1
|
Tyring S, Moore A, Morita A, Hong HCH, Song IH, Eccleston J, Levy G, Mohamed MEF, Qian Y, Wu T, Pan A, Hew K, Papp KA. Cedirogant in adults with psoriasis: a phase II, randomized, placebo-controlled clinical trial. Clin Exp Dermatol 2024; 49:1347-1355. [PMID: 38699939 DOI: 10.1093/ced/llae152] [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: 12/21/2023] [Revised: 02/28/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND Dysregulated interleukin (IL)-17/IL-23 signalling contributes to psoriasis pathogenesis. Cedirogant is an inverse agonist of nuclear receptor ROR-gamma isoform 2 (RORyt), a key transcription factor responsible for IL-17 synthesis and a regulator of the T helper 17 cell lineage programme. OBJECTIVES To evaluate the efficacy and safety of cedirogant to treat moderate-to-severe psoriasis. METHODS In this phase IIb, multicentre, double-blind, 16-week study (NCT05044234), adults aged 18-65 years were randomized 1 : 1 : 1 : 1 to once-daily oral cedirogant 75 mg, 150 mg, 375 mg or placebo. Assessments included: ≥ 50%/75%/90%/100% improvement from baseline in Psoriasis Area and Severity Index (PASI 50/75/90/100), static Physician's Global Assessment 0/1, Psoriasis Symptoms Scale 0 and improvements in itch; adverse events (AEs); pharmacokinetics; and IL-17A/F biomarker levels. Efficacy results based on observed cases were summarized descriptively. RESULTS Of 156 enrolled patients, most were male (70.5%); 39 patients were randomized to each treatment. Only 47 patients completed the study; the study was terminated early owing to preclinical findings. At week 16, PASI 75 achievement rates (primary endpoint) were 29%, 8% and 42% in the cedirogant 75-mg, 150-mg and 375-mg groups, respectively, and 0% in the placebo group. AE rates were similar in the cedirogant 75-mg, 150-mg and placebo groups, and higher in the cedirogant 375-mg group; most AEs were mild or moderate. CONCLUSIONS Patients with psoriasis who received cedirogant showed PASI improvement, and cedirogant was generally well tolerated. The results should be interpreted in the context of early study termination. Cedirogant development has been discontinued.
Collapse
Affiliation(s)
- Stephen Tyring
- Center for Clinical Studies, Department of Dermatology, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA
| | - Angela Moore
- Arlington Research Center, Arlington, TX, USA
- Department of Dermatology, Baylor University Medical Center, Dallas, TX, USA
| | - Akimchi Morita
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - H Chih-Ho Hong
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC, Canada
- Probity Medical Research, Surrey, BC, Canada
| | | | | | | | | | | | | | - Anqi Pan
- AbbVie Inc., North Chicago, IL, USA
| | | | - Kim A Papp
- Alliance Clinical Research and Probity Medical Research, Waterloo, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
2
|
Zhao Q, Feng W, Gao P, Han Y, Zhang S, Zhou A, Shi L, Zhang J. Deoxynivalenol-Induced Spleen Toxicity in Mice: Inflammation, Endoplasmic Reticulum Stress, Macrophage Polarization, and the Dysregulation of LncRNA Expression. Toxins (Basel) 2024; 16:432. [PMID: 39453208 PMCID: PMC11511314 DOI: 10.3390/toxins16100432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/05/2024] [Accepted: 10/07/2024] [Indexed: 10/26/2024] Open
Abstract
The spleen is a primary target of deoxynivalenol (DON) toxicity, but its underlying molecular mechanisms remain unclear. This study investigates the effects of DON on inflammation, splenic macrophage polarization, endoplasmic reticulum (ER) stress, and transcriptome changes (mRNA and lncRNAs) in mouse spleen. We found that DON exposure at doses of 2.5 or 5 mg/kg BW significantly induced inflammation and polarized splenic macrophages towards the M1 phenotype. Additionally, DON activated PERK-eIF2α-ATF4-mediated ER stress and upregulated apoptosis-related proteins (caspase-12, caspase-3). The ER stress inhibitor, 4-Phenylbutyric acid, significantly alleviated DON-induced ER stress, apoptosis, and the M1 polarization of splenic macrophages. Transcriptome analysis identified 1968 differentially expressed (DE) lncRNAs and 2664 DE mRNAs in mouse spleen following DON exposure. Functional enrichment analysis indicated that the upregulated genes were involved in pathways associated with immunity, including Th17 cell differentiation, TNF signaling, and IL-17 signaling, while downregulated mRNAs were linked to cell survival and growth pathways. Furthermore, 370 DE lncRNAs were predicted to target 255 DE target genes associated with immune processes, including the innate immune response, interferon-beta response, cytokine production regulation, leukocyte apoptosis, and NF-κB signaling genes. This study provides new insights into the mechanisms underlying DON toxicity and its effects on the immune system.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jing Zhang
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming & Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (Q.Z.); (W.F.); (P.G.); (Y.H.); (S.Z.); (A.Z.); (L.S.)
| |
Collapse
|
3
|
Walther K, Gröger S, Vogler JAH, Wöstmann B, Meyle J. Inflammation indices in association with periodontitis and cancer. Periodontol 2000 2024; 96:281-315. [PMID: 39317462 PMCID: PMC11579835 DOI: 10.1111/prd.12612] [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: 05/16/2024] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/26/2024]
Abstract
Inflammation is a complex physiological process that plays a pivotal role in many if not all pathological conditions, including infectious as well as inflammatory diseases, like periodontitis and autoimmune disorders. Inflammatory response to periodontal biofilms and tissue destruction in periodontitis is associated with the release of inflammatory mediators. Chronic inflammation can promote the development of cancer. Persistence of inflammatory mediators plays a crucial role in this process. Quantification and monitoring of the severity of inflammation in relation to cancer is essential. Periodontitis is mainly quantified based on the severity and extent of attachment loss and/or pocket probing depth, in addition with bleeding on probing. In recent years, studies started to investigate inflammation indices in association with periodontal diseases. To date, only few reviews have been published focusing on the relationship between blood cell count, inflammation indices, and periodontitis. This review presents a comprehensive overview of different systemic inflammation indices, their methods of measurement, and the clinical applications in relation to periodontitis and cancer. This review outlines the physiological basis of inflammation and the underlying cellular and molecular mechanisms of the parameters described. Key inflammation indices are commonly utilized in periodontology such as the neutrophil to lymphocyte ratio. Inflammation indices like the platelet to lymphocyte ratio, platelet distribution width, plateletcrit, red blood cell distribution width, lymphocyte to monocyte ratio, delta neutrophil index, and the systemic immune inflammation index are also used in hospital settings and will be discussed. The clinical roles and limitations, relationship to systemic diseases as well as their association to periodontitis and treatment response are described.
Collapse
Affiliation(s)
- Kay‐Arne Walther
- Department of Periodontology, Dental ClinicJustus Liebig University of GiessenGiessenGermany
- Department of Prosthodontics, Dental ClinicJustus Liebig University of GiessenGiessenGermany
| | - Sabine Gröger
- Department of Periodontology, Dental ClinicJustus Liebig University of GiessenGiessenGermany
- Department of Orthodontics, Dental ClinicJustus Liebig University of GiessenGiessenGermany
| | | | - Bernd Wöstmann
- Department of Periodontology, Dental ClinicJustus Liebig University of GiessenGiessenGermany
- Department of Prosthodontics, Dental ClinicJustus Liebig University of GiessenGiessenGermany
| | - Jörg Meyle
- Department of Periodontology, Dental ClinicJustus Liebig University of GiessenGiessenGermany
- Department of Periodontology, Dental ClinicUniversity of BernBernSwitzerland
| |
Collapse
|
4
|
Pan P, Wang Y, Nyirenda MH, Saiyed Z, Karimian Azari E, Sunderman A, Milling S, Harnett MM, Pineda M. Undenatured type II collagen protects against collagen-induced arthritis by restoring gut-joint homeostasis and immunity. Commun Biol 2024; 7:804. [PMID: 38961129 PMCID: PMC11222443 DOI: 10.1038/s42003-024-06476-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 06/20/2024] [Indexed: 07/05/2024] Open
Abstract
Oral administration of harmless antigens can induce suppression of reactive immune responses, a process that capitalises on the ability of the gastrointestinal tract to tolerate exposure to food and commensal microbiome without triggering inflammatory responses. Repeating exposure to type II collagen induces oral tolerance and inhibits induction of arthritis, a chronic inflammatory joint condition. Although some mechanisms underlying oral tolerance are described, how dysregulation of gut immune networks impacts on inflammation of distant tissues like the joints is unclear. We used undenatured type II collagen in a prophylactic regime -7.33 mg/kg three times/week- to describe the mechanisms associated with protective oral immune-therapy (OIT) in gut and joint during experimental Collagen-Induced Arthritis (CIA). OIT reduced disease incidence to 50%, with reduced expression of IL-17 and IL-22 in the joints of asymptomatic mice. Moreover, whilst the gut tissue of arthritic mice shows substantial damage and activation of tissue-specific immune networks, oral administration of undenatured type II collagen protects against gut pathology in all mice, symptomatic and asymptomatic, rewiring IL-17/IL-22 networks. Furthermore, gut fucosylation and microbiome composition were also modulated. These results corroborate the relevance of the gut-joint axis in arthritis, showing novel regulatory mechanisms linked to therapeutic OIT in joint disease.
Collapse
Affiliation(s)
- Piaopiao Pan
- Centre for the Cellular Microenvironment, School of Molecular Biology, University of Glasgow, Glasgow, UK
| | - Yilin Wang
- Department of Bacteriology and Immunology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis & Thoracic Tumor Research Institute, Beijing, China
| | - Mukanthu H Nyirenda
- Institute of Infection and Immunity, University of Glasgow, Glasgow, UK
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Zainulabedin Saiyed
- Research and Development, Lonza Greenwood LLC, North Emerald Road, Greenwood, SC, USA
| | - Elnaz Karimian Azari
- Research and Development, Lonza Greenwood LLC, North Emerald Road, Greenwood, SC, USA
| | - Amy Sunderman
- Research and Development, Lonza Greenwood LLC, North Emerald Road, Greenwood, SC, USA
| | - Simon Milling
- Institute of Infection and Immunity, University of Glasgow, Glasgow, UK
| | | | - Miguel Pineda
- Centre for the Cellular Microenvironment, School of Molecular Biology, University of Glasgow, Glasgow, UK.
| |
Collapse
|
5
|
He G, Chen J, Hao W, Hu W. Causal effect of gut microbiota and diabetic nephropathy: a Mendelian randomization study. Diabetol Metab Syndr 2024; 16:89. [PMID: 38658966 PMCID: PMC11044463 DOI: 10.1186/s13098-024-01327-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 04/02/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND The interaction of dysbiosis of gut microbiota (GM) with diabetic nephropathy (DN) drew our attention and a better understanding of GM on DN might provide potential therapeutic approaches. However, the exact causal effect of GM on DN remains unknown. METHODS We applied two-sample Mendelian Randomization (MR) analysis, including inverse variance weighted (IVW), MR-Egger methods, etc., to screen the significant bacterial taxa based on the GWAS data. Sensitivity analysis was conducted to assess the robustness of MR results. To identify the most critical factor on DN, Mendelian randomization-Bayesian model averaging (MR-BMA) method was utilized. Then, whether the reverse causality existed was verified by reverse MR analysis. Finally, transcriptome MR analysis was performed to investigate the possible mechanism of GM on DN. RESULTS At locus-wide significance levels, the results of IVW suggested that order Bacteroidales (odds ratio (OR) = 1.412, 95% confidence interval (CI): 1.025-1.945, P = 0.035), genus Akkermansia (OR = 1.449, 95% CI: 1.120-1.875, P = 0.005), genus Coprococcus 1 (OR = 1.328, 95% CI: 1.066-1.793, P = 0.015), genus Marvinbryantia (OR = 1.353, 95% CI: 1.037-1.777, P = 0.030) and genus Parasutterella (OR = 1.276, 95% CI: 1.022-1.593, P = 0.032) were risk factors for DN. Reversely, genus Eubacterium ventriosum (OR = 0.756, 95% CI: 0.594-0.963, P = 0.023), genus Ruminococcus gauvreauii (OR = 0.663, 95% CI: 0.506-0.870, P = 0.003) and genus Erysipelotrichaceae (UCG003) (OR = 0.801, 95% CI: 0.644-0.997, P = 0.047) were negatively associated with the risk of DN. Among these taxa, genus Ruminococcus gauvreauii played a crucial role in DN. No significant heterogeneity or pleiotropy in the MR result was found. Mapped genes (FDR < 0.05) related to GM had causal effects on DN, while FCGR2B and VNN2 might be potential therapeutic targets. CONCLUSIONS This work provided new evidence for the causal effect of GM on DN occurrence and potential biomarkers for DN. The significant bacterial taxa in our study provided new insights for the 'gut-kidney' axis, as well as unconventional prevention and treatment strategies for DN.
Collapse
Affiliation(s)
- Ganyuan He
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangdong Provincial Geriatrics Institute, Southern Medical University, Guangzhou, China
| | - Jiayi Chen
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangdong Provincial Geriatrics Institute, Southern Medical University, Guangzhou, China
| | - Wenke Hao
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangdong Provincial Geriatrics Institute, Southern Medical University, Guangzhou, China.
| | - Wenxue Hu
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangdong Provincial Geriatrics Institute, Southern Medical University, Guangzhou, China.
| |
Collapse
|
6
|
Gao Y, Kennelly JP, Xiao X, Whang E, Ferrari A, Bedard AH, Mack JJ, Nguyen AH, Weston T, Uchiyama LF, Lee MS, Young SG, Bensinger SJ, Tontonoz P. T cell cholesterol transport is a metabolic checkpoint that links intestinal immune responses to dietary lipid absorption. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.584164. [PMID: 38559079 PMCID: PMC10979874 DOI: 10.1101/2024.03.08.584164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The intrinsic pathways that control membrane organization in immune cells and the impact of such pathways on cellular function are not well defined. Here we report that the non-vesicular cholesterol transporter Aster-A links plasma membrane (PM) cholesterol availability in T cells to immune signaling and systemic metabolism. Aster-A is recruited to the PM during T-cell receptor (TCR) activation, where it facilitates the removal of newly generated "accessible" membrane cholesterol. Loss of Aster-A leads to excess PM cholesterol accumulation, resulting in enhanced TCR nano-clustering and signaling, and Th17 cytokine production. Finally, we show that the mucosal Th17 response is restrained by PM cholesterol remodeling. Ablation of Aster-A in T cells leads to enhanced IL-22 production, reduced intestinal fatty acid absorption, and resistance to diet-induced obesity. These findings delineate a multi-tiered regulatory scheme linking immune cell lipid flux to nutrient absorption and systemic physiology.
Collapse
|
7
|
Wen Y, Wang H, Tian D, Wang G. TH17 cell: a double-edged sword in the development of inflammatory bowel disease. Therap Adv Gastroenterol 2024; 17:17562848241230896. [PMID: 38390028 PMCID: PMC10883129 DOI: 10.1177/17562848241230896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 01/17/2024] [Indexed: 02/24/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic nonspecific inflammatory disease of the gastrointestinal tract, and its pathogenesis has not been fully understood. Extensive dysregulation of the intestinal mucosal immune system is critical in the development and progression of IBD. T helper (Th) 17 cells have the characteristics of plasticity. They can transdifferentiate into subpopulations with different functions in response to different factors in the surrounding environment, thus taking on different roles in regulating the intestinal immune responses. In this review, we will focus on the plasticity of Th17 cells as well as the function of Th17 cells and their related cytokines in IBD. We will summarize their pathogenic and protective roles in IBD under different conditions, respectively, hoping to further deepen the understanding of the pathological mechanisms underlying IBD and provide insights for future treatment.
Collapse
Affiliation(s)
- Yue Wen
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dean Tian
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Ge Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| |
Collapse
|
8
|
Wei L, Chen Z, Lv Q. Mucosal-associated invariant T cells display both pathogenic and protective roles in patients with inflammatory bowel diseases. Amino Acids 2023; 55:1819-1827. [PMID: 37819474 DOI: 10.1007/s00726-023-03344-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 09/26/2023] [Indexed: 10/13/2023]
Abstract
An important subtype of the innate-like T lymphocytes is mucosal-associated invariant T (MAIT) cells expressing a semi-invariant T cell receptor α (TCR-α) chain. MAIT cells could be activated mainly by TCR engagement or cytokines. They have been found to have essential roles in various immune mediated. There have been growing preclinical and clinical findings that show an association between MAIT cells and the physiopathology of inflammatory bowel diseases (IBD). Of note, published reports demonstrate contradictory findings regarding the role of MAIT cells in IBD patients. A number of reports suggests a protective effect, whereas others show a pathogenic impact. The present review article aimed to explore and discuss the findings of experimental and clinical investigations evaluating the effects of MAIT cells in IBD subjects and animal models. Findings indicate that MAIT cells could exert opposite effects in the course of IBD, including an anti-inflammatory protective effect of blood circulating MAIT cells and an effector pathogenic effect of colonic MAIT cells. Another important finding is that blood levels of MAIT cells can be considered as a potential biomarker in IBD patients.
Collapse
Affiliation(s)
- Lei Wei
- Department of General Surgery, Pudong New District Gongli Hospital of Shanghai, Shanghai, 200120, China
| | - Zhigang Chen
- Department of General Surgery, Pudong New District Gongli Hospital of Shanghai, Shanghai, 200120, China
| | - Qiang Lv
- Department of General Surgery, Pudong New District Gongli Hospital of Shanghai, Shanghai, 200120, China.
| |
Collapse
|
9
|
Bao W, Wang L, Liu X, Li M. Predicting diagnostic biomarkers associated with immune infiltration in Crohn's disease based on machine learning and bioinformatics. Eur J Med Res 2023; 28:255. [PMID: 37496049 PMCID: PMC10369716 DOI: 10.1186/s40001-023-01200-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/26/2023] [Indexed: 07/28/2023] Open
Abstract
OBJECTIVE The objective of this study is to investigate potential biomarkers of Crohn's disease (CD) and the pathological importance of infiltration of associated immune cells in disease development using machine learning. METHODS Three publicly accessible CD gene expression profiles were obtained from the GEO database. Inflammatory tissue samples were selected and differentiated between colonic and ileal tissues. To determine the differentially expressed genes (DEGs) between CD and healthy controls, the larger sample size was merged as a training unit. The function of DEGs was comprehended through disease enrichment (DO) and gene set enrichment analysis (GSEA) on DEGs. Promising biomarkers were identified using the support vector machine-recursive feature elimination and lasso regression models. To further clarify the efficacy of potential biomarkers as diagnostic genes, the area under the ROC curve was observed in the validation group. Additionally, using the CIBERSORT approach, immune cell fractions from CD patients were examined and linked with potential biomarkers. RESULTS Thirty-four DEGs were identified in colon tissue, of which 26 were up-regulated and 8 were down-regulated. In ileal tissues, 50 up-regulated and 50 down-regulated DEGs were observed. Disease enrichment of colon and ileal DEGs primarily focused on immunity, inflammatory bowel disease, and related pathways. CXCL1, S100A8, REG3A, and DEFA6 in colon tissue and LCN2 and NAT8 in ileum tissue demonstrated excellent diagnostic value and could be employed as CD gene biomarkers using machine learning methods in conjunction with external dataset validation. In comparison to controls, antigen processing and presentation, chemokine signaling pathway, cytokine-cytokine receptor interactions, and natural killer cell-mediated cytotoxicity were activated in colonic tissues. Cytokine-cytokine receptor interactions, NOD-like receptor signaling pathways, and toll-like receptor signaling pathways were activated in ileal tissues. NAT8 was found to be associated with CD8 T cells, while CXCL1, S100A8, REG3A, LCN2, and DEFA6 were associated with neutrophils, indicating that immune cell infiltration in CD is closely connected. CONCLUSION CXCL1, S100A8, REG3A, and DEFA6 in colonic tissue and LCN2 and NAT8 in ileal tissue can be employed as CD biomarkers. Additionally, immune cell infiltration is crucial for CD development.
Collapse
Affiliation(s)
- Wenhui Bao
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Spleen and Gastroenterology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, No.354 Beima Road, Hongqiao District, Tianjin, China
| | - Lin Wang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Nephrology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoxiao Liu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Department of Comprehensive Rehabilitation, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ming Li
- Spleen and Gastroenterology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, No.354 Beima Road, Hongqiao District, Tianjin, China.
| |
Collapse
|
10
|
Douglas A, Stevens B, Lynch L. Interleukin-17 as a key player in neuroimmunometabolism. Nat Metab 2023; 5:1088-1100. [PMID: 37488456 PMCID: PMC10440016 DOI: 10.1038/s42255-023-00846-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/14/2023] [Indexed: 07/26/2023]
Abstract
In mammals, interleukin (IL)-17 cytokines are produced by innate and adaptive lymphocytes. However, the IL-17 family has widespread expression throughout evolution, dating as far back as cnidaria, molluscs and worms, which predate lymphocytes. The evolutionary conservation of IL-17 suggests that it is involved in innate defence strategies, but also that this cytokine family has a fundamental role beyond typical host defence. Throughout evolution, IL-17 seems to have a major function in homeostatic maintenance at barrier sites. Most recently, a pivotal role has been identified for IL-17 in regulating cellular metabolism, neuroimmunology and tissue physiology, particularly in adipose tissue. Here we review the emerging role of IL-17 signalling in regulating metabolic processes, which may shine a light on the evolutionary role of IL-17 beyond typical immune responses. We propose that IL-17 helps to coordinate the cross-talk among the nervous, endocrine and immune systems for whole-body energy homeostasis as a key player in neuroimmunometabolism.
Collapse
Affiliation(s)
- Aaron Douglas
- School of Biochemistry and Immunology, TBSI, Trinity College Dublin, Dublin, Ireland
| | - Brenneth Stevens
- School of Biochemistry and Immunology, TBSI, Trinity College Dublin, Dublin, Ireland
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lydia Lynch
- School of Biochemistry and Immunology, TBSI, Trinity College Dublin, Dublin, Ireland.
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Abstract
Mucosal associated invariant T (MAIT) cells are innate-like T lymphocytes, strikingly enriched at mucosal surfaces and characterized by a semi-invariant αβ T cell receptor (TCR) recognizing microbial derived intermediates of riboflavin synthesis presented by the MHC-Ib molecule MR1. At barrier sites MAIT cells occupy a prime position for interaction with commensal microorganisms, comprising the microbiota. The microbiota is a rich source of riboflavin derived antigens required in early life to promote intra-thymic MAIT cell development and sustain a life-long population of tissue resident cells. A symbiotic relationship is thought to be maintained in health whereby microbes promote maturation and homeostasis, and in turn MAIT cells can engage a TCR-dependent "tissue repair" program in the presence of commensal organisms conducive to sustaining barrier function and integrity of the microbial community. MAIT cell activation can be induced in a MR1-TCR dependent manner or through MR1-TCR independent mechanisms via pro-inflammatory cytokines interleukin (IL)-12/-15/-18 and type I interferon. MAIT cells provide immunity against bacterial, fungal and viral pathogens. However, MAIT cells may have deleterious effects through insufficient or exacerbated effector activity and have been implicated in autoimmune, inflammatory and allergic conditions in which microbial dysbiosis is a shared feature. In this review we summarize the current knowledge on the role of the microbiota in the development and maintenance of circulating and tissue resident MAIT cells. We also explore how microbial dysbiosis, alongside changes in intestinal permeability and imbalance between pro- and anti-inflammatory components of the immune response are together involved in the potential pathogenicity of MAIT cells. Whilst there have been significant improvements in our understanding of how the microbiota shapes MAIT cell function, human data are relatively lacking, and it remains unknown if MAIT cells can conversely influence the composition of the microbiota. We speculate whether, in a human population, differences in microbiomes might account for the heterogeneity observed in MAIT cell frequency across mucosal sites or between individuals, and response to therapies targeting T cells. Moreover, we speculate whether manipulation of the microbiota, or harnessing MAIT cell ligands within the gut or disease-specific sites could offer novel therapeutic strategies.
Collapse
Affiliation(s)
- Maisha F. Jabeen
- Respiratory Medicine Unit, Experimental Medicine Division, Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, United Kingdom
| | - Timothy S. C. Hinks
- Respiratory Medicine Unit, Experimental Medicine Division, Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
12
|
Ouyang Y, Zhao J, Wang S. Multifunctional hydrogels based on chitosan, hyaluronic acid and other biological macromolecules for the treatment of inflammatory bowel disease: A review. Int J Biol Macromol 2023; 227:505-523. [PMID: 36495992 DOI: 10.1016/j.ijbiomac.2022.12.032] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/28/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Hydrogel is a three-dimensional network polymer material rich in water. It is widely used in the biomedical field because of its unique physical and chemical properties and good biocompatibility. In recent years, the incidence of inflammatory bowel disease (IBD) has gradually increased, and the disadvantages caused by traditional drug treatment of IBD have emerged. Therefore, there is an urgent need for new treatments to alleviate IBD. Hydrogel has become a potential therapeutic platform. However, there is a lack of comprehensive review of functional hydrogels for IBD treatment. This paper first summarizes the pathological changes in IBD sites. Then, the action mechanisms of hydrogels prepared from chitosan, sodium alginate, hyaluronic acid, functionalized polyethylene glycol, cellulose, pectin, and γ-polyglutamic acid on IBD were described from aspects of drug delivery, peptide and protein delivery, biologic therapies, loading probiotics, etc. In addition, the advanced functions of IBD treatment hydrogels were summarized, with emphasis on adhesion, synergistic therapy, pH sensitivity, particle size, and temperature sensitivity. Finally, the future development direction of IBD treatment hydrogels has been prospected.
Collapse
Affiliation(s)
- Yongliang Ouyang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093,China
| | - Jiulong Zhao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, China
| | - Shige Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093,China.
| |
Collapse
|
13
|
Immunological Aspects of Von Hippel-Lindau Disease: A Focus on Neuro-Oncology and Myasthenia Gravis. Diagnostics (Basel) 2023; 13:diagnostics13010144. [PMID: 36611440 PMCID: PMC9818211 DOI: 10.3390/diagnostics13010144] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
Von Hippel-Lindau (VHL) disease is an autosomal dominant condition that predisposes affected individuals to a variety of malignant and benign neoplasms. The pathogenetic turning point of this illness is the accumulation of hypoxia-inducible factor (HIF)-1α, a transcription factor of several genes involved in oncogenesis, angiogenesis, tissue regeneration, metabolic regulation, hematopoiesis, and inflammatory responses. From an oncological perspective, increased awareness of the molecular pathways underlying this disease is bringing us closer to the development of specific and targeted therapies. Meanwhile, on the surgical side, improved understanding can help to better identify the patients to be treated and the surgical timing. Overall, pathogenesis research is crucial for developing patient-tailored therapies. One of the actual key topics of interest is the link between the VHL/HIF axis and inflammation. The present study aims to outline the fundamental mechanisms that link VHL disease and immune disorders, as well as to explore the details of the overlap between VHL disease and myasthenia gravis (MG) pathogenetic pathways. As a result, MG becomes a paradigm for autoimmune disorders that might be related with VHL disease.
Collapse
|
14
|
Li C, Yu R, Ding Y. Association between Porphyromonas Gingivalis and systemic diseases: Focus on T cells-mediated adaptive immunity. Front Cell Infect Microbiol 2022; 12:1026457. [PMID: 36467726 PMCID: PMC9712990 DOI: 10.3389/fcimb.2022.1026457] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/25/2022] [Indexed: 12/01/2023] Open
Abstract
The association between periodontal disease and systemic disease has become a research hotspot. Porphyromonas gingivalis (P. gingivalis), a crucial periodontal pathogen, affects the development of systemic diseases. The pathogenicity of P. gingivalis is largely linked to interference with the host's immunity. This review aims to discover the role of P. gingivalis in the modulation of the host's adaptive immune system through a large number of virulence factors and the manipulation of cellular immunological responses (mainly mediated by T cells). These factors may affect the cause of large numbers of systemic diseases, such as atherosclerosis, hypertension, adverse pregnancy outcomes, inflammatory bowel disease, diabetes mellitus, non-alcoholic fatty liver disease, rheumatoid arthritis, and Alzheimer's disease. The point of view of adaptive immunity may provide a new idea for treating periodontitis and related systemic diseases.
Collapse
Affiliation(s)
- Cheng Li
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Ran Yu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Yumei Ding
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| |
Collapse
|
15
|
Nour-Eldine W, Ltaief SM, Abdul Manaph NP, Al-Shammari AR. In search of immune cellular sources of abnormal cytokines in the blood in autism spectrum disorder: A systematic review of case-control studies. Front Immunol 2022; 13:950275. [PMID: 36268027 PMCID: PMC9578337 DOI: 10.3389/fimmu.2022.950275] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 09/21/2022] [Indexed: 12/04/2022] Open
Abstract
Abnormal cytokine levels in circulating blood have been repeatedly reported in autism; however, the underlying cause remains unclear. This systematic review aimed to investigate cytokine levels in peripheral blood compartments and identify their potential immune cellular sources in subjects with autism through comparison with controls. We conducted an electronic database search (PubMed, Scopus, ProQuest Central, Ovid, SAGE Journals, and Wiley Online Library) from inception (no time limits) to July 9, 2020, and identified 75 relevant articles. Our qualitative data synthesis focused on results consistently described in at least three independent studies, and we reported the results according to the PRISMA protocol. We found that compared with controls, in subjects with autism, cytokines IL-6, IL-17, TNF-α, and IL-1β increased in the plasma and serum. We also identified monocytes, neutrophils, and CD4+ T cells as potential sources of these elevated cytokines in autism. Cytokines IFN-γ, TGF-β, RANTES, and IL-8 were increased in the plasma/serum of subjects with autism, and IFN-γ was likely produced by CD4+ T cells and natural killer (NK) cells, although conflicting evidence is present for IFN-γ and TGF-β. Other cytokines-IL-13, IL-10, IL-5, and IL-4-were found to be unaltered in the plasma/serum and post-stimulated blood immune cells in autistic individuals as compared with controls. The frequencies of T cells, monocytes, B cells, and NK cells were unchanged in subjects with autism as opposed to controls, suggesting that abnormal cytokines were unlikely due to altered cell numbers but might be due to altered functioning of these cells in autism. Our results support existing studies of abnormal cytokines in autism and provide comprehensive evidence of potential cellular sources of these altered cytokines in the context of autism. Systematic Review Registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42020205224, identifier [CRD42020205224].
Collapse
Affiliation(s)
| | | | | | - Abeer R. Al-Shammari
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| |
Collapse
|
16
|
Bai R, Li Z, Lv S, Hua W, Dai L, Wu H. Exploring the biological function of immune cell-related genes in human immunodeficiency virus (HIV)-1 infection based on weighted gene co-expression network analysis (WGCNA). BMC Med Genomics 2022; 15:200. [PMID: 36123690 PMCID: PMC9484082 DOI: 10.1186/s12920-022-01357-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/15/2022] [Indexed: 12/04/2022] Open
Abstract
Background Acquired immunodeficiency syndrome (AIDS) is a chronic infectious disease characterized by consistent immune dysfunction. The objective of this study is to determine whether immune cell-related genes can be used as biomarkers for the occurrence of AIDS and potential molecular mechanisms. Methods A weighted gene co-expression network analysis was performed using the GSE6740 dataset from the Gene Expression Synthesis Database to identify the Hub gene, which contained microarray data from HIV-1 positive (HIV-1+) and HIV-1 negative (HIV-1−) individuals. The HIV-1+-related differentially expressed genes were then identified using the limma package. Subsequently, the characteristic immune cell-related genes were identified as diagnostic biomarkers for HIV-1+ using the random forest model (RF), support vector machine model, and generalized linear model. Results MEdarkgreen exhibited the strongest correlation with HIV clinical features of any of these modules. As the best model for diagnosing HIV-1±, RF was used to select four critical immune cell-related genes, namely, ARRB1, DPEP2, LTBP3, and RGCC, and a nomogram model was created to predict the occurrence of HIV-1 infection based on four key immune cell-related genes. Diagnostic genes were shown to be engaged in immune-related pathways, suggesting that immunological molecules, immune cells, and immune pathways all have a role in HIV-1 infection. The CTD database was explored for prospective medications or molecular compounds that might be utilized to treat HIV-1+ patients. = Moreover, in HIV-1+ individuals, the ceRNA network revealed that ARRB1, DPEP2, LTBP3, and RGCC could be regulated by lncRNAs through the corresponding miRNAs. Ultimately, RT-PCR results from clinical blood samples demonstrated that the four diagnostic genes were significantly downregulated in HIV-1+ patients. Conclusion We screened four immune cell-related genes, ARRB1, DPEP2, LTBP3, and RGCC, which may be considered as the diagnostic markers for HIV-1/AIDS. Our findings reveal that immune related genes and pathways involved in HIV-1 pathogenesis were regulated on both genetic and epigenetic levels by constructing a ceRNA network associated with lncRNA. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01357-y.
Collapse
Affiliation(s)
- Ruojing Bai
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research On Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Zhen Li
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research On Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Shiyun Lv
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research On Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Wei Hua
- Travel Clinic, Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Lili Dai
- Travel Clinic, Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
| | - Hao Wu
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research On Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
17
|
Schiweck C, Edwin Thanarajah S, Aichholzer M, Matura S, Reif A, Vrieze E, Weigert A, Visekruna A. Regulation of CD4 + and CD8 + T Cell Biology by Short-Chain Fatty Acids and Its Relevance for Autoimmune Pathology. Int J Mol Sci 2022; 23:8272. [PMID: 35955407 PMCID: PMC9368239 DOI: 10.3390/ijms23158272] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 07/07/2022] [Indexed: 02/01/2023] Open
Abstract
The gut microbiota encodes a broad range of enzymes capable of synthetizing various metabolites, some of which are still uncharacterized. One well-known class of microbiota-derived metabolites are the short-chain fatty acids (SCFAs) such as acetate, propionate, butyrate and valerate. SCFAs have long been considered a mere waste product of bacterial metabolism. Novel results have challenged this long-held dogma, revealing a central role for microbe-derived SCFAs in gut microbiota-host interaction. SCFAs are bacterial signaling molecules that act directly on host T lymphocytes by reprogramming their metabolic activity and epigenetic status. They have an essential biological role in promoting differentiation of (intestinal) regulatory T cells and in production of the anti-inflammatory cytokine interleukin-10 (IL-10). These small molecules can also reach the circulation and modulate immune cell function in remote tissues. In experimental models of autoimmune and inflammatory diseases, such as inflammatory bowel disease, multiple sclerosis or diabetes, a strong therapeutic potential of SCFAs through the modulation of effector T cell function was observed. In this review, we discuss current research activities toward understanding a relevance of microbial SCFA for treating autoimmune and inflammatory pathologies from in vitro to human studies.
Collapse
Affiliation(s)
- Carmen Schiweck
- Department for Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt-Goethe University, 60528 Frankfurt, Germany; (S.E.T.); (M.A.); (S.M.); (A.R.)
| | - Sharmili Edwin Thanarajah
- Department for Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt-Goethe University, 60528 Frankfurt, Germany; (S.E.T.); (M.A.); (S.M.); (A.R.)
| | - Mareike Aichholzer
- Department for Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt-Goethe University, 60528 Frankfurt, Germany; (S.E.T.); (M.A.); (S.M.); (A.R.)
| | - Silke Matura
- Department for Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt-Goethe University, 60528 Frankfurt, Germany; (S.E.T.); (M.A.); (S.M.); (A.R.)
| | - Andreas Reif
- Department for Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt-Goethe University, 60528 Frankfurt, Germany; (S.E.T.); (M.A.); (S.M.); (A.R.)
| | - Elske Vrieze
- Department of Psychiatry and Neurosciences, UPC KU Leuven, Campus Gasthuisberg, KU Leuven, 3000 Leuven, Belgium;
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt/Main, 60590 Frankfurt, Germany;
| | - Alexander Visekruna
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, 35043 Marburg, Germany;
| |
Collapse
|
18
|
Lee AY, Foulsham W. Regulatory T Cells: Therapeutic Opportunities in Uveitis. FRONTIERS IN OPHTHALMOLOGY 2022; 2:901144. [PMID: 38983511 PMCID: PMC11182269 DOI: 10.3389/fopht.2022.901144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/28/2022] [Indexed: 07/11/2024]
Abstract
Regulatory T cells (Tregs) are critical for the maintenance of immune tolerance and the suppression of excessive inflammation. Many inflammatory autoimmune disorders, including autoimmune uveitis, involve the loss of the suppressive capacities of Tregs. Over the past decade, Tregs' therapeutic potential in uveitis has garnered increasing attention. Specific subsets of Tregs, including TIGIT+ and PD-1+ Tregs, have emerged as potent immunosuppressors that may be particularly well-suited to cell-based therapeutics. Studies have elucidated the interaction between Treg development and the gut microbiome as well as various intracellular signaling pathways. Numerous cell-based therapies and therapeutic molecules have been proposed and investigated using the murine experimental autoimmune uveitis (EAU) model. However, certain challenges remain to be addressed. Studies involving the use of Tregs in human patients with uveitis are lacking, and there are concerns regarding Tregs' production and purification for practical use, their plasticity towards inflammatory phenotypes, immunogenicity, and tumorigenicity. Nevertheless, recent research has brought Tregs closer to yielding viable treatment options for uveitis.
Collapse
Affiliation(s)
| | - William Foulsham
- Department of Ophthalmology, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
19
|
Okazaki S, Hoashi T, Saeki H, Kanda N. A Case of Autoimmune Hepatitis/Primary Biliary Cholangitis Overlap Syndrome during Treatment with Brodalumab for Generalized Pustular Psoriasis. J NIPPON MED SCH 2022; 88:569-573. [PMID: 34980743 DOI: 10.1272/jnms.jnms.2021_88-517] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by accelerated tumor necrosis factor-α (TNF-α) /interleukin (IL) -23/IL-17 axis, epidermal hyperproliferation, and dysregulated differentiation. Psoriasis is occasionally associated with autoimmune liver diseases such as autoimmune hepatitis (AIH) or primary biliary cholangitis (PBC), caused by autoimmunity against hepatocyte- or cholangiocyte-specific autoantigens, respectively. Overlap syndrome is a condition in which patients have features of both AIH and PBC. It has been reported that AIH, PBC, or the overlap syndrome can be triggered by certain drug therapies. A 65-year-old Japanese man developed increased serum levels of aspartate and alanine aminotransferases, and positive anti-nuclear and anti-mitochondrial M2 antibodies, along with neutropenia, at 4 weeks of treatment with an anti-IL-17 receptor A antibody brodalumab for generalized pustular psoriasis. Histological evaluation of the liver revealed interface hepatitis and non-suppurative destructive cholangitis, which is compatible with the overlap syndrome of AIH and PBC. This is the first case of AIH/PBC overlap syndrome during treatment with brodalumab for generalized pustular psoriasis. The relationship between brodalumab and AIH/PBC overlap syndrome should be further elucidated. The risk of autoimmune liver diseases in patients with psoriasis treated with brodalumab should be carefully considered.
Collapse
Affiliation(s)
- Shizuka Okazaki
- Department of Dermatology, Nippon Medical School Chiba Hokusoh Hospital
| | | | - Hidehisa Saeki
- Department of Dermatology, Nippon Medical School Hospital
| | - Naoko Kanda
- Department of Dermatology, Nippon Medical School Chiba Hokusoh Hospital
| |
Collapse
|
20
|
Shi L, Kidder K, Bian Z, Chiang SKT, Ouellette C, Liu Y. SIRPα sequesters SHP-2 to promote IL-4 and IL-13 signaling and the alternative activation of macrophages. Sci Signal 2021; 14:eabb3966. [PMID: 34582250 DOI: 10.1126/scisignal.abb3966] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Lei Shi
- Program of Immunology and Molecular Cellular Biology, Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | - Koby Kidder
- Program of Immunology and Molecular Cellular Biology, Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | - Zhen Bian
- Program of Immunology and Molecular Cellular Biology, Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | - Samantha Kuon Ting Chiang
- Program of Immunology and Molecular Cellular Biology, Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | - Corbett Ouellette
- Program of Immunology and Molecular Cellular Biology, Department of Biology, Georgia State University, Atlanta, GA 30302, USA.,Center of Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30302, USA
| | - Yuan Liu
- Program of Immunology and Molecular Cellular Biology, Department of Biology, Georgia State University, Atlanta, GA 30302, USA.,Center of Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30302, USA
| |
Collapse
|
21
|
Ait-Oufella H, Lavillegrand JR, Tedgui A. Regulatory T Cell-Enhancing Therapies to Treat Atherosclerosis. Cells 2021; 10:cells10040723. [PMID: 33805071 PMCID: PMC8064079 DOI: 10.3390/cells10040723] [Citation(s) in RCA: 9] [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: 02/11/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
Experimental studies have provided strong evidence that chronic inflammation triggered by the sub-endothelial accumulation of cholesterol-rich lipoproteins in arteries is essential in the initiation and progression of atherosclerosis. Recent clinical trials highlighting the efficacy of anti-inflammatory therapies in coronary patients have confirmed that this is also true in humans Monocytes/macrophages are central cells in the atherosclerotic process, but adaptive immunity, through B and T lymphocytes, as well as dendritic cells, also modulates the progression of the disease. Analysis of the role of different T cell subpopulations in murine models of atherosclerosis identified effector Th1 cells as proatherogenic, whereas regulatory T cells (Tregs) have been shown to protect against atherosclerosis. For these reasons, better understanding of how Tregs influence the atherosclerotic process is believed to provide novel Treg-targeted therapies to combat atherosclerosis. This review article summarizes current knowledge about the role of Tregs in atherosclerosis and discusses ways to enhance their function as novel immunomodulatory therapeutic approaches against cardiovascular disease.
Collapse
Affiliation(s)
- Hafid Ait-Oufella
- Paris Cardiovascular Research Center—PARCC, Université de Paris, INSERM UMR-S 970, 75012 Paris, France; (J.-R.L.); (A.T.)
- AP-HP (Assistance Publique-Hôpitaux de Paris), Hôpital Saint-Antoine, Sorbonne Université, 75012 Paris, France
- Correspondence: ; Tel.: +33-1-5398-8006; Fax: +33-1-5398-8052
| | - Jean-Rémi Lavillegrand
- Paris Cardiovascular Research Center—PARCC, Université de Paris, INSERM UMR-S 970, 75012 Paris, France; (J.-R.L.); (A.T.)
| | - Alain Tedgui
- Paris Cardiovascular Research Center—PARCC, Université de Paris, INSERM UMR-S 970, 75012 Paris, France; (J.-R.L.); (A.T.)
| |
Collapse
|
22
|
Gonçalves VS, Santos FDS, Dos Santos Junior AG, Piraine REA, Rodrigues PRC, Brasil CL, Conrad NL, Leite FPL. Recombinant bovine IL17A acts as an adjuvant for bovine herpesvirus vaccine. Res Vet Sci 2021; 136:185-191. [PMID: 33677208 DOI: 10.1016/j.rvsc.2021.02.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/23/2021] [Accepted: 02/14/2021] [Indexed: 12/24/2022]
Abstract
The Bovine herpes virus type 5 glycoprotein D (gD) is essential for viral penetration into host permissive cells. The Herpes virus gD glycoprotein has been used for bovine immunization, being efficient in reduction of viral replication, shedding and clinical signs, however sterilizing immunity is still not achieved. Recombinant subunit vaccines are, in general, poorly immunogenic requiring additional adjuvant components. Interleukin 17A (IL17A) is a pro-inflammatory cytokine produced by T helper 17 cells that mediate mucosal immunity. IL17 production during vaccine-induced immunity is a requirement for mucosal protection to several agents. In this study, we investigated the potential of a recombinant IL17A to act as an adjuvant for a recombinant BoHV-5 glycoprotein D vaccine in cattle. Three cattle groups were divided as: group 1) rgD5 + alumen + rIL-17A; 2) rgD5 + alumen; and 3) PBS + alumen. The cattle (3 per group) received two doses of their respective vaccines at an interval of 21 days. The group that received rIL17 in its vaccine formulation at the 7th day after the prime immunization had significant higher levels of specific rgD-IgG than the alumen group. Addition of rIL17 also led to a significant fold increase in specific anti-rgD IgG and neutralizing antibodies to the virus, respectively, when compared with the alumen group. Cells stimulated with rIL17A responded with IL17 transcription, as well IL2, IL4, IL10, IL15, Bcl6 and CXCR5. Our findings suggest that the rIL17A has adjuvant potential for use in vaccines against BoHV-5 as well as potentially other pathogens of cattle.
Collapse
Affiliation(s)
- Vitória Sequeira Gonçalves
- Núcleo de Biotecnologia - Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas (UFPel), Pelotas, RS, Brazil
| | - Francisco Denis Souza Santos
- Núcleo de Biotecnologia - Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas (UFPel), Pelotas, RS, Brazil
| | | | - Renan Eugênio Araujo Piraine
- Núcleo de Biotecnologia - Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas (UFPel), Pelotas, RS, Brazil
| | | | - Carolina Litchina Brasil
- Departamento de Microbiologia e Parasitologia, Universidade Federal de Pelotas (UFPel), Pelotas, RS, Brazil
| | - Neida Lucia Conrad
- Núcleo de Biotecnologia - Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas (UFPel), Pelotas, RS, Brazil
| | - Fábio Pereira Leivas Leite
- Núcleo de Biotecnologia - Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas (UFPel), Pelotas, RS, Brazil.
| |
Collapse
|
23
|
Rosolowski M, Oberle V, Ahnert P, Creutz P, Witzenrath M, Kiehntopf M, Loeffler M, Suttorp N, Scholz M. Dynamics of cytokines, immune cell counts and disease severity in patients with community-acquired pneumonia - Unravelling potential causal relationships. Cytokine 2020; 136:155263. [PMID: 32896803 DOI: 10.1016/j.cyto.2020.155263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Community acquired pneumonia (CAP) is a severe and often rapidly deteriorating disease. To better understand its dynamics and potential causal relationships, we analyzed time series data of cytokines, blood and clinical parameters in hospitalized CAP patients. METHODS Time series data of 10 circulating cytokines, blood counts and clinical parameters were related to baseline characteristics of 403 CAP patients using univariate mixed models. Bivariate mixed models were applied to analyze correlations between the time series. To identify potential causal relationships, we inferred cross-lagged relationships between pairs of parameters using latent curve models with structured residuals. RESULTS IL-6 levels decreased faster over time in younger patients (Padj = 0.06). IL-8, VCAM-1, and IL-6 correlated strongly with disease severity as assessed by the sequential organ failure assessment (SOFA) score (r = 0.49, 0.48, 0.46, respectively; all Padj < 0.001). IL-6 and bilirubin correlated with respect to their mean levels and slopes over time (r = 0.36 and r = 0.46, respectively; Padj < 0.001). A number of potential causal relationships were identified, e.g., a negative effect of ICAM-1 on MCP-1, or a positive effect of the level of creatinine on the subsequent VCAM-1 concentration (P < 0.001). CONCLUSIONS These results suggest that IL-6 trajectories of CAP patients are associated with age and run parallel to bilirubin levels. The time series analysis also unraveled directed, potentially causal relationships between cytokines, blood parameters and clinical outcomes. This will facilitate the development of mechanistic models of CAP, and with it, improvements in treatment or surveillance strategies for this disease. TRIAL REGISTRATION clinicaltrials.gov NCT02782013, May 25, 2016, retrospectively registered.
Collapse
Affiliation(s)
- Maciej Rosolowski
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, Leipzig, Germany.
| | - Volker Oberle
- Department of Clinical Chemistry and Laboratory Medicine, Jena University Hospital, Jena, Germany
| | - Peter Ahnert
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, Leipzig, Germany
| | - Petra Creutz
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Martin Witzenrath
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Michael Kiehntopf
- Integrated Biobank Jena (IBBJ) and Institute of Clinical Chemistry and Laboratory Diagnostics, Jena University Hospital, Jena, Germany
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, Leipzig, Germany
| | - Norbert Suttorp
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, Leipzig, Germany
| |
Collapse
|
24
|
Bhattacharya P, Ellegård R, Khalid M, Svanberg C, Govender M, Keita ÅV, Söderholm JD, Myrelid P, Shankar EM, Nyström S, Larsson M. Complement opsonization of HIV affects primary infection of human colorectal mucosa and subsequent activation of T cells. eLife 2020; 9:e57869. [PMID: 32876566 PMCID: PMC7492089 DOI: 10.7554/elife.57869] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023] Open
Abstract
HIV transmission via genital and colorectal mucosa are the most common routes of dissemination. Here, we explored the effects of free and complement-opsonized HIV on colorectal tissue. Initially, there was higher antiviral responses in the free HIV compared to complement-opsonized virus. The mucosal transcriptional response at 24 hr revealed the involvement of activated T cells, which was mirrored in cellular responses observed at 96 hr in isolated mucosal T cells. Further, HIV exposure led to skewing of T cell phenotypes predominantly to inflammatory CD4+ T cells, that is Th17 and Th1Th17 subsets. Of note, HIV exposure created an environment that altered the CD8+ T cell phenotype, for example expression of regulatory factors, especially when the virions were opsonized with complement factors. Our findings suggest that HIV-opsonization alters the activation and signaling pathways in the colorectal mucosa, which promotes viral establishment by creating an environment that stimulates mucosal T cell activation and inflammatory Th cells.
Collapse
Affiliation(s)
- Pradyot Bhattacharya
- Division of Molecular Medicine and Virology, Department of Clinical and Experimental Medicine, Linköping UniversityLinköpingSweden
| | - Rada Ellegård
- Division of Molecular Medicine and Virology, Department of Clinical and Experimental Medicine, Linköping UniversityLinköpingSweden
| | - Mohammad Khalid
- Division of Molecular Medicine and Virology, Department of Clinical and Experimental Medicine, Linköping UniversityLinköpingSweden
| | - Cecilia Svanberg
- Division of Molecular Medicine and Virology, Department of Clinical and Experimental Medicine, Linköping UniversityLinköpingSweden
| | - Melissa Govender
- Division of Molecular Medicine and Virology, Department of Clinical and Experimental Medicine, Linköping UniversityLinköpingSweden
| | - Åsa V Keita
- Division of Surgery, Orthopedics and Oncology, Linköping UniversityLinköpingSweden
| | - Johan D Söderholm
- Division of Surgery, Orthopedics and Oncology, Linköping UniversityLinköpingSweden
| | - Pär Myrelid
- Division of Surgery, Orthopedics and Oncology, Linköping UniversityLinköpingSweden
| | - Esaki M Shankar
- Center of Excellence for Research in AIDS (CERiA), University of Malaya, Lembah PantaiKuala LumpurMalaysia
- Division of Infection Biology and Medical Microbiology, Department of Life Sciences, Central University of Tamil NaduThiruvarurIndia
| | - Sofia Nyström
- Division of Molecular Medicine and Virology, Department of Clinical and Experimental Medicine, Linköping UniversityLinköpingSweden
- Department of Clinical Immunology and Transfusion Medicine and Department of Clinical and Experimental Medicine, Linköping UniversityLinköpingSweden
| | - Marie Larsson
- Division of Molecular Medicine and Virology, Department of Clinical and Experimental Medicine, Linköping UniversityLinköpingSweden
| |
Collapse
|
25
|
Chitrakar A, Budda SA, Henderson JG, Axtell RC, Zenewicz LA. E3 Ubiquitin Ligase Von Hippel-Lindau Protein Promotes Th17 Differentiation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:1009-1023. [PMID: 32690659 PMCID: PMC8167928 DOI: 10.4049/jimmunol.2000243] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 06/20/2020] [Indexed: 12/14/2022]
Abstract
Von Hippel-Lindau (VHL) is an E3 ubiquitin ligase that targets proteins, including HIF-1α, for proteasomal degradation. VHL and HIF regulate the balance between glycolysis and oxidative phosphorylation, which is critical in highly dynamic T cells. HIF-1α positively regulates Th17 differentiation, a complex process in which quiescent naive CD4 T cells undergo transcriptional changes to effector cells, which are commonly dysregulated in autoimmune diseases. The role of VHL in Th17 cells is not known. In this study, we hypothesized VHL negatively regulates Th17 differentiation and deletion of VHL in CD4 T cells would elevate HIF-1α and increase Th17 differentiation. Unexpectedly, we found that VHL promotes Th17 differentiation. Mice deficient in VHL in their T cells were resistant to an autoimmune disease, experimental autoimmune encephalomyelitis, often mediated by Th17 cells. In vitro Th17 differentiation was impaired in VHL-deficient T cells. In the absence of VHL, Th17 cells had decreased activation of STAT3 and SMAD2, suggesting that VHL indirectly or directly regulates these critical signaling molecules. Gene expression analysis revealed that in Th17 cells, VHL regulates many cellular pathways, including genes encoding proteins involved indirectly or directly in the glycolysis pathway. Compared with wild-type, VHL-deficient Th17 cells had elevated glycolysis and glycolytic capacity. Our finding has implications on the design of therapeutics targeting the distinct metabolic needs of T cells to combat chronic inflammatory diseases.
Collapse
Affiliation(s)
- Alisha Chitrakar
- Department of Microbiology and Immunology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104; and
| | - Scott A Budda
- Department of Microbiology and Immunology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104; and
| | - Jacob G Henderson
- Department of Microbiology and Immunology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104; and
| | - Robert C Axtell
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - Lauren A Zenewicz
- Department of Microbiology and Immunology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104; and
| |
Collapse
|
26
|
Menegon YA, Pinheiro NB, Santos LM, Rodrigues PRC, Avila LFC, Conceição FR, Leite FPL. Toxocara canis infection may impair bovine herpesvirus type 5 immunization. Res Vet Sci 2020; 132:268-270. [PMID: 32693251 DOI: 10.1016/j.rvsc.2020.06.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/12/2020] [Accepted: 06/30/2020] [Indexed: 11/17/2022]
Abstract
Helminths have developed complex mechanisms to suppress the host immune response. These mechanisms may impair the host vaccine response. This study aimed to evaluate the effect of Toxocara spp. infection on the vaccine immune response to bovine herpesvirus type 5 (BoHV-5). First, 30 heifers received two doses of an experimental BoHV-5 vaccine. At 42nd days after the primo vaccination the vaccine efficacy was evaluated, and the presence of anti-Toxocara antibodies. Second, 20 Balb/c mice were divided into two groups, one infected with T. canis and the other without infection. After infection, both groups received two doses of vaccine. The vaccine immune response was assessed by BoHV-5 serum neutralization and splenic cytokines transcription by qPCR. All heifers positive for Toxocara spp. (40%) showed BoHV-5 SN titer ≤1:32, whereas heifers negative for Toxocara spp. (60%) had BoHV-5 SN titer ≥1: 128. Infected T. canis mice showed BoHV-5 SN titer ≤1:2, whereas mice not infected with T. canis BoHV-5 SN titer ≥1:8. Splenocytes from control mice stimulated with BoHV-5 had a significant (p < .05) mRNA transcription for the cytokines IL-12, IL-17, and IL-23, whereas the same cytokines were down-regulated in T. canis infected mice. These results suggest that Toxocara spp. infection may impair BoHV-5 immunization and should be considered for efficient herd immunization.
Collapse
Affiliation(s)
- Y A Menegon
- Federal University of Pelotas, Center for Technological Development, Biotechnology, 96160-900 Capão do Leão, RS, Brazil
| | - N B Pinheiro
- Federal University of Pelotas, Parasitology Graduate Program, 96160-900 Capão do Leão, RS, Brazil
| | - L M Santos
- Federal University of Pelotas, Center for Technological Development, Biotechnology, 96160-900 Capão do Leão, RS, Brazil
| | - P R C Rodrigues
- Federal University of Pelotas, Veterinary School, Laboratory of Virology, 96160-900 Capão do Leão, RS, Brazil
| | - L F C Avila
- Federal University of Rio Grande, Medicine School, Laboratory of Parasitology, Rio Grande 96203-900, RS, Brazil
| | - F R Conceição
- Federal University of Pelotas, Center for Technological Development, Biotechnology, 96160-900 Capão do Leão, RS, Brazil
| | - F P L Leite
- Federal University of Pelotas, Center for Technological Development, Biotechnology, 96160-900 Capão do Leão, RS, Brazil; Federal University of Pelotas, Parasitology Graduate Program, 96160-900 Capão do Leão, RS, Brazil.
| |
Collapse
|
27
|
Xu W, Zhou W, Wang H, Liang S. Roles of Porphyromonas gingivalis and its virulence factors in periodontitis. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2020; 120:45-84. [PMID: 32085888 DOI: 10.1016/bs.apcsb.2019.12.001] [Citation(s) in RCA: 198] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Periodontitis is an infection-driven inflammatory disease, which is characterized by gingival inflammation and bone loss. Periodontitis is associated with various systemic diseases, including cardiovascular, respiratory, musculoskeletal, and reproductive system related abnormalities. Recent theory attributes the pathogenesis of periodontitis to oral microbial dysbiosis, in which Porphyromonas gingivalis acts as a critical agent by disrupting host immune homeostasis. Lipopolysaccharide, proteases, fimbriae, and some other virulence factors are among the strategies exploited by P. gingivalis to promote the bacterial colonization and facilitate the outgrowth of the surrounding microbial community. Virulence factors promote the coaggregation of P. gingivalis with other bacteria and the formation of dental biofilm. These virulence factors also modulate a variety of host immune components and subvert the immune response to evade bacterial clearance or induce an inflammatory environment. In this chapter, our focus is to discuss the virulence factors of periodontal pathogens, especially P. gingivalis, and their roles in regulating immune responses during periodontitis progression.
Collapse
Affiliation(s)
- Weizhe Xu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China; Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, United States
| | - Wei Zhou
- Department of Endodontics, Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, Pudong, China
| | - Huizhi Wang
- VCU Philips Institute for Oral Health Research, Department of Oral and Craniofacial Molecular Biology, Virginia Commonwealth University School of Dentistry, Richmond, VA, United States
| | - Shuang Liang
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, United States
| |
Collapse
|
28
|
Chen YL, Gutowska-Owsiak D, Hardman CS, Westmoreland M, MacKenzie T, Cifuentes L, Waithe D, Lloyd-Lavery A, Marquette A, Londei M, Ogg G. Proof-of-concept clinical trial of etokimab shows a key role for IL-33 in atopic dermatitis pathogenesis. Sci Transl Med 2019; 11:eaax2945. [PMID: 31645451 DOI: 10.1126/scitranslmed.aax2945] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/15/2019] [Accepted: 09/25/2019] [Indexed: 12/15/2022]
Abstract
Targeted inhibition of cytokine pathways provides opportunities to understand fundamental biology in vivo in humans. The IL-33 pathway has been implicated in the pathogenesis of atopy through genetic and functional associations. We investigated the role of IL-33 inhibition in a first-in-class phase 2a study of etokimab (ANB020), an IgG1 anti-IL-33 monoclonal antibody, in patients with atopic dermatitis (AD). Twelve adult patients with moderate to severe AD received a single systemic administration of etokimab. Rapid and sustained clinical benefit was observed, with 83% achieving Eczema Area and Severity Index 50 (EASI50), and 33% EASI75, with reduction in peripheral eosinophils at day 29 after administration. We noted significant reduction in skin neutrophil infiltration after etokimab compared with placebo upon skin challenge with house dust mite, reactivity to which has been implicated in the pathogenesis of AD. We showed that etokimab also inhibited neutrophil migration to skin interstitial fluid in vitro. Besides direct effects on neutrophil migration, etokimab revealed additional unexpected CXCR1-dependent effects on IL-8-induced neutrophil migration. These human in vivo findings confirm an IL-33 upstream role in modulating skin inflammatory cascades and define the therapeutic potential for IL-33 inhibition in human diseases, including AD.
Collapse
Affiliation(s)
- Yi-Ling Chen
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Danuta Gutowska-Owsiak
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
- Institute of Biotechnology UG, Intercollegiate Faculty of Biotechnology of University of Gdańsk and Medical University of Gdańsk, 80-307 Gdańsk, Poland
| | - Clare S Hardman
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | | | | | | | - Dominic Waithe
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | | | | | | | - Graham Ogg
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK.
- Oxford University Hospitals, Oxford OX3 7LE, UK
| |
Collapse
|
29
|
David SC, Laan Z, Minhas V, Chen AY, Davies J, Hirst TR, McColl SR, Alsharifi M, Paton JC. Enhanced safety and immunogenicity of a pneumococcal surface antigen A mutant whole-cell inactivated pneumococcal vaccine. Immunol Cell Biol 2019; 97:726-739. [PMID: 31050022 DOI: 10.1111/imcb.12257] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 03/21/2019] [Accepted: 04/29/2019] [Indexed: 01/14/2023]
Abstract
Existing capsular polysaccharide-based vaccines against pneumococcal disease are highly effective against vaccine-included serotypes, but they are unable to combat serotype replacement. We have developed a novel pneumococcal vaccine that confers serotype-independent protection, and could therefore constitute a "universal" vaccine formulation. This preparation is comprised of whole un-encapsulated pneumococci inactivated with gamma irradiation (γ-PN), and we have previously reported induction of cross-reactive immunity after nonadjuvanted intranasal vaccination. To further enhance vaccine immunogenicity and safety, we modified the pneumococcal vaccine strain to induce a stressed state during growth. Specifically, the substrate binding component of the psaBCA operon for manganese import was mutated to create a pneumococcal surface antigen A (psaA) defective vaccine strain. psaA mutation severely attenuated the growth of the vaccine strain in vitro without negatively affecting pneumococcal morphology, thereby enhancing vaccine safety. In addition, antibodies raised against vaccine preparations based on the modified strain [γ-PN(ΔPsaA)] showed more diversified reactivity to wild-type pneumococcal challenge strains compared to those induced by the original formulation. The modified vaccine also induced comparable protective TH 17 responses in the lung, and conferred greater protection against lethal heterologous pneumococcal challenge. Overall, the current study demonstrates successful refinement of a serotype-independent pneumococcal vaccine candidate to enhance safety and immunogenicity.
Collapse
Affiliation(s)
- Shannon C David
- Research Centre for Infectious Diseases, and Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Zoe Laan
- Research Centre for Infectious Diseases, and Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Vikrant Minhas
- Research Centre for Infectious Diseases, and Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Austen Y Chen
- Research Centre for Infectious Diseases, and Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Justin Davies
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, Australia
| | - Timothy R Hirst
- Research Centre for Infectious Diseases, and Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.,GPN Vaccines Pty Ltd, Yarralumla, ACT, Australia.,Gamma Vaccines Pty Ltd, Yarralumla, ACT, Australia
| | - Shaun R McColl
- Research Centre for Infectious Diseases, and Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Mohammed Alsharifi
- Research Centre for Infectious Diseases, and Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.,GPN Vaccines Pty Ltd, Yarralumla, ACT, Australia.,Gamma Vaccines Pty Ltd, Yarralumla, ACT, Australia
| | - James C Paton
- Research Centre for Infectious Diseases, and Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.,GPN Vaccines Pty Ltd, Yarralumla, ACT, Australia
| |
Collapse
|
30
|
Bassi PB, Araujo FF, Garcia GC, Costa E Silva MF, Bittar ER, Bertonha CM, Martins-Filho OA, Araujo MSS, Bittar JF. Haematological and immunophenotypic evaluation of peripheral blood cells of cattle naturally infected with bovine papillomavirus. Vet J 2019; 244:112-115. [PMID: 30825886 DOI: 10.1016/j.tvjl.2018.12.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 11/29/2018] [Accepted: 12/04/2018] [Indexed: 01/28/2023]
Abstract
Papillomaviruses are among the most widespread animal viruses, with many hosts harbouring multiple virus types. The present study aimed to evaluate the haematological and immunophenotypic profile of cattle infected with bovine papillomavirus (BPV). Blood samples were collected from 10 animals with clinical cutaneous BPV and without clinical papillomatosis (control). Haematological analysis demonstrated a significant reduction in haemoglobin and haematocrit for BPV-infected animals. The results also showed an increase of natural killer cells and a decrease of γδ+ T-cells and the CD4+/CD8+ ratio for the BPV group when compared to the control group. The infection was also found to stimulate a pro-inflammatory profile with the participation of CD8+T cells producing elevated IFN-γ and IL-17. These findings, although preliminary, provide a better understanding of the immune response of cattle infected with BPV.
Collapse
Affiliation(s)
- Paula B Bassi
- Universidade de Uberaba (UNIUBE), Medicina Veterinária, Mestrado em Sanidade e Produção Animal nos Trópicos - Avenida Nenê Sabino 1697/1698, 38055-500, Uberaba-MG, Brazil
| | - Fernanda F Araujo
- Universidade de Uberaba (UNIUBE), Medicina Veterinária, Mestrado em Sanidade e Produção Animal nos Trópicos - Avenida Nenê Sabino 1697/1698, 38055-500, Uberaba-MG, Brazil.; Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou - Fundação Oswaldo Cruz. Avenida Augusto de Lima nº 1715, 30190-002, Barro Preto, Belo Horizonte-MG, Brazil
| | - Guilherme C Garcia
- Universidade de Uberaba (UNIUBE), Medicina Veterinária, Mestrado em Sanidade e Produção Animal nos Trópicos - Avenida Nenê Sabino 1697/1698, 38055-500, Uberaba-MG, Brazil
| | - Matheus F Costa E Silva
- Universidade de Uberaba (UNIUBE), Medicina Veterinária, Mestrado em Sanidade e Produção Animal nos Trópicos - Avenida Nenê Sabino 1697/1698, 38055-500, Uberaba-MG, Brazil.; Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou - Fundação Oswaldo Cruz. Avenida Augusto de Lima nº 1715, 30190-002, Barro Preto, Belo Horizonte-MG, Brazil
| | - Eustaquio R Bittar
- Universidade de Uberaba (UNIUBE), Medicina Veterinária, Mestrado em Sanidade e Produção Animal nos Trópicos - Avenida Nenê Sabino 1697/1698, 38055-500, Uberaba-MG, Brazil
| | - Candice M Bertonha
- Universidade de Uberaba (UNIUBE), Medicina Veterinária, Mestrado em Sanidade e Produção Animal nos Trópicos - Avenida Nenê Sabino 1697/1698, 38055-500, Uberaba-MG, Brazil
| | - Olindo A Martins-Filho
- Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou - Fundação Oswaldo Cruz. Avenida Augusto de Lima nº 1715, 30190-002, Barro Preto, Belo Horizonte-MG, Brazil
| | - Marcio Sobreira Silva Araujo
- Universidade de Uberaba (UNIUBE), Medicina Veterinária, Mestrado em Sanidade e Produção Animal nos Trópicos - Avenida Nenê Sabino 1697/1698, 38055-500, Uberaba-MG, Brazil.; Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou - Fundação Oswaldo Cruz. Avenida Augusto de Lima nº 1715, 30190-002, Barro Preto, Belo Horizonte-MG, Brazil..
| | - Joely F Bittar
- Universidade de Uberaba (UNIUBE), Medicina Veterinária, Mestrado em Sanidade e Produção Animal nos Trópicos - Avenida Nenê Sabino 1697/1698, 38055-500, Uberaba-MG, Brazil
| |
Collapse
|
31
|
Huang J, Luo S, Huang M, Zhang T, Min Z, Liu C, Zhang Q, Yang J, Min X. Protection against fatal pneumonia through mucosal and subcutaneous immunization with the pneumococcal SP0148 protein. Microb Pathog 2019; 129:206-212. [PMID: 30772476 DOI: 10.1016/j.micpath.2019.02.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/13/2019] [Accepted: 02/13/2019] [Indexed: 12/14/2022]
Abstract
Streptococcus pneumoniae infection is associated with very high morbidity and mortality throughout the world. Vaccines are an effective measure for the reduction of S. pneumoniae infection. In particular, protein vaccines are attracting increasing attention because of their good immunogenicity and wide coverage of serotypes. Therefore, identifying effective protein vaccine targets is important for protein vaccine development. SP0148 is a promising protein vaccine target for S. pneumoniae and is capable of reducing S. pneumoniae colonization in the nasopharynx of mice through the IL-17A pathway. However, the protective effects of SP0148 in fatal pneumococcal infection have not been evaluated. This study used subcutaneous and nasal immunization routes to systematically evaluate the protective effects of the SP0148 protein in fatal pneumococcal infection. Subcutaneous and nasal mucosal immunization with recombinant SP0148 protein produced effective immune protection against infection with a lethal dose of S. pneumoniae and significantly prolonged survival time and increased the survival rate of mice. Furthermore, nasal immunization with SP0148 induced mouse splenocytes to secrete high levels of the cytokines IFN-γ and IL-17A. Both recombinant SP0148 protein and its antiserum inhibited the adhesion of S.pneumoniae D39 to A549 human lung epithelial cells in a dose-dependent manner. In summary, SP0148 induced mice to produce protective immune responses to fatal S. pneumoniae infection, and our results could contribute to the accumulating data on the use of SP0148 protein vaccines.
Collapse
Affiliation(s)
- Jian Huang
- Department of Laboratory Medicine, The First Affiliated Hospital, Zunyi Medical University, Zunyi, 563003, China
| | - Shilu Luo
- Department of Laboratory Medicine, The First Affiliated Hospital, Zunyi Medical University, Zunyi, 563003, China
| | - Meirong Huang
- Department of Blood Transfusion, The First Affiliated Hospital, Zunyi Medical University, Zunyi, 563003, China
| | - Tao Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital, Zunyi Medical University, Zunyi, 563003, China
| | - Zongsu Min
- Zunyi Maternal and Child Health Hospital, Zunyi, 563000, China
| | - Changjin Liu
- Department of Laboratory Medicine, The First Affiliated Hospital, Zunyi Medical University, Zunyi, 563003, China
| | - Qing Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital, Zunyi Medical University, Zunyi, 563003, China
| | - Jianru Yang
- Department of Laboratory Medicine, The First Affiliated Hospital, Zunyi Medical University, Zunyi, 563003, China
| | - Xun Min
- Department of Laboratory Medicine, The First Affiliated Hospital, Zunyi Medical University, Zunyi, 563003, China.
| |
Collapse
|
32
|
Ojha S, Kumar B. A review on nanotechnology based innovations in diagnosis and treatment of multiple sclerosis. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.jocit.2017.12.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
33
|
Verboket R, Herrera-Vizcaíno C, Thorwart K, Booms P, Bellen M, Al-Maawi S, Sader R, Marzi I, Henrich D, Ghanaati S. Influence of concentration and preparation of platelet rich fibrin on human bone marrow mononuclear cells (in vitro). Platelets 2018; 30:861-870. [PMID: 30359164 DOI: 10.1080/09537104.2018.1530346] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Large bone defects have always been a big challenge. The use of bone marrow mononuclear cells (BMCs) combined with an osteoconductive scaffold has been proved a good alternative for the treatment of large bone defects. Another autologous source for tissue engineering is platelet rich fibrin (PRF). PRF is a blood concentrate system obtained through a one-step centrifugation. The generated 3D matrix of the PRF clot serves as a reservoir of growth factors. Those growth factors might support the regenerative response of BMC, and therefore the effect of PRF, centrifuged with either high medium (208 g) or low (60 g) relative centrifugation force (RCF) on BMCs was evaluated in vitro in the present study. The two PRF matrices obtained were initially characterized and compared to human serum. Significantly increased concentrations of insulin-like growth factor (IGF), soluble intercellular adhesion molecule-1 (sICAM1) and transforming growth factor (TGF)-β were found in PRF compared to human serum whereas VEGF concentration was not significantly altered. A dose-response study revealed no further activation of BMC's metabolic activity, if concentration of both PRF matrices exceeded 10% (v/v). Effect of both PRF preparations [10%] on BMC was analyzed after 2, 7, and 14 days in comparison to human serum [10%]. Metabolic activity of BMC increased significantly in all groups on day 14. Furthermore, gene expression of matrix metalloproteinases (MMP)-2, -7, and -9 was significantly stimulated in BMC cultivated with the respective PRF matrices compared to human serum. Apoptotic activity of BMC incubated with PRF was not altered compared to BMC cultivated with serum. In conclusion, PRF could be used as a growth factor delivery system of autologous or allogeneic source with the capability of stimulating cells such as BMC.
Collapse
Affiliation(s)
- René Verboket
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt , Frankfurt , Germany
| | - Carlos Herrera-Vizcaíno
- Clinic for Maxillofacial and Plastic Surgery, FORM, Frankfurt Oral Regenerative Medicine, Johann Wolfgang Goethe University , Frankfurt Am Main , Germany
| | - Kirsten Thorwart
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt , Frankfurt , Germany
| | - Patrick Booms
- Clinic for Maxillofacial and Plastic Surgery, FORM, Frankfurt Oral Regenerative Medicine, Johann Wolfgang Goethe University , Frankfurt Am Main , Germany
| | - Marlene Bellen
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt , Frankfurt , Germany
| | - Sarah Al-Maawi
- Clinic for Maxillofacial and Plastic Surgery, FORM, Frankfurt Oral Regenerative Medicine, Johann Wolfgang Goethe University , Frankfurt Am Main , Germany
| | - Robert Sader
- Clinic for Maxillofacial and Plastic Surgery, FORM, Frankfurt Oral Regenerative Medicine, Johann Wolfgang Goethe University , Frankfurt Am Main , Germany
| | - Ingo Marzi
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt , Frankfurt , Germany
| | - Dirk Henrich
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt , Frankfurt , Germany
| | - Shahram Ghanaati
- Clinic for Maxillofacial and Plastic Surgery, FORM, Frankfurt Oral Regenerative Medicine, Johann Wolfgang Goethe University , Frankfurt Am Main , Germany
| |
Collapse
|
34
|
Shen Y, Li J, Wang SQ, Jiang W. Ambiguous roles of innate lymphoid cells in chronic development of liver diseases. World J Gastroenterol 2018; 24:1962-1977. [PMID: 29760540 PMCID: PMC5949710 DOI: 10.3748/wjg.v24.i18.1962] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 04/25/2018] [Accepted: 05/06/2018] [Indexed: 02/06/2023] Open
Abstract
Innate lymphoid cells (ILCs) are defined as a distinct arm of innate immunity. According to their profile of secreted cytokines and lineage-specific transcriptional factors, ILCs can be categorized into the following three groups: group 1 ILCs (including natural killer (NK) cells and ILC1s) are dependent on T-bet and can produce interferon-γ; group 2 ILCs (ILC2s) are dependent on GATA3 and can produce type 2 cytokines, including interleukin (IL)-5 and IL-13; and, group 3 ILCs (including lymphoid tissue-like cells and ILC3s) are dependent on RORγt and can produce IL-22 and IL-17. Collaborative with adaptive immunity, ILCs are highly reactive innate effectors that promptly orchestrate immunity, inflammation and tissue repair. Dysregulation of ILCs might result in inflammatory disorders. Evidence regarding the function of intrahepatic ILCs is emerging from longitudinal studies of inflammatory liver diseases wherein they exert both physiological and pathological functions, including immune homeostasis, defenses and surveillance. Their overall effect on the liver depends on the balance of their proinflammatory and antiinflammatory populations, specific microenvironment and stages of immune responses. Here, we review the current data about ILCs in chronic liver disease progression, to reveal their roles in different stages as well as to discuss their therapeutic potency as intervention targets.
Collapse
Affiliation(s)
- Yue Shen
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jing Li
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of Gastroenterology, Tongji Hospital, Tongji University, Shanghai 200000, China
| | - Si-Qi Wang
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wei Jiang
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
35
|
Rose DR, Yang H, Serena G, Sturgeon C, Ma B, Careaga M, Hughes HK, Angkustsiri K, Rose M, Hertz-Picciotto I, Van de Water J, Hansen RL, Ravel J, Fasano A, Ashwood P. Differential immune responses and microbiota profiles in children with autism spectrum disorders and co-morbid gastrointestinal symptoms. Brain Behav Immun 2018; 70:354-368. [PMID: 29571898 PMCID: PMC5953830 DOI: 10.1016/j.bbi.2018.03.025] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 03/08/2018] [Accepted: 03/19/2018] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES Many studies have reported the increased presence of gastrointestinal (GI) symptoms in children with autism spectrum disorders (ASD). Altered microbiome profiles, pro-inflammatory responses and impaired intestinal permeability have been observed in children with ASD and co-morbid GI symptoms, yet few studies have compared these findings to ASD children without GI issues or similarly aged typical developing children. The aim of this study was to determine whether there are biological signatures in terms of immune dysfunction and microbiota composition in children with ASD with GI symptoms. METHODS Children were enrolled in one of four groups: ASD and GI symptoms of irregular bowel habits (ASDGI), children with ASD but without current or previous GI symptoms (ASDNoGI), typically developing children with GI symptoms (TDGI) and typically developing children without current or previous GI symptoms (TDNoGI). Peripheral blood mononuclear cells (PBMC) were isolated from the blood, stimulated and assessed for cytokine production, while stool samples were analyzed for microbial composition. RESULTS Following Toll-Like receptor (TLR)-4 stimulation, the ASDGI group produced increased levels of mucosa-relevant cytokines including IL-5, IL-15 and IL-17 compared to ASDNoGI. The production of the regulatory cytokine TGFβ1 was decreased in the ASDGI group compared with both the ASDNoGI and TDNoGI groups. Analysis of the microbiome at the family level revealed differences in microbiome composition between ASD and TD children with GI symptoms; furthermore, a predictive metagenome functional content analysis revealed that pathways were differentially represented between ASD and TD subjects, independently of the presence of GI symptoms. The ASDGI also showed an over-representation of the gene encoding zonulin, a molecule regulating gut permeability, compared to the other groups. CONCLUSIONS Overall our findings suggest that children with ASD who experience GI symptoms have an imbalance in their immune response, possibly influenced by or influencing metagenomic changes, and may have a propensity to impaired gut barrier function which may contribute to their symptoms and clinical outcome.
Collapse
Affiliation(s)
- Destanie R Rose
- Department of Medical Microbiology and Immunology, University of California Davis, USA; MIND Institute, University of California Davis, USA
| | - Houa Yang
- Department of Medical Microbiology and Immunology, University of California Davis, USA; MIND Institute, University of California Davis, USA
| | - Gloria Serena
- Division of Pediatric Gastroenterology and Nutrition, Center for Celiac Research, MassGeneral Hospital for Children, Boston, MA, USA; Graduate Program in Life Sciences University of Maryland School of Medicine, Baltimore, MD, USA
| | - Craig Sturgeon
- Division of Pediatric Gastroenterology and Nutrition, Center for Celiac Research, MassGeneral Hospital for Children, Boston, MA, USA; Graduate Program in Life Sciences University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bing Ma
- Institute of Genomic Science, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Milo Careaga
- Department of Medical Microbiology and Immunology, University of California Davis, USA; MIND Institute, University of California Davis, USA
| | - Heather K Hughes
- Department of Medical Microbiology and Immunology, University of California Davis, USA; MIND Institute, University of California Davis, USA
| | - Kathy Angkustsiri
- MIND Institute, University of California Davis, USA; Department of Pediatrics, University of California Davis, USA; Children's Center for Environmental Health, University of California Davis, CA, USA
| | - Melissa Rose
- Children's Center for Environmental Health, University of California Davis, CA, USA; Public Health Sciences, University of California Davis, CA, USA
| | - Irva Hertz-Picciotto
- MIND Institute, University of California Davis, USA; Children's Center for Environmental Health, University of California Davis, CA, USA; Public Health Sciences, University of California Davis, CA, USA
| | - Judy Van de Water
- MIND Institute, University of California Davis, USA; Children's Center for Environmental Health, University of California Davis, CA, USA; Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis, CA, USA
| | - Robin L Hansen
- MIND Institute, University of California Davis, USA; Department of Pediatrics, University of California Davis, USA; Children's Center for Environmental Health, University of California Davis, CA, USA
| | - Jacques Ravel
- Institute of Genomic Science, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alessio Fasano
- Division of Pediatric Gastroenterology and Nutrition, Center for Celiac Research, MassGeneral Hospital for Children, Boston, MA, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California Davis, USA; MIND Institute, University of California Davis, USA; Children's Center for Environmental Health, University of California Davis, CA, USA.
| |
Collapse
|
36
|
Th17-lineage cells in pulmonary sarcoidosis and Löfgren's syndrome: Friend or foe? J Autoimmun 2018; 87:82-96. [DOI: 10.1016/j.jaut.2017.12.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 12/12/2017] [Indexed: 01/17/2023]
|
37
|
Interleukin-37 Ameliorates Coxsackievirus B3-induced Viral Myocarditis by Modulating the Th17/Regulatory T cell Immune Response. J Cardiovasc Pharmacol 2018; 69:305-313. [PMID: 28207428 DOI: 10.1097/fjc.0000000000000476] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Myocarditis is a heterogeneous group of disorders defined by inflammation of the heart muscle with an excessively activated immune response. Numerous interventions have been investigated for the treatment of myocarditis while success is limited. Interleukin-37 (IL-37), a novel member of the IL-1 cytokine family, is a natural inhibitor of innate immunity associated with autoimmune diseases. However, the modulatory effect of IL-37 in myocarditis is unknown. In this study, we investigated the immunological regulation of IL-37 in the coxsackievirus B3-induced model of murine viral myocarditis. The results show that IL-37 significantly ameliorates the signs of myocarditis with increased survival rate and bodyweight, improved histological changes, reduced activities of MB isoenzyme of creatine kinase and cardiac troponin I, and a suppressed response of Th17 cells and enhanced response of regulatory T cells (Tregs) in the spleen. Moreover, IL-37 down-regulates the expression of Th17-related cytokines IL-6 and IL-17A, while promoting Treg-related cytokine IL-10 levels in the heart. Therefore, IL-37 may exhibit anti-inflammatory activity in the murine model of myocarditis by regulating the balance between Th17 and Treg cells, thereby providing a possible novel therapeutic target in myocarditis.
Collapse
|
38
|
Patel AB, Tsilioni I, Weng Z, Theoharides TC. TNF stimulates IL-6, CXCL8 and VEGF secretion from human keratinocytes via activation of mTOR, inhibited by tetramethoxyluteolin. Exp Dermatol 2018; 27:135-143. [PMID: 29105195 DOI: 10.1111/exd.13461] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2017] [Indexed: 12/19/2022]
Abstract
Psoriasis is an autoimmune skin disease characterized by keratinocyte hyperproliferation and chronic inflammation. The pathogenesis of psoriasis involves proinflammatory cytokines, such as tumor necrosis factor (TNF), but the mechanism of keratinocyte activation is not well understood. Here, we show that TNF (10 or 50 ng/mL) stimulates a significant (P < .0001) gene expression and secretion of proinflammatory IL-6, CXCL8 and VEGF from both cultured human HaCaT and normal epidermal human keratinocytes (NHEKs). This effect occurs via activation of the mammalian target of rapamycin (mTOR) signalling complex as shown by Western blot analysis and phospho-ELISAs. Pretreatment with the novel natural flavonoid tetramethoxyluteolin (10-100 μmol L-1 ) significantly (P < .0001) inhibits gene expression and secretion (P < .0001) of all 3 mediators in a concentration-dependent manner. Moreover, tetramethoxyluteolin (50 μmol L-1 ) appears to be a potent inhibitor of the phosphorylated mTOR substrates (pmTORSer2448 , pp70S6KThr389 and p4EBP1Thr37/46 ) as compared to known mTOR inhibitors in keratinocytes. The present findings indicate that TNF stimulates skin inflammation via mTOR signalling. Inhibition by tetramethoxyluteolin may be used in the treatment for psoriasis.
Collapse
Affiliation(s)
- Arti B Patel
- Molecular Immunopharmacology and Drug Discovery Laboratory, Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA.,Graduate Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA
| | - Irene Tsilioni
- Molecular Immunopharmacology and Drug Discovery Laboratory, Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA
| | - Zuyi Weng
- Molecular Immunopharmacology and Drug Discovery Laboratory, Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA
| | - Theoharis C Theoharides
- Molecular Immunopharmacology and Drug Discovery Laboratory, Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA.,Graduate Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA.,Department of Internal Medicine, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
39
|
The Th17 Lineage: From Barrier Surfaces Homeostasis to Autoimmunity, Cancer, and HIV-1 Pathogenesis. Viruses 2017; 9:v9100303. [PMID: 29048384 PMCID: PMC5691654 DOI: 10.3390/v9100303] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 12/12/2022] Open
Abstract
The T helper 17 (Th17) cells represent a subset of CD4+ T-cells with unique effector functions, developmental plasticity, and stem-cell features. Th17 cells bridge innate and adaptive immunity against fungal and bacterial infections at skin and mucosal barrier surfaces. Although Th17 cells have been extensively studied in the context of autoimmunity, their role in various other pathologies is underexplored and remains an area of open investigation. This review summarizes the history of Th17 cell discovery and the current knowledge relative to the beneficial role of Th17 cells in maintaining mucosal immunity homeostasis. We further discuss the concept of Th17 pathogenicity in the context of autoimmunity, cancer, and HIV infection, and we review the most recent discoveries on molecular mechanisms regulating HIV replication/persistence in pathogenic Th17 cells. Finally, we stress the need for novel fundamental research discovery-based Th17-specific therapeutic interventions to treat pathogenic conditions associated with Th17 abnormalities, including HIV infection.
Collapse
|
40
|
Withers DR, Hepworth MR. Group 3 Innate Lymphoid Cells: Communications Hubs of the Intestinal Immune System. Front Immunol 2017; 8:1298. [PMID: 29085366 PMCID: PMC5649144 DOI: 10.3389/fimmu.2017.01298] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 09/27/2017] [Indexed: 12/12/2022] Open
Abstract
The maintenance of mammalian health requires the generation of appropriate immune responses against a broad range of environmental and microbial challenges, which are continually encountered at barrier tissue sites including the skin, lung, and gastrointestinal tract. Dysregulated barrier immune responses result in inflammation, both locally and systemically in peripheral organs. Group 3 innate lymphoid cells (ILC3) are constitutively present at barrier sites and appear to be highly specialized in their ability to sense a range of environmental and host-derived signals. Under homeostatic conditions, ILC3 respond to local cues to maintain tissue homeostasis and restrict inflammatory responses. In contrast, perturbations in the tissue microenvironment resulting from disease, infection, or tissue damage can drive dysregulated pro-inflammatory ILC3 responses and contribute to immunopathology. The tone of the ILC3 response is dictated by a balance of “exogenous” signals, such as dietary metabolites and commensal microbes, and “endogenous” host-derived signals from stromal cells, immune cells, and the nervous system. ILC3 must therefore have the capacity to simultaneously integrate a wide array of complex and dynamic inputs in order to regulate barrier function and tissue health. In this review, we discuss the concept of ILC3 as a “communications hub” in the intestinal tract and associated lymphoid tissues and address the variety of signals, derived from multiple biological systems, which are interpreted by ILC3 to modulate the release of downstream effector molecules and regulate cell–cell crosstalk. Successful integration of environmental cues by ILC3 and downstream propagation to the broader immune system is required to maintain a tolerogenic and anti-inflammatory tone and reinforce barrier function, whereas dysregulation of ILC3 responses can contribute to the onset or progression of clinically relevant chronic inflammatory diseases.
Collapse
Affiliation(s)
- David R Withers
- College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy (III), University of Birmingham, Birmingham, United Kingdom
| | - Matthew R Hepworth
- Manchester Collaborative Centre for Inflammation Research (MCCIR), Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
41
|
Kelley CF, Kraft CS, de Man TJ, Duphare C, Lee HW, Yang J, Easley KA, Tharp GK, Mulligan MJ, Sullivan PS, Bosinger SE, Amara RR. The rectal mucosa and condomless receptive anal intercourse in HIV-negative MSM: implications for HIV transmission and prevention. Mucosal Immunol 2017; 10:996-1007. [PMID: 27848950 PMCID: PMC5433931 DOI: 10.1038/mi.2016.97] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 09/23/2016] [Indexed: 02/04/2023]
Abstract
Most HIV transmissions among men who have sex with men (MSM), the group that accounted for 67% of new US infections in 2014, occur via exposure to the rectal mucosa. However, it is unclear how the act of condomless receptive anal intercourse (CRAI) may alter the mucosal immune environment in HIV-negative MSM. Here, we performed a comprehensive characterization of the rectal mucosal immune environment for the phenotype and production of pro-inflammatory cytokines by CD4 and CD8 T cells, global transcriptomic analyses, and the composition of microbiota in HIV-negative MSM. Our results show that compared with men who had never engaged in anal intercourse, the rectal mucosa of MSM engaging in CRAI has a distinct phenotype characterized by higher levels of Th17 cells, greater CD8+ T cell proliferation and production of pro-inflammatory cytokines, molecular signatures associated with mucosal injury and repair likely mediated by innate immune cells, and a microbiota enriched for the Prevotellaceae family. These data provide a high-resolution model of the immunological, molecular, and microbiological perturbations induced by CRAI, will have direct utility in understanding rectal HIV transmission among MSM, and will enhance the design of future biomedical prevention interventions, including candidate HIV vaccines.
Collapse
Affiliation(s)
- Colleen F. Kelley
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine,Department of Epidemiology, Rollins School of Public Health, Emory University,Corresponding author;
| | - Colleen S. Kraft
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine,Department of Pathology, Emory University School of Medicine
| | - Tom J.B. de Man
- Division of Healthcare Quality Promotion, Centers for Disease Control and Prevention
| | - Chandni Duphare
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University
| | - Hyun-Woo Lee
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University
| | - Jing Yang
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University
| | - Kirk A. Easley
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University
| | - Gregory K. Tharp
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University
| | - Mark J. Mulligan
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine
| | - Patrick S. Sullivan
- Department of Epidemiology, Rollins School of Public Health, Emory University
| | - Steven E. Bosinger
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University
| | - Rama R. Amara
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University,Department of Microbiology and Immunology, Emory University School of Medicine
| |
Collapse
|
42
|
Madecassic acid, the contributor to the anti-colitis effect of madecassoside, enhances the shift of Th17 toward Treg cells via the PPARγ/AMPK/ACC1 pathway. Cell Death Dis 2017; 8:e2723. [PMID: 28358365 PMCID: PMC5386545 DOI: 10.1038/cddis.2017.150] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 02/20/2017] [Accepted: 02/20/2017] [Indexed: 02/07/2023]
Abstract
The imbalance between Th17 and Treg cells substantially contributes to the intestinal immune disturbance and subsequent tissue injury in ulcerative colitis. The triterpenoid-rich fraction of Centella asiatica was able to ameliorate dextran sulfate sodium-induced colitis in mice. Here we explored its active ingredient and underlying mechanism with a focus on restoring the Th17/Treg balance. The four main triterpenoids occurring in C. asiatica were shown to attenuate colitis in mice by oral administration. The most effective ingredient madecassoside lost anti-colitis effect when applied topically in the colon, and madecassic acid was recognized to be the active form of madecassoside. Oral administration of madecassic acid decreased the percentage of Th17 cells and downregulated the expression of RORγt, IL-17A, IL-17F, IL-21 and IL-22 and increased the percentage of Treg cells and the expression of Foxp3 and IL-10 in the colons of mice with colitis, but it did not affect Th1 and Th2 cells. Under Th17-polarizing conditions, madecassic acid downregulated ACC1 expression and enhanced the shift of Th17 cells toward Treg cells, but it did not affect the differentiation of Treg cells under Treg-polarizing conditions. Both compound C and AMPK siRNA inhibited the madecassic acid-mediated downregulation of ACC1 expression and shift of Th17 cells to Treg cells under Th17-polarizing conditions. GW9662, T0070907 and PPARγ siRNA blocked the effect of madecassic acid on AMPK activation, ACC1 expression and shift of Th17 cells to Treg cells. Furthermore, madecassic acid was identified as a PPARγ agonist, as it promoted PPARγ transactivation. The correlation between activation of PPARγ and AMPK, downregulation of ACC1 expression, restoration of Th17/Treg balance and attenuation of colitis by madecassic acid was validated in mice with DSS-induced colitis. In conclusion, madecassic acid was the active form of madecassoside in ameliorating colitis by restoring the Th17/Treg balance via regulating the PPARγ/AMPK/ACC1 pathway.
Collapse
|
43
|
Franchi L, Monteleone I, Hao LY, Spahr MA, Zhao W, Liu X, Demock K, Kulkarni A, Lesch CA, Sanchez B, Carter L, Marafini I, Hu X, Mashadova O, Yuan M, Asara JM, Singh H, Lyssiotis CA, Monteleone G, Opipari AW, Glick GD. Inhibiting Oxidative Phosphorylation In Vivo Restrains Th17 Effector Responses and Ameliorates Murine Colitis. THE JOURNAL OF IMMUNOLOGY 2017; 198:2735-2746. [PMID: 28242647 DOI: 10.4049/jimmunol.1600810] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 01/30/2017] [Indexed: 12/21/2022]
Abstract
Integration of signaling and metabolic pathways enables and sustains lymphocyte function. Whereas metabolic changes occurring during T cell activation are well characterized, the metabolic demands of differentiated T lymphocytes are largely unexplored. In this study, we defined the bioenergetics of Th17 effector cells generated in vivo. These cells depend on oxidative phosphorylation (OXPHOS) for energy and cytokine production. Mechanistically, the essential role of OXPHOS in Th17 cells results from their limited capacity to increase glycolysis in response to metabolic stresses. This metabolic program is observed in mouse and human Th17 cells, including those isolated from Crohn disease patients, and it is linked to disease, as inhibiting OXPHOS reduces the severity of murine colitis and psoriasis. These studies highlight the importance of analyzing metabolism in effector lymphocytes within in vivo inflammatory contexts and suggest a therapeutic role for manipulating OXPHOS in Th17-driven diseases.
Collapse
Affiliation(s)
- Luigi Franchi
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109
| | - Ivan Monteleone
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | | | | | - Wenpu Zhao
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109
| | - Xikui Liu
- Lycera Corporation, Ann Arbor, MI 48109
| | | | | | | | | | | | - Irene Marafini
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Xiao Hu
- Lycera Corporation, Ann Arbor, MI 48109
| | - Oksana Mashadova
- Meyer Cancer Center, Weill Cornell Medical College, New York, NY 10065
| | - Min Yuan
- Division of Signal Transduction, Beth Israel Deaconess Medical Center, Boston, MA 02115
| | - John M Asara
- Department of Medicine, Harvard Medical School, Boston, MA 02115
| | - Harinder Singh
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109.,Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Anthony W Opipari
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, 48109; and
| | - Gary D Glick
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
44
|
Zenobia C, Hajishengallis G. Basic biology and role of interleukin-17 in immunity and inflammation. Periodontol 2000 2017; 69:142-59. [PMID: 26252407 DOI: 10.1111/prd.12083] [Citation(s) in RCA: 312] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2014] [Indexed: 02/06/2023]
Abstract
Interleukin-17 (also known as interleukin-17A) is a key cytokine that links T-cell activation to neutrophil mobilization and activation. As such, interleukin-17 can mediate protective innate immunity to pathogens or contribute to the pathogenesis of inflammatory diseases, such as psoriasis and rheumatoid arthritis. This review summarizes the basic biology of interleukin-17 and discusses its emerging role in periodontal disease. The current burden of evidence from human and animal model studies suggests that the net effect of interleukin-17 signaling promotes disease development. In addition to promoting neutrophilic inflammation, interleukin-17 has potent pro-osteoclastogenic effects that are likely to contribute to the pathogenesis of periodontitis, rheumatoid arthritis and other diseases involving bone immunopathology. Systemic treatments with anti-interleukin-17 biologics have shown promising results in clinical trials for psoriasis and rheumatoid arthritis; however, their impact on the highly prevalent periodontal disease has not been investigated or reported. Future clinical trials, preferably using locally administered interleukin-17 blockers, are required to implicate conclusivelyinterleukin-17 in periodontitis and, more importantly, to establish an effective adjunctive treatment for this oral inflammatory disease.
Collapse
|
45
|
Fasching P, Stradner M, Graninger W, Dejaco C, Fessler J. Therapeutic Potential of Targeting the Th17/Treg Axis in Autoimmune Disorders. Molecules 2017; 22:E134. [PMID: 28098832 PMCID: PMC6155880 DOI: 10.3390/molecules22010134] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 01/05/2017] [Accepted: 01/10/2017] [Indexed: 02/08/2023] Open
Abstract
A disruption of the crucial balance between regulatory T-cells (Tregs) and Th17-cells was recently implicated in various autoimmune disorders. Tregs are responsible for the maintenance of self-tolerance, thus inhibiting autoimmunity, whereas pro-inflammatory Th17-cells contribute to the induction and propagation of inflammation. Distortion of the Th17/Treg balance favoring the pro-inflammatory Th17 side is hence suspected to contribute to exacerbation of autoimmune disorders. This review aims to summarize recent data and advances in targeted therapeutic modification of the Th17/Treg-balance, as well as information on the efficacy of candidate therapeutics with respect to the treatment of autoimmune diseases.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Autoimmune Diseases/drug therapy
- Autoimmune Diseases/genetics
- Autoimmune Diseases/immunology
- Autoimmune Diseases/pathology
- Forkhead Transcription Factors/antagonists & inhibitors
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/immunology
- Gene Expression Regulation
- Humans
- Immunologic Factors/therapeutic use
- Inflammation
- Interleukin-17/antagonists & inhibitors
- Interleukin-17/genetics
- Interleukin-17/immunology
- Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/immunology
- Piperidines/therapeutic use
- Pyrimidines/therapeutic use
- Pyrroles/therapeutic use
- Signal Transduction
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Th17 Cells/pathology
- Ustekinumab/therapeutic use
Collapse
Affiliation(s)
- Patrizia Fasching
- Department of Rheumatology and Immunology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria.
| | - Martin Stradner
- Department of Rheumatology and Immunology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria.
| | - Winfried Graninger
- Department of Rheumatology and Immunology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria.
| | - Christian Dejaco
- Department of Rheumatology and Immunology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria.
| | - Johannes Fessler
- Department of Rheumatology and Immunology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria.
| |
Collapse
|
46
|
Thankam FG, Dilisio MF, Dougherty KA, Dietz NE, Agrawal DK. Triggering receptor expressed on myeloid cells and 5'adenosine monophosphate-activated protein kinase in the inflammatory response: a potential therapeutic target. Expert Rev Clin Immunol 2016; 12:1239-1249. [PMID: 27266327 PMCID: PMC5158012 DOI: 10.1080/1744666x.2016.1196138] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
INTRODUCTION The events in the cellular and molecular signaling triggered during inflammation mitigate tissue healing. The metabolic check-point control mediated by 5'-adenosine monophosphate-activated protein kinase (AMPK) is crucial for switching the cells into an activated state capable of mediating inflammatory events. The cell metabolism involved in the inflammatory response represents a potential therapeutic target for the pharmacologic management of inflammation. Areas covered: In this article, a critical review is presented on triggering receptor expressed on myeloid cell (TREM) receptors and their role in the inflammatory responses, as well as homeostasis between different TREM molecules and their regulation. Additionally, we discussed the relationship between TREM and AMPK to identify novel targets to limit the inflammatory response. Literature search was carried out from the National Library of Medicine's Medline database (using PubMed as the search engine) and Google Scholar and identified relevant studies up to 30 March 2016 using inflammation, TREM, AMPK, as the key words. Expert commentary: The prevention of phenotype switching of immune cells during inflammation by targeting AMPK and TREM-1 could be beneficial for developing novel management strategies for inflammation and associated complications.
Collapse
Affiliation(s)
- Finosh G Thankam
- Department of Clinical & Translational Science, Creighton University School of Medicine, Omaha, NE, USA
| | - Matthew F. Dilisio
- Department of Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, USA
| | | | - Nicholas E. Dietz
- Department of Pathology, Creighton University School of Medicine, Omaha, NE, USA
| | - Devendra K. Agrawal
- Department of Clinical & Translational Science, Creighton University School of Medicine, Omaha, NE, USA
| |
Collapse
|
47
|
Abboud A, Namas RA, Ramadan M, Mi Q, Almahmoud K, Abdul-Malak O, Azhar N, Zaaqoq A, Namas R, Barclay DA, Yin J, Sperry J, Peitzman A, Zamora R, Simmons RL, Billiar TR, Vodovotz Y. Computational Analysis Supports an Early, Type 17 Cell-Associated Divergence of Blunt Trauma Survival and Mortality. Crit Care Med 2016; 44:e1074-e1081. [PMID: 27513538 PMCID: PMC5201164 DOI: 10.1097/ccm.0000000000001951] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Blunt trauma patients may present with similar demographics and injury severity yet differ with regard to survival. We hypothesized that this divergence was due to different trajectories of systemic inflammation and utilized computational analyses to define these differences. DESIGN Retrospective clinical study and experimental study in mice. SETTING Level 1 trauma center and experimental laboratory. PATIENTS From a cohort of 493 victims of blunt trauma, we conducted a pairwise, retrospective, case-control study of patients who survived over 24 hours but ultimately died (nonsurvivors; n = 19) and patients who, after ICU admission, went on to be discharged(survivors; n = 19). INTERVENTIONS None in patients. Neutralizing anti-interleukin-17A antibody in mice. MEASUREMENTS AND MAIN RESULTS Data on systemic inflammatory mediators assessed within the first 24 hours and over 7 days were analyzed with computational modeling to infer dynamic networks of inflammation. Network density among inflammatory mediators in nonsurvivors increased in parallel with organ dysfunction scores over 7 days, suggesting the presence of early, self-sustaining, pathologic inflammation involving high-mobility group protein B1, interleukin-23, and the Th17 pathway. Survivors demonstrated a pattern commensurate with a self-resolving, predominantly lymphoid response, including higher levels of the reparative cytokine interleukin-22. Mice subjected to trauma/hemorrhage exhibited reduced organ damage when treated with anti-interleukin-17A. CONCLUSIONS Variable type 17 immune responses are hallmarks of organ damage, survival, and mortality after blunt trauma and suggest a lymphoid cell-based switch from self-resolving to self-sustaining inflammation.
Collapse
Affiliation(s)
- Andrew Abboud
- University of Pittsburgh, Department of Surgery, Pittsburgh, PA 15213
| | - Rami A. Namas
- University of Pittsburgh, Department of Surgery, Pittsburgh, PA 15213
| | - Mostafa Ramadan
- University of Pittsburgh, Department of Surgery, Pittsburgh, PA 15213
| | - Qi Mi
- University of Pittsburgh, Department of Mathematics, Pittsburgh, PA 15260
| | - Khalid Almahmoud
- University of Pittsburgh, Department of Surgery, Pittsburgh, PA 15213
| | | | - Nabil Azhar
- University of Pittsburgh, Department of Surgery, Pittsburgh, PA 15213
| | - Akram Zaaqoq
- University of Pittsburgh, Department of Critical Care Medicine, Pittsburgh, PA 15213
| | - Rajaie Namas
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, MI 48109
| | - Derek A. Barclay
- University of Pittsburgh, Department of Surgery, Pittsburgh, PA 15213
| | - Jinling Yin
- University of Pittsburgh, Department of Surgery, Pittsburgh, PA 15213
| | - Jason Sperry
- University of Pittsburgh, Department of Surgery, Pittsburgh, PA 15213
| | - Andrew Peitzman
- University of Pittsburgh, Department of Surgery, Pittsburgh, PA 15213
| | - Ruben Zamora
- University of Pittsburgh, Department of Surgery, Pittsburgh, PA 15213
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15219
| | | | | | - Yoram Vodovotz
- University of Pittsburgh, Department of Surgery, Pittsburgh, PA 15213
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15219
| |
Collapse
|
48
|
Banerjee S, Sindberg G, Wang F, Meng J, Sharma U, Zhang L, Dauer P, Chen C, Dalluge J, Johnson T, Roy S. Opioid-induced gut microbial disruption and bile dysregulation leads to gut barrier compromise and sustained systemic inflammation. Mucosal Immunol 2016; 9:1418-1428. [PMID: 26906406 PMCID: PMC4996771 DOI: 10.1038/mi.2016.9] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 12/30/2015] [Indexed: 02/04/2023]
Abstract
Morphine and its pharmacological derivatives are the most prescribed analgesics for moderate to severe pain management. However, chronic use of morphine reduces pathogen clearance and induces bacterial translocation across the gut barrier. The enteric microbiome has been shown to have a critical role in the preservation of the mucosal barrier function and metabolic homeostasis. Here, we show for the first time, using bacterial 16s rDNA sequencing, that chronic morphine treatment significantly alters the gut microbial composition and induces preferential expansion of Gram-positive pathogenic and reduction in bile-deconjugating bacterial strains. A significant reduction in both primary and secondary bile acid levels was seen in the gut, but not in the liver with morphine treatment. Morphine-induced microbial dysbiosis and gut barrier disruption was rescued by transplanting placebo-treated microbiota into morphine-treated animals, indicating that microbiome modulation could be exploited as a therapeutic strategy for patients using morphine for pain management.
Collapse
Affiliation(s)
- Santanu Banerjee
- Department of Surgery, 515 Delaware St SE, Moos 11-204, University
of Minnesota, MN 55455, USA
| | - Gregory Sindberg
- Department of Veterinary Medicine, 515 Delaware St SE, Moos 11-204,
University of Minnesota, MN 55455, USA
| | - Fuyuan Wang
- Department of Veterinary Medicine, 515 Delaware St SE, Moos 11-204,
University of Minnesota, MN 55455, USA
| | - Jingjing Meng
- Department of Surgery, 515 Delaware St SE, Moos 11-204, University
of Minnesota, MN 55455, USA
| | - Umakant Sharma
- Department of Surgery, 515 Delaware St SE, Moos 11-204, University
of Minnesota, MN 55455, USA
| | - Li Zhang
- Department of Pharmacology, 515 Delaware St SE, Moos 11-204,
University of Minnesota, MN 55455, USA
| | - Patricia Dauer
- Department of Pharmacology, 515 Delaware St SE, Moos 11-204,
University of Minnesota, MN 55455, USA
| | - Chi Chen
- Department of Food Science and Nutrition, 515 Delaware St SE, Moos
11-204, University of Minnesota, MN 55455, USA
| | - Joseph Dalluge
- Department of Chemistry, 515 Delaware St SE, Moos 11-204, University
of Minnesota, MN 55455, USA
| | - Timothy Johnson
- Department of Veterinary Medicine, 515 Delaware St SE, Moos 11-204,
University of Minnesota, MN 55455, USA
| | - Sabita Roy
- Department of Surgery, 515 Delaware St SE, Moos 11-204, University
of Minnesota, MN 55455, USA,Department of Pharmacology, 515 Delaware St SE, Moos 11-204,
University of Minnesota, MN 55455, USA,To whom correspondence should be addressed. Prof. Sabita
Roy, Director, Division of Infection, Inflammation and Vascular Biology,
Department of Surgery and Pharmacology Telephone Number: 612-624-4615, Fax
Number 612 626-4900,
| |
Collapse
|
49
|
The effects of konjac oligosaccharide on TNBS-induced colitis in rats. Int Immunopharmacol 2016; 40:385-391. [DOI: 10.1016/j.intimp.2016.08.040] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 08/28/2016] [Accepted: 08/31/2016] [Indexed: 12/25/2022]
|
50
|
Peloquin JM, Goel G, Kong L, Huang H, Haritunians T, Sartor RB, Daly MJ, Newberry RD, McGovern DP, Yajnik V, Lira SA, Xavier RJ. Characterization of candidate genes in inflammatory bowel disease-associated risk loci. JCI Insight 2016; 1:e87899. [PMID: 27668286 DOI: 10.1172/jci.insight.87899] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
GWAS have linked SNPs to risk of inflammatory bowel disease (IBD), but a systematic characterization of disease-associated genes has been lacking. Prior studies utilized microarrays that did not capture many genes encoded within risk loci or defined expression quantitative trait loci (eQTLs) using peripheral blood, which is not the target tissue in IBD. To address these gaps, we sought to characterize the expression of IBD-associated risk genes in disease-relevant tissues and in the setting of active IBD. Terminal ileal (TI) and colonic mucosal tissues were obtained from patients with Crohn's disease or ulcerative colitis and from healthy controls. We developed a NanoString code set to profile 678 genes within IBD risk loci. A subset of patients and controls were genotyped for IBD-associated risk SNPs. Analyses included differential expression and variance analysis, weighted gene coexpression network analysis, and eQTL analysis. We identified 116 genes that discriminate between healthy TI and colon samples and uncovered patterns in variance of gene expression that highlight heterogeneity of disease. We identified 107 coexpressed gene pairs for which transcriptional regulation is either conserved or reversed in an inflammation-independent or -dependent manner. We demonstrate that on average approximately 60% of disease-associated genes are differentially expressed in inflamed tissue. Last, we identified eQTLs with either genotype-only effects on expression or an interaction effect between genotype and inflammation. Our data reinforce tissue specificity of expression in disease-associated candidate genes, highlight genes and gene pairs that are regulated in disease-relevant tissue and inflammation, and provide a foundation to advance the understanding of IBD pathogenesis.
Collapse
Affiliation(s)
- Joanna M Peloquin
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease.,Center for Computational and Integrative Biology
| | - Gautam Goel
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease.,Center for Computational and Integrative Biology
| | - Lingjia Kong
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease.,Center for Computational and Integrative Biology
| | - Hailiang Huang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Talin Haritunians
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - R Balfour Sartor
- Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mark J Daly
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Rodney D Newberry
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Dermot P McGovern
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Vijay Yajnik
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease
| | - Sergio A Lira
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ramnik J Xavier
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease.,Center for Computational and Integrative Biology.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|