1
|
Huang J, Li L, Xu L, Feng L, Wang Y, SIK AG, Jin M, Wang R, Liu K, Li X. Methyl 3-Bromo-4,5-dihydroxybenzoate Attenuates Inflammatory Bowel Disease by Regulating TLR/NF-κB Pathways. Mar Drugs 2025; 23:47. [PMID: 39852549 PMCID: PMC11766471 DOI: 10.3390/md23010047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/11/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by uncontrolled, chronic relapsing inflammation in the gastrointestinal tract and has become a global healthcare problem. Here, we aimed to illustrate the anti-inflammatory activity and the underlying mechanism of methyl 3-bromo-4,5-dihydroxybenzoate (MBD), a compound derived from marine organisms, especially in IBD, using a zebrafish model. The results indicated that MBD could inhibit the inflammatory responses induced by CuSO4, tail amputation and LPS in zebrafish. Furthermore, MBD notably inhibited the intestinal migration of immune cells, enhanced the integrity of the gut mucosal barrier and improved intestinal peristalsis function in a zebrafish IBD model induced by trinitro-benzene-sulfonic acid (TNBS). In addition, MBD could inhibit ROS elevation induced by TNBS. Network pharmacology analysis, molecular docking, transcriptomics sequencing and RT-PCR were conducted to investigate the potential mechanism. The results showed that MBD could regulate the TLR/NF-κB pathways by inhibiting the mRNA expression of TNF-α, NF-κB, IL-1, IL-1β, IL6, AP1, IFNγ, IKKβ, MyD88, STAT3, TRAF1, TRAF6, NLRP3, NOD2, TLR3 and TLR4, and promoting the mRNA expression of IL4, IκBα and Bcl-2. In conclusion, these findings indicate that MBD could be a potential candidate for the treatment of IBD.
Collapse
Affiliation(s)
- Jing Huang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (J.H.); (L.L.); (L.X.); (L.F.); (Y.W.); (A.G.S.); (M.J.)
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan 250103, China
| | - Lei Li
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (J.H.); (L.L.); (L.X.); (L.F.); (Y.W.); (A.G.S.); (M.J.)
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan 250103, China
| | - Liyan Xu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (J.H.); (L.L.); (L.X.); (L.F.); (Y.W.); (A.G.S.); (M.J.)
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan 250103, China
| | - Lixin Feng
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (J.H.); (L.L.); (L.X.); (L.F.); (Y.W.); (A.G.S.); (M.J.)
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan 250103, China
| | - Yuxin Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (J.H.); (L.L.); (L.X.); (L.F.); (Y.W.); (A.G.S.); (M.J.)
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan 250103, China
| | - Attila Gabor SIK
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (J.H.); (L.L.); (L.X.); (L.F.); (Y.W.); (A.G.S.); (M.J.)
- Institute of Physiology, Medical School, University of Pecs, H-7624 Pecs, Hungary
- University Research and Innovation Center, Obuda University, H-1034 Budapest, Hungary
| | - Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (J.H.); (L.L.); (L.X.); (L.F.); (Y.W.); (A.G.S.); (M.J.)
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan 250103, China
| | - Rongchun Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (J.H.); (L.L.); (L.X.); (L.F.); (Y.W.); (A.G.S.); (M.J.)
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan 250103, China
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (J.H.); (L.L.); (L.X.); (L.F.); (Y.W.); (A.G.S.); (M.J.)
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan 250103, China
| | - Xiaobin Li
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (J.H.); (L.L.); (L.X.); (L.F.); (Y.W.); (A.G.S.); (M.J.)
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan 250103, China
| |
Collapse
|
2
|
Shao F, Wang Z, Ye L, Wu R, Wang J, Yu QX, Wusiman D, Tuo Z, Yoo KH, Shu Z, Wei W, Li D, Cho WC, Liu Z, Feng D. Basic helix-loop-helix ARNT like 1 regulates the function of immune cells and participates in the development of immune-related diseases. BURNS & TRAUMA 2025; 13:tkae075. [PMID: 39830193 PMCID: PMC11741524 DOI: 10.1093/burnst/tkae075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/26/2024] [Accepted: 11/01/2024] [Indexed: 01/22/2025]
Abstract
The circadian clock is an internal timekeeper system that regulates biological processes through a central circadian clock and peripheral clocks controlling various genes. Basic helix-loop-helix ARNT-like 1 (BMAL1), also known as aryl hydrocarbon receptor nuclear translocator-like protein 1 (ARNTL1), is a key component of the circadian clock. The deletion of BMAL1 alone can abolish the circadian rhythms of the human body. BMAL1 plays a critical role in immune cell function. Dysregulation of BMAL1 is linked to immune-related diseases such as autoimmune diseases, infectious diseases, and cancer, and vice versa. This review highlights the significant role of BMAL1 in governing immune cells, including their development, differentiation, migration, homing, metabolism, and effector functions. This study also explores how dysregulation of BMAL1 can have far-reaching implications and potentially contribute to the onset of immune-related diseases such as autoimmune diseases, infectious diseases, cancer, sepsis, and trauma. Furthermore, this review discusses treatments for immune-related diseases that target BMAL1 disorders. Understanding the impact of BMAL1 on immune function can provide insights into the pathogenesis of immune-related diseases and help in the development of more effective treatment strategies. Targeting BMAL1 has been demonstrated to achieve good efficacy in immune-related diseases, indicating its promising potential as a targetable therapeutic target in these diseases.
Collapse
Affiliation(s)
- Fanglin Shao
- Chengdu Basebio Company, Tianfu Third Street, High-Tech Zone, Chengdu 610041, China
| | - Zhipeng Wang
- Department of Urology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, First Ring Road, Qingyang District, Chengdu 610041, China
| | - Luxia Ye
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, West Gate Street, Linhai City 317000, Zhejiang Province, China
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Renmin South Road, Wuhou District, Chengdu 610041, China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Renmin South Road, Wuhou District, Chengdu 610041, China
| | - Qing-Xin Yu
- Department of Pathology, Ningbo Clinical Pathology Diagnosis Center, Huancheng North Road, Jiangbei District, Ningbo, Zhejiang Province, 315211, China
| | - Dilinaer Wusiman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN 47907, USA
- Purdue Institute for Cancer Research, Purdue University, 615 W. State Street, West Lafayette, IN 47907, USA
| | - Zhouting Tuo
- Chengdu Basebio Company, Tianfu Third Street, High-Tech Zone, Chengdu 610041, China
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Jinzhai South Road, Shushan District, Hefei, Anhui 230032, China
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, 26 Kyungheedae-Ro, Dongdaemun-Gu, South Korea
| | - Ziyu Shu
- Department of Earth Science and Engineering, Imperial College London, Exhibition Road, South Kensington, London SW7 2AZ, UK
- Joint International Research Laboratory of Green Buildings and Built Environments (Ministry of Education), Chongqing University, Shapingba Street, Shapingba District, Chongqing 400044, China
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Renmin South Road, Wuhou District, Chengdu 610041, China
| | - Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Renmin South Road, Wuhou District, Chengdu 610041, China
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Gascoigne Road, Yau Ma Tei, Kowloon, Hong Kong SAR, China
| | - Zhihong Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Renmin South Road, Wuhou District, Chengdu 610041, China
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Renmin South Road, Wuhou District, Chengdu 610041, China
- Division of Surgery & Interventional Science, University College London, Gower Street, London W1T 6JF, London W1W 7TS, UK
| |
Collapse
|
3
|
Ghorab RA, Fouad SH, Sherief AF, El-Sehsah EM, Shamloul S, Taha SI. MiR-146a (rs2910164) Gene Polymorphism and Its Impact on Circulating MiR-146a Levels in Patients with Inflammatory Bowel Diseases. Inflammation 2024:10.1007/s10753-024-02108-0. [PMID: 39103590 DOI: 10.1007/s10753-024-02108-0] [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: 06/22/2024] [Revised: 07/08/2024] [Accepted: 07/16/2024] [Indexed: 08/07/2024]
Abstract
MicroRNA-146a (miR-146a) has been involved in the pathophysiology of inflammatory bowel disease (IBD). However, the precise processes are still not entirely understood. Contradictory studies suggest that miR-146a expression could be influenced by the miR-146a rs2910164 C > G polymorphism. This case-control study aimed to investigate the association of miR-146a rs2910164 C > G gene polymorphism and its impact on circulating miR-146a expression levels in Egyptian IBD patients. We included 40 IBD patients and 30 matched healthy controls. Genotyping of miR-146a rs2910164 polymorphism and assessment of miR-146a expression level were done using quantitative real-time PCR in all participants. MiR-146a rs2910164 GG genotype and the G allele were reported in 47% and 70% of the IBD patient group, respectively. And they were associated with increased IBD risk. All the IBD patients with the CC genotype (100%) and most of those with the CG genotype (66.67%) had an inactive disease, while most IBD patients with the GG genotype (73.68%) had an active disease. The miR-146a expression level was the highest with the CC genotype and the lowest with the GG genotype. Also, miR-146a expression level decreased significantly in IBD patients than controls and with disease activity. Combined detection of fecal calprotectin with miR-146a expression level improved the diagnostic sensitivity and the negative predictive value in differentiating IBD patients with active disease from those inactive. Our study identified a strong association of miR-146a rs2910164 GG genotype and G allele with IBD-increased susceptibility and activity in the Egyptian population. The miR-146a rs2910164 polymorphism can reduce miR-146a expression levels in these patients as well. Further research on a larger sample size and different ethnic populations can be the key to progress in establishing this genetic association.
Collapse
Affiliation(s)
- Rasha Ahmed Ghorab
- Department of Clinical Pathology, Faculty of Medicine, Ain-Shams University, 11591 Abbasia, Cairo, Egypt
| | - Shaimaa H Fouad
- Department of Internal Medicine /Allergy and Clinical Immunology, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Ahmed F Sherief
- Department of Tropical Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Eman M El-Sehsah
- Department of Medical Microbiology and Immunology, Mansoura Faculty of Medicine, Mansoura, Egypt
| | - Sara Shamloul
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sara I Taha
- Department of Clinical Pathology, Faculty of Medicine, Ain-Shams University, 11591 Abbasia, Cairo, Egypt.
| |
Collapse
|
4
|
Zhang J, Yao Z. Immune cell trafficking: a novel perspective on the gut-skin axis. Inflamm Regen 2024; 44:21. [PMID: 38654394 DOI: 10.1186/s41232-024-00334-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024] Open
Abstract
Immune cell trafficking, an essential mechanism for maintaining immunological homeostasis and mounting effective responses to infections, operates under a stringent regulatory framework. Recent advances have shed light on the perturbation of cell migration patterns, highlighting how such disturbances can propagate inflammatory diseases from their origin to distal organs. This review collates and discusses current evidence that demonstrates atypical communication between the gut and skin, which are conventionally viewed as distinct immunological spheres, in the milieu of inflammation. We focus on the aberrant, reciprocal translocation of immune cells along the gut-skin axis as a pivotal factor linking intestinal and dermatological inflammatory conditions. Recognizing that the translation of these findings into clinical practices is nascent, we suggest that therapeutic strategies aimed at modulating the axis may offer substantial benefits in mitigating the widespread impact of inflammatory diseases.
Collapse
Affiliation(s)
- Jiayan Zhang
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhirong Yao
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Eladham MW, Selvakumar B, Saheb Sharif-Askari N, Saheb Sharif-Askari F, Ibrahim SM, Halwani R. Unraveling the gut-Lung axis: Exploring complex mechanisms in disease interplay. Heliyon 2024; 10:e24032. [PMID: 38268584 PMCID: PMC10806295 DOI: 10.1016/j.heliyon.2024.e24032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 12/18/2023] [Accepted: 01/02/2024] [Indexed: 01/26/2024] Open
Abstract
The link between gut and lung starts as early as during organogenesis. Even though they are anatomically distinct, essential bidirectional crosstalk via complex mechanisms supports GLA. Emerging studies have demonstrated the association of gut and lung diseases via multifaceted mechanisms. Advancements in omics and metagenomics technologies revealed a potential link between gut and lung microbiota, adding further complexity to GLA. Despite substantial studies on GLA in various disease models, mechanisms beyond microbial dysbiosis regulating the interplay between gut and lung tissues during disease conditions are not thoroughly reviewed. This review outlines disease specific GLA mechanisms, emphasizing research gaps with a focus on gut-to-lung direction based on current GLA literature. Moreover, the review discusses potential gut microbiota and their products like metabolites, immune modulators, and non-bacterial contributions as a basis for developing treatment strategies for lung diseases. Advanced experimental methods, modern diagnostic tools, and technological advancements are also highlighted as crucial areas for improvement in developing novel therapeutic approaches for GLA-related diseases. In conclusion, this review underscores the importance of exploring additional mechanisms within the GLA to gain a deeper understanding that could aid in preventing and treating a wide spectrum of lung diseases.
Collapse
Affiliation(s)
- Mariam Wed Eladham
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Balachandar Selvakumar
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Narjes Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Fatemeh Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Pharmacy Practice and Pharmaceutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | | | - Rabih Halwani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Prince Abdullah Ben Khaled Celiac Disease Research Chair, Department of Pediatrics, Faculty of Medicine, King Saud University, Saudi Arabia
| |
Collapse
|
6
|
Differential Homing Receptor Profiles of Lymphocytes Induced by Attenuated versus Live Plasmodium falciparum Sporozoites. Vaccines (Basel) 2022; 10:vaccines10101768. [PMID: 36298634 PMCID: PMC9611729 DOI: 10.3390/vaccines10101768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/02/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022] Open
Abstract
The onset of an adaptive immune response provides the signals required for differentiation of antigen-specific lymphocytes into effector cells and imprinting of these cells for re-circulation to the most appropriate anatomical site (i.e., homing). Lymphocyte homing is governed by the expression of tissue-specific lymphocyte homing receptors that bind to unique tissue-specific ligands on endothelial cells. In this study, a whole-parasite malaria vaccine (radiation-attenuated sporozoites (RAS)) was used as a model system to establish homing receptor signatures induced by the parasite delivered through mosquito bite to provide a benchmark of desirable homing receptors for malaria vaccine developers. This immunization regimen resulted in the priming of antigen-specific B cells and CD8+ T cells for homing primarily to the skin and T/B cell compartments of secondary lymphoid organs. Infection with live sporozoites, however, triggers the upregulation of homing receptor for the liver and the skin, demonstrating that there is a difference in the signal provided by attenuated vs. live sporozoites. This is the first report on imprinting of homing routes by Plasmodium sporozoites and, surprisingly, it also points to additional, yet to be identified, signals provided by live parasites that prime lymphocytes for homing to the liver. The data also demonstrate the utility of this method for assessing the potential of vaccine formulations to direct antigen-specific lymphocytes to the most relevant anatomical site, thus potentially impacting vaccine efficacy.
Collapse
|
7
|
Shigehiro T, Ueno M, Kijihira M, Takahashi R, Umemura C, Taha EA, Kurosaka C, Asayama M, Murakami H, Satoh A, Nakamura Y, Futami J, Masuda J. Immune State Conversion of the Mesenteric Lymph Node in a Mouse Breast Cancer Model. Int J Mol Sci 2022; 23:ijms231911035. [PMID: 36232335 PMCID: PMC9570492 DOI: 10.3390/ijms231911035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/17/2022] [Accepted: 09/18/2022] [Indexed: 11/16/2022] Open
Abstract
Secondary lymphoid tissues, such as the spleen and lymph nodes (LNs), contribute to breast cancer development and metastasis in both anti- and pro-tumoral directions. Although secondary lymphoid tissues have been extensively studied, very little is known about the immune conversion in mesenteric LNs (mLNs) during breast cancer development. Here, we demonstrate inflammatory immune conversion of mLNs in a metastatic 4T1 breast cancer model. Splenic T cells were significantly decreased and continuously suppressed IFN-γ production during tumor development, while myeloid-derived suppressor cells (MDSCs) were dramatically enriched. However, T cell numbers in the mLN did not decrease, and the MDSCs only moderately increased. T cells in the mLN exhibited conversion from a pro-inflammatory state with high IFN-γ expression to an anti-inflammatory state with high expression of IL-4 and IL-10 in early- to late-stages of breast cancer development. Interestingly, increased migration of CD103+CD11b+ dendritic cells (DCs) into the mLN, along with increased (1→3)-β-D-glucan levels in serum, was observed even in late-stage breast cancer. This suggests that CD103+CD11b+ DCs could prime cancer-reactive T cells. Together, the data indicate that the mLN is an important lymphoid tissue contributing to breast cancer development.
Collapse
Affiliation(s)
- Tsukasa Shigehiro
- Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Correspondence: (T.S.); (J.M.); Tel.: +81-47-121-4060 (T.S.); +81-86-251-8003 (J.M.)
| | - Maho Ueno
- Department of Applied Chemistry and Biotechnology, Faculty of Engineering, Okayama University, Okayama 700-8530, Japan
| | - Mayumi Kijihira
- Department of Applied Chemistry and Biotechnology, Faculty of Engineering, Okayama University, Okayama 700-8530, Japan
| | - Ryotaro Takahashi
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
| | - Chiho Umemura
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Eman A. Taha
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Chisaki Kurosaka
- Department of Applied Chemistry and Biotechnology, Faculty of Engineering, Okayama University, Okayama 700-8530, Japan
| | - Megumi Asayama
- Department of Applied Chemistry and Biotechnology, Faculty of Engineering, Okayama University, Okayama 700-8530, Japan
| | - Hiroshi Murakami
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Ayano Satoh
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Yoshimasa Nakamura
- Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan
| | - Junichiro Futami
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Junko Masuda
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
- Department of Pharmacology, Tokyo Women’s Medical University, Shinjuku, Tokyo 162-8666, Japan
- Correspondence: (T.S.); (J.M.); Tel.: +81-47-121-4060 (T.S.); +81-86-251-8003 (J.M.)
| |
Collapse
|
8
|
Dyavar SR, Singh R, Emani R, Pawar GP, Chaudhari VD, Podany AT, Avedissian SN, Fletcher CV, Salunke DB. Role of toll-like receptor 7/8 pathways in regulation of interferon response and inflammatory mediators during SARS-CoV2 infection and potential therapeutic options. Biomed Pharmacother 2021; 141:111794. [PMID: 34153851 PMCID: PMC8189763 DOI: 10.1016/j.biopha.2021.111794] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/27/2021] [Accepted: 05/29/2021] [Indexed: 12/17/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV2) is the causative agent of Corona Virus Disease 2019 (COVID-19). Lower production of type I and III interferons and higher levels of inflammatory mediators upon SARS-CoV2 infection contribute to COVID-19 pathogenesis. Optimal interferon production and controlled inflammation are essential to limit COVID-19 pathogenesis. However, the aggravated inflammatory response observed in COVID-19 patients causes severe damage to the host and frequently advances to acute respiratory distress syndrome (ARDS). Toll-like receptor 7 and 8 (TLR7/8) signaling pathways play a central role in regulating induction of interferons (IFNs) and inflammatory mediators in dendritic cells. Controlled inflammation is possible through regulation of TLR mediated response without influencing interferon production to reduce COVID-19 pathogenesis. This review focuses on inflammatory mediators that contribute to pathogenic effects and the role of TLR pathways in the induction of interferon and inflammatory mediators and their contribution to COVID-19 pathogenesis. We conclude that potential TLR7/8 agonists inducing antiviral interferon response and controlling inflammation are important therapeutic options to effectively eliminate SARS-CoV2 induced pathogenesis. Ongoing and future studies may provide additional evidence on their safety and efficacy to treat COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Shetty Ravi Dyavar
- University of Nebraska Medical Center (UNMC) Center for Drug Discovery, UNMC, Omaha, NE 68198, USA.
| | - Rahul Singh
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India
| | - Rohini Emani
- Buck Institute for Research on Ageing, Novato, CA, USA
| | - Ganesh P Pawar
- Division of Medicinal Chemistry, CSIR-Institute of Microbiology Technology Chandigarh, Sector-39A, Chandigarh,160036, India
| | - Vinod D Chaudhari
- Division of Medicinal Chemistry, CSIR-Institute of Microbiology Technology Chandigarh, Sector-39A, Chandigarh,160036, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Anthony T Podany
- University of Nebraska Medical Center (UNMC) Center for Drug Discovery, UNMC, Omaha, NE 68198, USA
| | - Sean N Avedissian
- University of Nebraska Medical Center (UNMC) Center for Drug Discovery, UNMC, Omaha, NE 68198, USA
| | - Courtney V Fletcher
- University of Nebraska Medical Center (UNMC) Center for Drug Discovery, UNMC, Omaha, NE 68198, USA
| | - Deepak B Salunke
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India; National Interdisciplinary Centre of Vaccine, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
9
|
Hinrichs AC, Blokland SLM, Lopes AP, Wichers CGK, Kruize AA, Pandit A, Radstake TRDJ, van Roon JAG. Transcriptome Analysis of CCR9+ T Helper Cells From Primary Sjögren's Syndrome Patients Identifies CCL5 as a Novel Effector Molecule. Front Immunol 2021; 12:702733. [PMID: 34386009 PMCID: PMC8354142 DOI: 10.3389/fimmu.2021.702733] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/09/2021] [Indexed: 12/27/2022] Open
Abstract
Introduction CCR9+ Tfh-like pathogenic T helper (Th) cells are elevated in patients with primary Sjögren’s syndrome (pSS) and indicated to play a role in pSS immunopathology. Here we delineate the CCR9+ Th cell-specific transcriptome to study the molecular dysregulation of these cells in pSS patients. Methods CCR9+, CXCR5+ and CCR9-CXCR5- Th cells from blood of 7 healthy controls (HC) and 7 pSS patients were FACS sorted and RNA sequencing was performed. Computational analysis was used to identify differentially expressed genes (DEGs), coherent gene expression networks and differentially regulated pathways. Target genes were replicated in additional cohorts. Results 5131 genes were differentially expressed between CCR9+ and CXCR5+ Th cells; 6493 and 4783 between CCR9+ and CCR9-CXCR5- and between CXCR5+ and CCR9-CXCR5-, respectively. In the CCR9+ Th cell subset 2777 DEGs were identified between HC and pSS patients, 1416 and 1077 in the CXCR5+ and CCR9-CXCR5- subsets, respectively. One gene network was selected based on eigengene expression differences between the Th cell subsets and pathways enriched for genes involved in migration and adhesion, cytokine and chemokine production. Selected DEGs of interest (HOPX, SOX4, ITGAE, ITGA1, NCR3, ABCB1, C3AR1, NT5E, CCR5 and CCL5) from this module were validated and found upregulated in blood CCR9+ Th cells, but were similarly expressed in HC and pSS patients. Increased frequencies of CCR9+ Th cells were shown to express higher levels of CCL5 than CXCR5+ and CCR9-CXCR5- Th cells, with the highest expression confined to effector CCR9+ Th cells. Antigenic triggering and stimulation with IL-7 of the Th cell subsets co-cultured with monocytes strongly induced CCL5 secretion in CCR9+ Th cell cocultures. Additionally, effector CCR9+ Th cells rapidly released CCL5 and secreted the highest CCL5 levels upon stimulation. Conclusion Transcriptomic analysis of circulating CCR9+ Th cells reveals CCR9-specific pathways involved in effector T cell function equally expressed in pSS patients and HC. Given the increased numbers of CCR9+ Th cells in the blood and inflamed glands of pSS patients and presence of inflammatory stimuli to activate these cells this suggests that CCR9-specific functions, such as cell recruitment upon CCL5 secretion, could significantly contribute to immunopathology in pSS.
Collapse
Affiliation(s)
- Anneline C Hinrichs
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Sofie L M Blokland
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Ana P Lopes
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Catharina G K Wichers
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Aike A Kruize
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Aridaman Pandit
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Timothy R D J Radstake
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Joel A G van Roon
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
10
|
Avery EG, Bartolomaeus H, Maifeld A, Marko L, Wiig H, Wilck N, Rosshart SP, Forslund SK, Müller DN. The Gut Microbiome in Hypertension: Recent Advances and Future Perspectives. Circ Res 2021; 128:934-950. [PMID: 33793332 DOI: 10.1161/circresaha.121.318065] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The pathogenesis of hypertension is known to involve a diverse range of contributing factors including genetic, environmental, hormonal, hemodynamic and inflammatory forces, to name a few. There is mounting evidence to suggest that the gut microbiome plays an important role in the development and pathogenesis of hypertension. The gastrointestinal tract, which houses the largest compartment of immune cells in the body, represents the intersection of the environment and the host. Accordingly, lifestyle factors shape and are modulated by the microbiome, modifying the risk for hypertensive disease. One well-studied example is the consumption of dietary fibers, which leads to the production of short-chain fatty acids and can contribute to the expansion of anti-inflammatory immune cells, consequently protecting against the progression of hypertension. Dietary interventions such as fasting have also been shown to impact hypertension via the microbiome. Studying the microbiome in hypertensive disease presents a variety of unique challenges to the use of traditional model systems. Integrating microbiome considerations into preclinical research is crucial, and novel strategies to account for reciprocal host-microbiome interactions, such as the wildling mouse model, may provide new opportunities for translation. The intricacies of the role of the microbiome in hypertensive disease is a matter of ongoing research, and there are several technical considerations which should be accounted for moving forward. In this review we provide insights into the host-microbiome interaction and summarize the evidence of its importance in the regulation of blood pressure. Additionally, we provide recommendations for ongoing and future research, such that important insights from the microbiome field at large can be readily integrated in the context of hypertension.
Collapse
Affiliation(s)
- Ellen G Avery
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Berlin, Germany (E.G.A.,H.B.,A.M.,L.M.,N.W.,S.K.F.,D.N.M.).,For Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (E.G.A.,H.B., N.W., S.K.F., D.N.M.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (E.G.A., H.B., A.M., L.M., N.W., S.K.F., D.N.M.).,Freie Universität Berlin, Department of Biology, Chemistry, Pharmacy, Berlin, Germany (E.G.A.)
| | - Hendrik Bartolomaeus
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Berlin, Germany (E.G.A.,H.B.,A.M.,L.M.,N.W.,S.K.F.,D.N.M.).,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany (H.B., A.M., L.M., N.W., S.K.F., D.N.M.).,For Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (E.G.A.,H.B., N.W., S.K.F., D.N.M.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (E.G.A., H.B., A.M., L.M., N.W., S.K.F., D.N.M.)
| | - Andras Maifeld
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Berlin, Germany (E.G.A.,H.B.,A.M.,L.M.,N.W.,S.K.F.,D.N.M.).,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany (H.B., A.M., L.M., N.W., S.K.F., D.N.M.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (E.G.A., H.B., A.M., L.M., N.W., S.K.F., D.N.M.)
| | - Lajos Marko
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Berlin, Germany (E.G.A.,H.B.,A.M.,L.M.,N.W.,S.K.F.,D.N.M.).,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany (H.B., A.M., L.M., N.W., S.K.F., D.N.M.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (E.G.A., H.B., A.M., L.M., N.W., S.K.F., D.N.M.)
| | - Helge Wiig
- Department of Biomedicine, University of Bergen, Norway (H.W.)
| | - Nicola Wilck
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Berlin, Germany (E.G.A.,H.B.,A.M.,L.M.,N.W.,S.K.F.,D.N.M.).,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany (H.B., A.M., L.M., N.W., S.K.F., D.N.M.).,For Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (E.G.A.,H.B., N.W., S.K.F., D.N.M.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (E.G.A., H.B., A.M., L.M., N.W., S.K.F., D.N.M.).,For Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany (N.W.)
| | - Stephan P Rosshart
- Medical Center-University of Freiburg, Department of Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, Freiburg, Germany (S.P.R.)
| | - Sofia K Forslund
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Berlin, Germany (E.G.A.,H.B.,A.M.,L.M.,N.W.,S.K.F.,D.N.M.).,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany (H.B., A.M., L.M., N.W., S.K.F., D.N.M.).,For Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (E.G.A.,H.B., N.W., S.K.F., D.N.M.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (E.G.A., H.B., A.M., L.M., N.W., S.K.F., D.N.M.)
| | - Dominik N Müller
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Berlin, Germany (E.G.A.,H.B.,A.M.,L.M.,N.W.,S.K.F.,D.N.M.).,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany (H.B., A.M., L.M., N.W., S.K.F., D.N.M.).,For Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (E.G.A.,H.B., N.W., S.K.F., D.N.M.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (E.G.A., H.B., A.M., L.M., N.W., S.K.F., D.N.M.)
| |
Collapse
|
11
|
van Hoffen E, Mercenier A, Vidal K, Benyacoub J, Schloesser J, Kardinaal A, Lucas-van de Bos E, van Alen I, Roggero I, Duintjer K, Berendts A, Albers R, Kleerebezem M, Ten Bruggencate S. Characterization of the pathophysiological determinants of diarrheagenic Escherichia coli infection using a challenge model in healthy adults. Sci Rep 2021; 11:6060. [PMID: 33723346 PMCID: PMC7960709 DOI: 10.1038/s41598-021-85161-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/15/2021] [Indexed: 11/09/2022] Open
Abstract
An experimental human challenge model with an attenuated diarrheagenic Escherichia coli (E. coli) strain has been used in food intervention studies aimed to increase resistance to E. coli infection. This study was designed to refine and expand this challenge model. In a double-blind study, healthy male subjects were orally challenged with 1E10 or 5E10 colony-forming units (CFU) of E. coli strain E1392/75-2A. Three weeks later, subjects were rechallenged with 1E10 CFU of E. coli. Before and after both challenges, clinical symptoms and infection- and immune-related biomarkers were analyzed. Subset analysis was performed on clinically high- and low-responders. Regardless of inoculation dose, the first challenge induced clinical symptoms for 2-3 days. In blood, neutrophils, CRP, CXCL10, and CFA/II-specific IgG were induced, and in feces calprotectin and CFA/II-specific IgA. Despite clinical differences between high- and low-responders, infection and immune biomarkers did not differ. The first inoculation induced protection at the second challenge, with a minor clinical response, and no change in biomarkers. The refined study design resulted in a larger dynamic range of symptoms, and identification of biomarkers induced by a challenge with the attenuated E. coli strain E1392/75-2A, which is of value for future intervention studies. Addition of a second inoculation allows to study the protective response induced by a primary infection.Clinicaltrials.gov registration: NCT02541695 (04/09/2015).
Collapse
Affiliation(s)
- Els van Hoffen
- Department of Nutrition and Health, NIZO, PO Box 20, 6710 BA, Ede, the Netherlands
| | - Annick Mercenier
- Nestlé Institute of Health Sciences, Gastrointestinal Health, Nestlé Research, Lausanne, Switzerland.,NutriLeads, Wageningen, The Netherlands
| | - Karine Vidal
- Nestlé Institute of Health Sciences, Gastrointestinal Health, Nestlé Research, Lausanne, Switzerland
| | - Jalil Benyacoub
- Nestlé Institute of Health Sciences, Gastrointestinal Health, Nestlé Research, Lausanne, Switzerland
| | - Joyce Schloesser
- Department of Nutrition and Health, NIZO, PO Box 20, 6710 BA, Ede, the Netherlands.
| | - Alwine Kardinaal
- Department of Nutrition and Health, NIZO, PO Box 20, 6710 BA, Ede, the Netherlands
| | | | - Ingrid van Alen
- Department of Nutrition and Health, NIZO, PO Box 20, 6710 BA, Ede, the Netherlands
| | - Iris Roggero
- Nestlé Institute of Health Sciences, Gastrointestinal Health, Nestlé Research, Lausanne, Switzerland
| | - Kim Duintjer
- Department of Nutrition and Health, NIZO, PO Box 20, 6710 BA, Ede, the Netherlands.,Host Microbe Interactomics Group, Department of Animal Sciences, Wageningen University and Research, Wageningen, the Netherlands
| | - Anneke Berendts
- Department of Nutrition and Health, NIZO, PO Box 20, 6710 BA, Ede, the Netherlands
| | | | - Michiel Kleerebezem
- Host Microbe Interactomics Group, Department of Animal Sciences, Wageningen University and Research, Wageningen, the Netherlands
| | | |
Collapse
|
12
|
Adapter Chimeric Antigen Receptor (AdCAR)-Engineered NK-92 Cells for the Multiplex Targeting of Bone Metastases. Cancers (Basel) 2021; 13:cancers13051124. [PMID: 33807875 PMCID: PMC7961358 DOI: 10.3390/cancers13051124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/15/2021] [Accepted: 03/04/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Metastatic disease remains one of the biggest challenges for tumor therapy. The aim of our study was the preclinical evaluation of adapter chimeric antigen receptor (AdCAR)-engineered NK-92 cell efficacy as a possible treatment strategy for various types of bone metastatic cancers. We confirmed that AdCAR NK-92 cells successfully induces tumor cell lysis in bone metastasis cell lines derived from mammary, renal cell and colorectal carcinoma as well as melanoma in a specific and controllable manner, thus, establishing a potent cellular product with universal applicability and quick clinical translation potential for the treatment of solid tumors, including metastases. Abstract Background: Since metastatic spreading of solid tumor cells often leads to a fatal outcome for most cancer patients, new approaches for patient-individualized, targeted immunotherapy are urgently needed. Methods: Here, we established cell lines from four bone metastases of different tumor entities. We assessed AdCAR NK-92-mediated cytotoxicity in vitro in standard cytotoxicity assays as well as 3D spheroid models Results: AdCAR-engineered NK-92 cells successfully demonstrated distinct and specific cytotoxic potential targeting different tumor antigens expressed on cell lines established from bone metastases of mammary, renal cell and colorectal carcinoma as well as melanomas. In that process AdCAR NK-92 cells produced a multitude of NK effector molecules as well as pro inflammatory cytokines. Furthermore, AdCAR NK-92 showed increased cytotoxicity in 3D spheroid models which can recapitulate in vivo architecture, thereby bridging the gap between in vitro and in vivo models. Conclusions: AdCAR NK-92 cells may provide an interesting and promising “off-the-shelf” cellular product for the targeted therapy of cancers metastasizing to the bone, while utilization of clinically approved, therapeutic antibodies, as exchangeable adapter molecules can facilitate quick clinical translation.
Collapse
|
13
|
Ortega Moreno L, Fernández-Tomé S, Chaparro M, Marin AC, Mora-Gutiérrez I, Santander C, Baldan-Martin M, Gisbert JP, Bernardo D. Profiling of Human Circulating Dendritic Cells and Monocyte Subsets Discriminates Between Type and Mucosal Status in Patients With Inflammatory Bowel Disease. Inflamm Bowel Dis 2021; 27:268-274. [PMID: 32548643 DOI: 10.1093/ibd/izaa151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Intestinal dendritic cells (DC) and macrophages drive disease progression in patients with inflammatory bowel disease (IBD). We aimed to characterize the activation and homing profile of human circulating DC and monocyte subsets in healthy control patients (CP) and IBD patients. METHODS Eighteen CP and 64 patients with IBD were categorized by diagnoses of Crohn disease (CD) and ulcerative colitis (UC), either endoscopically active (inflamed) or quiescent. Circulating type 1 conventional DC, type 2 conventional DC, plasmacytoid DC, classical monocytes, nonclassical monocytes, and intermediate monocytes were identified by flow cytometry in each individual and characterized for the expression of 18 markers. Association between DC/monocytes and IBD risk was tested by logistic regression. Discriminant canonical analyses were performed to classify the patients in their own endoscopy category considering all markers on each subset. RESULTS CCRL1, CCR3, and CCR5 expression on circulating type 1 DC; CCRL1 expression on nonclassical monocytes; and CCR9 and β7 expression on classical monocytes allowed us to discriminate among the different study groups. Indeed, the same markers (excluding β7) were also associated with IBD when all DC and monocyte subsets were considered at the same time. CONCLUSIONS Monitoring the phenotype of human circulating DC and monocyte subsets may provide novel tools as biomarkers for disease diagnosis (CD/UC) or mucosal status (inflamed/noninflamed) in the absence of an invasive colonoscopy.
Collapse
Affiliation(s)
- Lorena Ortega Moreno
- Servicio de Aparato Digestivo, Hospital Universitario de La Princesa e Instituto de Investigación Sanitaria Princesa & Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Madrid, Spain.,Departamento de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Samuel Fernández-Tomé
- Servicio de Aparato Digestivo, Hospital Universitario de La Princesa e Instituto de Investigación Sanitaria Princesa & Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Madrid, Spain
| | - María Chaparro
- Servicio de Aparato Digestivo, Hospital Universitario de La Princesa e Instituto de Investigación Sanitaria Princesa & Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Madrid, Spain.,Departamento de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Alicia C Marin
- Servicio de Aparato Digestivo, Hospital Universitario de La Princesa e Instituto de Investigación Sanitaria Princesa & Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Madrid, Spain
| | - Irene Mora-Gutiérrez
- Servicio de Aparato Digestivo, Hospital Universitario de La Princesa e Instituto de Investigación Sanitaria Princesa & Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Madrid, Spain
| | - Cecilio Santander
- Servicio de Aparato Digestivo, Hospital Universitario de La Princesa e Instituto de Investigación Sanitaria Princesa & Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Madrid, Spain.,Departamento de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Montserrat Baldan-Martin
- Servicio de Aparato Digestivo, Hospital Universitario de La Princesa e Instituto de Investigación Sanitaria Princesa & Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Madrid, Spain
| | - Javier P Gisbert
- Servicio de Aparato Digestivo, Hospital Universitario de La Princesa e Instituto de Investigación Sanitaria Princesa & Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Madrid, Spain.,Departamento de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - David Bernardo
- Servicio de Aparato Digestivo, Hospital Universitario de La Princesa e Instituto de Investigación Sanitaria Princesa & Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Madrid, Spain.,Mucosal Immunology Lab, Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
| |
Collapse
|
14
|
The Role of Inflammation in Crohn's Disease Recurrence after Surgical Treatment. J Immunol Res 2020; 2020:8846982. [PMID: 33426097 PMCID: PMC7781709 DOI: 10.1155/2020/8846982] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 02/08/2023] Open
Abstract
Introduction Postoperative recurrence after surgery for Crohn's disease (CD) is virtually inevitable, and its mechanism is poorly known. Aim To review the numerous factors involved in CD postoperative recurrence (POR) pathogenesis, focusing on single immune system components as well as the immune system as a whole and highlighting the clinical significance in terms of preventive strategies and future perspectives. Methods A systematic literature search on CD POR, followed by a review of the main findings. Results The immune system plays a pivotal role in CD POR, with many different factors involved. Memory T-lymphocytes retained in mesenteric lymph nodes seem to represent the main driving force. New pathophysiology-based preventive strategies in the medical and surgical fields may help reduce POR rates. In particular, surgical strategies have already been developed and are currently under investigation. Conclusions POR is a complex phenomenon, whose driving mechanisms are gradually being unraveled. New preventive strategies addressing these mechanisms seem promising.
Collapse
|
15
|
Schulz-Kuhnt A, Wirtz S, Neurath MF, Atreya I. Regulation of Human Innate Lymphoid Cells in the Context of Mucosal Inflammation. Front Immunol 2020; 11:1062. [PMID: 32655549 PMCID: PMC7324478 DOI: 10.3389/fimmu.2020.01062] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
Since their identification as a unique cell population, innate lymphoid cells (ILCs) have revolutionized our understanding of immune responses, leaving their impact on multiple inflammatory and fibrotic pathologies without doubt. Thus, a tightly controlled regulation of local ILC numbers and their activity is of crucial importance. Even though this has been extensively studied in murine ILCs in the last few years, our knowledge of human ILCs is still lagging behind. Our review article will therefore summarize recent insights into the function of human ILCs and will particularly focus on their regulation under inflammatory conditions. The quality and intensity of ILC involvement into local immune responses at mucosal sites of the human body can potentially be modulated via three different axes: (1) activation of tissue-resident mature ILCs, (2) plasticity and local transdifferentiation of specific ILC subsets, and (3) tissue migration and accumulation of peripheral ILCs. Despite a still ongoing scientific effort in this field, already existing data on the fate of human ILCs under different pathologic conditions clearly indicate that all three of these mechanisms are of relevance for the clinical course of chronic inflammatory and autoimmune diseases and might likewise provide new target structures for future therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | - Imke Atreya
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| |
Collapse
|
16
|
Rigoni R, Fontana E, Dobbs K, Marrella V, Taverniti V, Maina V, Facoetti A, D'Amico G, Al-Herz W, Cruz-Munoz ME, Schuetz C, Gennery AR, Garabedian EK, Giliani S, Draper D, Dbaibo G, Geha RS, Meyts I, Tousseyn T, Neven B, Moshous D, Fischer A, Schulz A, Finocchi A, Kuhns DB, Fink DL, Lionakis MS, Swamydas M, Guglielmetti S, Alejo J, Myles IA, Pittaluga S, Notarangelo LD, Villa A, Cassani B. Cutaneous barrier leakage and gut inflammation drive skin disease in Omenn syndrome. J Allergy Clin Immunol 2020; 146:1165-1179.e11. [PMID: 32311393 PMCID: PMC7649331 DOI: 10.1016/j.jaci.2020.04.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 03/11/2020] [Accepted: 04/06/2020] [Indexed: 12/30/2022]
Abstract
Background Severe early-onset erythroderma and gut inflammation, with massive tissue infiltration of oligoclonal activated T cells are the hallmark of Omenn syndrome (OS). Objective The impact of altered gut homeostasis in the cutaneous manifestations of OS remains to be clarified. Methods We analyzed a cohort of 15 patients with OS and the 129Sv/C57BL/6 knock-in Rag2R229Q/R229Q (Rag2R229Q) mouse model. Homing phenotypes of circulating lymphocytes were analyzed by flow cytometry. Inflammatory cytokines and chemokines were examined in the sera by ELISA and in skin biopsies by immunohistochemistry and in situ RNA hybridization. Experimental colitis was induced in mice by dextran sulfate sodium salt. Results We show that memory/activated T cells from patients with OS and from the Rag2R229Q mouse model of OS abundantly express the skin homing receptors cutaneous lymphocyte associated antigen and CCR4 (Ccr4), associated with high levels of chemokine C-C motif ligands 17 and 22. Serum levels of LPS are also elevated. A broad Th1/Th2/Th17 inflammatory signature is detected in the periphery and in the skin. Increased Tlr4 expression in the skin of Rag2R229Q mice is associated with enhanced cutaneous inflammation on local and systemic administration of LPS. Likewise, boosting colitis in Rag2R229Q mice results in increased frequency of Ccr4+ splenic T cells and worsening of skin inflammation, as indicated by epidermal thickening, enhanced epithelial cell activation, and dermal infiltration by Th1 effector T cells. Conclusions These results support the existence of an interplay between gut and skin that can sustain skin inflammation in OS.
Collapse
Affiliation(s)
- Rosita Rigoni
- Milan Unit, Institute for Genetic and Biomedical Research (IRGB) National Research Council (CNR), Milan, Italy; Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Elena Fontana
- Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Kerry Dobbs
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, Md
| | - Veronica Marrella
- Milan Unit, Institute for Genetic and Biomedical Research (IRGB) National Research Council (CNR), Milan, Italy; Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Valentina Taverniti
- Department of Food, Environmental, and Nutritional Sciences, University of Milan Milan, Italy
| | - Virginia Maina
- Milan Unit, Institute for Genetic and Biomedical Research (IRGB) National Research Council (CNR), Milan, Italy; Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Amanda Facoetti
- Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy; Humanitas University, Rozzano, Milan, Italy
| | - Giovanna D'Amico
- Centro Ricerca Tettamanti, Clinica Pediatrica, Università Milano-Bicocca, Monza, Italy
| | - Waleed Al-Herz
- Department of Pediatrics, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait; Allergy and Clinical Immunology Unit, Pediatric Department, Al-Sabah Hospital, Kuwait City, Kuwait
| | | | - Catharina Schuetz
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Andrew R Gennery
- Great North Children's Hospital, Clinical Resource Building, Newcastle upon Tyne, United Kingdom; Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Silvia Giliani
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Cytogenetic and Medical Genetics Unit, "A. Nocivelli" Institute for Molecular Medicine, Spedali Civili Hospital, Brescia, Italy
| | - Deborah Draper
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, Md
| | - Ghassan Dbaibo
- Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Raif S Geha
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Isabelle Meyts
- Department of Pediatrics, Universitair Ziekenhuis Leuven, University Hospitals Leuven, Leuven, Belgium; Laboratory for Inborn Errors of Immunity, Department of Immunology, Microbiology and Transplantation, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Thomas Tousseyn
- Lab for Translational Cell and Tissue Research, Department of Imaging and Pathology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Benedicte Neven
- Imagine Institute, Paris Descartes-Sorbonne Paris Cité University, Paris, France; Pediatric Immuno-Hematology Unit, Necker Children Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Despina Moshous
- Imagine Institute, Paris Descartes-Sorbonne Paris Cité University, Paris, France; Pediatric Immuno-Hematology Unit, Necker Children Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Alain Fischer
- Imagine Institute, Paris Descartes-Sorbonne Paris Cité University, Paris, France; Pediatric Immuno-Hematology Unit, Necker Children Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Ansgar Schulz
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Andrea Finocchi
- Department of Pediatrics, Children's Hospital Bambino Gesù, Rome, Italy
| | - Douglas B Kuhns
- Neutrophil Monitoring Laboratory, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, Md
| | - Danielle L Fink
- Neutrophil Monitoring Laboratory, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, Md
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Muthulekha Swamydas
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Simone Guglielmetti
- Department of Food, Environmental, and Nutritional Sciences, University of Milan Milan, Italy
| | - Julie Alejo
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Md
| | - Ian A Myles
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, Md
| | - Stefania Pittaluga
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Md
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, Md.
| | - Anna Villa
- Milan Unit, Institute for Genetic and Biomedical Research (IRGB) National Research Council (CNR), Milan, Italy; Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cells, and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Barbara Cassani
- Milan Unit, Institute for Genetic and Biomedical Research (IRGB) National Research Council (CNR), Milan, Italy; Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy.
| |
Collapse
|
17
|
A three-dimensional immunocompetent intestine-on-chip model as in vitro platform for functional and microbial interaction studies. Biomaterials 2019; 220:119396. [PMID: 31398556 DOI: 10.1016/j.biomaterials.2019.119396] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/08/2019] [Accepted: 07/28/2019] [Indexed: 12/31/2022]
Abstract
Alterations of the microbial composition in the gut and the concomitant dysregulation of the mucosal immune response are associated with the pathogenesis of opportunistic infections, chronic inflammation, and inflammatory bowel disease. To create a platform for the investigation of the underlying mechanisms, we established a three-dimensional microphysiological model of the human intestine. This model resembles organotypic microanatomical structures and includes tissue resident innate immune cells exhibiting features of mucosal macrophages and dendritic cells. The model displays the physiological immune tolerance of the intestinal lumen to microbial-associated molecular patterns and can, therefore, be colonised with living microorganisms. Functional studies on microbial interaction between probiotic Lactobacillus rhamnosus and the opportunistic pathogen Candida albicans show that pre-colonization of the intestinal lumen of the model by L. rhamnosus reduces C. albicans-induced tissue damage, lowers its translocation, and limits fungal burden. We demonstrate that microbial interactions can be efficiently investigated using the in vitro model creating a more physiological and immunocompetent microenvironment. The intestinal model allows a detailed characterisation of the immune response, microbial pathogenicity mechanisms, and quantification of cellular dysfunction attributed to alterations in the microbial composition.
Collapse
|
18
|
Di Fusco D, Dinallo V, Marafini I, Figliuzzi MM, Romano B, Monteleone G. Antisense Oligonucleotide: Basic Concepts and Therapeutic Application in Inflammatory Bowel Disease. Front Pharmacol 2019; 10:305. [PMID: 30983999 PMCID: PMC6450224 DOI: 10.3389/fphar.2019.00305] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/12/2019] [Indexed: 12/17/2022] Open
Abstract
Several molecular technologies aimed at regulating gene expression that have been recently developed as a strategy to combat inflammatory and neoplastic diseases. Among these, antisense technology is a specific, rapid, and potentially high-throughput approach for inhibiting gene expression through recognition of cellular RNAs. Advances in the understanding of the molecular mechanisms that drive tissue damage in different inflammatory diseases, including Crohn's disease (CD) and ulcerative colitis (UC), the two major inflammatory bowel diseases (IBDs) in humans, have facilitated the identification of novel druggable targets and offered interesting therapeutic perspectives for the treatment of patients. This short review provides a comprehensive understanding of the basic concepts underlying the mechanism of action of the oligonucleotide therapeutics, and summarizes the available pre-clinical and clinical data for oligonucleotide-based therapy in IBD.
Collapse
Affiliation(s)
- Davide Di Fusco
- Department of Systems Medicine, Gastroenterology, University of Tor Vergata, Rome, Italy
| | - Vincenzo Dinallo
- Department of Systems Medicine, Gastroenterology, University of Tor Vergata, Rome, Italy
| | - Irene Marafini
- Department of Systems Medicine, Gastroenterology, University of Tor Vergata, Rome, Italy
| | - Michele M Figliuzzi
- Department of Systems Medicine, Gastroenterology, University of Tor Vergata, Rome, Italy
| | - Barbara Romano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Giovanni Monteleone
- Department of Systems Medicine, Gastroenterology, University of Tor Vergata, Rome, Italy
| |
Collapse
|
19
|
Blokland SLM, Flessa CM, van Roon JAG, Mavragani CP. Emerging roles for chemokines and cytokines as orchestrators of immunopathology in Sjögren's syndrome. Rheumatology (Oxford) 2019; 60:3072-3087. [PMID: 30838419 DOI: 10.1093/rheumatology/key438] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/03/2018] [Indexed: 12/14/2022] Open
Abstract
In primary SS (pSS), chemokines and cytokines orchestrate immunopathology driven by a complex network of interacting inflammatory cells. In recent years, the importance of chemotactic and non-chemotactic cytokines that control function, movement and placing of all cells within the inflamed exocrine glands and directing immunopathology has become increasingly clear. This paper reviews the current knowledge on chemokines and focuses on the emerging roles of novel chemotactic and non-chemotactic mediators in pSS. It highlights their contribution to pathogenic processes such as B cell hyperactivity and the formation of ectopic lymphoid structures. To this end, the role of acquired (CXCR5/CCR9 Th-cell-mediated) and innate (inflammasome/IL-1/IL-18-mediated) pathways in steering immunopathology is discussed.
Collapse
Affiliation(s)
- Sofie L M Blokland
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, The Netherlands.,Laboratory of Translational Immunology, Department of Immunology, Utrecht University, Utrecht, The Netherlands
| | - Christina-Maria Flessa
- Department of Physiology, National and Kapodistrian University of Athens, Athens, Greece
| | - Joel A G van Roon
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, The Netherlands.,Laboratory of Translational Immunology, Department of Immunology, Utrecht University, Utrecht, The Netherlands
| | - Clio P Mavragani
- Department of Physiology, National and Kapodistrian University of Athens, Athens, Greece.,Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Joint Academic Rheumatology Program, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
20
|
Cosorich I, McGuire HM, Warren J, Danta M, King C. CCR9 Expressing T Helper and T Follicular Helper Cells Exhibit Site-Specific Identities During Inflammatory Disease. Front Immunol 2019; 9:2899. [PMID: 30662436 PMCID: PMC6329311 DOI: 10.3389/fimmu.2018.02899] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022] Open
Abstract
CD4+ T helper (Th) cells that express the gut homing chemokine receptor CCR9 are increased in the peripheral blood of patients with inflammatory bowel disease and Sjögren's syndrome and in the inflamed lesions of autoimmune diseases that affect the accessory organs of the digestive system. However, despite the important role of the GIT in both immunity and autoimmunity, the nature of CCR9-expressing cells in GIT lymphoid organs and their role in chronic inflammatory diseases remains unknown. In this study, we analyzed the characteristics of CCR9+ Th and T follicular helper (Tfh) cells in GIT associated lymphoid tissues in health, chronic inflammation and autoimmunity. Our findings reveal an association between the transcriptome and phenotype of CCR9+ Th in the pancreas and CCR9+ Tfh cells from GIT-associated lymphoid tissues. GIT CCR9+ Tfh cells exhibited characteristics, including a Th17-like transcriptome and production of effector cytokines, which indicated a microenvironment-specific signature. Both CCR9+ Tfh cells and CCR9+ Th cells from GIT-associated lymphoid tissues migrated to the pancreas. The expression of CCR9 was important for migration of both subsets to the pancreas, but Tfh cells that accumulated in the pancreas had downmodulated expression of CXCR5. Taken together, the findings provide evidence that CCR9+ Tfh cells and Th cells from the GIT exhibit plasticity and can accumulate in distal accessory organs of the digestive system where they may participate in autoimmunity.
Collapse
Affiliation(s)
- Ilaria Cosorich
- Department of Immunology, The Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Helen M McGuire
- Department of Immunology, The Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Joanna Warren
- Department of Immunology, The Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Mark Danta
- St Vincent's Clinical School, University of NSW, Sydney, NSW, Australia
| | - Cecile King
- Department of Immunology, The Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of NSW, Sydney, NSW, Australia
| |
Collapse
|
21
|
Abstract
The growing understanding of the immunopathogenesis of inflammatory bowel diseases (IBDs) has contributed to the identification of new targets whose expression/activity can be modulated for therapeutic purposes. Several approaches have been employed to develop selective pharmaceutical compounds; among these, antisense oligonucleotides (ASOs) or synthetic oligonucleotides represent a valid option for inhibiting or enhancing, respectively, the expression/function of molecules that have been implicated in the control of IBD-related inflammation. In this context, data have been accumulated for the following compounds: alicaforsen, an ASO targeting intercellular adhesion molecule-1, a transmembrane glycoprotein that regulates rolling and adhesion of leukocytes to inflamed intestine; DIMS0150 and BL-7040, two oligonucleotides that enhance Toll-like receptor-9 activity; Mongersen, an ASO that inhibits Smad7, thereby restoring transforming growth factor-β1/Smad-associated signaling; STNM01, a double-stranded RNA oligonucleotide silencing carbohydrate sulfotransferase, an enzyme involved in fibrogenic processes, and hgd40, a specific DNAzyme inhibiting expression of the transcription factor GATA3. In this article, we review the rationale and the available data relative to the use of these agents in IBD. Although pre-clinical and phase II trials in IBD support the use of oligonucleotide-based therapies for treating the pathogenic process occurring in the gut of patients with these disorders, further work is needed to establish whether and which patients can benefit from specific ASOs and identify biomarkers that could help optimize treatment.
Collapse
|
22
|
He P, Haque A, Lin S, Cominelli F, Yun CC. Inhibition of autotaxin alleviates inflammation and increases the expression of sodium-dependent glucose cotransporter 1 and Na +/H + exchanger 3 in SAMP1/Fc mice. Am J Physiol Gastrointest Liver Physiol 2018; 315:G762-G771. [PMID: 30118349 PMCID: PMC6293258 DOI: 10.1152/ajpgi.00215.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Crohn's disease (CD) is a chronic, relapsing, inflammatory disease that is often associated with malnutrition because of inflammation in the small intestine. Autotaxin (ATX) is a secreted enzyme that produces extracellular lysophosphatidic acid. Increasing evidence suggests that ATX is upregulated during inflammation, and inhibition of ATX has been effective in attenuating chronic inflammatory conditions, such as arthritis and pulmonary fibrosis. This study aims to determine whether inhibition of ATX alleviates CD-associated inflammation and malnutrition by using SAMP1/Fc mice, a model of CD-like ileitis. SAMP1/Fc mice were treated the ATX inhibitor PF-8380 for 4 wk. Inhibition of ATX led to increased weight gain in SAMP1/Fc mice, decreased T helper 2 cytokine expression, including IL-4, IL-5, and IL-13, and attenuated immune cell migration. SAMP1/Fc mice have low expression of Na+-dependent glucose transporter 1 (SGLT1), suggesting impaired nutrient absorption associated with ileitis. PF-8380 treatment significantly enhanced SGLT1 expression in SAMP1/Fc mice, which could reflect the increased weight changes. However, IL-4 or IL-13 did not alter SGLT1 expression in Caco-2 cells, ruling out their direct effects on SGLT1 expression. Immunofluorescence analysis showed that the expression of sucrase-isomaltase, a marker for intestinal epithelial cell (IEC) differentiation, was decreased in inflamed regions of SAMP1/Fc mice, which was partially restored by PF-8380. Moreover, expression of Na+/H+ exchanger 3 was also improved by PF-8380, suggesting that suppression of inflammation by PF-8380 enhanced IEC differentiation. Our study therefore suggests that ATX is a potential target for treating intestinal inflammation and restoration of the absorptive function of the intestine. NEW & NOTEWORTHY This study is the first, to our knowledge, to determine whether autotoxin (ATX) inhibition improves inflammation and body weights in SAMP1/Fc mice, a mouse model of ileitis. ATX inhibition increased body weights of SAMP1/Fc mice and increased Na+-dependent glucose transporter 1 (SGLT1) expression. Increased SGLT1 expression in the inflamed regions was not a direct effect of cytokines but an indirect effect of increased epithelial cell differentiation upon ATX inhibition.
Collapse
Affiliation(s)
- Peijian He
- 1Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,2Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - Abedul Haque
- 1Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,2Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - Songbai Lin
- 1Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,2Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - Fabio Cominelli
- 3Department of Medicine, Case Digestive Health Research Institute, Case Western University School of Medicine, Cleveland, Ohio
| | - C. Chris Yun
- 1Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,2Atlanta Veterans Affairs Medical Center, Decatur, Georgia,4Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
23
|
Fu J, Li G, Wu J, Wang Z. Intestinal IgA positive lymphocytes in acute liver necrosis decrease due to lymphocyte homing disturbance and apoptosis. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2018; 111:101-105. [PMID: 30318894 DOI: 10.17235/reed.2018.5656/2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
AIM the number of intestinal IgA+ lymphocytes are decreased in acute liver necrosis and the mechanism remains poorly understood. The purpose of this study was to observe the role of lymphocyte homing and apoptosis associated with decreased intestinal IgA positive lymphocytes in acute liver necrosis. METHODS the acute liver necrosis mouse model and LTβR pre-treatment were used to assess intestinal mucosal addressin cell adhesion molecule-1 (MAdCAM - 1) expression, cell apoptosis, IgA+ cells and secretory immunoglobulin A (SIgA). RESULTS MAdCAM - 1 mRNA and protein expression decreased significantly in the acute necrosis group; 0.57 ± 0.032 fold vs. baseline (p < 0.05) and 0.45 ± 0.072 fold vs. baseline (p < 0.05), respectively. LTβR pre-treatment could significantly improve the decline of MAdCAM - 1 mRNA and protein expression in the intestinal mucosa (1.83 ± 0.064 fold vs. baseline, p < 0.05 and 1.75 ± 0.046 fold vs. baseline, p < 0.05, respectively) and partially restore the decline in IgA+ lymphocytes and SIgA levels. There were increased rates of enterocyte apoptosis in both the acute liver necrosis and LTβR pre-treatment group; 0.79% vs. control (p < 0.05) and 0.77% vs. control (p < 0.05), respectively). CONCLUSION our results suggest that the dysfunction of lymphocyte homing and apoptosis are both involved with decreased intestinal IgA+ lymphocytes in acute liver necrosis. LTβR pre-treatment can partially restore IgA+ cells and SIgA by increasing MAdCAM - 1 expression, rather than inhibiting lymphocyte apoptosis.
Collapse
Affiliation(s)
- Jinlong Fu
- Gastroenterology, The Affiliated Hospital,Hangzhou Normal University, China
| | - Guodong Li
- Gastroenterology, The Affiliated Hospital,Hangzhou Normal University School of Medicine
| | - Jianliang Wu
- Gastroenterology, The Affiliated Hospital,Hangzhou Normal University School of Medicine, China
| | - Zhiyong Wang
- Gastroenterology, The Affiliated Hospital,Hangzhou Normal University School of Medicine, China
| |
Collapse
|
24
|
Anand S, Mande SS. Diet, Microbiota and Gut-Lung Connection. Front Microbiol 2018; 9:2147. [PMID: 30283410 PMCID: PMC6156521 DOI: 10.3389/fmicb.2018.02147] [Citation(s) in RCA: 261] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 08/21/2018] [Indexed: 12/19/2022] Open
Abstract
The gut microbial community (Gut microbiota) is known to impact metabolic functions as well as immune responses in our body. Diet plays an important role in determining the composition of the gut microbiota. Gut microbes help in assimilating dietary nutrients which are indigestible by humans. The metabolites produced by them not only modulate gastro-intestinal immunity, but also impact distal organs like lung and brain. Micro-aspiration of gut bacteria or movement of sensitized immune cells through lymph or bloodstream can also influence immune response of other organs. Dysbiosis in gut microbiota has been implicated in several lung diseases, including allergy, asthma and cystic fibrosis. The bi-directional cross-talk between gut and lung (termed as Gut-Lung axis) is best exemplified by intestinal disturbances observed in lung diseases. Some of the existing probiotics show beneficial effects on lung health. A deeper understanding of the gut microbiome which comprises of all the genetic material within the gut microbiota and its role in respiratory disorders is likely to help in designing appropriate probiotic cocktails for therapeutic applications.
Collapse
Affiliation(s)
- Swadha Anand
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services Ltd., Pune, India
| | - Sharmila S Mande
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services Ltd., Pune, India
| |
Collapse
|
25
|
Abstract
Inflammatory bowel diseases (IBD) are chronic inflammatory conditions of the gastrointestinal tract encompassing two main clinical entities: Crohn's disease (CD) and ulcerative colitis (UC). These disorders are characterized by various grades of tissue damage and development of local complications and extra-intestinal manifestations. The cause of IBD remains unknown but accumulating evidence indicates that both CD and UC arise in genetically predisposed individuals as a result of the action of multiple environmental factors, which ultimately trigger excessive and poorly controlled immune response against antigens of the luminal flora. Despite this realization, a full understanding of IBD pathogenesis is still out of reach and, consequently, treatment is far from optimal. However, in recent years, several pathways of intestinal damage have been delineated and the improved knowledge has contributed to the development of new therapies. Various approaches have been used to either inhibit the expression and/or function of inflammatory molecules or enhance counter-regulatory mechanisms. This review summarizes the available pre-clinical and clinical data for antisense oligonucleotides and oligonucleotide-based therapy to provide a comprehensive understanding of the rationale and mechanism of action of these compounds in IBD. Key messages Preclinical studies and clinical trials show that antisense oligonucleotide (ASO)-based therapy could be of benefit in inflammatory bowel diseases. ASOs have an excellent safety profile. Technical issues emerged from clinical trials suggest that changes in drug formulation and/or route of administration could improve ASO efficacy.
Collapse
Affiliation(s)
- Irene Marafini
- a Department of Systems Medicine , University of Rome Tor Vergata , Rome, Italy
| | - Giovanni Monteleone
- a Department of Systems Medicine , University of Rome Tor Vergata , Rome, Italy
| |
Collapse
|
26
|
Association of Three Polymorphisms rs11614913, rs2910146, and rs3746444 in miRNA-196a2, miRNA-146a, and miRNA-499 with Inflammatory Bowel Disease: A Systematic Review and Meta-Analysis. Gastroenterol Res Pract 2018; 2018:7295131. [PMID: 29706994 PMCID: PMC5863352 DOI: 10.1155/2018/7295131] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 11/02/2017] [Accepted: 11/14/2017] [Indexed: 02/07/2023] Open
Abstract
Background It has been found that single-nucleotide polymorphisms (SNPs) of microRNA might be involved in the development of inflammatory bowel diseases (IBDs). However, the related retrospective research has not been reported. In this work, we performed a meta-analysis to derive a more precise estimation of the associated relationship. Methods We searched the studies on the association of SNPs of microRNA with the hereditary susceptibility of IBD in PubMed and Embase; eligible research was selected by screening the abstract and full text. The meta-analysis was performed based on the statistical software Stata 14.0, and besides, the odds ratio and 95% confidence interval were calculated to evaluate the strength of the association. Results 159 papers were acquired from the PubMed and Embase databases, and five eligible articles containing nine case-control studies were selected. In the study, we first found that the association between miRNA-196a2 rs11614913 and IBD was insignificant. Then, the susceptibility of miRNA-146a rs2910146 to IBD increased significantly in allelic comparison, homozygote model, heterozygote model, and dominant model. Moreover, a positive relationship between miRNA-499 rs3746444 and IBD was identified in the homozygote model. Conclusion Our findings demonstrated that miRNA-146a rs2910146 (G>C) polymorphism was associated with the susceptibility to IBD and miRNA-196a2 rs11614913 (T>C) and miRNA-499 rs3746444 (A>G) did not reveal an obvious relationship with the IBD susceptibility.
Collapse
|
27
|
Bernardo D, Chaparro M, Gisbert JP. Human Intestinal Dendritic Cells in Inflammatory Bowel Diseases. Mol Nutr Food Res 2018; 62:e1700931. [PMID: 29336524 DOI: 10.1002/mnfr.201700931] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/03/2018] [Indexed: 12/21/2022]
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a serious, costly, and persistent health problem with an estimated prevalence in Western countries around 0.5% of the general population; its socioeconomic impact is comparable with that for chronic diseases such as diabetes. Conventional treatment involves escalating drug regimens with concomitant side effects followed, in some cases, by surgical interventions, which are often multiple, mainly in Crohn's disease. The goal of finding a targeted gut-specific immunotherapy for IBD patients is therefore an important unmet need. However, to achieve this goal we first must understand how dendritic cells (DC), the most potent antigen present cells of the immune system, control the immune tolerance in the gastrointestinal tract and how their properties are altered in those patients suffering from IBD. In this review, we summarize the current available information regarding human intestinal DC subsets composition, phenotype, and function in the human gastrointestinal tract describing how, in the IBD mucosa, DC display pro-inflammatory properties, which drive disease progression. A better understanding of the mechanisms inducing DC abnormal profile in IBD may provide us with novel tools to perform tissue specific immunomodulation.
Collapse
Affiliation(s)
- David Bernardo
- Gastroenterology Unit, Hospital Universitario de La Princesa and Instituto de Investigación Sanitaria Princesa (IIS-IP), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - María Chaparro
- Gastroenterology Unit, Hospital Universitario de La Princesa and Instituto de Investigación Sanitaria Princesa (IIS-IP), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Javier P Gisbert
- Gastroenterology Unit, Hospital Universitario de La Princesa and Instituto de Investigación Sanitaria Princesa (IIS-IP), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| |
Collapse
|
28
|
Dzutsev A, Hogg A, Sui Y, Solaymani-Mohammadi S, Yu H, Frey B, Wang Y, Berzofsky JA. Differential T cell homing to colon vs. small intestine is imprinted by local CD11c + APCs that determine homing receptors. J Leukoc Biol 2017; 102:1381-1388. [PMID: 28951425 DOI: 10.1189/jlb.1a1116-463rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 09/08/2017] [Accepted: 09/11/2017] [Indexed: 01/07/2023] Open
Abstract
Mechanisms that imprint T cell homing to the small intestine have been well studied; however, those for homing to the colon are poorly understood. Recently, we found that these are distinct subcompartments of the gut mucosal immune system, which implies differential homing. Here, we show that colonic CD11c+ APCs imprint CD8+ T cell preferential homing to the colon, in contrast to those from the small intestine that imprint CD8+ T cell homing to the small intestine, and that the differences are related to the variable ability of APCs to induce α4β7-integrin and CCR9 expression on T cells. Colon APCs also expressed lower levels of retinoic acid-producing enzymes that are known to control the mucosal homing of T cells. These findings are the first to our knowledge to directly demonstrate that colon APCs imprint T cells to selectively home to the large bowel, which is critical for the design of successful T cell-based therapies and vaccines, such as colon cancer immunotherapy and HIV vaccines.
Collapse
Affiliation(s)
- Amiran Dzutsev
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Alison Hogg
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Yongjun Sui
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | | | - Huifeng Yu
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Blake Frey
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Yichuan Wang
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| |
Collapse
|
29
|
Anti-NKG2D mAb: A New Treatment for Crohn's Disease? Int J Mol Sci 2017; 18:ijms18091997. [PMID: 28926962 PMCID: PMC5618646 DOI: 10.3390/ijms18091997] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 08/24/2017] [Accepted: 09/11/2017] [Indexed: 01/09/2023] Open
Abstract
Crohn’s disease (CD) and ulcerative colitis (UC) are immunologically-mediated, debilitating conditions resulting from destructive inflammation of the gastrointestinal tract. The pathogenesis of IBD is incompletely understood, but is considered to be the result of an abnormal immune response with a wide range of cell types and proteins involved. Natural Killer Group 2D (NKG2D) is an activating receptor constitutively expressed on human Natural Killer (NK), γδ T, mucosal-associated invariant T (MAIT), CD56+ T, and CD8+ T cells. Activation of NKG2D triggers cellular proliferation, cytokine production, and target cell killing. Research into the NKG2D mechanism of action has primarily been focused on cancer and viral infections where cytotoxicity evasion is a concern. In human inflammatory bowel disease (IBD) this system is less characterized, but the ligands have been shown to be highly expressed during intestinal inflammation and the following receptor activation may contribute to tissue degeneration. A recent phase II clinical trial showed that an antibody against NKG2D induced clinical remission of CD in some patients, suggesting NKG2D and its ligands to be of importance in the pathogenesis of CD. This review will describe the receptor and its ligands in intestinal tissues and the clinical potential of blocking NKG2D in Crohn’s disease.
Collapse
|
30
|
Blokland SLM, Hillen MR, Kruize AA, Meller S, Homey B, Smithson GM, Radstake TRDJ, van Roon JAG. Increased CCL25 and T Helper Cells Expressing CCR9 in the Salivary Glands of Patients With Primary Sjögren's Syndrome: Potential New Axis in Lymphoid Neogenesis. Arthritis Rheumatol 2017. [DOI: 10.1002/art.40182] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
| | | | - Aike A. Kruize
- University Medical Center Utrecht; Utrecht The Netherlands
| | | | | | | | | | | |
Collapse
|
31
|
Vadstrup K, Galsgaard ED, Jensen H, Lanier LL, Ryan JC, Chen SY, Nolan GP, Vester-Andersen MK, Pedersen JS, Gerwien J, Jensen T, Bendtsen F. NKG2D ligand expression in Crohn's disease and NKG2D-dependent stimulation of CD8 + T cell migration. Exp Mol Pathol 2017; 103:56-70. [PMID: 28684217 DOI: 10.1016/j.yexmp.2017.06.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 05/24/2017] [Accepted: 06/30/2017] [Indexed: 12/15/2022]
Abstract
Interaction between the activating NKG2D receptor on lymphocytes and its ligands MICA, MICB, and ULBP1-6 modulate T and NK cell activity and may contribute to the pathogenesis of Crohn's disease (CD). NKG2D ligands are generally not expressed on the cell surface of normal, non-stressed cells, but expression of MICA and MICB in CD intestine has been reported. In this exploratory study, we further characterize the expression of NKG2D and its ligands, including the less well-described ULBP4-6, in CD, and test if NKG2D ligand interactions are involved in the migration of activated T cells into the affected mucosal compartments. Intestinal tissue from CD patients and healthy controls were analyzed by flow cytometry, mass cytometry, and immunohistochemistry for expression of NKG2D and ligands, and for cytokine release. Furthermore, NKG2D-dependent chemotaxis of activated CD8+ T cells across a monolayer of ligand-expressing human intestinal endothelial cells was examined. Activated lymphocytes down-regulated NKG2D expression upon accumulation in inflamed CD intestine. NKG2D expression on CD56+ T and γδ T cells from inflamed tissue seemed inversely correlated with CRP levels and cytokine release. B cells, monocytes, mucosal epithelium, and vascular endothelium expressed NKG2D ligands in inflamed CD intestine. The expression of NKG2D ligands was correlated with cytokine release, but was highly variable between patients. Stimulation of vascular intestinal endothelial cells in vitro induced expression of NKG2D ligands, including MICA/B and ULBP2/6. Blockade of NKG2D on CD8+ T cells inhibited the migration over ligand-expressing endothelial cells. Intestinal induction of NKG2D ligands and ligand-induced down-regulation of NKG2D in CD suggest that the NKG2D-ligand interaction may be involved in both the activation and recruitment of NKG2D+ lymphocytes into the inflamed CD intestine.
Collapse
Affiliation(s)
- Kasper Vadstrup
- Gastrounit, Medical Division, Hvidovre University Hospital, DK-2650 Hvidovre, Denmark; Faculty of Health Sciences, The Panum Institute, University of Copenhagen, DK-2200 Copenhagen N, Denmark; Biopharmaceutical Research Unit, Novo Nordisk A/S, DK-2760 Maaloev, Denmark; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | | | - Helle Jensen
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lewis L Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - James C Ryan
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Medicine, Veterans Affairs Medical Center and University of California San Francisco, San Francisco, CA, USA
| | - Shih-Yu Chen
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | - Garry P Nolan
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | | | - Julie Steen Pedersen
- Gastrounit, Medical Division, Hvidovre University Hospital, DK-2650 Hvidovre, Denmark
| | - Jens Gerwien
- Biopharmaceutical Research Unit, Novo Nordisk A/S, DK-2760 Maaloev, Denmark
| | - Teis Jensen
- Biopharmaceutical Research Unit, Novo Nordisk A/S, DK-2760 Maaloev, Denmark
| | - Flemming Bendtsen
- Gastrounit, Medical Division, Hvidovre University Hospital, DK-2650 Hvidovre, Denmark; Faculty of Health Sciences, The Panum Institute, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| |
Collapse
|
32
|
Escudero-Hernández C, Peña AS, Bernardo D. Immunogenetic Pathogenesis of Celiac Disease and Non-celiac Gluten Sensitivity. Curr Gastroenterol Rep 2017; 18:36. [PMID: 27216895 DOI: 10.1007/s11894-016-0512-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Celiac disease is the most common oral intolerance in Western countries. It results from an immune response towards gluten proteins from certain cereals in genetically predisposed individuals (HLA-DQ2 and/or HLA-DQ8). Its pathogenesis involves the adaptive (HLA molecules, transglutaminase 2, dendritic cells, and CD4(+) T-cells) and the innate immunity with an IL-15-mediated response elicited in the intraepithelial compartment. At present, the only treatment is a permanent strict gluten-free diet (GFD). Multidisciplinary studies have provided a deeper insight of the genetic and immunological factors and their interaction with the microbiota in the pathogenesis of the disease. Similarly, a better understanding of the composition of the toxic gluten peptides has improved the ways to detect them in food and drinks and how to monitor GFD compliance via non-invasive approaches. This review, therefore, addresses the major findings obtained in the last few years including the re-discovery of non-celiac gluten sensitivity.
Collapse
Affiliation(s)
- Celia Escudero-Hernández
- Mucosal Immunology Laboratory, IBGM, Facultad de Medicina, Dpto. Pediatría e Inmunología, University of Valladolid-Consejo Superior de Investigaciones Científicas, (4th floor) Av. Ramón y Cajal 7, 47005, Valladolid, Spain
| | - Amado Salvador Peña
- VU Medical Center Amsterdam, Laboratory of Immunogenetics, Department of Medical Microbiology and Infection Control, VU University Medical Center, De Boelelaan 1108 Room 10E65, 1081 HZ, Amsterdam, The Netherlands
| | - David Bernardo
- Gastroenterology Unit, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, 28006, Spain.
| |
Collapse
|
33
|
Karuppuchamy T, Behrens EH, González-Cabrera P, Sarkisyan G, Gima L, Boyer JD, Bamias G, Jedlicka P, Veny M, Clark D, Peach R, Scott F, Rosen H, Rivera-Nieves J. Sphingosine-1-phosphate receptor-1 (S1P 1) is expressed by lymphocytes, dendritic cells, and endothelium and modulated during inflammatory bowel disease. Mucosal Immunol 2017; 10:162-171. [PMID: 27049060 PMCID: PMC5053832 DOI: 10.1038/mi.2016.35] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/07/2016] [Indexed: 02/04/2023]
Abstract
The sphingosine-1-phosphate receptor-1 (S1P1) agonist ozanimod ameliorates ulcerative colitis, yet its mechanism of action is unknown. Here, we examine the cell subsets that express S1P1 in intestine using S1P1-eGFP mice, the regulation of S1P1 expression in lymphocytes after administration of dextran sulfate sodium (DSS), after colitis induced by transfer of CD4+CD45RBhi cells, and by crossing a mouse with TNF-driven ileitis with S1P1-eGFP mice. We then assayed the expression of enzymes that regulate intestinal S1P levels, and the effect of FTY720 on lymphocyte behavior and S1P1 expression. We found that not only T and B cells express S1P1, but also dendritic (DC) and endothelial cells. Furthermore, chronic but not acute inflammatory signals increased S1P1 expression, while the enzymes that control tissue S1P levels in mice and humans with inflammatory bowel disease (IBD) were uniformly dysregulated, favoring synthesis over degradation. Finally, we observed that FTY720 reduced T-cell velocity and induced S1P1 degradation and retention of Naïve but not effector T cells. Our data demonstrate that chronic inflammation modulates S1P1 expression and tissue S1P levels and suggests that the anti-inflammatory properties of S1PR agonists might not be solely due to their lymphopenic effects, but also due to potential effects on DC migration and vascular barrier function.
Collapse
Affiliation(s)
- Thangaraj Karuppuchamy
- Inflammatory Bowel Disease Center, Division of Gastroenterology, University of California San Diego, La Jolla, CA,VA San Diego Healthcare System, San Diego, CA
| | - En-hui Behrens
- Inflammatory Bowel Disease Center, Division of Gastroenterology, University of California San Diego, La Jolla, CA,VA San Diego Healthcare System, San Diego, CA
| | - Pedro González-Cabrera
- Department of Chemical Physiology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA
| | - Gor Sarkisyan
- Department of Chemical Physiology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA
| | - Lauren Gima
- Inflammatory Bowel Disease Center, Division of Gastroenterology, University of California San Diego, La Jolla, CA,VA San Diego Healthcare System, San Diego, CA
| | - Joshua D. Boyer
- Inflammatory Bowel Disease Center, Division of Gastroenterology, University of California San Diego, La Jolla, CA,VA San Diego Healthcare System, San Diego, CA
| | - Giorgos Bamias
- Academic Department of Gastroenterology, Laikon Hospital, Athens, Greece
| | - Paul Jedlicka
- Department of Pathology, University of Colorado Health Sciences, Aurora, CO
| | - Marisol Veny
- Inflammatory Bowel Disease Center, Division of Gastroenterology, University of California San Diego, La Jolla, CA,VA San Diego Healthcare System, San Diego, CA
| | - David Clark
- Inflammatory Bowel Disease Center, Division of Gastroenterology, University of California San Diego, La Jolla, CA,VA San Diego Healthcare System, San Diego, CA
| | - Robert Peach
- Receptos Inc. 3033 Science Park Road, La Jolla, CA
| | - Fiona Scott
- Receptos Inc. 3033 Science Park Road, La Jolla, CA
| | - Hugh Rosen
- Department of Chemical Physiology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA
| | - Jesús Rivera-Nieves
- Inflammatory Bowel Disease Center, Division of Gastroenterology, University of California San Diego, La Jolla, CA,VA San Diego Healthcare System, San Diego, CA
| |
Collapse
|
34
|
Wu N, Song YL, Wang B, Zhang XY, Zhang XJ, Wang YL, Cheng YY, Chen DD, Xia XQ, Lu YS, Zhang YA. Fish gut-liver immunity during homeostasis or inflammation revealed by integrative transcriptome and proteome studies. Sci Rep 2016; 6:36048. [PMID: 27808112 PMCID: PMC5093735 DOI: 10.1038/srep36048] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 10/10/2016] [Indexed: 12/11/2022] Open
Abstract
The gut-associated lymphoid tissue, connected with liver via bile and blood, constructs a local immune environment of both defense and tolerance. The gut-liver immunity has been well-studied in mammals, yet in fish remains largely unknown, even though enteritis as well as liver and gallbladder syndrome emerged as a limitation in aquaculture. In this study, we performed integrative bioinformatic analysis for both transcriptomic (gut and liver) and proteomic (intestinal mucus and bile) data, in both healthy and infected tilapias. We found more categories of immune transcripts in gut than liver, as well as more adaptive immune in gut meanwhile more innate in liver. Interestingly reduced differential immune transcripts between gut and liver upon inflammation were also revealed. In addition, more immune proteins in bile than intestinal mucus were identified. And bile probably providing immune effectors to intestinal mucus upon inflammation was deduced. Specifically, many key immune transcripts in gut or liver as well as key immune proteins in mucus or bile were demonstrated. Accordingly, we proposed a hypothesized profile of fish gut-liver immunity, during either homeostasis or inflammation. Current data suggested that fish gut and liver may collaborate immunologically while keep homeostasis using own strategies, including potential unique mechanisms.
Collapse
Affiliation(s)
- Nan Wu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Yu-Long Song
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,Demorgen Bioinformation Technology Co. Ltd, Wuhan 430072, China
| | - Bei Wang
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Xiang-Yang Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xu-Jie Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Ya-Li Wang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Ying-Yin Cheng
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Dan-Dan Chen
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Xiao-Qin Xia
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Yi-Shan Lu
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yong-An Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,State Key Laboratory of Freshwater Ecology and Biotechnology, Wuhan 430072, China
| |
Collapse
|
35
|
Hauptmann M, Schaible UE. Linking microbiota and respiratory disease. FEBS Lett 2016; 590:3721-3738. [PMID: 27637588 DOI: 10.1002/1873-3468.12421] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 08/30/2016] [Accepted: 09/07/2016] [Indexed: 12/13/2022]
Abstract
An increasing body of evidence indicates the relevance of microbiota for pulmonary health and disease. Independent investigations recently demonstrated that the lung harbors a resident microbiota. Therefore, it is intriguing that a lung microbiota can shape pulmonary immunity and epithelial barrier functions. Here, we discuss the ways how the composition of the microbial community in the lung may influence pulmonary health and vice versa, factors that determine community composition. Prominent microbiota at other body sites such as the intestinal one may also contribute to pulmonary health and disease. However, it is difficult to discriminate between influences of lung vs. gut microbiota due to systemic mutuality between both communities. With focuses on asthma and respiratory infections, we discuss how microbiota of lung and gut can determine pulmonary immunity and barrier functions.
Collapse
Affiliation(s)
- Matthias Hauptmann
- Priority Program Infections, Cellular Microbiology, Research Center Borstel, Germany
| | - Ulrich E Schaible
- Priority Program Infections, Cellular Microbiology, Research Center Borstel, Germany.,German Centre for Infection Research, TTU-TB, Borstel, Germany
| |
Collapse
|
36
|
Brasseit J, Althaus-Steiner E, Faderl M, Dickgreber N, Saurer L, Genitsch V, Dolowschiak T, Li H, Finke D, Hardt WD, McCoy KD, Macpherson AJ, Corazza N, Noti M, Mueller C. CD4 T cells are required for both development and maintenance of disease in a new mouse model of reversible colitis. Mucosal Immunol 2016; 9:689-701. [PMID: 26376366 DOI: 10.1038/mi.2015.93] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 08/06/2015] [Indexed: 02/04/2023]
Abstract
Current therapies to treat inflammatory bowel diseases have limited efficacy, significant side effects, and often wane over time. Little is known about the cellular and molecular mechanisms operative in the process of mucosal healing from colitis. To study such events, we developed a new model of reversible colitis in which adoptive transfer of CD4(+)CD45RB(hi) T cells into Helicobacter typhlonius-colonized lymphopenic mice resulted in a rapid onset of colonic inflammation that was reversible through depletion of colitogenic T cells. Remission was associated with an improved clinical and histopathological score, reduced immune cell infiltration to the intestinal mucosa, altered intestinal gene expression profiles, regeneration of the colonic mucus layer, and the restoration of epithelial barrier integrity. Notably, colitogenic T cells were not only critical for induction of colitis but also for maintenance of disease. Depletion of colitogenic T cells resulted in a rapid drop in tumor necrosis factor α (TNFα) levels associated with reduced infiltration of inflammatory immune cells to sites of inflammation. Although neutralization of TNFα prevented the onset of colitis, anti-TNFα treatment of mice with established disease failed to resolve colonic inflammation. Collectively, this new model of reversible colitis provides an important research tool to study the dynamics of mucosal healing in chronic intestinal remitting-relapsing disorders.
Collapse
Affiliation(s)
- J Brasseit
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - E Althaus-Steiner
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - M Faderl
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - N Dickgreber
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - L Saurer
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - V Genitsch
- Division of Clinical Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - T Dolowschiak
- Institute of Microbiology, ETH Zürich, Zurich, Switzerland
| | - H Li
- Maurice E. Müller Laboratories, University Clinic for Visceral Surgery and Medicine, University of Bern, Bern, Switzerland
| | - D Finke
- Division of Developmental Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - W-D Hardt
- Institute of Microbiology, ETH Zürich, Zurich, Switzerland
| | - K D McCoy
- Maurice E. Müller Laboratories, University Clinic for Visceral Surgery and Medicine, University of Bern, Bern, Switzerland
| | - A J Macpherson
- Maurice E. Müller Laboratories, University Clinic for Visceral Surgery and Medicine, University of Bern, Bern, Switzerland
| | - N Corazza
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - M Noti
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - C Mueller
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| |
Collapse
|
37
|
Marafini I, Di Fusco D, Calabrese E, Sedda S, Pallone F, Monteleone G. Antisense approach to inflammatory bowel disease: prospects and challenges. Drugs 2016; 75:723-30. [PMID: 25911184 DOI: 10.1007/s40265-015-0391-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Despite the great success of anti-tumour necrosis factor-based therapies, the treatment of Crohn's disease (CD) and ulcerative colitis (UC) still remains a challenge for clinicians, as these drugs are not effective in all patients, their efficacy may wane with time, and their use can increase the risk of adverse events and be associated with the development of new immune-mediated diseases. Therefore, new therapeutic targets are currently being investigated both in pre-clinical studies and in clinical trials. Among the technologies used to build new therapeutic compounds, the antisense oligonucleotide (ASO) approach is slowly gaining space in the field of inflammatory bowel diseases (IBDs), and three ASOs have been investigated in clinical trials. Systemic administration of alicaforsen targeting intercellular adhesion molecule-1, a protein involved in the recruitment of leukocytes to inflamed intestine, was not effective in CD, even though the same compound was of benefit when given as an enema to UC patients. DIMS0150, targeting nuclear factor (NF) κB-p65, a transcription factor that promotes pro-inflammatory responses, was very promising in pre-clinical studies and is currently being tested in clinical trials. Oral mongersen, targeting Smad7, an intracellular protein that inhibits transforming growth factor (TGF)-β1 activity, was safe and well tolerated by CD patients, and the results of a phase II clinical trial showed the efficacy of the drug in inducing clinical remission in patients with active disease. In this leading article, we review the rationale and the clinical data available regarding these three agents, and we discuss the challenge of using ASOs in IBD.
Collapse
Affiliation(s)
- Irene Marafini
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | | | | | | | | | | |
Collapse
|
38
|
Bamias G, Pizarro TT, Cominelli F. Pathway-based approaches to the treatment of inflammatory bowel disease. Transl Res 2016; 167:104-15. [PMID: 26408803 PMCID: PMC4782917 DOI: 10.1016/j.trsl.2015.09.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 09/01/2015] [Accepted: 09/02/2015] [Indexed: 12/17/2022]
Abstract
Crohn's disease and ulcerative colitis, collectively termed inflammatory bowel disease (IBD), are immunologic disorders that represent the prototypes of chronic intestinal inflammation. Their pathogenesis involves the dysregulated interaction between the intestinal microbiota and the gut-associated mucosal immune system that takes place when genetically predisposed individuals are exposed to detrimental environmental triggers. In recent years, the therapeutic dogma in IBD has shifted away from the administration of nonspecific immunosuppressives toward a pathway-based approach. In this review, we present an outlook of IBD treatment based on this new conceptual approach. Firstly, we will provide an overview of the major aspects of IBD pathogenesis with emphasis on specific pathway-based defects. Secondly, we will examine in detail the development of novel therapeutic approaches that can be used to target genetics, dysbiosis, the epithelial barrier, proinflammatory cytokines, and leukocyte trafficking. Most of these strategies are still in the developmental phase, but promising approaches include fecal microbiota transplantation as a means to correct IBD-related dysbiosis; administration of modified phosphatidylcholine to enhance the function of the intestinal mucous and tighten the defective epithelial barrier; the reduction of over-reactive proinflammatory pathways through the blockade of novel, nontumor necrosis factor inflammatory mediators via monoclonal antibodies against the common p40 chain of interleukin (IL-12) and IL-23, Janus kinase inhibitors, or antisense oligonucleotides against inhibitors of the immunosuppressive cytokine transforming growth factor-β1; and finally, inhibition of leukocyte trafficking to the gut via neutralization of the gut-specific α4β7 integrin. Availability of such diverse treatment modalities with specific pathway-based targets will increase the therapeutic options for patients with IBD.
Collapse
Affiliation(s)
- Giorgos Bamias
- First Department of Gastroenterology, Ethnikon and Kapodistriakon University of Athens, Laikon Hospital, Athens, Greece
| | - Theresa T Pizarro
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio; Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio
| | - Fabio Cominelli
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio; Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
39
|
Samuelson DR, Welsh DA, Shellito JE. Regulation of lung immunity and host defense by the intestinal microbiota. Front Microbiol 2015; 6:1085. [PMID: 26500629 PMCID: PMC4595839 DOI: 10.3389/fmicb.2015.01085] [Citation(s) in RCA: 259] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/22/2015] [Indexed: 12/13/2022] Open
Abstract
Every year in the United States approximately 200,000 people die from pulmonary infections, such as influenza and pneumonia, or from lung disease that is exacerbated by pulmonary infection. In addition, respiratory diseases such as, asthma, affect 300 million people worldwide. Therefore, understanding the mechanistic basis for host defense against infection and regulation of immune processes involved in asthma are crucial for the development of novel therapeutic strategies. The identification, characterization, and manipulation of immune regulatory networks in the lung represents one of the biggest challenges in treatment of lung associated disease. Recent evidence suggests that the gastrointestinal (GI) microbiota plays a key role in immune adaptation and initiation in the GI tract as well as at other distal mucosal sites, such as the lung. This review explores the current research describing the role of the GI microbiota in the regulation of pulmonary immune responses. Specific focus is given to understanding how intestinal “dysbiosis” affects lung health.
Collapse
Affiliation(s)
- Derrick R Samuelson
- Section of Pulmonary/Critical Care and Allergy/Immunology, Department of Medicine, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - David A Welsh
- Section of Pulmonary/Critical Care and Allergy/Immunology, Department of Medicine, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Judd E Shellito
- Section of Pulmonary/Critical Care and Allergy/Immunology, Department of Medicine, Louisiana State University Health Sciences Center New Orleans, LA, USA
| |
Collapse
|
40
|
Evans-Marin HL, Cao AT, Yao S, Chen F, He C, Liu H, Wu W, Gonzalez MG, Dann SM, Cong Y. Unexpected Regulatory Role of CCR9 in Regulatory T Cell Development. PLoS One 2015; 10:e0134100. [PMID: 26230654 PMCID: PMC4521878 DOI: 10.1371/journal.pone.0134100] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 07/06/2015] [Indexed: 01/01/2023] Open
Abstract
T cells reactive to microbiota regulate the pathogenesis of inflammatory bowel disease (IBD). As T cell trafficking to intestines is regulated through interactions between highly specific chemokine-chemokine receptors, efforts have been made to develop intestine-specific immunosuppression based on blocking these key processes. CCR9, a gut-trophic chemokine receptor expressed by lymphocytes and dendritic cells, has been implicated in the regulation of IBD through mediating recruitment of T cells to inflamed sites. However, the role of CCR9 in inducing and sustaining inflammation in the context of IBD is poorly understood. In this study, we demonstrate that CCR9 deficiency in effector T cells and Tregs does not affect the development of colitis in a microbiota antigen-specific, T cell-mediated model. However, Treg cells express higher levels of CCR9 compared to those in effector T cells. Interestingly, CCR9 inhibits Treg cell development, in that CCR9-/- mice demonstrate a high level of Foxp3+ Tregs, and ligation of CCR9 by its ligand CCL25 inhibits Treg cell differentiation in vitro. Collectively, our data indicate that in addition to acting as a gut-homing molecule, CCR9 signaling shapes immune responses by inhibiting Treg cell development.
Collapse
Affiliation(s)
- Heather L. Evans-Marin
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Anthony T. Cao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Suxia Yao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Feidi Chen
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Chong He
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Gastroenterology, The Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
| | - Han Liu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Gastroenterology, The Qilu Hospital, Shandong University, Shandong, China
| | - Wei Wu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Gastroenterology, The Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
| | - Maria G. Gonzalez
- School of Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Sara M. Dann
- Department of Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
41
|
Kang SH, Jin BR, Kim HJ, Seo GY, Jang YS, Kim SJ, An SJ, Park SR, Kim WS, Kim PH. Lactoferrin Combined with Retinoic Acid Stimulates B1 Cells to Express IgA Isotype and Gut-homing Molecules. Immune Netw 2015; 15:37-43. [PMID: 25713507 PMCID: PMC4338266 DOI: 10.4110/in.2015.15.1.37] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 01/23/2015] [Accepted: 01/26/2015] [Indexed: 12/03/2022] Open
Abstract
It is well established that TGF-β1 and retinoic acid (RA) cause IgA isotype switching in mice. We recently found that lactoferrin (LF) also has an activity of IgA isotype switching in spleen B cells. The present study explored the effect of LF on the Ig production by mouse peritoneal B cells. LF, like TGF-β1, substantially increased IgA production in peritoneal B1 cells but little in peritoneal B2 cells. In contrast, LF increased IgG2b production in peritoneal B2 cells much more strongly than in peritoneal B1 cells. LF in combination with RA further enhanced the IgA production and, interestingly, this enhancement was restricted to IgA isotype and B1 cells. Similarly, the combination of the two molecules also led to expression of gut homing molecules α4β7 and CCR9 on peritoneal B1 cells, but not on peritoneal B2 cells. Thus, these results indicate that LF and RA can contribute to gut IgA response through stimulating IgA isotype switching and expression of gut-homing molecules in peritoneal B1 cells.
Collapse
Affiliation(s)
- Seong-Ho Kang
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 200-701, Korea
| | - Bo-Ra Jin
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 200-701, Korea
| | - Hyeon-Jin Kim
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 200-701, Korea
| | - Goo-Young Seo
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 200-701, Korea
| | - Young-Saeng Jang
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 200-701, Korea
| | - Sun-Jin Kim
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 200-701, Korea
| | - Sun-Jin An
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 200-701, Korea
| | - Seok-Rae Park
- Department of Microbiology, College of Medicine, Konyang University, Daejeon 302-718, Korea
| | - Woan-Sub Kim
- Department of Animal Life and Environmental Science, College of Agriculture and Life Science, Hankyong National University, Anseong 456-749, Korea
| | - Pyeung-Hyeun Kim
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 200-701, Korea
| |
Collapse
|
42
|
Meinhardt K, Kroeger I, Bauer R, Ganss F, Ovsiy I, Rothamer J, Büttner M, Atreya I, Waldner M, Bittrich M, Lehmann CH, Rieger MA, Beilhack A, Zeiser R, Edinger M, Dudziak D, Mackensen A, Rehli M, Ullrich E. Identification and characterization of the specific murine NK cell subset supporting graft- versus-leukemia- and reducing graft- versus-host-effects. Oncoimmunology 2015; 4:e981483. [PMID: 25949862 PMCID: PMC4368119 DOI: 10.4161/2162402x.2014.981483] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 10/23/2014] [Indexed: 11/19/2022] Open
Abstract
Clinical studies investigating the impact of natural killer (NK) cells in allogeneic hematopoietic stem cell transplantation settings have yielded promising results. However, NK cells are a functionally and phenotypically heterogeneous population. Therefore, we addressed the functional relevance of specific NK cell subsets distinguished by expression of CD117, CD27 and CD11b surface markers in graft-versus-leukemia (GVL)-reaction and graft-versus-host-disease (GVHD). Our results clearly demonstrate that the subset of c-Kit−CD27−CD11b+ NK cells expressed multiple cytotoxic pathway genes and provided optimal graft-versus-leukemia-effects, while significantly reducing T cell proliferation induced by allogeneic dendritic cells. Furthermore, these NK cells migrated to inflamed intestinal tissues where graft-versus-host-colitis was efficiently mitigated. For the first time, we identified the c-Kit−CD27−CD11b+ NK cell population as the specific effector NK cell subset capable of significantly diminishing GVHD in fully mismatched bone marrow transplantation settings. In conclusion, the subset of c-Kit−CD27−CD11b+ NK cells not only supports GVL, but also plays a unique role in the protection against GVHD by migrating to the peripheral GVHD target organs where they exert efficient immunoregulatory activities. These new insights demonstrate the importance of selecting the optimal NK cell subset for cellular immunotherapy following allogeneic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Kathrin Meinhardt
- Department of Internal Medicine 5; Hematology and Oncology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany ; Childrens Hospital; Department of Pediatric Hematology and Oncology; Johann Wolfgang Goethe University ; Frankfurt, Germany ; LOEWE Center for Cell and Gene Therapy; Johann Wolfgang Goethe University ; Frankfurt, Germany
| | - Irena Kroeger
- Department of Internal Medicine 5; Hematology and Oncology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany
| | - Ruth Bauer
- Department of Internal Medicine 5; Hematology and Oncology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany ; Childrens Hospital; Department of Pediatric Hematology and Oncology; Johann Wolfgang Goethe University ; Frankfurt, Germany ; LOEWE Center for Cell and Gene Therapy; Johann Wolfgang Goethe University ; Frankfurt, Germany
| | - Franziska Ganss
- Department of Internal Medicine 5; Hematology and Oncology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany ; Childrens Hospital; Department of Pediatric Hematology and Oncology; Johann Wolfgang Goethe University ; Frankfurt, Germany ; LOEWE Center for Cell and Gene Therapy; Johann Wolfgang Goethe University ; Frankfurt, Germany
| | - Ilja Ovsiy
- Childrens Hospital; Department of Pediatric Hematology and Oncology; Johann Wolfgang Goethe University ; Frankfurt, Germany ; LOEWE Center for Cell and Gene Therapy; Johann Wolfgang Goethe University ; Frankfurt, Germany
| | - Johanna Rothamer
- Department of Internal Medicine 5; Hematology and Oncology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany ; Childrens Hospital; Department of Pediatric Hematology and Oncology; Johann Wolfgang Goethe University ; Frankfurt, Germany ; LOEWE Center for Cell and Gene Therapy; Johann Wolfgang Goethe University ; Frankfurt, Germany
| | - Maike Büttner
- Department of Nephropathology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany
| | - Imke Atreya
- Department of Internal Medicine 1; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany
| | - Maximilian Waldner
- Department of Internal Medicine 1; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany
| | - Max Bittrich
- Department of Internal Medicine 5; Hematology and Oncology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany
| | - Christian Hk Lehmann
- Department of Dermatology; Laboratory of Dendritic Cell Biology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany
| | - Michael A Rieger
- LOEWE Center for Cell and Gene Therapy; Johann Wolfgang Goethe University ; Frankfurt, Germany ; Department of Hematology/Oncology; Johann Wolfgang Goethe University ; Frankfurt, Germany
| | - Andreas Beilhack
- IZKF Research Group for Experimental Stem Cell Transplantation; Department of Medicine II; University Hospital ; Würzburg, Germany
| | - Robert Zeiser
- Division of Hematology and Oncology; Department of Medicine; Freiburg University Medical Center; Albert-Ludwigs-University ; Freiburg, Germany
| | - Matthias Edinger
- Department of Internal Medicine 3; University Hospital Regensburg ; Regensburg; Germany ; Regensburg Center for Interventional Immunology (RCI); University Regensburg ; Regensburg, Germany
| | - Diana Dudziak
- Department of Dermatology; Laboratory of Dendritic Cell Biology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5; Hematology and Oncology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany
| | - Michael Rehli
- Department of Internal Medicine 3; University Hospital Regensburg ; Regensburg; Germany ; Regensburg Center for Interventional Immunology (RCI); University Regensburg ; Regensburg, Germany
| | - Evelyn Ullrich
- Department of Internal Medicine 5; Hematology and Oncology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany ; Childrens Hospital; Department of Pediatric Hematology and Oncology; Johann Wolfgang Goethe University ; Frankfurt, Germany ; LOEWE Center for Cell and Gene Therapy; Johann Wolfgang Goethe University ; Frankfurt, Germany
| |
Collapse
|
43
|
Abstract
BACKGROUND Tight junction proteins (TJPs) and dendritic cells (DC) are critical in the pathogenesis of inflammatory bowel diseases. The ileal pouch formed by restorative proctocolectomy provides a unique human model for studying the pathogenesis of inflammatory bowel diseases. Data implicate the microbiota in the pathogenesis of pouchitis, while the role of innate immune factors remains unclear. We performed longitudinal and cross-sectional studies of patients after restorative proctocolectomy and assessed TJP and DC characteristics in the ileal pouch. METHODS Mucosal biopsies were taken from the ileal pouch of patients with ulcerative colitis (UC) and familial adenomatous polyposis (n = 8). Of patients with UC, one group (n = 5) was followed longitudinally over the first year after ileostomy closure, another group had pouchitis (n = 15), and another group no inflammation (n = 18). Dendritic cell phenotype and epithelial cell TJP expression were assessed using flow cytometric analysis. RESULTS Increased epithelial expression of the "pore-forming" TJP claudin 2, and DC expression of gut-homing markers CCR 9 and integrin β7, occurred early after ileostomy closure. In patients with UC with pouchitis, epithelial expression of ZO-1 and claudin 1 were reduced, DC were activated with increased CD40, and Toll-like receptor 4 expression increased. In pouchitis, DC expressing CCR 9 were decreased, whereas DC expressing β7 increased. CONCLUSIONS Abnormalities were found in TJP expression in the pouch of patients with UC, in particular, increased expression of the pore-forming claudin 2 as an early event in the development of pouch inflammation and an aberrant DC phenotype was characterized in the ileal pouch of patients with UC.
Collapse
|
44
|
Cook KW, Letley DP, Ingram RJM, Staples E, Skjoldmose H, Atherton JC, Robinson K. CCL20/CCR6-mediated migration of regulatory T cells to the Helicobacter pylori-infected human gastric mucosa. Gut 2014; 63:1550-9. [PMID: 24436142 PMCID: PMC4173663 DOI: 10.1136/gutjnl-2013-306253] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Helicobacter pylori-induced peptic ulceration is less likely to occur in patients with a strong gastric anti-inflammatory regulatory T cell (Treg) response. Migration of Tregs into the gastric mucosa is therefore important. OBJECTIVE To identify the homing receptors involved in directing Tregs to the gastric mucosa, and investigate how H pylori stimulates the relevant chemokine responses. DESIGN Gastric biopsy samples and peripheral blood were donated by 84 H pylori-infected and 46 uninfected patients. Luminex assays quantified gastric biopsy chemokine concentrations. Flow cytometry was used to characterise homing receptors on CD4(+)CD25(hi) Tregs. H pylori wild-type and isogenic mutants were used to investigate the signalling mechanisms behind CCL20 and IL-8 induction in gastric epithelial cell lines. Transwell assays were used to quantify Treg migration towards chemokines in vitro. RESULTS CCL20, CXCL1-3 and IL-8 concentrations were significantly increased in gastric biopsy samples from H pylori-infected patients. CCR6 (CCL20 receptor), CXCR1 and CXCR2 (IL-8 and CXCL1-3 receptors) were expressed by a higher proportion of peripheral blood Tregs in infected patients. Most gastric Tregs expressed these receptors. H pylori induced CCL20 production by gastric epithelial cells via cag pathogenicity island (cagPAI)-dependent NF-κB signalling. Foxp3(+), but not Foxp3(-), CD4 cells from infected mice migrated towards recombinant CCL20 in vitro. CONCLUSIONS As well as increasing Treg numbers, H pylori infection induces a change in their characteristics. Expression of CCR6, CXCR1 and CXCR2 probably enables their migration towards CCL20 and IL-8 in the infected gastric mucosa. Such qualitative changes may also explain how H pylori protects against some extragastric inflammatory disorders.
Collapse
Affiliation(s)
- Katherine W Cook
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, The University of Nottingham, Nottingham, UK
| | - Darren P Letley
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, The University of Nottingham, Nottingham, UK
| | - Richard J M Ingram
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, The University of Nottingham, Nottingham, UK
| | - Emily Staples
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, The University of Nottingham, Nottingham, UK
| | - Helle Skjoldmose
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, The University of Nottingham, Nottingham, UK
| | - John C Atherton
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, The University of Nottingham, Nottingham, UK
| | - Karen Robinson
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, The University of Nottingham, Nottingham, UK
| |
Collapse
|
45
|
Gómez-Pérez GP, van Bruggen R, Grobusch MP, Dobaño C. Plasmodium falciparum malaria and invasive bacterial co-infection in young African children: the dysfunctional spleen hypothesis. Malar J 2014; 13:335. [PMID: 25158979 PMCID: PMC4161853 DOI: 10.1186/1475-2875-13-335] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 08/21/2014] [Indexed: 12/25/2022] Open
Abstract
Children with recent or acute malaria episodes are at increased risk of invasive bacterial infections (IBI). However, the exact nature of the malaria-IBI association is still unclear. Young children have an age-related spleen immunologic immaturity, mainly due to the still ongoing development of the marginal zone (MZ) B cell subset. By mounting a rapid antibody response against encapsulated bacteria, these cells are critical for the defence against highly pathogenic microorganisms that do not elicit classical T cell-dependent responses. There is increasing evidence that the anatomy of the spleen becomes disorganized during malaria infection, with complete dissolution of the MZ and apoptosis of MZ B cells. Correspondingly, a reduction in the frequency of the peripheral equivalent of the MZ B cells has been found in malaria endemic areas. A remarkable similarity exists in IBI susceptibility between African children with malaria and hyposplenic or splenectomized patients. However, studies specifically assessing the immune function of the spleen in controlling bacterial infections in young children with malaria are scarce. Here, it is hypothesized that Plasmodium falciparum malaria infection constitutes a detrimental factor in the still immature spleen function of young children, resulting in a factually hyposplenic state during malaria episodes, putting children with malaria at a high risk to develop life-threatening bacterial infections. Studies to confirm or reject this hypothesis are greatly needed, as well as the development of affordable and feasible tools to assess the immune spleen function against encapsulated bacteria in children with malaria.
Collapse
Affiliation(s)
- Gloria P Gómez-Pérez
- Barcelona Centre for International Health Research (CRESIB, Hospital Clínic-Universitat de Barcelona), Barcelona 08036, Spain.
| | | | | | | |
Collapse
|
46
|
Endoscopic photoconversion reveals unexpectedly broad leukocyte trafficking to and from the gut. Proc Natl Acad Sci U S A 2014; 111:6696-701. [PMID: 24753589 DOI: 10.1073/pnas.1405634111] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Given mounting evidence of the importance of gut-microbiota/immune-cell interactions in immune homeostasis and responsiveness, surprisingly little is known about leukocyte movements to, and especially from, the gut. We address this topic in a minimally perturbant manner using Kaede transgenic mice, which universally express a photoconvertible fluorescent reporter. Transcutaneous exposure of the cervical lymph nodes to violet light permitted punctual tagging of immune cells specifically therein, and subsequent monitoring of their immigration to the intestine; endoscopic flashing of the descending colon allowed specific labeling of intestinal leukocytes and tracking of their emigration. Our data reveal an unexpectedly broad movement of leukocyte subsets to and from the gut at steady state, encompassing all lymphoid and myeloid populations examined. Nonetheless, different subsets showed different trafficking proclivities (e.g., regulatory T cells were more restrained than conventional T cells in their exodus from the cervical lymph nodes). The novel endoscopic approach enabled us to evidence gut-derived Th17 cells in the spleens of K/BxN mice at the onset of their genetically determined arthritis, thereby furnishing a critical mechanistic link between the intestinal microbiota, namely segmented filamentous bacteria, and an extraintestinal autoinflammatory disease.
Collapse
|
47
|
Cárdeno A, Magnusson MK, Strid H, Alarcón de La Lastra C, Sánchez-Hidalgo M, Ohman L. The unsaponifiable fraction of extra virgin olive oil promotes apoptosis and attenuates activation and homing properties of T cells from patients with inflammatory bowel disease. Food Chem 2014; 161:353-60. [PMID: 24837962 DOI: 10.1016/j.foodchem.2014.04.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Revised: 03/05/2014] [Accepted: 04/02/2014] [Indexed: 10/25/2022]
Abstract
The unsaponifiable fraction (UF) of extra virgin olive oil (EVOO) possesses anti-inflammatory properties and exerts preventative effects in murine models of inflammatory bowel disease (IBD). The present study was designed to determine the in vitro effects of UF on blood and intestinal T cells from IBD patients and healthy subjects. The T cell phenotype was investigated by flow cytometry and cytokine secretion was determined by ELISA. The presence of UF of EVOO promoted apoptosis and attenuated activation of intestinal and blood T cells isolated from IBD patients, decreasing the frequency of CD69(+) and CD25(+) T cells and, also, the secretion of IFN-γ. Moreover, UF reduced the expression of the gut homing receptor integrin β7 on blood T cells from IBD patients. In conclusion, UF modulates the activity and the gut homing capacity of T cells, and might therefore be considered as a dietary complement with an anti-inflammatory role in IBD patients.
Collapse
Affiliation(s)
- A Cárdeno
- Dept. Microbiology and Immunology, Sahlgrenska Academy at the University of Gothenburg, Sweden; Dept. Pharmacology, School of Pharmacy, University of Seville, Spain
| | - M K Magnusson
- Dept. Microbiology and Immunology, Sahlgrenska Academy at the University of Gothenburg, Sweden; Dept. Internal Medicine and Clinical Nutrition, Sahlgrenska Academy at the University of Gothenburg, Sweden
| | - H Strid
- Dept. Internal Medicine and Clinical Nutrition, Sahlgrenska Academy at the University of Gothenburg, Sweden
| | | | - M Sánchez-Hidalgo
- Dept. Pharmacology, School of Pharmacy, University of Seville, Spain
| | - L Ohman
- Dept. Microbiology and Immunology, Sahlgrenska Academy at the University of Gothenburg, Sweden; Dept. Internal Medicine and Clinical Nutrition, Sahlgrenska Academy at the University of Gothenburg, Sweden.
| |
Collapse
|
48
|
Abstract
Inflammatory bowel disease (IBD) is an intestinal inflammatory disease of unknown etiology with two main distinguishable entities, Crohn's disease (CD) and ulcerative colitis (UC). Current diagnosis of IBD relies on the clinical, endoscopic, radiological, histological and biochemical features, but these approaches have shortcomings, especially in cases of overlapping symptoms of CD and UC. The detection of serological markers can improve the diagnosis of IBD, because an aberrant immune response against microbial or endogenous antigens in a genetically susceptible host seems to be implicated in IBD pathogenesis. Current evidence suggests that the detection of serum immunology markers is useful in differential diagnosis of CD versus UC and can be a valuable aid in stratifying patients according to disease phenotype and risk of complications.
Collapse
|
49
|
Buckner CM, Moir S, Kardava L, Ho J, Santich BH, Kim LJY, Funk EK, Nelson AK, Winckler B, Chairez CL, Theobald-Whiting NL, Anaya-O'Brien S, Alimchandani M, Quezado MM, Yao MD, Kovacs JA, Chun TW, Fauci AS, Malech HL, De Ravin SS. CXCR4/IgG-expressing plasma cells are associated with human gastrointestinal tissue inflammation. J Allergy Clin Immunol 2013; 133:1676-85.e5. [PMID: 24373354 DOI: 10.1016/j.jaci.2013.10.050] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 09/16/2013] [Accepted: 10/14/2013] [Indexed: 12/30/2022]
Abstract
BACKGROUND We previously reported abnormalities in circulating B cells in patients with chronic granulomatous disease (CGD) and those with HIV infection. Gastrointestinal complications are common to both diseases and likely involve perturbation of immune cells, including plasma cells (PCs). IgA is the most abundant immunoglobulin in the human body, with roles in protection and maintenance of intestinal homeostasis. IgA is produced primarily by PCs residing in mucosal tissues that are also thought to circulate in the blood. OBJECTIVE We sought to characterize and compare PCs in patients with infectious (HIV) and noninfectious (CGD and Crohn disease) diseases that have been associated with intestinal inflammation. METHODS Phenotypic and transcriptional analyses were performed on cells isolated from the blood and colon. RESULTS IgA-secreting CCR10-expressing PCs predominated in the guts of healthy subjects, whereas in patients with HIV, CGD, and Crohn disease, there was a significant increase in the proportion of IgG-secreting PCs. Where intestinal inflammation was present, IgG-secreting PCs expressed reduced levels of CCR10 and increased levels of CXCR4. The intensity of CXCR4 expression correlated with the frequency of IgG-expressing PCs and the frequency of CXCR4(+)/IgG(+) PCs was associated with the severity of intestinal inflammatory disease yet distinct from PCs and plasmablasts circulating in the blood. CONCLUSIONS These findings suggest that regardless of the underlying disease, the presence of CXCR4(+)/IgG(+) PCs in the gut is a strong yet localized indicator of intestinal inflammation. Furthermore, our findings suggest that CXCR4(+)/IgG(+) PCs might play a role in immune cell homeostasis during inflammatory processes of the gut.
Collapse
Affiliation(s)
- Clarisa M Buckner
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Md.
| | - Lela Kardava
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Jason Ho
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Brian H Santich
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Leo Jin Young Kim
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Emily K Funk
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Amy K Nelson
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Britanny Winckler
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Cheryl L Chairez
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Narda L Theobald-Whiting
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Sandra Anaya-O'Brien
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | | | - Martha M Quezado
- Laboratory of Pathology, National Cancer Institute, Bethesda, Md
| | - Michael D Yao
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Joseph A Kovacs
- Critical Care Medicine, National Institutes of Health Clinical Center, Bethesda, Md
| | - Tae-Wook Chun
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Anthony S Fauci
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Harry L Malech
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Suk See De Ravin
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, Bethesda, Md.
| |
Collapse
|
50
|
Valatas V, Vakas M, Kolios G. The value of experimental models of colitis in predicting efficacy of biological therapies for inflammatory bowel diseases. Am J Physiol Gastrointest Liver Physiol 2013; 305:G763-85. [PMID: 23989010 DOI: 10.1152/ajpgi.00004.2013] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
During the last decade, biological therapies have an increasing share in the modern therapeutics of various diseases including inflammatory bowel diseases (IBD). Animal models of IBD have often been used to identify the targets of biological therapies, to test their relevance to disease pathogenesis, to assess their therapeutic efficacy in vivo, and to check for drug toxicity. In the field of inflammatory diseases the majority of biologics under development have failed to reach the clinic. This review examines the ability of preclinical data from animal models of IBD to predict success or failure of biologics in human IBD. Specifically, it describes the murine models of IBD, the mechanism of disease induction, the phenotype of the disease, its relevance to human IBD, and the specific immunological features of disease pathogenesis in each model and mainly compares the results of the phase II and III trials of biologics in IBD with preclinical data obtained from studies in animal models. Finally, it examines the possible reasons for low success in translation from bench to bedside and offers some suggestions to improve translation rates.
Collapse
Affiliation(s)
- Vassilis Valatas
- Dept. of Gastroenterology, Univ. Hospital of Heraklion, PO Box 1352, Voutes, Heraklion, GR-71100, Crete, Greece.
| | | | | |
Collapse
|