1
|
Hu X, Xie S, Yi X, Ouyang Y, Zhao W, Yang Z, Zhang Z, Wang L, Huang X, Peng M, Yu F. Bidirectional Mendelian Randomization of Causal Relationship between Inflammatory Cytokines and Different Pathological Types of Lung Cancer. J Cancer 2024; 15:4969-4984. [PMID: 39132165 PMCID: PMC11310887 DOI: 10.7150/jca.98301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/07/2024] [Indexed: 08/13/2024] Open
Abstract
Prior research has proposed a potential association between lung cancer and inflammatory cytokines, yet the specific causal relationship remains unclear, especially across various lung cancer pathologies. This study utilized bidirectional Mendelian randomization (MR) to explore these causal connections, unveiling novel insights. Our research revealed distinctive inflammatory cytokine profiles for each subtype of lung cancer and identified potential biomarkers that could refine diagnostic and therapeutic approaches. We applied two-sample Mendelian randomization, leveraging genetic variance data from three extensive genome-wide association studies (GWAS) focusing on different lung cancer types (lung adenocarcinoma: 1590 cases and 314,193 controls of healthy individuals of European descent; lung squamous cell carcinoma: 1510 cases and 314,193 controls of European ancestry; small cell lung cancer: 717 cases and 314,193 controls of European ancestry). A separate GWAS summary on inflammatory cytokines from 8,293 healthy participants was also included. The inverse variance weighting method was utilized to examine causal relationships, with robustness confirmed through multiple sensitivity analyses, including MR-Egger, weighted median, and MR-PRESSO. Our analysis revealed that elevated levels of IL_1RA were associated with an increased risk of lung adenocarcinoma (OR: 1.29, 95% CI: 1.02-1.64, p = 0.031), while higher MCP_1_MCAF levels correlated with a decreased risk of lung squamous cell carcinoma (OR: 0.77, 95% CI: 0.61-0.98, p = 0.031). Furthermore, IL_10, IL_13, and TRAIL levels were positively associated with lung squamous cell carcinoma risk (IL_10: OR: 1.27, 95% CI: 1.06-1.53, p = 0.012; IL_13: OR: 1.15, 95% CI: 1.06-1.53, p = 0.036; TRAIL: OR: 1.15, 95% CI: 1.06-1.53, p = 0.043). No association was found between inflammatory cytokine levels and small cell lung cancer development, whereas SDF_1A and B-NGF were linked to an increased risk of this cancer type (SDF_1A: OR: 1.13, 95% CI: 1.05-1.21, p = 0.001; B-NGF: OR: 1.13, 95% CI: 1.01-1.27, p = 0.029). No significant relationship was observed between the 41 circulating inflammatory cytokines and lung adenocarcinoma or squamous cell carcinoma development. Our findings indicate distinct associations between specific inflammatory cytokines and different types of lung cancer. Elevated IL_1RA levels are a risk marker for lung adenocarcinoma, whereas higher MCP_1_MCAF levels appear protective against lung squamous cell carcinoma. Conversely, elevated levels of IL_10, IL_13, and TRAIL are linked with an increased risk of lung squamous cell carcinoma. The relationships of SDF_1A and B-NGF with small-cell lung cancer highlight the complexity of inflammatory markers in cancer development. This study provides a nuanced understanding of the role of inflammatory cytokines in lung cancer, underscoring their potential in refining diagnosis and treatment strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Muyun Peng
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Fenglei Yu
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha 410000, China
| |
Collapse
|
2
|
Jeong B, Pahan K. IL-12p40 Monomer: A Potential Player in Macrophage Regulation. IMMUNO 2024; 4:77-90. [PMID: 38435456 PMCID: PMC10907066 DOI: 10.3390/immuno4010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024] Open
Abstract
Macrophages are myeloid phagocytic leukocytes whose functions are to protect against infections, mediate T-cell responses, and maintain tissue homeostasis. IL-12p40 monomer is a cytokine that is largely produced by macrophages, and it has, for the longest time, been considered a largely non-functional cytokine of the IL-12 family. However, new research has emerged that demonstrates that this p40 monomer may play a bigger role in shaping immune environments. To shed light on the specific effects of p40 monomer on macrophages and their surrounding environment, we showed, through cell culture studies, qPCR, ELISA, and immunofluorescence analyses, that the direct administration of recombinant p40 monomer to RAW 264.7 cells and primary lung macrophages stimulated the production of both pro-inflammatory (TNFα) and anti-inflammatory (IL-10) signals. Accordingly, p40 monomer prevented the full pro-inflammatory effects of LPS, and the neutralization of p40 monomer by mAb a3-3a stimulated the pro-inflammatory effects of LPS. Furthermore, we demonstrated that the intranasal administration of p40 monomer upregulated TNFα+IL-10+ macrophages in vivo in the lungs of mice. Collectively, these results indicate an important immunoregulatory function of p40 monomer in the upregulation of both pro- and anti-inflammatory molecules in macrophages.
Collapse
Affiliation(s)
- Brian Jeong
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
3
|
Shih LJ, Yang CC, Liao MT, Lu KC, Hu WC, Lin CP. An important call: Suggestion of using IL-10 as therapeutic agent for COVID-19 with ARDS and other complications. Virulence 2023; 14:2190650. [PMID: 36914565 PMCID: PMC10026935 DOI: 10.1080/21505594.2023.2190650] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023] Open
Abstract
The global coronavirus disease 2019 (COVID-19) pandemic has a detrimental impact on public health. COVID-19 usually manifests as pneumonia, which can progress into acute respiratory distress syndrome (ARDS) related to uncontrolled TH17 immune reaction. Currently, there is no effective therapeutic agent to manage COVID-19 with complications. The currently available anti-viral drug remdesivir has an effectiveness of 30% in SARS-CoV-2-induced severe complications. Thus, there is a need to identify effective agents to treat COVID-19 and the associated acute lung injury and other complications. The host immunological pathway against this virus typically involves the THαβ immune response. THαβ immunity is triggered by type 1 interferon and interleukin-27 (IL-27), and the main effector cells of the THαβ immune response are IL10-CD4 T cells, CD8 T cells, NK cells, and IgG1-producing B cells. In particular, IL-10 exerts a potent immunomodulatory or anti-inflammatory effect and is an anti-fibrotic agent for pulmonary fibrosis. Concurrently, IL-10 can ameliorate acute lung injury or ARDS, especially those caused by viruses. Owing to its anti-viral activity and anti-pro-inflammatory effects, in this review, IL-10 is suggested as a possible treatment agent for COVID-19.
Collapse
Affiliation(s)
- Li-Jane Shih
- Department of Medical Laboratory, Taoyuan Armed Forces General Hospital, Taoyuan City, Taiwan
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei City, Taiwan
| | - Chun-Chun Yang
- Department of Laboratory Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Min-Tser Liao
- Department of Pediatrics, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
- National Defense Medical Center, Department of Pediatrics, Tri-Service General Hospital, Taipei, Taiwan
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Medicine, Fu-Jen Catholic University Hospital, New Taipei City, Taiwan
| | - Wan-Chung Hu
- Department of Clinical Pathology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Chih-Pei Lin
- Department of Laboratory Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- h Department of Biotechnology, Ming Chuan University, Taoyuan, Taiwan
| |
Collapse
|
4
|
Al-Janabi A, Martin P, Khan AR, Foulkes AC, Smith CH, Griffiths CEM, Morris AP, Eyre S, Warren RB. Integrated proteomics and genomics analysis of paradoxical eczema in psoriasis patients treated with biologics. J Allergy Clin Immunol 2023; 152:1237-1246. [PMID: 37536512 DOI: 10.1016/j.jaci.2023.07.011] [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: 02/20/2023] [Revised: 07/10/2023] [Accepted: 07/21/2023] [Indexed: 08/05/2023]
Abstract
BACKGROUND Few studies have explored the immunology and genetic risk of paradoxical eczema occurring as an adverse event of biologic therapy in patients with psoriasis. OBJECTIVES We sought to describe the systemic inflammatory signature of paradoxical eczema using proteomics and explore whether this is genetically mediated. METHODS This study used the Olink Target 96 Inflammation panel on 256 serum samples from 71 patients with psoriasis and paradoxical eczema, and 75 controls with psoriasis to identify differentially expressed proteins and enriched gene sets. Case samples from 1 or more time points (T1 prebiologic, T2 postbiologic, and T3 postparadoxical eczema) were matched 1:1 with control samples. Genes contributing to enriched gene sets were selected, and functional single nucleotide polymorphisms used to create polygenic risk scores in a genotyped cohort of 88 paradoxical eczema cases and 3124 psoriasis controls. RESULTS STAMBP expression was lower in cases at T1 than in controls (log-fold change: -0.44; adjusted P = .022); no other proteins reached statistical significance at equivalent time points. Eleven gene sets including cytokine and chemokine pathways were enriched in cases at T2 and 10 at T3. Of the 39 proteins contributing to enriched gene sets, the majority are associated with the atopic dermatitis serum proteome. A polygenic risk score including 38 functional single nucleotide polymorphisms linked to enriched gene sets was associated with paradoxical eczema (adjusted P = .046). CONCLUSIONS The paradoxical eczema systemic inflammatory proteome trends toward atopic dermatitis at a gene-set level and is detectable before onset of the phenotype. This signature could be genetically determined.
Collapse
Affiliation(s)
- Ali Al-Janabi
- Centre for Dermatology Research, Northern Care Alliance NHS Foundation Trust, The University of Manchester, Manchester Academic Health Science Centre, NIHR Manchester Biomedical Research Centre, Manchester, United Kingdom.
| | - Paul Martin
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, University of Manchester, Manchester, United Kingdom; The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | | | - Amy C Foulkes
- Centre for Dermatology Research, Northern Care Alliance NHS Foundation Trust, The University of Manchester, Manchester Academic Health Science Centre, NIHR Manchester Biomedical Research Centre, Manchester, United Kingdom
| | - Catherine H Smith
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, King's College London, United Kingdom; St. John's Institute of Dermatology, Guy's and St Thomas' National Health Service Foundation Trust, London, United Kingdom
| | - Christopher E M Griffiths
- Centre for Dermatology Research, Northern Care Alliance NHS Foundation Trust, The University of Manchester, Manchester Academic Health Science Centre, NIHR Manchester Biomedical Research Centre, Manchester, United Kingdom
| | - Andrew P Morris
- Centre for Dermatology Research, Northern Care Alliance NHS Foundation Trust, The University of Manchester, Manchester Academic Health Science Centre, NIHR Manchester Biomedical Research Centre, Manchester, United Kingdom; Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, University of Manchester, Manchester, United Kingdom
| | - Steve Eyre
- Centre for Dermatology Research, Northern Care Alliance NHS Foundation Trust, The University of Manchester, Manchester Academic Health Science Centre, NIHR Manchester Biomedical Research Centre, Manchester, United Kingdom; Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, University of Manchester, Manchester, United Kingdom
| | - Richard B Warren
- Centre for Dermatology Research, Northern Care Alliance NHS Foundation Trust, The University of Manchester, Manchester Academic Health Science Centre, NIHR Manchester Biomedical Research Centre, Manchester, United Kingdom
| |
Collapse
|
5
|
Carlini V, Noonan DM, Abdalalem E, Goletti D, Sansone C, Calabrone L, Albini A. The multifaceted nature of IL-10: regulation, role in immunological homeostasis and its relevance to cancer, COVID-19 and post-COVID conditions. Front Immunol 2023; 14:1161067. [PMID: 37359549 PMCID: PMC10287165 DOI: 10.3389/fimmu.2023.1161067] [Citation(s) in RCA: 46] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023] Open
Abstract
Interleukin-10 (IL-10) is a pleiotropic cytokine that has a fundamental role in modulating inflammation and in maintaining cell homeostasis. It primarily acts as an anti-inflammatory cytokine, protecting the body from an uncontrolled immune response, mostly through the Jak1/Tyk2 and STAT3 signaling pathway. On the other hand, IL-10 can also have immunostimulating functions under certain conditions. Given the pivotal role of IL-10 in immune modulation, this cytokine could have relevant implications in pathologies characterized by hyperinflammatory state, such as cancer, or infectious diseases as in the case of COVID-19 and Post-COVID-19 syndrome. Recent evidence proposed IL-10 as a predictor of severity and mortality for patients with acute or post-acute SARS-CoV-2 infection. In this context, IL-10 can act as an endogenous danger signal, released by tissues undergoing damage in an attempt to protect the organism from harmful hyperinflammation. Pharmacological strategies aimed to potentiate or restore IL-10 immunomodulatory action may represent novel promising avenues to counteract cytokine storm arising from hyperinflammation and effectively mitigate severe complications. Natural bioactive compounds, derived from terrestrial or marine photosynthetic organisms and able to increase IL-10 expression, could represent a useful prevention strategy to curb inflammation through IL-10 elevation and will be discussed here. However, the multifaceted nature of IL-10 has to be taken into account in the attempts to modulate its levels.
Collapse
Affiliation(s)
- Valentina Carlini
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), MultiMedica, Milan, Italy
| | - Douglas M. Noonan
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), MultiMedica, Milan, Italy
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Eslam Abdalalem
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), MultiMedica, Milan, Italy
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Clementina Sansone
- Stazione Zoologica Anton Dohrn, Istituto Nazionale di Biologia, Ecologia e Biotecnologie Marine, Napoli, Italy
| | - Luana Calabrone
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), MultiMedica, Milan, Italy
| | - Adriana Albini
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) European Institute of Oncology IEO-, Milan, Italy
| |
Collapse
|
6
|
Kavanaugh A, Baraliakos X, Gao S, Chen W, Sweet K, Chakravarty SD, Song Q, Shawi M, Rahman P. Genetic and Molecular Distinctions Between Axial Psoriatic Arthritis and Radiographic Axial Spondyloarthritis: Post Hoc Analyses from Four Phase 3 Clinical Trials. Adv Ther 2023; 40:2439-2456. [PMID: 36995469 PMCID: PMC10129963 DOI: 10.1007/s12325-023-02475-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/20/2023] [Indexed: 03/31/2023]
Abstract
INTRODUCTION Emerging evidence suggests psoriatic arthritis (PsA) with axial involvement (axPsA) and radiographic axial spondyloarthritis (r-axSpA) may possibly represent distinct disorders, with some differing clinical manifestations, genetic associations, and radiographic findings. Moreover, axPsA and r-axSpA may respond differently to therapies: guselkumab (interleukin [IL]-23p19 subunit inhibitor [i]) and ustekinumab (IL-12/23p40i) demonstrated improvements in axial symptoms in patients with PsA; however, neither risankizumab (IL-23p19i) nor ustekinumab demonstrated efficacy versus placebo in patients with r-axSpA. Current analyses aim to further understand potential molecular distinctions between axPsA and r-axSpA and examine the pharmacodynamic effects of guselkumab in patients with axPsA and those with PsA without axial involvement (non-axPsA). METHODS Post hoc analyses utilized biomarker data from blood and serum samples collected from a subset of participants in phase 3 studies of ustekinumab in r-axSpA and guselkumab in PsA (DISCOVER-1 and DISCOVER-2). Participants with axPsA were identified by investigator-verified sacroiliitis (imaging-confirmed) and axial symptoms. HLA mapping, serum cytokine analysis, and whole-blood RNA sequencing were conducted. RESULTS Relative to r-axSpA, patients with axPsA had a lower prevalence of HLA-B27, HLA-C01, and HLA-C02 alleles and a higher prevalence of HLA-B13, HLA-B38, HLA-B57, HLA-C06, and HLA-C12 alleles. Compared with r-axSpA, patients with axPsA had elevated baseline levels of serum IL-17A and IL-17F cytokines, enrichment of IL-17 and IL-10 pathway-associated genes, and neutrophil gene markers. Across axPsA and non-axPsA cohorts, reductions in cytokine levels and normalization of pathway-associated gene expression with guselkumab treatment were comparable. CONCLUSION The differences in HLA genetic associations, serum cytokines, and enrichment scores support the concept that axPsA and r-axSpA may be distinct disorders. The comparable pharmacodynamic effects of guselkumab on cytokine levels and pathway-associated genes observed in patients with axPsA and non-axPsA are consistent with demonstrated clinical improvements across PsA cohorts. These findings contribute to the understanding of potential genetic and molecular distinctions between axPsA and r-axSpA. TRIAL REGISTRATION ClinicalTrials.gov identifiers, NCT03162796, NCT0315828, NCT02437162, and NCT02438787.
Collapse
Affiliation(s)
- Arthur Kavanaugh
- University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | | | - Sheng Gao
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Warner Chen
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Kristen Sweet
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Soumya D Chakravarty
- Janssen Scientific Affairs, LLC, Horsham, PA, USA
- Drexel University College of Medicine, Philadelphia, PA, USA
| | - Qingxuan Song
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - May Shawi
- Immunology Global Medical Affairs, Janssen Pharmaceutical Companies of Johnson & Johnson, Horsham, PA, USA
| | - Proton Rahman
- Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|
7
|
Haghmorad D, Yousefi B, Eslami M, Rashidy-Pour A, Tarahomi M, Tavaf MJ, Soltanmohammadi A, Zargarani S, Kamyshnyi A, Oksenych V. Oral Administration of Myelin Oligodendrocyte Glycoprotein Attenuates Experimental Autoimmune Encephalomyelitis through Induction of Th2/Treg Cells and Suppression of Th1/Th17 Immune Responses. Curr Issues Mol Biol 2022; 44:5728-5740. [PMID: 36421672 PMCID: PMC9688797 DOI: 10.3390/cimb44110388] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 10/28/2024] Open
Abstract
Multiple Sclerosis (MS) is a demyelinating autoimmune disorder of the central nervous system (CNS). Experimental autoimmune encephalomyelitis (EAE) has been widely used to determine the pathogenesis of the disease and evaluate new treatment strategies for MS. Therefore, we investigated the efficacy of oral administration of a Myelin Oligodendrocyte Glycoprotein (MOG) in the treatment of EAE. Female C57BL/6 mice were utilized in three groups (Control group, received PBS orally; prevention group, oral administration of MOG35-55 two weeks before EAE induction; treatment group, oral administration of MOG35-55 after EAE induction). MOG administration, both as prevention and treatment, significantly controlled clinical score, weight loss, CNS inflammation, and demyelination, mainly through the modulation of T cell proliferation, and reduction in pro-inflammatory cytokines and transcription factors, including TNF-α, IFN-γ, IL-17, T-bet, and ROR-γt. MOG administration, both as prevention and treatment, also induced anti-inflammatory cytokines and transcription factors, including IL-4, TGF-β, GATA-3, and Foxp3. The results showed that oral administration of MOG, both as prevention and treatment, could efficiently control EAE development. Immunomodulatory mechanisms include the induction of Th2 and Treg cells and the suppression of pro-inflammatory Th1 and Th17 cells.
Collapse
Affiliation(s)
- Dariush Haghmorad
- Cancer Research Center, Semnan University of Medical Sciences, Semnan 35131, Iran
- Department of Immunology, Semnan University of Medical Sciences, Semnan 35131, Iran
| | - Bahman Yousefi
- Cancer Research Center, Semnan University of Medical Sciences, Semnan 35131, Iran
- Department of Immunology, Semnan University of Medical Sciences, Semnan 35131, Iran
| | - Majid Eslami
- Department of Bacteriology and Virology, Semnan University of Medical Sciences, Semnan 35131, Iran
| | - Ali Rashidy-Pour
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan 35131, Iran
| | - Mahdieh Tarahomi
- Department of Immunology, Semnan University of Medical Sciences, Semnan 35131, Iran
| | - Maryam Jadid Tavaf
- Department of Immunology, Semnan University of Medical Sciences, Semnan 35131, Iran
| | | | - Simin Zargarani
- Department of Immunology, Semnan University of Medical Sciences, Semnan 35131, Iran
| | - Aleksandr Kamyshnyi
- Department of Microbiology, Virology and Immunology, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
| | - Valentyn Oksenych
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway
| |
Collapse
|
8
|
Franken A, Van Mol P, Vanmassenhove S, Donders E, Schepers R, Van Brussel T, Dooms C, Yserbyt J, De Crem N, Testelmans D, De Wever W, Nackaerts K, Vansteenkiste J, Vos R, Humblet-Baron S, Lambrechts D, Wauters E. Single-cell transcriptomics identifies pathogenic T-helper 17.1 cells and pro-inflammatory monocytes in immune checkpoint inhibitor-related pneumonitis. J Immunother Cancer 2022; 10:jitc-2022-005323. [PMID: 36171010 PMCID: PMC9528720 DOI: 10.1136/jitc-2022-005323] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2022] [Indexed: 11/11/2022] Open
Abstract
Background Immune checkpoint inhibitor (ICI)-related pneumonitis is the most frequent fatal immune-related adverse event associated with programmed cell death protein-1/programmed death ligand-1 blockade. The pathophysiology however remains largely unknown, owing to limited and contradictory findings in existing literature pointing at either T-helper 1 or T-helper 17-mediated autoimmunity. In this study, we aimed to gain novel insights into the mechanisms of ICI-related pneumonitis, thereby identifying potential therapeutic targets. Methods In this prospective observational study, single-cell RNA and T-cell receptor sequencing was performed on bronchoalveolar lavage fluid of 11 patients with ICI-related pneumonitis and 6 demographically-matched patients with cancer without ICI-related pneumonitis. Single-cell transcriptomic immunophenotyping and cell fate mapping coupled to T-cell receptor repertoire analyses were performed. Results We observed enrichment of both CD4+ and CD8+ T cells in ICI-pneumonitis bronchoalveolar lavage fluid. The CD4+ T-cell compartment showed an increase of pathogenic T-helper 17.1 cells, characterized by high co-expression of TBX21 (encoding T-bet) and RORC (ROR-γ), IFN-G (IFN-γ), IL-17A, CSF2 (GM-CSF), and cytotoxicity genes. Type 1 regulatory T cells and naïve-like CD4+ T cells were also enriched. Within the CD8+ T-cell compartment, mainly effector memory T cells were increased. Correspondingly, myeloid cells in ICI-pneumonitis bronchoalveolar lavage fluid were relatively depleted of anti-inflammatory resident alveolar macrophages while pro-inflammatory ‘M1-like’ monocytes (expressing TNF, IL-1B, IL-6, IL-23A, and GM-CSF receptor CSF2RA, CSF2RB) were enriched compared with control samples. Importantly, a feedforward loop, in which GM-CSF production by pathogenic T-helper 17.1 cells promotes tissue inflammation and IL-23 production by pro-inflammatory monocytes and vice versa, has been well characterized in multiple autoimmune disorders but has never been identified in ICI-related pneumonitis. Conclusions Using single-cell transcriptomics, we identified accumulation of pathogenic T-helper 17.1 cells in ICI-pneumonitis bronchoalveolar lavage fluid—a phenotype explaining previous divergent findings on T-helper 1 versus T-helper 17 involvement in ICI-pneumonitis—, putatively engaging in detrimental crosstalk with pro-inflammatory ‘M1-like’ monocytes. This finding yields several novel potential therapeutic targets for the treatment of ICI-pneumonitis. Most notably repurposing anti-IL-23 merits further research as a potential efficacious and safe treatment for ICI-pneumonitis.
Collapse
Affiliation(s)
- Amelie Franken
- VIB - CCB Department of Human Genetics, KU Leuven, Leuven, Flemish Brabant, Belgium
| | - Pierre Van Mol
- VIB - CCB Department of Human Genetics, KU Leuven, Leuven, Flemish Brabant, Belgium.,Pneumology - Respiratory Oncology, Katholieke Universiteit Leuven Universitaire Ziekenhuizen Leuven, Leuven, Flemish Brabant, Belgium
| | - Sam Vanmassenhove
- VIB - CCB Department of Human Genetics, KU Leuven, Leuven, Flemish Brabant, Belgium
| | - Elena Donders
- VIB - CCB Department of Human Genetics, KU Leuven, Leuven, Flemish Brabant, Belgium.,Pneumology - Respiratory Oncology, Katholieke Universiteit Leuven Universitaire Ziekenhuizen Leuven, Leuven, Flemish Brabant, Belgium
| | - Rogier Schepers
- VIB - CCB Department of Human Genetics, KU Leuven, Leuven, Flemish Brabant, Belgium
| | - Thomas Van Brussel
- VIB - CCB Department of Human Genetics, KU Leuven, Leuven, Flemish Brabant, Belgium
| | - Christophe Dooms
- Pneumology - Respiratory Oncology, Katholieke Universiteit Leuven Universitaire Ziekenhuizen Leuven, Leuven, Flemish Brabant, Belgium.,Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Flemish Brabant, Belgium
| | - Jonas Yserbyt
- Pneumology, Katholieke Universiteit Leuven Universitaire Ziekenhuizen Leuven, Leuven, Flemish Brabant, Belgium
| | - Nico De Crem
- Pneumology, Katholieke Universiteit Leuven Universitaire Ziekenhuizen Leuven, Leuven, Flemish Brabant, Belgium
| | - Dries Testelmans
- Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Flemish Brabant, Belgium.,Pneumology, Katholieke Universiteit Leuven Universitaire Ziekenhuizen Leuven, Leuven, Flemish Brabant, Belgium
| | - Walter De Wever
- Department of Imaging & Pathology, KU Leuven, Leuven, Flemish Brabant, Belgium
| | - Kristiaan Nackaerts
- Pneumology - Respiratory Oncology, Katholieke Universiteit Leuven Universitaire Ziekenhuizen Leuven, Leuven, Flemish Brabant, Belgium.,Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Flemish Brabant, Belgium
| | - Johan Vansteenkiste
- Pneumology - Respiratory Oncology, Katholieke Universiteit Leuven Universitaire Ziekenhuizen Leuven, Leuven, Flemish Brabant, Belgium.,Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Flemish Brabant, Belgium
| | - Robin Vos
- Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Flemish Brabant, Belgium.,Pneumology, Katholieke Universiteit Leuven Universitaire Ziekenhuizen Leuven, Leuven, Flemish Brabant, Belgium
| | - Stéphanie Humblet-Baron
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Flemish Brabant, Belgium
| | - Diether Lambrechts
- VIB - CCB Department of Human Genetics, KU Leuven, Leuven, Flemish Brabant, Belgium
| | - Els Wauters
- Pneumology - Respiratory Oncology, Katholieke Universiteit Leuven Universitaire Ziekenhuizen Leuven, Leuven, Flemish Brabant, Belgium.,Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Flemish Brabant, Belgium
| |
Collapse
|
9
|
Modulatory effects of Porphyra-derived polysaccharides, oligosaccharides and their mixture on antigen-specific immune responses in ovalbumin-sensitized mice. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105209] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
10
|
Santos J, Calabrese DR, Greenland JR. Lymphocytic Airway Inflammation in Lung Allografts. Front Immunol 2022; 13:908693. [PMID: 35911676 PMCID: PMC9335886 DOI: 10.3389/fimmu.2022.908693] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/16/2022] [Indexed: 11/16/2022] Open
Abstract
Lung transplant remains a key therapeutic option for patients with end stage lung disease but short- and long-term survival lag other solid organ transplants. Early ischemia-reperfusion injury in the form of primary graft dysfunction (PGD) and acute cellular rejection are risk factors for chronic lung allograft dysfunction (CLAD), a syndrome of airway and parenchymal fibrosis that is the major barrier to long term survival. An increasing body of research suggests lymphocytic airway inflammation plays a significant role in these important clinical syndromes. Cytotoxic T cells are observed in airway rejection, and transcriptional analysis of airways reveal common cytotoxic gene patterns across solid organ transplant rejection. Natural killer (NK) cells have also been implicated in the early allograft damage response to PGD, acute rejection, cytomegalovirus, and CLAD. This review will examine the roles of lymphocytic airway inflammation across the lifespan of the allograft, including: 1) The contribution of innate lymphocytes to PGD and the impact of PGD on the adaptive immune response. 2) Acute cellular rejection pathologies and the limitations in identifying airway inflammation by transbronchial biopsy. 3) Potentiators of airway inflammation and heterologous immunity, such as respiratory infections, aspiration, and the airway microbiome. 4) Airway contributions to CLAD pathogenesis, including epithelial to mesenchymal transition (EMT), club cell loss, and the evolution from constrictive bronchiolitis to parenchymal fibrosis. 5) Protective mechanisms of fibrosis involving regulatory T cells. In summary, this review will examine our current understanding of the complex interplay between the transplanted airway epithelium, lymphocytic airway infiltration, and rejection pathologies.
Collapse
Affiliation(s)
- Jesse Santos
- Department of Medicine University of California, San Francisco, San Francisco, CA, United States
| | - Daniel R. Calabrese
- Department of Medicine University of California, San Francisco, San Francisco, CA, United States
- Medical Service, Veterans Affairs Health Care System, San Francisco, CA, United States
| | - John R. Greenland
- Department of Medicine University of California, San Francisco, San Francisco, CA, United States
- Medical Service, Veterans Affairs Health Care System, San Francisco, CA, United States
| |
Collapse
|
11
|
Ouyang K, Oparaugo N, Nelson AM, Agak GW. T Cell Extracellular Traps: Tipping the Balance Between Skin Health and Disease. Front Immunol 2022; 13:900634. [PMID: 35795664 PMCID: PMC9250990 DOI: 10.3389/fimmu.2022.900634] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/23/2022] [Indexed: 11/30/2022] Open
Abstract
The role of extracellular traps (ETs) in the innate immune response against pathogens is well established. ETs were first identified in neutrophils and have since been identified in several other immune cells. Although the mechanistic details are not yet fully understood, recent reports have described antigen-specific T cells producing T cell extracellular traps (TETs). Depending on their location within the cutaneous environment, TETs may be beneficial to the host by their ability to limit the spread of pathogens and provide protection against damage to body tissues, and promote early wound healing and degradation of inflammatory mediators, leading to the resolution of inflammatory responses within the skin. However, ETs have also been associated with worse disease outcomes. Here, we consider host-microbe ET interactions by highlighting how cutaneous T cell-derived ETs aid in orchestrating host immune responses against Cutibacterium acnes (C. acnes), a commensal skin bacterium that contributes to skin health, but is also associated with acne vulgaris and surgical infections following joint-replacement procedures. Insights on the role of the skin microbes in regulating T cell ET formation have broad implications not only in novel probiotic design for acne treatment, but also in the treatment for other chronic inflammatory skin disorders and autoimmune diseases.
Collapse
Affiliation(s)
- Kelsey Ouyang
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, United States
- Division of Dermatology, Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Nicole Oparaugo
- David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Amanda M. Nelson
- Department of Dermatology, Penn State University College of Medicine, Hershey, PA, United States
| | - George W. Agak
- Division of Dermatology, Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- *Correspondence: George W. Agak,
| |
Collapse
|
12
|
Heng AHS, Han CW, Abbott C, McColl SR, Comerford I. Chemokine-Driven Migration of Pro-Inflammatory CD4 + T Cells in CNS Autoimmune Disease. Front Immunol 2022; 13:817473. [PMID: 35250997 PMCID: PMC8889115 DOI: 10.3389/fimmu.2022.817473] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/25/2022] [Indexed: 12/13/2022] Open
Abstract
Pro-inflammatory CD4+ T helper (Th) cells drive the pathogenesis of many autoimmune conditions. Recent advances have modified views of the phenotype of pro-inflammatory Th cells in autoimmunity, extending the breadth of known Th cell subsets that operate as drivers of these responses. Heterogeneity and plasticity within Th1 and Th17 cells, and the discovery of subsets of Th cells dedicated to production of other pro-inflammatory cytokines such as GM-CSF have led to these advances. Here, we review recent progress in this area and focus specifically upon evidence for chemokine receptors that drive recruitment of these various pro-inflammatory Th cell subsets to sites of autoimmune inflammation in the CNS. We discuss expression of specific chemokine receptors by subsets of pro-inflammatory Th cells and highlight which receptors may be tractable targets of therapeutic interventions to limit pathogenic Th cell recruitment in autoimmunity.
Collapse
Affiliation(s)
- Aaron H S Heng
- The Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, Faculty of Science, The University of Adelaide, Adelaide, SA, Australia
| | - Caleb W Han
- The Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, Faculty of Science, The University of Adelaide, Adelaide, SA, Australia
| | - Caitlin Abbott
- The Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, Faculty of Science, The University of Adelaide, Adelaide, SA, Australia
| | - Shaun R McColl
- The Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, Faculty of Science, The University of Adelaide, Adelaide, SA, Australia
| | - Iain Comerford
- The Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, Faculty of Science, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
13
|
Soprano LL, Ferrero MR, Landoni M, García GA, Esteva MI, Couto AS, Duschak VG. Cruzipain Sulfotopes-Specific Antibodies Generate Cardiac Tissue Abnormalities and Favor Trypanosoma cruzi Infection in the BALB/c Mice Model of Experimental Chagas Disease. Front Cell Infect Microbiol 2022; 11:814276. [PMID: 35059328 PMCID: PMC8763857 DOI: 10.3389/fcimb.2021.814276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 12/07/2021] [Indexed: 11/13/2022] Open
Abstract
Trypanosoma cruzi cruzipain (Cz) bears a C-terminal domain (C-T) that contains sulfated epitopes “sulfotopes” (GlcNAc6S) on its unique N-glycosylation site. The effects of in vivo exposure to GlcNAc6S on heart tissue ultrastructure, immune responses, and along the outcome of infection by T. cruzi, were evaluated in a murine experimental model, BALB/c, using three independent strategies. First, mice were pre-exposed to C-T by immunization. C-T-immunized mice (C-TIM) showed IgG2a/IgG1 <1, induced the production of cytokines from Th2, Th17, and Th1 profiles with respect to those of dC-TIM, which only induced IL-10 respect to the control mice. Surprisingly, after sublethal challenge, both C-TIM and dC-TIM showed significantly higher parasitemia and mortality than the control group. Second, mice exposed to BSA-GlcNAc6S as immunogen (BSA-GlcNAc6SIM) showed: severe ultrastructural cardiac alterations while BSA-GlcNAcIM conserved the regular tissue architecture with slight myofibril changes; a strong highly specific humoral-immune-response reproducing the IgG-isotype-profile obtained with C-TIM; and a significant memory-T-cell-response demonstrating sulfotope-immunodominance with respect to BSA-GlcNAcIM. After sublethal challenge, BSA-GlcNAc6SIM showed exacerbated parasitemias, despite elevated IFN-γ levels were registered. In both cases, the abrogation of ultrastructural alterations when using desulfated immunogens supported the direct involvement of sulfotopes and/or indirect effect through their specific antibodies, in the induction of tissue damage. Finally, a third strategy using a passive transference of sulfotope-specific antibodies (IgG-GlcNAc6S) showed the detrimental activity of IgG-GlcNAc6S on mice cardiac tissue, and mice treated with IgG-GlcNAc6S after a sublethal dose of T. cruzi, surprisingly reached higher parasitemias than control groups. These findings confirmed the indirect role of the sulfotopes, via their IgG-GlcNAc6S, both in the immunopathogenicity as well as favoring T. cruzi infection.
Collapse
Affiliation(s)
- Luciana L Soprano
- Area of Biochemistry of Proteins and Glycobiology of Parasites, Research Department, National Institute of Parasitology "Dr. Mario Fatala Chaben", ANLIS-Malbrán, Health Department, Ciudad Autónoma de Buenos Aires (CABA, 1063), Buenos Aires, Argentina
| | - Maximiliano R Ferrero
- Area of Biochemistry of Proteins and Glycobiology of Parasites, Research Department, National Institute of Parasitology "Dr. Mario Fatala Chaben", ANLIS-Malbrán, Health Department, Ciudad Autónoma de Buenos Aires (CABA, 1063), Buenos Aires, Argentina
| | - Malena Landoni
- Organic Chemistry Department, Natural and Exact Sciences Faculty; Research Center in Carbohydrates (CIHIDECAR), University of Buenos Aires, Buenos Aires, Argentina.,Ministry of Science, Technology and Innovation, National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| | - Gabriela A García
- Area of Biochemistry of Proteins and Glycobiology of Parasites, Research Department, National Institute of Parasitology "Dr. Mario Fatala Chaben", ANLIS-Malbrán, Health Department, Ciudad Autónoma de Buenos Aires (CABA, 1063), Buenos Aires, Argentina.,Ministry of Science, Technology and Innovation, National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| | - Mónica I Esteva
- Area of Biochemistry of Proteins and Glycobiology of Parasites, Research Department, National Institute of Parasitology "Dr. Mario Fatala Chaben", ANLIS-Malbrán, Health Department, Ciudad Autónoma de Buenos Aires (CABA, 1063), Buenos Aires, Argentina
| | - Alicia S Couto
- Organic Chemistry Department, Natural and Exact Sciences Faculty; Research Center in Carbohydrates (CIHIDECAR), University of Buenos Aires, Buenos Aires, Argentina.,Ministry of Science, Technology and Innovation, National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| | - Vilma G Duschak
- Area of Biochemistry of Proteins and Glycobiology of Parasites, Research Department, National Institute of Parasitology "Dr. Mario Fatala Chaben", ANLIS-Malbrán, Health Department, Ciudad Autónoma de Buenos Aires (CABA, 1063), Buenos Aires, Argentina.,Ministry of Science, Technology and Innovation, National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| |
Collapse
|
14
|
Kuca-Warnawin E, Olesińska M, Szczȩsny P, Kontny E. Impact and Possible Mechanism(s) of Adipose Tissue-Derived Mesenchymal Stem Cells on T-Cell Proliferation in Patients With Rheumatic Disease. Front Physiol 2022; 12:749481. [PMID: 35095547 PMCID: PMC8793746 DOI: 10.3389/fphys.2021.749481] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/14/2021] [Indexed: 12/18/2022] Open
Abstract
Objectives: Systemic lupus erythematosus (SLE) and systemic sclerosis (SSc) are chronic wasting, incurable rheumatic diseases of autoimmune background, in which T cells play a critical pathogenic role. Autologous adipose tissue-derived mesenchymal stem cells (ASCs) may represent an alternative therapeutic option for SLE and SSc patients, but the biology of these cells is poorly understood. Methods: Herein, we evaluated the anti-proliferative impact of ASCs of healthy donors (HD/ASCs, 5 reference cell lines), SLE patients (n = 20), and SSc patients (n = 20) on T lymphocytes. To assess the direct and indirect pathway of ASCs action, peripheral blood mononuclear cells (PBMCs) and purified CD4+ T cells of HD were activated and co-cultured in cell-to-cell contact (C-C) and transwell (T-W) conditions with untreated or cytokine (TNF + IFNΥ, TI)-licensed ASCs, then analyzed by flow cytometry to rate the proliferation response of CD8+ and/or CD4+ T cells. The concentrations of kynurenines, prostaglandin E2 (PGE2), interleukin 10 (IL-10), and transforming growth factor β (TGFβ) were measured from culture supernatants. Specific inhibitors of these factors (1-MT, indomethacin, and cytokine-neutralizing antibody) were used to assess their contribution to anti-proliferative ASCs action. Results: All tested ASCs significantly decreased the number of proliferating CD4+ and CD8+ T cells, the number of division/proliferating cell (PI), and fold expansion (RI), and similarly upregulated kynurenines and PGE2, but not cytokine levels, in the co-cultures with both types of target cells. However, TI-treated SLE/ASCs and SSc/ASCs exerted a slightly weaker inhibitory effect on CD4+ T-cell replication than their respective HD/ASCs. All ASCs acted mainly via soluble factors. Their anti-proliferative effect was stronger, and kynurenine levels were higher in the T-W condition than the C-C condition. Blocking experiments indicated an involvement of kynurenine pathway in inhibiting the number of proliferating cells, PI, and RI values as well as PGE2 role in decreasing the number of proliferating cells. TGFβ did not contribute to ASCs anti-proliferative capabilities, while IL-10 seems to be involved in such activity of only SLE/ASCs. Conclusion: The results indicate that SLE/ASCs and SSc/ASCs retain their capability to restrain the expansion of allogeneic CD4+ and CD8+ T cells and act by similar mechanisms as ASCs of healthy donors and thus may have therapeutic value.
Collapse
Affiliation(s)
- Ewa Kuca-Warnawin
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Marzena Olesińska
- Clinic of Connective Tissue Diseases, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Piotr Szczȩsny
- Clinic of Connective Tissue Diseases, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Ewa Kontny
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| |
Collapse
|
15
|
Wang C, Yang J, Xie L, Saimaier K, Zhuang W, Han M, Liu G, Lv J, Shi G, Li N, Du C. Methyl Butyrate Alleviates Experimental Autoimmune Encephalomyelitis and Regulates the Balance of Effector T Cells and Regulatory T Cells. Inflammation 2021; 45:977-991. [PMID: 34786625 DOI: 10.1007/s10753-021-01596-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/31/2021] [Accepted: 11/03/2021] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS), characterized by demyelinating neuropathy. The etiology of MS is not yet clear and its treatment remains a major medical challenge. While we search for drugs that can effectively treat experimental autoimmune encephalomyelitis (EAE), the animal model of MS, we also hope to further explore its possible pathogenesis. In the present study, we investigated whether methyl butyrate (MB) could alleviate EAE and its potential mechanisms. In EAE mice, we found that administration of MB was effective in alleviating their clinical signs and improving histopathological manifestations of the CNS. In the CNS and intestinal lamina propria, we observed fewer effector T cells, including Th1 and Th17, in the MB-treated group. MB also increased the proportion of regulatory T cells and the secretion of IL-10 in peripheral immune organs. In vitro, MB led to suppression of Th1 cells and promotion of regulatory T cells in their differentiation. Given that MB had no direct effect on Th17 cell differentiation in vitro, we hypothesized that MB suppressed Th17 cells indirectly by inhibiting the secretion of IL-6, which was later confirmed both in vitro and in vivo. In addition, we found that MB treatment upregulated Maf gene expression in mice, which explained its promotion of IL-10 secretion. The above findings suggest that MB may provide new ideas for the study of the mechanism of MS and have positive implications for new drug development.
Collapse
Affiliation(s)
- Chun Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jingshu Yang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200433, China
| | - Ling Xie
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Kaidireya Saimaier
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Wei Zhuang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Institute of Biophysics, National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Mengyao Han
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Guangyu Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jie Lv
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Guangfeng Shi
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200433, China
| | - Ning Li
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200433, China.
| | - Changsheng Du
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
16
|
Krupa A, Kowalska I. The Kynurenine Pathway-New Linkage between Innate and Adaptive Immunity in Autoimmune Endocrinopathies. Int J Mol Sci 2021; 22:9879. [PMID: 34576041 PMCID: PMC8469440 DOI: 10.3390/ijms22189879] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 12/18/2022] Open
Abstract
The kynurenine pathway (KP) is highly regulated in the immune system, where it promotes immunosuppression in response to infection or inflammation. Indoleamine 2,3-dioxygenase 1 (IDO1), the main enzyme of KP, has a broad spectrum of activity on immune cells regulation, controlling the balance between stimulation and suppression of the immune system at sites of local inflammation, relevant to a wide range of autoimmune and inflammatory diseases. Various autoimmune diseases, among them endocrinopathies, have been identified to date, but despite significant progress in their diagnosis and treatment, they are still associated with significant complications, morbidity, and mortality. The precise cellular and molecular mechanisms leading to the onset and development of autoimmune disease remain poorly clarified so far. In breaking of tolerance, the cells of the innate immunity provide a decisive microenvironment that regulates immune cells' differentiation, leading to activation of adaptive immunity. The current review provided a comprehensive presentation of the known role of IDO1 and KP activation in the regulation of the innate and adaptive arms of the immune system. Significant attention has been paid to the immunoregulatory role of IDO1 in the most prevalent, organ-specific autoimmune endocrinopathies-type 1 diabetes mellitus (T1DM) and autoimmune thyroiditis.
Collapse
Affiliation(s)
- Anna Krupa
- Department of Internal Medicine and Metabolic Diseases, Medical University of Bialystok, M. Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland
| | - Irina Kowalska
- Department of Internal Medicine and Metabolic Diseases, Medical University of Bialystok, M. Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland
| |
Collapse
|
17
|
Zalosnik MI, Fabio MC, Bertoldi ML, Castañares CN, Degano AL. MeCP2 deficiency exacerbates the neuroinflammatory setting and autoreactive response during an autoimmune challenge. Sci Rep 2021; 11:10997. [PMID: 34040112 PMCID: PMC8155097 DOI: 10.1038/s41598-021-90517-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 05/12/2021] [Indexed: 02/04/2023] Open
Abstract
Rett syndrome is a severe and progressive neurological disorder linked to mutations in the MeCP2 gene. It has been suggested that immune alterations may play an active role in the generation and/or maintenance of RTT phenotypes. However, there is no clear consensus about which pathways are regulated in vivo by MeCP2 in the context of immune activation. In the present work we set to characterize the role of MeCP2 during the progression of Experimental Autoimmune Encephalomyelitis (EAE) using the MeCP2308/y mouse model (MUT), which represents a condition of "MeCP2 function deficiency". Our results showed that MeCP2 deficiency increased the susceptibility to develop EAE, along with a defective induction of anti-inflammatory responses and an exacerbated MOG-specific IFNγ expression in immune sites. In MUT-EAE spinal cord, we found a chronic increase in pro-inflammatory cytokines gene expression (IFNγ, TNFα and IL-1β) and downregulation of genes involved in immune regulation (IL-10, FoxP3 and CX3CR1). Moreover, our results indicate that MeCP2 acts intrinsically upon immune activation, affecting neuroimmune homeostasis by regulating the pro-inflammatory/anti-inflammatory balance in vivo. These results are relevant to identify the potential consequences of MeCP2 mutations on immune homeostasis and to explore novel therapeutic strategies for MeCP2-related disorders.
Collapse
Affiliation(s)
- M I Zalosnik
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
- Centro de Investigaciones en Química Biológica de Córdoba, Consejo Nacional de Investigaciones Científicas y Técnicas (CIQUIBIC, CONICET), Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - M C Fabio
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Córdoba (INIMEC-CONICET-UNC), Córdoba, Argentina
| | - M L Bertoldi
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
- Centro de Investigaciones en Química Biológica de Córdoba, Consejo Nacional de Investigaciones Científicas y Técnicas (CIQUIBIC, CONICET), Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - C N Castañares
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Córdoba (INIMEC-CONICET-UNC), Córdoba, Argentina
| | - A L Degano
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina.
- Centro de Investigaciones en Química Biológica de Córdoba, Consejo Nacional de Investigaciones Científicas y Técnicas (CIQUIBIC, CONICET), Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina.
| |
Collapse
|
18
|
Yuba E, Budina E, Katsumata K, Ishihara A, Mansurov A, Alpar AT, Watkins EA, Hosseinchi P, Reda JW, Lauterbach AL, Nguyen M, Solanki A, Kageyama T, Swartz MA, Ishihara J, Hubbell JA. Suppression of Rheumatoid Arthritis by Enhanced Lymph Node Trafficking of Engineered Interleukin-10 in Murine Models. Arthritis Rheumatol 2021; 73:769-778. [PMID: 33169522 PMCID: PMC11095083 DOI: 10.1002/art.41585] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 10/13/2020] [Accepted: 11/05/2020] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is a major autoimmune disease that causes synovitis and joint damage. Although clinical trials have been performed using interleukin-10 (IL-10), an antiinflammatory cytokine, as a potential treatment of RA, the therapeutic effects of IL-10 have been limited, potentially due to insufficient residence in lymphoid organs, where antigen recognition primarily occurs. This study was undertaken to engineer an IL-10-serum albumin (SA) fusion protein and evaluate its effects in 2 murine models of RA. METHODS SA-fused IL-10 (SA-IL-10) was recombinantly expressed. Mice with collagen antibody-induced arthritis (n = 4-7 per group) or collagen-induced arthritis (n = 9-15 per group) were injected intravenously with wild-type IL-10 or SA-IL-10, and the retention of SA-IL-10 in the lymph nodes (LNs), immune cell composition in the paws, and therapeutic effect of SA-IL-10 on mice with arthritis were assessed. RESULTS SA fusion to IL-10 led to enhanced accumulation in the mouse LNs compared with unmodified IL-10. Intravenous SA-IL-10 treatment restored immune cell composition in the paws to a normal status, elevated the frequency of suppressive alternatively activated macrophages, reduced IL-17A levels in the paw-draining LN, and protected joint morphology. Intravenous SA-IL-10 treatment showed similar efficacy as treatment with an anti-tumor necrosis factor antibody. SA-IL-10 was equally effective when administered intravenously, locally, or subcutaneously, which is a benefit for clinical translation of this molecule. CONCLUSION SA fusion to IL-10 is a simple but effective engineering strategy for RA therapy and has potential for clinical translation.
Collapse
Affiliation(s)
- Eiji Yuba
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
- Department of Applied Chemistry, Osaka Prefecture University, Osaka 599-8531, Japan
| | - Erica Budina
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Kiyomitsu Katsumata
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Ako Ishihara
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Aslan Mansurov
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Aaron T. Alpar
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Elyse A. Watkins
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Peyman Hosseinchi
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Joseph W. Reda
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Abigail L. Lauterbach
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Mindy Nguyen
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Ani Solanki
- Animal Resource Center, University of Chicago, Chicago, IL 60637, USA
| | - Takahiro Kageyama
- Department of Microbiology and Immunology, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA
| | - Melody A. Swartz
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
- Ben May Department of Cancer Research, University of Chicago, Chicago, IL 60637, USA
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Jun Ishihara
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
- Department of Bioengineering, Imperial College London, London W12 0BZ, UK
| | - Jeffrey A. Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
19
|
Regulatory and Effector Cell Disequilibrium in Patients with Acute Cellular Rejection and Chronic Lung Allograft Dysfunction after Lung Transplantation: Comparison of Peripheral and Alveolar Distribution. Cells 2021; 10:cells10040780. [PMID: 33916034 PMCID: PMC8065700 DOI: 10.3390/cells10040780] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/28/2021] [Accepted: 03/29/2021] [Indexed: 01/19/2023] Open
Abstract
Background: The immune mechanisms occurring during acute rejection (AR) and chronic lung allograft dysfunction are a challenge for research and the balance between effector and regulatory cells has not been defined completely. In this study, we aimed to elucidate the interaction of effector cells, mainly Th17, Th1 and Th2, and regulatory cells including (CD4+CD25+CD127low/−) T reg cells and phenotypes of B regs, CD19+CD24hiCD38hi, CD19+CD24hiCD27hi and CD19+CD5+CD1d+. Methods: Bronchoalveolar lavage cells (BAL) and peripheral blood mononuclear cells (PBMCs) from stable lung transplanted (LTx )subjects (n = 4), AR patients (n = 6) and bronchiolitis obliterans syndrome (BOS) (n = 6) were collected at the same time. Cellular subsets were detected through flow cytometry. Results: A predominance of Th17 cells subtypes in the PBMCs and BAL and a depletion of Tregs, that resulted in decrease Treg/Th17 ratio, was observed in the AR group. CD19+CD24hiCD38hi Bregs resulted increased in BAL of AR patients. Th1 cells predominance and a reduction of Tregs cells was observed in BAL from AR patients. Moreover, multivariate analysis showed interdependences within studied variables revealing that effector cells and regulatory cells can effectively discriminate patients’ immunological status. Conclusions: In AR, BOS and stable lung transplant, regulatory and effector cells clearly demonstrated different pathways of activation. Understanding of the balance of T cells and T and B regulatory cells can offers insights into rejection.
Collapse
|
20
|
Almeida RS, Ferreira MLB, Sonon P, Cordeiro MT, Sadissou I, Diniz GTN, Militão-Albuquerque MDFP, Franca RFDO, Donadi EA, Lucena-Silva N. Cytokines and Soluble HLA-G Levels in the Acute and Recovery Phases of Arbovirus-Infected Brazilian Patients Exhibiting Neurological Complications. Front Immunol 2021; 12:582935. [PMID: 33776990 PMCID: PMC7994272 DOI: 10.3389/fimmu.2021.582935] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 01/08/2021] [Indexed: 12/19/2022] Open
Abstract
Severe neurological complications following arbovirus infections have been a major concern in seasonal outbreaks, as reported in the Northeast region of Brazil, where the same mosquito transmitted Zika (ZIKV), Dengue (DENV), and Chikungunya (CHIKV) viruses. In this study, we evaluated the levels of 36 soluble markers, including cytokines, chemokines, growth factors, and soluble HLA-G (Luminex and ELISA) in: i) serum and cerebrospinal fluid (CSF), during the acute phase and two years after the infection (recovery phase, only serum), ii) the relationship among all soluble molecules in serum and CSF, and iii) serum of infected patients without neurological complications, during the acute infection. Ten markers (sHLA-G, IL-10, IL-22, IL-8, MIP-1α, MIP-1β, MCP-1, HGF, VEGF, and IL-1RA) exhibited differential levels between the acute and recovery phases, with pronounced increases in MIP-1α (P<0.0001), MCP-1 (P<0.0001), HGF (P= 0.0001), and VEGF (P<0.0001) in the acute phase. Fourteen molecules (IL-1β, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-9, IL-13, IL-15, IL-17A, IFN-α, TNF, and G-CSF) exhibited distinct levels between arbovirus patients presenting or not neurological complications. IL-8, EGF, IL-6, and MCP-1 levels were increased in CSF, while RANTES and Eotaxin levels were higher in serum. Soluble serum (IL-22, RANTES, Eotaxin) and CSF (IL-8, EGF, IL-3) mediators may discriminate putative risks for neurological complications following arbovirus infections. Neurological complications were associated with the presence of a predominant inflammatory profile, whereas in non-complicated patients an anti-inflammatory profile may predominate. Mediators associated with neuroregeneration (EGF and IL-3) may be induced in response to neurological damage. Broad spectrum immune checkpoint molecules (sHLA-G) interact with cytokines, chemokines, and growth factors. The identification of soluble markers may be useful to monitor neurological complications and may aid in the development of novel therapies against neuroinflammation.
Collapse
Affiliation(s)
- Renata Santos Almeida
- Laboratory of Immunogenetics, Department of Immunology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil
| | | | - Paulin Sonon
- Laboratory of Immunogenetics, Department of Immunology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil.,Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Marli Tenório Cordeiro
- Department of Virology and Experimental Therapy, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil
| | - Ibrahim Sadissou
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - George Tadeu Nunes Diniz
- Department of Collective Health, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil
| | | | | | | | - Norma Lucena-Silva
- Laboratory of Immunogenetics, Department of Immunology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil
| |
Collapse
|
21
|
Motavalli R, Etemadi J, Soltani-Zangbar MS, Ardalan MR, Kahroba H, Roshangar L, Nouri M, Aghebati-Maleki L, Khiavi FM, Abediazar S, Mehdizadeh A, Hojjat-Farsangi M, Mahmoodpoor A, Kafil HS, Zolfaghari M, Ahmadian Heris J, Yousefi M. Altered Th17/Treg ratio as a possible mechanism in pathogenesis of idiopathic membranous nephropathy. Cytokine 2021; 141:155452. [PMID: 33571932 DOI: 10.1016/j.cyto.2021.155452] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/26/2022]
Abstract
Idiopathic membranous nephropathy (IMN) as a single organ autoimmune disease is a main cause of nephrotic syndrome in adults which is determined through autoantibodies to podocytes proteins. Th17/regulatory T (Treg) balance has emerged as a prominent factor in the regulation of autoimmunity. In this study, we evaluated the balance of Th17 and Treg cells, expression level of related master transcription factors, cytokines and microRNAs in mononuclear cells of peripheral blood of 30 patients with IMN and 30 healthy individuals before treatment. No significant variation was observed in Th17 cell frequency, retinoic acid receptor-related orphan nuclear receptor γt (RORɣt), signal transducer and Activator of transcription 3(STAT3), IL-17, and IL-23, while IL-21, IL-4, and IL-10 had significant increase in mRNA expression and protein level of peripheral blood mononuclear cells in IMN cases. Reduction in the percentage of Treg cells was also accompanied with significantly decreased expression of Forkhead box P3(FOXP3) and Transforming growth factor beta(TGF-β) in IMN patients compared to the control group. Our study revealed that Th17 cells themselves might not be engaged in the pathogenesis of newly diagnosed patients with IMN; however, decreased T reg cells and increased ratio of Th17/Treg lymphocytes might display a role in the pathogenesis of IMN before treatment.
Collapse
Affiliation(s)
- Roza Motavalli
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jalal Etemadi
- Kidney Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Mohamad-Reza Ardalan
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Kidney Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Houman Kahroba
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Sima Abediazar
- Kidney Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hojjat-Farsangi
- Department of Oncology-Pathology, Immune and Gene Therapy Lab, Cancer Center Karolinska (CCK), Karolina University Hospital Solna and Karolinska Institute, Stockholm, Sweden
| | - Ata Mahmoodpoor
- Department of Anesthesiology and Critical Care Medicine, Faculty of Medicine, University of Medical Sciences, Tabriz, Iran
| | - Hossein Samadi Kafil
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohamadali Zolfaghari
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Ahmadian Heris
- Department of Pediatrics, School of Medicine, Children Medical Research and Training Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
22
|
Leipe J, Pirronello F, Schulze-Koops H, Skapenko A. Altered T cell plasticity favours Th17 cells in early arthritis. Rheumatology (Oxford) 2021; 59:2754-2763. [PMID: 32030419 DOI: 10.1093/rheumatology/kez660] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/18/2019] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES The predominance of differentiated Th17 cells has been implied as a key driver of autoimmune arthritis, including early RA. Because accumulating evidence suggests that Th cell differentiation is a plastic process, we investigated plasticity and underlying molecular mechanisms to address the shift towards the Th17 phenotype in early RA. METHODS A cohort of 61 patients with early, active, untreated RA and 45 age- and sex-matched healthy controls were studied. Viable in vitro- and in vivo-generated Th1, Th2 and Th17 cells were FACS-sorted and transdifferentiated under Th1-, Th2- or Th17-inducing conditions. The cytokine Th profile of the transdifferentiated cells was assessed by flow cytometry. Th cell-associated cytokine and transcription factor gene loci were analysed by chromatin immunoprecipitation assay and their expression by quantitative real-time PCR. RESULTS In vitro-generated Th cells showed substantial plasticity, which was similar between RA and healthy controls, whereas in vivo-derived Th1 and Th2 cells from RA patients demonstrated an enhanced plasticity towards IL-17-expressing phenotypes compared with healthy controls. Further, in vivo-generated Th17 cells from RA patients showed a resistance to transdifferentiate into Th1 or Th2 cells. The serum/glucocorticoid-regulated kinase 1-forkhead box protein O1-IL-23 receptor (SGK1-FOXO1-IL-23R) axis together with increased RORC expression was associated with the predominant Th17 phenotype in early RA. CONCLUSIONS Our data indicate that in vivo-originated Th subsets are prone to Th17 cell transdifferentiation in early RA, while Th17 cells are resistant to changes in their phenotype. Together, the data imply that an altered plasticity contributes to the Th17 shift in early RA.
Collapse
Affiliation(s)
- Jan Leipe
- Division of Rheumatology and Clinical Immunology, Medizinische Klinik and Poliklinik IV, University of Munich, Munich, Germany.,Division of Rheumatology, Department of Medicine V, University Hospital Mannheim, Mannheim, Germany ∗Jan Leipe and Fausto Pirronello contributed equally to this work
| | - Fausto Pirronello
- Division of Rheumatology and Clinical Immunology, Medizinische Klinik and Poliklinik IV, University of Munich, Munich, Germany
| | - Hendrik Schulze-Koops
- Division of Rheumatology and Clinical Immunology, Medizinische Klinik and Poliklinik IV, University of Munich, Munich, Germany
| | - Alla Skapenko
- Division of Rheumatology and Clinical Immunology, Medizinische Klinik and Poliklinik IV, University of Munich, Munich, Germany
| |
Collapse
|
23
|
Ala M, Kheyri Z. The rationale for selenium supplementation in inflammatory bowel disease: A mechanism-based point of view. Nutrition 2021; 85:111153. [PMID: 33578241 DOI: 10.1016/j.nut.2021.111153] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/19/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023]
Abstract
Management of inflammatory bowel disease (IBD) has always been a challenge for physicians. Current treatment protocols may cause numerous adverse effects. Selenium is known for its putative antiinflammatory properties. Selenium is needed for the biosynthesis of enzymatically active selenoproteins, which contribute to antioxidative defense, and effective function of immune systems. Several studies have shown that patients with IBD have a lower selenium level compared to healthy subjects. Hence, experimental studies mimicking ulcerative colitis and Crohn's disease investigated the effect of selenium supplementation on IBD. Previous studies indicated the following: 1) Selenoproteins can curb the inflammatory response and attenuate oxidative stress. This antiinflammatory property caused remission in animal models of colitis. 2) Selenium supports protective gut microbiota, which indirectly improves management of IBD. 3) Selenium may block some of the predominant tumorigenesis pathways proposed in colitis-associated colorectal cancer. 4) Selenium supplementation showed promising results in preliminary clinical studies, particularly in patients with selenium deficiency. While selenium supplementation seems to be beneficial for IBD, clinical studies have remained too preliminary in this regard. Randomized clinical trials are needed to measure the short-term and long-term effects of selenium on both active and quiescent IBD, particularly in patients with IBD who have documented selenium deficiency.
Collapse
Affiliation(s)
- Moein Ala
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Zahedin Kheyri
- Baharloo Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Jang SG, Lee J, Hong SM, Kwok SK, Cho ML, Park SH. Metformin enhances the immunomodulatory potential of adipose-derived mesenchymal stem cells through STAT1 in an animal model of lupus. Rheumatology (Oxford) 2020; 59:1426-1438. [PMID: 31904843 DOI: 10.1093/rheumatology/kez631] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 11/21/2019] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVES Mesenchymal stem cells (MSCs) are considered potential therapeutic agents for treating autoimmune disease because of their immunomodulatory capacities and anti-inflammatory effects. However, several studies have shown that there is no consistency in the effectiveness of the MSCs to treat autoimmune disease, including SLE. In this study, we investigated whether metformin could enhance the immunoregulatory function of MSCs, what mechanism is relevant, and whether metformin-treated MSCs could be effective in an animal lupus model. METHODS Adipose-derived (Ad)-MSCs were cultured for 72 h in the presence of metformin. Immunoregulatory factors expression was analysed by real-time PCR and ELISA. MRL/lpr mice weekly injected intravenously with 1 × 106 Ad-MSCs or metformin-treated Ad-MSCs for 8 weeks. 16-week-old mice were sacrificed and proteinuria, anti-dsDNA IgG antibody, glomerulonephritis, immune complex, cellular subset were analysed in each group. RESULTS Metformin enhanced the immunomodulatory functions of Ad-MSCs including IDO, IL-10 and TGF-β. Metformin upregulated the expression of p-AMPK, p-STAT1 and inhibited the expression of p-STAT3, p-mTOR in Ad-MSCs. STAT1 inhibition by siRNA strongly diminished IDO, IL-10, TGF-β in metformin-treated Ad-MSCs. As a result, metformin promoted the immunoregulatory effect of Ad-MSCs by enhancing STAT1 expression, which was dependent on the AMPK/mTOR pathway. Administration of metformin-treated Ad-MSCs resulted in significant disease activity improvement including inflammatory phenotype, glomerulonephritis, proteinuria and anti-dsDNA IgG antibody production in MRL/lpr mice. Moreover, metformin-treated Ad-MSCs inhibited CD4-CD8- T-cell expansion and Th17/Treg cell ratio. CONCLUSION Metformin optimized the immunoregulatory properties of Ad-MSCs and may be a novel therapeutic agent for the treatment of lupus.
Collapse
Affiliation(s)
- Se Gwang Jang
- The Rheumatism Research Center, Catholic Research Institute of Medical ScienceThe Catholic University of Korea, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jaeseon Lee
- The Rheumatism Research Center, Catholic Research Institute of Medical ScienceThe Catholic University of Korea, Seoul, Republic of Korea
| | - Seung-Min Hong
- The Rheumatism Research Center, Catholic Research Institute of Medical ScienceThe Catholic University of Korea, Seoul, Republic of Korea
| | - Seung-Ki Kwok
- The Rheumatism Research Center, Catholic Research Institute of Medical ScienceThe Catholic University of Korea, Seoul, Republic of Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical ScienceThe Catholic University of Korea, Seoul, Republic of Korea
| | - Sung-Hwan Park
- The Rheumatism Research Center, Catholic Research Institute of Medical ScienceThe Catholic University of Korea, Seoul, Republic of Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
25
|
Elashiry M, Elashiry MM, Elsayed R, Rajendran M, Auersvald C, Zeitoun R, Rashid MH, Ara R, Meghil MM, Liu Y, Arbab AS, Arce RM, Hamrick M, Elsalanty M, Brendan M, Pacholczyk R, Cutler CW. Dendritic cell derived exosomes loaded with immunoregulatory cargo reprogram local immune responses and inhibit degenerative bone disease in vivo. J Extracell Vesicles 2020; 9:1795362. [PMID: 32944183 PMCID: PMC7480413 DOI: 10.1080/20013078.2020.1795362] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Chronic bone degenerative diseases represent a major threat to the health and well-being of the population, particularly those with advanced age. This study isolated exosomes (EXO), natural nano-particles, from dendritic cells, the “directors” of the immune response, to examine the immunobiology of DC EXO in mice, and their ability to reprogram immune cells responsible for experimental alveolar bone loss in vivo. Distinct DC EXO subtypes including immune-regulatory (regDC EXO), loaded with TGFB1 and IL10 after purification, along with immune stimulatory (stimDC EXO) and immune “null” immature (iDCs EXO) unmodified after purification, were delivered via I.V. route or locally into the soft tissues overlying the alveolar bone. Locally administrated regDC EXO showed high affinity for inflamed sites, and were taken up by both DCs and T cells in situ. RegDC EXO-encapsulated immunoregulatory cargo (TGFB1 and IL10) was protected from proteolytic degradation. Moreover, maturation of recipient DCs and induction of Th17 effectors was suppressed by regDC EXO, while T-regulatory cell recruitment was promoted, resulting in inhibition of bone resorptive cytokines and reduction in osteoclastic bone loss. This work is the first demonstration of DC exosome-based therapy for a degenerative alveolar bone disease and provides the basis for a novel treatment strategy.
Collapse
Affiliation(s)
- Mahmoud Elashiry
- Department of Periodontics, Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia at Augusta University, Augusta, GA, USA
| | - Mohamed M Elashiry
- Department of Periodontics, Dental College of Georgia at Augusta University, GA, USA, Department of Endodontics, Faculty of Dentistry, Ain Shams University, Cairo, Egypt
| | - Ranya Elsayed
- Department of Periodontics, Dental College of Georgia at Augusta University, GA, USA
| | - Mythily Rajendran
- Department of Periodontics, Dental College of Georgia at Augusta University, GA, USA
| | - Carol Auersvald
- Department of Periodontics, Dental College of Georgia at Augusta University, GA, USA
| | - Rana Zeitoun
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia at Augusta University, Department of Fixed Prosthodontics, Faculty of Dentistry, Ain Shams University, Cairo, Egypt
| | - Mohammad H Rashid
- Department of Biochemistry & Molecular Biology, Georgia Cancer Center, Augusta, GA, USA
| | - Roxan Ara
- Department of Biochemistry & Molecular Biology, Georgia Cancer Center, Augusta, GA, USA
| | - Mohamed M Meghil
- Department of Periodontics, Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia at Augusta University, Augusta, GA, USA
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, GA, USA
| | - Ali S Arbab
- Department of Biochemistry & Molecular Biology, Georgia Cancer Center, Augusta, GA, USA
| | - Roger M Arce
- Department of Periodontics and Oral Hygiene, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Mark Hamrick
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, GA, USA
| | - Mohammed Elsalanty
- Department of Periodontics, Dental College of Georgia at Augusta University, GA, USA
| | - Marshall Brendan
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, GA, USA
| | - Rafal Pacholczyk
- Georgia Cancer Center, Augusta, GA, USA.,Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia at Augusta University, GA, USA
| | - Christopher W Cutler
- Department of Periodontics, Dental College of Georgia at Augusta University, GA, USA
| |
Collapse
|
26
|
Guo J, Wang LY, Wu J, Xu LF, Sun M. The JAK2 inhibitor AG490 regulates the Treg/Th17 balance and alleviates DSS-induced intestinal damage in IBD rats. Clin Exp Pharmacol Physiol 2020; 47:1374-1381. [PMID: 32215928 DOI: 10.1111/1440-1681.13311] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/17/2020] [Accepted: 03/19/2020] [Indexed: 01/26/2023]
Abstract
The pathogenesis of inflammatory bowel disease (IBD) remains unclear, and it is currently believed that an imbalance in regulatory T (Treg) cells/T helper 17 cells (Th17 cells) is related to the occurrence and development of IBD. Recently, the JAK2 inhibitor AG490 has been used in animal models such as rheumatoid arthritis and bronchial asthma models and shown to exert immunoregulatory functions that improve disorder in the Treg/Th17 cell balance. This study aimed to evaluate the effect of AG490 on the intestinal inflammatory process in an IBD rat model. A dextran sulfate sodium (DSS)-induced IBD rat model was established, and disease activity index (DAI) scores were calculated. The histopathological damage score was determined by haematoxylin-eosin (H&E) staining. Treg/Th17 cells in the spleen were detected by flow cytometry. The levels of interleukin (IL)-10, IL-6 and IL-17A were detected by enzyme-linked immunosorbent assay (ELISA). AG490 attenuated DSS-induced IBD injury by regulating the Treg/Th17 balance and related cytokine secretion to reduce the DAI and colonic tissue damage. Thus, AG490 may be a new method for effective treatment of IBD.
Collapse
Affiliation(s)
- Jing Guo
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Li-Yun Wang
- Department of Neonatology, The First People's Hospital of Lianyungang/The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, China
| | - Jie Wu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ling-Fen Xu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mei Sun
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
27
|
Yahaya T, Salisu T. Genes predisposing to type 1 diabetes mellitus and pathophysiology: a narrative review. MEDICAL JOURNAL OF INDONESIA 2020. [DOI: 10.13181/mji.rev.203732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The possibility of targeting the causal genes along with the mechanisms of pathogenically complex diseases has led to numerous studies on the genetic etiology of some diseases. In particular, studies have added more genes to the list of type 1 diabetes mellitus (T1DM) suspect genes, necessitating an update for the interest of all stakeholders. Therefore this review articulates T1DM suspect genes and their pathophysiology. Notable electronic databases, including Medline, Scopus, PubMed, and Google-Scholar were searched for relevant information. The search identified over 73 genes suspected in the pathogenesis of T1DM, with human leukocyte antigen, insulin gene, and cytotoxic T lymphocyte-associated antigen 4 accounting for most of the cases. Mutations in these genes, along with environmental factors, may produce a defective immune response in the pancreas, resulting in β-cell autoimmunity, insulin deficiency, and hyperglycemia. The mechanisms leading to these cellular reactions are gene-specific and, if targeted in diabetic individuals, may lead to improved treatment. Medical practitioners are advised to formulate treatment procedures that target these genes in patients with T1DM.
Collapse
|
28
|
Ahmed CM, Ildefonso CJ, Johnson HM, Lewin AS. A C-terminal peptide from type I interferon protects the retina in a mouse model of autoimmune uveitis. PLoS One 2020; 15:e0227524. [PMID: 32101556 PMCID: PMC7043762 DOI: 10.1371/journal.pone.0227524] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 02/04/2020] [Indexed: 01/26/2023] Open
Abstract
Experimental autoimmune uveitis (EAU) in rodents recapitulates many features of the disease in humans and has served as a useful tool for the development of therapeutics. A peptide from C-terminus of interferon α1, conjugated to palmitoyl-lysine for cell penetration, denoted as IFNα-C, was tested for its anti-inflammatory properties in ARPE-19 cells, followed by testing in a mouse model of EAU. Treatment with IFNα-C and evaluation by RT-qPCR showed the induction of anti-inflammatory cytokines and chemokine. Inflammatory markers induced by treatment with TNFα were suppressed when IFNα-C was simultaneously present. TNF-α mediated induction of NF-κB and signaling by IL-17A were attenuated by IFNα-C. Differentiated ARPE-19 cells were treated with TNFα in the presence or absence IFNα-C and analyzed by immmunhistochemistry. IFNα-C protected against the disruption integrity of tight junction proteins. Similarly, loss of transepithelial resistance caused by TNFα was prevented by IFNα-C. B10.RIII mice were immunized with a peptide from interphotoreceptor binding protein (IRBP) and treated by gavage with IFNα-C. Development of uveitis was monitored by histology, fundoscopy, SD-OCT, and ERG. Treatment with IFNα-C prevented uveitis in mice immunized with the IRBP peptide. Splenocytes isolated from mice with ongoing EAU exhibited antigen-specific T cell proliferation that was inhibited in the presence of IFNα-C. IFNα-C peptide exhibits anti-inflammatory properties and protects mice against damage to retinal structure and function suggesting that it has therapeutic potential for the treatment of autoimmune uveitis.
Collapse
Affiliation(s)
- Chulbul M. Ahmed
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, United States of America
| | - Cristhian J. Ildefonso
- Department of Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | - Howard M. Johnson
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, United States of America
| | - Alfred S. Lewin
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, United States of America
| |
Collapse
|
29
|
Wolbert J, Cheng MI, Meyer zu Horste G, Su MA. Deciphering immune mechanisms in chronic inflammatory demyelinating polyneuropathies. JCI Insight 2020; 5:132411. [PMID: 32051341 DOI: 10.1172/jci.insight.132411] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chronic inflammatory demyelinating polyneuropathy (CIDP) is an autoimmune disease of the peripheral nerves that presents with either chronic progression or relapsing disease. Recent studies in samples from patients with CIDP and mouse models have delineated how defects in central (thymic) and peripheral (extrathymic) immune tolerance mechanisms can cause PNS autoimmunity. Notably, nerve parenchymal cells actively contribute to local autoimmunity and also control disease outcome. Here, we outline how emerging technologies increasingly enable an integrated view of how immune cells and PNS parenchymal cells communicate in CIDP. We also relate the known heterogeneity of clinical presentation with specific underlying mechanisms. For example, a severe subtype of CIDP with tremor is associated with pathogenic IgG4 autoantibodies against nodal and paranodal proteins. An improved understanding of pathogenic mechanisms in CIDP will form the basis for more effective mechanism-based therapies.
Collapse
Affiliation(s)
- Jolien Wolbert
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Mandy I Cheng
- Department of Microbiology Immunology and Medical Genetics and
| | - Gerd Meyer zu Horste
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Maureen A Su
- Department of Microbiology Immunology and Medical Genetics and.,Department of Pediatrics, UCLA, Los Angeles, California, USA
| |
Collapse
|
30
|
Goischke HK. Vitamin D supplementation for the prevention or depletion of side effects of therapy with alemtuzumab in multiple sclerosis. Ther Clin Risk Manag 2019; 15:891-904. [PMID: 31371976 PMCID: PMC6636607 DOI: 10.2147/tcrm.s188941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/16/2019] [Indexed: 12/22/2022] Open
Abstract
PURPOSE OF REVIEW Not only the multiple sclerosis specialist but also the general neurologist and primary care practitioner are increasingly aware of possible adverse events (AEs) by treatment with alemtuzumab (over 47% risk of secondary autoimmune-mediated diseases). Vitamin D supplementation's effect (VDS) to reduce these autoimmune AEs is poorly performed in routine practice. This article seeks to justify why this simple, inexpensive, patient-friendly therapy should be seriously discussed. RECENT FINDINGS Patients who have developed autoimmunity also show a high basal level of IL-21, a cytokine which increases the growth of auto-reactive T-cells. For side effects such as thyroid dysfunction, autoimmune thrombocytopenia, autoimmune hemolytic anemia, autoimmune hepatitis, diabetes mellitus type 1, and alopecia areata/alopecia totalis, VDS may have an impact on the immunological mechanism, in particular lowering levels of IL-17 and IL-21. SUMMARY The potential role of vitamin D in influencing autoimmune diseases is evident. If a life-threatening side-effect can be prevented by high-dose VDS, it is ethical to initiate this add-on therapy despite contradictory results in studies on the effectiveness of VDS.
Collapse
Affiliation(s)
- Hans-Klaus Goischke
- Independent Research, Internal Medicine, Rehabilitation Medicine, Social Medicine, Bad Brückenau, Bavaria, Germany
| |
Collapse
|
31
|
Bunte K, Beikler T. Th17 Cells and the IL-23/IL-17 Axis in the Pathogenesis of Periodontitis and Immune-Mediated Inflammatory Diseases. Int J Mol Sci 2019; 20:ijms20143394. [PMID: 31295952 PMCID: PMC6679067 DOI: 10.3390/ijms20143394] [Citation(s) in RCA: 298] [Impact Index Per Article: 59.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/11/2019] [Accepted: 07/08/2019] [Indexed: 12/12/2022] Open
Abstract
Innate immunity represents the semi-specific first line of defense and provides the initial host response to tissue injury, trauma, and pathogens. Innate immunity activates the adaptive immunity, and both act highly regulated together to establish and maintain tissue homeostasis. Any dysregulation of this interaction can result in chronic inflammation and autoimmunity and is thought to be a major underlying cause in the initiation and progression of highly prevalent immune-mediated inflammatory diseases (IMIDs) such as psoriasis, rheumatoid arthritis, inflammatory bowel diseases among others, and periodontitis. Th1 and Th2 cells of the adaptive immune system are the major players in the pathogenesis of IMIDs. In addition, Th17 cells, their key cytokine IL-17, and IL-23 seem to play pivotal roles. This review aims to provide an overview of the current knowledge about the differentiation of Th17 cells and the role of the IL-17/IL-23 axis in the pathogenesis of IMIDs. Moreover, it aims to review the association of these IMIDs with periodontitis and briefly discusses the therapeutic potential of agents that modulate the IL-17/IL-23 axis.
Collapse
Affiliation(s)
- Kübra Bunte
- Department of Periodontics, Preventive and Restorative Dentistry, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Thomas Beikler
- Department of Periodontics, Preventive and Restorative Dentistry, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany.
| |
Collapse
|
32
|
Mahmoud HM, Osman M, Elshabrawy O, Abdallah HMI, Khairallah A. AM-1241 CB2 Receptor Agonist Attenuates Inflammation, Apoptosis and Stimulate Progenitor Cells in Bile Duct Ligated Rats. Open Access Maced J Med Sci 2019; 7:925-936. [PMID: 30976335 PMCID: PMC6454175 DOI: 10.3889/oamjms.2019.194] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/19/2019] [Accepted: 02/20/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND: The cannabinoid receptor 2 (CB2) plays a pleiotropic role in the innate immunity and is considered a crucial mediator of liver disease. Cannabinoid CB2 receptor activation has been reported to attenuate liver fibrosis in CCl4 exposed mice and also plays a potential role in liver regeneration in a mouse model of I/R and protection against alcohol-induced liver injury. AIM: In this study, we investigated the impact of CB2 receptors on the antifibrotic and regenerative process associated with cholestatic liver injury. METHODS: Twenty-six rats had bile duct ligation co-treated with silymarin and AM1241 for 3 consecutive weeks. Serum hepatotoxicity markers were determined, and histopathological evaluation was performed. RESULTS: Following bile duct ligation (BDL) for 3 weeks, there was increased aminotransferase levels, marked inflammatory infiltration and hepatocyte apoptosis with induced oxidative stress, as reflected by increased lipid peroxidation. Conversely, following treatment with the CB2 agonist, AM-1241, BDL rats displayed a reduction in liver injury and attenuation of fibrosis as reflected by expression of hydroxyproline and α-smooth muscle actin. AM1241 treatment also significantly attenuated lipid peroxidation end-products, p53-dependent apoptosis and also attenuated inflammatory process by stimulating IL-10 production. Moreover, AM1241 treated rats were associated with significant expression of hepatic progenitor/oval cell markers. CONCLUSION: In conclusion, this study points out that CB2 receptors reduce liver injury and promote liver regeneration via distinct mechanisms including IL-10 dependent inhibition of inflammation, reduction of p53-reliant apoptosis and through stimulation of oval/progenitor cells. These results suggest that CB2 agonists display potent hepatoregenrative properties, in addition to their antifibrogenic effects.
Collapse
Affiliation(s)
- Hesham M Mahmoud
- Cairo University Kasr Alainy, Faculty of Medicine, Pharmacology, Cairo, Egypt
| | - Mona Osman
- Cairo University Kasr Alainy, Faculty of Medicine, Pharmacology, Cairo, Egypt
| | | | | | - Ahmed Khairallah
- Pharmacology Department, National Research Centre, Dokki, Cairo 11211, Egypt
| |
Collapse
|
33
|
Dimitrijević M, Arsenović-Ranin N, Kosec D, Bufan B, Nacka-Aleksić M, Pilipović I, Leposavić G. Sexual dimorphism in Th17/Treg axis in lymph nodes draining inflamed joints in rats with collagen-induced arthritis. Brain Behav Immun 2019; 76:198-214. [PMID: 30476564 DOI: 10.1016/j.bbi.2018.11.311] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/22/2018] [Accepted: 11/22/2018] [Indexed: 01/11/2023] Open
Abstract
Collagen type II-induced arthritis (CIA) in Dark Agouti rats, a model of rheumatoid arthritis (RA), reproduces sexual dimorphism in the incidence and severity of the human disease. Th17 cells are central in the induction/propagation of autoimmune inflammation in CIA and RA. To assess mechanisms underlying this dimorphism in CIA rats, in lymph nodes draining inflamed joints and adjacent tissues (dLNs) from CIA rats of both sexes Th17/CD25+Foxp3+CD4+ T-regulatory cell (Treg) ratio, Th17 cell redifferentiation in functionally distinct subsets and Treg transdifferentiation into IL-17-producing cells (exTregs) were examined. In female rats (developing more severe CIA than their male counterparts) the higher frequency of all Th17 cells (reflecting partly their greater proliferation), followed by the higher frequency of highly pathogenic IFN-γ/GM-CSF-co-producing cells, but lower frequency of less pathogenic/immunoregulatory IL-10-producing cells among them was found. Additionally, compared with male rats, in female rats the lower frequency of Tregs was observed. Moreover, Tregs from female rats exhibited diminished proliferative and suppressive capacity (judging by PD-1 expression) and enhanced conversion into IL-17-producing cells. Given that TGF-β concentration was comparable in collagen-type II-stimulated dLN cell cultures from female and male rats, the shift in Th17/Treg ratio followed by augmented Th17 cell redifferentiation into IFN-γ/GM-CSF-co-producing cells and Treg transdifferentiation into IL-17-producing cells in female rats was associated with increased concentration of IL-6 in female rat dLN cell cultures, and the higher frequency of IL-1β- and IL-23-producing cells among their dLN cells. The lower frequency of IL-10-producing B cells, presumably B regulatory cells (Bregs) could also contribute to the shift in Th17/Treg ratio in female rat compared with male rat dLNs. Consistently, the lower expression of IL-35 (the cytokine promoting Treg expansion directly and indirectly, by favoring Breg expansion and conversion into IL-10/IL-35-producing cells) in female rat dLN cells was detected. Thus, the study identified putative cellular and molecular substrates of the sexual dimorphism in the immunopathogenesis and clinical outcome of CIA and suggested mechanisms to be targeted in females to improve control of Th17 response, and consequently clinical outcome of CIA, and possibly RA.
Collapse
Affiliation(s)
- Mirjana Dimitrijević
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar despota Stefana 142, Belgrade, Serbia.
| | - Nevena Arsenović-Ranin
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, Belgrade, Serbia
| | - Duško Kosec
- Immunology Research Center, Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Vojvode Stepe 458, Belgrade, Serbia
| | - Biljana Bufan
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, Belgrade, Serbia
| | - Mirjana Nacka-Aleksić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, Belgrade, Serbia
| | - Ivan Pilipović
- Immunology Research Center, Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Vojvode Stepe 458, Belgrade, Serbia
| | - Gordana Leposavić
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, Belgrade, Serbia.
| |
Collapse
|
34
|
Olive Oil Effects on Colorectal Cancer. Nutrients 2018; 11:nu11010032. [PMID: 30583613 PMCID: PMC6357067 DOI: 10.3390/nu11010032] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 12/11/2018] [Accepted: 12/15/2018] [Indexed: 02/08/2023] Open
Abstract
Colorectal cancer is the fourth cause of cancer-related death worldwide. A Mediterranean diet showed protective action against colorectal cancer due to the intake of different substances. Olive oil is a fundamental component of the Mediterranean diet. Olive oil is rich in high-value health compounds (such as monounsaturated free fatty acids, squalene, phytosterols, and phenols). Phenolic compounds exert favourable effects on free radicals, inflammation, gut microbiota, and carcinogenesis. The interaction between gut microbiota and olive oil consumption could modulate colonic microbial composition or activity, with a possible role in cancer prevention. Gut microbiota is able to degrade some substances found in olive oil, producing active metabolites with chemopreventive action. Further clinical research is needed to clarify the beneficial effects of olive oil and its components. A better knowledge of the compounds found in olive oil could lead to the development of nutritional supplements or chemotherapeutic agents with a potential in the prevention and treatment of colorectal cancer.
Collapse
|
35
|
Torabi S, Tamaddon M, Asadolahi M, Shokri G, Tavakoli R, Tasharrofi N, Rezaei R, Tavakolpour V, Sazegar H, Kouhkan F. miR-455-5p downregulation promotes inflammation pathways in the relapse phase of relapsing-remitting multiple sclerosis disease. Immunogenetics 2018; 71:87-95. [DOI: 10.1007/s00251-018-1087-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 09/21/2018] [Indexed: 01/01/2023]
|
36
|
Napier RJ, Lee EJ, Vance EE, Snow PE, Samson KA, Dawson CE, Moran AE, Stenzel P, Davey MP, Sakaguchi S, Rosenzweig HL. Nod2 Deficiency Augments Th17 Responses and Exacerbates Autoimmune Arthritis. THE JOURNAL OF IMMUNOLOGY 2018; 201:1889-1898. [PMID: 30150283 DOI: 10.4049/jimmunol.1700507] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 07/27/2018] [Indexed: 12/11/2022]
Abstract
Arthritis in a genetically susceptible SKG strain of mice models a theoretical paradigm wherein autoimmune arthritis arises because of interplay between preexisting autoreactive T cells and environmental stimuli. SKG mice have a point mutation in ZAP-70 that results in attenuated TCR signaling, altered thymic selection, and spontaneous production of autoreactive T cells that cause arthritis following exposure to microbial β-glucans. In this study, we identify Nod2, an innate immune receptor, as a critical suppressor of arthritis in SKG mice. SKG mice deficient in Nod2 (Nod2-/-SKG) developed a dramatically exacerbated form of arthritis, which was independent of sex and microbiota, but required the skg mutation in T cells. Worsened arthritis in Nod2-/-SKG mice was accompanied by expansion of Th17 cells, which to some measure coproduced TNF, GM-CSF, and IL-22, along with elevated IL-17A levels within joint synovial fluid. Importantly, neutralization of IL-17A mitigated arthritis in Nod2-/-SKG mice, indicating that Nod2-mediated protection occurs through suppression of the Th17 response. Nod2 deficiency did not alter regulatory T cell development or function. Instead, Nod2 deficiency resulted in an enhanced fundamental ability of SKG CD4+ T cells (from naive mice) to produce increased levels of IL-17 and to passively transfer arthritis to lymphopenic recipients on a single-cell level. These data reveal a previously unconsidered role for T cell-intrinsic Nod2 as an endogenous negative regulator of Th17 responses and arthritogenic T cells. Based on our findings, future studies aimed at understanding a negative regulatory function of Nod2 within autoreactive T cells could provide novel therapeutic strategies for treatment of patients with arthritis.
Collapse
Affiliation(s)
- Ruth J Napier
- Veterans Affairs Portland Health Care System, Portland, OR 97239.,Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239
| | - Ellen J Lee
- Veterans Affairs Portland Health Care System, Portland, OR 97239.,Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239
| | - Emily E Vance
- Veterans Affairs Portland Health Care System, Portland, OR 97239.,Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239
| | - Paige E Snow
- Veterans Affairs Portland Health Care System, Portland, OR 97239
| | - Kimberly A Samson
- Veterans Affairs Portland Health Care System, Portland, OR 97239.,Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239
| | - Clare E Dawson
- Veterans Affairs Portland Health Care System, Portland, OR 97239
| | - Amy E Moran
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239
| | - Peter Stenzel
- Department of Anatomic Pathology, Oregon Health & Science University, Portland, OR 97239
| | - Michael P Davey
- Veterans Affairs Portland Health Care System, Portland, OR 97239.,Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239.,Department of Medicine, Oregon Health & Science University, Portland, OR 97239; and
| | | | - Holly L Rosenzweig
- Veterans Affairs Portland Health Care System, Portland, OR 97239; .,Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239
| |
Collapse
|
37
|
Ai G, Yan W, Yu H, Xiao F, Xi D, Ma K, Huang J, Luo X, Wan X, Ning Q. Soluble Fgl2 restricts autoimmune hepatitis progression via suppressing Tc17 and conventional CD8+ T cell function. J Gene Med 2018; 20:e3023. [PMID: 29756667 DOI: 10.1002/jgm.3023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 04/17/2018] [Accepted: 05/03/2018] [Indexed: 12/28/2022] Open
Affiliation(s)
- Guo Ai
- Institute of Infectious Disease, Tongji Hospital of Tongji Medical College; Wuhan China
| | - Weiming Yan
- Institute of Infectious Disease, Tongji Hospital of Tongji Medical College; Wuhan China
| | - Haijing Yu
- Department of Infectious Disease, Tongji Hospital of Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| | - Fang Xiao
- Institute of Infectious Disease, Tongji Hospital of Tongji Medical College; Wuhan China
| | - Dong Xi
- Institute of Infectious Disease, Tongji Hospital of Tongji Medical College; Wuhan China
| | - Ke Ma
- Department of Infectious Disease, Tongji Hospital of Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| | - Jiaquan Huang
- Department of Infectious Disease, Tongji Hospital of Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| | - Xiaoping Luo
- Department of Pediatrics, Tongji Hospital of Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| | - Xiaoyang Wan
- Institute of Infectious Disease, Tongji Hospital of Tongji Medical College; Wuhan China
| | - Qin Ning
- Institute of Infectious Disease, Tongji Hospital of Tongji Medical College; Wuhan China
- Department of Infectious Disease, Tongji Hospital of Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| |
Collapse
|
38
|
Iacobaeus E, Douagi I, Jitschin R, Marcusson-Ståhl M, Andrén AT, Gavin C, Lefsihane K, Davies LC, Mougiakakos D, Kadri N, Le Blanc K. Phenotypic and functional alterations of myeloid-derived suppressor cells during the disease course of multiple sclerosis. Immunol Cell Biol 2018; 96:820-830. [PMID: 29569304 DOI: 10.1111/imcb.12042] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 03/15/2018] [Accepted: 03/15/2018] [Indexed: 12/20/2022]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system involving dysregulated encephalitogenic T cells. Myeloid-derived suppressor cells (MDSCs) have been recognized for their important function in regulating T-cell responses. Recent studies have indicated a role for MDSCs in autoimmune diseases, but their significance in MS is not clear. Here, we assessed the frequencies of CD14+ HLA-DRlow monocytic MDSCs (Mo-MDSCs) and CD33+ CD15+ CD11b+ HLA-DRlow granulocytic MDSCs (Gr-MDSCs) and investigated phenotypic and functional differences of Mo-MDSCs at different clinical stages of MS and in healthy subjects (HC). Increased frequencies of Mo-MDSCs (P < 0.05) and Gr-MDSCs (P < 0.05) were observed in relapsing-remitting MS patients during relapse (RRMS-relapse) compared to stable RRMS (RRMS-rem). Secondary progressive MS (SPMS) patients displayed a decreased frequency of Mo-MDSCs and Gr-MDSCs compared to HC (P < 0.05). Mo-MDSCs within RRMS patients expressed significantly higher cell surface protein levels of CD86 and CD163 compared to SPMS patients. Mo-MDSCs within SPMS exhibited decreased mRNA expression of interleukin-10 and heme oxygenase 1 compared to RRMS and HC. Analysis of T-cell regulatory function of Mo-MDSCs demonstrated T-cell suppressive capacity in RRMS and HCs, while Mo-MDSCs of SPMS promoted autologous T-cell proliferation, which aligned with a differential cytokine profile compared to RRMS and HCs. This study is the first to show phenotypic and functional shifts of MDSCs between clinical stages of MS, suggesting a role for MDSCs as a therapeutic target to prevent MS disease progression.
Collapse
Affiliation(s)
- Ellen Iacobaeus
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.,Division of Neurology, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Iyadh Douagi
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institute, Stockholm, Sweden
| | - Regina Jitschin
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Maritha Marcusson-Ståhl
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Anton Törnqvist Andrén
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Caroline Gavin
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Katia Lefsihane
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Lindsay C Davies
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Dimitrios Mougiakakos
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Nadir Kadri
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Katarina Le Blanc
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
39
|
Martin NM, Griffin DE. Interleukin-10 Modulation of Virus Clearance and Disease in Mice with Alphaviral Encephalomyelitis. J Virol 2018; 92:e01517-17. [PMID: 29263262 PMCID: PMC5827374 DOI: 10.1128/jvi.01517-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/11/2017] [Indexed: 02/07/2023] Open
Abstract
Alphaviruses are an important cause of mosquito-borne outbreaks of arthritis, rash, and encephalomyelitis. Previous studies in mice with a virulent strain (neuroadapted SINV [NSV]) of the alphavirus Sindbis virus (SINV) identified a role for Th17 cells and regulation by interleukin-10 (IL-10) in the pathogenesis of fatal encephalomyelitis (K. A. Kulcsar, V. K. Baxter, I. P. Greene, and D. E. Griffin, Proc Natl Acad Sci U S A 111:16053-16058, 2014, https://doi.org/10.1073/pnas.1418966111). To determine the role of virus virulence in generation of immune responses, we analyzed the modulatory effects of IL-10 on disease severity, virus clearance, and the CD4+ T cell response to infection with a recombinant strain of SINV of intermediate virulence (TE12). The absence of IL-10 during TE12 infection led to longer morbidity, more weight loss, higher mortality, and slower viral clearance than in wild-type mice. More severe disease and impaired virus clearance in IL-10-/- mice were associated with more Th1 cells, fewer Th2 cells, innate lymphoid type 2 cells, regulatory cells, and B cells, and delayed production of antiviral antibody in the central nervous system (CNS) without an effect on Th17 cells. Therefore, IL-10 deficiency led to more severe disease in TE12-infected mice by increasing Th1 cells and by hampering development of the local B cell responses necessary for rapid production of antiviral antibody and virus clearance from the CNS. In addition, the shift from Th17 to Th1 responses with decreased virus virulence indicates that the effects of IL-10 deficiency on immunopathologic responses in the CNS during alphavirus infection are influenced by virus strain.IMPORTANCE Alphaviruses cause mosquito-borne outbreaks of encephalomyelitis, but determinants of outcome are incompletely understood. We analyzed the effects of the anti-inflammatory cytokine IL-10 on disease severity and virus clearance after infection with an alphavirus strain of intermediate virulence. The absence of IL-10 led to longer illness, more weight loss, more death, and slower viral clearance than in mice that produced IL-10. IL-10 influenced development of disease-causing T cells and entry into the brain of B cells producing antiviral antibody. The Th1 pathogenic cell subtype that developed in IL-10-deficient mice infected with a less virulent virus was distinct from the Th17 subtype that developed in response to a more virulent virus, indicating a role for virus strain in determining the immune response. Slow production of antibody in the nervous system led to delayed virus clearance. Therefore, both the virus strain and the host response to infection are important determinants of outcome.
Collapse
Affiliation(s)
- Nina M Martin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Diane E Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
40
|
Wang W, Wang Z, Qin Y, Tang G, Cai G, Liu Y, Zhang J, Zhang P, Shen Q, Shen L, Yu W. Th17, synchronically increased with T regs and B regs , promoted by tumour cells via cell-contact in primary hepatic carcinoma. Clin Exp Immunol 2018; 192:181-192. [PMID: 29271479 DOI: 10.1111/cei.13094] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 12/09/2017] [Accepted: 12/13/2017] [Indexed: 01/14/2023] Open
Abstract
Documented reports about T helper type 17 (Th17) cells have revealed that Th17 plays a critical role in inflammation and autoimmunity diseases. However, the role of Th17 in cancer remains contradictory. The interplay between Th17 and tumour cells in the tumour microenvironment of primary hepatic carcinoma (PHC) needs to be explored further and the relationship between Th17, regulatory T cells (Tregs ) and regulatory B cells (Bregs ) has not been defined completely. In this study, numerous experiments were undertaken to elucidate the interaction of Th17 and Treg /Breg cells involved in PHC. Our work demonstrated that an increased Th17 was detected in the peripheral circulation and in tumour tissues in PHC patients. In addition, increases in peripheral blood Th17 corresponded with tumour-node-metastasis (TNM) stage progression. Also, further studies indicated that Th17 cells were promoted by tumour cells in the PHC tumour microenvironment through both contact-dependent and -independent mechanisms, but cell-contact played the major important role in promoting the production and proliferation of Th17. When isolated CD4+ CD25+ CD127low Tregs and CD4+ CD25- CD127+ non-Tregs were cultured with autologous tumour cells, it implied that the phenotype of Th17 and Tregs was modified by tumour cells in the tumour microenvironment. As well as this, Th17 cells were also found to correlate positively with CD4+ forkhead box protein 3+ Tregs and CD19+ CD5+ CD1dhi Bregs in PHC. Notably, Th17 increased synchronically with Tregs and Bregs in PHC. These findings may provide new clues to reveal the mechanisms of immune escape in PHC.
Collapse
Affiliation(s)
- W Wang
- Department of Clinical Laboratory, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Z Wang
- Department of Anesthesia, Eastern Hepatobiliary Hospital, Shanghai, China
| | - Y Qin
- Department of Laboratory Diagnosis, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - G Tang
- Department of Laboratory Diagnosis, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - G Cai
- Department of Laboratory Diagnosis, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Y Liu
- Department of Laboratory Diagnosis, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - J Zhang
- Department of Laboratory Diagnosis, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - P Zhang
- Department of Laboratory Diagnosis, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Q Shen
- Department of Laboratory Diagnosis, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - L Shen
- Department of Clinical Laboratory, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - W Yu
- Department of Anesthesia, Eastern Hepatobiliary Hospital, Shanghai, China
| |
Collapse
|
41
|
Zhou Y, Leng X, He Y, Li Y, Liu Y, Liu Y, Zou Q, Shi G, Wang Y. Loss of Perp in T Cells Promotes Resistance to Apoptosis of T Helper 17 Cells and Exacerbates the Development of Experimental Autoimmune Encephalomyelitis in Mice. Front Immunol 2018; 9:842. [PMID: 29740445 PMCID: PMC5925960 DOI: 10.3389/fimmu.2018.00842] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 04/05/2018] [Indexed: 01/03/2023] Open
Abstract
T helper 17 (Th17) cells are crucial for the pathogenesis of multiple sclerosis (MS) in humans and experimental autoimmune encephalomyelitis (EAE) in animals. High frequency of Th17 cells and low sensitivity to activation-induced cell death (AICD) are detected in MS patients. However, the mechanisms underlying apoptosis resistance of T cells remain unclear. Perp is an apoptosis-associated target of p53 and implicated in the development of cancers. Here, we show that loss of Perp in T cells does not affect Th1, Th17, or Treg cell differentiation, but does significantly increase the resistance of Perp-/- Th17 cells to AICD and anti-Fas in Lck-Cre × Perpfl/fl mice by inhibiting the caspase-dependent apoptotic pathway. Moreover, Lck-Cre × Perpfl/fl mice exhibited earlier onset of EAE and severe spinal cord inflammation and demyelination, accompanied by increased levels of pro-inflammatory cytokines and enlarged population of Th17 cells. Therefore, Perp deletion promoted Th17 responses and exacerbated the development and severity of EAE.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Emergency, West China Second University Hospital and Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Xiao Leng
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Yan He
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Xiamen University, Xiamen, China
| | - Yan Li
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Xiamen University, Xiamen, China
| | - Yuan Liu
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Xiamen University, Xiamen, China
| | - Yang Liu
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Qiang Zou
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Guixiu Shi
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Xiamen University, Xiamen, China
- *Correspondence: Guixiu Shi, ; Yantang Wang,
| | - Yantang Wang
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
- *Correspondence: Guixiu Shi, ; Yantang Wang,
| |
Collapse
|
42
|
Genc B, Bozan HR, Genc S, Genc K. Stem Cell Therapy for Multiple Sclerosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1084:145-174. [PMID: 30039439 DOI: 10.1007/5584_2018_247] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory, autoimmune, and neurodegenerative disease of the central nervous system (CNS). It is characterized by demyelination and neuronal loss that is induced by attack of autoreactive T cells to the myelin sheath and endogenous remyelination failure, eventually leading to functional neurological disability. Although recent evidence suggests that MS relapses are induced by environmental and exogenous triggers such as viral infections in a genetic background, its very complex pathogenesis is not completely understood. Therefore, the efficiency of current immunosuppression-based therapies of MS is too low, and emerging disease-modifying immunomodulatory agents such as fingolimod and dimethyl fumarate cannot stop progressive neurodegenerative process. Thus, the cell replacement therapy approach that aims to overcome neuronal cell loss and remyelination failure and to increase endogenous myelin repair capacity is considered as an alternative treatment option. A wide variety of preclinical studies, using experimental autoimmune encephalomyelitis model of MS, have recently shown that grafted cells with different origins including mesenchymal stem cells (MSCs), neural precursor and stem cells, and induced-pluripotent stem cells have the ability to repair CNS lesions and to recover functional neurological deficits. The results of ongoing autologous hematopoietic stem cell therapy studies, with the advantage of peripheral administration to the patients, have suggested that cell replacement therapy is also a feasible option for immunomodulatory treatment of MS. In this chapter, we overview cell sources and applications of the stem cell therapy for treatment of MS. We also discuss challenges including those associated with administration route, immune responses to grafted cells, integration of these cells to existing neural circuits, and risk of tumor growth. Finally, future prospects of stem cell therapy for MS are addressed.
Collapse
Affiliation(s)
- Bilgesu Genc
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Turkey
| | - Hemdem Rodi Bozan
- School of Medicine, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Sermin Genc
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey.,Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Kursad Genc
- Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University Health Campus, Izmir, Turkey.
| |
Collapse
|
43
|
Banerjee A, Bhattacharya P, Dagur PK, Karmakar S, Ismail N, Joshi AB, Akue AD, KuKuruga M, McCoy JP, Dey R, Nakhasi HL. Live Attenuated Leishmania donovani Centrin Gene-Deleted Parasites Induce IL-23-Dependent IL-17-Protective Immune Response against Visceral Leishmaniasis in a Murine Model. THE JOURNAL OF IMMUNOLOGY 2017; 200:163-176. [PMID: 29187586 DOI: 10.4049/jimmunol.1700674] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 10/26/2017] [Indexed: 12/18/2022]
Abstract
No vaccine exists against visceral leishmaniasis. To develop effective vaccines, we have previously reported protective role of live attenuated centrin gene-deleted Leishmania donovani (LdCen-/- ) parasites through induction of Th1 type immune response in mice, hamsters, and dogs. In this study, we specifically explored the role of Th17 cells in LdCen-/- -induced host protection in mice. Our results showed that compared with wild-type L. donovani infection, LdCen-/- parasites induce significantly higher expression of Th17 differentiation cytokines in splenic dendritic cells. There was also induction of IL-17 and its promoting cytokines in total splenocytes and in both CD4 and CD8 T cells following immunization with LdCen-/- Upon challenge with wild-type parasites, IL-17 and its differentiating cytokines were significantly higher in LdCen-/- -immunized mice compared with nonimmunized mice that resulted in parasite control. Alongside IL-17 induction, we observed induction of IFN-γ-producing Th1 cells as reported earlier. However, Th17 cells are generated before Th1 cells. Neutralization of either IL-17 or IFN-γ abrogated LdCen-/- -induced host protection further confirming the essential role of Th17 along with Th1 cytokines in host protection. Treatment with recombinant IL-23, which is required for stabilization and maintenance of IL-17, heightened Th17, and Tc17 responses in immunized mice splenocytes. In contrast, Th17 response was absent in immunized IL-23R-/- mice that failed to induce protection upon virulent Leishmania challenge suggesting that IL-23 plays an essential role in IL-17-mediated protection by LdCen-/- parasites. This study unveiled the role of IL-23-dependent IL-17 induction in LdCen-/- parasite-induced immunity and subsequent protection against visceral leishmaniasis.
Collapse
Affiliation(s)
- Antara Banerjee
- Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993.,Department of Zoology, Bangabasi College, Kolkata, 700016 West Bengal, India
| | - Parna Bhattacharya
- Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993
| | - Pradeep K Dagur
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Subir Karmakar
- Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993
| | - Nevien Ismail
- Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993
| | - Amritanshu B Joshi
- Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993
| | - Adovi D Akue
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993
| | - Mark KuKuruga
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993
| | - John Philip McCoy
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Ranadhir Dey
- Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993;
| | - Hira L Nakhasi
- Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993;
| |
Collapse
|
44
|
Hu D, Notarbartolo S, Croonenborghs T, Patel B, Cialic R, Yang TH, Aschenbrenner D, Andersson KM, Gattorno M, Pham M, Kivisakk P, Pierre IV, Lee Y, Kiani K, Bokarewa M, Tjon E, Pochet N, Sallusto F, Kuchroo VK, Weiner HL. Transcriptional signature of human pro-inflammatory T H17 cells identifies reduced IL10 gene expression in multiple sclerosis. Nat Commun 2017; 8:1600. [PMID: 29150604 PMCID: PMC5693957 DOI: 10.1038/s41467-017-01571-8] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 09/29/2017] [Indexed: 01/08/2023] Open
Abstract
We have previously reported the molecular signature of murine pathogenic TH17 cells that induce experimental autoimmune encephalomyelitis (EAE) in animals. Here we show that human peripheral blood IFN-γ+IL-17+ (TH1/17) and IFN-γ−IL-17+ (TH17) CD4+ T cells display distinct transcriptional profiles in high-throughput transcription analyses. Compared to TH17 cells, TH1/17 cells have gene signatures with marked similarity to mouse pathogenic TH17 cells. Assessing 15 representative signature genes in patients with multiple sclerosis, we find that TH1/17 cells have elevated expression of CXCR3 and reduced expression of IFNG, CCL3, CLL4, GZMB, and IL10 compared to healthy controls. Moreover, higher expression of IL10 in TH17 cells is found in clinically stable vs. active patients. Our results define the molecular signature of human pro-inflammatory TH17 cells, which can be used to both identify pathogenic TH17 cells and to measure the effect of treatment on TH17 cells in human autoimmune diseases. CD4+ T cells secreting interleukin-17 (TH17) have diverse functions in modulating autoimmune diseases. Here the authors show via transcriptome analyses that a subset of human TH 17 co-expressing interferon-γ (TH1/17) has a molecular signature similar to “pathogenic” mouse TH 17 but distinct from “non-pathogenic” mouse TH 17.
Collapse
Affiliation(s)
- Dan Hu
- Ann Romney Center for Neurologic Diseases and Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Samuele Notarbartolo
- Institute for Research in Biomedicine, Università della Svizzera italiana, via Vincenzo Vela 6, CH-6500, Bellinzona, Switzerland
| | - Tom Croonenborghs
- Program in Translational NeuroPsychiatric Genomics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,KU Leuven Technology Campus Geel, AdvISe, Kleinhoefstraat 4, 2440, Geel, Belgium.,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - Bonny Patel
- Ann Romney Center for Neurologic Diseases and Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Ron Cialic
- Ann Romney Center for Neurologic Diseases and Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Tun-Hsiang Yang
- Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA
| | - Dominik Aschenbrenner
- Institute for Research in Biomedicine, Università della Svizzera italiana, via Vincenzo Vela 6, CH-6500, Bellinzona, Switzerland.,Translational Gastroenterology Unit, NDM Experimental Medicine, University of Oxford, Headington, OX3 9DU, UK
| | - Karin M Andersson
- Department of Rheumatology and Inflammation Research, Sahlgrenska University Hospital, Gothenburg University, Box 480, 405 30, Gothenburg, Sweden
| | - Marco Gattorno
- Second Division of Pediatrics, G. Gaslini Scientific Institute, Largo Gerolamo Gaslini, 5, 16100, Genova(GE), Italy
| | - Minh Pham
- Ann Romney Center for Neurologic Diseases and Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Pia Kivisakk
- Ann Romney Center for Neurologic Diseases and Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Isabelle V Pierre
- Ann Romney Center for Neurologic Diseases and Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Youjin Lee
- Ann Romney Center for Neurologic Diseases and Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Karun Kiani
- Program in Translational NeuroPsychiatric Genomics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Maria Bokarewa
- Department of Rheumatology and Inflammation Research, Sahlgrenska University Hospital, Gothenburg University, Box 480, 405 30, Gothenburg, Sweden
| | - Emily Tjon
- Ann Romney Center for Neurologic Diseases and Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Nathalie Pochet
- Program in Translational NeuroPsychiatric Genomics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera italiana, via Vincenzo Vela 6, CH-6500, Bellinzona, Switzerland.,Institute of Microbiology, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093, Zürich, Switzerland
| | - Vijay K Kuchroo
- Ann Romney Center for Neurologic Diseases and Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases and Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
45
|
Kantono M, Guo B. Inflammasomes and Cancer: The Dynamic Role of the Inflammasome in Tumor Development. Front Immunol 2017; 8:1132. [PMID: 28955343 PMCID: PMC5600922 DOI: 10.3389/fimmu.2017.01132] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 08/28/2017] [Indexed: 12/20/2022] Open
Abstract
Chronic Inflammation in tumor microenvironments is not only associated with various stages of tumor development, but also has significant impacts on tumor immunity and immunotherapy. Inflammasome are an important innate immune pathway critical for the production of active IL-1β and interleukin 18, as well as the induction of pyroptosis. Although extensive studies have demonstrated that inflammasomes play a vital role in infectious and autoimmune diseases, their role in tumor progression remains elusive. Multiple studies using a colitis-associated colon cancer model show that inflammasome components provide protection against the development of colon cancer. However, very recent studies demonstrate that inflammasomes promote tumor progression in skin and breast cancer. These results indicate that inflammasomes can promote and suppress tumor development depending on types of tumors, specific inflammasomes involved, and downstream effector molecules. The complicated role of inflammasomes raises new opportunities and challenges to manipulate inflammasome pathways in the treatment of cancer.
Collapse
Affiliation(s)
- Melvin Kantono
- Department of Microbiology and Immunology, Medical University of South Carolina (MUSC), Charleston, SC, United States.,Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC, United States
| | - Beichu Guo
- Department of Microbiology and Immunology, Medical University of South Carolina (MUSC), Charleston, SC, United States.,Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC, United States
| |
Collapse
|
46
|
Vultaggio A, Nencini F, Pratesi S, Cammelli D, Totaro M, Romagnani S, Maggi E, Matucci A. IL-10–Producing Infliximab-Specific T Cells Regulate the Antidrug T Cell Response in Exposed Patients. THE JOURNAL OF IMMUNOLOGY 2017; 199:1283-1289. [DOI: 10.4049/jimmunol.1700008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 06/09/2017] [Indexed: 12/29/2022]
|
47
|
Nacka-Aleksić M, Stojić-Vukanić Z, Pilipović I, Vujnović I, Bufan B, Dimitrijević M, Leposavić G. Strain specificities in cellular and molecular immunopathogenic mechanisms underlying development of experimental autoimmune encephalomyelitis in aged rats. Mech Ageing Dev 2017; 164:146-163. [DOI: 10.1016/j.mad.2017.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 02/21/2017] [Accepted: 03/03/2017] [Indexed: 11/29/2022]
|
48
|
Paiatto LN, Silva FGD, Bier J, Brochetto-Braga MR, Yamada ÁT, Tamashiro WMSC, Simioni PU. Oral Tolerance Induced by OVA Intake Ameliorates TNBS-Induced Colitis in Mice. PLoS One 2017; 12:e0170205. [PMID: 28099498 PMCID: PMC5242488 DOI: 10.1371/journal.pone.0170205] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/01/2017] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Literature data have shown that the consumption of dietary proteins may cause modulatory effects on the host immune system, process denominated oral tolerance by bystander suppression. It has been shown that the bystander suppression induced by dietary proteins can improve inflammatory diseases such as experimental arthritis. Here, we evaluated the effects of oral tolerance induced by ingestion of ovalbumin (OVA) on TNBS-induced colitis in mice, an experimental model for human Crohn's disease. METHODS AND RESULTS Colitis was induced in BALB/c mice by instilling a single dose of TNBS (100 mg/kg) in ethanol into the colon. Tolerized mice received OVA (4mg/mL) dissolved in the drinking water for seven consecutive days, prior to or concomitantly with the intrarectal instillation. Control groups received protein-free water and ethanol by intrarectal route. We observed that either the prior or concomitant induction of oral tolerance were able to reduce the severity of colitis as noted by recovery of body weight gain, improvement of clinical signs and reduction of histological abnormalities. The in vitro proliferation of spleen cells from tolerant colitic mice was lower than that of control mice, the same as the frequencies of CD4+ T cells secreting IL-17 and IFN-γ. The frequencies of regulatory T cells and T cells secreting IL-10 have increased significantly in mice orally treated with OVA. The levels of inflammatory cytokines (IL-17A, TNF-α, IL-6 and IFN-γ) were lower in supernatants of cells from tolerant colitic mice, whereas IL-10 levels were higher. CONCLUSION Our data show that the modulation of immune response induced by oral tolerance reduces the severity of experimental colitis. Such modulation may be partially attributed to the increase of Treg cells and reduction of pro-inflammatory cytokines in peripheral lymphoid organs of tolerant mice by bystander suppression.
Collapse
Affiliation(s)
- Lisiery N. Paiatto
- Department of Genetics, Evolution and Bioagents, Institute of Biology, State University of Campinas, UNICAMP, Campinas, São Paulo, Brazil
- Institute of Biosciences, Universidade Estadual Paulista, UNESP, Rio Claro, São Paulo, Brazil
| | - Fernanda G. D. Silva
- Faculty of Food Engineering, University of Campinas, UNICAMP, Campinas, São Paulo, Brazil
| | - Julia Bier
- Department of Genetics, Evolution and Bioagents, Institute of Biology, State University of Campinas, UNICAMP, Campinas, São Paulo, Brazil
| | | | - Áureo T. Yamada
- Department of Histology and Embryology, Institute of Biology, State University of Campinas, UNICAMP, Campinas, São Paulo, Brazil
| | - Wirla M. S. C. Tamashiro
- Department of Genetics, Evolution and Bioagents, Institute of Biology, State University of Campinas, UNICAMP, Campinas, São Paulo, Brazil
| | - Patricia U. Simioni
- Department of Genetics, Evolution and Bioagents, Institute of Biology, State University of Campinas, UNICAMP, Campinas, São Paulo, Brazil
- Institute of Biosciences, Universidade Estadual Paulista, UNESP, Rio Claro, São Paulo, Brazil
- Department of Biomedical Science, Faculty of Americana, FAM, Americana, São Paulo, Brazil
| |
Collapse
|
49
|
Stanisavljević S, Lukić J, Soković S, Mihajlovic S, Mostarica Stojković M, Miljković D, Golić N. Correlation of Gut Microbiota Composition with Resistance to Experimental Autoimmune Encephalomyelitis in Rats. Front Microbiol 2016; 7:2005. [PMID: 28018327 PMCID: PMC5156687 DOI: 10.3389/fmicb.2016.02005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 11/30/2016] [Indexed: 12/28/2022] Open
Abstract
Multiple sclerosis is a chronic inflammatory disease of the central nervous system (CNS). It is widely accepted that autoimmune response against the antigens of the CNS is the essential pathogenic force in the disease. It has recently become increasingly appreciated that activated encephalitogenic cells tend to migrate toward gut associated lymphoid tissues (GALTs) and that interrupted balance between regulatory and inflammatory immunity within the GALT might have decisive role in the initiation and propagation of the CNS autoimmunity. Gut microbiota composition and function has the major impact on the balance in the GALT. Thus, our aim was to perform analyses of gut microbiota in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. Albino Oxford (AO) rats that are highly resistant to EAE induction and Dark Agouti (DA) rats that develop EAE after mild immunization were compared for gut microbiota composition in different phases after EAE induction. Microbial analyses of the genus Lactobacillus and related lactic acid bacteria showed higher diversity of Lactobacillus spp. in EAE-resistant AO rats, while some members of Firmicutes and Proteobacteria (Undibacterium oligocarboniphilum) were detected only in feces of DA rats at the peak of the disease (between 13 and 16 days after induction). Interestingly, in contrast to our previous study where Turicibacter sp. was found exclusively in non-immunized AO, but not in DA rats, in this study it was detected in DA rats that remained healthy 16 days after induction, as well as in four of 12 DA rats at the peak of the disease. Similar observation was obtained for the members of Lachnospiraceae. Further, production of a typical regulatory cytokine interleukin-10 was compared in GALT cells of AO and DA rats, and higher production was observed in DA rats. Our data contribute to the idea that gut microbiota and GALT considerably influence multiple sclerosis pathogenesis.
Collapse
Affiliation(s)
- Suzana Stanisavljević
- Department of Immunology, Institute for Biological Research “Siniša Stanković," University of BelgradeBelgrade, Serbia
| | - Jovanka Lukić
- Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of BelgradeBelgrade, Serbia
| | - Svetlana Soković
- Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of BelgradeBelgrade, Serbia
| | - Sanja Mihajlovic
- Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of BelgradeBelgrade, Serbia
| | | | - Djordje Miljković
- Department of Immunology, Institute for Biological Research “Siniša Stanković," University of BelgradeBelgrade, Serbia
- *Correspondence: Djordje Miljković, Natasa Golić,
| | - Natasa Golić
- Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of BelgradeBelgrade, Serbia
- *Correspondence: Djordje Miljković, Natasa Golić,
| |
Collapse
|