1
|
Saleki K, Alijanizadeh P, Javanmehr N, Rezaei N. The role of Toll-like receptors in neuropsychiatric disorders: Immunopathology, treatment, and management. Med Res Rev 2024; 44:1267-1325. [PMID: 38226452 DOI: 10.1002/med.22012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 10/20/2023] [Accepted: 12/20/2023] [Indexed: 01/17/2024]
Abstract
Neuropsychiatric disorders denote a broad range of illnesses involving neurology and psychiatry. These disorders include depressive disorders, anxiety, schizophrenia, bipolar disorder, attention deficit hyperactivity disorder, autism spectrum disorders, headaches, and epilepsy. In addition to their main neuropathology that lies in the central nervous system (CNS), lately, studies have highlighted the role of immunity and neuroinflammation in neuropsychiatric disorders. Toll-like receptors (TLRs) are innate receptors that act as a bridge between the innate and adaptive immune systems via adaptor proteins (e.g., MYD88) and downstream elements; TLRs are classified into 13 families that are involved in normal function and illnesses of the CNS. TLRs expression affects the course of neuropsychiatric disorders, and is influenced during their pharmacotherapy; For example, the expression of multiple TLRs is normalized during the major depressive disorder pharmacotherapy. Here, the role of TLRs in neuroimmunology, treatment, and management of neuropsychiatric disorders is discussed. We recommend longitudinal studies to comparatively assess the cell-type-specific expression of TLRs during treatment, illness progression, and remission. Also, further research should explore molecular insights into TLRs regulation and related pathways.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
- Department of e-Learning, Virtual School of Medical Education and Management, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Parsa Alijanizadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Nima Javanmehr
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
2
|
Feng S, Zhang C, Chen S, He R, Chao G, Zhang S. TLR5 Signaling in the Regulation of Intestinal Mucosal Immunity. J Inflamm Res 2023; 16:2491-2501. [PMID: 37337514 PMCID: PMC10276996 DOI: 10.2147/jir.s407521] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/23/2023] [Indexed: 06/21/2023] Open
Abstract
Toll-like receptor 5 (TLR5) is a pattern recognition receptor that specifically recognizes flagellin and consequently plays a crucial role in the control of intestinal homeostasis by activating innate and adaptive immune responses. TLR5 overexpression, on the other hand, might disrupt the intestinal mucosal barrier, which serves as the first line of defense against harmful microbes. The intestine symbiotic bacteria, mucous layer, intestinal epithelial cells (IECs), adherens junctions (such as tight junctions and peripheral membrane proteins), the intestinal mucosal immune system, and cytokines make up the intestinal mucosal barrier. Impaired barrier function has been linked to intestinal illnesses such as inflammatory bowel disease (IBD). IBD is a persistent non-specific inflammatory illness of the digestive system with an unknown cause. It is now thought to be linked to infection, environment, genes, immune system, and the gut microbiota. The significance of immunological dysfunction in IBD has received more attention in recent years. The purpose of this paper is to explore TLR5's position in the intestinal mucosal barrier and its relevance to IBD.
Collapse
Affiliation(s)
- Shuyan Feng
- Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Chi Zhang
- Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Shanshan Chen
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, People’s Republic of China
| | - Ruonan He
- Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Guanqun Chao
- Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou, 310018, People’s Republic of China
| | - Shuo Zhang
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, People’s Republic of China
| |
Collapse
|
3
|
Ciaston I, Dobosz E, Potempa J, Koziel J. The subversion of toll-like receptor signaling by bacterial and viral proteases during the development of infectious diseases. Mol Aspects Med 2022; 88:101143. [PMID: 36152458 PMCID: PMC9924004 DOI: 10.1016/j.mam.2022.101143] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/29/2022] [Accepted: 09/09/2022] [Indexed: 02/05/2023]
Abstract
Toll-like receptors (TLRs) are pattern recognition receptors (PRRs) that respond to pathogen-associated molecular patterns (PAMPs). The recognition of specific microbial ligands by TLRs triggers an innate immune response and also promotes adaptive immunity, which is necessary for the efficient elimination of invading pathogens. Successful pathogens have therefore evolved strategies to subvert and/or manipulate TLR signaling. Both the impairment and uncontrolled activation of TLR signaling can harm the host, causing tissue destruction and allowing pathogens to proliferate, thus favoring disease progression. In this context, microbial proteases are key virulence factors that modify components of the TLR signaling pathway. In this review, we discuss the role of bacterial and viral proteases in the manipulation of TLR signaling, highlighting the importance of these enzymes during the development of infectious diseases.
Collapse
Affiliation(s)
- Izabela Ciaston
- Department of Microbiology Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Ewelina Dobosz
- Department of Microbiology Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jan Potempa
- Department of Microbiology Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland; Department of Oral Health and Systemic Disease, University of Louisville School of Dentistry, University of Louisville, Louisville, KY, USA.
| | - Joanna Koziel
- Department of Microbiology Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
4
|
Xia P, Lian S, Wu Y, Yan L, Quan G, Zhu G. Zinc is an important inter-kingdom signal between the host and microbe. Vet Res 2021; 52:39. [PMID: 33663613 PMCID: PMC7931793 DOI: 10.1186/s13567-021-00913-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 02/08/2021] [Indexed: 12/15/2022] Open
Abstract
Zinc (Zn) is an essential trace element in living organisms and plays a vital role in the regulation of both microbial virulence and host immune responses. A growing number of studies have shown that zinc deficiency or the internal Zn concentration does not meet the needs of animals and microbes, leading to an imbalance in zinc homeostasis and intracellular signalling pathway dysregulation. Competition for zinc ions (Zn2+) between microbes and the host exists in the use of Zn2+ to maintain cell structure and physiological functions. It also affects the interplay between microbial virulence factors and their specific receptors in the host. This review will focus on the role of Zn in the crosstalk between the host and microbe, especially for changes in microbial pathogenesis and nociceptive neuron-immune interactions, as it may lead to new ways to prevent or treat microbial infections.
Collapse
Affiliation(s)
- Pengpeng Xia
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, Yangzhou, 225009, China. .,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China. .,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China.
| | - Siqi Lian
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, Yangzhou, 225009, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China
| | - Yunping Wu
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, Yangzhou, 225009, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China
| | - Li Yan
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, Yangzhou, 225009, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China
| | - Guomei Quan
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, Yangzhou, 225009, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China
| | - Guoqiang Zhu
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, Yangzhou, 225009, China. .,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China. .,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
5
|
Severity of Experimental Autoimmune Uveitis Is Reduced by Pretreatment with Live Probiotic Escherichia coli Nissle 1917. Cells 2020; 10:cells10010023. [PMID: 33375578 PMCID: PMC7823395 DOI: 10.3390/cells10010023] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/16/2020] [Accepted: 12/22/2020] [Indexed: 02/07/2023] Open
Abstract
Non-infectious uveitis is considered an autoimmune disease responsible for a significant burden of blindness in developed countries and recent studies have linked its pathogenesis to dysregulation of the gut microbiota. We tested the immunomodulatory properties of two probiotics, Escherichia coli Nissle 1917 (EcN) and E. coli O83:K24:H31 (EcO), in a model of experimental autoimmune uveitis (EAU). To determine the importance of bacterial viability and treatment timing, mice were orally treated with live or autoclaved bacteria in both preventive and therapeutic schedules. Disease severity was assessed by ophthalmoscopy and histology, immune phenotypes in mesenteric and cervical lymph nodes were analyzed by flow cytometry and the gut immune environment was analyzed by RT-PCR and/or gut tissue culture. EcN, but not EcO, protected against EAU but only as a live organism and only when administered before or at the time of disease induction. Successful prevention of EAU was accompanied by a decrease in IRBP-specific T cell response in the lymph nodes draining the site of immunization as early as 7 days after the immunization and eye-draining cervical lymph nodes when the eye inflammation became apparent. Furthermore, EcN promoted an anti-inflammatory response in Peyer’s patches, increased gut antimicrobial peptide expression and decreased production of inducible nitric oxide synthase in macrophages. In summary, we show here that EcN controls inflammation in EAU and suggest that probiotics may have a role in regulating the gut–eye axis.
Collapse
|
6
|
Rasaei R, Sarodaya N, Kim KS, Ramakrishna S, Hong SH. Importance of Deubiquitination in Macrophage-Mediated Viral Response and Inflammation. Int J Mol Sci 2020; 21:ijms21218090. [PMID: 33138315 PMCID: PMC7662591 DOI: 10.3390/ijms21218090] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 01/14/2023] Open
Abstract
Ubiquitination and deubiquitination play a fundamental role in the signaling pathways associated with innate and adaptive immune responses. Macrophages are key sentinels for the host defense, triggering antiviral and inflammatory responses against various invading pathogens. Macrophages recognize the genetic material of these pathogens as pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs) through the activation of its pattern recognition receptors (PRRs), initiating the cascade of immune signaling, which leads to the production of pro- and anti-inflammatory cytokines that initiates the appropriate immune response. Macrophage-mediated immune response is highly regulated and tightly controlled by the ubiquitin system since its abnormal activation or dysregulation may result in the severe pathogenesis of numerous inflammatory and autoimmune diseases. Deubiquitinating enzymes (DUBs) play a crucial role in reversing the ubiquitination and controlling the magnitude of the immune response. During infection, pathogens manipulate the host defense system by regulating DUBs to obtain nutrients and increase proliferation. Indeed, the regulation of DUBs by small molecule inhibitors has been proposed as an excellent way to control aberrant activation of immune signaling molecules. This review is focused on the complex role of DUBs in macrophage-mediated immune response, exploring the potential use of DUBs as therapeutic targets in autoimmune and inflammatory diseases by virtue of small molecule DUB inhibitors.
Collapse
Affiliation(s)
- Roya Rasaei
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea;
| | - Neha Sarodaya
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (N.S.); (K.-S.K.)
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (N.S.); (K.-S.K.)
- College of Medicine, Hanyang University, Seoul 04763, Korea
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (N.S.); (K.-S.K.)
- College of Medicine, Hanyang University, Seoul 04763, Korea
- Correspondence: or (S.R.); or (S.-H.H.)
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea;
- Correspondence: or (S.R.); or (S.-H.H.)
| |
Collapse
|
7
|
Differential miRNA-Gene Expression in M Cells in Response to Crohn's Disease-Associated AIEC. Microorganisms 2020; 8:microorganisms8081205. [PMID: 32784656 PMCID: PMC7466023 DOI: 10.3390/microorganisms8081205] [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: 07/15/2020] [Revised: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 12/21/2022] Open
Abstract
Adherent-invasive Escherichia coli (AIEC), which abnormally colonize the ileal mucosa of Crohn’s disease (CD) patients, are able to invade intestinal epithelial cells (IECs) and translocate through M cells overlying Peyer’s patches. The levels of microRNA (miRNA) and gene expression in IECs and M cells upon AIEC infection have not been investigated. Here, we used human intestinal epithelial Caco-2 monolayers and an in vitro M-cell model of AIEC translocation to analyze comprehensive miRNA and gene profiling under basal condition and upon infection with the reference AIEC LF82 strain. Our results showed that AIEC LF82 translocated through M cells but not Caco-2 monolayers. Both differential gene expression and miRNA profile in M cells compared to Caco-2 cells were obtained. In addition, AIEC infection induces changes in gene and miRNA profiles in both Caco-2 and M cells. In silico analysis showed that certain genes dysregulated upon AIEC infection were potential targets of AIEC-dysregulated miRNAs, suggesting a miRNA-mediated regulation of gene expression during AIEC infection in Caco-2, as well as M cells. This study facilitates the discovery of M cell-specific and AIEC response-specific gene-miRNA signature and enhances the molecular understanding of M cell biology under basal condition and in response to infection with CD-associated AIEC.
Collapse
|
8
|
Mansilla F, Takagi M, Garcia-Castillo V, Aso H, Nader-Macias ME, Vignolo G, Kitazawa H, Villena J. Modulation of Toll-like receptor-mediated innate immunity in bovine intestinal epithelial cells by lactic acid bacteria isolated from feedlot cattle. Benef Microbes 2020; 11:269-282. [PMID: 32363914 DOI: 10.3920/bm2019.0189] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The ability of lactobacilli isolated from feedlot cattle environment to differentially modulate the innate immune response triggered by Toll-like receptors (TLRs) activation in bovine intestinal epithelial (BIE) cells was evaluated. BIE cells were stimulated with Lactobacillus mucosae CRL2069, Lactobacillus acidophilus CRL2074, Lactobacillus fermentum CRL2085 or Lactobacillus rhamnosus CRL2084 and challenged with heat-stable pathogen associated molecular patterns (PAMPs) from enterotoxigenic Escherichia coli (ETEC) to induce the activation of TLR4 or with polyinosinic:polycytidylic acid (poly(I:C)) to activate TLR3. Type I interferons, cytokines, chemokines and negative regulators of TLR signalling were studied by RT-PCR. L. mucosae CRL2069 significantly reduced the expression of interleukin (IL)-8 and monocyte chemoattractant protein (MCP)-1 in BIE cells in the context of TLR3 activation. L. mucosae CRL2069 also reduced the expression of tumour necrosis factor-α, IL-β, MCP-1, and IL-8 in heat-stable ETEC PAMPs-challenged BIE cells. In addition, reduced expressions of IL-6, MCP-1, and IL-8 were found in BIE cells stimulated with L. rhamnosus CRL2084, although its effect was significantly lower than that observed for the CRL2069 strain. The reduced levels of pro-inflammatory factors in BIE cells induced by the CRL2069 and CRL2085 strains was related to their ability of increasing the expression of TLR negative regulators. L. mucosae CRL2069 significantly improved the expression of A20-binding inhibitor of NFκ-B activation 3 (ABIN-3), interleukin-1 receptor-associated kinase M (IRAK-M) and mitogen-activated protein kinase 1 (MKP-1) while L. rhamnosus CRL2084 augmented ABIN-3 expression in BIE cells. The results of this work suggest that among the studied strains, L. mucosae CRL2069 was able to regulate TLR3-mediated innate immune response and showed a remarkable capacity to modulate TLR4-mediated inflammation in BIE cells. The CRL2069 strain induce the up-regulation of three TLR negative regulators that would influence nuclear factor kB and mitogen-activated protein kinases signalling pathways while reducing the expression of pro-inflammatory cytokines and chemokines. Therefore, L. mucosae CRL2069 is an interesting immunobiotic candidate for the protection of the bovine host against TLR-mediated intestinal inflammatory damage.
Collapse
Affiliation(s)
- F Mansilla
- Reference Centre for Lactobacilli (CERELA-CONICET), Chacabuco 145, San Miguel de Tucuman, 4000 Tucuman, Argentina
| | - M Takagi
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki Aza Aoba, Aoba-ku, Sendai, Miyagi, 980-8572, Japan
| | - V Garcia-Castillo
- Reference Centre for Lactobacilli (CERELA-CONICET), Chacabuco 145, San Miguel de Tucuman, 4000 Tucuman, Argentina.,Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki Aza Aoba, Aoba-ku, Sendai, Miyagi, 980-8572, Japan
| | - H Aso
- Cell Biology Laboratory, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki Aza Aoba, Aoba-ku, Sendai, Miyagi, 980-8572, Japan.,Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki Aza Aoba, Aoba-ku, Sendai, Miyagi, 980-8572, Japan
| | - M E Nader-Macias
- Reference Centre for Lactobacilli (CERELA-CONICET), Chacabuco 145, San Miguel de Tucuman, 4000 Tucuman, Argentina
| | - G Vignolo
- Reference Centre for Lactobacilli (CERELA-CONICET), Chacabuco 145, San Miguel de Tucuman, 4000 Tucuman, Argentina
| | - H Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki Aza Aoba, Aoba-ku, Sendai, Miyagi, 980-8572, Japan.,Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki Aza Aoba, Aoba-ku, Sendai, Miyagi, 980-8572, Japan
| | - J Villena
- Reference Centre for Lactobacilli (CERELA-CONICET), Chacabuco 145, San Miguel de Tucuman, 4000 Tucuman, Argentina.,Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki Aza Aoba, Aoba-ku, Sendai, Miyagi, 980-8572, Japan
| |
Collapse
|
9
|
Al Nabhani Z, Eberl G. Imprinting of the immune system by the microbiota early in life. Mucosal Immunol 2020; 13:183-189. [PMID: 31988466 DOI: 10.1038/s41385-020-0257-y] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 02/04/2023]
Abstract
The ontogeny and maturation of the immune system is modulated by the microbiota. During fetal life, the mother's microbiota produces compounds that are transferred to the fetus and offspring, and enhance the generation of innate immune cells. After birth, the colonizing microbiota induces the development of intestinal lymphoid tissues and maturation of myeloid and lymphoid cells, and imprints the immune system with a reactivity level that persists long after weaning into adulthood. When the cross-talk between host and microbiota is perturbed early in life, a pathological imprinting may develop that is characterized by excessive immune reactivity in adulthood, which translates into increased susceptibility to inflammatory pathologies. In this review, we discuss the recent data that demonstrate the existence of a time window of opportunity early in life during which mice and human have to be exposed to microbiota in order to develop a balanced immune system. We also discuss the factors involved in imprinting, such as the microbiota, immune cells and stromal cells, as well as the nature of imprinting.
Collapse
Affiliation(s)
- Ziad Al Nabhani
- Microenvironment and Immunity Unit, Institut Pasteur, INSERM U1224, Paris, France
| | - Gérard Eberl
- Microenvironment and Immunity Unit, Institut Pasteur, INSERM U1224, Paris, France.
| |
Collapse
|
10
|
Kanmani P, Kim H. Functional capabilities of probiotic strains on attenuation of intestinal epithelial cell inflammatory response induced by TLR4 stimuli. Biofactors 2019; 45:223-235. [PMID: 30537409 DOI: 10.1002/biof.1475] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/10/2018] [Accepted: 10/12/2018] [Indexed: 12/28/2022]
Abstract
Intestinal epithelial cells (IECs) respond to intruders and their cellular molecules by displaying inflammatory state that can be abrogated by probiotics. However, the molecular mechanisms underlying the beneficial activity of probiotic strains have yet to be elucidated. This study was conducted to investigate whether probiotic strains have immunoregulatory effects in IECs, and how they respond to bacterial lipopolysaccharide (LPS) in vitro. Caco2 cells were stimulated with LABs and followed by LPS. The expression of different cytokines that involved in toll-like receptor (TLR) signaling was analyzed. Caco2 cells that were exposed to LPS showed upregulated expression of IL-6, CXCL8, CCL2, and BPI that were counteracted by LAB strains through the modulation of TLR negative regulators (A20, Tollip, SIGIRR, and IRAKM), p38 MAPK and p65 NF-κB signaling. Lactobacillus plantarum, L. farciminis, and L. pentosus unveiled better activity as compared to other strains. Moreover, LAB strains were the potent inducers of immunoregulatory cytokines in coculture system. The expression of IL-10 and TGF-β were found to be higher as compared with LPS. Conversely, TNF-α at the protein level was dampened by LAB strains in both the apical and basolateral compartments. Collectively, our results demonstrated that the selected LAB strains exert profound immunoregulatory effects in response to LPS on IECs; however, further studies in vivo and in clinical settings are important to corroborate these effects. © 2018 BioFactors, 45(2):223-235, 2019.
Collapse
Affiliation(s)
- Paulraj Kanmani
- Department of Rehabilitation Medicine of Korean Medicine, Dongguk University Ilsan Hospital, Goyang, Gyeonggi, Republic of Korea
| | - Hojun Kim
- Department of Rehabilitation Medicine of Korean Medicine, Dongguk University Ilsan Hospital, Goyang, Gyeonggi, Republic of Korea
| |
Collapse
|
11
|
Li Y, Mooney EC, Holden SE, Xia XJ, Cohen DJ, Walsh SW, Ma A, Sahingur SE. A20 Orchestrates Inflammatory Response in the Oral Mucosa through Restraining NF-κB Activity. THE JOURNAL OF IMMUNOLOGY 2019; 202:2044-2056. [PMID: 30760622 DOI: 10.4049/jimmunol.1801286] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 01/16/2019] [Indexed: 12/15/2022]
Abstract
Deregulated immune response to a dysbiotic resident microflora within the oral cavity leads to chronic periodontal disease, local tissue destruction, and various systemic complications. To preserve tissue homeostasis, inflammatory signaling pathways involved in the progression of periodontitis must be tightly regulated. A20 (TNFAIP3), a ubiquitin-editing enzyme, has emerged as one of the key regulators of inflammation. Yet, the function of A20 in the oral mucosa and the biological pathways in which A20 mitigates periodontal inflammation remain elusive. Using a combination of in vivo and ex vivo disease models, we report in this study that A20 regulates inflammatory responses to a keystone oral bacterium, Porphyromonas gingivalis, and restrains periodontal inflammation through its effect on NF-κB signaling and cytokine production. Depletion of A20 using gene editing in human macrophage-like cells (THP-1) significantly increased cytokine secretion, whereas A20 overexpression using lentivirus infection dampened the cytokine production following bacterial challenge through modulating NF-κB activity. Similar to human cells, bone marrow-derived macrophages from A20-deficient mice infected with P. gingivalis displayed increased NF-κB activity and cytokine production compared with the cells isolated from A20-competent mice. Subsequent experiments using a murine ligature-induced periodontitis model showed that even a partial loss of A20 promotes an increased inflammatory phenotype and more severe bone loss, further verifying the critical function of A20 in the oral mucosa. Collectively, to our knowledge, these findings reveal the first systematic evidence of a physiological role for A20 in the maintenance of oral tissue homeostasis as a negative regulator of inflammation.
Collapse
Affiliation(s)
- Yajie Li
- Department of Periodontics, School of Dentistry, Virginia Commonwealth University, Richmond, VA 23298
| | - Erin C Mooney
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, VA 23298
| | - Sara E Holden
- Department of Periodontics, School of Dentistry, Virginia Commonwealth University, Richmond, VA 23298
| | - Xia-Juan Xia
- Department of Periodontics, School of Dentistry, Virginia Commonwealth University, Richmond, VA 23298
| | - David J Cohen
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284
| | - Scott W Walsh
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298.,Departments of Obstetrics and Gynecology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298; and
| | - Averil Ma
- Department of Medicine, School of Medicine, University of California San Francisco, San Francisco, CA 94143
| | - Sinem E Sahingur
- Department of Periodontics, School of Dentistry, Virginia Commonwealth University, Richmond, VA 23298; .,Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, VA 23298
| |
Collapse
|
12
|
Kanmani P, Kim H. Protective Effects of Lactic Acid Bacteria Against TLR4 Induced Inflammatory Response in Hepatoma HepG2 Cells Through Modulation of Toll-Like Receptor Negative Regulators of Mitogen-Activated Protein Kinase and NF-κB Signaling. Front Immunol 2018; 9:1537. [PMID: 30022981 PMCID: PMC6039550 DOI: 10.3389/fimmu.2018.01537] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/21/2018] [Indexed: 12/12/2022] Open
Abstract
The beneficial effects of probiotics in several liver diseases have been investigated in both animal and clinical models; however, the precise mechanisms responsible for their effects have not yet been elucidated. Gut transmitted endotoxins such as LPS have been shown to play critical roles in hepatic inflammation and injury. Therefore, in this study, we investigated the beneficial role of selected lactic acid bacteria (LABs) on reduction of hepatic steatosis (HS) and attenuation of LPS induced inflammatory response in vitro. Total cellular fluid (TCF) of LABs treatment reduced HS by decreasing the amount of lipid accumulation in vitro. Additionally, HepG2 cells exposed to LPS showed increased expression of exacerbated inflammatory cytokines, such as IL-6, CXCL8, CCL2, and TNF-α, but these effects were counteracted when cells were treated with TCF of LABs prior to LPS challenge. Moreover, TCF of LABs was able to modulate mRNA levels of TLR negative regulators and protein levels of p38 MAPK and p65 NF-κB transcription factors. However, these modulations were differed remarkably between both free fatty acid treated and untreated HepG2 cells. Heat-killed LABs were also indirectly suppressed THP-1 cells to produce higher level of IL-10, TLR4, and lower at genes level of TGF-β, IL-1β, and IL-6, and at protein level of TNF-α in response to LPS. Taken together, our findings indicate that selected LABs exhibit profound immunoregulatory effects on liver cells via modulation of TLR negative regulators of the MAPK and NF-κB pathways.
Collapse
Affiliation(s)
- Paulraj Kanmani
- Department of Korean Medicine, Dongguk University, Goyang, South Korea
| | - Hojun Kim
- Department of Korean Medicine, Dongguk University, Goyang, South Korea
| |
Collapse
|
13
|
THP-1-derived macrophages render lung epithelial cells hypo-responsive to Legionella pneumophila - a systems biology study. Sci Rep 2017; 7:11988. [PMID: 28931863 PMCID: PMC5607273 DOI: 10.1038/s41598-017-12154-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 09/04/2017] [Indexed: 11/25/2022] Open
Abstract
Immune response in the lung has to protect the huge alveolar surface against pathogens while securing the delicate lung structure. Macrophages and alveolar epithelial cells constitute the first line of defense and together orchestrate the initial steps of host defense. In this study, we analysed the influence of macrophages on type II alveolar epithelial cells during Legionella pneumophila-infection by a systems biology approach combining experimental work and mathematical modelling. We found that L. pneumophila-infected THP-1-derived macrophages provoke a pro-inflammatory activation of neighboring lung epithelial cells, but in addition render them hypo-responsive to direct infection with the same pathogen. We generated a kinetic mathematical model of macrophage activation and identified a paracrine mechanism of macrophage-secreted IL-1β inducing a prolonged IRAK-1 degradation in lung epithelial cells. This intercellular crosstalk may help to avoid an overwhelming inflammatory response by preventing excessive local secretion of pro-inflammatory cytokines and thereby negatively regulating the recruitment of immune cells to the site of infection. This suggests an important but ambivalent immunomodulatory role of macrophages in lung infection.
Collapse
|
14
|
赵 娜, 黄 彪, 吴 巧, 唐 勇, 余 曙. 蛋白修饰与炎症性肠病. Shijie Huaren Xiaohua Zazhi 2017; 25:1521-1527. [DOI: 10.11569/wcjd.v25.i17.1521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
近年来炎症性肠病(inflammatory bowel disease, IBD)的发病率明显呈持续上升趋势, 越来越多的证据表明, 肠道内蛋白质的异常表达或蛋白修饰的异常与IBD的发病有关. 蛋白修饰是指蛋白质通过翻译后修饰改变自身的空间构象、活性、稳定性及与其他分子相互作用等方面的性能, 从而参与调节机体多样化的生命过程. 虽然蛋白修饰不会改变DNA的序列, 但可以影响相关基因的表达. 研究显示, 蛋白修饰可能通过患者的饮食、环境及肠道微生物等多方面影响基因表型从而参与IBD的发病过程. 本文就蛋白修饰在IBD发病过程中所起的作用做一综述.
Collapse
|
15
|
Sun KY, Xu DH, Xie C, Plummer S, Tang J, Yang XF, Ji XH. Lactobacillus paracasei modulates LPS-induced inflammatory cytokine release by monocyte-macrophages via the up-regulation of negative regulators of NF-kappaB signaling in a TLR2-dependent manner. Cytokine 2017; 92:1-11. [PMID: 28088611 DOI: 10.1016/j.cyto.2017.01.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 12/28/2016] [Accepted: 01/03/2017] [Indexed: 01/02/2023]
Abstract
The application of the probiotic lactobacillus is suggested in the treatment of some inflammatory diseases of intestines due to its potential ability to attenuate inflammation. However, the mechanism is not completely understood. In PBMCs, Lactobacillus paracasei (L. Paracasei) down-regulated the LPS-induced production of TNF-α and IL-6. Using a macrophage-like differentiated THP-1 cell line induced by PMA, we investigated the effect of L. paracasei on the production of pro-inflammatory cytokines by monocyte-macrophages. Treatment of the differentiated THP-1 cells with L. paracasei either concurrently with or before LPS challenge attenuated the LPS-induced secretion of TNF-α and IL-1β. This effect was due to a decrease in IκB phosphorylation and NF-κB nuclear translocation. Furthermore, treatment of the differentiated THP-1 cells with L. paracasei induced the expression of negative regulators of the NF-κB signaling pathway, including the deubiquitinating enzyme A20, suppressor of cytokine signaling (SOCS) 1, SOCS3, and IL-1 receptor-associated kinase (IRAK) 3. Pretreatment with an IRAK4 inhibitor suppressed the L. paracasei-induced expression of these negative regulators and further increased the LPS-mediated expressions of TNF-α and IL-1β. Moreover, treatment with an antibody against Toll-like receptor (TLR) 2 reversed the effect of L. paracasei on inducing negative regulators and inhibiting TNF-α and IL-1β productions. Our findings suggest that L. paracasei inhibits the production of pro-inflammatory cytokines by monocyte-macrophages via the induction of negative regulators of the NF-κB signaling pathway in a TLR2-IRAK4-dependent manner.
Collapse
Affiliation(s)
- Ke-Yi Sun
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Dong-Hua Xu
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Chao Xie
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | | | | | - Xiao Fan Yang
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Xiao Hui Ji
- Department of Immunology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
16
|
Zheng CF, Shi JR, Huang Y, Wang SN. A20 inhibits lipopolysaccharide-induced inflammation in enterocytes. World J Gastrointest Pharmacol Ther 2016; 7:540-549. [PMID: 27867687 PMCID: PMC5095573 DOI: 10.4292/wjgpt.v7.i4.540] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 07/14/2016] [Accepted: 08/16/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To examine the role of A20 in the regulation of intestinal epithelial cells (IECs) inflammation.
METHODS Using gene transfection, both stable overexpression and knockdown A20-expressed HT-29 cell lines were established. Accordingly, the cells were divided into the following groups: The control group, the A20 overexpression group, the A20 knockdown group and the respective controls. A20 was stimulated with lipopolysaccharide (LPS) in a dose- and time-dependent manner and was detected using western blotting and real-time polymerase chain reaction (PCR) analyses. Immunofluorescence and western blotting analyses were performed to investigate the role of A20 in the regulation of nuclear factor (NF)-κB activation and translocation into the nucleus. ELISA and real-time PCR were performed to examine A20 in regulating the release of the following inflammatory cytokines: Tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6 and IL-8.
RESULTS The expression of A20 in IECs was inducible. When intestinal epithelial cells were subjected to the stimulation of LPS, the expression of A20 was increased, and the expression of A20 was induced in a dose- and time-dependent manner. The expression of A20 was very low in HT-29 cells without LPS stimulation but rapidly increased and was maintained at a high level 2-4 h after stimulation with LPS. These levels gradually declined with a change in time-course, and the expression of A20 increased with increasing LPS stimulation. Western blotting and immunofluorescence revealed that overexpression of A20 can inhibit NF-κB activation and its translocation to the nucleus. The overexpression of A20 can reduce the levels of proinflammatory cytokines involved in the pathophysiology of inflammatory bowel disease. There was no significant difference in the expression of IL-8 mRNA in the control group, A20 overexpression group or A20 knockdown group without LPS stimulation (P > 0.05); however, while after 2 h, 4 h and 8 h stimulation with LPS, the expression of IL-8 in the A20 overexpression group was lower than the control group and the A20 knockdown group (P < 0.05 or P < 0.01). The expression of TNF-α was different at different time points after 8 h of LPS stimulation (F = 31.33, DF = 5, P < 0.001), and the expression of TNF-α increased as the LPS stimulation time increased. Upon LPS stimulation, lower levels of TNF-α were detected in the A20 overexpression cell lines (P < 0.05). There were no significant differences in the induction of IL-6 and IL-1β among the control group, A20 overexpression group and A20 knockdown group (P > 0.05).
CONCLUSION A20 plays an important role in limiting inflammation by inhibiting LPS-induced NF-κB responses in the gut luminal. A20 may be a potential therapeutic tool for inflammatory diseases.
Collapse
|
17
|
Liu L, Zhang C, Hu Y, Zhou L, Tan Q. Changes in gut toll-like receptor-4 and nod-like receptor family pyrin domain containing-3 innate pathways in liver cirrhosis rats with bacterial translocation. Clin Res Hepatol Gastroenterol 2016; 40:575-583. [PMID: 27061671 DOI: 10.1016/j.clinre.2016.02.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 01/31/2016] [Accepted: 02/29/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND Bacterial translocation (BT) plays a vital role in the development of liver cirrhosis (LC); however, little is known regarding the role of gut innate immunity in cirrhosis. AIMS To observe the influence of BT on multiple vital organs in LC and changes in the gut toll-like receptor-4 and nod-like receptor family pyrin domain containing-3 innate pathways in liver cirrhosis rats with bacterial translocation. METHODS Wistar rats were divided into control and liver cirrhosis with and without bacterial translocation groups. Functional analysis was conducted using peripheral serum. Hematoxylin-eosin staining was performed to evaluate morphological changes in vital organs. Immunohistochemistry was performed to identify the distribution of toll-like receptor-4, nuclear factor-κ-gene binding-p65, nod-like receptor family pyrin domain containing-3, and its adaptor molecule Asc in the ileum. An enzyme-linked immunosorbent assay was conducted to measure tumor necrosis factor-α, interleukin-1β and interleukin-18 levels. RESULTS A significant decrease in the serum albumin level and an increase in the serum ammonia level were detected in the BT-positive rats compared with the BT-negative rats. Pathological injuries of the intestines and livers of the BT-positive rats were greatly increased compared with the BT-negative rats. The cirrhotic rats with bacterial translocation exhibited increased expression of toll-like receptor-4, nuclear factor-κ-gene binding-p65, nod-like receptor family pyrin domain containing-3, Asc and tumor necrosis factor-α in the intestines compared with the negative group. There were no significant differences of interleukin-1β and interleukin-18 in the intestines between these two groups. CONCLUSIONS Excessive activation of nuclear factor-κ-gene binding-p65/tumor necrosis factor-α and impaired activation of nod-like receptor family pyrin domain containing-3/Asc/interleukin-1β, interleukin-18 in the intestinal mucosa were observed in the cirrhotic rats with BT. Regulation of innate immunity in the intestinal mucosa may represent a novel strategy for the prevention and treatment of LC and its complications.
Collapse
Affiliation(s)
- Ling Liu
- Department of gastroenterology and hepatology, West China hospital, Sichuan university, Guoxue Lane 37(#), 610041 Chengdu, China.
| | - Chiqian Zhang
- Department of gastroenterology and hepatology, Guiyang medical university, 550004 Guiyang, Guizhou province, China.
| | - Yanyan Hu
- Department of gastroenterology, general hospital of Fengfeng Jizhong energy group, 056107 Handan, China.
| | - Li Zhou
- Department of gastroenterology and hepatology, Guiyang medical university, 550004 Guiyang, Guizhou province, China.
| | - Qinghua Tan
- Department of gastroenterology and hepatology, West China hospital, Sichuan university, Guoxue Lane 37(#), 610041 Chengdu, China.
| |
Collapse
|
18
|
Dobosz E, Wilamowski M, Lech M, Bugara B, Jura J, Potempa J, Koziel J. MCPIP-1, Alias Regnase-1, Controls Epithelial Inflammation by Posttranscriptional Regulation of IL-8 Production. J Innate Immun 2016; 8:564-578. [PMID: 27513529 DOI: 10.1159/000448038] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 06/28/2016] [Indexed: 12/14/2022] Open
Abstract
Pattern recognition receptors are critical for the detection of invading microorganisms. They activate multiple pathways that lead to the induction of proinflammatory responses and pathogen clearance. The intensity and duration of this immune reaction must be tightly controlled spatially and temporally in every tissue by different negative regulators. We hypothesized that monocyte chemoattractant protein-1-induced protein-1 (MCPIP-1) might play a role in maintaining immune homeostasis in the epithelium both under physiological conditions and upon bacterial infection. To this end, we examined the distribution of the MCPIP-1 transcript and protein in various tissues. The MCPIP-1 protein level was higher in epithelial cells than in myeloid cells. MCPIP-1 exerted RNase activity towards the interleukin (IL)-8 transcript and the lifespan of IL-8 was determined by the presence of the stem-loops/hairpin structures at the 3'UTR region of IL-8 mRNA. Moreover, using fully active, purified recombinant MCPIP-1 protein, we elucidated the mechanism by which MCPIP-1 controls the IL-8 mRNA level. In conclusion, we uncovered a novel IL-8-dependent mechanism via which MCPIP-1 maintains epithelial homeostasis. This study reveals for the first time that MCPIP-1 plays a crucial anti-inflammatory role not only in myeloid cells but also in epithelial cells.
Collapse
Affiliation(s)
- Ewelina Dobosz
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Innate immune responses are key to maintain adequate host-microbial interactions. However, those signals are needed to efficiently trigger rapid and targeted antimicrobial responses in case of pathogen encounter. Several molecules have evolved to regulate intensity and coordinate signaling to avoid detrimental consequences to the host. Regulation can occur at the cell surface, within the cytoplasm, and at the transcriptional level. Innate immune regulation seems to be equally important than stimulation, as disruption of immunoregulatory molecules modulates the risk for several diseases. This is the case for colitis and inflammatory bowel disease but also colorectal cancer and intestinal infections. In this review, we recapitulate the molecular mechanisms underlying regulation of innate immune signals and mention their implications in several disease states including inflammatory bowel disease.
Collapse
|
20
|
Neagos J, Standiford TJ, Newstead MW, Zeng X, Huang SK, Ballinger MN. Epigenetic Regulation of Tolerance to Toll-Like Receptor Ligands in Alveolar Epithelial Cells. Am J Respir Cell Mol Biol 2016; 53:872-81. [PMID: 25965198 DOI: 10.1165/rcmb.2015-0057oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
To protect the host against exuberant inflammation and injury responses, cells have the ability to become hyporesponsive or "tolerized" to repeated stimulation by microbial and nonmicrobial insults. The lung airspace is constantly exposed to a variety of exogenous and endogenous Toll-like receptor (TLR) ligands, yet the ability of alveolar epithelial cells (AECs) to be tolerized has yet to be examined. We hypothesize that type II AECs will develop a tolerance phenotype upon repeated TLR agonist exposure. To test this hypothesis, primary AECs isolated from the lungs of mice and a murine AEC cell line (MLE-12) were stimulated with either a vehicle control or a TLR ligand for 18 hours, washed, then restimulated with either vehicle or TLR ligand for an additional 6 hours. Tolerance was assessed by measurement of TLR ligand-stimulated chemokine production (monocyte chemoattractant protein [MCP]-1/CCL2, keratinocyte chemoattractant [KC]/CXCL1, and macrophage inflammatory protein [MIP]-2/CXCL2). Sequential treatment of primary AECs or MLE-12 cells with TLR agonists resulted in induction of either tolerance or cross-tolerance. The induction of tolerance was not due to expression of specific negative regulators of TLR signaling (interleukin-1 receptor associated kinase [IRAK]-M, Toll-interacting protein [Tollip], single Ig IL-1-related receptor [SIGIRR], or suppressor of cytokine signaling [SOCS]), inhibitory microRNAs (miRs; specifically, miR-155 and miR146a), or secretion of inhibitory or regulatory soluble mediators (prostaglandin E2, IL-10, transforming growth factor-β, or IFN-α/β). Moreover, inhibition of histone demethylation or DNA methylation did not prevent the development of tolerance. However, treatment of AECs with the histone deacetylase inhibitors trichostatin A or suberoylanilide hyrozamine resulted in reversal of the tolerance phenotype. These findings indicate a novel mechanism by which epigenetic modification regulates the induction of tolerance in AECs.
Collapse
Affiliation(s)
- Jacqueline Neagos
- 1 Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan; and
| | - Theodore J Standiford
- 1 Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan; and
| | - Michael W Newstead
- 1 Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan; and
| | - Xianying Zeng
- 1 Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan; and
| | - Steven K Huang
- 1 Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan; and
| | - Megan N Ballinger
- 2 Department of Internal Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Ohio State University, Columbus, Ohio
| |
Collapse
|
21
|
Li J, Zhang L, Zhang Y, Liu Y, Zhang H, Wei L, Shen T, Jiang C, Zhu D. A20 deficiency leads to angiogenesis of pulmonary artery endothelial cells through stronger NF-κB activation under hypoxia. J Cell Mol Med 2016; 20:1319-28. [PMID: 26991692 PMCID: PMC4929300 DOI: 10.1111/jcmm.12816] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 01/17/2016] [Indexed: 12/11/2022] Open
Abstract
A20 is a zinc finger protein associated with hypoxia. As chronic hypoxia is responsible for intimal hyperplasia and disordered angiogenesis of pulmonary artery, which are histological hallmarks of pulmonary artery hypertension, we intended to explore the role of A20 in angiogenesis of pulmonary artery endothelial cells (ECs). Here, we found a transient elevation of A20 expression in the lung tissues from hypoxic rats compared with normoxic controls. This rapid enhancement was mainly detected in the endothelium, and similar results were reproduced in vitro. During early hypoxia, genetic inhibition of A20 increased proliferation in pulmonary artery ECs, linking to advanced cell cycle progression as well as microtubule polymerization, and aggravated angiogenic effects including tube formation, cell migration and adhesion molecules expression. In addition, a negative feedback loop between nuclear factor-kappa B and A20 was confirmed. Our findings provide evidence for an adaptive role of A20 against pulmonary artery ECs angiogenesis via nuclear factor-kappa B activation.
Collapse
Affiliation(s)
- Jing Li
- Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang, China.,Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, Heilongjiang, China
| | - Linlin Zhang
- Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang, China
| | - Yueming Zhang
- Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang, China.,Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, Heilongjiang, China
| | - Ying Liu
- Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang, China.,Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongyue Zhang
- Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang, China.,Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, Heilongjiang, China
| | - Liuping Wei
- Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang, China.,Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, Heilongjiang, China
| | - Tingting Shen
- Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang, China.,Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, Heilongjiang, China
| | - Chun Jiang
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Daling Zhu
- Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang, China.,Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
22
|
Secretory IgA in complex with Lactobacillus rhamnosus potentiates mucosal dendritic cell-mediated Treg cell differentiation via TLR regulatory proteins, RALDH2 and secretion of IL-10 and TGF-β. Cell Mol Immunol 2016; 14:546-556. [PMID: 26972771 DOI: 10.1038/cmi.2015.110] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 12/02/2015] [Accepted: 12/02/2015] [Indexed: 12/16/2022] Open
Abstract
The importance of secretory IgA in controlling the microbiota is well known, yet how the antibody affects the perception of the commensals by the local immune system is still poorly defined. We have previously shown that the transport of secretory IgA in complex with bacteria across intestinal microfold cells results in an association with dendritic cells in Peyer's patches. However, the consequences of such an interaction on dendritic cell conditioning have not been elucidated. In this study, we analyzed the impact of the commensal Lactobacillus rhamnosus, alone or associated with secretory IgA, on the responsiveness of dendritic cells freshly recovered from mouse Peyer's patches, mesenteric lymph nodes, and spleen. Lactobacillus rhamnosus-conditioned mucosal dendritic cells are characterized by increased expression of Toll-like receptor regulatory proteins [including single immunoglobulin interleukin-1 receptor-related molecule, suppressor of cytokine signaling 1, and Toll-interacting molecule] and retinaldehyde dehydrogenase 2, low surface expression of co-stimulatory markers, high anti- versus pro-inflammatory cytokine production ratios, and induction of T regulatory cells with suppressive function. Association with secretory IgA enhanced the anti-inflammatory/regulatory Lactobacillus rhamnosus-induced conditioning of mucosal dendritic cells, particularly in Peyer's patches. At the systemic level, activation of splenic dendritic cells exposed to Lactobacillus rhamnosus was partially dampened upon association with secretory IgA. These data suggest that secretory IgA, through coating of commensal bacteria, contributes to the conditioning of mucosal dendritic cells toward tolerogenic profiles essential for the maintenance of intestinal homeostasis.
Collapse
|
23
|
Srivastav S, Saha A, Barua J, Ukil A, Das PK. IRAK-M regulates the inhibition of TLR-mediated macrophage immune response during late in vitro Leishmania donovani infection. Eur J Immunol 2015; 45:2787-97. [PMID: 26140693 DOI: 10.1002/eji.201445336] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 06/19/2015] [Accepted: 06/30/2015] [Indexed: 12/18/2022]
Abstract
Intramacrophage protozoan parasite Leishmania donovani, causative agent of visceral leishmaniasis, escapes Toll-like receptor (TLR) dependent early host immune response by inducing the deubiquitinating enzyme A20, which is sustained up to 6 h postinfection only. Therefore, Leishmania must apply other means to deactivate late host responses. Here, we elucidated the role of IL-1 receptor-associated kinase M (IRAK-M), a negative regulator of TLR signaling, in downregulating macrophage proinflammatory response during late hours of in vitro infection. Our data reveal a sharp decline in IRAK1 and IRAK4 phosphorylation at 24 h postinfection along with markedly reduced association of IRAK1-TNF receptor associated factor 6, which is mandatory for TLR activation. In contrast, IRAK-M was induced after A20 levels decreased and reached a maximum at 24 h postinfection. IRAK-M induction coincided with increased stimulation of TGF-β, a hallmark cytokine of visceral infection. TGF-β-dependent signaling-mediated induction of SMAD family of proteins, 2, 3, and 4 plays important roles in transcriptional upregulation of IRAK-M. In infected macrophages, siRNA-mediated silencing of IRAK-M displayed enhanced IRAK1 and IRAK4 phosphorylation with a concomitant increase in downstream NF-κB activity and reduced parasite survival. Taken together, the results suggest that IRAK-M may be targeted by L. donovani to inhibit TLR-mediated proinflammatory response late during in vitro infection.
Collapse
Affiliation(s)
- Supriya Srivastav
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Amrita Saha
- Department of Biochemistry, University of Calcutta, Kolkata, India
| | - Jayita Barua
- Department of Biochemistry, University of Calcutta, Kolkata, India
| | - Anindita Ukil
- Department of Biochemistry, University of Calcutta, Kolkata, India
| | - Pijush K Das
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
24
|
Miceli-Richard C, Criswell LA. Genetic, genomic and epigenetic studies as tools for elucidating disease pathogenesis in primary Sjögren's syndrome. Expert Rev Clin Immunol 2014; 10:437-44. [PMID: 24646085 DOI: 10.1586/1744666x.2014.901888] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Primary Sjögren's syndrome (pSS) is characterized by lymphoid infiltration of lacrimal and salivary glands leading to xerophthalmia and xerostomia. pSS is a complex disease involving both genetic and environmental risk factors. Technological advances over the past 10 years have revolutionized genetics and genomics research enabling high-throughput characterization and analysis of DNA and RNA in patient samples on a genome-wide scale. Further, application of high-throughput methods for characterization of epigenetic variation, such as DNA methylation status, is increasingly being applied to AID populations and will likely further define additional risk factors for disease risk and outcome. Main results obtain in pSS through these various approaches are reviewed here.
Collapse
|
25
|
Li C, Guo S, Gao J, Guo Y, Du E, Lv Z, Zhang B. Maternal high-zinc diet attenuates intestinal inflammation by reducing DNA methylation and elevating H3K9 acetylation in the A20 promoter of offspring chicks. J Nutr Biochem 2014; 26:173-83. [PMID: 25541535 DOI: 10.1016/j.jnutbio.2014.10.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 10/02/2014] [Accepted: 10/02/2014] [Indexed: 12/12/2022]
Abstract
A20 is an anti-inflammatory protein that suppresses ubiquitin-dependent nuclear factor κB (NF-κB) signaling, which can be regulated by the microelement zinc (Zn). In mammals, Zn deficiency contributes to a decrease in A20 abundance, which impairs the gut mucosa barrier. However, it is unclear whether the epigenetic reprogramming of the A20 promoter is involved in enhanced Zn-induced intestinal immunity, especially in avian species. Herein, we show that maternal organic Zn exposure resulted in significantly improved intestinal morphological characteristics, increased mucin 2 (MUC2) abundance and secretory IgA (sIgA) production in progeny jejunums. Maternal and offspring Zn supplementation partially alleviated Zn-deficiency-induced inflammatory response, accompanied by repression of NF-κB signaling. Additionally, we observed DNA hypomethylation and histone H3 at lysine 9 (H3K9) hyperacetylation at the A20 promoter region and subsequent activated A20 expression in Zn-supplemented hens compared with control. Notably, maternal dietary organic Zn exposure exhibited greater attenuation of gut impairment, along with increased MUC2 expression and sIgA level, and decreased the abundance of TNF-α and A20 relative to the inorganic-Zn group. Furthermore, enhanced acetylated H3K9 and A20 transcription at day 14 was found in the offspring adequate dietary Zn group. Thus, A20 may be a novel inflammatory-suppressed factor of chick gut that is persistently promoted by dietary Zn supplementation via epigenetic modifications at A20 promoter.
Collapse
Affiliation(s)
- Changwu Li
- State key Laboratory of Animal Nutrition, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, Yuanmingyuan West Road, Haidian District, Beijing, People's Republic of China, 100193
| | - Shuangshuang Guo
- State key Laboratory of Animal Nutrition, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, Yuanmingyuan West Road, Haidian District, Beijing, People's Republic of China, 100193
| | - Jing Gao
- State key Laboratory of Animal Nutrition, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, Yuanmingyuan West Road, Haidian District, Beijing, People's Republic of China, 100193
| | - Yuming Guo
- State key Laboratory of Animal Nutrition, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, Yuanmingyuan West Road, Haidian District, Beijing, People's Republic of China, 100193.
| | - Encun Du
- State key Laboratory of Animal Nutrition, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, Yuanmingyuan West Road, Haidian District, Beijing, People's Republic of China, 100193
| | - Zengpeng Lv
- State key Laboratory of Animal Nutrition, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, Yuanmingyuan West Road, Haidian District, Beijing, People's Republic of China, 100193
| | - Beibei Zhang
- State key Laboratory of Animal Nutrition, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, Yuanmingyuan West Road, Haidian District, Beijing, People's Republic of China, 100193
| |
Collapse
|
26
|
Liu MW, Wang YH, Qian CY, Li H. Xuebijing exerts protective effects on lung permeability leakage and lung injury by upregulating Toll-interacting protein expression in rats with sepsis. Int J Mol Med 2014; 34:1492-504. [PMID: 25269519 PMCID: PMC4214342 DOI: 10.3892/ijmm.2014.1943] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Accepted: 09/16/2014] [Indexed: 01/07/2023] Open
Abstract
Xuebijing (XBJ) is a type of traditional Tibetan medicine, and previous pharmacological studies have shown that the ethanol extract is derived from Chuanxiong, Chishao, Danshen and Honghua. Chuanxiong, Chishao, Danshen and Honghua possesses potent anti-inflammatory activity, and has been used in the treatment of inflammatory infectious diseases. In the present study, we investigated the effects of XBJ on pulmonary permeability and lung injury in cecal ligation and puncture (CLP)-induced sepsis in rats. A CLP sepsis model was established for the control and treatment groups, respectively. Approximately 2 h prior to surgery, an amount of 100 mg/kg XBJ injection was administered to the treatment group. Reverse transcription polymerase chain reaction (PT-PCR) and western blot analysis were used to examine the expression of Toll-interacting protein (Tollip), interleukin-1 receptor-associated kinase 1 (IRAK1), Toll-like receptor 4 (TLR4), nuclear factor-κB65 (NF-κB65) and TNF receptor-associated factor 6 (TRAF6) in lung tissue. ELISA was applied to detect changes of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), interleukin-1 (IL-1), interleukin-4 (IL-4) and interleukin-10 (IL-10) levels in bronchoalveolar lavage (BAL) fluid, and intercellular adhesion molecule 1 (ICAM-1) and von Willebrand factor (vWF) in serum. The number of neutrophils, albumin and total cells in the BAL fluid were measured. For histological analysis, hematoxylin and eosin (H&E) stains were evaluated. Lung permeability, the wet/dry weight ratio (W/D) and the lung pathology score were determined following the induction of ALI by CLP for 24 h. The results demonstrated that XBJ upregulated Tollip expression and blocked the activity of IRAK1, TLR4, NF-κβ65 and TRAF6. Additionally, the number of neutrophils and total cells were significantly decreased in the XBJ group compared to that in the control group. Lung permeability, the wet/dry weight ratio (W/D) and the lung pathology score were significantly decreased in the XBJ group. The histological results also demonstrated the attenuation effect of XBJ on CLP-induced lung inflammation. The results of the present study indicated that XBJ has a significantly reduced CLP-induced lung permeability by upregulating Tollip expression. The protective effects of XBJ suggest its therapeutic potential in CLP-induced acute lung injury treatment.
Collapse
Affiliation(s)
- Ming-Wei Liu
- Department of Emergency, The First Hospital Affiliated To Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Yun-Hui Wang
- Department of Emergency, The First Hospital Affiliated To Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Chuan-Yun Qian
- Department of Emergency, The First Hospital Affiliated To Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Hui Li
- Surgical Intensive Care Unit, The Second Hospital Affiliated To Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| |
Collapse
|
27
|
Günaltay S, Nyhlin N, Kumawat AK, Tysk C, Bohr J, Hultgren O, Hörnquist EH. Differential expression of interleukin-1/Toll-like receptor signaling regulators in microscopic and ulcerative colitis. World J Gastroenterol 2014; 20:12249-12259. [PMID: 25232259 PMCID: PMC4161810 DOI: 10.3748/wjg.v20.i34.12249] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 05/05/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate Toll-like receptor (TLR) signaling regulators in microscopic and ulcerative colitis patients.
METHODS: Total RNA and microRNA were isolated from fresh frozen colonic biopsies of non-inflamed controls and patients with active or in-remission collagenous colitis (CC), lymphocytic colitis (LC), or ulcerative colitis (UC). We compared expressions of interleukin-1 receptor-associated kinase (IRAK)-2, IRAK-M, interleukin (IL)-37, microRNA (miR)-146a, miR-155, and miR-21 using quantitative real time reverse transcription polymerase chain reaction.
RESULTS: IRAK-M expression was increased in LC patients with active disease in histopathological remission (LC-HR; P = 0.02) and UC patients (P = 0.01), but no differences in IRAK-2 expression were detected compared to controls. miR-146a, -155 and -21 expressions were increased in LC-HR (P = 0.04, 0.07, and 0.004) and UC (P = 0.02, 0.04 and 0.03) patients. miR-146a and miR-21 expressions were significantly enhanced in UC patients compared to UC remission (UC-R; P = 0.01 and 0.04). Likewise, active CC patients showed significantly increased expression of miR-155 (P = 0.003) and miR-21 (P = 0.006). IL-37 expression was decreased in both CC (P = 0.03) and LC (P = 0.04) patients with a similar trend in UC patients but not statistically significant, whilst it was increased in UC-R patients compared to controls (P = 0.02) and active UC (P = 0.001).
CONCLUSION: The identification of differentially expressed miRNAs, IL-37, and IRAK-M suggests different pathophysiologic mechanisms in various disease stages in LC, CC, and UC.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Biopsy
- Case-Control Studies
- Colitis, Collagenous/diagnosis
- Colitis, Collagenous/genetics
- Colitis, Collagenous/immunology
- Colitis, Lymphocytic/diagnosis
- Colitis, Lymphocytic/genetics
- Colitis, Lymphocytic/immunology
- Colitis, Ulcerative/diagnosis
- Colitis, Ulcerative/genetics
- Colitis, Ulcerative/immunology
- Colon/immunology
- Colon/pathology
- Female
- Humans
- Inflammation Mediators/analysis
- Interleukin-1/analysis
- Interleukin-1/metabolism
- Interleukin-1 Receptor-Associated Kinases/analysis
- Intestinal Mucosa/immunology
- Intestinal Mucosa/pathology
- Male
- MicroRNAs/analysis
- Middle Aged
- Signal Transduction
- Toll-Like Receptors/metabolism
Collapse
|
28
|
Villena J, Aso H, Kitazawa H. Regulation of toll-like receptors-mediated inflammation by immunobiotics in bovine intestinal epitheliocytes: role of signaling pathways and negative regulators. Front Immunol 2014; 5:421. [PMID: 25228903 PMCID: PMC4151153 DOI: 10.3389/fimmu.2014.00421] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 08/19/2014] [Indexed: 12/13/2022] Open
Abstract
Intestinal epithelial cells (IECs) detect bacterial and viral associated molecular patterns via germline-encoded pattern-recognition receptors (PRRs) and are responsible for maintaining immune tolerance to the communities of resident commensal bacteria while being also capable to mount immune responses against pathogens. Toll-like receptors (TLRs) are a major class of PRRs expressed on IECs and immune cells, which are involved in the induction of both tolerance and inflammation. In the last decade, experimental and clinical evidence was generated to support the application of probiotics with immunoregulatory capacities (immunobiotics) for the prevention and treatment of several gastrointestinal inflammatory disorders in which TLRs exert a significant role. The majority of these studies were performed in mouse and human cell lines, and despite the growing interest in the bovine immune system due to the economic importance of cattle as livestock, only few studies have been conducted on cattle. In this regard, our group has established a bovine intestinal epithelial (BIE) cell line originally derived from fetal bovine intestinal epitheliocytes and used this cell line to evaluate the impact of immunobiotics in TLR-mediated inflammation. This review aims to summarize the current knowledge of the beneficial effects of immunobiotics in the regulation of intestinal inflammation/infection in cattle. Especially, we discuss the role of TLRs and their negative regulators in both the inflammatory response and the beneficial effects of immunobiotics in bovine IECs. This review article emphasizes the cellular and molecular interactions of immunobiotics with BIE cells through TLRs and gives the scientific basis for the development of immunomodulatory feed for bovine healthy development.
Collapse
Affiliation(s)
- Julio Villena
- Immunobiotics Research Group , Tucuman , Argentina ; Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET) , Tucuman , Argentina
| | - Hisashi Aso
- Cell Biology Laboratory, Graduate School of Agricultural Science, Tohoku University , Sendai , Japan
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University , Sendai , Japan
| |
Collapse
|
29
|
Zhang X, Su Y, Song H, Yu Z, Zhang B, Chen H. Attenuated A20 expression of acute myeloid leukemia-derived dendritic cells increased the anti-leukemia immune response of autologous cytolytic T cells. Leuk Res 2014; 38:673-81. [DOI: 10.1016/j.leukres.2014.03.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 03/13/2014] [Accepted: 03/15/2014] [Indexed: 11/26/2022]
|
30
|
Airway structural cells regulate TLR5-mediated mucosal adjuvant activity. Mucosal Immunol 2014; 7:489-500. [PMID: 24064672 DOI: 10.1038/mi.2013.66] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 07/16/2013] [Accepted: 08/13/2013] [Indexed: 02/04/2023]
Abstract
Antigen-presenting cell (APC) activation is enhanced by vaccine adjuvants. Most vaccines are based on the assumption that adjuvant activity of Toll-like receptor (TLR) agonists depends on direct, functional activation of APCs. Here, we sought to establish whether TLR stimulation in non-hematopoietic cells contributes to flagellin's mucosal adjuvant activity. Nasal administration of flagellin enhanced T-cell-mediated immunity, and systemic and secretory antibody responses to coadministered antigens in a TLR5-dependent manner. Mucosal adjuvant activity was not affected by either abrogation of TLR5 signaling in hematopoietic cells or the presence of flagellin-specific, circulating neutralizing antibodies. We found that flagellin is rapidly degraded in conducting airways, does not translocate into lung parenchyma and stimulates an early immune response, suggesting that TLR5 signaling is regionalized. The flagellin-specific early response of lung was regulated by radioresistant cells expressing TLR5 (particularly the airway epithelial cells). Flagellin stimulated the epithelial production of a small set of mediators that included the chemokine CCL20, which is known to promote APC recruitment in mucosal tissues. Our data suggest that (i) the adjuvant activity of TLR agonists in mucosal vaccination may require TLR stimulation of structural cells and (ii) harnessing the effect of adjuvants on epithelial cells can improve mucosal vaccines.
Collapse
|
31
|
Zou XL, Pei DA, Yan JZ, Xu G, Wu P. A20 overexpression inhibits lipopolysaccharide-induced NF-κB activation, TRAF6 and CD40 expression in rat peritoneal mesothelial cells. Int J Mol Sci 2014; 15:6592-608. [PMID: 24747594 PMCID: PMC4013649 DOI: 10.3390/ijms15046592] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 03/04/2014] [Accepted: 03/24/2014] [Indexed: 12/31/2022] Open
Abstract
Zinc finger protein A20 is a key negative regulator of inflammation. However, whether A20 may affect inflammation during peritoneal dialysis (PD)-associated peritonitis is still unclear. This study was aimed to investigate the effect of A20 overexpression on lipopolysaccharide (LPS)-induced inflammatory response in rat peritoneal mesothelial cells (RPMCs). Isolated and cultured RPMCs in vitro. Plasmid pGEM-T easy-A20 was transfected into RPMCs by Lipofectamine™2000. The protein expression of A20, phospho-IκBα, IκBα, TNF receptor-associated factor (TRAF) 6 and CD40 were analyzed by Western blot. The mRNA expression of TRAF6, CD40, interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) were determined by real time-PCR. NF-κB p65 DNA binding activity, IL-6 and TNF-α levels in cells culture supernatant were determined by ELISA. Our results revealed that RPMCs overexpression of A20 lead to significant decrease of LPS-induced IκBα phosphorylation and NF-κB DNA binding activity (all p<0.01). In addition, A20 also attenuated the expression of TRAF6, CD40, IL-6 and TNF-α as well as levels of IL-6 and TNF-α in cells culture supernatant (all p<0.05). However, A20 only partly inhibited CD40 expression. Our study indicated that A20 overexpression may depress the inflammatory response induced by LPS in cultured RPMCs through negatively regulated the relevant function of adaptors in LPS signaling pathway.
Collapse
Affiliation(s)
- Xun-Liang Zou
- Department of Nephrology, the Affiliated Hospital, Hangzhou Normal University, Hangzhou 310015, Zhejiang, China.
| | - De-An Pei
- Division of Cardiology, Hangzhou Red Cross Hospital, Hangzhou 310003, Zhejiang, China.
| | - Ju-Zhen Yan
- Department of Nephrology, the Affiliated Hospital, Hangzhou Normal University, Hangzhou 310015, Zhejiang, China.
| | - Gang Xu
- Department of Nephrology, the Affiliated Hospital, Hangzhou Normal University, Hangzhou 310015, Zhejiang, China.
| | - Ping Wu
- Department of Nephrology, the Affiliated Hospital, Hangzhou Normal University, Hangzhou 310015, Zhejiang, China.
| |
Collapse
|
32
|
Kamba A, Lee IA, Mizoguchi E. Potential association between TLR4 and chitinase 3-like 1 (CHI3L1/YKL-40) signaling on colonic epithelial cells in inflammatory bowel disease and colitis-associated cancer. Curr Mol Med 2014; 13:1110-21. [PMID: 23170831 DOI: 10.2174/1566524011313070006] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 11/02/2012] [Accepted: 11/19/2012] [Indexed: 12/19/2022]
Abstract
Inflammatory bowel disease (IBD) is a group of inflammatory disorders in the small and large intestines. Several studies have proved that persistent and disregulated host/microbial interactions are required for the development of IBD. It is well known that chronic IBD is strongly associated with an increased risk of developing colorectal cancer by 0.5-1% annually, 8-10 years after the initial diagnosis. To detect the tiny dysplasia or early stage of cancer in chronic IBD patients, a tremendous amount of effort is currently directed for improving colonoscopic technology and noninvasive serological marker development. However, there is only a limited amount of data available to understand the exact mechanism of how long term chronic colitis is connected to the development of colorectal tumors. Recently, our group has identified significantly increased expression of chitinase 3-like 1 (CHI3L1) molecule in non-dysplastic mucosa from patients with IBD and remote dysplasia/cancer, compared to patients with IBD without dysplasia or healthy controls. CHI3L1 seems to contribute to the proliferation, migration, and neoplastic progression of colonic epithelial cells (CECs) under inflammatory conditions. Furthermore, the TLR4-mediated intracellular signaling cascade is likely to interact with CHI3L1 signaling in CECs. In this review article, we have concisely summarized the cellular and molecular mechanisms underlining the development of IBD and colitis-associated cancer, with particular focus on the TLR4- and CHI3L1-signaling pathways in CECs.
Collapse
Affiliation(s)
- A Kamba
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | | | |
Collapse
|
33
|
Hu J, Wang G, Liu X, Zhou L, Jiang M, Yang L. A20 is critical for the induction of Pam3CSK4-tolerance in monocytic THP-1 cells. PLoS One 2014; 9:e87528. [PMID: 24489933 PMCID: PMC3905037 DOI: 10.1371/journal.pone.0087528] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 12/30/2013] [Indexed: 01/24/2023] Open
Abstract
A20 functions to terminate Toll-like receptor (TLR)-induced immune response, and play important roles in the induction of lipopolysacchride (LPS)-tolerance. However, the molecular mechanism for Pam3CSK4-tolerance is uncertain. Here we report that TLR1/2 ligand Pam3CSK4 induced tolerance in monocytic THP-1 cells. The pre-treatment of THP-1 cells with Pam3CSK4 down-regulated the induction of pro-inflammatory cytokines induced by Pam3CSK4 re-stimulation. Pam3CSK4 pre-treatment also down-regulated the signaling transduction of JNK, p38 and NF-κB induced by Pam3CSK4 re-stimulation. The activation of TLR1/2 induced a rapid and robust up-regulation of A20, suggesting that A20 may contribute to the induction of Pam3CSK4-tolerance. This hypothesis was proved by the observation that the over-expression of A20 by gene transfer down-regulated Pam3CSK4-induced inflammatory responses, and the down-regulation of A20 by RNA interference inhibited the induction of tolerance. Moreover, LPS induced a significant up-regulation of A20, which contributed to the induction of cross-tolerance between LPS and Pam3CSK4. A20 was also induced by the treatment of THP-1 cells with TNF-α and IL-1β. The pre-treatment with TNF-α and IL-1β partly down-regulated Pam3CSK4-induced activation of MAPKs. Furthermore, pharmacologic inhibition of GSK3 signaling down-regulated Pam3CSK4-induced A20 expression, up-regulated Pam3CSK4-induced inflammatory responses, and partly reversed Pam3CSK4 pre-treatment-induced tolerance, suggesting that GSK3 is involved in TLR1/2-induced tolerance by up-regulation of A20 expression. Taken together, these results indicated that A20 is a critical regulator for TLR1/2-induced pro-inflammatory responses.
Collapse
Affiliation(s)
- Jinyue Hu
- Medical Research Center, Changsha Central Hospital, Changsha, Hunan, China
- * E-mail: (JH); (LY)
| | - Guihua Wang
- Cancer Center, Changsha Central Hospital, Changsha, Hunan, China
| | - Xueting Liu
- Medical Research Center, Changsha Central Hospital, Changsha, Hunan, China
| | - Lina Zhou
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Manli Jiang
- Medical Research Center, Changsha Central Hospital, Changsha, Hunan, China
| | - Li Yang
- Tuberculosis Research Center, Changsha Central Hospital, Changsha, Hunan, China
- * E-mail: (JH); (LY)
| |
Collapse
|
34
|
Lu P, Sodhi CP, Hackam DJ. Toll-like receptor regulation of intestinal development and inflammation in the pathogenesis of necrotizing enterocolitis. ACTA ACUST UNITED AC 2013; 21:81-93. [PMID: 24365655 DOI: 10.1016/j.pathophys.2013.11.007] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Toll-like receptors (TLRs) are a structurally related family of molecules that respond to a wide variety of endogenous and exogenous ligands, and which serve as important components of the innate immune system. While TLRs have established roles in host defense, these molecules have also been shown to play important roles in the development of various disease states. A particularly important example of the role of TLRs in disease induction includes necrotizing enterocolitis (NEC), which is the most common gastrointestinal disease in preterm infants, and which is associated with extremely high morbidity and mortality rates. The development of NEC is thought to reflect an abnormal interaction between microorganisms and the immature intestinal epithelium, and emerging evidence has clearly placed the spotlight on an important and exciting role for TLRs, particularly TLR4, in NEC pathogenesis. In premature infants, TLR4 signaling within the small intestinal epithelium regulates apoptosis, proliferation and migration of enterocytes, affects the differentiation of goblet cells, and reduces microcirculatory perfusion, which in combination result in the development of NEC. This review will explore the signaling properties of TLRs on hematopoietic and non-hematopoietic cells, and will examine the role of TLR4 signaling in the development of NEC. In addition, the effects of dampening TLR4 signaling using synthetic and endogenous TLR4 inhibitors and active components from amniotic fluid and human milk on NEC severity will be reviewed. In so doing, we hope to present a balanced approach to the understanding of the role of TLRs in both immunity and disease pathogenesis, and to dissect the precise roles for TLR4 in both the cause and therapeutic intervention of necrotizing enterocolitis.
Collapse
Affiliation(s)
- Peng Lu
- Departments of Surgery, University of Pittsburgh School of Medicine, United States
| | - Chhinder P Sodhi
- Departments of Surgery, University of Pittsburgh School of Medicine, United States; Division of Pediatric Surgery, Children's Hospital of Pittsburgh of UPMC, United States
| | - David J Hackam
- Departments of Surgery, University of Pittsburgh School of Medicine, United States; Division of Pediatric Surgery, Children's Hospital of Pittsburgh of UPMC, United States.
| |
Collapse
|
35
|
Biasi F, Leonarduzzi G, Oteiza PI, Poli G. Inflammatory bowel disease: mechanisms, redox considerations, and therapeutic targets. Antioxid Redox Signal 2013; 19:1711-47. [PMID: 23305298 PMCID: PMC3809610 DOI: 10.1089/ars.2012.4530] [Citation(s) in RCA: 193] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Oxidative stress is thought to play a key role in the development of intestinal damage in inflammatory bowel disease (IBD), because of its primary involvement in intestinal cells' aberrant immune and inflammatory responses to dietary antigens and to the commensal bacteria. During the active disease phase, activated leukocytes generate not only a wide spectrum of pro-inflammatory cytokines, but also excess oxidative reactions, which markedly alter the redox equilibrium within the gut mucosa, and maintain inflammation by inducing redox-sensitive signaling pathways and transcription factors. Moreover, several inflammatory molecules generate further oxidation products, leading to a self-sustaining and auto-amplifying vicious circle, which eventually impairs the gut barrier. The current treatment of IBD consists of long-term conventional anti-inflammatory therapy and often leads to drug refractoriness or intolerance, limiting patients' quality of life. Immune modulators or anti-tumor necrosis factor α antibodies have recently been used, but all carry the risk of significant side effects and a poor treatment response. Recent developments in molecular medicine point to the possibility of treating the oxidative stress associated with IBD, by designing a proper supplementation of specific lipids to induce local production of anti-inflammatory derivatives, as well as by developing biological therapies that target selective molecules (i.e., nuclear factor-κB, NADPH oxidase, prohibitins, or inflammasomes) involved in redox signaling. The clinical significance of oxidative stress in IBD is now becoming clear, and may soon lead to important new therapeutic options to lessen intestinal damage in this disease.
Collapse
Affiliation(s)
- Fiorella Biasi
- 1 Department of Clinical and Biological Sciences, University of Turin , San Luigi Gonzaga Hospital, Orbassano, Italy
| | | | | | | |
Collapse
|
36
|
Xu N, Yu ZH, Yao QS, Wang ZQ, Qu HL, Sun Y, Liu J, Jin CZ. PPAR-γ and tollip are associated with toll-like receptors in colitis rats. J Immunoassay Immunochem 2013; 34:219-31. [PMID: 23656243 DOI: 10.1080/15321819.2012.699490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
To elucidate the significance of Toll-like receptors and their negative regulating factors PPAR-γ and Tollip on the pathogenesis of colitis. Colitis model was induced by TNBS in rat. The expression of TLR2, TLR4, NF-κBp65, PPAR-γ and Tollip was examined by immunohistochemistry (IHC) and reverse-transcription polymerase chain reaction (RT-PCR). RT-PCR revealed a significant increased expression of TLR2, TLR4, and NF-κBp65 in the colitis group compared with the normal group (TLR2: 1.057 ± 0.092, 0.463 ± 0.101, t = 4.125, P = 0.001; TLR4: 0.376 ± 0.029, 0.215 ± 0.049, t = 2.731, P = 0.013; NF-κBp65: 0.746 ± 0.049, 0.206 ± 0.063, t = 6.055, P = 0.000). The expression was positively correlated with the generally damage score and the histological injury score correspondingly (TLR2: r = 0.573, r = 0.559; TLR4: r = 0.754, r = 0.866; NF-κBp65: r = 0.548, r = 0.919). The Tollip mRNA wasn't obviously diversity between the normal and colitis groups by RT-PCR (Tollip: 0.288 ± 0.050, 0.140 ± 0.046, t = 1.993, P = 0.061). While the Tollip protein was mainly assembled in the lamina propriaand higher in the colitis group compared with the normal group by IHC. The expression of PPAR-γ in the colitis group was obviously lower than that in the normal group (PPAR-γ: 0.255 ± 0.065, 0.568 ± 0.072, t = 2.882, P = 0.010). The expression of Tollip and PPAR-γ was negative correlated with the generally damage score and histological injury score correspondingly (Tollip: r = -0.497, r = -0.551; PPAR-γ: r = -0.683, r = -0.853). The disbalance between TLRs and their negative regulating factors PPAR-γ and Tollip was closely associated with the course of colitis.
Collapse
Affiliation(s)
- Ning Xu
- Department of Gastroenterology , Affiliated Yantai Yu Huang Ding Hospital of Qingdao University Medical School, Yantai, PR China
| | | | | | | | | | | | | | | |
Collapse
|
37
|
AZIZ MONOWAR, ISHIHARA SHUNJI, ANSARY MESBAHUDDIN, SONOYAMA HIROKI, TADA YASUMASA, OKA AKIHIKO, KUSUNOKI RYUSAKU, TAMAGAWA YUJI, FUKUBA NOBUHIKO, MISHIMA YOSHIYUKI, MISHIRO TSUYOSHI, OSHIMA NAOKI, MORIYAMA ICHIRO, ISHIMURA NORIHISA, SATO SHUICHI, YUKI TAKAFUMI, KAWASHIMA KOUSAKU, KINOSHITA YOSHIKAZU. Crosstalk between TLR5 and Notch1 signaling in epithelial cells during intestinal inflammation. Int J Mol Med 2013; 32:1051-62. [PMID: 24048326 DOI: 10.3892/ijmm.2013.1501] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 08/27/2013] [Indexed: 11/06/2022] Open
|
38
|
Murata K, Villena J, Tomosada Y, Hara R, Chiba E, Shimazu T, Aso H, Suda Y, Iwabuchi N, Xiao JZ, Saito T, Kitazawa H. Bifidobacteria Upregulate Expression of Toll-Like Receptor Negative Regulators Counteracting Enterotoxigenic <i>Escherichia coli</i> Mediated Inflammation in Bovine Intestinal Epitheliocytes. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/ojvm.2013.32023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
39
|
Li N, Quidgley MC, Kobeissy FH, Joseph J, Neu J. Microbial cell components induced tolerance to flagellin-stimulated inflammation through Toll-like receptor pathways in intestinal epithelial cells. Cytokine 2012; 60:806-11. [PMID: 22944462 PMCID: PMC3572906 DOI: 10.1016/j.cyto.2012.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 06/29/2012] [Accepted: 08/06/2012] [Indexed: 01/01/2023]
Abstract
In the intestine, bacterial components activate innate responses that protect the host. We hypothesize that bacterial components reduce Interleukin-8 (IL-8) production in intestinal epithelial cells stimulated by flagellin via the Toll-like receptor (TLR) signaling pathway. Caco-2 cells were pretreated with various doses of lipopolysaccharide (LPS), lipoteichoic acid (LTA), or low-dose flagellin (LDFL) for 24h. Cells were then treated with flagellin (FL) 500 ng/ml (HDFL) for another 48 h. IL-8 production was measured in the cell culture medium by ELISA. Eighty-four genes in the TLR pathway were evaluated by RT Profiler PCR Array. Pathway Studio 8.0 software was used for altered pathway analysis. HDFL induced IL-8 production by 19-fold (p<0.01). Pretreatment with LDFL at 20, 10 or 1 ng/ml reduced HDFL-induced IL-8 production by 61%, 52% and 40%, respectively (p<0.05). LPS at 50 μg/ml decreased HDFL-induced IL-8 production by 38% (p<0.05). HDFL up-regulated CXCL10, IL1B, IL-8, IRAK2, NF-κB1 and I-κB (all p<0.05). Pathway Studio analysis showed that HDFL induced cell processes including inflammation, cell death and apoptosis. Pretreatment with LDFL at 10 ng/ml down-regulated FADD, FOS, MAP4K4, MyD88, TLR2, TLR3 and TNFERSF1A compared to HDFL (all p<0.05). These down-regulated genes are integral for numerous cell functions including inflammatory response, cell death, apoptosis and infection. These results demonstrate that LPS and LDFL provoke tolerance to HDFL-induced IL-8 production. This tolerance effect was accompanied by a complex interaction of multiple genes related to inflammatory as well as other responses in the TLR pathway rather than a single gene alteration.
Collapse
Affiliation(s)
- Nan Li
- Department of Pediatrics, University of Florida, 1600 SW Archer Road, Gainesville FL, USA 32610
| | - Maria C. Quidgley
- Department of Pediatrics, University of Florida, 1600 SW Archer Road, Gainesville FL, USA 32610
| | - Firas H. Kobeissy
- Department of Psychiatry, University of Florida, 1600 SW Archer Road, Gainesville FL, USA 32610
| | - Jessica Joseph
- Department of Pediatrics, University of Florida, 1600 SW Archer Road, Gainesville FL, USA 32610
| | - Josef Neu
- Department of Pediatrics, University of Florida, 1600 SW Archer Road, Gainesville FL, USA 32610
| |
Collapse
|
40
|
Abstract
The healthy human gut supports a complex and diverse microbiota, dominated by bacterial phylotypes belonging to Bacteroidetes and Firmicutes. In the inflamed gut, overall diversity decreases, coincident with a greater representation of Proteobacteria. There is growing evidence supporting an important role for human gut bacteria in mucosal immunity; interactions at the level of both intestinal and colonic epithelial cells, dendritic cells, and T and B immune cells have been documented. These interactions influence gut barrier and defense mechanisms that include antimicrobial peptide and secretory IgA synthesis. The functional effects of commensal bacteria on T helper cell differentiation have led to the emerging concept that microbiota composition determines T effector- and T regulatory-cell balance, immune responsiveness, and homeostasis. The importance of this biology in relation to immune homeostasis, inflammatory bowel disease, and the rising incidence of autoimmune diseases will be discussed. The detailed description of the human gut microbiota, integrated with evidence-based mechanisms of immune modulation, provides an exciting platform for the identification of next-generation probiotics and related pharmaceutical products.
Collapse
Affiliation(s)
- Denise Kelly
- Rowett Institute of Nutrition & Health, University of Aberdeen, Foresterhill, Aberdeen, Scotland, UK.
| | | |
Collapse
|
41
|
Chen C, Yang G, Geng XR, Wang X, Liu Z, Yang PC. TNFAIP3 facilitates degradation of microbial antigen SEB in enterocytes. PLoS One 2012; 7:e45941. [PMID: 23029332 PMCID: PMC3454357 DOI: 10.1371/journal.pone.0045941] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 08/23/2012] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND AIMS The enterocytes have the potential to absorb noxious substances, such as microbial products, from the gut lumen. How the enterocytes process the substances to harmless materials is not fully understood. This study aims to elucidate the role of ubiquitin E3 ligase TNFAIP3 (TNFAIP3) in facilitating the degradation of endocytic microbial products in enterocytes. METHODS Human intestinal epithelial cell line, HT-29 cells, was cultured to monolayers using as an in vitro model to observe the endocytosis and degradation of microbial products, Staphylococcal enterotoxin B (SEB) in epithelial cells. The RNA interference was employed to knock down the TNFAIP3 gene in HT-29 cells to observe the role of TNFAIP3 in the degradation of endocytic SEB. The role of TNFAIP3 in facilitating the endosome/lysosome fusion was observed by immunocytochemistry. RESULTS Upon the absorption of SEB, the expression of TNFAIP3 was increased in HT-29 cells. Silencing the TNFAIP3 gene in HT-29 cells resulted in a large quantity of SEB to be transported across the HT-29 monolayers to the transwell basal chambers; the transportation was via the intracellular pathway. TNFAIP3 was required in the fusion of SEB-carrying endosomes and lysosomes. CONCLUSIONS TNFAIP3 plays a critical role in the degradation of endocytic SEB in enterocytes.
Collapse
Affiliation(s)
- Chi Chen
- Department of Gastroenterology, the Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
| | - Gui Yang
- Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Xiao-Rui Geng
- Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Xingpeng Wang
- Department of Gastroenterology, the Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
| | - Zhanju Liu
- Department of Gastroenterology, the Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- * E-mail: (PCY); (ZL)
| | - Ping-Chang Yang
- Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
- * E-mail: (PCY); (ZL)
| |
Collapse
|
42
|
Chen W, Saxena A, Li N, Sun J, Gupta A, Lee DW, Tian Q, Dobaczewski M, Frangogiannis NG. Endogenous IRAK-M attenuates postinfarction remodeling through effects on macrophages and fibroblasts. Arterioscler Thromb Vasc Biol 2012; 32:2598-608. [PMID: 22995519 DOI: 10.1161/atvbaha.112.300310] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Effective postinfarction repair requires timely suppression of innate immune signals to prevent the catastrophic consequences of uncontrolled inflammation on cardiac geometry and function. In macrophages, interleukin-1 receptor-associated kinase (IRAK)-M acts as a functional decoy preventing uncontrolled toll-like receptor /interleukin-1-mediated responses. Our study investigates the role of IRAK-M as a negative regulator of the postinfarction inflammatory response and as a modulator of cardiac remodeling. METHODS AND RESULTS In wild-type mouse infarcts IRAK-M was upregulated in infiltrating macrophages and fibroblasts exhibiting a biphasic response. When compared with wild-type animals, infarcted IRAK-M(-/-) mice had enhanced adverse remodeling and worse systolic dysfunction; however, acute infarct size was comparable between groups. Adverse remodeling in IRAK-M(-/-) animals was associated with enhanced myocardial inflammation and protease activation. The protective actions of IRAK-M involved phenotypic modulation of macrophages and fibroblasts. IRAK-M(-/-) infarcts showed increased infiltration with proinflammatory CD11b+/Ly6C(hi) monocytes; leukocytes harvested from IRAK-M-null infarcts exhibited accentuated cytokine expression. In vitro, IRAK-M expression was upregulated in cytokine-stimulated murine cardiac fibroblasts and suppressed their matrix-degrading properties without affecting their inflammatory activity. CONCLUSIONS Endogenous IRAK-M attenuates adverse postinfarction remodeling suppressing leukocyte inflammatory activity, while inhibiting fibroblast-mediated matrix degradation.
Collapse
Affiliation(s)
- Wei Chen
- The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, 1300 Morris Park Avenue Forchheimer G46B, Bronx, NY 10461, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Rhee L, Murphy SF, Kolodziej LE, Grimm WA, Weber CR, Lodolce JP, Chang JE, Bartulis SJ, Messer JS, Schneider JR, Paski S, Nero TM, Boone DL. Expression of TNFAIP3 in intestinal epithelial cells protects from DSS- but not TNBS-induced colitis. Am J Physiol Gastrointest Liver Physiol 2012; 303:G220-7. [PMID: 22595989 PMCID: PMC3404569 DOI: 10.1152/ajpgi.00077.2012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intestinal epithelial cells (IEC) maintain gastrointestinal homeostasis by providing a physical and functional barrier between the intestinal lumen and underlying mucosal immune system. The activation of NF-κB and prevention of apoptosis in IEC are required to maintain the intestinal barrier and prevent colitis. How NF-κB activation in IEC prevents colitis is not fully understood. TNFα-induced protein 3 (TNFAIP3) is a NF-κB-induced gene that acts in a negative-feedback loop to inhibit NF-κB activation and also to inhibit apoptosis; therefore, we investigated whether TNFAIP3 expression in the intestinal epithelium impacts susceptibility of mice to colitis. Transgenic mice expressing TNFAIP3 in IEC (villin-TNFAIP3 Tg mice) were exposed to dextran sodium sulfate (DSS) or 2,4,6-trinitrobenzene sulfonic acid (TNBS), and the severity and characteristics of mucosal inflammation and barrier function were compared with wild-type mice. Villin-TNFAIP3 Tg mice were protected from DSS-induced colitis and displayed reduced production of NF-κB-dependent inflammatory cytokines. Villin-TNFAIP3 Tg mice were also protected from DSS-induced increases in intestinal permeability and induction of IEC death. Villin-TNFAIP3 Tg mice were not protected from colitis induced by TNBS. These results indicate that TNFAIP3 expression in IEC prevents colitis involving DSS-induced IEC death, but not colitis driven by T cell-mediated inflammation. As TNFAIP3 inhibits NF-κB activation and IEC death, expression of TNFAIP3 in IEC may provide an avenue to inhibit IEC NF-κB activation without inducing IEC death and inflammation.
Collapse
Affiliation(s)
- Lesley Rhee
- 1Department of Medicine, University of Chicago, Chicago, Illinois; and
| | - Stephen F. Murphy
- 1Department of Medicine, University of Chicago, Chicago, Illinois; and
| | | | - Wesley A. Grimm
- 1Department of Medicine, University of Chicago, Chicago, Illinois; and
| | | | - James P. Lodolce
- 1Department of Medicine, University of Chicago, Chicago, Illinois; and
| | - Jonathan E. Chang
- 1Department of Medicine, University of Chicago, Chicago, Illinois; and
| | - Sarah J. Bartulis
- 1Department of Medicine, University of Chicago, Chicago, Illinois; and
| | | | - Jeff R. Schneider
- 1Department of Medicine, University of Chicago, Chicago, Illinois; and
| | - Shirley Paski
- 1Department of Medicine, University of Chicago, Chicago, Illinois; and
| | - Thomas M. Nero
- 1Department of Medicine, University of Chicago, Chicago, Illinois; and
| | - David L. Boone
- 1Department of Medicine, University of Chicago, Chicago, Illinois; and
| |
Collapse
|
44
|
Shah JA, Vary JC, Chau TTH, Bang ND, Yen NTB, Farrar JJ, Dunstan SJ, Hawn TR. Human TOLLIP regulates TLR2 and TLR4 signaling and its polymorphisms are associated with susceptibility to tuberculosis. THE JOURNAL OF IMMUNOLOGY 2012; 189:1737-46. [PMID: 22778396 DOI: 10.4049/jimmunol.1103541] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Tuberculosis, one of the leading causes of death worldwide, stimulates inflammatory responses with beneficial and pathologic consequences. The regulation and nature of an optimal inflammatory response to Mycobacterium tuberculosis remains poorly understood in humans. Insight into mechanisms of negative regulation of the TLR-mediated innate immune response to M. tuberculosis could provide significant breakthroughs in the design of new vaccines and drugs. We hypothesized that TOLLIP and its common variants negatively regulate TLR signaling in human monocytes and are associated with susceptibility to tuberculosis. Using short hairpin RNA knockdown of TOLLIP in peripheral blood human monocytes, we found that TOLLIP suppresses TNF and IL-6 production after stimulation with TLR2 and TLR4 ligands. In contrast, secretion of the anti-inflammatory cytokine IL-10 was induced by TOLLIP. We also discovered two common polymorphisms that are associated with either decreased levels of mRNA expression (rs3750920) or increased IL-6 production (rs5743899) in a sample of 56 healthy volunteers. Furthermore, in a case-population study in Vietnam with 760 cord blood samples and 671 TB case patients, we found that SNPs rs3750920 and rs5743899 were associated with susceptibility to tuberculosis (p = 7.03 × 10(-16) and 6.97 × 10(-7), respectively). These data demonstrate that TOLLIP has an anti-inflammatory effect on TLR signaling in humans and that TOLLIP deficiency is associated with an increased risk of tuberculosis. To our knowledge, these data also show the first associations of TOLLIP polymorphisms with any infectious disease. These data also implicate an unexpected mechanism of negative regulation of TLR signaling in human tuberculosis pathogenesis.
Collapse
Affiliation(s)
- Javeed A Shah
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98195, USA.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Miron N, Cristea V. Enterocytes: active cells in tolerance to food and microbial antigens in the gut. Clin Exp Immunol 2012; 167:405-12. [PMID: 22288583 DOI: 10.1111/j.1365-2249.2011.04523.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Enterocytes used to be studied particularly in terms of digestion protagonists. However, as the immune functions of the intestinal tract were better understood, it became clear that enterocytes are not mere bystanders concerning the induction of immune tolerance to dietary peptides and gut microbiota. In fact, enterocytes are involved actively in shaping the intestinal immune environment, designed for maintaining a non-belligerent state. This tolerant milieu of the gut immune system is achieved by keeping a balance between suppression and stimulation of the inflammatory responses. Our review presents the current state of knowledge concerning the relationship between enterocytes and immune cells (dendritic cells, lymphocytes), with emphasis on the enterocytes' impact on the mechanisms leading to the induction of oral tolerance.
Collapse
Affiliation(s)
- N Miron
- Department of Immunology, University of Medicine and Pharmacy, Iuliu Haţieganu Cluj-Napoca, Romania.
| | | |
Collapse
|
46
|
Wu Q, Jiang D, Smith S, Thaikoottathil J, Martin RJ, Bowler RP, Chu HW. IL-13 dampens human airway epithelial innate immunity through induction of IL-1 receptor-associated kinase M. J Allergy Clin Immunol 2011; 129:825-833.e2. [PMID: 22154382 DOI: 10.1016/j.jaci.2011.10.043] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 09/26/2011] [Accepted: 10/31/2011] [Indexed: 10/14/2022]
Abstract
BACKGROUND Impaired airway mucosal immunity can contribute to increased respiratory tract infections in asthmatic patients, but the involved molecular mechanisms have not been fully clarified. Airway epithelial cells serve as the first line of respiratory mucosal defense to eliminate inhaled pathogens through various mechanisms, including Toll-like receptor (TLR) pathways. Our previous studies suggest that impaired TLR2 function in T(H)2 cytokine-exposed airways might decrease immune responses to pathogens and subsequently exacerbate allergic inflammation. IL-1 receptor-associated kinase M (IRAK-M) negatively regulates TLR signaling. However, IRAK-M expression in airway epithelium from asthmatic patients and its functions under a T(H)2 cytokine milieu remain unclear. OBJECTIVES We sought to evaluate the role of IRAK-M in IL-13-inhibited TLR2 signaling in human airway epithelial cells. METHODS We examined IRAK-M protein expression in epithelia from asthmatic patients versus that in normal airway epithelia. Moreover, IRAK-M regulation and function in modulating innate immunity (eg, TLR2 signaling) were investigated in cultured human airway epithelial cells with or without IL-13 stimulation. RESULTS IRAK-M protein levels were increased in asthmatic airway epithelium. Furthermore, in primary human airway epithelial cells, IL-13 consistently upregulated IRAK-M expression, largely through activation of phosphoinositide 3-kinase pathway. Specifically, phosphoinositide 3-kinase activation led to c-Jun binding to human IRAK-M gene promoter and IRAK-M upregulation. Functionally, IL-13-induced IRAK-M suppressed airway epithelial TLR2 signaling activation (eg, TLR2 and human β-defensin 2), partly through inhibiting activation of nuclear factor κB. CONCLUSIONS Our data indicate that epithelial IRAK-M overexpression in T(H)2 cytokine-exposed airways inhibits TLR2 signaling, providing a novel mechanism for the increased susceptibility of infections in asthmatic patients.
Collapse
Affiliation(s)
- Qun Wu
- Department of Medicine, National Jewish Health and the University of Colorado Denver, Denver, CO 80206, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Takagi M. Toll-like receptor--a potent driving force behind rheumatoid arthritis. J Clin Exp Hematop 2011; 51:77-92. [PMID: 22104306 DOI: 10.3960/jslrt.51.77] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Toll like receptor (TLR), one of the key functions of innate immune system, can recognize not only exogenous pathogen-associated molecular patterns, namely PAMPs, but also endogenous molecules created upon tissue injury, sterile inflammation and degeneration. Endogenous TLR ligands are called as damage-associated molecular patters (DAMPs), including endogenous molecules released by activated and necrotic cells, and extracellular matrix molecules. DAMPs are also known as alarmins. TLR research has brought about new insights in the rheumatic diseases. Previous reports suggest that TLRs and the signal pathways intensively contribute to the pathogenesis of rheumatoid arthritis (RA) and other arthritic conditions with interaction of various TLR ligands. Accumulated knowledge of TLR system is summarized to overlook TLRs and the signaling pathway in arthritis conditions, with special reference to RA.
Collapse
Affiliation(s)
- Michiaki Takagi
- Department of Orthopaedic Surgery, Yamagata University School of Medicine, Japan.
| |
Collapse
|
48
|
Kolodziej LE, Lodolce JP, Chang JE, Schneider JR, Grimm WA, Bartulis SJ, Zhu X, Messer JS, Murphy SF, Reddy N, Turner JR, Boone DL. TNFAIP3 maintains intestinal barrier function and supports epithelial cell tight junctions. PLoS One 2011; 6:e26352. [PMID: 22031828 PMCID: PMC3198775 DOI: 10.1371/journal.pone.0026352] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 09/25/2011] [Indexed: 02/07/2023] Open
Abstract
Tight junctions between intestinal epithelial cells mediate the permeability of the intestinal barrier, and loss of intestinal barrier function mediated by TNF signaling is associated with the inflammatory pathophysiology observed in Crohn's disease and celiac disease. Thus, factors that modulate intestinal epithelial cell response to TNF may be critical for the maintenance of barrier function. TNF alpha-induced protein 3 (TNFAIP3) is a cytosolic protein that acts in a negative feedback loop to regulate cell signaling induced by Toll-like receptor ligands and TNF, suggesting that TNFAIP3 may play a role in regulating the intestinal barrier. To investigate the specific role of TNFAIP3 in intestinal barrier function we assessed barrier permeability in TNFAIP3−/− mice and LPS-treated villin-TNFAIP3 transgenic mice. TNFAIP3−/− mice had greater intestinal permeability compared to wild-type littermates, while villin-TNFAIP3 transgenic mice were protected from increases in permeability seen within LPS-treated wild-type littermates, indicating that barrier permeability is controlled by TNFAIP3. In cultured human intestinal epithelial cell lines, TNFAIP3 expression regulated both TNF-induced and myosin light chain kinase-regulated tight junction dynamics but did not affect myosin light chain kinase activity. Immunohistochemistry of mouse intestine revealed that TNFAIP3 expression inhibits LPS-induced loss of the tight junction protein occludin from the apical border of the intestinal epithelium. We also found that TNFAIP3 deubiquitinates polyubiquitinated occludin. These in vivo and in vitro studies support the role of TNFAIP3 in promoting intestinal epithelial barrier integrity and demonstrate its novel ability to maintain intestinal homeostasis through tight junction protein regulation.
Collapse
Affiliation(s)
- Lauren E. Kolodziej
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - James P. Lodolce
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Jonathan E. Chang
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Jeffrey R. Schneider
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Wesley A. Grimm
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Sarah J. Bartulis
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Xiaorong Zhu
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Jeannette S. Messer
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Stephen F. Murphy
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Nishith Reddy
- Illinois Math and Science Academy, Aurora, Illinois, United States of America
| | - Jerrold R. Turner
- Department of Pathology, University of Chicago, Chicago, Illinois, United States of America
| | - David L. Boone
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
49
|
Xiong Y, Medvedev AE. Induction of endotoxin tolerance in vivo inhibits activation of IRAK4 and increases negative regulators IRAK-M, SHIP-1, and A20. J Leukoc Biol 2011; 90:1141-8. [PMID: 21934070 DOI: 10.1189/jlb.0611273] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
TLRs mediate host defense against microbial pathogens by eliciting production of inflammatory mediators and activating expression of MHC, adhesion, and costimulatory molecules. Endotoxin tolerance limits excessive TLR-driven inflammation during sepsis and reprograms macrophage responses to LPS, decreasing expression of proinflammatory cytokines without inhibiting anti-inflammatory and antimicrobial mediators. Molecular mechanisms of reprogramming of TLR4 signaling upon in vivo induction of endotoxin tolerance are incompletely understood. We used an in vivo model of endotoxin tolerance, whereby C57BL/6 mice were i.p.-inoculated with LPS or PBS, followed by in vitro challenge of peritoneal or splenic macrophages with LPS to examine activation of IRAK4 and expression of negative regulatory molecules. Administration of LPS in vivo-induced endotoxin tolerance in peritoneal and splenic macrophages, as evidenced by decreased degradation of IκBα, suppressed phosphorylation of p38 and reduced expression of TNF-α, IL-6, and KC mRNA upon in vitro LPS challenge. Macrophages from control and endotoxin-tolerant mice exhibited comparable TLR4 mRNA levels and similar expression of IL-1RA and IL-10 genes. Endotoxin tolerization in vivo blocked TLR4-driven IRAK4 phosphorylation and activation in macrophages, while increasing expression of IRAK-M, SHIP-1, A20 mRNA, and A20 protein. Thus, induction of endotoxin tolerance in vivo inhibits expression of proinflammatory mediators via impaired activation of IRAK4, p38, and NF-κB and increases expression of negative regulators of TLR4 pathways.
Collapse
Affiliation(s)
- Yanbao Xiong
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | |
Collapse
|
50
|
Nishio H, Kanno S, Onoyama S, Ikeda K, Tanaka T, Kusuhara K, Fujimoto Y, Fukase K, Sueishi K, Hara T. Nod1 Ligands Induce Site-Specific Vascular Inflammation. Arterioscler Thromb Vasc Biol 2011; 31:1093-9. [DOI: 10.1161/atvbaha.110.216325] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Objective—
The goal of this study was to investigate the effects of stimulants for a nucleotide-binding domain, leucine-rich repeat-containing (NLR) protein family on human artery endothelial cells and murine arteries.
Methods and Results—
Human coronary artery endothelial cells were challenged in vitro with microbial components that stimulate NLRs or Toll-like receptors. We found stimulatory effects of NLR and Toll-like receptor ligands on the adhesion molecule expression and cytokine secretion by human coronary artery endothelial cells. On the basis of these results, we examined the in vivo effects of these ligands in mice. Among them, FK565, 1 of the nucleotide-binding oligomerization domain (Nod)-1 ligands induced strong site-specific inflammation in the aortic root. Furthermore, coronary arteritis/valvulitis developed after direct oral administration or ad libitum drinking of FK565. The degree of the respective vascular inflammation was associated with persistent high expression of proinflammatory chemokine/cytokine and matrix metallopeptidase (
Mmp
) genes in each tissue in vivo by microarray analysis.
Conclusion—
This is the first coronary arteritis animal model induced by oral administration of a pure synthetic Nod1 ligand. The present study has demonstrated an unexpected role of Nod1 in the development of site-specific vascular inflammation, especially coronary arteritis. These findings might lead to the clarification of the pathogenesis and pathophysiology of coronary artery disease in humans.
Collapse
Affiliation(s)
- Hisanori Nishio
- From the Departments of Pediatrics (H.N., S.K., S.O., K.I., T.T., T.H.) and Pathophysiological and Experimental Pathology (H.N., K.S.), Graduate School of Medical Sciences, Kyushu University, Fukuoka; Department of Pediatrics, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Kitakyushu (K.K.); Department of Chemistry, Graduate School of Science, Osaka University, Osaka, Japan (Y.F., K.F.); Japan Society for the Promotion of Science (T.H.), Tokyo, Japan
| | - Shunsuke Kanno
- From the Departments of Pediatrics (H.N., S.K., S.O., K.I., T.T., T.H.) and Pathophysiological and Experimental Pathology (H.N., K.S.), Graduate School of Medical Sciences, Kyushu University, Fukuoka; Department of Pediatrics, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Kitakyushu (K.K.); Department of Chemistry, Graduate School of Science, Osaka University, Osaka, Japan (Y.F., K.F.); Japan Society for the Promotion of Science (T.H.), Tokyo, Japan
| | - Sagano Onoyama
- From the Departments of Pediatrics (H.N., S.K., S.O., K.I., T.T., T.H.) and Pathophysiological and Experimental Pathology (H.N., K.S.), Graduate School of Medical Sciences, Kyushu University, Fukuoka; Department of Pediatrics, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Kitakyushu (K.K.); Department of Chemistry, Graduate School of Science, Osaka University, Osaka, Japan (Y.F., K.F.); Japan Society for the Promotion of Science (T.H.), Tokyo, Japan
| | - Kazuyuki Ikeda
- From the Departments of Pediatrics (H.N., S.K., S.O., K.I., T.T., T.H.) and Pathophysiological and Experimental Pathology (H.N., K.S.), Graduate School of Medical Sciences, Kyushu University, Fukuoka; Department of Pediatrics, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Kitakyushu (K.K.); Department of Chemistry, Graduate School of Science, Osaka University, Osaka, Japan (Y.F., K.F.); Japan Society for the Promotion of Science (T.H.), Tokyo, Japan
| | - Tamami Tanaka
- From the Departments of Pediatrics (H.N., S.K., S.O., K.I., T.T., T.H.) and Pathophysiological and Experimental Pathology (H.N., K.S.), Graduate School of Medical Sciences, Kyushu University, Fukuoka; Department of Pediatrics, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Kitakyushu (K.K.); Department of Chemistry, Graduate School of Science, Osaka University, Osaka, Japan (Y.F., K.F.); Japan Society for the Promotion of Science (T.H.), Tokyo, Japan
| | - Koichi Kusuhara
- From the Departments of Pediatrics (H.N., S.K., S.O., K.I., T.T., T.H.) and Pathophysiological and Experimental Pathology (H.N., K.S.), Graduate School of Medical Sciences, Kyushu University, Fukuoka; Department of Pediatrics, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Kitakyushu (K.K.); Department of Chemistry, Graduate School of Science, Osaka University, Osaka, Japan (Y.F., K.F.); Japan Society for the Promotion of Science (T.H.), Tokyo, Japan
| | - Yukari Fujimoto
- From the Departments of Pediatrics (H.N., S.K., S.O., K.I., T.T., T.H.) and Pathophysiological and Experimental Pathology (H.N., K.S.), Graduate School of Medical Sciences, Kyushu University, Fukuoka; Department of Pediatrics, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Kitakyushu (K.K.); Department of Chemistry, Graduate School of Science, Osaka University, Osaka, Japan (Y.F., K.F.); Japan Society for the Promotion of Science (T.H.), Tokyo, Japan
| | - Koichi Fukase
- From the Departments of Pediatrics (H.N., S.K., S.O., K.I., T.T., T.H.) and Pathophysiological and Experimental Pathology (H.N., K.S.), Graduate School of Medical Sciences, Kyushu University, Fukuoka; Department of Pediatrics, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Kitakyushu (K.K.); Department of Chemistry, Graduate School of Science, Osaka University, Osaka, Japan (Y.F., K.F.); Japan Society for the Promotion of Science (T.H.), Tokyo, Japan
| | - Katsuo Sueishi
- From the Departments of Pediatrics (H.N., S.K., S.O., K.I., T.T., T.H.) and Pathophysiological and Experimental Pathology (H.N., K.S.), Graduate School of Medical Sciences, Kyushu University, Fukuoka; Department of Pediatrics, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Kitakyushu (K.K.); Department of Chemistry, Graduate School of Science, Osaka University, Osaka, Japan (Y.F., K.F.); Japan Society for the Promotion of Science (T.H.), Tokyo, Japan
| | - Toshiro Hara
- From the Departments of Pediatrics (H.N., S.K., S.O., K.I., T.T., T.H.) and Pathophysiological and Experimental Pathology (H.N., K.S.), Graduate School of Medical Sciences, Kyushu University, Fukuoka; Department of Pediatrics, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Kitakyushu (K.K.); Department of Chemistry, Graduate School of Science, Osaka University, Osaka, Japan (Y.F., K.F.); Japan Society for the Promotion of Science (T.H.), Tokyo, Japan
| |
Collapse
|