1
|
Marrufo AM, Flores-Mireles AL. Macrophage fate: to kill or not to kill? Infect Immun 2024; 92:e0047623. [PMID: 38829045 PMCID: PMC11385966 DOI: 10.1128/iai.00476-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024] Open
Abstract
Macrophages are dynamic innate immune cells that either reside in tissue, serving as sentinels, or recruited as monocytes from bone marrow into inflamed and infected tissue. In response to cues in the tissue microenvironment (TME), macrophages polarize on a continuum toward M1 or M2 with diverse roles in progression and resolution of disease. M1-like macrophages exhibit proinflammatory functions with antimicrobial and anti-tumorigenic activities, while M2-like macrophages have anti-inflammatory functions that generally resolve inflammatory responses and orchestrate a tissue healing process. Given these opposite phenotypes, proper spatiotemporal coordination of macrophage polarization in response to cues within the TME is critical to effectively resolve infectious disease and regulate wound healing. However, if this spatiotemporal coordination becomes disrupted due to persistent infection or dysregulated coagulation, macrophages' inappropriate response to these cues will result in the development of diseases with clinically unfavorable outcomes. Since plasticity and heterogeneity are hallmarks of macrophages, they are attractive targets for therapies to reprogram toward specific phenotypes that could resolve disease and favor clinical prognosis. In this review, we discuss how basic science studies have elucidated macrophage polarization mechanisms in TMEs during infections and inflammation, particularly coagulation. Therefore, understanding the dynamics of macrophage polarization within TMEs in diseases is important in further development of targeted therapies.
Collapse
Affiliation(s)
- Armando M. Marrufo
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | | |
Collapse
|
2
|
Zhou X, Zhang X, Yu J. Gut mycobiome in metabolic diseases: Mechanisms and clinical implication. Biomed J 2024; 47:100625. [PMID: 37364760 PMCID: PMC11332988 DOI: 10.1016/j.bj.2023.100625] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/22/2023] [Accepted: 06/21/2023] [Indexed: 06/28/2023] Open
Abstract
Obesity, type 2 diabetes mellitus (T2DM) and non-alcoholic fatty liver disease (NAFLD) are three common metabolic diseases with high prevalence worldwide. Emerging evidence suggests that gut dysbiosis may influence the development of metabolic diseases, in which gut fungal microbiome (mycobiome) is actively involved. In this review, we summarize the studies exploring the composition changes of gut mycobiome in metabolic diseases and mechanisms by which fungi affect the development of metabolic diseases. The current mycobiome-based therapies, including probiotic fungi, fungal products, anti-fungal agents and fecal microbiota transplantation (FMT), and their implication in treating metabolic diseases are discussed. We highlight the unique role of gut mycobiome in metabolic diseases, providing perspectives for future research on gut mycobiome in metabolic diseases.
Collapse
Affiliation(s)
- Xingyu Zhou
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiang Zhang
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
3
|
Santiso A, Heinemann A, Kargl J. Prostaglandin E2 in the Tumor Microenvironment, a Convoluted Affair Mediated by EP Receptors 2 and 4. Pharmacol Rev 2024; 76:388-413. [PMID: 38697857 DOI: 10.1124/pharmrev.123.000901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 05/05/2024] Open
Abstract
The involvement of the prostaglandin E2 (PGE2) system in cancer progression has long been recognized. PGE2 functions as an autocrine and paracrine signaling molecule with pleiotropic effects in the human body. High levels of intratumoral PGE2 and overexpression of the key metabolic enzymes of PGE2 have been observed and suggested to contribute to tumor progression. This has been claimed for different types of solid tumors, including, but not limited to, lung, breast, and colon cancer. PGE2 has direct effects on tumor cells and angiogenesis that are known to promote tumor development. However, one of the main mechanisms behind PGE2 driving cancerogenesis is currently thought to be anchored in suppressed antitumor immunity, thus providing possible therapeutic targets to be used in cancer immunotherapies. EP2 and EP4, two receptors for PGE2, are emerging as being the most relevant for this purpose. This review aims to summarize the known roles of PGE2 in the immune system and its functions within the tumor microenvironment. SIGNIFICANCE STATEMENT: Prostaglandin E2 (PGE2) has long been known to be a signaling molecule in cancer. Its presence in tumors has been repeatedly associated with disease progression. Elucidation of its effects on immunological components of the tumor microenvironment has highlighted the potential of PGE2 receptor antagonists in cancer treatment, particularly in combination with immune checkpoint inhibitor therapeutics. Adjuvant treatment could increase the response rates and the efficacy of immune-based therapies.
Collapse
Affiliation(s)
- Ana Santiso
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Akos Heinemann
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Julia Kargl
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| |
Collapse
|
4
|
Babalola KT, Arora M, Ganugula R, Agarwal SK, Mohan C, Kumar MNVR. Leveraging Lymphatic System Targeting in Systemic Lupus Erythematosus for Improved Clinical Outcomes. Pharmacol Rev 2024; 76:228-250. [PMID: 38351070 PMCID: PMC10877736 DOI: 10.1124/pharmrev.123.000938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 11/21/2023] [Accepted: 12/06/2023] [Indexed: 02/16/2024] Open
Abstract
The role of advanced drug delivery strategies in drug repositioning and minimizing drug attrition rates, when applied early in drug discovery, is poised to increase the translational impact of various therapeutic strategies in disease prevention and treatment. In this context, drug delivery to the lymphatic system is gaining prominence not only to improve the systemic bioavailability of various pharmaceutical drugs but also to target certain specific diseases associated with the lymphatic system. Although the role of the lymphatic system in lupus is known, very little is done to target drugs to yield improved clinical benefits. In this review, we discuss recent advances in drug delivery strategies to treat lupus, the various routes of drug administration leading to improved lymph node bioavailability, and the available technologies applied in other areas that can be adapted to lupus treatment. Moreover, this review also presents some recent findings that demonstrate the promise of lymphatic targeting in a preclinical setting, offering renewed hope for certain pharmaceutical drugs that are limited by efficacy in their conventional dosage forms. These findings underscore the potential and feasibility of such lymphatic drug-targeting approaches to enhance therapeutic efficacy in lupus and minimize off-target effects of the pharmaceutical drugs. SIGNIFICANCE STATEMENT: The World Health Organization estimates that there are currently 5 million humans living with some form of lupus. With limited success in lupus drug discovery, turning to effective delivery strategies with existing drug molecules, as well as those in the early stage of discovery, could lead to better clinical outcomes. After all, effective delivery strategies have been proven to improve treatment outcomes.
Collapse
Affiliation(s)
- K T Babalola
- The Center for Convergent Bioscience and Medicine (CCBM) (K.T.B., M.A., R.G., M.N.V.R.K.), Division of Translational Science and Medicine, College of Community Health Sciences (K.T.B., M.A., R.G., M.N.V.R.K.), Alabama Life Research Institute (K.T.B., M.A., R.G., M.N.V.R.K.), and Department of Biological Sciences (M.A., R.G., M.N.V.R.K.), The University of Alabama, Tuscaloosa, Alabama; Section of Immunology, Allergy and Rheumatology, Department of Medicine, Biology of Inflammation Baylor College of Medicine, One Baylor Plaza, Houston, Texas (S.K.A.); Department of Biomedical Engineering, University of Houston, Houston, Texas (C.M.); Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama (M.N.V.R.K.); and Center for Free Radical Biology (M.N.V.R.K.) and Nephrology Research and Training Center, Division of Nephrology, Department of Medicine (M.N.V.R.K.), University of Alabama at Birmingham, Birmingham, Alabama
| | - M Arora
- The Center for Convergent Bioscience and Medicine (CCBM) (K.T.B., M.A., R.G., M.N.V.R.K.), Division of Translational Science and Medicine, College of Community Health Sciences (K.T.B., M.A., R.G., M.N.V.R.K.), Alabama Life Research Institute (K.T.B., M.A., R.G., M.N.V.R.K.), and Department of Biological Sciences (M.A., R.G., M.N.V.R.K.), The University of Alabama, Tuscaloosa, Alabama; Section of Immunology, Allergy and Rheumatology, Department of Medicine, Biology of Inflammation Baylor College of Medicine, One Baylor Plaza, Houston, Texas (S.K.A.); Department of Biomedical Engineering, University of Houston, Houston, Texas (C.M.); Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama (M.N.V.R.K.); and Center for Free Radical Biology (M.N.V.R.K.) and Nephrology Research and Training Center, Division of Nephrology, Department of Medicine (M.N.V.R.K.), University of Alabama at Birmingham, Birmingham, Alabama
| | - R Ganugula
- The Center for Convergent Bioscience and Medicine (CCBM) (K.T.B., M.A., R.G., M.N.V.R.K.), Division of Translational Science and Medicine, College of Community Health Sciences (K.T.B., M.A., R.G., M.N.V.R.K.), Alabama Life Research Institute (K.T.B., M.A., R.G., M.N.V.R.K.), and Department of Biological Sciences (M.A., R.G., M.N.V.R.K.), The University of Alabama, Tuscaloosa, Alabama; Section of Immunology, Allergy and Rheumatology, Department of Medicine, Biology of Inflammation Baylor College of Medicine, One Baylor Plaza, Houston, Texas (S.K.A.); Department of Biomedical Engineering, University of Houston, Houston, Texas (C.M.); Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama (M.N.V.R.K.); and Center for Free Radical Biology (M.N.V.R.K.) and Nephrology Research and Training Center, Division of Nephrology, Department of Medicine (M.N.V.R.K.), University of Alabama at Birmingham, Birmingham, Alabama
| | - S K Agarwal
- The Center for Convergent Bioscience and Medicine (CCBM) (K.T.B., M.A., R.G., M.N.V.R.K.), Division of Translational Science and Medicine, College of Community Health Sciences (K.T.B., M.A., R.G., M.N.V.R.K.), Alabama Life Research Institute (K.T.B., M.A., R.G., M.N.V.R.K.), and Department of Biological Sciences (M.A., R.G., M.N.V.R.K.), The University of Alabama, Tuscaloosa, Alabama; Section of Immunology, Allergy and Rheumatology, Department of Medicine, Biology of Inflammation Baylor College of Medicine, One Baylor Plaza, Houston, Texas (S.K.A.); Department of Biomedical Engineering, University of Houston, Houston, Texas (C.M.); Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama (M.N.V.R.K.); and Center for Free Radical Biology (M.N.V.R.K.) and Nephrology Research and Training Center, Division of Nephrology, Department of Medicine (M.N.V.R.K.), University of Alabama at Birmingham, Birmingham, Alabama
| | - C Mohan
- The Center for Convergent Bioscience and Medicine (CCBM) (K.T.B., M.A., R.G., M.N.V.R.K.), Division of Translational Science and Medicine, College of Community Health Sciences (K.T.B., M.A., R.G., M.N.V.R.K.), Alabama Life Research Institute (K.T.B., M.A., R.G., M.N.V.R.K.), and Department of Biological Sciences (M.A., R.G., M.N.V.R.K.), The University of Alabama, Tuscaloosa, Alabama; Section of Immunology, Allergy and Rheumatology, Department of Medicine, Biology of Inflammation Baylor College of Medicine, One Baylor Plaza, Houston, Texas (S.K.A.); Department of Biomedical Engineering, University of Houston, Houston, Texas (C.M.); Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama (M.N.V.R.K.); and Center for Free Radical Biology (M.N.V.R.K.) and Nephrology Research and Training Center, Division of Nephrology, Department of Medicine (M.N.V.R.K.), University of Alabama at Birmingham, Birmingham, Alabama
| | - M N V Ravi Kumar
- The Center for Convergent Bioscience and Medicine (CCBM) (K.T.B., M.A., R.G., M.N.V.R.K.), Division of Translational Science and Medicine, College of Community Health Sciences (K.T.B., M.A., R.G., M.N.V.R.K.), Alabama Life Research Institute (K.T.B., M.A., R.G., M.N.V.R.K.), and Department of Biological Sciences (M.A., R.G., M.N.V.R.K.), The University of Alabama, Tuscaloosa, Alabama; Section of Immunology, Allergy and Rheumatology, Department of Medicine, Biology of Inflammation Baylor College of Medicine, One Baylor Plaza, Houston, Texas (S.K.A.); Department of Biomedical Engineering, University of Houston, Houston, Texas (C.M.); Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama (M.N.V.R.K.); and Center for Free Radical Biology (M.N.V.R.K.) and Nephrology Research and Training Center, Division of Nephrology, Department of Medicine (M.N.V.R.K.), University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
5
|
Liu D, Wang L, Hu Z, Ma Z, Yang Q. Association Between SPARC Polymorphisms and Ankylosing Spondylitis and Its mRNA and Protein Expression in a Chinese Han Population: A Case-Control Study. Int J Gen Med 2023; 16:3533-3542. [PMID: 37605781 PMCID: PMC10440112 DOI: 10.2147/ijgm.s419094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/05/2023] [Indexed: 08/23/2023] Open
Abstract
Objective We explore the association of polymorphisms in Secreted protein acidic and rich in cysteine (SPARC) with ankylosing spondylitis (AS) and detect SPARC mRNA and protein expression in a Chinese Han population. Methods Nine single-nucleotide polymorphisms (SNPs) of SPARC were genotyped in 768 AS patients and 768 controls by TaqMan genotyping assay. mRNA expression of SPARC was detected by real-time polymerase chain reaction (RT-PCR), and serum level of SPARC protein was detected by ELISA. Results The frequency of A allele of rs171121187 was significantly higher in AS patients than in controls (Pc=0.003, odds ratio [OR]=1.45, 95% confidence interval [95% CI] = 1.18-1.77), the AA and AC genotypes increased the risk of AS when compared with CC genotype (Pc=0.003, OR=3.96, 95% CI=1.80-8.75, and Pc=0.003, OR=1.27, 95% CI=1.01-1.61, respectively). The frequency of G allele of rs4958487 was significantly lower in AS than in controls (Pc=0.001, OR=0.60, 95% CI=0.47-0.68), the GG and GA genotypes reduced the risk of AS when compared with AA genotype (Pc=0.005, OR=0.46, 95% CI 0.18-1.14, and Pc=0.005, OR=0.60, 95% CI=0.45-0.79, respectively). The haplotype AA of rs17112187/rs4958487 significantly increased the risk of AS (P=2.31E-5, OR=1.60, 95% CI=1.28-1.98), while haplotype CG decreased the risk of AS (P=5.42E-5, OR=0.55, 95% CI=0.41-0.74). Expression levels of SPARC mRNA were significantly lower in both Peripheral blood mononuclear cells (PBMC) and granulocytes in AS patients than in controls (P=0.008 and P=0.005, respectively). SPARC protein levels were also reduced in AS patients versus the controls (P=0.002). Conclusion This study indicates that polymorphisms in SPARC are associated with AS susceptibility, and both mRNA and protein levels of SPARC are decreased in AS patients in a Chinese Han population.
Collapse
Affiliation(s)
- Dongxia Liu
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People’s Republic of China
| | - Liya Wang
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People’s Republic of China
- Department of Rheumatology and Immunology, Heze Medical College, Heze, 274000, People’s Republic of China
| | - Zhongdan Hu
- Tianjin Zhonghe Clinic Co. Ltd. of Heping District, Tianjin, 300450, People’s Republic of China
| | - Zhenzhen Ma
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People’s Republic of China
| | - Qingrui Yang
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People’s Republic of China
| |
Collapse
|
6
|
Hartmann P, Schnabl B. Fungal infections and the fungal microbiome in hepatobiliary disorders. J Hepatol 2023; 78:836-851. [PMID: 36565724 PMCID: PMC10033447 DOI: 10.1016/j.jhep.2022.12.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022]
Abstract
Liver and biliary diseases affect more than a billion people worldwide, with high associated morbidity and mortality. The impact of the intestinal bacterial microbiome on liver diseases has been well established. However, the fungal microbiome, or mycobiome, has been overlooked for a long time. Recently, several studies have shed light on the role of the mycobiome in the development and progression of hepatobiliary diseases. In particular, the fungal genus Candida has been found to be involved in the pathogenesis of multiple hepatobiliary conditions. Herein, we compare colonisation and infection, describe mycobiome findings in the healthy state and across the various hepatobiliary conditions, and point toward communalities. We detail how quantitation of immune responses to fungal antigens can be employed to predict disease severity, e.g. using antibodies to Saccharomyces cerevisiae or specific anti-Candida albicans antibodies. We also show how fungal products (e.g. beta-glucans, candidalysin) activate the host's immune system to exacerbate liver and biliary diseases. Finally, we describe how the gut mycobiome can be modulated to ameliorate hepatobiliary conditions.
Collapse
Affiliation(s)
- Phillipp Hartmann
- Department of Medicine, University of California San Diego, La Jolla, CA, USA; Department of Pediatrics, University of California San Diego, La Jolla, CA, USA; Division of Gastroenterology, Hepatology & Nutrition, Rady Children's Hospital San Diego, San Diego, CA, USA
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA; Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
7
|
Comparative immunophenotyping of Saccharomyces cerevisiae and Candida spp. strains from Crohn's disease patients and their interactions with the gut microbiome. J Transl Autoimmun 2020; 3:100036. [PMID: 32743520 PMCID: PMC7388382 DOI: 10.1016/j.jtauto.2020.100036] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/19/2019] [Accepted: 01/05/2020] [Indexed: 02/06/2023] Open
Abstract
Investigation of the fungal communities in animal models of Inflammatory Bowel Diseases (IBD) showed a controversial role of Saccharomyces cerevisiae and Candida spp. In health and disease. These conflicting observations could be ascribed to immunogenic differences among co-specific strains. To assess the relevance of intra-strains differences on yeast immunogenicity and impact on the microbiota, we screened S. cerevisiae and Candida spp. Strains isolated from fecal samples of IBD patients. We compared the cytokine profiles, obtained upon stimulation of Peripheral Blood Mononuclear Cells (PBMCs) and Dendritic Cells with different yeast strains, and evaluated the relationship between strain’s cell wall sugar amount and immune response. Moreover, the gut microbiota composition was explored in relation to fungal isolation from fecal samples by metabarcoding analysis. The comparison of cytokine profiles showed strain dependent rather than species-dependent differences in immune responses. Differences in immunogenicity correlated with the cell wall composition of S. cerevisiae intestinal strains. Stimulation of human healthy PBMCs with different strains showed a pro-inflammatory IL-6 response counterbalanced by IL-10 production. Interestingly, Crohn’s (CD) patients responded differently to “self” and “non-self” strains, eliciting pure Th1 or Th17 cytokine patterns. The differences observed in vitro were recapitulated in vivo, where different strains contributed in dramatically different ways to local epithelial activity and to the inflammation of wild type and Interleukin-deficient mice. Furthermore, we observed that the gut microbiota profiles significantly differentiated according to the presence of Saccharomyces or Candida spp. or the absence of fungal isolates in fecal samples. Our results show the importance to deepen metagenomics and immunophenotyping analyses to the strain level, to elucidate the role of fungal and bacterial communities in health and disease. Previous studies indicated an involvement of gut mycobiome in IBD pathogenesis. We screened for immunomodulatory properties S. cerevisiae and Candida strains from IBD patients. The fungal immunomodulation depends on strain-rather than species-specific traits. Differences in immunogenicity correlate with the cell wall composition of gut strains. CD patients responded differently to “self” and “non-self” strains.
Collapse
|
8
|
Abraham A, Ostroff G, Levitz SM, Oyston PCF. A novel vaccine platform using glucan particles for induction of protective responses against Francisella tularensis and other pathogens. Clin Exp Immunol 2019; 198:143-152. [PMID: 31400225 PMCID: PMC6797901 DOI: 10.1111/cei.13356] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2019] [Indexed: 12/13/2022] Open
Abstract
Vaccines are considered the bedrock of preventive medicine. However, for many pathogens, it has been challenging to develop vaccines that stimulate protective, long-lasting immunity. We have developed a novel approach using β-1,3-D-glucans (BGs), natural polysaccharides abundantly present in fungal cell walls, as a biomaterial platform for vaccine delivery. BGs simultaneously provide for receptor-targeted antigen delivery to specialized antigen-presenting cells together with adjuvant properties to stimulate antigen-specific and trained non-specific immune responses. This review focuses on various approaches of using BG particles (GPs) to develop bacterial and fungal vaccine candidates. A special case history for the development of an effective GP tularaemia vaccine candidate is highlighted.
Collapse
Affiliation(s)
- A. Abraham
- University of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - G. Ostroff
- University of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - S. M. Levitz
- University of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - P. C. F. Oyston
- CBR Division, Defence Science and Technology Laboratory, Porton DownSalisburyUK
| |
Collapse
|
9
|
Tan TG, Lim YS, Tan A, Leong R, Pavelka N. Fungal Symbionts Produce Prostaglandin E 2 to Promote Their Intestinal Colonization. Front Cell Infect Microbiol 2019; 9:359. [PMID: 31681635 PMCID: PMC6813641 DOI: 10.3389/fcimb.2019.00359] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 10/03/2019] [Indexed: 02/04/2023] Open
Abstract
Candida albicans is a ubiquitous fungal symbiont that resides on diverse human barrier surfaces. Both mammalian and fungal cells can convert arachidonic acid into the lipid mediator, prostaglandin E2 (PGE2), but the physiological significance of fungus-derived PGE2 remains elusive. Here we report that a C. albicans mutant deficient in PGE2 production suffered a loss of competitive fitness in the murine gastrointestinal (GI) tract and that PGE2 supplementation mitigated this fitness defect. Impaired fungal PGE2 production affected neither the in vitro fitness of C. albicans nor hyphal morphogenesis and virulence in either systemic or mucosal infection models. Instead, fungal production of PGE2 was associated with enhanced fungal survival within phagocytes. Consequently, ablation of colonic phagocytes abrogated the intra-GI fitness boost conferred by fungal PGE2. These observations suggest that C. albicans has evolved the capacity to produce PGE2 from arachidonic acid, a host-derived precursor, to promote its own colonization of the host gut. Analogous mechanisms might undergird host-microbe interactions of other symbiont fungi.
Collapse
Affiliation(s)
- Tze Guan Tan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | | | | | | | - Norman Pavelka
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
10
|
Abstract
Eicosanoids are bioactive lipid mediators generated in almost all mammalian cells from the oxidation of arachidonic acid and other related twenty-carbon polyunsaturated fatty acids (PUFA). Eicosanoids regulate various physiological functions, including cellular homoeostasis and modulation of inflammatory responses in mammals. The mode of action of these lipid mediators depend on their binding to different G-protein coupled receptors. The three main enzymatic pathways associated with their production are the COX pathway, LOX pathway and cytochrome P450 pathway. Interestingly, investigations have also revealed that several human pathogenic fungi are capable of producing these bioactive lipid mediators; however, the exact biosynthetic pathways and their function in pathogenicity are not yet extensively characterized. The aim of the current review is to summarize the recent discoveries pertaining to eicosanoid production by human pathogenic yeasts with a special focus on the opportunistic human fungal pathogen Candida parapsilosis.
Collapse
Affiliation(s)
- Tanmoy Chakraborty
- Interdisciplinary Excellence Centre, Department of Microbiology, University of Szeged, Szeged, Hungary
| | - Renáta Tóth
- Interdisciplinary Excellence Centre, Department of Microbiology, University of Szeged, Szeged, Hungary
| | - Attila Gácser
- Interdisciplinary Excellence Centre, Department of Microbiology, University of Szeged, Szeged, Hungary.,MTA-SZTE "Lendület" "Mycobiome" Research Group, University of Szeged, Szeged, Hungary
| |
Collapse
|
11
|
Ueno A, Jeffery L, Kobayashi T, Hibi T, Ghosh S, Jijon H. Th17 plasticity and its relevance to inflammatory bowel disease. J Autoimmun 2018; 87:38-49. [DOI: 10.1016/j.jaut.2017.12.004] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 12/03/2017] [Indexed: 02/08/2023]
|
12
|
Rahman MA, Thomas R. The SKG model of spondyloarthritis. Best Pract Res Clin Rheumatol 2017; 31:895-909. [DOI: 10.1016/j.berh.2018.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/06/2018] [Indexed: 12/21/2022]
|
13
|
Wakayama N, Matsune S, Okubo K. Delayed type of allergic skin reaction to Candida albicans in eosinophilic rhinosinusitis cases. Auris Nasus Larynx 2017; 45:111-115. [PMID: 28552275 DOI: 10.1016/j.anl.2017.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 05/11/2017] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Eosinophilic chronic rhinosinusitis (ECRS) is frequently complicated by asthma, and recognized as refractory and persistent rhinosinusitis. However, the detailed pathophysiology of ECRS has not been elucidated yet. In this study, we investigated the association between recurrent ECRS and intradermal testing to multi-antigens including Candida albicans. METHODS The subjects were 49 cases of bilateral chronic rhinosinusitis including 24 ECRS cases. They underwent endoscopic sinus surgery and submitted to pathological examination. Prior to surgery, peripheral blood eosinophil count, total and antigen-specific IgE levels (11 categories), and intradermal tests (5 categories) were carried out in all patients. These patients were followed-up for longer than 3 months. We compared the results of preoperative and postoperative clinical examination data between ECRS and non-ECRS (NECRS) cases. RESULTS Positive reaction of the delayed type of intradermal testing to C. albicans was significantly more often observed in ECRS than NECRS cases. (P<0.01) Additionally, these positive reaction cases exhibited significantly higher recurrence of nasal polyps and symptoms of ECRS (P<0.05). CONCLUSION These results suggest the involvement of (Coombs) type IV allergic reaction to C. albicans in the pathophysiology of ECRS.
Collapse
Affiliation(s)
- Nozomu Wakayama
- Department of Otolaryngology, Nippon Medical School Musashikosugi Hospital, 1-396, Kosugimachi, Nakahara-ku Kawasaki-shi, Kanagawa Prefecture 211-0063, Japan.
| | - Shoji Matsune
- Department of Otolaryngology, Nippon Medical School Musashikosugi Hospital, 1-396, Kosugimachi, Nakahara-ku Kawasaki-shi, Kanagawa Prefecture 211-0063, Japan
| | - Kimihiro Okubo
- Department of Otolaryngology, Nippon Medical School Musashikosugi Hospital, 1-396, Kosugimachi, Nakahara-ku Kawasaki-shi, Kanagawa Prefecture 211-0063, Japan
| |
Collapse
|
14
|
Bhaumik S, Basu R. Cellular and Molecular Dynamics of Th17 Differentiation and its Developmental Plasticity in the Intestinal Immune Response. Front Immunol 2017; 8:254. [PMID: 28408906 PMCID: PMC5374155 DOI: 10.3389/fimmu.2017.00254] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 02/21/2017] [Indexed: 01/15/2023] Open
Abstract
After emerging from the thymus, naive CD4 T cells circulate through secondary lymphoid tissues, including gut-associated lymphoid tissue of the intestine. The activation of naïve CD4 T cells by antigen-presenting cells offering cognate antigen initiate differentiation programs that lead to the development of highly specialized T helper (Th) cell lineages. Although initially believed that developmental programing of effector T cells such as T helper 1 (Th1) or T helper 2 (Th2) resulted in irreversible commitment to a fixed fate, subsequent studies have demonstrated greater flexibility, or plasticity, in effector T cell stability than originally conceived. This is particularly so for the Th17 subset, differentiation of which is a highly dynamic process with overlapping developmental axes with inducible regulatory T (iTreg), T helper 22 (Th22), and Th1 cells. Accordingly, intermediary stages of Th17 cells are found in various tissues, which co-express lineage-specific transcription factor(s) or cytokine(s) of developmentally related CD4 T cell subsets. A highly specialized tissue like that of the intestine, which harbors the largest immune compartment of the body, adds several layers of complexity to the intricate process of Th differentiation. Due to constant exposure to millions of commensal microbes and periodic exposure to pathogens, the intestinal mucosa maintains a delicate balance between regulatory and effector T cells. It is becoming increasingly clear that equilibrium between tolerogenic and inflammatory axes is maintained in the intestine by shuttling the flexible genetic programming of a developing CD4 T cell along the developmental axis of iTreg, Th17, Th22, and Th1 subsets. Currently, Th17 plasticity remains an unresolved concern in the field of clinical research as targeting Th17 cells to cure immune-mediated disease might also target its related subsets. In this review, we discuss the expanding sphere of Th17 plasticity through its shared developmental axes with related cellular subsets such as Th22, Th1, and iTreg in the context of intestinal inflammation and also examine the molecular and epigenetic features of Th17 cells that mediate these overlapping developmental programs.
Collapse
Affiliation(s)
- Suniti Bhaumik
- Division of Anatomic Pathology, Department of Pathology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Rajatava Basu
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| |
Collapse
|
15
|
Milanez FM, Saad CGS, Viana VT, Moraes JCB, Périco GV, Sampaio-Barros PD, Goncalves CR, Bonfá E. IL-23/Th17 axis is not influenced by TNF-blocking agents in ankylosing spondylitis patients. Arthritis Res Ther 2016; 18:52. [PMID: 26912133 PMCID: PMC4765065 DOI: 10.1186/s13075-016-0949-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 02/08/2016] [Indexed: 12/31/2022] Open
Abstract
Background Advances in pathophysiology and treatment of ankylosing spondylitis (AS) was recently demonstrated. However, the effect of anti-TNF in the newly described inflammatory pathways involved pathogenesis of this disease remains to be determined. The aim of our study was, therefore, to investigate long-term influence of anti-TNF drugs in IL-23/IL-17 axis of AS patients and their possible correlation with treatment, clinical, laboratory and radiographic parameters. Methods Eighty-six AS anti-TNF naïve patients, 47 referred for anti-TNF therapy (active-AS; BASDAI ≥ 4) and 39 with BASDAI < 4 (control-AS) were included. The active group was evaluated at baseline, 12-months and 24-months after TNF blockade and compared at baseline to control-AS group and to 47 healthy age- and gender-matched controls. Plasma levels of IL-17A, IL-22, IL-23 and PGE2 were measured. Non-steroidal anti-inflammatory drugs (NSAIDs) intake were recorded every 6 months. Radiographic severity and progression was assessed by mSASSS at baseline and 24 months after therapy. Results At baseline, active-AS group presented higher IL-23 and PGE2 levels compared to control-AS group (p < 0.001 and p = 0.008) and to healthy controls (p < 0.001 and p = 0.02). After 24-months of TNF blockade, IL-23 and PGE2 remained elevated with higher levels compared with the healthy group (p < 0.001 and p = 0.03) in spite of significant improvements in all clinical/inflammatory parameters (p < 0.001). Further analysis of 27 anti-TNF-treated patients who achieved a good response (ASDAS-CRP < 2.1,with a drop ≥ 1.1) at 24-months revealed that IL-23 plasma levels remained higher than healthy controls (p < 0.001) and higher than control-AS group with similar disease activity (ASDAS-CRP < 2.1, p = 0.01). In active-AS group (n = 47), there was a strong correlation between IL-23 and IL-17A at baseline, 12-months and 24-months after anti-TNF therapy (p ≤ 0.001). Conclusion This study provides novel data demonstrating that the IL-23/IL-17 axis is not influenced by TNF blockade in AS patients despite clinical and inflammation improvements and NSAID intake.
Collapse
Affiliation(s)
- Fernanda Manente Milanez
- Division of Rheumatology, Faculdade de Medicina da Universidade de São Paulo - Reumatologia, Av. Dr. Arnaldo, n° 455, 3° andar, sala 3192, São Paulo, SP, 05403-010, Brazil.
| | - Carla G S Saad
- Division of Rheumatology, Faculdade de Medicina da Universidade de São Paulo - Reumatologia, Av. Dr. Arnaldo, n° 455, 3° andar, sala 3192, São Paulo, SP, 05403-010, Brazil.
| | - Vilma T Viana
- Division of Rheumatology, Faculdade de Medicina da Universidade de São Paulo - Reumatologia, Av. Dr. Arnaldo, n° 455, 3° andar, sala 3192, São Paulo, SP, 05403-010, Brazil.
| | - Júlio C B Moraes
- Division of Rheumatology, Faculdade de Medicina da Universidade de São Paulo - Reumatologia, Av. Dr. Arnaldo, n° 455, 3° andar, sala 3192, São Paulo, SP, 05403-010, Brazil.
| | - Grégory Vinícius Périco
- URC - Unidade Radiológica Criciúma, Rua Antonio de Lucca, 139 - Centro - Criciúma, Santa Catarina, SC, 88811-503, Brazil.
| | - Percival Degrava Sampaio-Barros
- Division of Rheumatology, Faculdade de Medicina da Universidade de São Paulo - Reumatologia, Av. Dr. Arnaldo, n° 455, 3° andar, sala 3192, São Paulo, SP, 05403-010, Brazil.
| | - Célio R Goncalves
- Division of Rheumatology, Faculdade de Medicina da Universidade de São Paulo - Reumatologia, Av. Dr. Arnaldo, n° 455, 3° andar, sala 3192, São Paulo, SP, 05403-010, Brazil.
| | - Eloísa Bonfá
- Division of Rheumatology, Faculdade de Medicina da Universidade de São Paulo - Reumatologia, Av. Dr. Arnaldo, n° 455, 3° andar, sala 3192, São Paulo, SP, 05403-010, Brazil.
| |
Collapse
|
16
|
Li Y, Sheng K, Chen J, Wu Y, Zhang F, Chang Y, Wu H, Fu J, Zhang L, Wei W. Regulation of PGE2 signaling pathways and TNF-alpha signaling pathways on the function of bone marrow-derived dendritic cells and the effects of CP-25. Eur J Pharmacol 2015; 769:8-21. [PMID: 26415983 DOI: 10.1016/j.ejphar.2015.09.036] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Revised: 09/22/2015] [Accepted: 09/23/2015] [Indexed: 12/29/2022]
Abstract
This study was to investigate PGE2 and TNF-alpha signaling pathway involving in the maturation and activation of bone marrow dendritic cells (DCs) and the effect of CP-25. Bone marrow DCs were isolated and stimulated by PGE2 and TNF-alpha respectively. The markers of maturation and activation expressed on DCs, such as CD40, CD80, CD83, CD86, MHC-II, and the ability of antigen uptake of DCs were analyzed by flow cytometry. The proliferation of T cells co-cultured with DCs, the signaling pathways of PGE2-EP4-cAMP and TNF-alpha-TRADD-TRAF2-NF-κB in DCs were analyzed. The results showed that both PGE2 and TNF-alpha up-regulated the expressions of CD40, CD80, CD83, CD86, and MHC-II, decreased the antigen uptake of DCs, and DCs stimulated by PGE2 or TNF-alpha could increase T cell proliferation. CP-25 (10(-5), 10(-6), and 10(-7)mol/l) decreased significantly the expressions of CD40, CD80, CD83, CD86 and MHC-II, increased the antigen uptake of DCs, and suppressed T cell proliferation induced by DCs. PGE2 increased the expressions of EP4, NF-κB and down-regulated cAMP level of DCs. TNF-alpha could also up-regulate TNFR1, TRADD, TRAF2, and NF-κB expression of DCs. CP-25 (10(-5), 10(-6), and 10(-7)mol/l) decreased the expressions of EP4 and NF-κB, increased cAMP level in DCs stimulated by PGE2. CP-25 (10(-5), 10(-6), and 10(-7)mol/l) also could down-regulate significantly TNFR1, TRADD, TRAF2, and NF-κB expression in DCs stimulated by TNF-alpha. These results demonstrate that PGE2 and TNF-alpha could enhance DCs functions by mediating PGE2-EP4-cAMP pathway, TNF-alpha-TNFR1-TRADD-TRAF2-NF-κB pathway respectively. CP-25 might inhibit the function of DCs through regulating PGE2-EP4-cAMP and TNF-alpha-TNFR1-TRADD-TRAF2-NF-κB pathways.
Collapse
Affiliation(s)
- Ying Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, Anhui Province, China
| | - Kangliang Sheng
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, Anhui Province, China
| | - Jingyu Chen
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, Anhui Province, China
| | - Yujing Wu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, Anhui Province, China
| | - Feng Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, Anhui Province, China
| | - Yan Chang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, Anhui Province, China
| | - Huaxun Wu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, Anhui Province, China
| | - Jingjing Fu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, Anhui Province, China
| | - Lingling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, Anhui Province, China.
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, Anhui Province, China.
| |
Collapse
|
17
|
Shen L, Liu Y. Prostaglandin E2 blockade enhances the pulmonary anti-Cryptococcus neoformans immune reaction via the induction of TLR-4. Int Immunopharmacol 2015; 28:376-81. [PMID: 26122137 DOI: 10.1016/j.intimp.2015.06.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 06/18/2015] [Accepted: 06/18/2015] [Indexed: 10/23/2022]
Abstract
The present study aimed to explore whether the inhibition of prostaglandin E2 enhances pulmonary anti-Cryptococcus neoformans immunity. Lung colony forming unit (CFU) assays demonstrated that the cryptococcal infection was dramatically depressed in mice given EP2 and EP4 or single EP antagonist treatment compared to the untreated wild type mice (p<0.05), leading to the increased survival of the infected mice by 8-9 days or 2-4 days, respectively. RT-PCR and flow cytometry assays showed that the expression of IFN-γ, IL-17, IL-22 in M1 macrophages and IL-10 in M2 macrophages increased significantly at 1 week post-infection in mice with either EP2 or EP4 blockade (p<0.05). The polarization of alveolar macrophages showed that, at 1 week post infection, the alveolar macrophages in untreated wild type mice, TLR4(-/-) mice and TLR4(-/-) mice with EP2 and EP4 blockade were strongly M2 polarized, whereas the alveolar macrophages in wild type mice with EP2 and EP4 blockade were M1 polarized. In conclusion, the blockade of EP2 and EP4 promotes mouse survival after cryptococcus infection by promoting the production of cytokines via TLR4, as well as the enhanced M1 polarization of alveolar macrophages.
Collapse
Affiliation(s)
- Liyun Shen
- Department of Hematology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, China.
| | - Ying Liu
- Department of Pneumology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, China
| |
Collapse
|
18
|
Ardeshir A, Narayan NR, Méndez-Lagares G, Lu D, Rauch M, Huang Y, Van Rompay KKA, Lynch SV, Hartigan-O'Connor DJ. Breast-fed and bottle-fed infant rhesus macaques develop distinct gut microbiotas and immune systems. Sci Transl Med 2015; 6:252ra120. [PMID: 25186175 DOI: 10.1126/scitranslmed.3008791] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Diet has a strong influence on the intestinal microbiota in both humans and animal models. It is well established that microbial colonization is required for normal development of the immune system and that specific microbial constituents prompt the differentiation or expansion of certain immune cell subsets. Nonetheless, it has been unclear how profoundly diet might shape the primate immune system or how durable the influence might be. We show that breast-fed and bottle-fed infant rhesus macaques develop markedly different immune systems, which remain different 6 months after weaning when the animals begin receiving identical diets. In particular, breast-fed infants develop robust populations of memory T cells as well as T helper 17 (TH17) cells within the memory pool, whereas bottle-fed infants do not. These findings may partly explain the variation in human susceptibility to conditions with an immune basis, as well as the variable protection against certain infectious diseases.
Collapse
Affiliation(s)
- Amir Ardeshir
- California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA
| | - Nicole R Narayan
- California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA. Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA
| | - Gema Méndez-Lagares
- California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA. Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA
| | - Ding Lu
- California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA. Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA
| | - Marcus Rauch
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yong Huang
- Department of Bioengineering and Therapeutic Sciences, School of Pharmacy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Koen K A Van Rompay
- California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA
| | - Susan V Lynch
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dennis J Hartigan-O'Connor
- California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA. Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA. Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
19
|
Du B, Lin C, Bian Z, Xu B. An insight into anti-inflammatory effects of fungal beta-glucans. Trends Food Sci Technol 2015. [DOI: 10.1016/j.tifs.2014.09.002] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
20
|
Benham H, Rehaume LM, Hasnain SZ, Velasco J, Baillet AC, Ruutu M, Kikly K, Wang R, Tseng HW, Thomas GP, Brown MA, Strutton G, McGuckin MA, Thomas R. Interleukin-23 mediates the intestinal response to microbial β-1,3-glucan and the development of spondyloarthritis pathology in SKG mice. Arthritis Rheumatol 2014; 66:1755-67. [PMID: 24664521 DOI: 10.1002/art.38638] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 03/18/2014] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Spondyloarthritides (SpA) occur in 1% of the population and include ankylosing spondylitis (AS) and arthropathy of inflammatory bowel disease (IBD), with characteristic spondylitis, arthritis, enthesitis, and IBD. Genetic studies implicate interleukin-23 (IL-23) receptor signaling in the development of SpA and IBD, and IL-23 overexpression in mice is sufficient for enthesitis, driven by entheseal-resident T cells. However, in genetically prone individuals, it is not clear where IL-23 is produced and how it drives the SpA syndrome, including IBD or subclinical gut inflammation of AS. Moreover, it is unclear why specific tissue involvement varies between patients with SpA. We undertook this study to determine the location of IL-23 production and its role in SpA pathogenesis in BALB/c ZAP-70(W163C)-mutant (SKG) mice injected intraperitoneally with β-1,3-glucan (curdlan). METHODS Eight weeks after curdlan injection in wild-type or IL-17A(-/-) SKG or BALB/c mice, pathology was scored in tissue sections. Mice were treated with anti-IL-23 or anti-IL-22. Cytokine production and endoplasmic reticulum (ER) stress were determined in affected organs. RESULTS In curdlan-treated SKG mice, arthritis, enthesitis, and ileitis were IL-23 dependent. Enthesitis was specifically dependent on IL-17A and IL-22. IL-23 was induced in the ileum, where it amplified ER stress, goblet cell dysfunction, and proinflammatory cytokine production. IL-17A was pathogenic, while IL-22 was protective against ileitis. IL-22+CD3- innate-like cells were increased in lamina propria mononuclear cells of ileitis-resistant BALB/c mice, which developed ileitis after curdlan injection and anti-IL-22. CONCLUSION In response to systemic β-1,3-glucan, intestinal IL-23 provokes local mucosal dysregulation and cytokines driving the SpA syndrome, including IL-17/IL-22-dependent enthesitis. Innate IL-22 production promotes ileal tolerance.
Collapse
Affiliation(s)
- Helen Benham
- University of Queensland and Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Kim YG, Udayanga KGS, Totsuka N, Weinberg JB, Núñez G, Shibuya A. Gut dysbiosis promotes M2 macrophage polarization and allergic airway inflammation via fungi-induced PGE₂. Cell Host Microbe 2014; 15:95-102. [PMID: 24439901 DOI: 10.1016/j.chom.2013.12.010] [Citation(s) in RCA: 278] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 10/02/2013] [Accepted: 12/24/2013] [Indexed: 11/16/2022]
Abstract
Although imbalances in gut microbiota composition, or "dysbiosis," are associated with many diseases, the effects of gut dysbiosis on host systemic physiology are less well characterized. We report that gut dysbiosis induced by antibiotic (Abx) treatment promotes allergic airway inflammation by shifting macrophage polarization in the lung toward the alternatively activated M2 phenotype. Adoptive transfer of alveolar macrophages derived from Abx-treated mice was sufficient to increase allergic airway inflammation. Abx treatment resulted in the overgrowth of a commensal fungal Candida species in the gut and increased plasma concentrations of prostaglandin E₂ (PGE₂), which induced M2 macrophage polarization in the lung. Suppression of PGE₂ synthesis by the cyclooxygenase inhibitors aspirin and celecoxib suppressed M2 macrophage polarization and decreased allergic airway inflammatory cell infiltration in Abx-treated mice. Thus, Abx treatment can cause overgrowth of particular fungal species in the gut and promote M2 macrophage activation at distant sites to influence systemic responses including allergic inflammation.
Collapse
Affiliation(s)
- Yun-Gi Kim
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Japan Science and Technology Agency, Core Research for Evolutional Science and Technology (CREST), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Kankanam Gamage Sanath Udayanga
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Japan Science and Technology Agency, Core Research for Evolutional Science and Technology (CREST), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Naoya Totsuka
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Japan Science and Technology Agency, Core Research for Evolutional Science and Technology (CREST), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Jason B Weinberg
- Department of Pediatrics and Communicable Diseases, Microbiology, and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Gabriel Núñez
- Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Akira Shibuya
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Japan Science and Technology Agency, Core Research for Evolutional Science and Technology (CREST), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Life Science Center of Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan.
| |
Collapse
|
22
|
Abstract
Spondyloarthritis (SpA) is a group of inflammatory rheumatic diseases whose main clinical feature is inflammation of the axial spine. Articular, periarticular, and extra-articular manifestations can also occur, depending on the type of spondyloarthritis. The most common clinical subsets of SpA are ankylosing spondylitis (AS) and psoriatic arthritis (PsA). SpA is a major health challenge given the propensity to affect young adults and the potential requirement for lifelong treatment. Although the precise etiology of SpA is unknown, there is mounting evidence that these diseases are a result of complex interplay of genetic, environmental, and immunological factors. In this review on SpA, we will discuss genetic variants with genome-wide significance, highlight potential clinical application of genetic variants, and discuss challenges in further elucidating the genetic basis of SpA.
Collapse
|
23
|
Suram S, Silveira LJ, Mahaffey S, Brown GD, Bonventre JV, Williams DL, Gow NAR, Bratton DL, Murphy RC, Leslie CC. Cytosolic phospholipase A(2)α and eicosanoids regulate expression of genes in macrophages involved in host defense and inflammation. PLoS One 2013; 8:e69002. [PMID: 23950842 PMCID: PMC3742295 DOI: 10.1371/journal.pone.0069002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 06/03/2013] [Indexed: 12/20/2022] Open
Abstract
The role of Group IVA cytosolic phospholipase A2 (cPLA2α)
activation in regulating macrophage transcriptional responses to
Candida
albicans infection was investigated.
cPLA2α releases arachidonic acid for the production of
eicosanoids. In mouse resident peritoneal macrophages, prostacyclin,
prostaglandin E2 and leukotriene C4 were produced within
minutes of C.
albicans addition before cyclooxygenase 2
expression. The production of TNFα was lower in C.
albicans-stimulated cPLA2α+/+
than cPLA2α-/- macrophages due to an autocrine effect of
prostaglandins that increased cAMP to a greater extent in
cPLA2α+/+ than cPLA2α-/-
macrophages. For global insight, differential gene expression in
C.
albicans-stimulated
cPLA2α+/+ and cPLA2α-/-
macrophages (3 h) was compared by microarray. cPLA2α+/+
macrophages expressed 86 genes at lower levels and 181 genes at higher levels
than cPLA2α-/- macrophages (≥2-fold, p<0.05). Several
pro-inflammatory genes were expressed at lower levels (Tnfα,
Cx3cl1, Cd40, Ccl5,
Csf1, Edn1, CxCr7, Irf1,
Irf4, Akna, Ifnγ, several IFNγ-inducible
GTPases). Genes that dampen inflammation (Socs3,
Il10, Crem, Stat3,
Thbd, Thbs1, Abca1) and
genes involved in host defense (Gja1, Csf3,
Trem1, Hdc) were expressed at higher
levels in cPLA2α+/+ macrophages. Representative genes
expressed lower in cPLA2α+/+ macrophages (Tnfα,
Csf1) were increased by treatment with a prostacyclin receptor
antagonist and protein kinase A inhibitor, whereas genes expressed at higher
levels (Crem, Nr4a2, Il10,
Csf3) were suppressed. The results suggest that
C.
albicans stimulates an autocrine loop in
macrophages involving cPLA2α, cyclooxygenase 1-derived prostaglandins
and increased cAMP that globally effects expression of genes involved in host
defense and inflammation.
Collapse
Affiliation(s)
- Saritha Suram
- Department of Pediatrics, National Jewish Health, Denver, Colorado,
United States of America
| | - Lori J. Silveira
- Division of Biostatistics and Bioinformatics, National Jewish Health,
Denver, Colorado, United States of America
| | - Spencer Mahaffey
- Department of Pediatrics, National Jewish Health, Denver, Colorado,
United States of America
| | - Gordon D. Brown
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United
Kingdom
| | - Joseph V. Bonventre
- Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts,
United States of America
| | - David L. Williams
- Department of Surgery, James H. Quillen College of Medicine, Johnson
City, Tennessee, United States of America
| | - Neil A. R. Gow
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United
Kingdom
| | - Donna L. Bratton
- Department of Pediatrics, National Jewish Health, Denver, Colorado,
United States of America
| | - Robert C. Murphy
- Department of Pharmacology, University of Colorado Denver, Aurora,
Colorado, United States of America
| | - Christina C. Leslie
- Department of Pediatrics, National Jewish Health, Denver, Colorado,
United States of America
- Department of Pharmacology, University of Colorado Denver, Aurora,
Colorado, United States of America
- Department of Pathology, University of Colorado Denver, Aurora, Colorado,
United States of America
- * E-mail:
| |
Collapse
|
24
|
Yokoyama U, Iwatsubo K, Umemura M, Fujita T, Ishikawa Y. The prostanoid EP4 receptor and its signaling pathway. Pharmacol Rev 2013; 65:1010-52. [PMID: 23776144 DOI: 10.1124/pr.112.007195] [Citation(s) in RCA: 203] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
The EP4 prostanoid receptor is one of four receptor subtypes for prostaglandin E2. It belongs to the family of G protein-coupled receptors. It was originally identified, similar to the EP2 receptor as a G(s)α-coupled, adenylyl cyclase-stimulating receptor. EP4 signaling plays a variety of roles through cAMP effectors, i.e., protein kinase A and exchange protein activated by cAMP. However, emerging evidence from studies using pharmacological approaches and genetically modified mice suggests that EP4, unlike EP2, can also be coupled to G(i)α, phosphatidylinositol 3-kinase, β-arrestin, or β-catenin. These signaling pathways constitute unique roles for the EP4 receptor. EP4 is widely distributed in the body and thus plays various physiologic and pathophysiologic roles. In particular, EP4 signaling is closely related to carcinogenesis, cardiac hypertrophy, vasodilation, vascular remodeling, bone remodeling, gastrointestinal homeostasis, renal function, and female reproductive function. In addition to the classic anti-inflammatory action of EP4 on mononuclear cells and T cells, recent evidence has shown that EP4 signaling contributes to proinflammatory action as well. The aim of this review is to present current findings on the biologic functions of the EP4 receptor. In particular, we will discuss its diversity from the standpoint of EP4-mediated signaling.
Collapse
Affiliation(s)
- Utako Yokoyama
- Cardiovascular Research Institute, Yokohama City University, Yokohama, Kanagawa, Japan
| | | | | | | | | |
Collapse
|
25
|
|
26
|
Lipinski T, Fitieh A, St. Pierre J, Ostergaard HL, Bundle DR, Touret N. Enhanced Immunogenicity of a Tricomponent Mannan Tetanus Toxoid Conjugate Vaccine Targeted to Dendritic Cells via Dectin-1 by Incorporating β-Glucan. THE JOURNAL OF IMMUNOLOGY 2013; 190:4116-28. [DOI: 10.4049/jimmunol.1202937] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
27
|
Poloso NJ, Urquhart P, Nicolaou A, Wang J, Woodward DF. PGE2 differentially regulates monocyte-derived dendritic cell cytokine responses depending on receptor usage (EP2/EP4). Mol Immunol 2013; 54:284-95. [PMID: 23337716 DOI: 10.1016/j.molimm.2012.12.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 12/04/2012] [Accepted: 12/14/2012] [Indexed: 02/07/2023]
Abstract
Dendritic cells (DCs) are central players in coordinating immune responses, both innate and adaptive. While the role of lipid mediators in the immune response has been the subject of many investigations, the precise role of prostaglandins has often been plagued by contradictory studies. In this study, we examined the role of PGE(2) on human DC function. Although studies have suggested that PGE(2) specifically plays a role in DC motility and cytokine release profile, the precise receptor usage and signaling pathways involved remain unclear. In this report we found that irrespective of the human donor, monocyte-derived dendritic cells (MoDCs) express three of the four PGE(2) receptor subtypes (EP(2-4)), although only EP(2) and EP(4) were active with respect to cytokine production. Using selective EP receptor antagonists and agonists, we demonstrate that PGE(2) coordinates control of IL-23 release (a promoter of Th17, an autoimmune associated T cell subset) in a dose-dependent manner by differential use of EP(2) and EP(4) receptors in LPS-activated MoDCs. This is in contrast to IL-12, which is dose dependently inhibited by PGE(2) through both receptor subtypes. Low concentrations (∼1-10nM) of PGE(2) promoted IL-23 production via EP(4) receptors, while at higher (>50 nM), but still physiologically relevant concentrations, IL-23 is suppressed by an EP(2) dependent mechanism. These results can be explained by differential regulation of the common subunit, IL-12p40, and IL-23p19, by EP(2) and EP(4). By these means, PGE(2) can act as a regulatory switch of immune responses depending on its concentration in the microenvironment. In addition, we believe these results may also explain why seemingly conflicting biological functions assigned to PGE(2) have been reported in the literature, as the concentration of ligand (PGE(2)) fundamentally alters the nature of the response. This finding also highlights the potential of designing therapeutics which differentially target these receptors.
Collapse
Affiliation(s)
- Neil J Poloso
- 2525 DuPont Dr, RD3-2B, Department Biological Sciences, Allergan Inc., Irvine, CA 92612, USA.
| | | | | | | | | |
Collapse
|
28
|
Robinson PC, Brown MA. The genetics of ankylosing spondylitis and axial spondyloarthritis. Rheum Dis Clin North Am 2012; 38:539-53. [PMID: 23083754 DOI: 10.1016/j.rdc.2012.08.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Ankylosing spondylitis (AS) and spondyloarthritis are strongly genetically determined. The long-standing association with HLA-B27 is well described, although the mechanism by which that association induces AS remains uncertain. Recent developments include the description of HLA-B27 tag single nucleotide polymorphisms in European and Asian populations. An increasing number of non-MHC genetic associations have been reported, which provided amongst other things the first evidence of the involvement of the IL-23 pathway in AS. The association with ERAP1 is now known to be restricted to HLA-B27 positive disease. Preliminary studies on the genetics of axial spondyloarthritis demonstrate a lower HLA-B27 carriage rate compared with AS. Studies with larger samples and including non-European ethnic groups are likely to further advance the understanding of the genetics of AS and spondyloarthritis.
Collapse
Affiliation(s)
- Philip C Robinson
- University of Queensland Diamantina Institute, Princess Alexandra Hospital, Brisbane, Woolloongabba, Queensland 4102, Australia
| | | |
Collapse
|
29
|
Arachidonic acid metabolites in pathogenic yeasts. Lipids Health Dis 2012; 11:100. [PMID: 22873782 PMCID: PMC3475069 DOI: 10.1186/1476-511x-11-100] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 08/03/2012] [Indexed: 01/18/2023] Open
Abstract
Although most of what is known about the biology and function of arachidonic acid metabolites comes from the study of mammalian biology, these compounds can also be produced by lower eukaryotes, including yeasts and other fungi. It is also in this group of organisms that the least is known about the metabolic pathways leading to the production of these compounds as well as the functions of these compounds in the biology of fungi and yeasts. This review will deal with the discovery of oxylipins from polyunsaturated fatty acids, and more specifically the arachidonic acid derived eicosanoids, such as 3-hydroxy eicosatetraenoic acid, prostaglandin F2α and prostaglandin E2, in yeasts starting in the early 1990s. This review will also focus on what is known about the metabolic pathways and/or proteins involved in the production of these compounds in pathogenic yeasts. The possible roles of these compounds in the biology, including the pathology, of these organisms will be discussed.
Collapse
|
30
|
Valdez PA, Vithayathil PJ, Janelsins BM, Shaffer AL, Williamson PR, Datta SK. Prostaglandin E2 suppresses antifungal immunity by inhibiting interferon regulatory factor 4 function and interleukin-17 expression in T cells. Immunity 2012; 36:668-79. [PMID: 22464170 PMCID: PMC3334441 DOI: 10.1016/j.immuni.2012.02.013] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Revised: 12/08/2011] [Accepted: 02/02/2012] [Indexed: 11/20/2022]
Abstract
T helper 17 (Th17) cells play an important role in mucosal host defense through production of the signature cytokines IL-17 and IL-22. Prostaglandin E2 (PGE2) has been shown to enhance IL-17 production by mature Th17 cells. However, when present during Th17 cell differentiation, we found that PGE2 inhibited the transcription factor IRF4 and suppressed production of IL-17 but not IL-22. We show that IRF4 was required for IL-17 expression but inhibited IL-22 expression, highlighting the potential for discordant regulation of these two cytokines in Th17 cells. The pathogenic fungus Cryptococcus neoformans produces PGE2, and we found that it uses PGE2- and IRF4-dependent mechanisms to specifically inhibit induction of IL-17 during Th17 cell differentiation. Blockade of host PGE2 during infection led to increased IL-17 production from CD4(+) T cells and increased survival of mice. These findings suggest that host- or pathogen-derived PGE2 can act directly on Th17 cells during differentiation to inhibit IL-17-dependent antimicrobial responses.
Collapse
Affiliation(s)
- Patricia A Valdez
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
31
|
Stefanelli P, Teloni R, Carannante A, Mariotti S, Nisini R, Gagliardi MC. Neisseria gonorrhoeae triggers the PGE2/IL-23 pathway and promotes IL-17 production by human memory T cells. Prostaglandins Other Lipid Mediat 2012; 99:24-9. [PMID: 22542425 DOI: 10.1016/j.prostaglandins.2012.04.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 03/26/2012] [Accepted: 04/10/2012] [Indexed: 12/27/2022]
Abstract
PGE2 is a potent modulator of the T helper (Th)17 immune response that plays a critical role in the host defense against bacterial, fungal and viral infections. We recently showed high serum levels of interleukin (IL)-17 in patients with gonococcal infection and we hypothesized that Neisseria gonorrhoeae could exploit a PGE2 mediated mechanism to promote IL-17 production. Here we show that N. gonorrhoeae induces human dendritic cell (DC) maturation, secretion of prostaglandin E2 and proinflammatory cytokines, including the pro-Th17 cytokine IL-23. Blocking PGE2 endogenous synthesis selectively reduces IL-23 production by DC in response to gonococcal stimulation, confirming recent data on PGE2/IL-23 crosstalk. N. gonorrhoeae stimulated DC induce a robust IL-17 production by memory CD4(+) T cells and this function correlates with PGE2 production. Our findings delineate a previously unknown role for PGE2 in the immune response to N. gonorrhoeae, suggesting its contribute via Th17 cell expansion.
Collapse
Affiliation(s)
- Paola Stefanelli
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | | | | | | | | | | |
Collapse
|
32
|
Abstract
Ankylosing spondylitis (AS), psoriasis and inflammatory bowel disease (IBD) often coexist in the same patient and in their families. In AS, genes within the MHC region, in particular HLA-B27, account for nearly 25% of disease hereditability, with additional small contributions from genes outside of the MHC locus, including those involved in intracellular antigen processing (that is, ERAP1, which interacts with HLA-B27) and cytokine genes such as those involved in the IL-17-IL-23 pathway. Similar to AS, the strongest genetic signal of susceptibility to psoriasis and psoriatic arthritis also emanates from the MHC region (attributable mostly to HLA-C(*)06:02 although other genes have been implicated), and gene-gene interaction of HLA-C with ERAP1. The remaining hereditary load is from genes involved in cytokine production, specifically genes in the IL-17-IL-23 pathway, the NFκB pathway and the type 2 T-helper pathway. In IBD, similar genetic influences are operative. Indeed, genes important in the regulation of the IL-17-IL-23 pathway and, in Crohn's disease, genes important for autophagy (that is, NOD2 and ATG16L1 and IRGM) have a role in conferring susceptibility of individuals to these diseases. Thus, AS, psoriasis and IBD seem to share similar pathogenic mechanisms of aberrant intracellular antigen processing or elimination of intracellular bacteria and cytokine production, especially in the IL-17-IL-23 pathway.
Collapse
Affiliation(s)
- John D Reveille
- The University of Texas Health Science Center at Houston, MSB 5.270, 6431 Fannin, Houston, TX 77030, USA.
| |
Collapse
|
33
|
Gaston JSH, Goodall JC, Baeten D. Interleukin-23: A central cytokine in the pathogenesis of spondylarthritis. ACTA ACUST UNITED AC 2011; 63:3668-71. [DOI: 10.1002/art.30600] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
34
|
Smeekens SP, van de Veerdonk FL, Joosten LAB, Jacobs L, Jansen T, Williams DL, van der Meer JWM, Kullberg BJ, Netea MG. The classical CD14⁺⁺ CD16⁻ monocytes, but not the patrolling CD14⁺ CD16⁺ monocytes, promote Th17 responses to Candida albicans. Eur J Immunol 2011; 41:2915-24. [PMID: 21695694 DOI: 10.1002/eji.201141418] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 05/30/2011] [Accepted: 06/17/2011] [Indexed: 01/09/2023]
Abstract
In the present study, we investigated the functional differences between cluster of differentiation (CD)14(++) CD16(-) and CD14(+) CD16(+) monocytes during anti-Candida host defense. CD14(++) CD16(-) are the "classical" monocytes and represent the majority of circulating monocytes in humans, while CD14(+) CD16(+) monocytes patrol the vasculature for maintenance of tissue integrity and repair. Both monocyte subsets inhibited the germination of live Candida albicans, and there was no difference in their capacity to phagocytose and kill Candida. Although production of IL-6 and IL-10 induced by C. albicans was found to be similar between monocyte subsets, IL-1β and prostaglandin E2 (PGE2) production was higher in CD14(++) CD16(-) compared with CD14(+) CD16(+) monocytes. In line with the increased production of IL-1β and PGE2, central mediators for inducing Th17 responses, CD14(++) CD16(-) monocytes induced greater Th17 responses upon stimulation with heat-killed C. albicans yeast. The percentage of cells that expressed mannose receptor (MR) was higher in the CD14(++) CD16(-) monocyte subset, and MR-specific stimulation induced higher Th17 responses only in co-cultures of CD14(++) CD16(-) monocytes and CD4 lymphocytes. In conclusion, both monocyte subsets have potent innate antifungal properties, but only CD14(++) CD16(-) monocytes are capable of inducing a potent Th17 response to C. albicans, an important component of antifungal host defense.
Collapse
Affiliation(s)
- Sanne P Smeekens
- Department of Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Evans DM, Spencer CCA, Pointon JJ, Su Z, Harvey D, Kochan G, Oppermann U, Opperman U, Dilthey A, Pirinen M, Stone MA, Appleton L, Moutsianas L, Moutsianis L, Leslie S, Wordsworth T, Kenna TJ, Karaderi T, Thomas GP, Ward MM, Weisman MH, Farrar C, Bradbury LA, Danoy P, Inman RD, Maksymowych W, Gladman D, Rahman P, Morgan A, Marzo-Ortega H, Bowness P, Gaffney K, Gaston JSH, Smith M, Bruges-Armas J, Couto AR, Sorrentino R, Paladini F, Ferreira MA, Xu H, Liu Y, Jiang L, Lopez-Larrea C, Díaz-Peña R, López-Vázquez A, Zayats T, Band G, Bellenguez C, Blackburn H, Blackwell JM, Bramon E, Bumpstead SJ, Casas JP, Corvin A, Craddock N, Deloukas P, Dronov S, Duncanson A, Edkins S, Freeman C, Gillman M, Gray E, Gwilliam R, Hammond N, Hunt SE, Jankowski J, Jayakumar A, Langford C, Liddle J, Markus HS, Mathew CG, McCann OT, McCarthy MI, Palmer CNA, Peltonen L, Plomin R, Potter SC, Rautanen A, Ravindrarajah R, Ricketts M, Samani N, Sawcer SJ, Strange A, Trembath RC, Viswanathan AC, Waller M, Weston P, Whittaker P, Widaa S, Wood NW, McVean G, Reveille JD, Wordsworth BP, Brown MA, Donnelly P. Interaction between ERAP1 and HLA-B27 in ankylosing spondylitis implicates peptide handling in the mechanism for HLA-B27 in disease susceptibility. Nat Genet 2011; 43:761-7. [PMID: 21743469 PMCID: PMC3640413 DOI: 10.1038/ng.873] [Citation(s) in RCA: 689] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 06/03/2011] [Indexed: 02/07/2023]
Abstract
Ankylosing spondylitis is a common form of inflammatory arthritis predominantly affecting the spine and pelvis that occurs in approximately 5 out of 1,000 adults of European descent. Here we report the identification of three variants in the RUNX3, LTBR-TNFRSF1A and IL12B regions convincingly associated with ankylosing spondylitis (P < 5 × 10(-8) in the combined discovery and replication datasets) and a further four loci at PTGER4, TBKBP1, ANTXR2 and CARD9 that show strong association across all our datasets (P < 5 × 10(-6) overall, with support in each of the three datasets studied). We also show that polymorphisms of ERAP1, which encodes an endoplasmic reticulum aminopeptidase involved in peptide trimming before HLA class I presentation, only affect ankylosing spondylitis risk in HLA-B27-positive individuals. These findings provide strong evidence that HLA-B27 operates in ankylosing spondylitis through a mechanism involving aberrant processing of antigenic peptides.
Collapse
Affiliation(s)
- David M Evans
- Medical Research Council (MRC) Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Lang R, Schoenen H, Desel C. Targeting Syk-Card9-activating C-type lectin receptors by vaccine adjuvants: findings, implications and open questions. Immunobiology 2011; 216:1184-91. [PMID: 21742403 DOI: 10.1016/j.imbio.2011.06.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2011] [Accepted: 06/13/2011] [Indexed: 12/19/2022]
Abstract
Pathogen recognition by the innate immune system is essential for the induction of adaptive T cell responses. A diverse range of pathogen-associated molecular patterns (PAMPs) are recognized by a variety of pathogen recognition receptors (PRRs). Among these are the well known Toll-like receptors (TLR) and the more recently described C-type lectin receptors (CLR) which utilize distinct signaling pathways leading to a diverse repertoire of effector molecules produced. The composition of the inflammatory juice released from activated innate immune cells has a major impact on the polarization of Th cell responses. Defined PAMPs may therefore be used as adjuvants to direct adaptive immune responses to subunit vaccines. Targeting CLR is an alternative or complementary strategy to TLR-triggering adjuvants that will benefit the development of more efficient subunit vaccines for prevention of major human infectious diseases. In this short review, we discuss the potential of CLRs activating APC via the Syk-Card9 pathway as receptors for adjuvants that direct the development of robust Th17 and Th1 responses to subunit vaccines.
Collapse
Affiliation(s)
- Roland Lang
- Institute of Clinical Microbiology, Immunology and Hygiene, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Wasserturmstr. 3-5, 91054 Erlangen, Germany.
| | | | | |
Collapse
|
37
|
Nakagawa T. Roles of prostaglandin E2 in the cochlea. Hear Res 2011; 276:27-33. [PMID: 21295127 DOI: 10.1016/j.heares.2011.01.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 01/15/2011] [Accepted: 01/19/2011] [Indexed: 12/20/2022]
Abstract
Prostaglandins are one of the major groups of chemical mediators in the mammalian body. Among prostaglandins, prostaglandin E2 (PGE2) is the most abundant prostanoid in humans and involved in regulating many different fundamental biological functions. PGE2 signaling is mediated by four distinct E-prostanoid receptors (EPs) namely EP1-4. Recently, accumulating evidence indicates critical, but complex roles of EP signaling in the pathogenesis of neuronal diseases depending on the context of neuronal injury. Four distinct EPs are expressed in the stria vascularis, spiral ligament, spiral ganglion and organ of Corti, indicating an involvement of EP signaling in the cochlear function. Activation of EP4 in cochleae significantly attenuates noise-induced damage in cochleae, and activation of EP2 or EP4 induces the formation of vascular endothelial growth factor in cochleae. These findings strongly suggest that individual EP signaling may be involved in the maintenance of the cochlear sensory system similarly to the central nervous system. This review highlights recent findings on EP signaling in the central nervous system, and presents its possible roles in regulation of blood flow, protection of sensory cells and immune responses in cochleae.
Collapse
Affiliation(s)
- Takayuki Nakagawa
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kawaharacho 54, Shogoin, Sakyoku, Kyoto 606-8507, Japan.
| |
Collapse
|