1
|
Paton H, Sarkar P, Gurung P. An overview of host immune responses against Leishmania spp. infections. Hum Mol Genet 2025:ddaf043. [PMID: 40287829 DOI: 10.1093/hmg/ddaf043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Leishmania spp. infections pose a significant global health challenge, affecting approximately 1 billion people across more than 88 endemic countries. This unicellular, obligate intracellular parasite causes a spectrum of diseases, ranging from localized cutaneous lesions to systemic visceral infections. Despite advancements in modern medicine and increased understanding of the parasite's etiology and associated diseases, treatment options remain limited to pentavalent antimonials, liposomal amphotericin B, and miltefosine. A deeper understanding of the interactions between immune and non-immune cells involved in the clearance of Leishmania spp. infections could uncover novel therapeutic strategies for this debilitating disease. This review highlights recent progress in elucidating how various cell types contribute to the regulation and resolution of Leishmania spp. infections.
Collapse
Affiliation(s)
- Hanna Paton
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
- Immunology Graduate Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
| | - Prabuddha Sarkar
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
| | - Prajwal Gurung
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
- Immunology Graduate Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Center for Immunology and Immune Based Disease, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Iowa City Veterans Affairs (VA) Medical Center, 601 US-6, Iowa City, IA 52246, United States
| |
Collapse
|
2
|
Herppich S, Hoenicke L, Kern F, Kruse F, Smout J, Greweling-Pils MC, Geffers R, Burton OT, Liston A, Keller A, Floess S, Huehn J. Zfp362 potentiates murine colonic inflammation by constraining Treg cell function rather than promoting Th17 cell differentiation. Eur J Immunol 2023; 53:e2250270. [PMID: 37366299 DOI: 10.1002/eji.202250270] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 05/02/2023] [Accepted: 06/09/2023] [Indexed: 06/28/2023]
Abstract
Mucosal barrier integrity and pathogen clearance is a complex process influenced by both Th17 and Treg cells. Previously, we had described the DNA methylation profile of Th17 cells and identified Zinc finger protein (Zfp)362 to be uniquely demethylated. Here, we generated Zfp362-/- mice to unravel the role of Zfp362 for Th17 cell biology. Zfp362-/- mice appeared clinically normal, showed no phenotypic alterations in the T-cell compartment, and upon colonization with segmented filamentous bacteria, no effect of Zfp362 deficiency on Th17 cell differentiation was observed. By contrast, Zfp362 deletion resulted in increased frequencies of colonic Foxp3+ Treg cells and IL-10+ and RORγt+ Treg cell subsets in mesenteric lymph nodes. Adoptive transfer of naïve CD4+ T cells from Zfp362-/- mice into Rag2-/- mice resulted in a significantly lower weight loss when compared with controls receiving cells from Zfp362+/+ littermates. However, this attenuated weight loss did not correlate with alterations of Th17 cells but instead was associated with an increase of effector Treg cells in mesenteric lymph nodes. Together, these results suggest that Zfp362 plays an important role in promoting colonic inflammation; however, this function is derived from constraining the effector function of Treg cells rather than directly promoting Th17 cell differentiation.
Collapse
Affiliation(s)
- Susanne Herppich
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Lisa Hoenicke
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Fabian Kern
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, Saarland University, Saarbrücken, Germany
- Department of Clinical Bioinformatics, Saarland University, Homburg, Germany
| | - Friederike Kruse
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Justine Smout
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Robert Geffers
- Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Oliver T Burton
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
| | - Adrian Liston
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
| | - Andreas Keller
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, Saarland University, Saarbrücken, Germany
- Department of Clinical Bioinformatics, Saarland University, Homburg, Germany
| | - Stefan Floess
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jochen Huehn
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
3
|
Rizvi ZA, Madan U, Tripathy MR, Goswami S, Mani S, Awasthi A, Dikshit M. Evaluation of Ayush-64 (a Polyherbal Formulation) and Its Ingredients in the Syrian Hamster Model for SARS-CoV-2 Infection Reveals the Preventative Potential of Alstonia scholaris. Pharmaceuticals (Basel) 2023; 16:1333. [PMID: 37765142 PMCID: PMC10534577 DOI: 10.3390/ph16091333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/16/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
In the current study, we evaluated the efficacy of Ayush-64 (A64), a polyherbal formulation containing Alstonia scholaris (L.) R. Br. (A. scholaris), Caesalpinia crista L. (C. crista), Picrorhiza kurroa Royle ex Benth (P. kurroa), and Swertia chirata (Roxb.) H. Karst. (S. chirata) against COVID-19 in a Syrian hamster infection model. Preventative use of A64 resulted in the late-phase recovery of body weight loss in severe acquired respiratory syndrome coronavirus-2 (SARS-CoV-2)-infected hamsters, suppression of pro-inflammatory cytokines, and blunted pulmonary pathology. In addition, we also investigated the efficacy of individual ingredients of A64, viz., A. scholaris, C. crista, P. kurroa, and S. chirata, in the hamster model. The hamster challenge data showed robust anti-viral and immunomodulatory potential in A. scholaris, followed by P. kurroa. However, C. crista and S. chirata of A64 showed prominent immunomodulatory potential without limiting the lung viral load. In order to better understand the immunomodulatory potential of these herbal extracts, we used an in vitro assay of helper T cell differentiation and found that A. scholaris mediated a more profound suppression of Th1, Th2, and Th17 cell differentiation as compared to A64 and other ingredients. Taken together, our animal study data identifies the ameliorative potential of A64 in mitigating coronavirus disease-19 (COVID-19) pulmonary pathology. A. scholaris, a constituent extract of A64, showed relatively higher anti-viral and immunomodulatory potential against COVID-19. The present study warrants further investigations to identify the active pharmaceutical ingredients of A. scholaris for further studies.
Collapse
Affiliation(s)
- Zaigham Abbas Rizvi
- Immuno-Biology Lab, Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute (THSTI), NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, India; (U.M.); (M.R.T.); (S.G.)
- Immunology-Core Lab, Translational Health Science and Technology Institute (THSTI), NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, India
| | - Upasna Madan
- Immuno-Biology Lab, Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute (THSTI), NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, India; (U.M.); (M.R.T.); (S.G.)
- Immunology-Core Lab, Translational Health Science and Technology Institute (THSTI), NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, India
| | - Manas Ranjan Tripathy
- Immuno-Biology Lab, Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute (THSTI), NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, India; (U.M.); (M.R.T.); (S.G.)
- Immunology-Core Lab, Translational Health Science and Technology Institute (THSTI), NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, India
| | - Sandeep Goswami
- Immuno-Biology Lab, Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute (THSTI), NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, India; (U.M.); (M.R.T.); (S.G.)
- Immunology-Core Lab, Translational Health Science and Technology Institute (THSTI), NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, India
| | - Shailendra Mani
- Non-Communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, India;
| | - Amit Awasthi
- Immuno-Biology Lab, Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute (THSTI), NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, India; (U.M.); (M.R.T.); (S.G.)
- Immunology-Core Lab, Translational Health Science and Technology Institute (THSTI), NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, India
| | - Madhu Dikshit
- Non-Communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, India;
- Pharmacology Department, CSIR-Central Drug Research Institute, Sitapur Rd., Sector 10, Jankipuram Extension, Lucknow 226031, India
| |
Collapse
|
4
|
Packialakshmi B, Hira S, Lund K, Zhang AH, Halterman J, Feng Y, Scott DW, Lees JR, Zhou X. NFAT5 contributes to the pathogenesis of experimental autoimmune encephalomyelitis (EAE) and decrease of T regulatory cells in female mice. Cell Immunol 2022; 375:104515. [DOI: 10.1016/j.cellimm.2022.104515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 11/03/2022]
|
5
|
Omokanye A, Ong LC, Lebrero-Fernandez C, Bernasconi V, Schön K, Strömberg A, Bemark M, Saelens X, Czarnewski P, Lycke N. Clonotypic analysis of protective influenza M2e-specific lung resident Th17 memory cells reveals extensive functional diversity. Mucosal Immunol 2022; 15:717-729. [PMID: 35260804 PMCID: PMC8903128 DOI: 10.1038/s41385-022-00497-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 01/31/2022] [Accepted: 02/06/2022] [Indexed: 02/06/2023]
Abstract
The fate of tissue-resident memory CD4 T cells (Trm) has been incompletely investigated. Here we show that intranasal, but not parenteral, immunization with CTA1-3M2e-DD stimulated M2e-specific Th17 Trm cells, which conferred strong protection against influenza virus infection in the lung. These cells rapidly expanded upon infection and effectively restricted virus replication as determined by CD4 T cell depletion studies. Single-cell RNAseq transcriptomic and TCR VDJ-analysis of M2e-tetramer-sorted CD4 T cells on day 3 and 8 post infection revealed complete Th17-lineage dominance (no Th1 or Tregs) with extensive functional diversity and expression of gene markers signifying mature resident Trm cells (Cd69, Nfkbid, Brd2, FosB). Unexpectedly, the same TCR clonotype hosted cells with different Th17 subcluster functions (IL-17, IL-22), regulatory and cytotoxic cells, suggesting a tissue and context-dependent differentiation of reactivated Th17 Trm cells. A gene set enrichment analysis demonstrated up-regulation of regulatory genes (Lag3, Tigit, Ctla4, Pdcd1) in M2e-specific Trm cells on day 8, indicating a tissue damage preventing function. Thus, contrary to current thinking, lung M2e-specific Th17 Trm cells are sufficient for controlling infection and for protecting against tissue injury. These findings will have strong implications for vaccine development against respiratory virus infections and influenza virus infections, in particular.
Collapse
Affiliation(s)
- Ajibola Omokanye
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Li Ching Ong
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Cristina Lebrero-Fernandez
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Valentina Bernasconi
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Karin Schön
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Anneli Strömberg
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Mats Bemark
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Xavier Saelens
- grid.5342.00000 0001 2069 7798VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium and Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Paulo Czarnewski
- grid.10548.380000 0004 1936 9377Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Nils Lycke
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
6
|
Brate AA, Boyden AW, Jensen IJ, Badovinac VP, Karandikar NJ. A Functionally Distinct CXCR3 +/IFN-γ +/IL-10 + Subset Defines Disease-Suppressive Myelin-Specific CD8 T Cells. THE JOURNAL OF IMMUNOLOGY 2021; 206:1151-1160. [PMID: 33558376 DOI: 10.4049/jimmunol.2001143] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/06/2021] [Indexed: 12/19/2022]
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the CNS. We have previously demonstrated that CNS-specific CD8 T cells possess a disease-suppressive function in MS and variations of its animal model, experimental autoimmune encephalomyelitis (EAE), including the highly clinically relevant relapsing-remitting EAE disease course. Regulatory CD8 T cell subsets have been identified in EAE and other autoimmune diseases, but studies vary in defining phenotypic properties of these cells. In relapsing-remitting EAE, PLP178-191 CD8 T cells suppress disease, whereas PLP139-151 CD8 T cells lack this function. In this study, we used this model to delineate the unique phenotypic properties of CNS-specific regulatory PLP178-191 CD8 T cells versus nonregulatory PLP139-151 or OVA323-339 CD8 T cells. Using multiparametric flow cytometric analyses of phenotypic marker expression, we identified a CXCR3+ subpopulation among activated regulatory CD8 T cells, relative to nonregulatory counterparts. This subset exhibited increased degranulation and IFN-γ and IL-10 coproduction. A similar subset was also identified in C57BL/6 mice within autoregulatory PLP178-191 CD8 T cells but not within nonregulatory OVA323-339 CD8 T cells. This disease-suppressing CD8 T cell subpopulation provides better insights into functional regulatory mechanisms, and targeted enhancement of this subset could represent a novel immunotherapeutic approach for MS.
Collapse
Affiliation(s)
- Ashley A Brate
- Department of Pathology, University of Iowa Health Care, Iowa City, IA 52241.,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52241.,Iowa City Veterans Affairs Medical Center, Iowa City, IA 52241; and
| | - Alexander W Boyden
- Department of Pathology, University of Iowa Health Care, Iowa City, IA 52241.,Iowa City Veterans Affairs Medical Center, Iowa City, IA 52241; and
| | - Isaac J Jensen
- Department of Pathology, University of Iowa Health Care, Iowa City, IA 52241.,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52241
| | - Vladimir P Badovinac
- Department of Pathology, University of Iowa Health Care, Iowa City, IA 52241.,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52241.,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52241
| | - Nitin J Karandikar
- Department of Pathology, University of Iowa Health Care, Iowa City, IA 52241; .,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52241.,Iowa City Veterans Affairs Medical Center, Iowa City, IA 52241; and
| |
Collapse
|
7
|
Zheng Y, Verhoeff TA, Perez Pardo P, Garssen J, Kraneveld AD. The Gut-Brain Axis in Autism Spectrum Disorder: A Focus on the Metalloproteases ADAM10 and ADAM17. Int J Mol Sci 2020; 22:ijms22010118. [PMID: 33374371 PMCID: PMC7796333 DOI: 10.3390/ijms22010118] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022] Open
Abstract
Autism Spectrum Disorder (ASD) is a spectrum of disorders that are characterized by problems in social interaction and repetitive behavior. The disease is thought to develop from changes in brain development at an early age, although the exact mechanisms are not known yet. In addition, a significant number of people with ASD develop problems in the intestinal tract. A Disintegrin And Metalloproteases (ADAMs) include a group of enzymes that are able to cleave membrane-bound proteins. ADAM10 and ADAM17 are two members of this family that are able to cleave protein substrates involved in ASD pathogenesis, such as specific proteins important for synapse formation, axon signaling and neuroinflammation. All these pathological mechanisms are involved in ASD. Besides the brain, ADAM10 and ADAM17 are also highly expressed in the intestines. ADAM10 and ADAM17 have implications in pathways that regulate gut permeability, homeostasis and inflammation. These metalloproteases might be involved in microbiota-gut-brain axis interactions in ASD through the regulation of immune and inflammatory responses in the intestinal tract. In this review, the potential roles of ADAM10 and ADAM17 in the pathology of ASD and as targets for new therapies will be discussed, with a focus on the gut-brain axis.
Collapse
Affiliation(s)
- Yuanpeng Zheng
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584CG Utrecht, The Netherlands; (Y.Z.); (T.A.V.); (P.P.P.); (J.G.)
| | - Tessa A. Verhoeff
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584CG Utrecht, The Netherlands; (Y.Z.); (T.A.V.); (P.P.P.); (J.G.)
| | - Paula Perez Pardo
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584CG Utrecht, The Netherlands; (Y.Z.); (T.A.V.); (P.P.P.); (J.G.)
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584CG Utrecht, The Netherlands; (Y.Z.); (T.A.V.); (P.P.P.); (J.G.)
- Global Centre of Excellence Immunology, Danone Nutricia Research B.V., 3584CT Utrecht, The Netherlands
| | - Aletta D. Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584CG Utrecht, The Netherlands; (Y.Z.); (T.A.V.); (P.P.P.); (J.G.)
- Correspondence: ; Tel.: +31-(0)3-02534509
| |
Collapse
|
8
|
Wadhwa R, Pandey P, Gupta G, Aggarwal T, Kumar N, Mehta M, Satija S, Gulati M, Madan JR, Dureja H, Balusamy SR, Perumalsamy H, Maurya PK, Collet T, Tambuwala MM, Hansbro PM, Chellappan DK, Dua K. Emerging Complexity and the Need for Advanced Drug Delivery in Targeting Candida Species. Curr Top Med Chem 2019; 19:2593-2609. [DOI: 10.2174/1568026619666191026105308] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 07/15/2019] [Accepted: 09/07/2019] [Indexed: 02/07/2023]
Abstract
Background:Candida species are the important etiologic agents for candidiasis, the most prevalent cause of opportunistic fungal infections. Candida invasion results in mucosal to systemic infections through immune dysfunction and helps in further invasion and proliferation at several sites in the host. The host defence system utilizes a wide array of the cells, proteins and chemical signals that are distributed in blood and tissues which further constitute the innate and adaptive immune system. The lack of antifungal agents and their limited therapeutic effects have led to high mortality and morbidity related to such infections.Methods:The necessary information collated on this review has been gathered from various literature published from 1995 to 2019.Results:This article sheds light on novel drug delivery approaches to target the immunological axis for several Candida species (C. albicans, C. glabrata, C. parapsilosis, C. tropicalis, C. krusei, C. rugose, C. hemulonii, etc.).Conclusion:It is clear that the novel drug delivery approaches include vaccines, adoptive transfer of primed immune cells, recombinant cytokines, therapeutic antibodies, and nanoparticles, which have immunomodulatory effects. Such advancements in targeting various underpinning mechanisms using the concept of novel drug delivery will provide a new dimension to the fungal infection clinic particularly due to Candida species with improved patient compliance and lesser side effects. This advancement in knowledge can also be extended to target various other similar microbial species and infections.
Collapse
Affiliation(s)
- Ridhima Wadhwa
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Parijat Pandey
- Shri Baba Mastnath Institute of Pharmaceutical Sciences and Research, Baba Mastnath University, Rohtak 124001, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura 302 017, Jaipur, India
| | - Taru Aggarwal
- Amity Institute of Biotechnology, Amity University, Noida 201303, India
| | - Nitesh Kumar
- Amity Institute for Advanced Research & Studies (M&D), Amity University, Noida 201303, India
| | - Meenu Mehta
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar, Delhi G.T. Road (NH-1), Phagwara-144411, Punjab, India
| | - Saurabh Satija
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar, Delhi G.T. Road (NH-1), Phagwara-144411, Punjab, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar, Delhi G.T. Road (NH-1), Phagwara-144411, Punjab, India
| | - Jyotsna R. Madan
- Department of Pharmaceutics, Smt. Kashibai Navale College of Pharmacy, Kondhwa, Pune, 411048, Maharashtra, India
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharishi Dayanand University, Rohtak, Haryana 124001, India
| | - Sri R. Balusamy
- Department of Food Science and Biotechnology, Sejong University, Gwangjin-gu, Seoul, 05006, Korea
| | - Haribalan Perumalsamy
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin, 446-701, Korea
| | - Pawan K. Maurya
- Department of Biochemistry, Central University of Haryana, Jant-Pali, Mahendergarh District 123031, Haryana, India
| | - Trudi Collet
- Innovative Medicines Group, Institute of Health & Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Brisbane, Queensland 4059, Australia
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County Londonderry, BT52 1SA, Northern Ireland, United Kingdom
| | - Philip M. Hansbro
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Kamal Dua
- School of Pharmaceutical Sciences, Shoolini University, Bajhol, Sultanpur, Solan, Himachal Pradesh 173 229, Australia
| |
Collapse
|
9
|
Yang YX, Shen HH, Cao F, Xie LY, Zhu GL, Sam NB, Wang DG, Pan HF. Therapeutic potential of enhancer of zeste homolog 2 in autoimmune diseases. Expert Opin Ther Targets 2019; 23:1015-1030. [PMID: 31747802 DOI: 10.1080/14728222.2019.1696309] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: Autoimmune diseases (ADs) are idiopathic and heterogeneous disorders with contentious pathophysiology. Great strides have been made in epigenetics and its involvement in ADs. Zeste homolog 2 (EZH2) has sparked extensive interest because of its pleiotropic roles in distinct pathologic contexts.Areas covered: This review summarizes the epigenetic functions and the biological significance of EZH2 in the etiology of rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), type 1 diabetes (T1D), inflammatory bowel disease (IBD), multiple sclerosis (MS), and systemic sclerosis (SSc). A brief recapitulation of the therapeutic potential of EZH2 targeting is provided.Expert opinion: There are questions marks and controversies surrounding the feasibility and safety of EZH2 targeting; it is recommended in RA and SLE, but queried in T1D, IBD, MS, and SSc. Future work should focus on contrast studies, systematic analyses and preclinical studies with optimizing methodologies. Selective research studies conducted in a stage-dependent manner are necessary because of the relapsing-remitting clinical paradigms.
Collapse
Affiliation(s)
- Yue-Xin Yang
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hui-Hui Shen
- Department of Clinical Medicine, The second School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Fan Cao
- Department of Clinical Medicine, The second School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Liang-Yu Xie
- Department of Clinical Medicine, The second School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Guang-Lin Zhu
- Department of Clinical Medicine, The second School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Napoleon Bellua Sam
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, China
| | - De-Guang Wang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, China
| |
Collapse
|
10
|
Daryabor G, Kabelitz D, Kalantar K. An update on immune dysregulation in obesity-related insulin resistance. Scand J Immunol 2019; 89:e12747. [PMID: 30593678 DOI: 10.1111/sji.12747] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/22/2018] [Accepted: 12/25/2018] [Indexed: 12/29/2022]
Abstract
Obesity is associated with chronic low-grade inflammation of the adipose tissue (AT) that might develop into systemic inflammation, insulin resistance (IR) and an increased risk of type 2 diabetes mellitus (T2DM) in severe obese rodents and humans. In the lean state, small normal adipocytes and AT macrophages interact with each other to maintain metabolic homeostasis but during obesity, enlarged adipocytes secrete inflammatory mediators and express immune receptors to recruit immune cells and aggravate the inflammation. The better understanding of the obesity-related inflammatory milieu and the sequential events leading to IR could be helpful in designing new preventive and therapeutic strategies. The present review will discuss the cellular and molecular abnormalities participating in the pathogenesis of obesity in obese individuals as well as high-fat diet (HFD)-fed mice, a mouse model of obesity.
Collapse
Affiliation(s)
- Gholamreza Daryabor
- Department of Immunology, Shiraz Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Kurosh Kalantar
- Department of Immunology, Shiraz Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
11
|
Huang XT, Wang B, Zhang WH, Peng MQ, Lin D. Total glucosides of paeony suppresses experimental autoimmune uveitis in association with inhibition of Th1 and Th2 cell function in mice. Int J Immunopathol Pharmacol 2018; 32:394632017751547. [PMID: 29363368 PMCID: PMC5849247 DOI: 10.1177/0394632017751547] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Total glucosides of paeony (TGP) are active components extracted from the roots
of Paeonia lactiflora Pall. In this study, we
investigated the role and mechanisms of TGP in experimental autoimmune uveitis
(EAU) model of mice. The C57BL/6 mice were randomly divided into three groups:
sham group, EAU-control group, and EAU-TGP group. Clinical score of images of
the eye fundus were taken on 7, 14, 21, and 28 days after induction of EAU. The
concentrations of proinflammatory cytokines in intraocular fluid were measured
at 14 days after EAU induction with the use of a multiplex assay system. Flow
cytometry was used to analyze the frequency of CD4+, CD8+, interferon-gamma
(IFN-γ), and CD4+/CD8+ ratio in spleen and lymph nodes. Western blotting was
used to measure expressions of mitogen-activated protein kinase (MAPK)
pathway-related proteins in retina. Clinical scores for uveitis were lower in
TGP-treated EAU mice than those without TGP treatment. Importantly, the
concentrations of cytokines induced by T-helper 1 (Th1) and T-helper 2 (Th2)
cells in intraocular fluid were reduced in EAU mice treated with TGP.
Furthermore, the frequency of CD4+, IFN-γ, and CD4+/CD8+ ratio was decreased and
the frequency of CD8+ was increased in spleen and lymph nodes of mice treated
with TGP. The anti-inflammatory effects of TGP were mediated by inhibiting the
MAPK signaling pathways. Our results showed that TGP suppressed uveitis in mice
via the inhibition of Th1 and Th2 cell function. Thus, TGP may be a promising
therapeutic strategy for uveitis, as well as other ocular inflammatory
diseases.
Collapse
Affiliation(s)
- Xue-Tao Huang
- 1 Department of Ophthalmology, Changsha Aier Eye Hospital, Aier School of Ophthalmology, Central South University, Changsha, China.,2 Department of Ophthalmology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Bin Wang
- 3 Department of Dermatology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Wen-Hua Zhang
- 4 Department of Ophthalmology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Man-Qiang Peng
- 1 Department of Ophthalmology, Changsha Aier Eye Hospital, Aier School of Ophthalmology, Central South University, Changsha, China
| | - Ding Lin
- 1 Department of Ophthalmology, Changsha Aier Eye Hospital, Aier School of Ophthalmology, Central South University, Changsha, China
| |
Collapse
|
12
|
Chung BH, Yang CW, Cho ML. Clinical significance of Th17 cells in kidney transplantation. Korean J Intern Med 2018; 33:860-866. [PMID: 29843491 PMCID: PMC6129639 DOI: 10.3904/kjim.2018.095] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022] Open
Abstract
Transplantation research has focused on cytotoxic T-cell and plasma cell/B-cell-targeted strategies, but little attention has been paid to the role of T helper 17 (Th17) cells in allograft dysfunction. However, accumulating evidence suggests that Th17 cells contribute to the development of acute and chronic allograft injury after transplantation of various organs, including the kidney. This review summarizes recent reports on the role of Th17 cells in kidney transplantation. Means of improving allograft outcomes by targeting the Th17 pathway are also suggested.
Collapse
Affiliation(s)
- Byung Ha Chung
- Convergent Research Consortium for Immunologic Disease, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
- Transplant Research Center, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Chul Woo Yang
- Convergent Research Consortium for Immunologic Disease, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
- Transplant Research Center, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Mi-La Cho
- Convergent Research Consortium for Immunologic Disease, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
- Correspondence to Mi-La Cho, Ph.D. Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea Tel: +82-2-2258-7467 Fax: +82-2-599-4287 E-mail:
| |
Collapse
|
13
|
Lawrence SM, Wynn JL. Chorioamnionitis, IL-17A, and fetal origins of neurologic disease. Am J Reprod Immunol 2017; 79:e12803. [PMID: 29271527 DOI: 10.1111/aji.12803] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 11/28/2017] [Indexed: 12/23/2022] Open
Abstract
The Centers for Disease Control and Prevention estimate that 1 in 323 infants have cerebral palsy. Highly correlated to intrauterine infection and inflammation, the incidence of cerebral palsy has remained constant over the last few decades despite significant advances in neonatal intensive care including improved ventilator techniques, surfactant therapy, maternal steroid administration, and use of intrapartum empiric antimicrobials. Recent advances in our understanding of immune responses to infection and inflammation have identified the cytokine IL-17A as a crucial component of early proinflammatory mediators that cause brain injury associated with neurologic impairment. Remarkably, maternal inflammatory responses to in utero inflammation and infection can also lead to potentially debilitating neurologic conditions in the offspring, which often become clinically apparent during childhood and/or early adulthood. This review details the role of IL-17A in fetal and maternal proinflammatory responses that lead to fetal brain injury and neurologic sequelae, including cerebral palsy. Recent findings regarding the role of maternal inflammatory responses in the development of childhood and adult neurologic conditions, such as autism, schizophrenia, and multiple sclerosis, will also be highlighted.
Collapse
Affiliation(s)
- Shelley M Lawrence
- College of Medicine, Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of California, San Diego, CA, USA.,Department of Pediatrics, Division of Host-Microbe Systems and Therapeutics, University of California, San Diego, CA, USA
| | - James L Wynn
- College of Medicine, Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Florida, Gainesville, FL, USA.,Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
14
|
Tibbitt C, Falconer J, Stoop J, van Eden W, Robinson JH, Hilkens CMU. Reduced TCR-dependent activation through citrullination of a T-cell epitope enhances Th17 development by disruption of the STAT3/5 balance. Eur J Immunol 2017; 46:1633-43. [PMID: 27173727 PMCID: PMC4949576 DOI: 10.1002/eji.201546217] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 03/22/2016] [Accepted: 05/09/2016] [Indexed: 01/10/2023]
Abstract
Citrullination is a post‐translational modification of arginine that commonly occurs in inflammatory tissues. Because T‐cell receptor (TCR) signal quantity and quality can regulate T‐cell differentiation, citrullination within a T‐cell epitope has potential implications for T‐cell effector function. Here, we investigated how citrullination of an immunedominant T‐cell epitope affected Th17 development. Murine naïve CD4+ T cells with a transgenic TCR recognising p89‐103 of the G1 domain of aggrecan (agg) were co‐cultured with syngeneic bone marrow‐derived dendritic cells (BMDC) presenting the native or citrullinated peptides. In the presence of pro‐Th17 cytokines, the peptide citrullinated on residue 93 (R93Cit) significantly enhanced Th17 development whilst impairing the Th2 response, compared to the native peptide. T cells responding to R93Cit produced less IL‐2, expressed lower levels of the IL‐2 receptor subunit CD25, and showed reduced STAT5 phosphorylation, whilst STAT3 activation was unaltered. IL‐2 blockade in native p89‐103‐primed T cells enhanced the phosphorylated STAT3/STAT5 ratio, and concomitantly enhanced Th17 development. Our data illustrate how a post‐translational modification of a TCR contact point may promote Th17 development by altering the balance between STAT5 and STAT3 activation in responding T cells, and provide new insight into how protein citrullination may influence effector Th‐cell development in inflammatory disorders.
Collapse
Affiliation(s)
- Christopher Tibbitt
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, U.K
| | - Jane Falconer
- Rheumatology Research Group, School of Immunity and Infection, University of Birmingham, U.K.,Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE), U.K
| | - Jeroen Stoop
- Department of Rheumatology, Leiden University, The Netherlands
| | - Willem van Eden
- Institute of Infectious Diseases and Immunology, Utrecht University, The Netherlands
| | - John H Robinson
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, U.K
| | - Catharien M U Hilkens
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, U.K.,Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE), U.K
| |
Collapse
|
15
|
Zhou X, Packialakshmi B, Xiao Y, Nurmukhambetova S, Lees JR. Progression of experimental autoimmune encephalomyelitis is associated with up-regulation of major sodium transporters in the mouse kidney cortex under a normal salt diet. Cell Immunol 2017; 317:18-25. [PMID: 28438314 DOI: 10.1016/j.cellimm.2017.04.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 04/13/2017] [Accepted: 04/13/2017] [Indexed: 01/11/2023]
Abstract
Recent demonstrations of exacerbation of experimental autoimmune encephalomyelitis (EAE) by high salt diets prompted us to study whether EAE stimulated Na absorption by the renal cortex, a primary regulatory site for Na balance, even under a normal NaCl diet. We found that as EAE progressed from mild to severe symptoms, there were parallel increases in the protein abundance of NHE3 and αENaC and the Na,K-ATPase activity with an affiliated elevation of its β1-subunit protein. These effects are associated with increases in the protein levels of the well-known regulators SGK1 and scaffold NHERF2, and phosphorylation of ERK1/2. These effects of EAE could not be explained by reduction in water or food intake. We conclude that EAE progression is associated with up-regulation of major Na transporters, which is most likely driven by increased expression of SGK1 and NHERF2 and activation of ERK1/2. These data suggest that EAE progression increases Na absorption by the renal cortex.
Collapse
Affiliation(s)
- Xiaoming Zhou
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.
| | - Balamurugan Packialakshmi
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Yao Xiao
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Saule Nurmukhambetova
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Jason R Lees
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
16
|
Karantanos T, Chistofides A, Barhdan K, Li L, Boussiotis VA. Regulation of T Cell Differentiation and Function by EZH2. Front Immunol 2016; 7:172. [PMID: 27199994 PMCID: PMC4853381 DOI: 10.3389/fimmu.2016.00172] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 04/18/2016] [Indexed: 12/17/2022] Open
Abstract
The enhancer of zeste homolog 2 (EZH2), one of the polycomb-group proteins, is the catalytic subunit of Polycomb-repressive complex 2 (PRC2) and induces the trimethylation of the histone H3 lysine 27 (H3K27me3) promoting epigenetic gene silencing. EZH2 contains a SET domain promoting the methyltransferase activity, while the three other protein components of PRC2, namely EED, SUZ12, and RpAp46/48, induce compaction of the chromatin permitting EZH2 enzymatic activity. Numerous studies highlight the role of this evolutionary conserved protein as a master regulator of differentiation in humans involved in the repression of the homeotic gene and the inactivation of X-chromosome. Through its effects in the epigenetic regulation of critical genes, EZH2 has been strongly linked to cell cycle progression, stem cell pluripotency, and cancer biology, being currently at the cutting edge of research. Most recently, EZH2 has been associated with hematopoietic stem cell proliferation and differentiation, thymopoiesis and lymphopoiesis. Several studies have evaluated the role of EZH2 in the regulation of T cell differentiation and plasticity as well as its implications in the development of autoimmune diseases and graft-versus-host disease (GVHD). The aim of this review is to summarize the current knowledge regarding the role of EZH2 in the regulation of the differentiation and function of T cells focusing on possible applications in various immune-mediated conditions, including autoimmune disorders and GVHD.
Collapse
Affiliation(s)
- Theodoros Karantanos
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; General Internal Medicine Section, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA
| | - Anthos Chistofides
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kankana Barhdan
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Lequn Li
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Vassiliki A Boussiotis
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Barros-Martins J, Schmolka N, Fontinha D, Pires de Miranda M, Simas JP, Brok I, Ferreira C, Veldhoen M, Silva-Santos B, Serre K. Effector γδ T Cell Differentiation Relies on Master but Not Auxiliary Th Cell Transcription Factors. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 196:3642-52. [PMID: 26994218 DOI: 10.4049/jimmunol.1501921] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 02/23/2016] [Indexed: 11/19/2022]
Abstract
γδ T lymphocytes are programmed into distinct IFN-γ-producing CD27(+) (γδ27(+)) and IL-17-producing CD27(-) (γδ27(-)) subsets that play key roles in protective or pathogenic immune responses. Although the signature cytokines are shared with their αβ Th1 (for γδ27(+)) and Th17 (for γδ27(-)) cell counterparts, we dissect in this study similarities and differences in the transcriptional requirements of murine effector γδ27(+), γδ27(-)CCR6(-), and γδ27(-)CCR6(+) γδ T cell subsets and αβ T cells. We found they share dependence on the master transcription factors T-bet and RORγt for IFN-γ and IL-17 production, respectively. However, Eomes is fully dispensable for IFN-γ production by γδ T cells. Furthermore, the Th17 cell auxiliary transcription factors RORα and BATF are not required for IL-17 production by γδ27(-) cell subsets. We also show that γδ27(-) (but not γδ27(+)) cells become polyfunctional upon IL-1β plus IL-23 stimulation, cosecreting IL-17A, IL-17F, IL-22, GM-CSF, and IFN-γ. Collectively, our in vitro and in vivo data firmly establish the molecular segregation between γδ27(+) and γδ27(-) T cell subsets and provide novel insight on the nonoverlapping transcriptional networks that control the differentiation of effector γδ versus αβ T cell subsets.
Collapse
Affiliation(s)
- Joana Barros-Martins
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Nina Schmolka
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Diana Fontinha
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Marta Pires de Miranda
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - J Pedro Simas
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Ingrid Brok
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Cristina Ferreira
- Laboratory for Lymphocyte Signalling and Development, Babraham Institute, Cambridge CB22 3AT, United Kingdom; and
| | - Marc Veldhoen
- Laboratory for Lymphocyte Signalling and Development, Babraham Institute, Cambridge CB22 3AT, United Kingdom; and
| | - Bruno Silva-Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto Gulbenkian de Ciência, 2781-901 Oeiras, Portugal
| | - Karine Serre
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal;
| |
Collapse
|
18
|
Choi GB, Yim YS, Wong H, Kim S, Kim H, Kim SV, Hoeffer CA, Littman DR, Huh JR. The maternal interleukin-17a pathway in mice promotes autism-like phenotypes in offspring. Science 2016; 351:933-9. [PMID: 26822608 PMCID: PMC4782964 DOI: 10.1126/science.aad0314] [Citation(s) in RCA: 840] [Impact Index Per Article: 93.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 01/14/2016] [Indexed: 12/13/2022]
Abstract
Viral infection during pregnancy has been correlated with increased frequency of autism spectrum disorder (ASD) in offspring. This observation has been modeled in rodents subjected to maternal immune activation (MIA). The immune cell populations critical in the MIA model have not been identified. Using both genetic mutants and blocking antibodies in mice, we show that retinoic acid receptor-related orphan nuclear receptor gamma t (RORγt)-dependent effector T lymphocytes [for example, T helper 17 (TH17) cells] and the effector cytokine interleukin-17a (IL-17a) are required in mothers for MIA-induced behavioral abnormalities in offspring. We find that MIA induces an abnormal cortical phenotype, which is also dependent on maternal IL-17a, in the fetal brain. Our data suggest that therapeutic targeting of TH17 cells in susceptible pregnant mothers may reduce the likelihood of bearing children with inflammation-induced ASD-like phenotypes.
Collapse
MESH Headings
- Animals
- Antibodies, Blocking/immunology
- Antibodies, Blocking/therapeutic use
- Autism Spectrum Disorder/genetics
- Autism Spectrum Disorder/immunology
- Autism Spectrum Disorder/prevention & control
- Behavior, Animal
- Behavioral Symptoms/immunology
- Cerebral Cortex/abnormalities
- Cerebral Cortex/drug effects
- Cerebral Cortex/immunology
- Female
- Interleukin-17/biosynthesis
- Interleukin-17/immunology
- Interleukin-17/pharmacology
- Male
- Maternal-Fetal Exchange/immunology
- Mice
- Mutation
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/immunology
- Phenotype
- Pregnancy
- Prenatal Exposure Delayed Effects/immunology
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Receptors, Retinoic Acid/genetics
- Receptors, Retinoic Acid/immunology
- Signal Transduction
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Retinoic Acid Receptor gamma
Collapse
Affiliation(s)
- Gloria B Choi
- The McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yeong S Yim
- The McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Helen Wong
- Center for Neural Science, New York University, New York, NY 10003, USA. Institute for Behavioral Genetics, Department of Integrated Physiology, University of Colorado, Boulder, CO 80303, USA
| | - Sangdoo Kim
- Division of Infectious Diseases and Immunology and Program in Innate Immunity, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Hyunju Kim
- Division of Infectious Diseases and Immunology and Program in Innate Immunity, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Sangwon V Kim
- The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Charles A Hoeffer
- Center for Neural Science, New York University, New York, NY 10003, USA. Institute for Behavioral Genetics, Department of Integrated Physiology, University of Colorado, Boulder, CO 80303, USA.
| | - Dan R Littman
- The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA. Howard Hughes Medical Institute, New York, NY 10016, USA.
| | - Jun R Huh
- Division of Infectious Diseases and Immunology and Program in Innate Immunity, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA. The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
19
|
Evonuk KS, Baker BJ, Doyle RE, Moseley CE, Sestero CM, Johnston BP, De Sarno P, Tang A, Gembitsky I, Hewett SJ, Weaver CT, Raman C, DeSilva TM. Inhibition of System Xc(-) Transporter Attenuates Autoimmune Inflammatory Demyelination. THE JOURNAL OF IMMUNOLOGY 2015; 195:450-463. [PMID: 26071560 DOI: 10.4049/jimmunol.1401108] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 05/11/2015] [Indexed: 01/15/2023]
Abstract
T cell infiltration into the CNS is a significant underlying pathogenesis in autoimmune inflammatory demyelinating diseases. Several lines of evidence suggest that glutamate dysregulation in the CNS is an important consequence of immune cell infiltration in neuroinflammatory demyelinating diseases; yet, the causal link between inflammation and glutamate dysregulation is not well understood. A major source of glutamate release during oxidative stress is the system Xc(-) transporter; however, this mechanism has not been tested in animal models of autoimmune inflammatory demyelination. We find that pharmacological and genetic inhibition of system Xc(-) attenuates chronic and relapsing-remitting experimental autoimmune encephalomyelitis (EAE). Remarkably, pharmacological blockade of system Xc(-) 7 d after induction of EAE attenuated T cell infiltration into the CNS, but not T cell activation in the periphery. Mice harboring a Slc7a11 (xCT) mutation that inactivated system Xc(-) were resistant to EAE, corroborating a central role for system Xc(-) in mediating immune cell infiltration. We next examined the role of the system Xc(-) transporter in the CNS after immune cell infiltration. Pharmacological inhibitors of the system Xc(-) transporter administered during the first relapse in a SJL animal model of relapsing-remitting EAE abrogated clinical disease, inflammation, and myelin loss. Primary coculture studies demonstrate that myelin-specific CD4(+) Th1 cells provoke microglia to release glutamate via the system Xc(-) transporter, causing excitotoxic death to mature myelin-producing oligodendrocytes. Taken together, these studies support a novel role for the system Xc(-) transporter in mediating T cell infiltration into the CNS as well as promoting myelin destruction after immune cell infiltration in EAE.
Collapse
Affiliation(s)
- Kirsten S Evonuk
- Center for Glial Biology in Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294.,Department of Physical Medicine Rehabilitation, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Brandi J Baker
- Center for Glial Biology in Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294.,Department of Physical Medicine Rehabilitation, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Ryan E Doyle
- Center for Glial Biology in Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294.,Department of Physical Medicine Rehabilitation, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Carson E Moseley
- Department of Pathology, University of Alabama at Birmingham, AL, 35294
| | - Christine M Sestero
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294.,Department of Biology, Chemistry, and Mathematics, University of Montevallo, Montevallo, AL 35115
| | - Bryce P Johnston
- Center for Glial Biology in Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294.,Department of Physical Medicine Rehabilitation, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Patrizia De Sarno
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Andrew Tang
- Department of Physical Medicine Rehabilitation, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Igor Gembitsky
- Department of Physical Medicine Rehabilitation, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Sandra J Hewett
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, NY, 13244
| | - Casey T Weaver
- Department of Pathology, University of Alabama at Birmingham, AL, 35294
| | - Chander Raman
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Tara M DeSilva
- Center for Glial Biology in Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294.,Department of Physical Medicine Rehabilitation, University of Alabama at Birmingham, Birmingham, AL, 35294.,Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294
| |
Collapse
|
20
|
Wu C, Goodall JC, Busch R, Gaston JSH. Relationship of CD146 expression to secretion of interleukin (IL)-17, IL-22 and interferon-γ by CD4(+) T cells in patients with inflammatory arthritis. Clin Exp Immunol 2015; 179:378-91. [PMID: 25113810 PMCID: PMC4337671 DOI: 10.1111/cei.12434] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2014] [Indexed: 12/14/2022] Open
Abstract
Expression of the adhesion molecule, CD146/MCAM/MelCAM, on T cells has been associated with recent activation, memory subsets and T helper type 17 (Th17) effector function, and is elevated in inflammatory arthritis. Th17 cells have been implicated in the pathogenesis of rheumatoid arthritis (RA) and spondyloarthritides (SpA). Here, we compared the expression of CD146 on CD4(+) T cells between healthy donors (HD) and patients with RA and SpA [ankylosing spondylitis (AS) or psoriatic arthritis (PsA)] and examined correlations with surface markers and cytokine secretion. Peripheral blood mononuclear cells (PBMC) were obtained from patients and controls, and synovial fluid mononuclear cells (SFMC) from patients. Cytokine production [elicited by phorbol myristate acetate (PMA)/ionomycin] and surface phenotypes were evaluated by flow cytometry. CD146(+) CD4(+) and interleukin (IL)-17(+) CD4(+) T cell frequencies were increased in PBMC of PsA patients, compared with HD, and in SFMC compared with PBMC. CD146(+) CD4(+) T cells were enriched for secretion of IL-17 [alone or with IL-22 or interferon (IFN)-γ] and for some putative Th17-associated surface markers (CD161 and CCR6), but not others (CD26 and IL-23 receptor). CD4(+) T cells producing IL-22 or IFN-γ without IL-17 were also present in the CD146(+) subset, although their enrichment was less marked. Moreover, a majority of cells secreting these cytokines lacked CD146. Thus, CD146 is not a sensitive or specific marker of Th17 cells, but rather correlates with heterogeneous cytokine secretion by subsets of CD4(+) helper T cells.
Collapse
Affiliation(s)
- C Wu
- Department of Medicine, University of Cambridge, Cambridge, UK; Department of Rheumatology and Immunology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | | | | | | |
Collapse
|
21
|
Jin Y, Wan Y, Chen G, Chen L, Zhang MQ, Deng L, Zhang JC, Xiong XZ, Xin JB. Treg/IL-17 ratio and Treg differentiation in patients with COPD. PLoS One 2014; 9:e111044. [PMID: 25329073 PMCID: PMC4199736 DOI: 10.1371/journal.pone.0111044] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 09/28/2014] [Indexed: 12/02/2022] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is characterized by chronic pulmonary and systematic inflammation. An abnormal adaptive immune response leads to an imbalance between pro- and anti-inflammatory processes. T-helper (Th), T-cytotoxic (Tc) and T-regulatory (Treg) cells may play important roles in immune and inflammatory responses. This study was conducted to clarify the changes and imbalance of cytokines and T lymphocyte subsets in patients with COPD, especially during acute exacerbations (AECOPD). Methods Twenty-three patients with stable COPD (SCOPD) and 21 patients with AECOPD were enrolled in the present study. In addition, 20 age-, sex- and weight-matched non-smoking healthy volunteers were included as controls. The serum levels of selected cytokines (TGF-β, IL-10, TNF-α, IL-17 and IL-9) were measured by enzyme-linked immunosorbent assay (ELISA) kits. Furthermore, the T lymphocyte subsets collected from peripheral blood samples were evaluated by flow cytometry after staining with anti-CD3-APC, anti-CD4-PerCP, anti-CD8- PerCP, anti-CD25-FITC and anti-FoxP3-PE monoclonal antibodies. Importantly, to remove the confounding effects of inflammatory factors, the authors introduced a concept of “inflammation adjustment” and corrected each measured value using representative inflammatory markers, such as TNF-α and IL-17. Results Unlike the other cytokines, serum TGF-β levels were considerably higher in patients with AECOPD relative to the control group regardless of adjustment. There were no significant differences in the percentages of either CD4+ or CD8+ T cells among the three groups. Although Tregs were relatively upregulated during acute exacerbations, their capacities of generation and differentiation were far from sufficient. Finally, the authors noted that the ratios of Treg/IL-17 were similar among groups. Conclusions These observations suggest that in patients with COPD, especially during acute exacerbations, both pro-inflammatory and anti-inflammatory reactions are strengthened, with the pro-inflammatory reactions dominating. Although the Treg/IL-17 ratios were normal, the regulatory T cells were still insufficient to suppress the accompanying increases in inflammation. All of these changes suggest a complicated mechanism of pro- and anti-inflammatory imbalance which needs to be further investigated.
Collapse
Affiliation(s)
- Yang Jin
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Wan
- Department of Respiratory and Critical Care Medicine, Wuhan No. 1 Hospital, Wuhan, China
| | - Gang Chen
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Long Chen
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming-Qiang Zhang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Deng
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Chu Zhang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xian-Zhi Xiong
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- * E-mail:
| | - Jian-Bao Xin
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Abstract
The human pathogenic fungus Candida albicans is the predominant cause of both superficial and invasive forms of candidiasis. C. albicans primarily infects immunocompromised individuals as a result of either immunodeficiency or intervention therapy, which highlights the importance of host immune defences in preventing fungal infections. The host defence system utilises a vast communication network of cells, proteins, and chemical signals distributed in blood and tissues, which constitute innate and adaptive immunity. Over the last decade the identity of many key molecules mediating host defence against C. albicans has been identified. This review will discuss how the host recognises this fungus, the events induced by fungal cells, and the host innate and adaptive immune defences that ultimately resolve C. albicans infections during health.
Collapse
|
23
|
Purvis HA, Anderson AE, Young DA, Isaacs JD, Hilkens CMU. A negative feedback loop mediated by STAT3 limits human Th17 responses. THE JOURNAL OF IMMUNOLOGY 2014; 193:1142-50. [PMID: 24973454 DOI: 10.4049/jimmunol.1302467] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The transcription factor STAT3 is critically required for the differentiation of Th17 cells, a T cell subset involved in various chronic inflammatory diseases. In this article, we report that STAT3 also drives a negative-feedback loop that limits the formation of IL-17-producing T cells within a memory population. By activating human memory CD4(+)CD45RO(+) T cells at a high density (HiD) or a low density (LoD) in the presence of the pro-Th17 cytokines IL-1β, IL-23, and TGF-β, we observed that the numbers of Th17 cells were significantly higher under LoD conditions. Assessment of STAT3 phosphorylation revealed a more rapid and stronger STAT3 activation in HiD cells than in LoD cells. Transient inhibition of active STAT3 in HiD cultures significantly enhanced Th17 cell numbers. Expression of the STAT3-regulated ectonucleotidase CD39, which catalyzes ATP hydrolysis, was higher in HiD, than in LoD, cell cultures. Interestingly, inhibition of CD39 ectonucleotidase activity enhanced Th17 responses under HiD conditions. Conversely, blocking the ATP receptor P2X7 reduced Th17 responses in LoD cultures. These data suggest that STAT3 negatively regulates Th17 cells by limiting the availability of ATP. This negative-feedback loop may provide a safety mechanism to limit tissue damage by Th17 cells during chronic inflammation. Furthermore, our results have relevance for the design of novel immunotherapeutics that target the STAT3-signaling pathway, because inhibition of this pathway may enhance, rather than suppress, memory Th17 responses.
Collapse
Affiliation(s)
- Harriet A Purvis
- Institute of Cellular Medicine, Musculoskeletal Research Group, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Amy E Anderson
- Institute of Cellular Medicine, Musculoskeletal Research Group, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - David A Young
- Institute of Cellular Medicine, Musculoskeletal Research Group, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - John D Isaacs
- Institute of Cellular Medicine, Musculoskeletal Research Group, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Catharien M U Hilkens
- Institute of Cellular Medicine, Musculoskeletal Research Group, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| |
Collapse
|
24
|
Hebecker B, Naglik JR, Hube B, Jacobsen ID. Pathogenicity mechanisms and host response during oral Candida albicans infections. Expert Rev Anti Infect Ther 2014; 12:867-79. [DOI: 10.1586/14787210.2014.916210] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Betty Hebecker
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knoell Institute Jena (HKI),
Beutenbergstrasse 11a, 07745 Jena, Germany
- Department Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knoell Institute Jena (HKI),
Beutenbergstrasse 11a, 07745 Jena, Germany
| | - Julian R Naglik
- Mucosal and Salivary Biology Group, King’s College London Dental Institute, King’s College London,
London SE1 1UL, UK
| | - Bernhard Hube
- Department Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knoell Institute Jena (HKI),
Beutenbergstrasse 11a, 07745 Jena, Germany
- Integrated Research and Treatment Center, Sepsis und Sepsisfolgen, Center for Sepsis Control and Care (CSCC), Universitätsklinikum Jena
- Friedrich Schiller University,
Jena, Germany
| | - Ilse D Jacobsen
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knoell Institute Jena (HKI),
Beutenbergstrasse 11a, 07745 Jena, Germany
| |
Collapse
|
25
|
|
26
|
Ryba-Stanisławowska M, Stanisławowski M, Myśliwska J. Effector and regulatory T cell subsets in diabetes-associated inflammation. Is there a connection with ST2/IL-33 axis? Perspective. Autoimmunity 2014; 47:361-71. [PMID: 24547981 DOI: 10.3109/08916934.2014.886198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Type 1 diabetes (DM1) is a chronic inflammatory disease, which when progresses leads to the development of late vascular complications. The disease involves impairments in regulatory and effector subsets of T lymphocytes, which suppress and maintain inflammatory response, respectively. ST2/IL-33 pathway is involved in T-cell-mediated immune response and might regulate the inflammatory process in several diseases. This review presents the latest research findings regarding effector and regulatory T cell subsets in the context of inflammation accompanying DM1 with particular focus on the ST2/IL-33 network and its possible association with T cell-mediated immunity.
Collapse
|
27
|
Higher Circulating Levels of Chemokine CCL20 in Patients with Multiple Sclerosis: Evaluation of the Influences of Chemokine Gene Polymorphism, Gender, Treatment and Disease Pattern. J Mol Neurosci 2014; 53:500-5. [DOI: 10.1007/s12031-013-0214-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 12/15/2013] [Indexed: 01/01/2023]
|
28
|
Abstract
Understanding how T cells generate productive and long-lasting responses, and how these mechanisms are dysregulated in autoimmune and inflammatory disorders is crucial for prevention and treatment of these diseases. MicroRNAs (miRNAs) are short noncoding RNA species able to suppress gene expression post-transcriptionally. Hundreds of different miRNAs are produced in a cell starting from longer precursors. While the role of miRNAs has been clearly established in the regulation of the differentiation, proliferation and effector functions of a variety of immune cells, here I will focus specifically on miRNAs known to be involved in regulating the biology of CD4 T helper lymphocytes.
Collapse
|
29
|
Myneni SR, Settem RP, Sharma A. Bacteria take control of tolls and T cells to destruct jaw bone. Immunol Invest 2013; 42:519-31. [DOI: 10.3109/08820139.2013.822761] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
30
|
Wei CC, You FT, Mei LY, Jian S, Qiang CY. Total glucosides of paeony prevents juxta-articular bone loss in experimental arthritis. Altern Ther Health Med 2013; 13:186. [PMID: 23870279 PMCID: PMC3728075 DOI: 10.1186/1472-6882-13-186] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 07/09/2013] [Indexed: 12/29/2022]
Abstract
Background Total glucosides of paeony (TGP) is a biologically active compound extracted from Paeony root. TGP has been used in rheumatoid arthritis therapy for many years. However, the mechanism by which TGP prevents bone loss has been less explored. Methods TGP was orally administered for 3 months to New Zealand rabbits with antigen-induced arthritis (AIA). Digital x-ray knee images and bone mineral density (BMD) measurements of the subchondral knee bone were performed before sacrifice. Chondrocytes were observed using transmission electron microscopy (TEM). Histological analysis and mRNA expression of receptor activator of nuclear factor-B ligand (RANKL) and osteoprotegerin (OPG) were evaluated in joint tissues. Results The BMD value in TGP rabbits was significantly higher compared with that seen in the AIA model rabbits. In addition, the subchondral bone plate was almost completely preserved by TGP treatment, while there was a decrease in bone plate integrity in AIA rabbits. There was less damage to the chondrocytes of the TGP treated group. Immunohistochemical examination of the TGP group showed that a higher percentage of TGP treated chondrocytes expressed OPG as compared to the chondrocytes isolated from AIA treated animals. In contrast, RANKL expression was significantly decreased in the TGP treated group compared to the AIA group. In support of the immunohistochemistry data, the expression of RANKL mRNA was decreased and OPG mRNA expression was enhanced in the TGP group when compared to that of the AIA model group. Conclusion These results reveal that TGP suppresses juxta-articular osteoporosis and prevents subchondral bone loss. The decreased RANKL and increased OPG expression seen in TGP treated animals could explain how administration of TGP maintains higher BMD.
Collapse
|
31
|
Chen WC, Lai YH, Chen HY, Guo HR, Su IJ, Chen HHW. Interleukin-17-producing cell infiltration in the breast cancer tumour microenvironment is a poor prognostic factor. Histopathology 2013; 63:225-33. [PMID: 23738752 DOI: 10.1111/his.12156] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 03/29/2013] [Indexed: 12/11/2022]
Abstract
AIMS Interleukin-17 (IL-17) is a proinflammatory cytokine that is most prominently produced by T-helper type 17 (Th17) cells, a distinct CD4+ T-helper cell subset. The aim of this study was to investigate the level of IL-17-producing cells in the breast cancer tumour microenvironment and its prognostic role. METHODS AND RESULTS A total of 207 breast carcinoma specimens were assessed by IL-17 immunohistochemistry, and the findings were correlated with clinicopathological parameters. We found that increased numbers of IL-17-producing cells were correlated with high histological grade, negative ER/PR status, and triple-negative molecular subtypes segregated by immunoprofiles. However, they did not correlate with stage, tumour size, nodal status, HER2 status, or histological type. Patients with tumours with high numbers of IL-17-producing cells had shorter disease-free survival (DFS) than patients with tumours with low numbers of IL-17-producing cells (P < 0.01). In multivariate analysis, high IL-17 level [hazard ratio (HR) 2.24; 95% CI 1.06-4.75], advanced T stage (HR 2.73; 95% CI 1.30-5.73), positive HER2 status (HR 4.88; 95% CI 1.47-16.18) and triple-negative subtype (HR 7.46; 95% CI 1.38-40.36) were significant prognostic factors for DFS. CONCLUSIONS Our results indicate that a high level of IL-17-producing cells in the breast cancer tumour microenvironment is a poor prognostic factor.
Collapse
Affiliation(s)
- Wen-Chung Chen
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | | | |
Collapse
|
32
|
Ryba-Stanisławowska M, Skrzypkowska M, Myśliwiec M, Myśliwska J. Loss of the balance between CD4+Foxp3+ regulatory T cells and CD4+IL17A+ Th17 cells in patients with type 1 diabetes. Hum Immunol 2013; 74:701-7. [DOI: 10.1016/j.humimm.2013.01.024] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 01/02/2013] [Accepted: 01/24/2013] [Indexed: 01/10/2023]
|