1
|
Liu AR, Sarkar N, Cress JD, de Jesus TJ, Vadlakonda A, Centore JT, Griffith AD, Rohr B, McCormick TS, Cooper KD, Ramakrishnan P. NF-κB c-Rel is a critical regulator of TLR7-induced inflammation in psoriasis. EBioMedicine 2024; 110:105452. [PMID: 39586195 PMCID: PMC11625363 DOI: 10.1016/j.ebiom.2024.105452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/28/2024] [Accepted: 10/30/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Nuclear factor kappa B (NF-κB) c-Rel is a psoriasis susceptibility locus, however mechanisms underlying c-Rel transactivation during disease are poorly understood. Inflammation in psoriasis can be triggered following Toll-like Receptor 7 (TLR7) signalling in dendritic cells (DCs), and c-Rel is a critical regulator of DC function. Here, we studied the mechanism of TLR7-induced c-Rel-mediated inflammation in DCs. METHODS The overall expression of c-Rel was analysed in skin sections from patients with psoriasis in human transcriptomics datasets as well as the imiquimod-induced psoriasis mouse model. The function of c-Rel in DCs following TLR7 stimulation was determined by c-Rel CRISPR/Cas9 knockout DC2.4 immortalised cells and primary bone marrow derived dendritic cells from c-Rel knockout C57BL6/J mice. FINDINGS c-Rel is highly expressed in lesional skin of patients with psoriasis and TLR7-induced psoriatic lesions in mice. c-Rel deficiency protected mice from the disease, and specifically compromised TLR7-induced, and not TLR9- or TLR3-induced, inflammation in dendritic cells. Mechanistically, c-Rel deficiency disrupted activating NF-κB dimers and allowed binding of inhibitory NF-κB homodimers to the IL-1β and IL-6 promoters thus inhibiting their expression. This functionally compromises the ability of c-Rel deficient DCs to induce Th17 polarisation, which is critical in psoriasis pathogenesis. INTERPRETATION Our findings reveal that c-Rel is a key regulator of TLR7-mediated dendritic cell-dependent inflammation, and that targeting c-Rel-dependent signalling could prove an effective strategy to dampen excessive inflammation in TLR7-related skin inflammation. FUNDING A complete list of funding sources that contributed to this study can be found in the Acknowledgements section.
Collapse
Affiliation(s)
- Angela Rose Liu
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, Ohio 44106, USA
| | - Nandini Sarkar
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, Ohio 44106, USA
| | - Jordan D Cress
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, Ohio 44106, USA
| | - Tristan J de Jesus
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, Ohio 44106, USA
| | - Ananya Vadlakonda
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, Ohio 44106, USA
| | - Joshua T Centore
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, Ohio 44106, USA
| | - Alexis D Griffith
- Department of Dermatology, Case Western Reserve University, 2109 Adelbert Road, Cleveland, Ohio 44106, USA
| | - Bethany Rohr
- Department of Dermatology, Case Western Reserve University, 2109 Adelbert Road, Cleveland, Ohio 44106, USA; University Hospitals-Cleveland Medical Center, 11100 Euclid Ave, Cleveland, Ohio 44106, USA
| | - Thomas S McCormick
- Department of Dermatology, Case Western Reserve University, 2109 Adelbert Road, Cleveland, Ohio 44106, USA; University Hospitals-Cleveland Medical Center, 11100 Euclid Ave, Cleveland, Ohio 44106, USA
| | - Kevin D Cooper
- Department of Dermatology, Case Western Reserve University, 2109 Adelbert Road, Cleveland, Ohio 44106, USA; University Hospitals-Cleveland Medical Center, 11100 Euclid Ave, Cleveland, Ohio 44106, USA
| | - Parameswaran Ramakrishnan
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, Ohio 44106, USA; The Case Comprehensive Cancer Center, Case Western Reserve University, 2103 Cornell Road, Cleveland, Ohio 44106, USA; Department of Biochemistry, Case Western Reserve University, 2109 Adelbert Road, Cleveland, Ohio 44106, USA; University Hospitals-Cleveland Medical Center, 11100 Euclid Ave, Cleveland, Ohio 44106, USA; Louis Stokes Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, Ohio 44106, USA.
| |
Collapse
|
2
|
Yosri M, Dokhan M, Aboagye E, Al Moussawy M, Abdelsamed HA. Mechanisms governing bystander activation of T cells. Front Immunol 2024; 15:1465889. [PMID: 39669576 PMCID: PMC11635090 DOI: 10.3389/fimmu.2024.1465889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/31/2024] [Indexed: 12/14/2024] Open
Abstract
The immune system is endowed with the capacity to distinguish between self and non-self, so-called immune tolerance or "consciousness of the immune system." This type of awareness is designed to achieve host protection by eliminating cells expressing a wide range of non-self antigens including microbial-derived peptides. Such a successful immune response is associated with the secretion of a whole spectrum of soluble mediators, e.g., cytokines and chemokines, which not only contribute to the clearance of infected host cells but also activate T cells that are not specific to the original cognate antigen. This kind of non-specific T-cell activation is called "bystander activation." Although it is well-established that this phenomenon is cytokine-dependent, there is evidence in the literature showing the involvement of peptide/MHC recognition depending on the type of T-cell subset (naive vs. memory). Here, we will summarize our current understanding of the mechanism(s) of bystander T-cell activation as well as its biological significance in a wide range of diseases including microbial infections, cancer, auto- and alloimmunity, and chronic inflammatory diseases such as atherosclerosis.
Collapse
Affiliation(s)
- Mohammed Yosri
- The Regional Center for Mycology and Biotechnology, Al-Azhar University, Cairo, Egypt
| | - Mohamed Dokhan
- Immunology Center of Georgia (IMMCG), Medical College of Georgia (MCG), Augusta University, Augusta, GA, United States
| | - Elizabeth Aboagye
- Immunology Center of Georgia (IMMCG), Medical College of Georgia (MCG), Augusta University, Augusta, GA, United States
| | - Mouhamad Al Moussawy
- Starzl Transplantation Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Hossam A. Abdelsamed
- Immunology Center of Georgia (IMMCG), Medical College of Georgia (MCG), Augusta University, Augusta, GA, United States
- Department of Physiology, Augusta University, Augusta, GA, United States
| |
Collapse
|
3
|
Chen L, Ruan G, Cheng Y, Yi A, Chen D, Wei Y. The role of Th17 cells in inflammatory bowel disease and the research progress. Front Immunol 2023; 13:1055914. [PMID: 36700221 PMCID: PMC9870314 DOI: 10.3389/fimmu.2022.1055914] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/28/2022] [Indexed: 01/11/2023] Open
Abstract
Th17 cells play an important role in the abnormal immune response in inflammatory bowel disease (IBD) and are involved in the development and progression of inflammation and fibrosis. An increasing amount of data has shown that gut microbes are important parts of intestinal immunity and regulators of Th17 cellular immunity. Th17 cell differentiation is regulated by intestinal bacteria and cytokines, and Th17 cells regulate the intestinal mucosal immune microenvironment by secreting cytokines, such as IL-17, IL-21, and IL-26. Solid evidence showed that, regarding the treatment of IBD by targeting Th17 cells, the therapeutic effect of different biological agents varies greatly. Fecal bacteria transplantation (FMT) in the treatment of IBD has been a popular research topic in recent years and is safe and effective with few side effects. To further understand the role of Th17 cells in the progression of IBD and associated therapeutic prospects, this review will discuss the progress of related research on Th17 cells in IBD by focusing on the interaction and immune regulation between Th17 cells and gut microbiota.
Collapse
Affiliation(s)
| | | | | | | | | | - Yanling Wei
- *Correspondence: Yanling Wei, ; Dongfeng Chen,
| |
Collapse
|
4
|
Wang M, Su T, Sun H, Cheng H, Jiang C, Guo P, Zhu Z, Fang R, He F, Ge M, Guan Q, Wei W, Wang Q. Regulating Th17/Treg Balance Contributes to the Therapeutic Effect of Ziyuglycoside I on Collagen-Induced Arthritis. Int J Mol Sci 2022; 23:16105. [PMID: 36555745 PMCID: PMC9786935 DOI: 10.3390/ijms232416105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/11/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
To investigate the therapeutic effect and primary pharmacological mechanism of Ziyuglycoside I (Ziyu I) on collagen-induced arthritis (CIA) mice. CIA mice were treated with 5, 10, or 20 mg/kg of Ziyu I or 2 mg/kg of methotrexate (MTX), and clinical manifestations, as well as pathological changes, were observed. T cell viability and subset type were determined, and serum levels of transforming growth factor-beta (TGF-β) and interleukin-17 (IL-17) were detected. The mRNA expression of retinoid-related orphan receptor-γt (RORγt) and transcription factor forkhead box protein 3 (Foxp3) in mouse spleen lymphocytes was ascertained by the real-time reverse transcriptase-polymerase chain reaction (RT-qPCR). Molecular docking was used to detect whether there was a molecular interaction between Ziyu I and protein kinase B (Akt). The activation of mechanistic target of rapamycin (mTOR) in T cells was verified by Western blotting or immunofluorescence. Ziyu I treatment effectively alleviated arthritis symptoms of CIA mice, including body weight, global score, arthritis index, and a number of swollen joints. Similarly, pathological changes of joints and spleens in arthritic mice were improved. The thymic index, T cell activity, and RORγt production of Ziyu I-treated mice were significantly reduced. Notably, through molecular docking, western blotting, and immunofluorescence data analysis, it was found that Ziyu I could interact directly with Akt to reduce downstream mTOR activation and inhibit helper T cell 17 (Th17) differentiation, thereby regulating Th17/regulatory T cell (Treg) balance and improving arthritis symptoms. Ziyu I effectively improves arthritic symptoms in CIA mice by inhibiting mTOR activation, thereby affecting Th17 differentiation and regulating Th17/Treg balance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei 230032, China
| | - Qingtong Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei 230032, China
| |
Collapse
|
5
|
Bhatnagar S, Revuri V, Shah M, Larson P, Shao Z, Yu D, Prabha S, Griffith TS, Ferguson D, Panyam J. Combination of STING and TLR 7/8 Agonists as Vaccine Adjuvants for Cancer Immunotherapy. Cancers (Basel) 2022; 14:cancers14246091. [PMID: 36551577 PMCID: PMC9777055 DOI: 10.3390/cancers14246091] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 11/30/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Immunostimulatory adjuvants that potently activate antigen-presenting cells and (in turn) prime cytotoxic T cells are a key component of anticancer vaccines. In this study, we investigated a multi-adjuvant approach combining a TLR 7/8 agonist (522) and a STING agonist (DMXAA) to promote enhanced antigen cross-presentation, stimulate specific antitumor T-cell responses, and provide improved anticancer efficacy. In vitro experiments using bone marrow-derived dendritic cells (BMDCs) confirmed enhanced activation with the 522-DMXAA combination based on both co-stimulatory molecule expression and pro-inflammatory cytokine secretion. The immunization of mice with vaccines comprising both 522 and DMXAA resulted in greater antitumor efficacy in B16F10 melanoma and MB49 bladder tumor models relative to mono-agonist vaccines. Flow cytometry-based analysis of immune cells from immunized mice revealed the significant activation of antigen-presenting cells, increased numbers of activated and Ag-specific CD8+ T cells in the spleen and lymph nodes, modest NK cell activation, and an overall reduction in CD206+ macrophages. These results were supported by an increase in the levels of IFN-γ and a reduction in IL-10 levels in the sera. Taken together, these findings demonstrate the potential of the TLR7/8 and STING agonist combination as vaccine adjuvants to activate both innate and adaptive immune responses.
Collapse
Affiliation(s)
- Shubhmita Bhatnagar
- Department of Pharmaceutical Sciences, School of Pharmacy, Temple University, Philadelphia, PA 19140, USA
| | - Vishnu Revuri
- Department of Pharmaceutical Sciences, School of Pharmacy, Temple University, Philadelphia, PA 19140, USA
| | - Manan Shah
- Department of Pharmaceutical Sciences, School of Pharmacy, Temple University, Philadelphia, PA 19140, USA
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Peter Larson
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Zekun Shao
- Department of Pharmaceutical Sciences, School of Pharmacy, Temple University, Philadelphia, PA 19140, USA
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daohai Yu
- Center for Biostatistics and Epidemiology, Lewis-Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Swayam Prabha
- Fels Cancer Institute for Personalized Medicine, Lewis-Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Fox Chase Comprehensive Cancer Institute, Temple University, Philadelphia, PA 19111, USA
| | - Thomas S. Griffith
- Department of Urology, Medical School, University of Minnesota, Minneapolis, MN 55455, USA
| | - David Ferguson
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jayanth Panyam
- Department of Pharmaceutical Sciences, School of Pharmacy, Temple University, Philadelphia, PA 19140, USA
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
- Fox Chase Comprehensive Cancer Institute, Temple University, Philadelphia, PA 19111, USA
- Correspondence: ; Tel.: +1-215-926-2006
| |
Collapse
|
6
|
Lo SM, Hwang YS, Liu CL, Shen CN, Hong WH, Yang WC, Lee MH, Shen CR. Inhibiting TLR7 Expression in the Retinal Pigment Epithelium Suppresses Experimental Autoimmune Uveitis. Front Immunol 2022; 12:736261. [PMID: 35069523 PMCID: PMC8766412 DOI: 10.3389/fimmu.2021.736261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/13/2021] [Indexed: 11/16/2022] Open
Abstract
Experimental autoimmune uveitis (EAU), a model of human uveitis, is an organ-specific, T cell-mediated autoimmune disease. Autoreactive T cells can penetrate the blood-retinal barrier, which is a physical defense composed of tight junction-linked retinal pigment epithelial (RPE) cells. RPE cells serve as antigen-presenting cells (APCs) in the eye since they express MHC class I and II and Toll-like receptors (TLRs). Although previous studies have shown that supplementation with TLR agonists exacerbates uveitis, little is known about how TLR signaling in the RPE contributes to the development of uveitis. In this study, we isolated the RPE from EAU mice, which were induced by active immunization (aEAU) or adoptive transfer of antigen-specific T cells (tEAU). The expression of TLRs on RPE was determined, and both aEAU and tEAU mice exhibited induced tlr7 expression. The TLR7 agonist R848 was shown to induce aggressive disease progression, along with significantly elevated levels of the uveopathogenic cytokine IL-17. Furthermore, not only IL-17 but also R848 appeared to enhance the inflammatory response and to impair the barrier function of the RPE, indicating that TLR7 signaling is involved in the pathogenesis of EAU by affecting the behaviors of the RPE and consequently allowing the infiltration of autoreactive T cells intraocularly. Finally, local application of shRNA against TLR7 delivered by recombinant AAV effectively inhibited disease severity and reduced IFN-γ and IL-17. Our findings highlight an immunomodulatory role of RPE TLR7 in EAU development and provide a potential therapeutic strategy for autoimmune uveitis.
Collapse
Affiliation(s)
- Sheng-Min Lo
- Department and Graduate Institute of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Yih-Shiou Hwang
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Department of Ophthalmology, Lin-Kou Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Chao-Lin Liu
- Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan.,Biochemical Technology R&D Center, Ming Chi University of Technology, New Taipei City, Taiwan
| | - Chia-Ning Shen
- Genomic Research Center, Academia Sinica, Taipei, Taiwan
| | - Wei-Hsin Hong
- Department and Graduate Institute of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Wei-Cheng Yang
- Department and Graduate Institute of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Meng-Hua Lee
- Department and Graduate Institute of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Chia-Rui Shen
- Department and Graduate Institute of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Department of Ophthalmology, Lin-Kou Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan City, Taiwan
| |
Collapse
|
7
|
Tian Y, Huang B, Li J, Tian X, Zeng X. Identification of the Association Between Toll-Like Receptors and T-Cell Activation in Takayasu’s Arteritis. Front Immunol 2022; 12:792901. [PMID: 35126357 PMCID: PMC8812403 DOI: 10.3389/fimmu.2021.792901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/22/2021] [Indexed: 12/26/2022] Open
Abstract
To explore the relationships between Toll-like receptors (TLRs) and the activation and differentiation of T-cells in Takayasu’s arteritis (TAK), using real-time fluorescence quantitative polymerase chain reaction, mRNA abundance of 29 target genes in peripheral blood mononuclear cells (PBMCs) were detected from 27 TAK patients and 10 healthy controls. Compared with the healthy control group, the untreated TAK group and the treated TAK group had an increased mRNA level of TLR2 and TLR4. A sample-to-sample matrix revealed that 80% of healthy controls could be separated from the TAK patients. Correlation analysis showed that the inactive-treated TAK group exhibited a unique pattern of inverse correlations between the TLRs gene clusters (including TLR1/2/4/6/8, BCL6, TIGIT, NR4A1, etc) and the gene cluster associated with T-cell activation and differentiation (including TCR, CD28, T-bet, GATA3, FOXP3, CCL5, etc). The dynamic gene co-expression network indicated the TAK groups had more active communication between TLRs and T-cell activation than healthy controls. BCL6, CCL5, FOXP3, GATA3, CD28, T-bet, TIGIT, IκBα, and NR4A1 were likely to have a close functional relation with TLRs at the inactive stage. The co-expression of TLR4 and TLR6 could serve as a biomarker of disease activity in treated TAK (the area under curve/sensitivity/specificity, 0.919/100%/90.9%). The largest gene co-expression cluster of the inactive-treated TAK group was associated with TLR signaling pathways, while the largest gene co-expression cluster of the active-treated TAK group was associated with the activation and differentiation of T-cells. The miRNA sequencing of the plasma exosomes combining miRDB, DIANA-TarBase, and miRTarBase databases suggested that the miR-548 family miR-584, miR-3613, and miR-335 might play an important role in the cross-talk between TLRs and T-cells at the inactive stage. This study found a novel relation between TLRs and T-cell in the pathogenesis of autoimmune diseases, proposed a new concept of TLR-co-expression signature which might distinguish different disease activity of TAK, and highlighted the miRNA of exosomes in TLR signaling pathway in TAK.
Collapse
Affiliation(s)
- Yixiao Tian
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital (PUMCH), Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Biqing Huang
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital (PUMCH), Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Jing Li
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital (PUMCH), Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
- *Correspondence: Jing Li, ; Xiaofeng Zeng,
| | - Xinping Tian
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital (PUMCH), Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital (PUMCH), Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
- *Correspondence: Jing Li, ; Xiaofeng Zeng,
| |
Collapse
|
8
|
Ryan L, Mills KHG. Sex differences regulate immune responses in experimental autoimmune encephalomyelitis and multiple sclerosis. Eur J Immunol 2021; 52:24-33. [PMID: 34727577 DOI: 10.1002/eji.202149589] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/30/2021] [Accepted: 10/29/2021] [Indexed: 12/26/2022]
Abstract
MS is an autoimmune disease of the CNS that afflicts over 2.5 million people worldwide. There are striking sex differences in the susceptibility to and progression of this disease in humans. Females are twice as likely to develop MS than males, whereas disease progression and disability is more rapid in males compared with females; however, the latter is still controversial. There is growing evidence, mainly from animal models, that innate and adaptive immune responses are different in males and females, and that this can influence the outcome of a range of diseases including infection, cancer, and autoimmunity. Since MS is an immune-mediated disease, sex differences in pathogenic immune responses may account for some of the differences in susceptibility to and progression seen in men versus women. Indeed, data from the mouse model of MS, EAE, have already provided some evidence that female mice have earlier disease onset associated with stronger Th17 responses. This review will discuss the possible immunological basis of sex differences in susceptibility and disease outcome in EAE and MS and how a better understanding of sex differences in the responses to disease-modifying therapies may lead to improved patient treatment.
Collapse
Affiliation(s)
- Lucy Ryan
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Kingston H G Mills
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
9
|
Umar S, Palasiewicz K, Van Raemdonck K, Volin MV, Romay B, Amin MA, Zomorrodi RK, Arami S, Gonzalez M, Rao V, Zanotti B, Fox DA, Sweiss N, Shahrara S. IRAK4 inhibition: a promising strategy for treating RA joint inflammation and bone erosion. Cell Mol Immunol 2021; 18:2199-2210. [PMID: 32415262 PMCID: PMC8429735 DOI: 10.1038/s41423-020-0433-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 03/30/2020] [Indexed: 01/22/2023] Open
Abstract
Flares of joint inflammation and resistance to currently available biologic therapeutics in rheumatoid arthritis (RA) patients could reflect activation of innate immune mechanisms. Herein, we show that a TLR7 GU-rich endogenous ligand, miR-Let7b, potentiates synovitis by amplifying RA monocyte and fibroblast (FLS) trafficking. miR-Let7b ligation to TLR7 in macrophages (MΦs) and FLSs expanded the synovial inflammatory response. Moreover, secretion of M1 monokines triggered by miR-Let7b enhanced Th1/Th17 cell differentiation. We showed that IRAK4 inhibitor (i) therapy attenuated RA disease activity by blocking TLR7-induced M1 MΦ or FLS activation, as well as monokine-modulated Th1/Th17 cell polarization. IRAK4i therapy also disrupted RA osteoclastogenesis, which was amplified by miR-Let7b ligation to joint myeloid TLR7. Hence, the effectiveness of IRAK4i was compared with that of a TNF inhibitor (i) or anti-IL-6R treatment in collagen-induced arthritis (CIA) and miR-Let7b-mediated arthritis. We found that TNF or IL-6R blocking therapies mitigated CIA by reducing the infiltration of joint F480+iNOS+ MΦs, the expression of certain monokines, and Th1 cell differentiation. Unexpectedly, these biologic therapies were unable to alleviate miR-Let7b-induced arthritis. The superior efficacy of IRAK4i over anti-TNF or anti-IL-6R therapy in miR-Let7b-induced arthritis or CIA was due to the ability of IRAK4i therapy to restrain the migration of joint F480+iNOS+ MΦs, vimentin+ fibroblasts, and CD3+ T cells, in addition to negating the expression of a wide range of monokines, including IL-12, MIP2, and IRF5 and Th1/Th17 lymphokines. In conclusion, IRAK4i therapy may provide a promising strategy for RA therapy by disconnecting critical links between inflammatory joint cells.
Collapse
Affiliation(s)
- Sadiq Umar
- Jesse Brown VA Medical Center, Chicago, IL, 60612, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Karol Palasiewicz
- Jesse Brown VA Medical Center, Chicago, IL, 60612, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Katrien Van Raemdonck
- Jesse Brown VA Medical Center, Chicago, IL, 60612, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Michael V Volin
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, 60515, USA
| | - Bianca Romay
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - M Asif Amin
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, MI, 481096, USA
| | - Ryan K Zomorrodi
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Shiva Arami
- Jesse Brown VA Medical Center, Chicago, IL, 60612, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Mark Gonzalez
- Department of Orthopedic Surgery, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Vikram Rao
- Pfizer Research, Cambridge, MA, 02139, USA
| | - Brian Zanotti
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, 60515, USA
| | - David A Fox
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, MI, 481096, USA
| | - Nadera Sweiss
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Shiva Shahrara
- Jesse Brown VA Medical Center, Chicago, IL, 60612, USA.
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
10
|
Wu N, Morsey BM, Emanuel KM, Fox HS. Sequence-specific extracellular microRNAs activate TLR7 and induce cytokine secretion and leukocyte migration. Mol Cell Biochem 2021; 476:4139-4151. [PMID: 34313894 DOI: 10.1007/s11010-021-04220-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023]
Abstract
Toll-like receptors (TLRs) can contribute to central nervous system disease pathologies via recognition of microRNAs (miRNAs); however, it remains to be determined which miRNAs are able to activate this signaling. Here we report that numerous miRNAs induced the production of tumor necrosis factor alpha in multiple myeloid cell types, including microglia, and that this effect was abolished in cells deficient in TLR7. Examination of closely related miRNAs that differed in their ability to activate TLR7 resulted in the identification of a motif (UGCUUAU) in miR-20a-5p and specific nucleotides (all the uridines and surprisingly the cytosine as well) in a key area of miR-20a-5p and miR-148b-3p that were vital for the secretion of cytokines via TLR7 stimulation. A 10-nucleotide sequence including this motif was identified to be the shortest single-stranded RNA to signal via TLR7. An miRNA containing this motif induced the secretion of multiple proinflammatory molecules, which was dependent on the phosphoinositide 3-kinase, mitogen-activated protein kinase, and nuclear factor kappa-light-chain-enhancer of activated B cell signaling pathways. Wild-type mice administered miR-20a-5p, which contained this motif, demonstrated increased leukocyte migration. This effect was significantly ameliorated in TLR7-knockout mice, and mice administered miR-20b-5p, in which the motif was mutated, did not exhibit leukocyte migration. We provide a detailed analysis of miRNAs that activate endosomal TLR7 and identify key nucleotide features of a sequence motif recognized by TLR7.
Collapse
Affiliation(s)
- Niming Wu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 42nd and Emile, Omaha, NE, 68198, USA
| | - Brenda M Morsey
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 42nd and Emile, Omaha, NE, 68198, USA
| | - Katy M Emanuel
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 42nd and Emile, Omaha, NE, 68198, USA
| | - Howard S Fox
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 42nd and Emile, Omaha, NE, 68198, USA.
| |
Collapse
|
11
|
Marks KE, Cho K, Stickling C, Reynolds JM. Toll-like Receptor 2 in Autoimmune Inflammation. Immune Netw 2021; 21:e18. [PMID: 34277108 PMCID: PMC8263214 DOI: 10.4110/in.2021.21.e18] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 06/22/2021] [Indexed: 12/11/2022] Open
Abstract
TLR signaling is critical for broad scale immune recognition of pathogens and/or danger molecules. TLRs are particularly important for the activation and the maturation of cells comprising the innate immune response. In recent years it has become apparent that several different TLRs regulate the function of lymphocytes as well, albeit to a lesser degree compared to innate immunity. TLR2 heterodimerizes with either TLR1 or TLR6 to broadly recognize bacterial lipopeptides as well as several danger-associated molecular patterns. In general, TLR2 signaling promotes immune cell activation leading to tissue inflammation, which is advantageous for combating an infection. Conversely, inappropriate or dysfunctional TLR2 signaling leading to an overactive inflammatory response could be detrimental during sterile inflammation and autoimmune disease. This review will highlight and discuss recent research advances linking TLR2 engagement to autoimmune inflammation.
Collapse
Affiliation(s)
- Kathryne E Marks
- Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Kaylin Cho
- Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Courtney Stickling
- Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Joseph M Reynolds
- Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| |
Collapse
|
12
|
Zenobia C, Herpoldt KL, Freire M. Is the oral microbiome a source to enhance mucosal immunity against infectious diseases? NPJ Vaccines 2021; 6:80. [PMID: 34078913 PMCID: PMC8172910 DOI: 10.1038/s41541-021-00341-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/24/2021] [Indexed: 12/14/2022] Open
Abstract
Mucosal tissues act as a barrier throughout the oral, nasopharyngeal, lung, and intestinal systems, offering first-line protection against potential pathogens. Conventionally, vaccines are applied parenterally to induce serotype-dependent humoral response but fail to drive adequate mucosal immune protection for viral infections such as influenza, HIV, and coronaviruses. Oral mucosa, however, provides a vast immune repertoire against specific microbial pathogens and yet is shaped by an ever-present microbiome community that has co-evolved with the host over thousands of years. Adjuvants targeting mucosal T-cells abundant in oral tissues can promote soluble-IgA (sIgA)-specific protection to confer increased vaccine efficacy. Th17 cells, for example, are at the center of cell-mediated immunity and evidence demonstrates that protection against heterologous pathogen serotypes is achieved with components from the oral microbiome. At the point of entry where pathogens are first encountered, typically the oral or nasal cavity, the mucosal surfaces are layered with bacterial cohabitants that continually shape the host immune profile. Constituents of the oral microbiome including their lipids, outer membrane vesicles, and specific proteins, have been found to modulate the Th17 response in the oral mucosa, playing important roles in vaccine and adjuvant designs. Currently, there are no approved adjuvants for the induction of Th17 protection, and it is critical that this research is included in the preparedness for the current and future pandemics. Here, we discuss the potential of oral commensals, and molecules derived thereof, to induce Th17 activity and provide safer and more predictable options in adjuvant engineering to prevent emerging infectious diseases.
Collapse
Affiliation(s)
| | | | - Marcelo Freire
- Departments of Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, USA.
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
13
|
Oner F, Alvarez C, Yaghmoor W, Stephens D, Hasturk H, Firatli E, Kantarci A. Resolvin E1 Regulates Th17 Function and T Cell Activation. Front Immunol 2021; 12:637983. [PMID: 33815391 PMCID: PMC8009993 DOI: 10.3389/fimmu.2021.637983] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
Resolvin E1 (RvE1) is a specialized pro-resolving lipid mediator derived from eicosapentaenoic acid and plays a critical role in resolving inflammation and tissue homeostasis. Th17 cells are a distinct group of T helper (Th) cells with tissue-destructive functions in autoimmune and chronic inflammatory diseases via the secretion of IL-17. Dendritic cell (DC)-mediated antigen presentation regulates the Th17-induced progression of inflammation and tissue destruction. In this study, we hypothesized that the RvE1 would restore homeostatic balance and inflammation by targeting the Th17 function. We designed three experiments to investigate the impact of RvE1 on different phases of Th17 response and the potential role of DCs: First CD4+ T cells were induced by IL-6/TGFβ to measure the effect of RvE1 on Th17 differentiation in an inflammatory milieu. Second, we measured the impact of RvE1 on DC-stimulated Th17 differentiation in a co-culture model. Third, we measured the effect of RvE1 on DC maturation. RvE1 blocked the CD25, CCR6 and IL-17 expression; IL-17, IL-21, IL-10, and IL-2 production, suggesting inhibition of T cell activation, Th17 stimulation and chemoattraction. RvE1 also suppressed the activation of DCs by limiting their pro-inflammatory cytokine production. Our findings collectively demonstrated that the RvE1 targeted the Th17 activation and the DC function as a potential mechanism for inflammatory resolution and acquired immune response.
Collapse
Affiliation(s)
- Fatma Oner
- The Forsyth Institute, Cambridge, MA, United States.,Department of Periodontology, School of Dentistry, Istanbul University, Istanbul, Turkey
| | - Carla Alvarez
- The Forsyth Institute, Cambridge, MA, United States.,Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Wael Yaghmoor
- The Forsyth Institute, Cambridge, MA, United States.,Faculty of Dentistry, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | | | - Erhan Firatli
- Department of Periodontology, School of Dentistry, Istanbul University, Istanbul, Turkey
| | - Alpdogan Kantarci
- The Forsyth Institute, Cambridge, MA, United States.,School of Dental Medicine, Harvard University, Boston, MA, United States
| |
Collapse
|
14
|
Babic M, Dimitropoulos C, Hammer Q, Stehle C, Heinrich F, Sarsenbayeva A, Eisele A, Durek P, Mashreghi MF, Lisnic B, Van Snick J, Löhning M, Fillatreau S, Withers DR, Gagliani N, Huber S, Flavell RA, Polic B, Romagnani C. NK cell receptor NKG2D enforces proinflammatory features and pathogenicity of Th1 and Th17 cells. J Exp Med 2021; 217:151818. [PMID: 32453422 PMCID: PMC7398170 DOI: 10.1084/jem.20190133] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/24/2020] [Accepted: 04/24/2020] [Indexed: 12/24/2022] Open
Abstract
NKG2D is a danger sensor expressed on different subsets of innate and adaptive lymphocytes. Despite its established role as a potent activator of the immune system, NKG2D-driven regulation of CD4+ T helper (Th) cell-mediated immunity remains unclear. In this study, we demonstrate that NKG2D modulates Th1 and proinflammatory T-bet+ Th17 cell effector functions in vitro and in vivo. In particular, NKG2D promotes higher production of proinflammatory cytokines by Th1 and T-bet+ Th17 cells and reinforces their transcription of type 1 signature genes, including Tbx21. Conditional deletion of NKG2D in T cells impairs the ability of antigen-specific CD4+ T cells to promote inflammation in vivo during antigen-induced arthritis and experimental autoimmune encephalomyelitis, indicating that NKG2D is an important target for the amelioration of Th1- and Th17-mediated chronic inflammatory diseases.
Collapse
Affiliation(s)
- Marina Babic
- Innate Immunity, German Rheumatism Research Centre-a Leibniz Institute, Berlin, Germany.,Division of Gastroenterology, Infectiology and Rheumatology, Medical Department I, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Quirin Hammer
- Innate Immunity, German Rheumatism Research Centre-a Leibniz Institute, Berlin, Germany
| | - Christina Stehle
- Innate Immunity, German Rheumatism Research Centre-a Leibniz Institute, Berlin, Germany
| | - Frederik Heinrich
- Therapeutic Gene Regulation, German Rheumatism Research Centre-a Leibniz Institute, Berlin, Germany
| | - Assel Sarsenbayeva
- Innate Immunity, German Rheumatism Research Centre-a Leibniz Institute, Berlin, Germany
| | - Almut Eisele
- Innate Immunity, German Rheumatism Research Centre-a Leibniz Institute, Berlin, Germany
| | - Pawel Durek
- Cell Biology, German Rheumatism Research Centre-a Leibniz Institute, Berlin, Germany
| | - Mir-Farzin Mashreghi
- Therapeutic Gene Regulation, German Rheumatism Research Centre-a Leibniz Institute, Berlin, Germany
| | - Berislav Lisnic
- Center for Proteomics, University of Rijeka, Rijeka, Croatia.,Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Croatia
| | | | - Max Löhning
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Centre-a Leibniz Institute, Berlin, Germany.,Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Simon Fillatreau
- Institut Necker-Enfants Malades, INSERM U1151/CNRS UMR8253, Faculté de Médecine Paris Descartes, Paris, France
| | - David R Withers
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Nicola Gagliani
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Samuel Huber
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT.,Howard Hughes Medical Institute, Yale University, New Haven, CT
| | - Bojan Polic
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Croatia
| | - Chiara Romagnani
- Innate Immunity, German Rheumatism Research Centre-a Leibniz Institute, Berlin, Germany.,Division of Gastroenterology, Infectiology and Rheumatology, Medical Department I, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
15
|
Katwal P, Uprety T, Okda F, Antony L, Thomas M, Chase C, Diel DG, Nelson E, Young A, Li F, Scaria J, Kaushik RS. Characterization of bovine ileal epithelial cell line for lectin binding, susceptibility to enteric pathogens, and TLR mediated immune responses. Comp Immunol Microbiol Infect Dis 2020; 74:101581. [PMID: 33260019 DOI: 10.1016/j.cimid.2020.101581] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 10/29/2020] [Accepted: 11/03/2020] [Indexed: 12/19/2022]
Abstract
In this study, primary and immortalized bovine intestinal epithelial cells (BIECs) were characterized for the expression of surface carbohydrate moieties. Primary BIEC-c4 cells showed staining greater than 90 % for 16 lectins but less than 50 % staining for four lectins. Immortalized BIECs showed significantly different lectin binding profile for few lectins compared to BIEC-c4 cells. BIEC-c4 cells were studied for infectivity to E. coli, Salmonella enterica, bovine rotavirus, bovine coronavirus, and bovine viral diarrhea virus. Bovine strain E. coli B41 adhered to BIEC-c4 cells and Salmonella strains S. Dublin and S. Mbandaka showed strong cell invasion. BIEC-c4 cells were susceptible to bovine rotavirus. LPS stimulation upregulated IL-10, IL-8, and IL-6 expression and Poly I:C upregulated TLR 8 and TLR 9 expression. This study provides important knowledge on the glycoconjugate expression profile of primary and immortalized BIECs and infectivity and immune responses of primary BIECs to bacterial and viral pathogens or ligands.
Collapse
Affiliation(s)
- Pratik Katwal
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, 57007, USA
| | - Tirth Uprety
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, 57007, USA
| | - Faten Okda
- Dept of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, 57007, USA; Dept. of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA; National Research Center, Giza, Egypt
| | - Linto Antony
- Dept of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, 57007, USA
| | - Milton Thomas
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, 57007, USA
| | - Christopher Chase
- Dept of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, 57007, USA
| | - Diego G Diel
- Dept of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, 57007, USA; Department of Population Medicine and Diagnostic Sciences, Cornell University, College of Veterinary Medicine, Ithaca, NY, 14853, USA
| | - Eric Nelson
- Dept of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, 57007, USA
| | - Alan Young
- Dept of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, 57007, USA
| | - Feng Li
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, 57007, USA; Department of Veterinary Science, M.H. Gluck Equine Research Center, University of Kentucky, Lexington, KY, 40546, USA
| | - Joy Scaria
- Dept of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, 57007, USA
| | - Radhey S Kaushik
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, 57007, USA.
| |
Collapse
|
16
|
Murray LM, Yerkovich ST, Ferreira MA, Upham JW. Risks for cold frequency vary by sex: role of asthma, age, TLR7 and leukocyte subsets. Eur Respir J 2020; 56:13993003.02453-2019. [PMID: 32513781 DOI: 10.1183/13993003.02453-2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 05/17/2020] [Indexed: 11/05/2022]
Abstract
Viral respiratory infections are usually benign but can trigger asthma exacerbations. The factors associated with upper respiratory tract infection (cold) frequency are not fully understood, nor is it clear whether such factors differ between women and men.To determine which immunological and clinical variables associate with the frequency of self-reported respiratory infections (colds), 150 asthma cases and 151 controls were recruited. Associations between antiviral immune response variables: toll-like receptor (TLR)7/8 gene expression, plasmacytoid dendritic cell (pDC) numbers and interferon-α, tumour necrosis factor and interleukin-12 production, and asthma were then examined that might explain cold frequency.People with asthma cases reported more colds per year (median 3 versus 2; p<0.001) and had lower baseline TLR7 gene expression (odds ratio 0.12; p=0.02) than controls. Associations between many variables and cold frequency differed between women and men. In women, high blood neutrophil counts (β=0.096, p=0.002), and younger age (β=-0.017, p<0.001), but not exposure to children, were independently associated with more frequent colds. In men, low TLR7 expression (β=-0.96, p=0.041) and high CLEC4C gene expression (a marker of pDC; β=0.88, p=0.008) were independently associated with more frequent colds. Poor asthma symptom control was independently associated with reduced TLR8 gene expression (β=-1.4, p=0.036) and high body mass index (β=0.041, p=0.004).Asthma, age and markers of inflammation and antiviral immunity in peripheral blood are associated with frequent colds. Interestingly, the variables associated with cold frequency differed between women and men.
Collapse
Affiliation(s)
- Liisa M Murray
- Diamantina Institute, The University of Queensland, Brisbane, Australia
| | | | | | - John W Upham
- Diamantina Institute, The University of Queensland, Brisbane, Australia.,Respiratory and Sleep Medicine, Princess Alexandra Hospital, Brisbane, Australia
| |
Collapse
|
17
|
Stögerer T, Stäger S. Innate Immune Sensing by Cells of the Adaptive Immune System. Front Immunol 2020; 11:1081. [PMID: 32547564 PMCID: PMC7274159 DOI: 10.3389/fimmu.2020.01081] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 05/05/2020] [Indexed: 01/05/2023] Open
Abstract
Sensing of microbes or of danger signals has mainly been attributed to myeloid innate immune cells. However, T and B cells also express functional pattern recognition receptors (PRRs). In these cells, PRRs mediate signaling cascades that result in different functions depending on the cell's activation and/or differentiation status, on the environment, and on the ligand/agonist. Some of these functions are beneficial for the host; however, some are detrimental and are exploited by pathogens to establish persistent infections. In this review, we summarize the available literature on innate immune sensing by cells of the adaptive immune system and discuss possible implications for chronic infections.
Collapse
Affiliation(s)
- Tanja Stögerer
- INRS Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada
| | - Simona Stäger
- INRS Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada
| |
Collapse
|
18
|
Evans-Marin H, Rogier R, Koralov SB, Manasson J, Roeleveld D, van der Kraan PM, Scher JU, Koenders MI, Abdollahi-Roodsaz S. Microbiota-Dependent Involvement of Th17 Cells in Murine Models of Inflammatory Arthritis. Arthritis Rheumatol 2019; 70:1971-1983. [PMID: 29975009 PMCID: PMC6587816 DOI: 10.1002/art.40657] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 06/28/2018] [Indexed: 12/20/2022]
Abstract
Objective Intestinal microbiota are associated with the development of inflammatory arthritis. The aim of this study was to dissect intestinal mucosal immune responses in the preclinical phase of arthritis and determine whether the presence of Th17 cells, beyond involvement of the cytokine interleukin‐17 (IL‐17), is required for arthritis development, and whether the involvement of Th17 cells in arthritis depends on the composition of the host microbiota. Methods Mucosal T cell production of IL‐17, interferon‐γ, tumor necrosis factor α (TNFα), IL‐22, and granulocyte–macrophage colony‐stimulating factor (GM‐CSF) was analyzed by flow cytometry and Luminex assay before arthritis onset in mice immunized to develop collagen‐induced arthritis (CIA). Pathogenic features of arthritis in mice with CIA and mice with antigen‐induced arthritis were compared between Th17 cell–deficient (CD4‐Cre+Rorcflox/flox) and Th17 cell–sufficient (CD4‐Cre−Rorcflox/flox) mice. In addition, the impact of intestinal microbiota on the Th17 cell dependence of CIA was assessed. Results Lamina propria CD4 T cells were activated before the onset of arthritis in mice with CIA, with marked up‐regulation of several cytokines, including IL‐17A, TNFα, and GM‐CSF. CD4‐Cre+Rorcflox/flox mice showed a specific reduction in intestinal mucosal levels of Th17 cells and partially reduced levels of IL‐17–producing CD8 T cells. However, total levels of IL‐17A, mostly produced by γδ T cells and neutrophils, were unaffected. The severity of arthritis was significantly reduced in Th17 cell–deficient mice, suggesting that Th17 cells have additional, IL‐17A–independent roles in inflammatory arthritis. Accordingly, antigen‐stimulated T cells from Th17 cell–deficient mice produced less IL‐17A, IL‐17F, and GM‐CSF. Importantly, the dependence of CIA on the involvement of Th17 cells was mitigated in the presence of an alternative microbiome. Conclusion These data from murine models suggest that activation of mucosal immunity precedes the development of arthritis, and also that Th17 cells have a microbiota‐dependent role in arthritis. Therefore, a microbiome‐guided stratification of patients might improve the efficacy of Th17‐targeted therapies.
Collapse
Affiliation(s)
| | - Rebecca Rogier
- Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Julia Manasson
- New York University School of Medicine, New York, New York
| | | | | | - Jose U Scher
- New York University School of Medicine, New York, New York
| | | | - Shahla Abdollahi-Roodsaz
- New York University School of Medicine and Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
19
|
Adaptive innate immunity or innate adaptive immunity? Clin Sci (Lond) 2019; 133:1549-1565. [DOI: 10.1042/cs20180548] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 07/05/2019] [Accepted: 07/10/2019] [Indexed: 12/19/2022]
Abstract
Abstract
The innate immunity is frequently accepted as a first line of relatively primitive defense interfering with the pathogen invasion until the mechanisms of ‘privileged’ adaptive immunity with the production of antibodies and activation of cytotoxic lymphocytes ‘steal the show’. Recent advancements on the molecular and cellular levels have shaken the traditional view of adaptive and innate immunity. The innate immune memory or ‘trained immunity’ based on metabolic changes and epigenetic reprogramming is a complementary process insuring adaptation of host defense to previous infections.
Innate immune cells are able to recognize large number of pathogen- or danger- associated molecular patterns (PAMPs and DAMPs) to behave in a highly specific manner and regulate adaptive immune responses. Innate lymphoid cells (ILC1, ILC2, ILC3) and NK cells express transcription factors and cytokines related to subsets of T helper cells (Th1, Th2, Th17). On the other hand, T and B lymphocytes exhibit functional properties traditionally attributed to innate immunity such as phagocytosis or production of tissue remodeling growth factors. They are also able to benefit from the information provided by pattern recognition receptors (PRRs), e.g. γδT lymphocytes use T-cell receptor (TCR) in a manner close to PRR recognition. Innate B cells represent another example of limited combinational diversity usage participating in various innate responses. In the view of current knowledge, the traditional black and white classification of immune mechanisms as either innate or an adaptive needs to be adjusted and many shades of gray need to be included.
Collapse
|
20
|
Protective effects of delayed intraventricular TLR7 agonist administration on cerebral white and gray matter following asphyxia in the preterm fetal sheep. Sci Rep 2019; 9:9562. [PMID: 31267031 PMCID: PMC6606639 DOI: 10.1038/s41598-019-45872-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 06/13/2019] [Indexed: 01/08/2023] Open
Abstract
Preterm brain injury is highly associated with inflammation, which is likely related in part to sterile responses to hypoxia-ischemia. We have recently shown that neuroprotection with inflammatory pre-conditioning in the immature brain is associated with induction of toll-like receptor 7 (TLR7). We therefore tested the hypothesis that central administration of a synthetic TLR7 agonist, gardiquimod (GDQ), after severe hypoxia-ischemia in preterm-equivalent fetal sheep would improve white and gray matter recovery. Fetal sheep at 0.7 of gestation received sham asphyxia or asphyxia induced by umbilical cord occlusion for 25 minutes, followed by a continuous intracerebroventricular infusion of GDQ or vehicle from 1 to 4 hours (total dose 1.8 mg/kg). Sheep were killed 72 hours after asphyxia for histology. GDQ significantly improved survival of immature and mature oligodendrocytes (2′,3′-cyclic-nucleotide 3′-phosphodiesterase, CNPase) and total oligodendrocytes (oligodendrocyte transcription factor 2, Olig-2) within the periventricular and intragyral white matter. There were reduced numbers of cells showing cleaved caspase-3 positive apoptosis and astrogliosis (glial fibrillary acidic protein, GFAP) in both white matter regions. Neuronal survival was increased in the dentate gyrus, caudate and medial thalamic nucleus. Central infusion of GDQ was associated with a robust increase in fetal plasma concentrations of the anti-inflammatory cytokines, interferon-β (IFN-β) and interleukin-10 (IL-10), with no significant change in the concentration of the pro-inflammatory cytokine, tumor necrosis factor-α (TNF-α). In conclusion, delayed administration of the TLR7 agonist, GDQ, after severe hypoxia-ischemia in the developing brain markedly ameliorated white and gray matter damage, in association with upregulation of anti-inflammatory cytokines. These data strongly support the hypothesis that modulation of secondary inflammation may be a viable therapeutic target for injury of the preterm brain.
Collapse
|
21
|
Tripathi SK, Välikangas T, Shetty A, Khan MM, Moulder R, Bhosale SD, Komsi E, Salo V, De Albuquerque RS, Rasool O, Galande S, Elo LL, Lahesmaa R. Quantitative Proteomics Reveals the Dynamic Protein Landscape during Initiation of Human Th17 Cell Polarization. iScience 2018; 11:334-355. [PMID: 30641411 PMCID: PMC6330361 DOI: 10.1016/j.isci.2018.12.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 08/08/2018] [Accepted: 12/20/2018] [Indexed: 12/18/2022] Open
Abstract
Th17 cells contribute to the pathogenesis of inflammatory and autoimmune diseases and cancer. To reveal the Th17 cell-specific proteomic signature regulating Th17 cell differentiation and function in humans, we used a label-free mass spectrometry-based approach. Furthermore, a comprehensive analysis of the proteome and transcriptome of cells during human Th17 differentiation revealed a high degree of overlap between the datasets. However, when compared with corresponding published mouse data, we found very limited overlap between the proteins differentially regulated in response to Th17 differentiation. Validations were made for a panel of selected proteins with known and unknown functions. Finally, using RNA interference, we showed that SATB1 negatively regulates human Th17 cell differentiation. Overall, the current study illustrates a comprehensive picture of the global protein landscape during early human Th17 cell differentiation. Poor overlap with mouse data underlines the importance of human studies for translational research. Quantitative proteomics analysis of early human Th17 cell polarization The proteome and transcriptome highly correlate during early Th17 polarization Poor overlap of proteome profiles of human and mouse during early Th17 polarization The results underline the importance of human studies for translational research
Collapse
Affiliation(s)
- Subhash K Tripathi
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6, FI-20520 Turku, Finland
| | - Tommi Välikangas
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6, FI-20520 Turku, Finland; Doctoral Programme in Mathematics and Computer Sciences (MATTI), University of Turku, University Hill, FI-20014 Turku, Finland
| | - Ankitha Shetty
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6, FI-20520 Turku, Finland; Centre of Excellence in Epigenetics, Department of Biology, Indian Institute of Science Education and Research (IISER), Pune 411008, India
| | - Mohd Moin Khan
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6, FI-20520 Turku, Finland; Turku Doctoral Programme of Molecular Medicine (TuDMM), University of Turku, Tykistökatu 6, FI-20520 Turku, Finland
| | - Robert Moulder
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6, FI-20520 Turku, Finland
| | - Santosh D Bhosale
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6, FI-20520 Turku, Finland; Turku Doctoral Programme of Molecular Medicine (TuDMM), University of Turku, Tykistökatu 6, FI-20520 Turku, Finland
| | - Elina Komsi
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6, FI-20520 Turku, Finland
| | - Verna Salo
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6, FI-20520 Turku, Finland; Turku Doctoral Programme of Molecular Medicine (TuDMM), University of Turku, Tykistökatu 6, FI-20520 Turku, Finland
| | - Rafael Sales De Albuquerque
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6, FI-20520 Turku, Finland
| | - Omid Rasool
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6, FI-20520 Turku, Finland
| | - Sanjeev Galande
- Centre of Excellence in Epigenetics, Department of Biology, Indian Institute of Science Education and Research (IISER), Pune 411008, India
| | - Laura L Elo
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6, FI-20520 Turku, Finland.
| | - Riitta Lahesmaa
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6, FI-20520 Turku, Finland.
| |
Collapse
|
22
|
Boeck A, Landgraf-Rauf K, Vogelsang V, Siemens D, Prazeres da Costa O, Klucker E, von Mutius E, Buch T, Mansmann U, Schaub B. Ca 2+ and innate immune pathways are activated and differentially expressed in childhood asthma phenotypes. Pediatr Allergy Immunol 2018; 29:823-833. [PMID: 30102794 DOI: 10.1111/pai.12971] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/26/2018] [Accepted: 07/26/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Asthma is the most common chronic disease in children. Underlying immunologic mechanisms-in particular of different phenotypes-are still just partly understood. The objective of the study was the identification of distinct cellular pathways in allergic asthmatics (AA) and nonallergic asthmatics (NA) vs healthy controls (HC). METHODS Peripheral blood mononuclear cells (PBMCs) of steroid-naïve children (n(AA/NA/HC) = 35/13/34)) from the CLARA study (n = 275) were stimulated (anti-CD3/CD28, LpA) or kept unstimulated. Gene expression was investigated by transcriptomics and quantitative RT-PCR. Differentially regulated pathways between phenotypes were assessed after adjustment for sex and age (KEGG pathways). Networks based on correlations of gene expression were built using force-directed graph drawing. RESULTS Allergic asthmatics vs NA and asthmatics overall vs HC showed significantly different expression of Ca2+ and innate immunity-associated pathways. PCR analysis confirmed significantly increased Ca2+ -associated gene regulation (ORMDL3 and ATP2A3) in asthmatics vs HC, most prominent in AA. Innate immunity receptors (LY75, TLR7), relevant for virus infection, were also upregulated in AA and NA compared to HC. AA and NA could be differentiated by increased ATP2A3 and FPR2 in AA, decreased CLEC4E in AA, and increased IFIH1 expression in NA following anti-CD3/28 stimulation vs unstimulated (fold change). CONCLUSIONS Ca2+ regulation and innate immunity response pattern to viruses were activated in PBMCs of asthmatics. Asthma phenotypes were differentially characterized by distinct regulation of ATP2A3 and expression of innate immune receptors (FPR2, CLEC4E, IFIH1). These genes may present promising targets for future in-depth investigation with the long-term goal of more phenotype-specific therapeutic interventions in asthmatics.
Collapse
Affiliation(s)
- Andreas Boeck
- Department of Asthma & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany
| | - Katja Landgraf-Rauf
- Department of Asthma & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany.,Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Vanessa Vogelsang
- Department of Asthma & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany
| | - Diana Siemens
- Department of Asthma & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany
| | | | - Elisabeth Klucker
- Department of Asthma & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany
| | - Erika von Mutius
- Department of Asthma & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany.,Member of the German Center for Lung Research (DZL), Munich, Germany.,Institute for Asthma and Allergy Prevention, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Thorsten Buch
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - Ulrich Mansmann
- Institute for Medical Information Processing, Biometry and Epidemiology, LMU, Munich, Germany
| | - Bianca Schaub
- Department of Asthma & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany.,Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
23
|
Bystander T Cells: A Balancing Act of Friends and Foes. Trends Immunol 2018; 39:1021-1035. [PMID: 30413351 DOI: 10.1016/j.it.2018.10.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/27/2018] [Accepted: 10/04/2018] [Indexed: 02/07/2023]
Abstract
T cell responses are essential for appropriate protection against pathogens. T cell immunity is achieved through the ability to discriminate between foreign and self-molecules, and this relies heavily on stringent T cell receptor (TCR) specificity. Recently, bystander activated T lymphocytes, that are specific for unrelated epitopes during an antigen-specific response, have been implicated in diverse diseases. Numerous infection models have challenged the classic dogma of T cell activation as being solely dependent on TCR and major histocompatibility complex (MHC) interactions, indicating an unappreciated role for pathogen-associated receptors on T cells. We discuss here the specific roles of bystander activated T cells in pathogenesis, shedding light on the ability of these cells to modulate disease severity independently from TCR recognition.
Collapse
|
24
|
Mo C, Zeng Z, Deng Q, Ding Y, Xiao R. Imbalance between T helper 17 and regulatory T cell subsets plays a significant role in the pathogenesis of systemic sclerosis. Biomed Pharmacother 2018; 108:177-183. [PMID: 30219674 DOI: 10.1016/j.biopha.2018.09.037] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 09/06/2018] [Accepted: 09/07/2018] [Indexed: 02/06/2023] Open
Abstract
Systemic sclerosis (SSc) is a rare autoimmune disease that is characterized by fibrosis, inflammation, and vasculopathy of the skin and internal organs. The etiopathogenesis of SSc remains unclear. However, the pivotal role of T lymphocytes with an aberrant immune response in SSc is well established. Among T cells, IL-17-producing helper T (Th17) cell and regulatory T (Treg) cell subsets have recently been found to play crucial roles in SSc pathogenesis. Generally speaking, Th17 cell subsets up-regulate inflammation, fibrosis, and autoimmunity, which are present in SSc, while Treg cell subsets have an immunosuppressive function and resist the immunological performance of Th17 cells. Up-to-date evidence has pointed out that the imbalance and abnormal functions of Th17/Treg cells may contribute to SSc. Therefore, this review aims to summarize the current understanding of the vital cytokines and signaling pathways that are involved in Th17/Treg differentiation and functions, and their roles in the pathogenesis of SSc, thus providing novel insights about targeting the Th17/Treg balance as a potential therapy for SSc treatment in the near future.
Collapse
Affiliation(s)
- Cuiling Mo
- Department of Dermatology, Second Xiangya Hospital, Central South University, 139 Ren-Min Road, Changsha 410011, China; Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, 410078, China.
| | - Zhuotong Zeng
- Department of Dermatology, Second Xiangya Hospital, Central South University, 139 Ren-Min Road, Changsha 410011, China.
| | - Qiancheng Deng
- Department of Dermatology, Second Xiangya Hospital, Central South University, 139 Ren-Min Road, Changsha 410011, China.
| | - Yan Ding
- Department of Dermatology, Hainan Provincial Dermatology Disease Hospital, 15 LongKun-Nan Road, Haikou 570206, China.
| | - Rong Xiao
- Department of Dermatology, Second Xiangya Hospital, Central South University, 139 Ren-Min Road, Changsha 410011, China.
| |
Collapse
|
25
|
Ghasiyari H, Rostami-Nejad M, Amani D, Rostami K, Pourhoseingholi MA, Asadzadeh-Aghdaei H, Zali MR. Diverse Profiles of Toll-Like Receptors 2, 4, 7, and 9 mRNA in Peripheral Blood and Biopsy Specimens of Patients with Celiac Disease. J Immunol Res 2018; 2018:7587095. [PMID: 30057921 PMCID: PMC6051003 DOI: 10.1155/2018/7587095] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/21/2018] [Accepted: 05/28/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND AIMS Both adaptive and innate immunity are involved in the development of celiac disease (CD). Altered Toll-like receptors (TLR) expression and activation may be partially responsible for the inflammation and subsequently crypt hyperplasia, but the main driver for inflammation is gliadin-reactive T-cells. Therefore, the aim of this study was to investigate the TLRs 2, 4, 7, and 9 gene expressions in both peripheral blood and intestinal mucosa of patients with celiac disease compared to healthy control (HC). MATERIAL AND METHODS Blood samples from 120 confirmed active CD patients and 120 age- and sex-matched healthy volunteers served as control group were collected during 2015-2016. Also, 20 biopsy specimens from the study group were randomly collected. Total RNA was isolated using a standard commercial kit. The mRNA expression of TLRs was quantified by relative qPCR with β2 microglobulin (β2m) as a reference gene. RESULTS TLR4 (P = 0.01) and TLR9 (P = 0.02) mRNA were significantly elevated in blood samples from CD patients compared to the healthy controls. Moreover, TLR2 (P = 0.03) and TLR4 (P = 0.0003) expression level was increased in CD biopsy specimens compared to controls, whereas expression of TLR9 mRNA was significantly decreased in CD patients. There was no significant difference in the expression of TLR7 in biopsy and blood specimens. CONCLUSIONS The alteration of TLR4 and TLR9 expression in the blood and biopsy samples of patients with CD supports the critical role of the innate immune system in the pathogenesis of this disease. Upregulation of TLR4 and TLR9 suggests the contribution of gut microbiota or dysregulation of the immune response to commensal flora in small bowel mucosa in celiac patients.
Collapse
Affiliation(s)
- Haniye Ghasiyari
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Rostami-Nejad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davar Amani
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamran Rostami
- Department of Gastroenterology, Milton Keynes University Hospital, Milton Keynes, UK
| | - Mohamad Amin Pourhoseingholi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh-Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
Sciorati C, Monno A, Doglio MG, Rigamonti E, Ascherman DP, Manfredi AA, Rovere-Querini P. Exacerbation of Murine Experimental Autoimmune Myositis by Toll-Like Receptor 7/8. Arthritis Rheumatol 2018; 70:1276-1287. [PMID: 29569859 DOI: 10.1002/art.40503] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 03/15/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Toll-like receptor 7 (TLR-7), TLR-8, and interferon (IFN)-induced genes are expressed in patients with idiopathic inflammatory myositis. This study was undertaken to investigate whether their activation influences the natural history of the disease. METHODS Experimental autoimmune myositis was induced in mice by injection of the amino-terminal portion of the murine histidyl-transfer RNA synthetase (HisRS). Disease was compared in the presence or the absence of the TLR-7/8 agonist R-848 in wild-type mice and in mice that fail to express the IFNα/β receptor (IFNα/βR-null mice). RESULTS Experimental autoimmune myositis induced by a single intramuscular immunization with HisRS spontaneously abated after 7-8 weeks. In contrast, levels of anti-HisRS autoantibodies, endomysial/perimysial leukocyte infiltration, and myofiber regeneration persisted at the end of the follow-up period (22 weeks after immunization) in mice immunized with HisRS in the presence of R-848. Myofiber major histocompatibility complex (MHC) class I molecules were detectable only in mice immunized with both HisRS and R-848. MHC up-regulation occurred early and in muscles that were not directly injected with HisRS. Muscle MHC expression paralleled with leukocyte infiltration. MHC class I molecules were selectively up-regulated in myotubes challenged with R-848 in vitro. Type I IFN was necessary for the prolonged autoantibody response and for the spreading of the autoimmune response, as demonstrated using IFNα/βR-null mice. Muscle infiltration was maintained in the injected muscle up to the end of the follow-up period. CONCLUSION TLR-7/8 activation is necessary to induce and maintain a systemic autoimmune response targeting the skeletal muscle. This experimental autoimmune myositis model reproduces many characteristics of human idiopathic inflammatory myopathies and may represent a tool for preclinical studies.
Collapse
Affiliation(s)
- Clara Sciorati
- IRCCS Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Antonella Monno
- IRCCS Ospedale San Raffaele Scientific Institute, Milan, Italy
| | | | - Elena Rigamonti
- IRCCS Ospedale San Raffaele Scientific Institute, Milan, Italy
| | | | - Angelo A Manfredi
- IRCCS Ospedale San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milan, Italy
| | - Patrizia Rovere-Querini
- IRCCS Ospedale San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
27
|
The effects of resiquimod in an ovalbumin-induced allergic rhinitis model. Int Immunopharmacol 2018; 59:233-242. [PMID: 29665497 DOI: 10.1016/j.intimp.2018.04.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/30/2018] [Accepted: 04/09/2018] [Indexed: 12/14/2022]
Abstract
Growing evidence indicates that the Toll-like receptor7/8(TLR7/8) agonist resiquimod (R848) is a potential inhibitor of type-2 immunity. However, the mechanisms mediating its therapeutic effects are not fully understood. This study investigated the effects of R848 on OVA-induced allergic rhinitis(AR) mice and the expression of IL-25, IL-33, TSLP, T-cell immunoglobulin mucin1 (TIM1) and T-cell immunoglobulin mucin3 (TIM3). BALB/c mice were intranasally sensitized and challenged with ovalbumin (OVA), and R848 was intraperitoneally injected into AR mice. Histological changes in the nasal mucosa were evaluated by hematoxylin and eosin (H & E) and Periodic Acid-Schiff (PAS) staining; cytokine levels in serum were measured with enzyme-linked immunosorbent assays (ELISAs);the mRNA expression levels of IFN-γ, IL-17 and Foxp3 in the spleen determined by quantitative real-time RT-PCR (qRT-PCR); the proportions of Th1, Th2, Th17, Treg and TIM3 + IFN-γ + Th1 cells in the spleen were assessed with flow cytometry; TIM1, TIM3 and IL-33 expression levels in the nasal mucosa were evaluated with immunofluorescence staining(IF).R848 alleviated the nasal allergic symptoms; reduced eosinophil cell infiltration, goblet cell hyperplasia in the nasal mucosa; reduced IL-13, IL-17, IL-25 and IL-33 levels in serum; upregulated the relative mRNA expression of IFN-γ and Foxp3, and downregulated the relative mRNA expression of IL-17 in the spleen; decreased Th2, Th17 and TIM3 + IFN-γ + Th1 cells ratios, increased the proportion of Th1 and Treg cells in the spleen; suppressed TIM1 and TIM3,but increased IL-33 expression in the nasal mucosa in OVA-induced AR mice. R848 suppresses IL-25, IL-33 released and TIM1, TIM3 expression, which may contribute to its anti-allergic effects.
Collapse
|
28
|
Huang L, Xu H, Peng G. TLR-mediated metabolic reprogramming in the tumor microenvironment: potential novel strategies for cancer immunotherapy. Cell Mol Immunol 2018; 15:428-437. [PMID: 29553135 PMCID: PMC6068099 DOI: 10.1038/cmi.2018.4] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 12/28/2017] [Accepted: 12/29/2017] [Indexed: 02/06/2023] Open
Abstract
Cellular energy metabolism not only promotes tumor cell growth and metastasis but also directs immune cell survival, proliferation and the ability to perform specific and functional immune responses within the tumor microenvironment. A better understanding of the molecular regulation of metabolism in different cell components in the tumor-suppressive microenvironment is critical for the development of effective strategies for human cancer treatments. Toll-like receptors (TLRs) have recently been recognized as critical factors involved in tumor pathogenesis, regulating both tumor cells and tumor-infiltrating innate and adaptive immune cells. However, little is known about the molecular crosstalk between TLR signaling and tumor or/and immune cell metabolism, although there is abundant expression of TLRs in these cells. In this review, we explore the functional role of TLR signaling in reprogramming cell metabolism in the tumor microenvironment. In particular, we discuss how malignant tumors regulate metabolism to support their growth and survival, summarize more recently identified metabolic profiles of different immune cell subsets and TLR-mediated regulation of cellular metabolism in both tumor and immune cells, and further explore potential strategies targeting cell metabolism for TLR-based cancer therapy. An improved understanding of these issues should open new avenues for the development of novel strategies via TLR-mediated metabolic reprogramming of the tumor microenvironment for cancer immunotherapy.
Collapse
Affiliation(s)
- Lan Huang
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal Medicine, Saint Louis University School of Medicine, 63104, Saint Louis, MO, USA.,Department of Microbiology and Immunology, Jiangsu University School of Medicine, 212013, Zhenjiang, China
| | - Huaxi Xu
- Department of Microbiology and Immunology, Jiangsu University School of Medicine, 212013, Zhenjiang, China
| | - Guangyong Peng
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal Medicine, Saint Louis University School of Medicine, 63104, Saint Louis, MO, USA.
| |
Collapse
|
29
|
Dolasia K, Bisht MK, Pradhan G, Udgata A, Mukhopadhyay S. TLRs/NLRs: Shaping the landscape of host immunity. Int Rev Immunol 2017; 37:3-19. [PMID: 29193992 DOI: 10.1080/08830185.2017.1397656] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Innate immune system provides the first line of defense against pathogenic organisms. It has a varied and large collection of molecules known as pattern recognition receptors (PRRs) which can tackle the pathogens promptly and effectively. Toll-like receptors (TLRs) and NOD-like receptors (NLRs) are members of the PRR family that recognize pathogen associated molecular patterns (PAMPs) and play pivotal roles to mediate defense against infections from bacteria, fungi, virus and various other pathogens. In this review, we discuss the critical roles of TLRs and NLRs in the regulation of host immune-effector functions such as cytokine production, phagosome-lysosome fusion, inflammasome activation, autophagy, antigen presentation, and B and T cell immune responses that are known to be essential for mounting a protective immune response against the pathogens. This review may be helpful to design TLRs/NLRs based immunotherapeutics to control various infections and pathophysiological disorders.
Collapse
Affiliation(s)
- Komal Dolasia
- a Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD) , Tuljaguda Complex, Nampally, Hyderabad , India
| | - Manoj K Bisht
- a Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD) , Tuljaguda Complex, Nampally, Hyderabad , India
| | - Gourango Pradhan
- a Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD) , Tuljaguda Complex, Nampally, Hyderabad , India
| | - Atul Udgata
- a Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD) , Tuljaguda Complex, Nampally, Hyderabad , India
| | - Sangita Mukhopadhyay
- a Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD) , Tuljaguda Complex, Nampally, Hyderabad , India
| |
Collapse
|