of cathelicidins allows them to interact with a wide range of microbial targets, thereby enhancing their antimicrobial efficacy. Distinct species produce specific cathelicidins, each adapted to meet their unique immune requirements. Cathelicidins primarily function by disrupting microbial membranes, leading to cell lysis. Beyond their direct antimicrobial action, they possess immunomodulatory properties that bolster host defense mechanisms. These properties include promoting chemotaxis, enhancing phagocytosis, and inducing cytokine production, thereby modulating the host immune response. The therapeutic potential of cathelicidins is significant, especially in light of the growing challenge of antimicrobial resistance (AMR). As conventional antibiotics lose efficacy, cathelicidins emerge as promising alternatives due to their unique mechanisms of action and reduced likelihood of inducing resistance. Recent research underscores their potential in treating infections, inflammatory diseases, and even cancer. Advances in synthetic biology offer promising prospects for effective cathelicidin-based therapies in the future. This review summarizes the diversity, modes of action, and clinical prospects of cathelicidins specific to farm animals.
Collapse
Affiliation(s)
- Ram Parsad
- ICAR-National Bureau of Animal Genetic Resources, Karnal, India
| | - Sonika Ahlawat
- ICAR-National Bureau of Animal Genetic Resources, Karnal, India.
| | - Meena Bagiyal
- ICAR-National Bureau of Animal Genetic Resources, Karnal, India
| | - Ritika Gera
- ICAR-National Bureau of Animal Genetic Resources, Karnal, India
| | - Pooja Chhabra
- ICAR-National Bureau of Animal Genetic Resources, Karnal, India
| | - Upasna Sharma
- ICAR-National Bureau of Animal Genetic Resources, Karnal, India
| | - Reena Arora
- ICAR-National Bureau of Animal Genetic Resources, Karnal, India
| | - Rekha Sharma
- ICAR-National Bureau of Animal Genetic Resources, Karnal, India
| |
Collapse
|
3
|
Shih CC, Liao WC, Ke HY, Kuo CW, Tsao CM, Tsai WC, Chiu YL, Huang HC, Wu CC. Antimicrobial peptide cathelicidin LL-37 preserves intestinal barrier and organ function in rats with heat stroke. Biomed Pharmacother 2023; 161:114565. [PMID: 36958193 DOI: 10.1016/j.biopha.2023.114565] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023] Open
Abstract
Global warming increases the incidence of heat stroke (HS) and HS causes the reduction of visceral blood flow during hyperthermia, leading to intestinal barrier disruption, microbial translocation, systemic inflammation and multiple organ failure. Cathelicidin LL-37 exhibits antimicrobial activities, helps innate immunity within the gut to maintain intestinal homeostasis, and augments intestinal wound healing and barrier function. Thus, we evaluated the effects and possible mechanisms of cathelicidin LL-37 on HS. Wistar rats were placed in a heating-chamber of 42 ̊C to induce HS. Changes in rectal temperature, hemodynamic parameters, and survival rate were measured during the experimental period. Blood samples and ilea were collected to analyze the effects of LL-37 on systemic inflammation, multiple organ dysfunction, and intestinal injury. Furthermore, LS174T and HT-29 cells were used to assess the underlying mechanisms. Our data showed cathelicidin LL-37 ameliorated the damage of intestinal cells induced by HS. Intestinal injury, systemic inflammation, and nitrosative stress (high nitric oxide level) caused by continuous hyperthermia were attenuated in HS rats treated with cathelicidin LL-37, and hence, improved multiple organ dysfunction, coagulopathy, and survival rate. These beneficial effects of cathelicidin LL-37 were attributed to the protection of intestinal goblet cells (by increasing transepithelial resistance, mucin-2 and Nrf2 expression) and the improvement of intestinal barrier function (less cyclooxygenase-2 expression and FITC-dextran translocation). Interestingly, high cathelicidin expression in the ileal samples of inflammatory bowel disease patients was associated with better clinical outcome. These results suggest that cathelicidin LL-37 could prevent heat stress-induced intestinal damage and heat-related illnesses.
Collapse
Affiliation(s)
- Chih-Chin Shih
- Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei, Taiwan, ROC.
| | - Wei-Chieh Liao
- Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Hung-Yen Ke
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chia-Wen Kuo
- Department of Nephrology, Taichung Armed Forces General Hospital, Taichung, Taiwan, ROC
| | - Cheng-Ming Tsao
- Department of Anesthesiology, Taipei Veterans General Hospital and National Yang-Ming Chiao-Tung University, Taipei, Taiwan, ROC
| | - Wen-Chiuan Tsai
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Yi-Lin Chiu
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Hsieh-Chou Huang
- Department of Anesthesiology, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
| | - Chin-Chen Wu
- Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei, Taiwan, ROC.
| |
Collapse
|
4
|
Cardoso MH, Meneguetti BT, Oliveira-Júnior NG, Macedo MLR, Franco OL. Antimicrobial peptide production in response to gut microbiota imbalance. Peptides 2022; 157:170865. [PMID: 36038014 DOI: 10.1016/j.peptides.2022.170865] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/18/2022]
Abstract
The gut microbiota presents essential functions in the immune response. The gut epithelium acts as a protective barrier and, therefore, can produce several antimicrobial peptides (AMPs) that can act against pathogenic microorganisms, including bacteria. Several factors cause a disturbance in gut microbiota, including the exacerbated and erroneous use of antibiotics. Antibiotic therapy has been closely related to bacterial resistance and is also correlated with undesired side-effects to the host, including the eradication of commensal bacteria. Consequently, this results in gut microbiota imbalance and inflammatory bowel diseases (IBD) development. In this context, AMPs in the gut epithelium play a restructuring role for gut microbiota. Some naturally occurring AMPs are selective for pathogenic bacteria, thus preserving the health microbiota. Therefore, AMPs produced by the host's epithelial cells represent effective molecules in treating gut bacterial infections. Bearing this in mind, this review focused on describing the importance of the host's AMPs in gut microbiota modulation and their role as anti-infective agents against pathogenic bacteria.
Collapse
Affiliation(s)
- Marlon H Cardoso
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS 79117900, Brazil; Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF 70790160, Brazil; Laboratório de Purificação de Proteínas e suas Funções Biológicas, Universidade Federal de Mato Grosso do Sul, Cidade Universitária, 79070900 Campo Grande, Mato Grosso do Sul, Brazil.
| | - Beatriz T Meneguetti
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS 79117900, Brazil
| | - Nelson G Oliveira-Júnior
- Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF 70790160, Brazil
| | - Maria L R Macedo
- Laboratório de Purificação de Proteínas e suas Funções Biológicas, Universidade Federal de Mato Grosso do Sul, Cidade Universitária, 79070900 Campo Grande, Mato Grosso do Sul, Brazil
| | - Octávio L Franco
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS 79117900, Brazil; Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF 70790160, Brazil.
| |
Collapse
|
5
|
Martell EM, González-Garcia M, Ständker L, Otero-González AJ. Host defense peptides as immunomodulators: The other side of the coin. Peptides 2021; 146:170644. [PMID: 34464592 DOI: 10.1016/j.peptides.2021.170644] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 12/13/2022]
Abstract
Host defense peptides (HDPs) exhibit a broad range of antimicrobial and immunomodulatory activities. In this sense, both functions are like different sides of the same coin. The direct antimicrobial side was discovered first, and widely studied for the development of anti-infective therapies. In contrast, the immunomodulatory side was recognized later and in the last 20 years the interest in this field has been continuously growing. Different to their antimicrobial activities, the immunomodulatory activities of host defense peptides are more effective in vivo. They offer a great opportunity for new therapeutic applications in the fields of anti-infective therapy, chronic inflammatory diseases treatment, novel vaccine adjuvants development and anticancer immunotherapy. These immune related functions of HDPs includes chemoattraction of leukocytes, modulation of inflammation, enhancement of antigen presentation and polarization of adaptive immune responses. Our attempt with this review is to make a careful evaluation of different aspects of the less explored, but attractive immunomodulatory side of the HDP functional coin.
Collapse
Affiliation(s)
- Ernesto M Martell
- Center for Protein Studies, Faculty of Biology, Havana University, Cuba
| | | | - Ludger Ständker
- Core Facility Functional Peptidomics (CFP), Ulm University Medical Center, Ulm, Germany
| | | |
Collapse
|
6
|
Fabisiak N, Fabisiak A, Chmielowiec-Korzeniowska A, Tymczyna L, Kamysz W, Kordek R, Bauer M, Kamysz E, Fichna J. Anti-inflammatory and antibacterial effects of human cathelicidin active fragment KR-12 in the mouse models of colitis: a novel potential therapy of inflammatory bowel diseases. Pharmacol Rep 2020; 73:163-171. [PMID: 33219923 PMCID: PMC7862075 DOI: 10.1007/s43440-020-00190-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 10/31/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022]
Abstract
Introduction Inflammatory bowel diseases (IBD) are a group of chronic gastrointestinal tract disorders with complex etiology, with intestinal dysbiosis as the most prominent factor. In this study, we assessed the anti-inflammatory and antibacterial actions of the human cathelicidin LL-37 and its shortest active fragment, KR-12 in the mouse models of colitis. Materials and methods Mouse models of colitis induced by 2,4,6-trinitrobenzenesulfonic acid (TNBS) and dextran sulfate sodium (DSS) were used in the study. The extent of inflammation was evaluated based on the macro- and microscopic scores, quantification of myeloperoxidase (MPO) activity and microbiological analysis of stool samples. Results A preliminary study with LL-37 and KR-12 (1 mg/kg, ip, twice daily) showed a decrease in macroscopic and ulcer scores in the acute TNBS-induced model of colitis. We observed that KR-12 (5 mg/kg, ip, twice daily) reduced microscopic and ulcer scores in the semi-chronic and chronic TNBS-induced models of colitis compared with inflamed mice. Furthermore, qualitative and quantitative changes in colonic microbiota were observed: KR-12 (5 mg/kg, ip, twice daily) decreased the overall number of bacteria, Escherichia coli and coli group bacteria. In the semi-chronic DSS-induced model, KR-12 attenuated intestinal inflammation as demonstrated by a reduction in macroscopic score and colon damage score and MPO activity. Conclusions We demonstrated that KR-12 alleviates inflammation in four different mouse models of colitis what suggests KR-12 and cathelicidins as a whole are worth being considered as a potential therapeutic option in the treatment of IBD.
Collapse
Affiliation(s)
- Natalia Fabisiak
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 6/8, 92-215, Lodz, Poland
- Department of Gastroenterology, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Adam Fabisiak
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 6/8, 92-215, Lodz, Poland
- Department of Digestive Tract Diseases, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | | | - Leszek Tymczyna
- Department of Animal Hygiene and Environment, University of Agriculture in Lublin, Lublin, Poland
| | - Wojciech Kamysz
- Department of Inorganic Chemistry, Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| | - Radzisław Kordek
- Department of Pathology, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Marta Bauer
- Department of Inorganic Chemistry, Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| | - Elżbieta Kamysz
- Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 6/8, 92-215, Lodz, Poland.
| |
Collapse
|
7
|
Scheenstra MR, van Harten RM, Veldhuizen EJA, Haagsman HP, Coorens M. Cathelicidins Modulate TLR-Activation and Inflammation. Front Immunol 2020; 11:1137. [PMID: 32582207 PMCID: PMC7296178 DOI: 10.3389/fimmu.2020.01137] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/11/2020] [Indexed: 12/30/2022] Open
Abstract
Cathelicidins are short cationic peptides that are part of the innate immune system. At first, these peptides were studied mostly for their direct antimicrobial killing capacity, but nowadays they are more and more appreciated for their immunomodulatory functions. In this review, we will provide a comprehensive overview of the various effects cathelicidins have on the detection of damage- and microbe-associated molecular patterns, with a special focus on their effects on Toll-like receptor (TLR) activation. We review the available literature based on TLR ligand types, which can roughly be divided into lipidic ligands, such as LPS and lipoproteins, and nucleic-acid ligands, such as RNA and DNA. For both ligand types, we describe how direct cathelicidin-ligand interactions influence TLR activation, by for instance altering ligand stability, cellular uptake and receptor interaction. In addition, we will review the more indirect mechanisms by which cathelicidins affect downstream TLR-signaling. To place all this information in a broader context, we discuss how these cathelicidin-mediated effects can have an impact on how the host responds to infectious organisms as well as how these effects play a role in the exacerbation of inflammation in auto-immune diseases. Finally, we discuss how these immunomodulatory activities can be exploited in vaccine development and cancer therapies.
Collapse
Affiliation(s)
- Maaike R Scheenstra
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, Netherlands
| | - Roel M van Harten
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, Netherlands
| | - Edwin J A Veldhuizen
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, Netherlands
| | - Henk P Haagsman
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, Netherlands
| | - Maarten Coorens
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.,Department of Clinical Microbiology, Karolinska University Laboratory, Stockholm, Sweden
| |
Collapse
|
8
|
Yang B, Good D, Mosaiab T, Liu W, Ni G, Kaur J, Liu X, Jessop C, Yang L, Fadhil R, Yi Z, Wei MQ. Significance of LL-37 on Immunomodulation and Disease Outcome. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8349712. [PMID: 32509872 PMCID: PMC7246396 DOI: 10.1155/2020/8349712] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 03/04/2020] [Accepted: 04/03/2020] [Indexed: 02/07/2023]
Abstract
LL-37, also called cathelicidin, is an important part of the human immune system, which can resist various pathogens. A plethora of experiments have demonstrated that it has the multifunctional effects of immune regulation, in addition to antimicrobial activity. Recently, there have been increasing interest in its immune function. It was found that LL-37 can have two distinct functions in different tissues and different microenvironments. Thus, it is necessary to investigate LL-37 immune functions from the two sides of the same coin. On the one side, LL-37 promotes inflammation and immune response and exerts its anti-infective and antitumor effects; on the other side, it has the ability to inhibit inflammation and promote carcinogenesis. This review presents a brief summary of its expression, structure, and immunomodulatory effects as well as brief discussions on the role of this small peptide as a key factor in the development and treatment of various inflammation-related diseases and cancers.
Collapse
Affiliation(s)
- Binbin Yang
- School of Medical Laboratory, Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong, Weifang Medical University, Weifang 261053, China
- School of Medical Science & Menzies Health Institute Queensland, Griffith University, Gold Coast, Qld 4215, Australia
| | - David Good
- School of Medical Science & Menzies Health Institute Queensland, Griffith University, Gold Coast, Qld 4215, Australia
- School of Allied Health, Australian Catholic University, Brisbane, Qld 4014, Australia
| | - Tamim Mosaiab
- School of Medical Science & Menzies Health Institute Queensland, Griffith University, Gold Coast, Qld 4215, Australia
- Institute for Glycomics, Griffith University, Gold Coast, Qld 4215, Australia
| | - Wei Liu
- School of Medical Laboratory, Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong, Weifang Medical University, Weifang 261053, China
- School of Medical Science & Menzies Health Institute Queensland, Griffith University, Gold Coast, Qld 4215, Australia
| | - Guoying Ni
- School of Medical Science & Menzies Health Institute Queensland, Griffith University, Gold Coast, Qld 4215, Australia
- The First Affiliated Hospital/School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
- School of Health and Sport Sciences, University of the Sunshine Coast, Maroochydore DC, Qld 4558, Australia
| | - Jasmine Kaur
- School of Medical Science & Menzies Health Institute Queensland, Griffith University, Gold Coast, Qld 4215, Australia
| | - Xiaosong Liu
- The First Affiliated Hospital/School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
- School of Health and Sport Sciences, University of the Sunshine Coast, Maroochydore DC, Qld 4558, Australia
- Cancer Research Institute, First People's Hospital of Foshan, Foshan 528000, China
| | - Calvin Jessop
- School of Medical Science & Menzies Health Institute Queensland, Griffith University, Gold Coast, Qld 4215, Australia
| | - Lu Yang
- School of Medical Laboratory, Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong, Weifang Medical University, Weifang 261053, China
- School of Medical Science & Menzies Health Institute Queensland, Griffith University, Gold Coast, Qld 4215, Australia
| | - Rushdi Fadhil
- School of Medical Science & Menzies Health Institute Queensland, Griffith University, Gold Coast, Qld 4215, Australia
| | - Zhengjun Yi
- School of Medical Laboratory, Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong, Weifang Medical University, Weifang 261053, China
| | - Ming Q. Wei
- School of Medical Science & Menzies Health Institute Queensland, Griffith University, Gold Coast, Qld 4215, Australia
| |
Collapse
|
9
|
Mookherjee N, Anderson MA, Haagsman HP, Davidson DJ. Antimicrobial host defence peptides: functions and clinical potential. Nat Rev Drug Discov 2020; 19:311-332. [DOI: 10.1038/s41573-019-0058-8] [Citation(s) in RCA: 425] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2019] [Indexed: 12/18/2022]
|
10
|
Lee KH, Ahn BS, Cha D, Jang WW, Choi E, Park S, Park JH, Oh J, Jung DE, Park H, Park JH, Suh Y, Jin D, Lee S, Jang YH, Yoon T, Park MK, Seong Y, Pyo J, Yang S, Kwon Y, Jung H, Lim CK, Hong JB, Park Y, Choi E, Shin JI, Kronbichler A. Understanding the immunopathogenesis of autoimmune diseases by animal studies using gene modulation: A comprehensive review. Autoimmun Rev 2020; 19:102469. [PMID: 31918027 DOI: 10.1016/j.autrev.2020.102469] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 09/20/2019] [Indexed: 12/21/2022]
Abstract
Autoimmune diseases are clinical syndromes that result from pathogenic inflammatory responses driven by inadequate immune activation by T- and B-cells. Although the exact mechanisms of autoimmune diseases are still elusive, genetic factors also play an important role in the pathogenesis. Recently, with the advancement of understanding of the immunological and molecular basis of autoimmune diseases, gene modulation has become a potential approach for the tailored treatment of autoimmune disorders. Gene modulation can be applied to regulate the levels of interleukins (IL), tumor necrosis factor (TNF), cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), interferon-γ and other inflammatory cytokines by inhibiting these cytokine expressions using short interfering ribonucleic acid (siRNA) or by inhibiting cytokine signaling using small molecules. In addition, gene modulation delivering anti-inflammatory cytokines or cytokine antagonists showed effectiveness in regulating autoimmunity. In this review, we summarize the potential target genes for gene or immunomodulation in autoimmune diseases including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), inflammatory bowel diseases (IBD) and multiple sclerosis (MS). This article will give a new perspective on understanding immunopathogenesis of autoimmune diseases not only in animals but also in human. Emerging approaches to investigate cytokine regulation through gene modulation may be a potential approach for the tailored immunomodulation of some autoimmune diseases near in the future.
Collapse
Affiliation(s)
- Keum Hwa Lee
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byung Soo Ahn
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dohyeon Cha
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Won Woo Jang
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eugene Choi
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soohyun Park
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jun Hyeong Park
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Junseok Oh
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Da Eun Jung
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Heeryun Park
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ju Ha Park
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Youngsong Suh
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dongwan Jin
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Siyeon Lee
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Hwan Jang
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tehwook Yoon
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min-Kyu Park
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoonje Seong
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jihoon Pyo
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sunmo Yang
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Youngin Kwon
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyunjean Jung
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chae Kwang Lim
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jun Beom Hong
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yeoeun Park
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eunjin Choi
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Andreas Kronbichler
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
11
|
Zong X, Cao X, Wang H, Xiao X, Wang Y, Lu Z. Cathelicidin-WA Facilitated Intestinal Fatty Acid Absorption Through Enhancing PPAR-γ Dependent Barrier Function. Front Immunol 2019; 10:1674. [PMID: 31379865 PMCID: PMC6650583 DOI: 10.3389/fimmu.2019.01674] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 07/04/2019] [Indexed: 01/28/2023] Open
Abstract
The molecular mechanisms underlying the cellular uptake of long-chain fatty acids and the regulation of this process have been debated in recent decades. Here, we established an intestinal barrier dysfunction model in mice and Caco2 cell line by Lipopolysaccharide (LPS), and evaluated the fatty acid uptake capacity of the intestine. We found that LPS stimulation restricted the absorption of long chain fatty acid (LCFA), while Cathelicidin-WA (CWA) pretreatment facilitated this physiological process. At the molecular level, our results demonstrated that the stimulatory effects of CWA on intestinal lipid absorption were dependent on cluster determinant 36 and fatty acid transport protein 4, but not fatty acid–binding protein. Further, an enhanced intestinal barrier was observed in vivo and in vitro when CWA alleviated the fatty acid absorption disorder induced by LPS stimulation. Mechanistically, peroxisome proliferator-activated receptor (PPAR-γ) signaling was considered as a key pathway for CWA to enhance LCFA absorption and barrier function. Treatment with a PPAR-γ inhibitor led to impaired intestinal barrier function and suppressed LCFA uptake. Moreover, once PPAR-γ signaling was blocked, CWA pretreatment could not maintain the stability of the intestinal epithelial cell barrier or LCFA uptake after LPS stimulation. Collectively, these findings suggested that PPAR-γ may serve as a target for specific therapies aimed at alleviating fatty acid uptake disorder, and CWA showed considerable potential as a new PPAR-γ agonist to strengthen intestinal barrier function against fatty acid malabsorption.
Collapse
Affiliation(s)
- Xin Zong
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Xiaoxuan Cao
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Hong Wang
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Xiao Xiao
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yizhen Wang
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Zeqing Lu
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
12
|
Huynh E, Penney J, Caswell J, Li J. Protective Effects of Protegrin in Dextran Sodium Sulfate-Induced Murine Colitis. Front Pharmacol 2019; 10:156. [PMID: 30873029 PMCID: PMC6403130 DOI: 10.3389/fphar.2019.00156] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 02/08/2019] [Indexed: 12/12/2022] Open
Abstract
Cathelicidins, a class of antimicrobial peptides, have been widely studied for their antimicrobial role in innate immune responses during infection and inflammation. At sub-antimicrobial concentrations, various cathelicidins from different species have been reported to exert chemotactic activity on neutrophils, monocytes, dendritic cells and T-cells, and also enhance angiogenesis and wound healing. To date, the role of the pig cathelicidin, protegrin-1 (PG-1), in immune modulation and tissue repair in the intestinal tract has not been investigated. The aim of the present study was to examine the potential protective effects of recombinant PG-1 in a mouse dextran sodium sulfate (DSS)-induced colitis inflammation model. This is the first report showing the protective effects of PG-1 in its various forms (pro-, cathelin-, and mature-forms) in attenuating significant body weight loss associated with DSS-induced colitis (p < 0.05). PG-1 treatment improved histological scores (P < 0.05) and influenced the gene expression of inflammatory mediators and tissue repair factors such as trefoil factor 3 (TFF3) and mucin (MUC-2). Protegrin treatment also altered the metabolite profile, returning the metabolite levels closer to untreated control levels. These findings lay the foundation for future oral application of recombinant PG-1 to potentially treat intestinal damage and inflammation.
Collapse
Affiliation(s)
- Evanna Huynh
- Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON, Canada
| | - Jenna Penney
- Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON, Canada
| | - Jeff Caswell
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Julang Li
- Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON, Canada
- Department of Life Science and Engineering, Foshan University, Foshan, China
| |
Collapse
|
13
|
Miranda PM, De Palma G, Serkis V, Lu J, Louis-Auguste MP, McCarville JL, Verdu EF, Collins SM, Bercik P. High salt diet exacerbates colitis in mice by decreasing Lactobacillus levels and butyrate production. MICROBIOME 2018; 6:57. [PMID: 29566748 PMCID: PMC5865374 DOI: 10.1186/s40168-018-0433-4] [Citation(s) in RCA: 198] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 03/05/2018] [Indexed: 05/19/2023]
Abstract
BACKGROUND Changes in hygiene and dietary habits, including increased consumption of foods high in fat, simple sugars, and salt that are known to impact the composition and function of the intestinal microbiota, may explain the increase in prevalence of chronic inflammatory diseases. High salt consumption has been shown to worsen autoimmune encephalomyelitis and colitis in mouse models through p38/MAPK signaling pathway. However, the effect of high salt diet (HSD) on gut microbiota and on intestinal immune homeostasis, and their roles in determining vulnerability to intestinal inflammatory stimuli are unknown. Here, we investigate the role of gut microbiota alterations induced by HSD on the severity of murine experimental colitis. RESULTS Compared to control diet, HSD altered fecal microbiota composition and function, reducing Lactobacillus sp. relative abundance and butyrate production. Moreover, HSD affected the colonic, and to a lesser extent small intestine mucosal immunity by enhancing the expression of pro-inflammatory genes such as Rac1, Map2k1, Map2k6, Atf2, while suppressing many cytokine and chemokine genes, such as Ccl3, Ccl4, Cxcl2, Cxcr4, Ccr7. Conventionally raised mice fed with HSD developed more severe DSS- (dextran sodium sulfate) and DNBS- (dinitrobenzene sulfonic acid) induced colitis compared to mice on control diet, and this effect was absent in germ-free mice. Transfer experiments into germ-free mice indicated that the HSD-associated microbiota profile is critically dependent on continued exposure to dietary salt. CONCLUSIONS Our results indicate that the exacerbation of colitis induced by HSD is associated with reduction in Lactobacillus sp. and protective short-chain fatty acid production, as well as changes in host immune status. We hypothesize that these changes alter gut immune homeostasis and lead to increased vulnerability to inflammatory insults.
Collapse
Affiliation(s)
- Pedro M. Miranda
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario Canada
- Graduate Program in Areas of Basic and Applied Biology, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Giada De Palma
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario Canada
| | - Viktoria Serkis
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario Canada
| | - Jun Lu
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario Canada
| | - Marc P. Louis-Auguste
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario Canada
| | - Justin L. McCarville
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario Canada
| | - Elena F. Verdu
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario Canada
| | - Stephen M. Collins
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario Canada
| | - Premysl Bercik
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario Canada
| |
Collapse
|
14
|
Shen J, Xiao Z. Cathelicidin in Gastrointestinal Disorders. ANTIMICROBIAL PEPTIDES IN GASTROINTESTINAL DISEASES 2018:61-76. [DOI: 10.1016/b978-0-12-814319-3.00004-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
15
|
Li Y, Chu X, Liu C, Huang W, Yao Y, Xia Y, Sun P, Long Q, Feng X, Li K, Yang X, Bai H, Sun W, Ma Y. Exogenous murine antimicrobial peptide CRAMP significantly exacerbates Ovalbumin-induced airway inflammation but ameliorates oxazolone-induced intestinal colitis in BALB/c mice. Hum Vaccin Immunother 2017; 14:146-158. [PMID: 29049008 DOI: 10.1080/21645515.2017.1386823] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Cathelicidin has been reported to be multifunctional. The current study aimed to investigate the influences of exogenous cathelicidin-related antimicrobial peptide (CRAMP) on inflammatory responses in different disease models. In OVA-induced allergic airway inflammation, CRAMP significantly enhanced the infiltration of inflammatory cells and accumulation of proinflammatory Th2 cytokine IL-13 and IL-33 in bronchial alveolar lavage fluid (BALF), exacerbated lung tissue inflammation and airway goblet cell hyperplasia, and elevated OVA-specific IgE level in serum. In oxazolone-induced intestinal colitis, the expression levels of CRAMP and its receptor FPR2 significantly increased in comparison with those of TNBS-induced mice, vesicle and normal controls. Exogenous CRAMP significantly prevented the development of ulcerative colitis, evidenced by improved body weight regain, decreased colons weight/length ratio, elevated epithelial integrity, and ameliorated colon tissue inflammation. In addition, pro-inflammatory cytokines TNF-α, IL-1β, IL-4 and IL-13, as well as chemokines CXCL2 and CXCL5 for neutrophils recruitment were significantly decreased in CRAMP-treated mice, and epithelial repair-related factors MUC2 and Claudin1 were increased, determined by real time-PCR and ELISAs. The results indicated that although CRAMP has pro-inflammatory effects in airway, local application of exogenous CRAMP might be a potential approach for the treatment of ulcerative colitis.
Collapse
Affiliation(s)
- Yang Li
- a Laboratory of Molecular Immunology , Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College , Kunming , China.,b Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases , Kunming , China.,c Yunnan Engineering Research Center of Vaccine Research and Development on Severe Infectious Diseases , Kunming , China
| | - Xiaojie Chu
- a Laboratory of Molecular Immunology , Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College , Kunming , China.,b Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases , Kunming , China.,c Yunnan Engineering Research Center of Vaccine Research and Development on Severe Infectious Diseases , Kunming , China
| | - Cunbao Liu
- a Laboratory of Molecular Immunology , Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College , Kunming , China.,b Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases , Kunming , China.,c Yunnan Engineering Research Center of Vaccine Research and Development on Severe Infectious Diseases , Kunming , China
| | - Weiwei Huang
- a Laboratory of Molecular Immunology , Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College , Kunming , China.,b Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases , Kunming , China.,c Yunnan Engineering Research Center of Vaccine Research and Development on Severe Infectious Diseases , Kunming , China
| | - Yufeng Yao
- a Laboratory of Molecular Immunology , Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College , Kunming , China.,b Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases , Kunming , China.,c Yunnan Engineering Research Center of Vaccine Research and Development on Severe Infectious Diseases , Kunming , China
| | - Ye Xia
- a Laboratory of Molecular Immunology , Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College , Kunming , China.,b Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases , Kunming , China.,c Yunnan Engineering Research Center of Vaccine Research and Development on Severe Infectious Diseases , Kunming , China
| | - Pengyan Sun
- a Laboratory of Molecular Immunology , Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College , Kunming , China.,b Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases , Kunming , China.,c Yunnan Engineering Research Center of Vaccine Research and Development on Severe Infectious Diseases , Kunming , China
| | - Qiong Long
- a Laboratory of Molecular Immunology , Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College , Kunming , China.,b Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases , Kunming , China.,c Yunnan Engineering Research Center of Vaccine Research and Development on Severe Infectious Diseases , Kunming , China
| | - Xuejun Feng
- a Laboratory of Molecular Immunology , Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College , Kunming , China.,b Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases , Kunming , China.,c Yunnan Engineering Research Center of Vaccine Research and Development on Severe Infectious Diseases , Kunming , China
| | - Kui Li
- a Laboratory of Molecular Immunology , Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College , Kunming , China.,b Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases , Kunming , China.,c Yunnan Engineering Research Center of Vaccine Research and Development on Severe Infectious Diseases , Kunming , China
| | - Xu Yang
- a Laboratory of Molecular Immunology , Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College , Kunming , China.,b Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases , Kunming , China.,c Yunnan Engineering Research Center of Vaccine Research and Development on Severe Infectious Diseases , Kunming , China
| | - Hongmei Bai
- a Laboratory of Molecular Immunology , Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College , Kunming , China.,b Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases , Kunming , China.,c Yunnan Engineering Research Center of Vaccine Research and Development on Severe Infectious Diseases , Kunming , China
| | - Wenjia Sun
- a Laboratory of Molecular Immunology , Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College , Kunming , China.,b Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases , Kunming , China.,c Yunnan Engineering Research Center of Vaccine Research and Development on Severe Infectious Diseases , Kunming , China
| | - Yanbing Ma
- a Laboratory of Molecular Immunology , Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College , Kunming , China.,b Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases , Kunming , China.,c Yunnan Engineering Research Center of Vaccine Research and Development on Severe Infectious Diseases , Kunming , China
| |
Collapse
|
16
|
MUC2 Mucin and Butyrate Contribute to the Synthesis of the Antimicrobial Peptide Cathelicidin in Response to Entamoeba histolytica- and Dextran Sodium Sulfate-Induced Colitis. Infect Immun 2017; 85:IAI.00905-16. [PMID: 28069814 DOI: 10.1128/iai.00905-16] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 12/21/2016] [Indexed: 12/14/2022] Open
Abstract
Embedded in the colonic mucus are cathelicidins, small cationic peptides secreted by colonic epithelial cells. Humans and mice have one cathelicidin-related antimicrobial peptide (CRAMP) each, LL-37/hCAP-18 and Cramp, respectively, with related structure and functions. Altered production of MUC2 mucin and antimicrobial peptides is characteristic of intestinal amebiasis. The interactions between MUC2 mucin and cathelicidins in conferring innate immunity against Entamoeba histolytica are not well characterized. In this study, we quantified whether MUC2 expression and release could regulate the expression and secretion of cathelicidin LL-37 in colonic epithelial cells and in the colon. The synthesis of LL-37 was enhanced with butyrate (a product of bacterial fermentation) and interleukin-1β (IL-1β) (a proinflammatory cytokine in colitis) in the presence of exogenously added purified MUC2. The LL-37 responses to butyrate and IL-1β were higher in high-MUC2-producing cells than in lentivirus short hairpin RNA (shRNA) MUC2-silenced cells. Activation of cyclic adenylyl cyclase (AMP) and mitogen-activated protein kinase (MAPK) signaling pathways was necessary for the simultaneous expression of MUC2 and cathelicidins. In Muc2 mucin-deficient (Muc2-/-) mice, murine cathelicidin (Cramp) was significantly reduced compared to that in Muc2+/- and Muc2+/+ littermates. E. histolytica-induced acute inflammation in colonic loops stimulated high levels of cathelicidin in Muc2+/+ but not in Muc2-/- littermates. In dextran sodium sulfate (DSS)-induced colitis in Muc2+/+ mice, which depletes the mucus barrier and goblet cell mucin, Cramp expression was significantly enhanced during restitution. These studies demonstrate regulatory mechanisms between MUC2 and cathelicidins in the colonic mucosa where an intact mucus barrier is essential for expression and secretion of cathelicidins in response to E. histolytica- and DSS-induced colitis.
Collapse
|
17
|
Pinheiro da Silva F, Machado MCC. The dual role of cathelicidins in systemic inflammation. Immunol Lett 2017; 182:57-60. [PMID: 28082134 DOI: 10.1016/j.imlet.2017.01.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/11/2016] [Accepted: 01/04/2017] [Indexed: 01/10/2023]
Abstract
Antimicrobial peptides are key components of the innate immune system. They act as broad-spectrum antimicrobial agents against Gram-positive and -negative bacteria, viruses, and fungi. More recently, antimicrobial peptides have been ascribed immunomodulatory functions, including roles in wound healing, induction of cytokines, and altering host gene expression. Cathelicidins are a class of antimicrobial peptide found in humans, mice, and rats, among others. Known as LL-37 in humans and cathelin-related antimicrobial peptide (CRAMP) in rodents, cathelicidins are produced by many different cells, including macrophages, neutrophils, and epithelial cells. The role of cathelicidins is somewhat confounding, as they exhibit both pro-and anti-inflammatory activity. A major obstacle in the study of cathelicidins is the inability of exogenous LL-37 or CRAMP to mimic the activity of their endogenous counterparts. Nevertheless, studies have shown that LL-37 is recognized by multiple receptors, and may stabilize or modulate Toll-like receptor signaling. In addition, cathelicidins play a role in apoptosis, inflammasome activation, and phagocytosis. However, many studies are revealing the dual effects of cathelicidins. For example, CRAMP appears to be protective in models of group A Streptococcus skin infection, pneumonia, and meningitis, but detrimental in cases of severe bacterial infection, such as septic shock. It is becoming increasingly clear that the activity of cathelicidins is modulated by complex interactions with the microenvironment, as well as the disease background. This article reviews what is currently known about the activity of cathelicidins in an attempt to understand their complex roles in systemic diseases.
Collapse
|
18
|
Yi H, Hu W, Chen S, Lu Z, Wang Y. Cathelicidin-WA Improves Intestinal Epithelial Barrier Function and Enhances Host Defense against Enterohemorrhagic Escherichia coli O157:H7 Infection. THE JOURNAL OF IMMUNOLOGY 2017; 198:1696-1705. [PMID: 28062699 DOI: 10.4049/jimmunol.1601221] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 12/06/2016] [Indexed: 01/28/2023]
Abstract
Impaired epithelial barrier function disrupts immune homeostasis and increases inflammation in intestines, leading to many intestinal diseases. Cathelicidin peptides suppress intestinal inflammation and improve intestinal epithelial barrier function independently of their antimicrobial activity. In this study, we investigated the effects of Cathelicidin-WA (CWA) on intestinal epithelial barrier function, as well as the underlying mechanism, by using enterohemorrhagic Escherichia coli (EHEC)-infected mice and intestinal epithelial cells. The results showed that CWA attenuated EHEC-induced clinical symptoms and intestinal colitis, as did enrofloxacin (Enro). CWA decreased IL-6 production in the serum, jejunum, and colon of EHEC-infected mice. Additionally, CWA alleviated the EHEC-induced disruption of mucin-2 and goblet cells in the intestine. Interestingly, CWA increased the mucus layer thickness, which was associated with increasing expression of trefoil factor 3, in the jejunum of EHEC-infected mice. CWA increased the expression of tight junction proteins in the jejunum of EHEC-infected mice. Using intestinal epithelial cells and a Rac1 inhibitor in vitro, we demonstrated that the CWA-mediated increases in the tight junction proteins might depend on the Rac1 pathway. Furthermore, CWA improved the microbiota and short-chain fatty acid concentrations in the cecum of EHEC-infected mice. Although Enro and CWA had similar effects on intestinal inflammation, CWA was superior to Enro with regard to improving intestinal epithelial barrier and microbiota in the intestine. In conclusion, CWA attenuated EHEC-induced inflammation, intestinal epithelial barrier damage, and microbiota disruption in the intestine of mice, suggesting that CWA may be an effective therapy for many intestinal diseases.
Collapse
Affiliation(s)
- Hongbo Yi
- Institute of Feed Science, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310058, China; and.,Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong 510640, China
| | - Wangyang Hu
- Institute of Feed Science, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310058, China; and
| | - Shan Chen
- Institute of Feed Science, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310058, China; and
| | - Zeqing Lu
- Institute of Feed Science, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310058, China; and
| | - Yizhen Wang
- Institute of Feed Science, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310058, China; and
| |
Collapse
|
19
|
Ozyurt C, Bora B, Ugurlu O, Evran S. Pathogen-specific nucleic acid aptamers as targeting components of antibiotic and gene delivery systems. NANOSTRUCTURES FOR DRUG DELIVERY 2017:551-577. [DOI: 10.1016/b978-0-323-46143-6.00018-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
20
|
Abstract
Human cathelicidin LL-37, the only member of the cathelicidin family of host defense peptides expressed in humans, plays a crucial role in host defense against pathogen invasion, as well as in regulating the functions of anti-inflammation, antitumorigenesis, and tissue repair. It is primarily produced by phagocytic leukocytes and epithelial cells, and mediates a wide range of biological responses. Emerging evidence from several studies indicates that LL-37 plays a prominent and complex role in inflammatory bowel disease (IBD). Although overexpression of LL-37 has been implicated in the inflamed and noninflamed colon mucosa in patients with ulcerative colitis, LL-37 expression was not changed in the inflamed or noninflamed colon or ileal mucosa in patients with Crohn's disease. Furthermore, studies in animal models and human patients further characterized the protective effect of cathelicidins both in ulcerative colitis and Crohn's disease. These data suggest the intricate functions of LL-37 in IBD. They will also create many strategies and opportunities for therapeutic intervention in IBD in the future. This review aims to elucidate the structure and bioactivity of LL-37 and also discuss the recent progress in understanding the relationship between LL-37 and IBD.
Collapse
|
21
|
Yi H, Zhang L, Gan Z, Xiong H, Yu C, Du H, Wang Y. High therapeutic efficacy of Cathelicidin-WA against postweaning diarrhea via inhibiting inflammation and enhancing epithelial barrier in the intestine. Sci Rep 2016; 6:25679. [PMID: 27181680 PMCID: PMC4867772 DOI: 10.1038/srep25679] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 04/20/2016] [Indexed: 12/12/2022] Open
Abstract
Diarrhea is a leading cause of death among young mammals, especially during weaning. Here, we investigated the effects of Cathelicidin-WA (CWA) on diarrhea, intestinal morphology, inflammatory responses, epithelial barrier and microbiota in the intestine of young mammals during weaning. Piglets with clinical diarrhea were selected and treated with saline (control), CWA or enrofloxacin (Enro) for 4 days. Both CWA and Enro effectively attenuated diarrhea. Compared with the control, CWA decreased IL-6, IL-8 and IL-22 levels and reduced neutrophil infiltration into the jejunum. CWA inhibited inflammation by down-regulating the TLR4-, MyD88- and NF-κB-dependent pathways. Additionally, CWA improved intestinal morphology by increasing villus and microvillus heights and enhancing intestinal barrier function by increasing tight junction (TJ) protein expression and augmenting wound-healing ability in intestinal epithelial cells. CWA also improved microbiota composition and increased short-chain fatty acid (SCFA) levels in feces. By contrast, Enro not only disrupted the intestinal barrier but also negatively affected microbiota composition and SCFA levels in the intestine. In conclusion, CWA effectively attenuated inflammation, enhanced intestinal barrier function, and improved microbiota composition in the intestines of weaned piglets. These results suggest that CWA could be an effective and safe therapy for diarrhea or other intestinal diseases in young mammals.
Collapse
Affiliation(s)
- Hongbo Yi
- Institute of Feed Science, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Lin Zhang
- Institute of Feed Science, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Zhenshun Gan
- Institute of Feed Science, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Haitao Xiong
- Institute of Feed Science, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Caihua Yu
- Institute of Feed Science, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Huahua Du
- Institute of Feed Science, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Yizhen Wang
- Institute of Feed Science, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
22
|
Zhang L, Wu WKK, Gallo RL, Fang EF, Hu W, Ling TKW, Shen J, Chan RLY, Lu L, Luo XM, Li MX, Chan KM, Yu J, Wong VWS, Ng SC, Wong SH, Chan FKL, Sung JJY, Chan MTV, Cho CH. Critical Role of Antimicrobial Peptide Cathelicidin for Controlling Helicobacter pylori Survival and Infection. THE JOURNAL OF IMMUNOLOGY 2016; 196:1799-1809. [DOI: 10.4049/jimmunol.1500021] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
The antimicrobial peptide cathelicidin is critical for protection against different kinds of microbial infection. This study sought to elucidate the protective action of cathelicidin against Helicobacter pylori infection and its associated gastritis. Exogenous cathelicidin was found to inhibit H. pylori growth, destroy the bacteria biofilm, and induce morphological alterations in H. pylori membrane. Additionally, knockdown of endogenous cathelicidin in human gastric epithelial HFE-145 cells markedly increased the intracellular survival of H. pylori. Consistently, cathelicidin knockout mice exhibited stronger H. pylori colonization, higher expression of proinflammatory cytokines IL-6, IL-1β, and ICAM1, and lower expression of the anti-inflammatory cytokine IL-10 in the gastric mucosa upon H. pylori infection. In wild-type mice, H. pylori infection also stimulated gastric epithelium-derived cathelicidin production. Importantly, pretreatment with bioengineered Lactococcus lactis that actively secretes cathelicidin significantly increased mucosal cathelicidin levels and reduced H. pylori infection and the associated inflammation. Moreover, cathelicidin strengthened the barrier function of gastric mucosa by stimulating mucus synthesis. Collectively, these findings indicate that cathelicidin plays a significant role as a potential natural antibiotic for H. pylori clearance and a therapeutic agent for chronic gastritis.
Collapse
Affiliation(s)
- Lin Zhang
- *Institute of Digestive Diseases and State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- †Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- ‡CUHK Shenzhen Research Institute, Shenzhen 518057, China
| | - William K. K. Wu
- *Institute of Digestive Diseases and State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- ‡CUHK Shenzhen Research Institute, Shenzhen 518057, China
- §Department of Anesthesia and Intensive Care, Chinese University of Hong Kong, Hong Kong, China
| | - Richard L. Gallo
- ¶Division of Dermatology, University of California, San Diego, La Jolla, CA 92093
| | - Evandro F. Fang
- ‖Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224
| | - Wei Hu
- #School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China; and
| | - Thomas K. W. Ling
- **Department of Microbiology, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong, China
| | - Jing Shen
- #School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China; and
| | - Ruby L. Y. Chan
- #School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China; and
| | - Lan Lu
- #School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China; and
| | - Xiao M. Luo
- #School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China; and
| | - Ming X. Li
- #School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China; and
| | - Kam M. Chan
- #School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China; and
| | - Jun Yu
- *Institute of Digestive Diseases and State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- †Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- ‡CUHK Shenzhen Research Institute, Shenzhen 518057, China
| | - Vincent W. S. Wong
- *Institute of Digestive Diseases and State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- †Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
| | - Siew C. Ng
- *Institute of Digestive Diseases and State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- †Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
| | - Sunny H. Wong
- *Institute of Digestive Diseases and State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- †Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- ‡CUHK Shenzhen Research Institute, Shenzhen 518057, China
| | - Francis K. L. Chan
- *Institute of Digestive Diseases and State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- †Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- ‡CUHK Shenzhen Research Institute, Shenzhen 518057, China
| | - Joseph J. Y. Sung
- *Institute of Digestive Diseases and State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- †Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- ‡CUHK Shenzhen Research Institute, Shenzhen 518057, China
| | - Matthew T. V. Chan
- §Department of Anesthesia and Intensive Care, Chinese University of Hong Kong, Hong Kong, China
| | - Chi H. Cho
- #School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China; and
| |
Collapse
|
23
|
Colonic MUC2 mucin regulates the expression and antimicrobial activity of β-defensin 2. Mucosal Immunol 2015; 8:1360-72. [PMID: 25921338 PMCID: PMC4762903 DOI: 10.1038/mi.2015.27] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 03/18/2015] [Indexed: 02/04/2023]
Abstract
In this study we identified mechanisms at the colonic mucosa by which MUC2 mucin regulated the production of β-defensin in a proinflammatory milieu but functionally protected susceptible bacteria from its antimicrobial effects. The regulator role of MUC2 on production of β-defensin 2 in combination with the proinflammatory cytokine interleukin-1β (IL-1β) was confirmed using purified human colonic MUC2 mucin and colonic goblet cells short hairpin RNA (shRNA) silenced for MUC2. In vivo, Muc2(-/-) mice showed impaired β-defensin mRNA expression and peptide localization in the colon as compared with Muc2(+/-) and Muc2(+/+) littermates. Importantly, purified MUC2 mucin abrogated the antimicrobial activity of β-defensin 2 against nonpathogenic and enteropathogenic Escherichia coli. Sodium metaperiodate oxidation of MUC2 removed the capacity of MUC2 to stimulate β-defensin production and MUC2's inhibition of defensin antimicrobial activity. This study highlights that a defective MUC2 mucin barrier, typical in inflammatory bowel diseases, may lead to deficient stimulation of β-defensin 2 and an unbalanced microbiota that favor the growth of β-defensin-resistant microbes such as Clostridium difficile.
Collapse
|
24
|
Zhang H, Xia X, Han F, Jiang Q, Rong Y, Song D, Wang Y. Cathelicidin-BF, a Novel Antimicrobial Peptide from Bungarus fasciatus, Attenuates Disease in a Dextran Sulfate Sodium Model of Colitis. Mol Pharm 2015; 12:1648-61. [PMID: 25807257 DOI: 10.1021/acs.molpharmaceut.5b00069] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Antimicrobial peptides are molecules of innate immunity. Cathelicidin-BF is the first cathelicidin peptide found in reptiles. However, the immunoregulatory and epithelial barrier protective properties of C-BF have not been reported. Inflammatory bowel diseases, including ulcerative colitis and Crohn's disease, can lead to colon cancer, the third most common malignant tumor. The objective is to develop the new found cathelicidin-BF as a therapeutic to patients of ulcerative colitis. The morphology of the colon epithelium was observed by H&E staining; apoptosis index and infiltration of inflammatory cells in colonic epithelium were measured by TUNEL and immunohistochemistry; the expression level of endogenous mCRAMP was analyzed by immunofluorescence; and phosphorylation of the transcription factors c-jun and NF-κB in colon were analyzed by Western blot. Our results showed that the morphology of the colon epithelium in the C-BF+DSS group was improved compared with the DSS group. Apoptosis and infiltration of inflammatory cells in colonic epithelium were also significantly attenuated in the C-BF+DSS group compared with the DSS group, and the expression level of endogenous mCRAMP in the DSS group was significantly higher than other groups. DSS-induced phosphorylation level of c-jun and NF-κB while C-BF effectively inhibited phosphorylation of NF-κB (p65). The barrier protective effect of C-BF was still excellent. In conclusion, C-BF effectively attenuated inflammation and improved disrupted barrier function. Notably, this is the first report to demonstrate that C-BF attenuates DSS-induced UC both through the regulation of intestinal immune and retention of barrier function, and the exact pathway was through NF-κB.
Collapse
Affiliation(s)
- Haiwen Zhang
- Key Laboratory of Animal Nutrition and Feed Science (Hua Dong), Ministry of Agriculture College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xi Xia
- Key Laboratory of Animal Nutrition and Feed Science (Hua Dong), Ministry of Agriculture College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Feifei Han
- Key Laboratory of Animal Nutrition and Feed Science (Hua Dong), Ministry of Agriculture College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qin Jiang
- Key Laboratory of Animal Nutrition and Feed Science (Hua Dong), Ministry of Agriculture College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yili Rong
- Key Laboratory of Animal Nutrition and Feed Science (Hua Dong), Ministry of Agriculture College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Deguang Song
- Key Laboratory of Animal Nutrition and Feed Science (Hua Dong), Ministry of Agriculture College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yizhen Wang
- Key Laboratory of Animal Nutrition and Feed Science (Hua Dong), Ministry of Agriculture College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
25
|
Kopp ZA, Jain U, Van Limbergen J, Stadnyk AW. Do antimicrobial peptides and complement collaborate in the intestinal mucosa? Front Immunol 2015; 6:17. [PMID: 25688244 PMCID: PMC4311685 DOI: 10.3389/fimmu.2015.00017] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/08/2015] [Indexed: 12/13/2022] Open
Abstract
It is well understood that multiple antimicrobial peptides (AMPs) are constitutively deployed by the epithelium to bolster the innate defenses along the entire length of the intestines. In addition to this constitutive/homeostatic production, AMPs may be inducible and levels changed during disease. In contrast to this level of knowledge on AMP sources and roles in the intestines, our understanding of the complement cascade in the healthy and diseased intestines is rudimentary. Epithelial cells make many complement proteins and there is compelling evidence that complement becomes activated in the lumen. With the common goal of defending the host against microbes, the opportunities for cross-talk between these two processes is great, both in terms of actions on the target microbes but also on regulating the synthesis and secretion of the alternate family of molecules. This possibility is beginning to become apparent with the finding that colonic epithelial cells possess anaphylatoxin receptors. There still remains much to be learned about the possible points of collaboration between AMPs and complement, for example, whether there is reciprocal control over expression in the intestinal mucosa in homeostasis and restoring the balance following infection and inflammation.
Collapse
Affiliation(s)
- Zoë A Kopp
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University , Halifax, NS , Canada
| | - Umang Jain
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University , Halifax, NS , Canada
| | - Johan Van Limbergen
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University , Halifax, NS , Canada ; Department of Pediatrics, Faculty of Medicine, Dalhousie University , Halifax, NS , Canada
| | - Andrew W Stadnyk
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University , Halifax, NS , Canada ; Department of Pediatrics, Faculty of Medicine, Dalhousie University , Halifax, NS , Canada
| |
Collapse
|
26
|
Cheng M, Ho S, Yoo JH, Tran DHY, Bakirtzi K, Su B, Tran DHN, Kubota Y, Ichikawa R, Koon HW. Cathelicidin suppresses colon cancer development by inhibition of cancer associated fibroblasts. Clin Exp Gastroenterol 2014; 8:13-29. [PMID: 25565877 PMCID: PMC4274046 DOI: 10.2147/ceg.s70906] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Cathelicidin (LL-37 in humans and mCRAMP in mice) represents a family of endogenous antimicrobial and anti-inflammatory peptides. Cancer-associated fibroblasts can promote the proliferation of colon cancer cells and growth of colon cancer tumors. METHODS We examined the role of cathelicidin in the development of colon cancer, using subcutaneous human HT-29 colon-cancer-cell-derived tumor model in nude mice and azoxymethane- and dextran sulfate-mediated colon cancer model in C57BL/6 mice. We also determined the indirect antitumoral mechanism of cathelicidin via the inhibition of epithelial-mesenchymal transition (EMT) of colon cancer cells and fibroblast-supported colon cancer cell proliferation. RESULTS Intravenous administration of cathelicidin expressing adeno-associated virus significantly reduced the size of tumors, tumor-derived collagen expression, and tumor-derived fibroblast expression in HT-29-derived subcutaneous tumors in nude mice. Enema administration of the mouse cathelicidin peptide significantly reduced the size and number of colonic tumors in azoxymethane- and dextran sulfate-treated mice without inducing apoptosis in tumors and the adjacent normal colonic tissues. Cathelicidin inhibited the collagen expression and vimentin-positive fibroblast expression in colonic tumors. Cathelicidin did not directly affect HT-29 cell viability, but did significantly reduce tumor growth factor-β1-induced EMT of colon cancer cells. Media conditioned by the human colonic CCD-18Co fibroblasts promoted human colon cancer HT-29 cell proliferation. Cathelicidin pretreatment inhibited colon cancer cell proliferation mediated by media conditioned by human colonic CCD-18Co fibroblasts. Cathelicidin disrupted tubulin distribution in colonic fibroblasts. Disruption of tubulin in fibroblasts reduced fibroblast-supported colon cancer cell proliferation. CONCLUSION Cathelicidin effectively inhibits colon cancer development by interfering with EMT and fibroblast-supported colon cancer cell proliferation.
Collapse
Affiliation(s)
- Michelle Cheng
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Samantha Ho
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jun Hwan Yoo
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA ; Digestive Disease Center, CHA University Bundang Medical Center, Seongnam, Republic of Korea
| | - Deanna Hoang-Yen Tran
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Kyriaki Bakirtzi
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Bowei Su
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Diana Hoang-Ngoc Tran
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Yuzu Kubota
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Ryan Ichikawa
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Hon Wai Koon
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
27
|
Ostaff MJ, Stange EF, Wehkamp J. Antimicrobial peptides and gut microbiota in homeostasis and pathology. EMBO Mol Med 2013; 5:1465-83. [PMID: 24039130 PMCID: PMC3799574 DOI: 10.1002/emmm.201201773] [Citation(s) in RCA: 245] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 06/19/2013] [Accepted: 07/04/2013] [Indexed: 12/17/2022] Open
Abstract
We survive because we adapted to a world of microorganisms. All our epithelial surfaces participate in keeping up an effective barrier against microbes while not initiating ongoing inflammatory processes and risking collateral damage to the host. Major players in this scenario are antimicrobial peptides (AMPs). Such broad-spectrum innate antibiotics are in part produced by specialized cells but also widely sourced from all epithelia as well as circulating inflammatory cells. AMPs belong to an ancient defense system found in all organisms and participated in a preservative co-evolution with a complex microbiome. Particularly interesting interactions between host barrier and microbiota can be found in the gut. The intestinal cell lining not only has to maintain a tightly regulated homeostasis during its high-throughput regeneration, but also a balanced relationship towards an extreme number of mutualistic or commensal inhabitants. Recent research suggests that advancing our understanding of the circumstances of such balanced and sometimes imbalanced interactions between gut microbiota and host AMPs should have therapeutic implications for different intestinal disorders.
Collapse
Affiliation(s)
- Maureen J Ostaff
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany and University of Tuebingen, Germany
| | | | | |
Collapse
|
28
|
Chow JYC, Li ZJ, Kei WK, Cho CH. Cathelicidin a potential therapeutic peptide for gastrointestinal inflammation and cancer. World J Gastroenterol 2013; 19:2731-2735. [PMID: 23687409 PMCID: PMC3653146 DOI: 10.3748/wjg.v19.i18.2731] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 04/04/2013] [Indexed: 02/06/2023] Open
Abstract
Cathelicidins, are host defense peptides synthesized and stored in circulating leukocytes and numerous types of epithelial tissues in particular the gastrointestinal (GI) tract and skin. They have been known for their antimicrobial activities against a variety of microbes. Recently it was discovered that they have other significant biological functions and produce appealing pharmacological actions against inflammation and cancer in the GI tract through defined mechanisms. Experimental evidence shows that these actions could be tissue and disease specific and concentration dependent. This article reviews some of the physiological functions of cathelicidins and also their therapeutic potential in the treatment of inflammation and cancer and also the delivery system for this peptide as targeted therapy for various disorders in the GI tract both in animals and humans.
Collapse
|
29
|
Ahluwalia A, Tarnawski AS. Cathelicidin gene therapy: a new therapeutic option in ulcerative colitis and beyond? Gene Ther 2013; 20:119-120. [PMID: 22456327 DOI: 10.1038/gt.2012.23] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
30
|
Zhang L, Yu J, Wong CCM, Ling TKW, Li ZJ, Chan KM, Ren SX, Shen J, Chan RLY, Lee CC, Li MSM, Cheng ASL, To KF, Gallo RL, Sung JJY, Wu WKK, Cho CH. Cathelicidin protects against Helicobacter pylori colonization and the associated gastritis in mice. Gene Ther 2012; 20:751-60. [PMID: 23254369 DOI: 10.1038/gt.2012.92] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 08/31/2012] [Accepted: 10/08/2012] [Indexed: 12/29/2022]
Abstract
Cathelicidin, an antimicrobial peptide of the innate immune system, has been shown to modulate microbial growth, wound healing and inflammation. However, whether cathelicidin controls Helicobacter pylori infection in vivo remains unexplored. This study sought to elucidate the role of endogenous and exogenous mouse cathelicidin (CRAMP) in the protection against H. pylori infection and the associated gastritis in mice. Results showed that genetic ablation of CRAMP in mice significantly increased the susceptibility of H. pylori colonization and the associated gastritis as compared with the wild-type control. Furthermore, replenishment with exogenous CRAMP, delivered via a bioengineered CRAMP-secreting strain of Lactococcus lactis, reduced H. pylori density in the stomach as well as the associated inflammatory cell infiltration and cytokine production. Collectively, these findings indicate that cathelicidin protects against H. pylori infection and its associated gastritis in vivo. Our study also demonstrates the feasibility of using the transformed food-grade bacteria to deliver cathelicidin, which may have potential clinical applications in the treatment of H. pylori infection in humans.
Collapse
Affiliation(s)
- L Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Ren SX, Cheng ASL, To KF, Tong JHM, Li MS, Shen J, Shen J, Wong CCM, Zhang L, Chan RLY, Wang XJ, Ng SSM, Chiu LCM, Marquez VE, Gallo RL, Chan FKL, Yu J, Sung JJY, Wu WKK, Cho CH. Host immune defense peptide LL-37 activates caspase-independent apoptosis and suppresses colon cancer. Cancer Res 2012; 72:6512-23. [PMID: 23100468 DOI: 10.1158/0008-5472.can-12-2359] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cathelicidins are a family of bacteriocidal polypeptides secreted by macrophages and polymorphonuclear leukocytes (PMN). LL-37, the only human cathelicidin, has been implicated in tumorigenesis, but there has been limited investigation of its expression and function in cancer. Here, we report that LL-37 activates a p53-mediated, caspase-independent apoptotic cascade that contributes to suppression of colon cancer. LL-37 was expressed strongly in normal colon mucosa but downregulated in colon cancer tissues, where in both settings its expression correlated with terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling-positive apoptotic cells. Exposure of colon cancer cells to LL-37 induced phosphatidylserine externalization and DNA fragmentation in a manner independent of caspase activation. Apoptogenic function was mediated by nuclear translocation of the proapoptotic factors, apoptosis-inducing factor (AIF) and endonuclease G (EndoG), through p53-dependent upregulation of Bax and Bak and downregulation of Bcl-2 via a pertussis toxin-sensitive G-protein-coupled receptor (GPCR) pathway. Correspondingly, colonic mucosa of cathelicidin-deficient mice exhibited reduced expression of p53, Bax, and Bak and increased expression of Bcl-2 together with a lower basal level of apoptosis. Cathelicidin-deficient mice exhibited an increased susceptibility to azoxymethane-induced colon tumorigenesis, establishing pathophysiologic relevance in colon cancer. Collectively, our findings show that LL-37 activates a GPCR-p53-Bax/Bak/Bcl-2 signaling cascade that triggers AIF/EndoG-mediated apoptosis in colon cancer cells.
Collapse
Affiliation(s)
- Shun X Ren
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|