1
|
Li Y, Xu Y, Su W, Xu J, Ye Z, Wang Z, Liu Q, Chen F. Exploring the immuno-nano nexus: A paradigm shift in tumor vaccines. Biomed Pharmacother 2025; 184:117897. [PMID: 39921945 DOI: 10.1016/j.biopha.2025.117897] [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: 10/31/2024] [Revised: 01/17/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025] Open
Abstract
Tumor vaccines have become a crucial strategy in cancer immunotherapy. Challenges of traditional tumor vaccines include inadequate immune activation and low efficacy of antigen delivery. Nanoparticles, with their tunable properties and versatile functionalities, have redefined the landscape of tumor vaccine design. In this review, we outline the multifaceted roles of nanoparticles in tumor vaccines, ranging from their capacity as delivery vehicles to their function as immunomodulatory adjuvants capable of stimulating anti-tumor immunity. We discuss how this innovative approach significantly boosts antigen presentation by leveraging tailored nanoparticles that facilitate efficient uptake by antigen-presenting cells. These nanoparticles have been meticulously designed to overcome biological barriers, ensuring optimal delivery to lymph nodes and effective interaction with the immune system. Overall, this review highlights the transformative power of nanotechnology in redefining the principles of tumor vaccines. The intent is to inform more efficacious and precise cancer immunotherapies. The integration of these advanced nanotechnological strategies should unlock new frontiers in tumor vaccine development, enhancing their potential to elicit robust and durable anti-tumor immunity.
Collapse
Affiliation(s)
- Yuanyuan Li
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Yike Xu
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Wenwen Su
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Jia Xu
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Zifei Ye
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Zhuoyi Wang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Qihui Liu
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| | - Fangfang Chen
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
2
|
Pluetrattanabha N, Direksunthorn T, Ahmad I, Jyothi SR, Shit D, Singh AK, Chauhan AS. Inflammasome activation in melanoma progression: the latest update concerning pathological role and therapeutic value. Arch Dermatol Res 2025; 317:258. [PMID: 39820618 DOI: 10.1007/s00403-025-03802-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 12/11/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025]
Abstract
The progression of melanoma is a complex process influenced by both internal and external cues which encourage the transition of tumour cells, uncontrolled growth, migration, and metastasis. Additionally, inflammation allows tumours to evade the immune system, contributing to cancer development. The inflammasome, a complex of many proteins, is crucial in enhancing immune responses to external and internal triggers. As a critical inflammatory mechanism, it contributes to the development of melanoma. These mechanisms may be triggered via various internal and external stimuli, causing the induction of specific enzymes such as caspase-1, caspase-11, or caspase-8. This, in turn, leads to the release of interleukin (IL)-1β and IL-18 and cell death by apoptosis and pyroptosis. Proper inflammasome stimulation is crucial for the host to deal with invading pathogens or tissue injury. However, inappropriate inflammasome stimulation can result in unregulated tissue reactions, thus easing many diseases, including melanoma. Hence, keeping a delicate equilibrium between the stimulation and prohibition of inflammasomes is crucial, necessitating meticulous control of the assembly and functional aspects of inflammasomes. This review examines the latest advancements in inflammasome studies, specifically focusing on the molecular processes that control inflammasome formation, signalling, and modulation in melanoma.
Collapse
Affiliation(s)
| | | | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
- Health and Medical Research Center, King Khalid University, P.O. Box 960, AlQura'a, Abha, Saudi Arabia
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics, JAIN (Deemed to be University) School of Sciences, Bangalore, Karnataka, India
| | - Debasish Shit
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| | | | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of research and innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| |
Collapse
|
3
|
Gao J, Zhang C, Xu H, Zhang T, Wang H, Wang Y, Wang X. Dissecting the Role of the Hydroxyl Moiety at C14 in (+)-Opioid-Based TLR4 Antagonists via Wet-Lab Experiments and Molecular Dynamics Simulations. J Chem Inf Model 2024; 64:5273-5284. [PMID: 38921627 DOI: 10.1021/acs.jcim.4c00692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
Toll-like receptor 4 (TLR4) is pivotal as an innate immune receptor, playing a critical role in mediating neuropathic pain and drug addiction through its regulation of the neuroinflammatory response. The nonclassical (+)-opioid isomers represent a unique subset of TLR4 antagonists known for their effective blood-brain barrier permeability. Despite growing interest in the structure-activity relationship of these (+)-opioid-based TLR4 antagonists, the specific impact of heteroatoms on their TLR4 antagonistic activities has not been fully explored. This study investigated the influence of the hydroxyl group at C14 in six (+)-opioid TLR4 antagonists (1-6) using wet-lab experiments and in silico simulations. The corresponding C14-deoxy derivatives (7-12) were synthesized, and upon comparison with their corresponding counterparts (1-6), it was discovered that their TLR4 antagonistic activities were significantly diminished. Molecular dynamics simulations showed that the (+)-opioid TLR4 antagonists (1-6) possessed more negative binding free energies to the TLR4 coreceptor MD2, which was responsible for ligand recognition. This was primarily attributed to the formation of a hydrogen bond between the hydroxyl group at the C-14 position of the antagonists (1-6) and the R90 residue of MD2 during the binding process. Such an interaction facilitated the entry and subsequent binding of these molecules within the MD2 cavity. In contrast, the C14-deoxy derivatives (7-12), lacking the hydroxyl group at the C-14 position, missed this crucial hydrogen bond interaction with the R90 residue of MD2, leading to their egression from the MD2 cavity during simulations. This study underscores the significant role of the C14 hydroxyl moiety in enhancing the effectiveness of (+)-opioid TLR4 antagonists, which provides insightful guidance for designing future (+)-isomer opioid-derived TLR4 antagonists.
Collapse
Affiliation(s)
- Jingwei Gao
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Cong Zhang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Hangyu Xu
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, China
| | - Tianshu Zhang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Hongshuang Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, China
| | - Yibo Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, China
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
- Beijing National Laboratory for Molecular Sciences, Beijing 100190, China
| |
Collapse
|
4
|
Gao J, Lin C, Zhang C, Zhang X, Wang Y, Xu H, Zhang T, Li H, Wang H, Wang X. Exploring the Function of (+)-Naltrexone Precursors: Their Activity as TLR4 Antagonists and Potential in Treating Morphine Addiction. J Med Chem 2024; 67:3127-3143. [PMID: 38306598 DOI: 10.1021/acs.jmedchem.3c02316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2024]
Abstract
Disruptions in the toll-like receptor 4 (TLR4) signaling pathway are linked to chronic inflammation, neuropathic pain, and drug addiction. (+)-Naltrexone, an opioid-derived TLR4 antagonist with a (+)-isomer configuration, does not interact with classical opioid receptors and has moderate blood-brain barrier permeability. Herein, we developed a concise 10-step synthesis for (+)-naltrexone and explored its precursors, (+)-14-hydroxycodeinone (1) and (+)-14-hydroxymorphinone (3). These precursors exhibited TLR4 antagonistic activities 100 times stronger than (+)-naltrexone, particularly inhibiting the TLR4-TRIF pathway. In vivo studies showed that these precursors effectively reduced behavioral effects of morphine, like sensitization and conditioned place preference by suppressing microglial activation and TNF-α expression in the medial prefrontal cortex and ventral tegmental area. Additionally, 3 displayed a longer half-life and higher oral bioavailability than 1. Overall, this research optimized (+)-naltrexone synthesis and identified its precursors as potent TLR4 antagonists, offering potential treatments for morphine addiction.
Collapse
Affiliation(s)
- Jingwei Gao
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Cong Lin
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Cong Zhang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xiaozheng Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Yibo Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Hangyu Xu
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Tianshu Zhang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Haohong Li
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou, Zhejiang 311121, China
| | - Hongshuang Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
- Beijing National Laboratory for Molecular Sciences, Beijing 100190, China
| |
Collapse
|
5
|
Sun Z, Zhao H, Ma L, Shi Y, Ji M, Sun X, Ma D, Zhou W, Huang T, Zhang D. The quest for nanoparticle-powered vaccines in cancer immunotherapy. J Nanobiotechnology 2024; 22:61. [PMID: 38355548 PMCID: PMC10865557 DOI: 10.1186/s12951-024-02311-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/26/2024] [Indexed: 02/16/2024] Open
Abstract
Despite recent advancements in cancer treatment, this disease still poses a serious threat to public health. Vaccines play an important role in preventing illness by preparing the body's adaptive and innate immune responses to combat diseases. As our understanding of malignancies and their connection to the immune system improves, there has been a growing interest in priming the immune system to fight malignancies more effectively and comprehensively. One promising approach involves utilizing nanoparticle systems for antigen delivery, which has been shown to potentiate immune responses as vaccines and/or adjuvants. In this review, we comprehensively summarized the immunological mechanisms of cancer vaccines while focusing specifically on the recent applications of various types of nanoparticles in the field of cancer immunotherapy. By exploring these recent breakthroughs, we hope to identify significant challenges and obstacles in making nanoparticle-based vaccines and adjuvants feasible for clinical application. This review serves to assess recent breakthroughs in nanoparticle-based cancer vaccinations and shed light on their prospects and potential barriers. By doing so, we aim to inspire future immunotherapies for cancer that harness the potential of nanotechnology to deliver more effective and targeted treatments.
Collapse
Affiliation(s)
- Zhe Sun
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Hui Zhao
- Department of Endodontics, East Branch of Jinan Stomatological Hospital, Jinan, 250000, Shandong, China
| | - Li Ma
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yanli Shi
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Mei Ji
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Xiaodong Sun
- Department of Endodontics, Gaoxin Branch of Jinan Stomatological Hospital, Jinan, 250000, Shandong, China
| | - Dan Ma
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Wei Zhou
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Tao Huang
- Department of Biomedical Engineering, Graeme Clark Institute, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Dongsheng Zhang
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
6
|
Dai Y, Zhou J, Shi C. Inflammasome: structure, biological functions, and therapeutic targets. MedComm (Beijing) 2023; 4:e391. [PMID: 37817895 PMCID: PMC10560975 DOI: 10.1002/mco2.391] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 10/12/2023] Open
Abstract
Inflammasomes are a group of protein complex located in cytoplasm and assemble in response to a wide variety of pathogen-associated molecule patterns, damage-associated molecule patterns, and cellular stress. Generally, the activation of inflammasomes will lead to maturation of proinflammatory cytokines and pyroptotic cell death, both associated with inflammatory cascade amplification. A sensor protein, an adaptor, and a procaspase protein interact through their functional domains and compose one subunit of inflammasome complex. Under physiological conditions, inflammasome functions against pathogen infection and endogenous dangers including mtROS, mtDNA, and so on, while dysregulation of its activation can lead to unwanted results. In recent years, advances have been made to clarify the mechanisms of inflammasome activation, the structural details of them and their functions (negative/positive) in multiple disease models in both animal models and human. The wide range of the stimuli makes the function of inflammasome diverse and complex. Here, we review the structure, biological functions, and therapeutic targets of inflammasomes, while highlight NLRP3, NLRC4, and AIM2 inflammasomes, which are the most well studied. In conclusion, this review focuses on the activation process, biological functions, and structure of the most well-studied inflammasomes, summarizing and predicting approaches for disease treatment and prevention with inflammasome as a target. We aim to provide fresh insight into new solutions to the challenges in this field.
Collapse
Affiliation(s)
- Yali Dai
- Institute of Rocket Force MedicineState Key Laboratory of Trauma and Chemical PoisoningArmy Medical UniversityChongqingChina
| | - Jing Zhou
- Institute of Rocket Force MedicineState Key Laboratory of Trauma and Chemical PoisoningArmy Medical UniversityChongqingChina
- Institute of ImmunologyArmy Medical UniversityChongqingChina
| | - Chunmeng Shi
- Institute of Rocket Force MedicineState Key Laboratory of Trauma and Chemical PoisoningArmy Medical UniversityChongqingChina
| |
Collapse
|
7
|
Kumar V, Bauer C, Stewart JH. TIME Is Ticking for Cervical Cancer. BIOLOGY 2023; 12:941. [PMID: 37508372 PMCID: PMC10376148 DOI: 10.3390/biology12070941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023]
Abstract
Cervical cancer (CC) is a major health problem among reproductive-age females and comprises a leading cause of cancer-related deaths. Human papillomavirus (HPV) is the major risk factor associated with CC incidence. However, lifestyle is also a critical factor in CC pathogenesis. Despite HPV vaccination introduction, the incidence of CC is increasing worldwide. Therefore, it becomes critical to understand the CC tumor immune microenvironment (TIME) to develop immune cell-based vaccination and immunotherapeutic approaches. The current article discusses the immune environment in the normal cervix of adult females and its role in HPV infection. The subsequent sections discuss the alteration of different immune cells comprising CC TIME and their targeting as future therapeutic approaches.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), 1700 Tulane Avenue, New Orleans, LA 70012, USA
| | - Caitlin Bauer
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), 1700 Tulane Avenue, New Orleans, LA 70012, USA
| | - John H Stewart
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), 1700 Tulane Avenue, New Orleans, LA 70012, USA
- Louisiana Children's Medical Center Cancer Center, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), 1700 Tulane Avenue, New Orleans, LA 70012, USA
| |
Collapse
|
8
|
Matsuda E, Takada K, Kobayashi O, Nakajima T, Ikeda Y, Asai-Sato M, Kawakami K, Komatsu A, Chishima F, Komine-Aizawa S, Hayakawa S, Kawana K. Pathological roles of antimicrobial peptides and pro-inflammatory factors secreted from the cervical epithelium in Gardnerella vaginalis-abundant vaginal flora in pregnancy. J Reprod Immunol 2023; 156:103797. [PMID: 36645984 DOI: 10.1016/j.jri.2023.103797] [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: 06/27/2022] [Revised: 12/13/2022] [Accepted: 01/05/2023] [Indexed: 01/08/2023]
Abstract
Bacterial vaginosis due to Gardnerella vaginalis (GV) is one of the main causes of preterm birth. Antimicrobial function of the cervical glands prevents ascending pathogen infection. This study investigated the effect of GV on the cervical gland cells. We examined the correlation between GV and neutrophil elastase in the cervical mucous obtained from pregnant women's clinical samples. Culture supernatants (sup) of GV and Lactobacillus crispatus (LC) were added to human immortalized cervical gland cells (EndoCx). Quantitative reverse transcription PCR (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA) were used to examine the effects on the production of antimicrobial peptides (AMPs), secretory leukocyte peptidase inhibitor (SLPI), and Elafin. mRNA microarray analysis revealed the expression profile of GV-exposed EndoCx. Moreover, the antimicrobial activity of Elafin against LC and GV was investigated. In the clinical samples, neutrophil elastase was increased in the GV-positive cervical mucous. In an in vitro assay, RT-qPCR and ELISA showed that GV-sup enhanced the secretion of Elafin, but not SLPI, from EndoCx, whereas LC-sup did not. mRNA microarray assay and ELISA results demonstrated that GV-sup enhanced the proinflammatory pathway and interleukin (IL)- 8 secretion from EndoCx as well as cell adhesion and tight junction pathways. Moreover, GV-sup directly enhanced Elafin and IL-8 secretion from the cervical gland cells. In the GV-abundant vaginal flora, IL-8 level increased the neutrophil elastase activity and Elafin inhibited the elastase activity to protect from tissue damage and infection. Thus, the balance of IL-8-induced neutrophil and Elafin-induced antiprotease activities may be crucial in preterm labor.
Collapse
Affiliation(s)
- Erina Matsuda
- Department of Obstetrics and Gynecology, Nihon University School of Medicine, Japan
| | - Kazuhide Takada
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Japan
| | - Osamu Kobayashi
- Department of Obstetrics and Gynecology, Nihon University School of Medicine, Japan
| | - Takahiro Nakajima
- Department of Obstetrics and Gynecology, Nihon University School of Medicine, Japan
| | - Yuji Ikeda
- Department of Obstetrics and Gynecology, Nihon University School of Medicine, Japan
| | - Mikiko Asai-Sato
- Department of Obstetrics and Gynecology, Nihon University School of Medicine, Japan
| | - Kaori Kawakami
- Department of Obstetrics and Gynecology, Nihon University School of Medicine, Japan
| | - Atsushi Komatsu
- Department of Obstetrics and Gynecology, Nihon University School of Medicine, Japan
| | - Fumihisa Chishima
- Department of Obstetrics and Gynecology, Nihon University School of Medicine, Japan
| | - Shihoko Komine-Aizawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Japan
| | - Satoshi Hayakawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Japan
| | - Kei Kawana
- Department of Obstetrics and Gynecology, Nihon University School of Medicine, Japan.
| |
Collapse
|
9
|
Pezeshki PS, Nowroozi A, Razi S, Rezaei N. Asthma and Allergy. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00002-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
10
|
Huang W, Zhang Y, Li Z, Li M, Li F, Mortimer M, Guo LH. Silver and Hyaluronic Acid-Coated Gold Nanoparticles Modulate the Metabolism of a Model Human Gut Bacterium Lactobacillus casei. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3377. [PMID: 36234503 PMCID: PMC9565723 DOI: 10.3390/nano12193377] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/11/2022] [Accepted: 09/23/2022] [Indexed: 05/19/2023]
Abstract
Medical applications of nanotechnology are promising in creating efficient and targeted therapies. However, so far, nanodrug design has not taken into consideration possible effects on human microbiota. The beneficial functions of bacteria could be stimulated by nanodrugs while negative effects on beneficial bacteria could cause risks to human health. Here, simulated intestinal fluid (IF) was optimized for culturing a human commensal and probiotic bacterial strain, Lactobacillus casei, to study the effects of medically relevant NPs—Ag and hyaluronic acid-coated Au NPs (HA-Au NPs)—in conditions pertinent to the gastrointestinal tract. When cultivated either aerobically or anaerobically, the specific growth rates of L. casei were ~0.2 h−1 in IF and ~0.4 h−1 in the standard medium of lactobacilli (MRS). Ag NPs inhibited the growth of L. casei in IF at lower concentrations (EC50 ~ 65 and 15 mg/L in aerobic and anaerobic conditions, respectively) than in MRS (EC50 > 100 mg/L), likely caused by differences in the composition of the two media and different intrinsic growth rates of bacteria in IF and MRS. Ag NP dissolution in IF and MRS did not explain the differences in growth inhibition, implying NP-specific effects. HA-Au NPs were not growth-inhibitory to L. casei up to 250 mg/L. Still, both NPs at sub-growth-inhibitory concentrations suppressed the expression of bacteriocin genes in L. casei, suggesting an inhibitory effect of NPs on the probiotic properties of L. casei, i.e., its competitiveness in microbial communities. However, HA-Au NPs did not appear to affect or even stimulated the immunomodulatory properties of L. casei in human intestinal epithelial cells. Thus, medically relevant NPs at low, sub-bacteriostatic levels can affect the metabolism of beneficial human bacteria and potentially induce changes in the microbiota and immune signaling.
Collapse
Affiliation(s)
- Wenqian Huang
- College of Life Science, China Jiliang University, Hangzhou 310018, China
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou 310018, China
| | - Yirong Zhang
- College of Life Science, China Jiliang University, Hangzhou 310018, China
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou 310018, China
| | - Zhi Li
- College of Life Science, China Jiliang University, Hangzhou 310018, China
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou 310018, China
| | - Minjie Li
- College of Quality and Safety Engineering, China Jiliang University, Hangzhou 310018, China
| | - Fangfang Li
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou 310018, China
- College of Quality and Safety Engineering, China Jiliang University, Hangzhou 310018, China
| | - Monika Mortimer
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou 310018, China
- College of Quality and Safety Engineering, China Jiliang University, Hangzhou 310018, China
| | - Liang-Hong Guo
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou 310018, China
- College of Quality and Safety Engineering, China Jiliang University, Hangzhou 310018, China
| |
Collapse
|
11
|
Galler AI, Suchodolski JS, Steiner JM, Sung CH, Hittmair KM, Richter B, Burgener IA. Microbial dysbiosis and fecal metabolomic perturbations in Yorkshire Terriers with chronic enteropathy. Sci Rep 2022; 12:12977. [PMID: 35902689 PMCID: PMC9334271 DOI: 10.1038/s41598-022-17244-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 07/22/2022] [Indexed: 01/08/2023] Open
Abstract
Dysbiosis and perturbations of fecal metabolic profiles have been reported in dogs with inflammatory bowel disease. Currently the incidence of dysbiosis and the fecal metabolomic profile in Yorkshire Terriers with chronic enteropathy (YTE) and the effects of treatment are unknown. This prospective observational study analyzed the dysbiosis index (DI) and fecal bile acid, sterol and fatty acid profiles in 14 Yorkshire Terriers with active YTE, 11 dogs in clinical remission, and 26 healthy Yorkshire Terriers. YTE was associated with dysbiosis and a significant increase in fatty acids (docosanoate, p = 0.002; gondoate, p = 0.026; erucate, p < 0.001; nervonate, p < 0.001; linolenate, p < 0.001), and plant sterols (campesterol, p < 0.001; brassicasterol, p = 0.024). The abundances of Fusobacterium (p < 0.001) and Cl. hiranonis (p = 0.018) and the concentrations of the secondary bile acid ursodeoxycholic acid (p = 0.033) and the plant sterol sitostanol (p = 0.003) were significantly decreased compared to healthy dogs. Dysbiosis, abundances of Fusobacterium, Cl. hiranonis and fecal concentrations of bile acids and sterols did not recover after treatment, while fecal fatty acid concentrations decreased in treated dogs. YTE is associated with dysbiosis and changes in bile acid, fatty acid, and sterol metabolism. These changes only recovered partially despite clinical remission. They might be breed-specific and involved in the pathogenesis of YTE.
Collapse
Affiliation(s)
- Alexandra I Galler
- Small Animal Internal Medicine, University of Veterinary Medicine, Vienna, Austria.
| | - Jan S Suchodolski
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | - Joerg M Steiner
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | - Chi-Hsuan Sung
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | - Katharina M Hittmair
- Clinical Unit of Diagnostic Imaging, University of Veterinary Medicine, Vienna, Austria
| | - Barbara Richter
- Institute of Pathology, University of Veterinary Medicine, Vienna, Austria
| | - Iwan A Burgener
- Small Animal Internal Medicine, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
12
|
Shu Y, Jiang H, Yuen CNT, Wang W, He J, Zhang H, Liu G, Wei L, Chen L, Wu H. Microcystin-leucine arginine induces skin barrier damage and reduces resistance to pathogenic bacteria in Lithobates catesbeianus tadpoles. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 238:113584. [PMID: 35512477 DOI: 10.1016/j.ecoenv.2022.113584] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/22/2022] [Accepted: 04/27/2022] [Indexed: 06/14/2023]
Abstract
Despite the importance of the skin mucosal barrier and commensal microbiota for the health of amphibians, the potential of environmental contaminants to disrupt the skin mucosal barrier and microbiota have rarely been studied in toxicology. In this study, tadpoles (Lithobates catesbeianus) were exposed to 0, 0.5, and 2 μg/L of microcystin-leucine arginine (MC-LR) for 30 days to explore the impacts of environmentally realistic MC-LR concentrations on the physical skin barrier, immune barrier, commensal microbiota, and skin resistance to pathogenic bacterial invasion. MC-LR exposure significantly reduced the collagen fibrils in the dermis of skin tissues and down-regulated tight junction and stratum corneum-related gene transcriptions, suggesting the damage caused by MC-LR to the physical barrier of the skin. Increased skin eosinophils and upregulated transcriptions of inflammation-related genes in the exposed tadpoles underline the development of skin inflammation resulting from MC-LR exposure even at environmentally realistic concentrations. Comparative transcriptome and immunobiochemical analyses found that antimicrobial peptides (Brevinin-1PLc, Brevinin-2GHc, and Ranatuerin-2PLa) and lysozyme were down-regulated in the exposed groups, while complement, pattern recognition receptor, and specific immune processes were up-regulated. However, the content of endotoxin lipopolysaccharide produced by bacteria increased in a dose-dependent pattern. The disc diffusion test showed a reduced ability of skin supernatant to inhibit pathogenic bacteria in the exposed groups. Analysis of microbial 16 S rRNA gene by high-throughput sequencing revealed that MC-LR interfered with the abundance, composition, and diversity of the skin commensal microbiota, which favored the growth of pathogen-containing genera Rhodococcus, Acinetobacter, and Gordonibacter. In summary, the current study provides the first clues about the impact of MC-LR on the integrity and function of skin barrier of amphibians. These new toxicological evidences can facilitate a more comprehensive evaluation of the ecological risk of MC-LR to amphibians.
Collapse
Affiliation(s)
- Yilin Shu
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China; State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Huiling Jiang
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China
| | - Calista N T Yuen
- State Key Laboratory in Marine Pollution, City University of Hong Kong, Department of Chemistry, Hong Kong, China
| | - Wenchao Wang
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China
| | - Jun He
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China
| | - Huijuan Zhang
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China
| | - Guangxuan Liu
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China
| | - Luting Wei
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China
| | - Lianguo Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| | - Hailong Wu
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China.
| |
Collapse
|
13
|
Galler AI, Klavins K, Burgener IA. A Preliminary Metabolomic Study of Yorkshire Terrier Enteropathy. Metabolites 2022; 12:metabo12030264. [PMID: 35323707 PMCID: PMC8954012 DOI: 10.3390/metabo12030264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 11/16/2022] Open
Abstract
Perturbations of metabolite profiles in human and canine enteropathies have been reported before. However, data in dogs are scarce and inconsistent. Currently, the metabolite profile in Yorkshire Terrier enteropathy (YTE) and the impact of treatment is unknown. The objective of this study was to investigate the plasma metabolome of 13 Yorkshire Terriers with YTE and compare it to 20 healthy Yorkshire Terriers. Furthermore, we studied the impact of treatment on the metabolome. In this prospective observational study, plasma metabolite profiles were analyzed by flow injection analysis-tandem mass spectrometry (FIA-MS/MS) and liquid chromatography-tandem mass spectrometry (LC-MS/MS) using a targeted metabolomics kit. Metabolite analysis revealed that YTE is accompanied by changes in lipid and bile acid metabolism. YTE was associated with a significant decrease of long-chain fatty acids (octadecenoic acid, eicosadienoic acid, eicosatrienoic acid) and lower levels of long-chain acylcarnitines (tetradecanoylcarnitine, hexadecanoylcarnitine, hexadecenoylcarnitine, octadecenoylcarnitine) compared with healthy controls. Furthermore, taurodeoxycholic acid, a secondary bile acid, was decreased in plasma from YTE patients. These changes might be breed-specific and might be involved in the pathogenesis of YTE. Interestingly, changes in metabolite levels were not recovered after treatment and differed considerably from healthy controls.
Collapse
Affiliation(s)
- Alexandra I. Galler
- Division of Small Animal Internal Medicine, University of Veterinary Medicine, 1210 Vienna, Austria;
- Correspondence:
| | - Kristaps Klavins
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Pulka St 3, LV-1007 Riga, Latvia;
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Pulka St 3, LV-1007 Riga, Latvia
| | - Iwan A. Burgener
- Division of Small Animal Internal Medicine, University of Veterinary Medicine, 1210 Vienna, Austria;
| |
Collapse
|
14
|
Suda Y, Sasaki N, Kagawa K, Elean M, Zhou B, Tomokiyo M, Islam MA, Rajoka MSR, Kober AKMH, Shimazu T, Egusa S, Terashima Y, Aso H, Ikeda-Ohtsubo W, Villena J, Kitazawa H. Immunobiotic Feed Developed with Lactobacillus delbrueckii subsp. delbrueckii TUA4408L and the Soymilk By-Product Okara Improves Health and Growth Performance in Pigs. Microorganisms 2021; 9:microorganisms9050921. [PMID: 33923082 PMCID: PMC8145491 DOI: 10.3390/microorganisms9050921] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/20/2021] [Accepted: 04/23/2021] [Indexed: 01/04/2023] Open
Abstract
Lactobacillus delbrueckii subsp. delbrueckii TUA4408L is able to differentially modulate the innate immune response of porcine intestinal epithelial cells triggered by TLR4 activation. This strain also has a remarkable ability to grow on plant substrates. These two immunological and biotechnological characteristics prompted us to evaluate whether the soymilk by-product okara fermented with the TUA4408L strain can serve as an immunobiotic feed with the ability to beneficially modulate the intestinal immunity of piglets after weaning to improve their productivity. Our in vivo studies demonstrated that the administration of immunobiotic TUA4408L-fermented okara feed significantly increased piglet growth performance and meat quality. These positive effects were associated with the ability of the TUA4408L-fermented okara feed to beneficially modulate both intestinal microbiota and immunity in pigs. The immunobiotic feed improved the abundance of the beneficial bacteria Lactobacillus and Lactococcus in the gut of pigs, reduced blood markers of inflammation, and differentially regulated the expression of inflammatory and regulatory cytokines in the intestinal mucosa. These findings indicate that the immunobiotic TUA4408L-fermented okara feed could be an economical and environmentally friendly option to improve the growth performance and immune health of pigs.
Collapse
Affiliation(s)
- Yoshihito Suda
- Department of Food Resource Development, School of Food Industrial Sciences, Miyagi University, Sendai 982-0215, Japan; (Y.S.); (N.S.); (K.K.)
| | - Nana Sasaki
- Department of Food Resource Development, School of Food Industrial Sciences, Miyagi University, Sendai 982-0215, Japan; (Y.S.); (N.S.); (K.K.)
| | - Kyoma Kagawa
- Department of Food Resource Development, School of Food Industrial Sciences, Miyagi University, Sendai 982-0215, Japan; (Y.S.); (N.S.); (K.K.)
- Graduate School of Food, Agricultural and Environmental Sciences, Miyagi University, Sendai 982-0215, Japan
| | - Mariano Elean
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman CP4000, Argentina;
| | - Binghui Zhou
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (B.Z.); (M.T.); (M.A.I.); (M.S.R.R.); (A.K.M.H.K.); (W.I.-O.)
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan;
| | - Mikado Tomokiyo
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (B.Z.); (M.T.); (M.A.I.); (M.S.R.R.); (A.K.M.H.K.); (W.I.-O.)
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan;
| | - Md. Aminul Islam
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (B.Z.); (M.T.); (M.A.I.); (M.S.R.R.); (A.K.M.H.K.); (W.I.-O.)
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan;
- Department of Medicine, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh-2202, Bangladesh
| | - Muhammad Shahid Riaz Rajoka
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (B.Z.); (M.T.); (M.A.I.); (M.S.R.R.); (A.K.M.H.K.); (W.I.-O.)
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan;
| | - A. K. M. Humayun Kober
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (B.Z.); (M.T.); (M.A.I.); (M.S.R.R.); (A.K.M.H.K.); (W.I.-O.)
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan;
- Department of Dairy and Poultry Science, Faculty of Veterinary Medicine, Chittagong Veterinary and Animal Sciences University, Khulshi, Chittagong 4225, Bangladesh
| | - Tomoyuki Shimazu
- Department of Food Science and Business, School of Food Industrial Sciences, Miyagi University, Sendai 982-0215, Japan;
| | - Shintaro Egusa
- Research and Development Division, Marusan-Ai Co., Ltd., Okazaki 444-2193, Japan; (S.E.); (Y.T.)
| | - Yuji Terashima
- Research and Development Division, Marusan-Ai Co., Ltd., Okazaki 444-2193, Japan; (S.E.); (Y.T.)
| | - Hisashi Aso
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan;
- Laboratory of Animal Health Science, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Wakako Ikeda-Ohtsubo
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (B.Z.); (M.T.); (M.A.I.); (M.S.R.R.); (A.K.M.H.K.); (W.I.-O.)
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan;
| | - Julio Villena
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman CP4000, Argentina;
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (B.Z.); (M.T.); (M.A.I.); (M.S.R.R.); (A.K.M.H.K.); (W.I.-O.)
- Correspondence: (J.V.); (H.K.)
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (B.Z.); (M.T.); (M.A.I.); (M.S.R.R.); (A.K.M.H.K.); (W.I.-O.)
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan;
- Correspondence: (J.V.); (H.K.)
| |
Collapse
|
15
|
Săsăran MO, Meliț LE, Dobru ED. MicroRNA Modulation of Host Immune Response and Inflammation Triggered by Helicobacter pylori. Int J Mol Sci 2021; 22:ijms22031406. [PMID: 33573346 PMCID: PMC7866828 DOI: 10.3390/ijms22031406] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 01/26/2021] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
Helicobacter pylori (H. pylori) remains the most-researched etiological factor for gastric inflammation and malignancies. Its evolution towards gastric complications is dependent upon host immune response. Toll-like receptors (TLRs) recognize surface and molecular patterns of the bacterium, especially the lipopolysaccharide (LPS), and act upon pathways, which will finally lead to activation of the nuclear factor-kappa B (NF-kB), a transcription factor that stimulates release of inflammatory cytokines. MicroRNAs (MiRNAs) finely modulate TLR signaling, but their expression is also modulated by activation of NF-kB-dependent pathways. This review aims to focus upon several of the most researched miRNAs on this subject, with known implications in host immune responses caused by H. pylori, including let-7 family, miRNA-155, miRNA-146, miRNA-125, miRNA-21, and miRNA-221. TLR-LPS interactions and their afferent pathways are regulated by these miRNAs, which can be considered as a bridge, which connects gastric inflammation to pre-neoplastic and malignant lesions. Therefore, they could serve as potential non-invasive biomarkers, capable of discriminating H. pylori infection, as well as its associated complications. Given that data on this matter is limited in children, as well as for as significant number of miRNAs, future research has yet to clarify the exact involvement of these entities in the progression of H. pylori-associated gastric conditions.
Collapse
Affiliation(s)
- Maria Oana Săsăran
- Department of Pediatrics III, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, Gheorghe Marinescu Street no 38, 540136 Târgu Mureș, Romania;
| | - Lorena Elena Meliț
- Department of Pediatrics I, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technol-ogy of Târgu Mureș, Gheorghe Marinescu Street no 38, 540136 Târgu Mureș, Romania
- Correspondence: ; Tel.: +40-742-984744
| | - Ecaterina Daniela Dobru
- Department of Internal Medicine VII, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, Gheorghe Marinescu Street no 38, 540136 Târgu Mureș, Romania;
| |
Collapse
|
16
|
Li J, Chen Y, Gu W, Xu F, Li H, Shan S, Sun X, Yin M, Yang G, Chen L. Characterization of a common carp intelectin gene with bacterial binding and agglutination activity. FISH & SHELLFISH IMMUNOLOGY 2021; 108:32-41. [PMID: 33249124 DOI: 10.1016/j.fsi.2020.11.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 10/27/2020] [Accepted: 11/24/2020] [Indexed: 06/12/2023]
Abstract
Intelectin (ITLN) is a type of glycan-binding lectin involved in many physiological processes and some human diseases. Here we report a common carp intelectin (cITLN). Like other orthologs, cITLN also contains a conserved fibrinogen-related domain (FReD) and a unique intelectin domain, expresses in all the tissues tested with the highest level in the hindgut, and responds to bacterial challenge in the acute phase. We also expressed cITLN in Escherichia coli (E. coli) system, and the purified recombinant cITLN could neither affect the surface of bacteria nor inhibit the growth of bacteria, but it can agglutinate both gram-positive and gram-negative bacteria in a calcium-dependent manner. The cITLN's ability of agglutination of gram-positive bacteria is stronger than that of gram-negative bacteria. This is probably because recombinant cITLN could binding peptidoglycan (PGN) with a higher degree to lipopolysaccharide (LPS). Our results of cITLN provided new insight into the function of intelectin in the intestinal mucosal immunity.
Collapse
Affiliation(s)
- Jinyi Li
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan, 250014, PR China
| | - Yanru Chen
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan, 250014, PR China
| | - Wei Gu
- Shandong Key Laboratory of Animal Microecological Preparation, Shandong Baolai-Leelai Bio-Tech Co., Ltd, No.28th, Chuangye Street, Taishan District, Tai'an, 271000, PR China
| | - Fojiao Xu
- Ramon V. del Rosario College of Business, G/F Faculty Center, 2401 Taft Avenue, 1004, Manila, Philippines
| | - Hua Li
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan, 250014, PR China
| | - Shijuan Shan
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan, 250014, PR China
| | - Xiaojie Sun
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan, 250014, PR China
| | - Miao Yin
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan, 250014, PR China
| | - Guiwen Yang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan, 250014, PR China
| | - Lei Chen
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan, 250014, PR China.
| |
Collapse
|
17
|
Caiazzo E, Cerqua I, Riemma MA, Turiello R, Ialenti A, Schrader J, Fiume G, Caiazza C, Roviezzo F, Morello S, Cicala C. Exacerbation of Allergic Airway Inflammation in Mice Lacking ECTO-5'-Nucleotidase (CD73). Front Pharmacol 2020; 11:589343. [PMID: 33328996 PMCID: PMC7734328 DOI: 10.3389/fphar.2020.589343] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/20/2020] [Indexed: 12/15/2022] Open
Abstract
The airways are a target tissue of type I allergies and atopy is the main etiological factor of bronchial asthma. A predisposition to allergy and individual response to allergens are dependent upon environmental and host factors. Early studies performed to clarify the role of extracellular adenosine in the airways highlighted the importance of adenosine-generating enzymes CD73, together with CD39, as an innate protection system against lung injury. In experimental animals, deletion of CD73 has been associated with immune and autoimmune diseases. Our experiments have been performed to investigate the role of CD73 in the assessment of allergic airway inflammation following sensitization. We found that in CD73−/− mice sensitization, induced by subcutaneous ovalbumin (OVA) administration, increased signs of airway inflammation and atopy developed, characterized by high IgE plasma levels and increased pulmonary cytokines, reduced frequency of lung CD4+CD25+Foxp3+ T cells, but without bronchial hyperreactivity, compared to sensitized wild type mice. Our results provide evidence that the lack of CD73 causes an uncontrolled allergic sensitization, suggesting that CD73 is a key molecule at the interface between innate and adaptive immune response. The knowledge of host immune factors controlling allergic sensitization is of crucial importance and might help to find preventive interventions that could act before an allergy develops.
Collapse
Affiliation(s)
- Elisabetta Caiazzo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Ida Cerqua
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Maria Antonietta Riemma
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Roberta Turiello
- Department of Pharmacy, University of Salerno, Salerno, Italy.,PhD Program in Drug Discovery and Development, University of Salerno, Salerno, Italy
| | - Armando Ialenti
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Jurgen Schrader
- Department of Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
| | - Giuseppe Fiume
- Department of Experimental and Clinical Medicine, University of Catanzaro Magna Graecia, Catanzaro, Italy
| | - Carmen Caiazza
- Department of Molecular Medicine and Medical Biotechnologies, School of Medicine and Surgery, University of Naples 'Federico II', Naples, Italy
| | - Fiorentina Roviezzo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Silvana Morello
- Department of Pharmacy, University of Salerno, Salerno, Italy
| | - Carla Cicala
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| |
Collapse
|
18
|
Mohammadi G, Rafiee G, Abdelrahman HA. Effects of dietary Lactobacillus plantarum (KC426951) in biofloc and stagnant-renewal culture systems on growth performance, mucosal parameters, and serum innate responses of Nile tilapia Oreochromis niloticus. FISH PHYSIOLOGY AND BIOCHEMISTRY 2020; 46:1167-1181. [PMID: 32133574 DOI: 10.1007/s10695-020-00777-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 02/20/2020] [Indexed: 06/10/2023]
Abstract
Effects of dietary Lactobacillus plantarum (KC426951) on growth and innate responses of Nile tilapia Oreochromis niloticus were evaluated in biofloc technology system and stagnant-renewal culture system (SRCS). The 90-day-long experiment contained four treatments: SRCS without probiotic (T1), SRCS with probiotic (T2), biofloc without probiotic (T3), and biofloc with probiotic (T4). The administration dose of probiotic was 2 × 108 CFU kg-1 diet. At the end of experiment, the mean final weights, specific growth rates, feed conversion ratios, and total biomass were significantly (P < 0.05) better in BFT treatments, with no significant effect of probiotic on these parameters in both culture systems. Meanwhile, skin mucosal parameters including total protein (TP), lysozyme (LYZ), alkaline phosphatase (ALP), and protease (PRO) activity were significantly enhanced following probiotic supplementation. T4 treatment displayed a significantly higher LYZ and ALP activity in mucus versus other treatments. Also, serum alternative complement activity was significantly heightened in probiotic-supplemented fish. Superoxide dismutase activity in T4 was detected higher than that of SRCS groups. The results of the current study demonstrated the enhancement of some mucosal and serum innate responses of Nile tilapia in both culture systems upon L. plantarum (KC426951) supplementation.
Collapse
Affiliation(s)
- Ghasem Mohammadi
- Department of Fisheries Sciences, Faculty of Natural Resources, University of Tehran, Karaj, Iran.
| | - Gholamreza Rafiee
- Department of Fisheries Sciences, Faculty of Natural Resources, University of Tehran, Karaj, Iran.
| | - Hisham A Abdelrahman
- Department of Veterinary Hygiene and Management, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
19
|
Wang J, Li P, Yu Y, Fu Y, Jiang H, Lu M, Sun Z, Jiang S, Lu L, Wu MX. Pulmonary surfactant-biomimetic nanoparticles potentiate heterosubtypic influenza immunity. Science 2020; 367:367/6480/eaau0810. [PMID: 32079747 DOI: 10.1126/science.aau0810] [Citation(s) in RCA: 202] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 05/03/2019] [Accepted: 12/23/2019] [Indexed: 12/12/2022]
Abstract
Current influenza vaccines only confer protection against homologous viruses. We synthesized pulmonary surfactant (PS)-biomimetic liposomes encapsulating 2',3'-cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), an agonist of the interferon gene inducer STING (stimulator of interferon genes). The adjuvant (PS-GAMP) vigorously augmented influenza vaccine-induced humoral and CD8+ T cell immune responses in mice by simulating the early phase of viral infection without concomitant excess inflammation. Two days after intranasal immunization with PS-GAMP-adjuvanted H1N1 vaccine, strong cross-protection was elicited against distant H1N1 and heterosubtypic H3N2, H5N1, and H7N9 viruses for at least 6 months while maintaining lung-resident memory CD8+ T cells. Adjuvanticity was then validated in ferrets. When alveolar epithelial cells (AECs) lacked Sting or gap junctions were blocked, PS-GAMP-mediated adjuvanticity was substantially abrogated in vivo. Thus, AECs play a pivotal role in configuring heterosubtypic immunity.
Collapse
Affiliation(s)
- Ji Wang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA 02114, USA.,Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou 510080, China
| | - Peiyu Li
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA 02114, USA.,Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Biosafety Level 3 Laboratory, Fudan University, Shanghai 200032, China
| | - Yang Yu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA 02114, USA
| | - Yuhong Fu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Biosafety Level 3 Laboratory, Fudan University, Shanghai 200032, China
| | - Hongye Jiang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA 02114, USA
| | - Min Lu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA 02114, USA
| | - Zhiping Sun
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Biosafety Level 3 Laboratory, Fudan University, Shanghai 200032, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Biosafety Level 3 Laboratory, Fudan University, Shanghai 200032, China
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Biosafety Level 3 Laboratory, Fudan University, Shanghai 200032, China.
| | - Mei X Wu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
20
|
Varga JFA, Bui-Marinos MP, Katzenback BA. Frog Skin Innate Immune Defences: Sensing and Surviving Pathogens. Front Immunol 2019; 9:3128. [PMID: 30692997 PMCID: PMC6339944 DOI: 10.3389/fimmu.2018.03128] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 12/18/2018] [Indexed: 01/26/2023] Open
Abstract
Amphibian skin is a mucosal surface in direct and continuous contact with a microbially diverse and laden aquatic and/or terrestrial environment. As such, frog skin is an important innate immune organ and first line of defence against pathogens in the environment. Critical to the innate immune functions of frog skin are the maintenance of physical, chemical, cellular, and microbiological barriers and the complex network of interactions that occur across all the barriers. Despite the global decline in amphibian populations, largely as a result of emerging infectious diseases, we understand little regarding the cellular and molecular mechanisms that underlie the innate immune function of amphibian skin and defence against pathogens. In this review, we discuss the structure, cell composition and cellular junctions that contribute to the skin physical barrier, the antimicrobial peptide arsenal that, in part, comprises the chemical barrier, the pattern recognition receptors involved in recognizing pathogens and initiating innate immune responses in the skin, and the contribution of commensal microbes on the skin to pathogen defence. We briefly discuss the influence of environmental abiotic factors (natural and anthropogenic) and pathogens on the immunocompetency of frog skin defences. Although some aspects of frog innate immunity, such as antimicrobial peptides are well-studied; other components and how they contribute to the skin innate immune barrier, are lacking. Elucidating the complex network of interactions occurring at the interface of the frog's external and internal environments will yield insight into the crucial role amphibian skin plays in host defence and the environmental factors leading to compromised barrier integrity, disease, and host mortality.
Collapse
Affiliation(s)
- Joseph F A Varga
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | | | | |
Collapse
|
21
|
Borghini L, Hibberd M, Davila S. Changes in H3K27ac following lipopolysaccharide stimulation of nasopharyngeal epithelial cells. BMC Genomics 2018; 19:969. [PMID: 30587130 PMCID: PMC6307289 DOI: 10.1186/s12864-018-5295-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 11/21/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The epithelium is the first line of defense against pathogens. Notably the epithelial cells lining the respiratory track are crucial in sensing airborne microbes and mounting an effective immune response via the expression of target genes such as cytokines and chemokines. Gene expression regulation following microbial recognition is partly regulated by chromatin re-organization and has been described in immune cells but data from epithelial cells is not as detailed. Here, we report genome-wide changes of the H3K27ac mark, characteristic of activated enhancers and promoters, after stimulation of nasopharyngeal epithelial cells with the bacterial endotoxin Lipopolysaccharide (LPS). RESULTS In this study, we have identified 626 regions where the H3K27ac mark showed reproducible increase following LPS induction in epithelial cells. This indicated that sensing of LPS led to opening of the chromatin in our system. Moreover, this phenomenon seemed to happen extensively at enhancers regions and we could observe instances of Super-enhancer formation. As expected, LPS-increased H3K27ac regions were found in the vicinity of genes relevant for LPS response and these changes correlated with up-regulation of their expression. In addition, we found the induction of H3K27ac mark to overlap with the binding of one of the NF-kB members and key regulator of the innate immune response, RELA, following LPS sensing. Indeed, inhibiting the NF-kB pathway abolished the deposition of H3K27ac at the TNF locus, a target of RELA, suggesting that these two phenomena are associated. CONCLUSIONS Enhancers' selection and activation following microbial or inflammatory stimuli has been described previously and shown to be mediated via the NF-kB pathway. Here, we demonstrate that this is also likely to occur in the case of LPS-sensing by nasopharyngeal epithelial cells as well. In addition to validating previous findings, we generated a valuable data set relevant to the host immune response to epithelial cell colonizing or infecting pathogens.
Collapse
Affiliation(s)
- Lisa Borghini
- Human Genetics, Genome Institute of Singapore, Singapore, 138672, Singapore. .,Infectious Disease, Genome Institute of Singapore, Singapore, 138672, Singapore.
| | - Martin Hibberd
- Infectious Disease, Genome Institute of Singapore, Singapore, 138672, Singapore.,Present Address: Pathogen Molecular Biology, Infectious & Tropical Disease, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Sonia Davila
- Human Genetics, Genome Institute of Singapore, Singapore, 138672, Singapore.,Present Address: SingHealth Duke-NUS Institute of Precision Medicine (PRISM), Singapore, 169609, Singapore
| |
Collapse
|
22
|
Ebner F, Kuhring M, Radonić A, Midha A, Renard BY, Hartmann S. Silent Witness: Dual-Species Transcriptomics Reveals Epithelial Immunological Quiescence to Helminth Larval Encounter and Fostered Larval Development. Front Immunol 2018; 9:1868. [PMID: 30158930 PMCID: PMC6104121 DOI: 10.3389/fimmu.2018.01868] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 07/30/2018] [Indexed: 11/17/2022] Open
Abstract
Gastrointestinal nematodes are among the most prevalent parasites infecting humans and livestock worldwide. Infective larvae of the soil-transmitted nematode Ascaris spp. enter the host and start tissue migration by crossing the intestinal epithelial barrier. The initial interaction of the intestinal epithelium with the parasite, however, has received little attention. In a time-resolved interaction model of porcine intestinal epithelial cells (IPEC-J2) and infective Ascaris suum larvae, we addressed the early transcriptional changes occurring simultaneously in both organisms using dual-species RNA-Seq. Functional analysis of the host response revealed an overall induction of metabolic activity, without induction of immune responsive genes or immune signaling pathways and showing suppression of chemotactic genes like CXCL8/IL-8 or CHI3L1. Ascaris larvae, when getting in contact with the epithelium, showed induction of genes that orchestrate motor activity and larval development, such as myosin, troponin, myoglobin, and protein disulfide isomerase 2 (PDI-2). In addition, excretory-secretory products that likely facilitate parasite invasion were increased, among them, aspartic protease 6 or hyaluronidase. Integration of host and pathogen data in an interspecies gene co-expression network indicated links between nematode fatty acid biosynthesis and host ribosome assembly/protein synthesis. In summary, our study provides new molecular insights into the early factors of parasite invasion, while at the same time revealing host immunological unresponsiveness. Reproducible software for dual RNA-Seq analysis of non-model organisms is available at https://gitlab.com/mkuhring/project_asuum and can be applied to similar studies.
Collapse
Affiliation(s)
- Friederike Ebner
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Mathias Kuhring
- Bioinformatics Unit (MF1), Department for Methods Development and Research Infrastructure, Robert Koch Institute, Berlin, Germany.,Core Unit Bioinformatics, Berlin Institute of Health (BIH), Berlin, Germany.,Berlin Institute of Health Metabolomics Platform, Berlin Institute of Health (BIH), Berlin, Germany.,Max Delbrück Center (MDC) for Molecular Medicine, Berlin, Germany
| | - Aleksandar Radonić
- Center for Biological Threats and Special Pathogens: Highly Pathogenic Viruses (ZBS 1), Robert Koch Institute, Berlin, Germany
| | - Ankur Midha
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Bernhard Y Renard
- Bioinformatics Unit (MF1), Department for Methods Development and Research Infrastructure, Robert Koch Institute, Berlin, Germany
| | - Susanne Hartmann
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
23
|
Peiravan A, Bertolini F, Rothschild MF, Simpson KW, Jergens AE, Allenspach K, Werling D. Genome-wide association studies of inflammatory bowel disease in German shepherd dogs. PLoS One 2018; 13:e0200685. [PMID: 30028859 PMCID: PMC6054420 DOI: 10.1371/journal.pone.0200685] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 05/14/2018] [Indexed: 12/12/2022] Open
Abstract
Canine Inflammatory Bowel Disease (IBD) is considered a multifactorial disease caused by complex interactions between the intestinal immune system, intestinal microbiota and environmental factors in genetically susceptible individuals. Although IBD can affect any breed, German shepherd dogs (GSD) in the UK are at increased risk of developing the disease. Based on previous evidence, the aim of the present study was to identify single nucleotide polymorphisms (SNPs), which may confer genetic susceptibility or resistance to IBD using a genome-wide association study (GWAS). Genomic DNA was extracted from EDTA blood or saliva samples of 96 cases and 98 controls. Genotyping of cases and controls was performed on the Canine Illumina HD SNP array and data generated was analyzed using PLINK. Several SNPs and regions on chromosomes 7,9,11 and 13 were detected to be associated with IBD using different SNP-by-SNP association methods and FST windows approach. Searching one Mb up-and down-stream of the most significant SNPs, as identified by single SNP analysis as well as 200Kb before and after the start and the end position of the associated regions identified by FST windows approach, we identified 63 genes. Using a combination of pathways analysis and a list of genes that have been reported to be involved in human IBD, we identified 16 candidate genes potentially associated with IBD in GSD.
Collapse
Affiliation(s)
- Atiyeh Peiravan
- Department of Pathology and Pathogen Biology, Royal Veterinary College, University of London, North Mymms, United Kingdom
| | - Francesca Bertolini
- Department of Animal Science, Iowa State University, Ames, Iowa, United States of America
| | - Max F. Rothschild
- Department of Animal Science, Iowa State University, Ames, Iowa, United States of America
| | - Kenneth W. Simpson
- College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Albert E. Jergens
- College of Veterinary Medicine, Iowa State University, Ames, Iowa, United States of America
| | - Karin Allenspach
- College of Veterinary Medicine, Iowa State University, Ames, Iowa, United States of America
| | - Dirk Werling
- Department of Pathology and Pathogen Biology, Royal Veterinary College, University of London, North Mymms, United Kingdom
| |
Collapse
|
24
|
Borghini L, Lu J, Hibberd M, Davila S. Variation in Genome-Wide NF-κB RELA Binding Sites upon Microbial Stimuli and Identification of a Virus Response Profile. THE JOURNAL OF IMMUNOLOGY 2018; 201:1295-1305. [PMID: 29959281 DOI: 10.4049/jimmunol.1800246] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 06/04/2018] [Indexed: 12/22/2022]
Abstract
NF-κB transcription factors are master regulators of the innate immune response. Activated downstream of pathogen recognition receptors, they regulate the expression of genes to help fight infections as well as recruit the adaptive immune system. NF-κB responds to a wide variety of signals, but the processes by which stimulus specificity is attained remain unclear. In this article, we characterized the response of one NF-κB member, RELA, to four stimuli mimicking infection in human nasopharyngeal epithelial cells. Comparing genome-wide RELA binding, we observed stimulus-specific sites, although most sites overlapped across stimuli. Specifically, the response to poly I:C (mimicking viral dsRNA and signaling through TLR3) induced a distinct RELA profile, binding in the vicinity of antiviral genes and correlating with corresponding gene expression. This group of binding sites was also enriched in IFN regulatory factor motifs and showed overlapping with IFN regulatory factor binding sites. A novel NF-κB target, OASL, was further validated and showed TLR3-specific activation. This work showed that some RELA DNA binding sites varied in activation response following different stimulations and that interaction with more specialized factors could help achieve this stimulus-specific activity. Our data provide a genomic view of regulated host response to different pathogen stimuli.
Collapse
Affiliation(s)
- Lisa Borghini
- Department of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore; .,Department of Infectious Diseases, Genome Institute of Singapore, Singapore 138672, Singapore; and
| | - Jinhua Lu
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine and Immunology Program, National University of Singapore, Singapore 119077
| | - Martin Hibberd
- Department of Infectious Diseases, Genome Institute of Singapore, Singapore 138672, Singapore; and
| | - Sonia Davila
- Department of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore;
| |
Collapse
|
25
|
Effects of 18 β-Glycyrrhetinic Acid on Fungal Protease-Induced Airway Inflammatory Responses. Mediators Inflamm 2018; 2018:6461032. [PMID: 29861658 PMCID: PMC5976916 DOI: 10.1155/2018/6461032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/21/2018] [Accepted: 03/28/2018] [Indexed: 01/30/2023] Open
Abstract
Airway epithelial cells secrete diverse inflammatory mediators in response to various stimuli. Thus, early regulation of immune responses in the airway epithelium is likely critical for the control of chronic inflammatory diseases. The purpose of the present study was to evaluate the effects of 18β-glycyrrhetinic acid (GA) on inflammatory responses generated in response to a fungal protease allergen that induces epithelial damage. To understand the underlying mechanisms, we also investigated the inhibitory effects of GA on the production of mitochondrial reactive oxygen species (ROS) in the human bronchial epithelial cell line BEAS2B. In this study, GA treatment reduced cytokine production and the human neutrophil cell line HL60 migration through decreased mitochondrial ROS production. In addition, GA significantly reduced inflammatory cell infiltration and cytokine levels in the bronchoalveolar lavage (BAL) fluid of fungal allergen-administered mice. Inhibitory effects of GA are dependent on the mitochondrial ROS/MAPK axis. Moreover, the effect of GA on the regulation of mitochondrial ROS depends on the expression of uncoupling protein-2 (UCP-2). Taken together, GA might represent a potential therapeutic agent for blocking inflammatory responses in airways.
Collapse
|
26
|
Lei-Leston AC, Murphy AG, Maloy KJ. Epithelial Cell Inflammasomes in Intestinal Immunity and Inflammation. Front Immunol 2017; 8:1168. [PMID: 28979266 PMCID: PMC5611393 DOI: 10.3389/fimmu.2017.01168] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/04/2017] [Indexed: 12/12/2022] Open
Abstract
Pattern recognition receptors (PRR), such as NOD-like receptors (NLRs), sense conserved microbial signatures, and host danger signals leading to the coordination of appropriate immune responses. Upon activation, a subset of NLR initiate the assembly of a multimeric protein complex known as the inflammasome, which processes pro-inflammatory cytokines and mediates a specialized form of cell death known as pyroptosis. The identification of inflammasome-associated genes as inflammatory bowel disease susceptibility genes implicates a role for the inflammasome in intestinal inflammation. Despite the fact that the functional importance of inflammasomes within immune cells has been well established, the contribution of inflammasome expression in non-hematopoietic cells remains comparatively understudied. Given that intestinal epithelial cells (IEC) act as a barrier between the host and the intestinal microbiota, inflammasome expression by these cells is likely important for intestinal immune homeostasis. Accumulating evidence suggests that the inflammasome plays a key role in shaping epithelial responses at the host-lumen interface with many inflammasome components highly expressed by IEC. Recent studies have exposed functional roles of IEC inflammasomes in mucosal immune defense, inflammation, and tumorigenesis. In this review, we present the main features of the predominant inflammasomes and their effector mechanisms contributing to intestinal homeostasis and inflammation. We also discuss existing controversies in the field and open questions related to their implications in disease. A comprehensive understanding of the molecular basis of intestinal inflammasome signaling could hold therapeutic potential for clinical translation.
Collapse
Affiliation(s)
- Andrea C Lei-Leston
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Alison G Murphy
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Kevin J Maloy
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
27
|
Chi W, Hua X, Chen X, Bian F, Yuan X, Zhang L, Wang X, Chen D, Deng R, Li Z, Liu Y, de Paiva CS, Pflugfelder SC, Li DQ. Mitochondrial DNA oxidation induces imbalanced activity of NLRP3/NLRP6 inflammasomes by activation of caspase-8 and BRCC36 in dry eye. J Autoimmun 2017; 80:65-76. [PMID: 28238526 DOI: 10.1016/j.jaut.2017.02.006] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 12/19/2016] [Accepted: 02/14/2017] [Indexed: 12/27/2022]
Abstract
The concept of innate immunity has been expanded to recognize environmental pathogens other than microbial components. However, whether and how the innate immunity is initiated by epithelium in response to environmental physical challenges such as low humidity and high osmolarity in an autoimmune disease, dry eye, is still largely unknown. Using two experimental dry eye models, primary human corneal epithelial cultures exposed to hyperosmolarity and mouse ocular surface facing desiccating stress, we uncovered novel innate immunity pathway by ocular surface epithelium, where oxidized mitochondrial DNA induces imbalanced activation of NLRP3/NLRP6 inflammasomes via stimulation of caspase-8 and BRCC36 in response to environmental stress. Activated NLRP3 with suppressed NLRP6 stimulates caspase-1 activation that leads to IL-1β and IL-18 maturation and secretion. NLRP3-independent caspase-8 noncanonically activates caspase-1 via reciprocal regulation of NLRP3/NLRP6-mediated inflammasomes. Reactive oxygen species-induced mitochondrial DNA oxidative damage and BRCC36 deubiquitinating activity provide a missing link and mechanism by which innate immunity responds to environmental stress via caspase-8-involved NLRP3/NLRP6 inflammasomes.
Collapse
Affiliation(s)
- Wei Chi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China; Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA.
| | - Xia Hua
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA; Second Hospital of Tianjin Medical University, Tianjin, China.
| | - Xin Chen
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA; School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China.
| | - Fang Bian
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA.
| | - Xiaoyong Yuan
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA.
| | - Lili Zhang
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - Xiaoran Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China; Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA.
| | - Ding Chen
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA; School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China.
| | - Ruzhi Deng
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA; School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China.
| | - Zhijie Li
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
| | - Yizhi Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | - Cintia S de Paiva
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA.
| | - Stephen C Pflugfelder
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA.
| | - De-Quan Li
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
28
|
Kim YH, Lee SH. Mitochondrial reactive oxygen species regulate fungal protease-induced inflammatory responses. Toxicology 2017; 378:86-94. [PMID: 28087464 DOI: 10.1016/j.tox.2017.01.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 01/06/2017] [Accepted: 01/09/2017] [Indexed: 01/27/2023]
Abstract
Epidemiological studies have shown that fungal infections are a main cause of respiratory tract diseases, such as asthma, bronchopneumonia, intoxication, and invasive fungal disease. Fungi such as Aspergillus and Candida species have become increasingly important pathogens as the global climate changes. Accordingly, in this study, we evaluated the toxicological potential of Aspergillus protease in the lower respiratory tract. Exposure of Aspergillus protease to A549 cells induced upregulation of tumor necrosis factor (TNF)-α, monocyte chemoattractant protein (MCP)-1, and intercellular adhesion molecule (ICAM)-1 mRNAs and increased production of interleukin (IL)-8 and MCP-1 protein through enhanced mitochondrial reactive oxygen species (ROS) generation and activation of mitogen-activated protein kinase (MAPK) and activator protein (AP)-1. Furthermore, the mitochondrial ROS scavenger Mito-TEMPO, which inhibited MAPK and AP-1, significantly reduced MCP-1 and IL-1β mRNA expression and reduced HL-60 cell migration through the suppression of MCP-1 and IL-8 protein secretion. Thus, our results demonstrated that mitochondria were an important source of Aspergillus protease-stimulated ROS and that regulation of mitochondrial ROS modulated inflammatory responses by preventing activation of MAPK and AP-1 in A549 cells.
Collapse
Affiliation(s)
- Yun Hee Kim
- Graduate School of Medical Science and Engineering, Biomedical Research Center, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea; Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 34054, Republic of Korea
| | - Seung-Hyo Lee
- Graduate School of Medical Science and Engineering, Biomedical Research Center, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.
| |
Collapse
|
29
|
Lauriano ER, Pergolizzi S, Capillo G, Kuciel M, Alesci A, Faggio C. Immunohistochemical characterization of Toll-like receptor 2 in gut epithelial cells and macrophages of goldfish Carassius auratus fed with a high-cholesterol diet. FISH & SHELLFISH IMMUNOLOGY 2016; 59:250-255. [PMID: 27818343 DOI: 10.1016/j.fsi.2016.11.003] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 10/28/2016] [Accepted: 11/02/2016] [Indexed: 06/06/2023]
Abstract
Toll-like receptors (TLRs) are a group of pattern recognition molecules that play a crucial role in innate immunity. The structural conservation of the archaic TLR system suggests that the regulation of the immune response might be similar in fish and mammals. Several TLRs (TLR-1, -2, and -4) are expressed by activated macrophages, "foam cells" in human atherosclerotic lesions. To date, 20 different TLRs were identified in more than a dozen different fish species. In this study we found that feeding goldfish, Carrassius auratus, a high-cholesterol diet (HCD) resulted macrophage foam cell formation in the intestinal tissues. The expression of TLR2 has been found in foam cells and in the cytoplasm of enterocytes, however the staining was more intense at the apical surface of polarized intestinal epithelial cells and in the lamina propria. In the intestinal epithelial cells and in the lamina propria cells of the control fish the TLR2 was expressed at low levels. The intestinal epithelium is directly involved in the mucosal immune response through its expression of proinflammatory genes, release of inflammatory cytokines, and recruitment of inflammatory cells.
Collapse
Affiliation(s)
- E R Lauriano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - S Pergolizzi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - G Capillo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - M Kuciel
- Department of Comparative Anatomy, Institute of Zoology, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland
| | - A Alesci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - C Faggio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy.
| |
Collapse
|
30
|
Abstract
OBJECTIVE An adequate bowel preparation is essential for a successful colonoscopy, but to date, only scarce information exists on the impact of the bowel cleansing on the gut microbiota, in particular, 1 month after the procedure. PATIENTS AND METHODS Through 16S rDNA Ion Torrent profiling of fecal samples of 10 patients, we evaluated changes that occurred in the gut microbiota composition immediately after a 4 liter polyethylene glycol-based (SELG Esse) bowel lavage and 1 month thereafter. We studied the gut microbiota at the phylum, class, and family level. RESULTS At the phyla level, we found a significant decrease in Firmicutes abundance and an increase in Proteobacteria abundance immediately after the colon cleansing and 1 month after the colonoscopy, whereas, at the class level, a significant increase in γ-Proteobacteria immediately after the colonoscopy was observed. Interestingly, 1 month after the endoscopic examination, this bacterial class was decreased 2.5-fold compared with samples before colonoscopy, as well as α-Proteobacteria. At the family level, a significant reduction in Lactobacillaceae and an increase in Enterobacteriaceae abundance were observed immediately after the colonoscopy, whereas 1 month after the bowel cleansing, these families were significantly lower compared with samples collected before the colonoscopy. Moreover, the abundance of Rikenellaceae and Eubacteriaceae has been observed to be significantly higher compared with samples collected before the bowel lavage. Finally, Streptococcaceae were increased 4.0-fold 1 month after the bowel lavage compared with fecal samples collected before the colonoscopy. CONCLUSION We provide clear evidence that, in normal individuals, a high-volume polyethylene glycol bowel cleansing preparation has a long-lasting effect on the gut microbiota composition and homeostasis, in particular, with a decrease in the Lactobacillaceae abundance, a population of protective bacteria. Further studies are required to assess whether these changes have any metabolic, immunological, or clinical consequence.
Collapse
|
31
|
Association of NOD1 and NOD2 polymorphisms with Guillain-Barré syndrome in Northern Indian population. J Neurol Sci 2016; 363:57-62. [PMID: 27000222 DOI: 10.1016/j.jns.2016.02.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 01/30/2016] [Accepted: 02/11/2016] [Indexed: 11/24/2022]
Abstract
BACKGROUND Nucleotide oligomerization domain (NOD) proteins are cytosolic pattern recognition receptors that respond to bacterial substrate and induce NF-κB activation in host. Association of NOD polymorphisms have been studied in many autoimmune disorders, however its role in Guillain-Barré syndrome (GBS) remains unknown. We have investigated NOD1 Glu266Lys and NOD2 (Arg702Trp and Gly908Arg) gene polymorphisms among patients with GBS. MATERIALS AND METHOD Polymorphisms in NOD-1 (Glu266Lys) and NOD-2 (Arg702Trp and Gly908Arg) genes were studied using polymerase chain reaction-restriction fragment length polymorphism in 105 patients with GBS and 100 healthy controls. RESULTS Homozygous genotype (Lys/Lys) of NOD1 was significantly associated with GBS (p=0.013); and its subtypes viz. acute motor axonal neuropathy (AMAN) and acute inflammatory demyelinating polyneuropathy (AIDP) (p=0.008 and p=0.024 respectively) than controls. In NOD2 (Arg702Trp and Gly908Arg) polymorphisms, only heterozygous genotype (Arg/Trp and Gly/Arg) showed significant association with GBS (p=0.001 and p=0.01 respectively); subtypes AMAN, acute motor-sensory axonal neuropathy (AMSAN) and AIDP showed association with heterozygote Arg702Trp (p=0.001; p=0.029 and p=0.001 respectively) whereas only AIDP was associated with heterozygote genotype Gly908Arg (p=0.003). CONCLUSION NOD1 (Glu266Lys) and NOD2 (Arg702Trp and Gly908Arg) polymorphisms were associated with an increased susceptibility to GBS. These polymorphisms could be genetic marker to GBS susceptibility.
Collapse
|
32
|
Abstract
Eukaryotes have evolved strategies to detect microbial intrusion and instruct immune responses to limit damage from infection. Recognition of microbes and cellular damage relies on the detection of microbe-associated molecular patterns (MAMPs, also called PAMPS, or pathogen-associated molecular patterns) and so-called "danger signals" by various families of host pattern recognition receptors (PRRs). Members of the recently identified protein family of nucleotide-binding domain andleucine-rich-repeat-containing proteins (NLR), including Nod1, Nod2, NLRP3, and NLRC4, have been shown to detect specific microbial motifs and danger signals for regulating host inflammatory responses. Moreover, with the discovery that polymorphisms in NOD1, NOD2, NLRP1, and NLRP3 are associated with susceptibility to chronic inflammatory disorders, the view has emerged that NLRs act not only as sensors butalso can serve as signaling platforms for instructing and balancing host immune responses. In this chapter, we explore the functions of these intracellular innate immune receptors and examine their implication in inflammatory diseases.
Collapse
|
33
|
Nizard M, Diniz MO, Roussel H, Tran T, Ferreira LC, Badoual C, Tartour E. Mucosal vaccines: novel strategies and applications for the control of pathogens and tumors at mucosal sites. Hum Vaccin Immunother 2015; 10:2175-87. [PMID: 25424921 DOI: 10.4161/hv.29269] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The mucosal immune system displays several adaptations reflecting the exposure to the external environment. The efficient induction of mucosal immune responses also requires specific approaches, such as the use of appropriate administration routes and specific adjuvants and/or delivery systems. In contrast to vaccines delivered via parenteral routes, experimental, and clinical evidences demonstrated that mucosal vaccines can efficiently induce local immune responses to pathogens or tumors located at mucosal sites as well as systemic response. At least in part, such features can be explained by the compartmentalization of mucosal B and T cell populations that play important roles in the modulation of local immune responses. In the present review, we discuss molecular and cellular features of the mucosal immune system as well as novel immunization approaches that may lead to the development of innovative and efficient vaccines targeting pathogens and tumors at different mucosal sites.
Collapse
Affiliation(s)
- Mevyn Nizard
- a INSERM U970; Universite Paris Descartes; Sorbonne Paris-Cité; Paris, France
| | | | | | | | | | | | | |
Collapse
|
34
|
Association between nucleotide oligomerisation domain two (Nod2) gene polymorphisms and canine inflammatory bowel disease. Vet Immunol Immunopathol 2014; 161:32-41. [DOI: 10.1016/j.vetimm.2014.06.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Revised: 06/06/2014] [Accepted: 06/19/2014] [Indexed: 02/07/2023]
|
35
|
Song-Zhao GX, Srinivasan N, Pott J, Baban D, Frankel G, Maloy KJ. Nlrp3 activation in the intestinal epithelium protects against a mucosal pathogen. Mucosal Immunol 2014; 7:763-774. [PMID: 24280937 PMCID: PMC4048180 DOI: 10.1038/mi.2013.94] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 10/01/2013] [Indexed: 02/04/2023]
Abstract
Polymorphisms in the intracellular pattern recognition receptor gene NLRP3 (NLR family, pyrin domain containing 3) have been associated with susceptibility to Crohn's disease, a type of inflammatory bowel disease. Following tissue damage or infection, NLRP3 triggers the formation of inflammasomes, containing NLRP3, ASC (apoptosis-associated speck-like protein containing a CARD domain), and caspase-1, that mediate secretion of interleukin (IL)-1β and IL-18. However, the precise role of NLRP3 inflammasomes in mucosal inflammation and barrier protection remains unclear. Here we show that upon infection with the attaching/effacing intestinal pathogen Citrobacter rodentium, Nlrp3(-/-) and Asc(-/-) mice displayed increased bacterial colonization and dispersion, more severe weight loss, and exacerbated intestinal inflammation. Analyses of irradiation bone marrow chimeras revealed that protection from disease was mediated through Nlrp3 activation in nonhematopoietic cells and was initiated very early after infection. Thus, early activation of Nlrp3 in intestinal epithelial cells limits pathogen colonization and prevents subsequent pathology, potentially providing a functional link between NLRP3 polymorphisms and susceptibility to inflammatory bowel disease.
Collapse
Affiliation(s)
- George X. Song-Zhao
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK.
| | - Naren Srinivasan
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK.
| | - Johanna Pott
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK.
| | - Dilair Baban
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK.
| | - Gad Frankel
- MRC Centre for Molecular Bacteriology and Infection, Division of Cell and Molecular Biology, Imperial College London, London, UK.
| | - Kevin J. Maloy
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK.
,Corresponding author: T: +44-(0)1865-275589 F: +44-(0)1865-275591
| |
Collapse
|
36
|
Suda Y, Villena J, Takahashi Y, Hosoya S, Tomosada Y, Tsukida K, Shimazu T, Aso H, Tohno M, Ishida M, Makino S, Ikegami S, Kitazawa H. Immunobiotic Lactobacillus jensenii as immune-health promoting factor to improve growth performance and productivity in post-weaning pigs. BMC Immunol 2014; 15:24. [PMID: 24943108 PMCID: PMC4068960 DOI: 10.1186/1471-2172-15-24] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 06/09/2014] [Indexed: 11/10/2022] Open
Abstract
Background Immunoregulatory probiotics (immunobiotics) have been proposed to improve piglets’ immune system to avoid intestinal infections and reduce unproductive inflammation after weaning. Previously, it was demonstrated that Lactobacillus jensenii TL2937 (LjTL2937) attenuated the inflammatory response triggered by activation of Toll-like receptor 4 (TLR-4) in porcine intestinal epithelial (PIE) cells and antigen presenting cells (APCs) from porcine Peyer’s patches (PP). Objective In view of the critical importance of PIE-APCs interactions in the regulation of intestinal immune responses, we aimed to examine the effect of LjTL2937 on activation patterns of APCs from swine PPs in co-cultures with PIE cells. In addition, we investigated whether LjTL2937 was able to beneficially modulate intestinal immunity of piglets after weaning to improve immune-health status. Results Stimulation of PIE-APCs co-cultures with LjTL2937 increased the expression of MHC-II, CD80/86, IL-10, and Bcl-3 in CD172a+CD11R1- and CD172a+CD11R1high APCs. In addition, the TL2937 strain caused the upregulation of three negative regulators of TLR4 in PIE cells: MKP-1, Bcl-3 and A20. These changes significantly reduced the inflammatory response triggered by TLR4 activation in PIE-APCs co-cultures. The in vivo experiments using castrated male piglets (crossbreeding (LWD) with Landrace (L), Large Yorkshire (W) and Duroc (D))of 3 weeks of age demonstrated that feeding with LjTL2937 significantly reduced blood complement activity and C reactive protein concentrations while no changes were observed in blood leukocytes, ratio of granulocytes to lymphocyte numbers, macrophages’ activity and antibody levels. In addition, treatment with LjTL2937 significantly improved growth performance and productivity, and increased carcass quality. Conclusions We demonstrated that the use of immunobiotics strains like LjTL2937, as supplemental additives for piglets feedings, could be used as a strategy to maintain and improve intestinal homeostasis; that is important for the development of the pig and for health and performance throughout the productive life of the animal.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science Tohoku University, 981-8555 Sendai, Japan.
| |
Collapse
|
37
|
Identification of selective small molecule inhibitors of the nucleotide-binding oligomerization domain 1 (NOD1) signaling pathway. PLoS One 2014; 9:e96737. [PMID: 24806487 PMCID: PMC4013053 DOI: 10.1371/journal.pone.0096737] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 04/10/2014] [Indexed: 12/28/2022] Open
Abstract
NOD1 is an intracellular pattern recognition receptor that recognizes diaminopimelic acid (DAP), a peptidoglycan component in gram negative bacteria. Upon ligand binding, NOD1 assembles with receptor-interacting protein (RIP)-2 kinase and initiates a signaling cascade leading to the production of pro-inflammatory cytokines. Increased NOD1 signaling has been associated with a variety of inflammatory disorders suggesting that small-molecule inhibitors of this signaling complex may have therapeutic utility. We utilized a cell-based screening approach with extensive selectivity profiling to search for small molecule inhibitors of the NOD1 signaling pathway. Via this process we identified three distinct chemical series, xanthines (SB711), quinazolininones (GSK223) and aminobenzothiazoles (GSK966) that selectively inhibited iE-DAP-stimulated IL-8 release via the NOD1 signaling pathway. All three of the newly identified compound series failed to block IL-8 secretion in cells following stimulation with ligands for TNF receptor, TLR2 or NOD2 and, in addition, none of the compound series directly inhibited RIP2 kinase activity. Our initial exploration of the structure-activity relationship and physicochemical properties of the three series directed our focus to the quinazolininone biarylsulfonamides (GSK223). Further investigation allowed for the identification of significantly more potent analogs with the largest boost in activity achieved by fluoro to chloro replacement on the central aryl ring. These results indicate that the NOD1 signaling pathway, similarly to activation of NOD2, is amenable to modulation by small molecules that do not target RIP2 kinase. These compounds should prove useful tools to investigate the importance of NOD1 activation in various inflammatory processes and have potential clinical utility in diseases driven by hyperactive NOD1 signaling.
Collapse
|
38
|
Airway structural cells regulate TLR5-mediated mucosal adjuvant activity. Mucosal Immunol 2014; 7:489-500. [PMID: 24064672 DOI: 10.1038/mi.2013.66] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 07/16/2013] [Accepted: 08/13/2013] [Indexed: 02/04/2023]
Abstract
Antigen-presenting cell (APC) activation is enhanced by vaccine adjuvants. Most vaccines are based on the assumption that adjuvant activity of Toll-like receptor (TLR) agonists depends on direct, functional activation of APCs. Here, we sought to establish whether TLR stimulation in non-hematopoietic cells contributes to flagellin's mucosal adjuvant activity. Nasal administration of flagellin enhanced T-cell-mediated immunity, and systemic and secretory antibody responses to coadministered antigens in a TLR5-dependent manner. Mucosal adjuvant activity was not affected by either abrogation of TLR5 signaling in hematopoietic cells or the presence of flagellin-specific, circulating neutralizing antibodies. We found that flagellin is rapidly degraded in conducting airways, does not translocate into lung parenchyma and stimulates an early immune response, suggesting that TLR5 signaling is regionalized. The flagellin-specific early response of lung was regulated by radioresistant cells expressing TLR5 (particularly the airway epithelial cells). Flagellin stimulated the epithelial production of a small set of mediators that included the chemokine CCL20, which is known to promote APC recruitment in mucosal tissues. Our data suggest that (i) the adjuvant activity of TLR agonists in mucosal vaccination may require TLR stimulation of structural cells and (ii) harnessing the effect of adjuvants on epithelial cells can improve mucosal vaccines.
Collapse
|
39
|
Elliott DE, Siddique SS, Weinstock JV. Innate immunity in disease. Clin Gastroenterol Hepatol 2014; 12:749-55. [PMID: 24632348 PMCID: PMC4083246 DOI: 10.1016/j.cgh.2014.03.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 03/07/2014] [Indexed: 02/07/2023]
Abstract
Cells can innately recognize generic products of viruses, bacteria, fungi, or injured tissue by engagement of pattern recognition receptors. Innate immune cells rapidly respond to this engagement to control commensals, thwart pathogens, and/or prompt repair. Insufficient or excessive activation of the innate immune response results in disease. This review focuses on pattern recognition receptors and cells of the innate immune system that are important for intestinal function. Our improving knowledge pertaining to this important aspect of our immune response is opening potential important new therapeutic opportunities for the treatment of disease.
Collapse
Affiliation(s)
- David E Elliott
- Division of Gastroenterology/Hepatology, Department of Internal Medicine, University of Iowa, Iowa City, Iowa
| | - Sana S Siddique
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Tufts Medical Center, Boston, Massachusetts
| | - Joel V Weinstock
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Tufts Medical Center, Boston, Massachusetts.
| |
Collapse
|
40
|
Bergmann-Leitner ES, Leitner WW. Adjuvants in the Driver's Seat: How Magnitude, Type, Fine Specificity and Longevity of Immune Responses Are Driven by Distinct Classes of Immune Potentiators. Vaccines (Basel) 2014; 2:252-96. [PMID: 26344620 PMCID: PMC4494256 DOI: 10.3390/vaccines2020252] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 03/20/2014] [Accepted: 03/28/2014] [Indexed: 12/16/2022] Open
Abstract
The mechanism by which vaccine adjuvants enhance immune responses has historically been considered to be the creation of an antigen depot. From here, the antigen is slowly released and provided to immune cells over an extended period of time. This "depot" was formed by associating the antigen with substances able to persist at the injection site, such as aluminum salts or emulsions. The identification of Pathogen-Associated Molecular Patterns (PAMPs) has greatly advanced our understanding of how adjuvants work beyond the simple concept of extended antigen release and has accelerated the development of novel adjuvants. This review focuses on the mode of action of different adjuvant classes in regards to the stimulation of specific immune cell subsets, the biasing of immune responses towards cellular or humoral immune response, the ability to mediate epitope spreading and the induction of persistent immunological memory. A better understanding of how particular adjuvants mediate their biological effects will eventually allow them to be selected for specific vaccines in a targeted and rational manner.
Collapse
Affiliation(s)
- Elke S Bergmann-Leitner
- US Military Malaria Research Program, Malaria Vaccine Branch, 503 Robert Grant Ave, 3W65, Silver Spring, MD 20910, USA.
| | - Wolfgang W Leitner
- Division on Allergy, Immunology and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 6610 Rockledge Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
41
|
Villena J, Kitazawa H. Modulation of Intestinal TLR4-Inflammatory Signaling Pathways by Probiotic Microorganisms: Lessons Learned from Lactobacillus jensenii TL2937. Front Immunol 2014; 4:512. [PMID: 24459463 PMCID: PMC3890654 DOI: 10.3389/fimmu.2013.00512] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 12/26/2013] [Indexed: 12/21/2022] Open
Abstract
The intestinal mucosa plays a critical role in the host’s interactions with innocuous commensal microbiota and invading pathogenic microorganisms. Intestinal epithelial cells (IECs) and gut associated immune cells recognize the bacterial components via pattern-recognition receptors (PRRs) and are responsible for maintaining tolerance to the large communities of resident luminal bacteria while being also able to mount inflammatory responses against pathogens. Toll-like receptors (TLRs) are a major class of PRRs that are present on IECs and immune cells which are involved in the induction of both tolerance and inflammation. A growing body of experimental and clinical evidence supports the therapeutic and preventive application of probiotics for several gastrointestinal inflammatory disorders in which TLRs exert a significant role. This review aims to summarize the current knowledge of the beneficial effects of probiotic microorganisms with the capacity to modulate the immune system (immunobiotics) in the regulation of intestinal inflammation in pigs, which are very important as both livestock and human model. Especially we discuss the role of TLRs, their signaling pathways, and their negative regulators in both the inflammatory intestinal injury and the beneficial effects of immunobiotics in general, and Lactobacillus jensenii TL2937 in particular. This review article emphasizes the cellular and molecular interactions of immunobiotics with IECs and immune cells through TLRs and their application for improving animal and human health.
Collapse
Affiliation(s)
- Julio Villena
- Immunobiotics Research Group , Tucuman , Argentina ; Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET) , Tucuman , Argentina
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Department of Science of Food Function and Health, Graduate School of Agricultural Science, Tohoku University , Sendai , Japan
| |
Collapse
|
42
|
Christophers E, Meyer T. Pustular skin diseases refl ect distinct innate defense pathways. ACTA ACUST UNITED AC 2014. [DOI: 10.1586/17469872.3.4.465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
43
|
Ribeiro A, Wörnle M. Viral nucleic acid recognition in human nonimmune cells: in vitro systems. Methods Mol Biol 2014; 1169:67-75. [PMID: 24957230 DOI: 10.1007/978-1-4939-0882-0_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The innate immune system detects viral infections through a variety of receptors that sense the virus via their nucleic acid genome and evokes initial antiviral responses. Nonimmune cells possess pathogen-recognition mechanisms that enable them to respond to virus by the expression of RNA or DNA recognition receptors, thereby initiating the first steps in the host-pathogen interaction and inducing the production of pro-inflammatory and antiviral mediators. To better understand the activation of innate-immune defense mechanisms by nonimmune cells, we describe an experimental procedure that mimics viral infection, using four human nonimmune cell culture models under stimulation with synthetic analogues of RNA and DNA virus. Furthermore, we exemplify two viral infection models using cultured nonimmune cells and hepatitis C virus (HCV) or polyomavirus BK (BKV). In addition, we report the importance of siRNA technique in gene regulation in order to identify specific markers involved in innate antiviral responses in these cells.
Collapse
Affiliation(s)
- Andrea Ribeiro
- Medizinische Klinik und Poliklinik Campus Innenstadt, Klinikum der LMU, Nußbaumstraße 20, 80336, Munich, Germany,
| | | |
Collapse
|
44
|
Nikolova M, Ambrozova G, Kratchanova M, Denev P, Kussovski V, Ciz M, Lojek A. Effects of pectic polysaccharides isolated from leek on the production of reactive oxygen and nitrogen species by phagocytes. J Med Food 2013; 16:711-8. [PMID: 23905651 DOI: 10.1089/jmf.2012.0234] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The current survey investigates the effect of four polysaccharides isolated from fresh leek or alcohol insoluble substances (AIS) of leek on the production of reactive oxygen species (ROS) and reactive nitrogen species (RNS) from phagocytes. The ability of the polysaccharides to activate serum complement was also investigated. Despite the lack of antioxidant activity, the pectic polysaccharides significantly decreased the production of ROS by human neutrophils. Polysaccharides isolated from AIS markedly activated RAW 264.7 macrophages for RNS production in a concentration-dependent manner. The Western blot analysis revealed that this effect was due to the stimulation of the inducible nitric oxide synthase protein expression of macrophages. The polysaccharides extracted from AIS with water showed the ability to fix serum complement, especially through the alternative pathway. It was found that the polysaccharide that has the highest complement-fixing effect is characterized by the highest content of uronic acids and the highest molecular weight.
Collapse
Affiliation(s)
- Mariana Nikolova
- Center of Phytochemistry, Institute of Organic Chemistry, Bulgarian Academy of Sciences, Plovdiv, Bulgaria
| | | | | | | | | | | | | |
Collapse
|
45
|
Hato T, El-Achkar TM, Dagher PC. Sisters in arms: myeloid and tubular epithelial cells shape renal innate immunity. Am J Physiol Renal Physiol 2013; 304:F1243-51. [PMID: 23515715 DOI: 10.1152/ajprenal.00101.2013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The importance of innate immunity for survival is underscored by its presence at almost every level of the evolutionary tree of life. The task of "danger" recognition by the innate immune system is carried out by a broad class of pattern recognition receptors. These receptors are expressed in both hematopoietic and nonhematopoietic cells such as renal epithelial cells. Upon activation, pattern recognition receptors induce essentially two types of defensive responses: inflammation and phagocytosis. In this review, we highlight evidence that renal epithelial cells are endowed with such defensive capabilities and as such fully participate in renal innate immune responses.
Collapse
Affiliation(s)
- Takashi Hato
- Department of Medicine, Indiana University, Indianapolis, IN, USA
| | | | | |
Collapse
|
46
|
Crossing the barrier: Targeting epithelial receptors for enhanced oral vaccine delivery. J Control Release 2012; 160:431-9. [DOI: 10.1016/j.jconrel.2012.02.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 02/02/2012] [Indexed: 01/09/2023]
|
47
|
TLR5 risk-associated haplotype for canine inflammatory bowel disease confers hyper-responsiveness to flagellin. PLoS One 2012; 7:e30117. [PMID: 22279566 PMCID: PMC3261174 DOI: 10.1371/journal.pone.0030117] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 12/09/2011] [Indexed: 02/06/2023] Open
Abstract
Single nucleotide polymorphisms (SNP) in the TLR5 gene have been associated with human inflammatory bowel disease (IBD) and animal models of this disease. We recently demonstrated a significant association between three non-synonymous SNPs in the canine TLR5 gene and IBD in German shepherd dogs (GSDs). However, so far, no direct link between these SNPs and a disturbance in TLR5 function was shown. In the present study, we determined the functional significance of the canine TLR5 SNPs by transfecting the identified risk-protective and risk-associated haplotype into human embryonic kidney cells (HEK) and assessed nuclear factor-kappa B (NF-κB) activation and CXCL8 production after stimulation. In addition, a whole blood assay for TLR5 activation was developed using blood derived from carrier dogs of either haplotype. There was a significant increase in NF-kB activity when cells transfected with the risk-associated TLR5 haplotype were stimulated with flagellin compared to the cells expressing the risk-protective TLR5 haplotype. This difference in NFkB activation correlated with CXCL8 expression in the supernatant measured by ELISA. Furthermore, whole blood taken from carrier dogs of the risk-associated TLR5 haplotype produced significantly more TNF after stimulation with flagellin compared to that taken from carriers of the risk-protective haplotype. Thus, we show for the first time a direct functional impact of the canine IBD risk-associated TLR5 haplotype, which results in hyper-responsiveness to flagellin compared to the IBD risk-protective TLR5 haplotype. Our data potentially suggest that similarly to human IBD and experimental models, TLR5 may also play a role in canine IBD. Blocking the hyper-responsive receptor found in susceptible dogs with IBD may alleviate the inappropriate inflammation seen in this disease.
Collapse
|
48
|
Duerr CU, Hornef MW. The mammalian intestinal epithelium as integral player in the establishment and maintenance of host-microbial homeostasis. Semin Immunol 2011; 24:25-35. [PMID: 22138188 DOI: 10.1016/j.smim.2011.11.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Only one single layer of epithelial cells separates the densely colonized and environmentally exposed intestinal lumen from the largely sterile subepithelial tissue. Together with the overlaying mucus and the subepithelial mucosal immune system the epithelium has evolved to maintain homeostasis in the presence of the enteric microbiota. It also contributes to rapid and efficient antimicrobial host defence in the event of infection with pathogenic microorganisms. Both, epithelial antimicrobial host defence and homeostasis rely on signalling pathways induced by innate immune receptors demonstrating the active role of epithelial cells in the host-microbial interplay. The interaction of epithelial cells with professional immune cells illustrates the integrated function within the mucosal tissue. In the present review we focus on structural and functional changes of the intestinal epithelium during the fetal-neonatal transition and infancy and try to delineate its role in the induction and maintenance of host-microbial homeostasis. We also address factors that impair epithelial functions and may lead to disruption of the mucosal barrier, tissue damage and the development of symptomatic disease.
Collapse
Affiliation(s)
- Claudia U Duerr
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Carl-Neuberg Str. 1, D-30625 Hannover, Germany
| | | |
Collapse
|
49
|
Kathrani A, House A, Catchpole B, Murphy A, Werling D, Allenspach K. Breed-independent toll-like receptor 5 polymorphisms show association with canine inflammatory bowel disease. ACTA ACUST UNITED AC 2011; 78:94-101. [DOI: 10.1111/j.1399-0039.2011.01707.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
50
|
Devriendt B, De Geest BG, Cox E. Designing oral vaccines targeting intestinal dendritic cells. Expert Opin Drug Deliv 2011; 8:467-83. [DOI: 10.1517/17425247.2011.561312] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|