1
|
Hong BK, You S, Kim JG, Kim M, Lee N, Lee K, Baek IP, Ju JH, Kim WU, Kim HY. Upregulation of interferon-γ response genes in monocytes and T cells identified by single-cell transcriptomics in patients with anti-citrullinated peptide antibody-positive early rheumatoid arthritis. Front Immunol 2025; 15:1439082. [PMID: 39877346 PMCID: PMC11772891 DOI: 10.3389/fimmu.2024.1439082] [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: 05/27/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction Our aim was to investigate the insufficiently understood differences in the immune system between anti-citrullinated peptide antibody (ACPA)-positive (ACPA+) and ACPA-negative (ACPA-) early rheumatoid arthritis (eRA) patients. Methods We performed multiple cytokine assays using sera from drug-naïve ACPA+ and ACPA- eRA patients. Additionally, we conducted single-cell RNA sequencing of CD45+ cells from peripheral blood samples to analyze and compare the distribution and functional characteristics of the cell subsets based on the ACPA status. Results Serum concentrations of interferon-γ (IFN-γ) and interleukin (IL)-12 were higher in ACPA+ eRA than in ACPA- eRA. Single-cell transcriptome analysis of 37,318 cells identified 17 distinct cell types and revealed the expansion of IL1B+ proinflammatory monocytes, IL7R+ T cells, and CD8+ CCL4+ T cells in ACPA+ eRA. Furthermore, we observed an enrichment of IFN-γ response genes in nearly all monocytes and T cells of ACPA+ eRA subsets. Heightened interactions between IFN-γ and IFN-γ receptors were observed in ACPA+ eRA, particularly between monocytes and T cells. We examined IFITM2 and IFITM3 as potential key markers in ACPA+ eRA given their pronounced upregulation and association with the IFN response. Specifically, the expression of these genes was elevated in IL1B+ proinflammatory monocytes (likely M1 monocytes), correlating with serum IFN-γ levels. Discussion Compared to ACPA- eRA, ACPA+ eRA showed higher serum IFN-γ and IL-12 levels, upregulated IFN-γ response genes, and enhanced IFN-γ-driven monocyte-T cell interactions. These distinct immune features of the peripheral circulation in ACPA+ eRA suggest a role for type 1 helper T cell-related immunity in its pathogenesis.
Collapse
Affiliation(s)
- Bong-Ki Hong
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sungyong You
- Urology and Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Jung Gon Kim
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Rheumatology, Department of Internal Medicine, Inje University Ilsan Paik Hospital, Goyang, Republic of Korea
| | - Minhyung Kim
- Urology and Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Naeun Lee
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kijun Lee
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- YiPSCELL, Inc., Seoul, Republic of Korea
| | | | - Ji Hyeon Ju
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- YiPSCELL, Inc., Seoul, Republic of Korea
- Department of Internal Medicine, The Catholic University of Korea, Seoul St. Mary’s Hospital, Seoul, Republic of Korea
| | - Wan-Uk Kim
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Internal Medicine, The Catholic University of Korea, Seoul St. Mary’s Hospital, Seoul, Republic of Korea
| | - Ho-Youn Kim
- The Catholic University of Korea and Ho-Youn Kim’s Clinic for Arthritis Rheumatism, Seoul, Republic of Korea
| |
Collapse
|
2
|
Ahmed B, Aliyu M, Getso MI, Bala JA, Ahmed RJ, Kabuga AI, Adamu AMY, Yusuf AA. Exploring the impact of interferon-gamma single nucleotide polymorphisms on HTLV-1 infection: Unraveling genetic influences in viral pathogenesis. Crit Rev Oncol Hematol 2025; 207:104614. [PMID: 39798937 DOI: 10.1016/j.critrevonc.2025.104614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/26/2024] [Accepted: 01/05/2025] [Indexed: 01/15/2025] Open
Abstract
Human T-lymphotropic virus-1 (HTLV-1) induces neoplastic adult T-cell leukemia/lymphoma (ATLL) and neurological HTLV-1 associated myelopathy (HAM) in approximately 3 %-5 % of infected individuals. The precise factors that facilitate disease manifestation are still unknown; interaction between the virus and the host's immune response is key. Cytokines regulates physiological activities and their dysregulation may initiate the pathogenesis of various malignant and infectious diseases. Genetic variations, particularly polymorphisms in gene regulatory regions, lead to varying cytokine production patterns. Interferon-gamma (IFN-γ), a key cytokine in HTLV-1 infection, is a signature cytokine for T-helper 1 (Th1) cells that interferes with viral replication and enhances innate and adaptive immune responses during viral infections. The IFNG gene possesses several single nucleotide polymorphisms (SNPs), among which the + 874 A/T SNP has been widely studied for its functional role in HTLV-1 infection. The purpose of this review was to provide insight into the impact of IFNG SNPs on HTLV-1 Infection.
Collapse
Affiliation(s)
- Bilkisu Ahmed
- Department of Medical Microbiology and Parasitology, Faculty of Basic Clinical Sciences, Bayero University Kano, P.M.B. 3011, Kano, Nigeria
| | - Mansur Aliyu
- Department of Medical Microbiology and Parasitology, Faculty of Basic Clinical Sciences, Bayero University Kano, P.M.B. 3011, Kano, Nigeria.
| | - Muhammad Ibrahim Getso
- Department of Medical Microbiology and Parasitology, Faculty of Basic Clinical Sciences, Bayero University Kano, P.M.B. 3011, Kano, Nigeria
| | - Jamilu Abubakar Bala
- Department of Medical Laboratory Science, Faculty of Allied Health Sciences, Bayero University Kano, P.M.B. 3011, Kano, Nigeria; Centre for Infectious Diseases Research, Bayero University Kano, P.M.B 3011, Kano, Nigeria
| | - Ramat Jummai Ahmed
- Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Ahmadu Bello University Zaria, P.M.B 1044, Zaria, Kaduna State, Nigeria
| | - Auwal Idris Kabuga
- Department of Medical Microbiology and Parasitology, Faculty of Basic Clinical Sciences, Bayero University Kano, P.M.B. 3011, Kano, Nigeria
| | - Al-Muktar Yahuza Adamu
- Department of Medical Microbiology and Parasitology, Faculty of Basic Clinical Sciences, Bayero University Kano, P.M.B. 3011, Kano, Nigeria
| | - Aminu Abba Yusuf
- Department of Haematology, Bayero University Kano and Aminu Kano Teaching Hospital, Kano, Nigeria
| |
Collapse
|
3
|
Cebeci S, Polat T, Ünübol N. Roles of NET Peptides With Known Antimicrobial Activity and Toxicity in Immune Response. J Immunol Res 2024; 2024:5528446. [PMID: 39759156 PMCID: PMC11698612 DOI: 10.1155/jimr/5528446] [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: 08/24/2024] [Accepted: 11/09/2024] [Indexed: 01/07/2025] Open
Abstract
Antimicrobial peptides (AMPs) are crucial components of the innate immune system in all living organisms, playing a vital role in the body's defense against diseases and infections. The immune system's primary functions include preventing disease-causing agents from entering the body and eliminating them without causing harm. These peptides exhibit broad-spectrum activity against bacteria, viruses, fungi, parasites, and cancer cells. They are secreted by innate and epithelial cells and contribute to host defense by inducing cellular activities such as cell migration, proliferation, differentiation, cytokine production, angiogenesis, and wound healing. In response to the growing challenge of bacterial resistance to antimicrobial agents, alternative drugs and new antibacterial molecules are being explored. In a previous study, NET AMPs were synthesized and their antimicrobial effects were determined. The current study extends this work by assessing the effects of these peptides on the immune system through cell culture experiments and ELISA. Specifically, the study investigated how different concentrations of these peptides influence the secretion of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and interferon-γ (IFN-γ) in mouse macrophages. Among the synthesized peptides, NET1 and NET2 demonstrated low cytotoxicity in TIB-71 RAW 264.7 macrophages. These peptides induced an anti-inflammatory response and reduced IL-6 expression in the absence of LPS stimulation, while simultaneously increasing IFN-γ and TNF-α secretion. These findings suggest that NET1 and NET2 peptides possess both anti-inflammatory and pro-inflammatory properties, highlighting their potential role in modulating immune responses.
Collapse
Affiliation(s)
- Sinan Cebeci
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Türkiye
| | - Tuba Polat
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Türkiye
| | - Nihan Ünübol
- Department of Medical Microbiology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Türkiye
- Medical Laboratory Technician Program, Vocational School of Health Services, Acibadem Mehmet Ali Aydinlar University, Istanbul, Türkiye
| |
Collapse
|
4
|
Tian Y, Dai J, Yang Y, Guo X, Wang W, Li F, Wang J, Liu R. Relationship between the risk of intestinal mucosal Epstein-Barr virus and/or cytomegalovirus infection and peripheral blood NK cells numbers in patients with ulcerative colitis: a cross-sectional study in Chinese population. Front Microbiol 2024; 15:1498483. [PMID: 39697654 PMCID: PMC11652489 DOI: 10.3389/fmicb.2024.1498483] [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: 09/19/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024] Open
Abstract
Objective This study aimed to analyze the relationship between the risk of common opportunistic pathogens Epstein-Barr virus (EBV) and cytomegalovirus (CMV) infection in intestinal mucosal tissues of Ulcerative Colitis (UC) patients and the number of peripheral blood NK cells. Methods UC patients admitted to a third-grade class-A hospital from January 2018 to December 2023 were selected as research population. Clinical data of the patients were collected from the electronic medical record system. Additionally, samples of intestinal mucosal tissues were obtained for real-time fluorescence quantitative PCR to detect and analyze the viral load of CMV and EBV. Blood samples were collected for lymphocyte subsets analysis. Multivariable logistic regression models analyses was used to determine the odds ratio (OR) and 95% confidence interval (95% CI) for the independent association between NK cells and EBV/CMV infections in UC. We further applied the restricted cubic spline analysis and smooth curve fitting to examine the non-linear relationship between them. Results 378 UC patients were enrolled. Of these patients, there were 194 patients (51.32%) with EBV /CMV infection. In multivariable logistic regression analyses NK cells was independently associated with EBV and/or CMV infection after adjusted potential confounders (OR 8.24, 95% CI 3.75-18.13, p < 0.001). A nonlinear relationship was found between NK cells and EBV/CMV infections, which had a threshold around 10.169. The effect sizes and CIs below and above the threshold were 0.535 (0.413-0.692), p < 0.001 and 1.034 (0.904-1.183), p > 0.05, respectively. Conclusion There was a non-linear relationship between NK cells and EBV/CMV infections. The risk for EBV/CMV infections was not increased with increasing NK cells in individuals with NK cells ≥ 10.169%, whereas the risk for EBV and/or CMV infection was increased with an decreasing NK cells in those with NK cells < 10.169%. The risk of EBV/CMV infections increases when NK cells were below a certain level.
Collapse
Affiliation(s)
- Ye Tian
- Department of Gastroenterology, Shanxi Provincial People’s Hospital, National Clinical Research Center for Digestive Diseases, Shanxi Inflammatory Bowel Disease Center, Taiyuan, China
| | - Jinghua Dai
- School of Nursing, Shanxi Medical University, Shanxi Provincial People’s Hospital, Taiyuan, China
| | - Yunfeng Yang
- Department of Gastroenterology, Shanxi Provincial People’s Hospital, National Clinical Research Center for Digestive Diseases, Shanxi Inflammatory Bowel Disease Center, Taiyuan, China
| | - Xiaofeng Guo
- Department of Gastroenterology, Shanxi Provincial People’s Hospital, National Clinical Research Center for Digestive Diseases, Shanxi Inflammatory Bowel Disease Center, Taiyuan, China
| | - Wei Wang
- Department of Laboratory Medicine, Shanxi Provincial People’s Hospital, Taiyuan, China
| | - Fengxia Li
- Department of Gastroenterology, Shanxi Provincial People’s Hospital, National Clinical Research Center for Digestive Diseases, Shanxi Inflammatory Bowel Disease Center, Taiyuan, China
| | - Juzi Wang
- Nursing Department, Shanxi Provincial People’s Hospital, Taiyuan, China
| | - Ruiyun Liu
- Shanxi Children’s Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| |
Collapse
|
5
|
Wu Z, Lin X, Yuan G, Li N, Xu R. Innate lymphoid cells: New players in osteoimmunology. Eur J Immunol 2024; 54:e2350381. [PMID: 38234001 DOI: 10.1002/eji.202350381] [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: 03/22/2023] [Revised: 12/21/2023] [Accepted: 01/09/2024] [Indexed: 01/19/2024]
Abstract
Innate lymphoid cells (ILCs) are the most recently identified immune cell types existing in lymphoid and nonlymphoid organs. Albeit they lack the expression of antigen receptors, ILCs play vital roles in innate immune responses by producing multiple effector cytokines. The ILC family includes conventional natural killer cells and cytokine-producing ILCs, which are divided into group 1, group 2, and group 3 ILCs based on their effector cytokines and developmental requirements. Emerging evidence has indicated that ILCs are essential immune regulators of bone homeostasis, playing a critical role in osteoimmunology. In this mini-review, we discuss recent advances in the understanding of ILC functions in bone homeostasis under physiological and pathological conditions, with an emphasis on the communication between ILCs and bone cells including osteoclasts and osteoblasts, as well as the underlying immunoregulatory networks involving ILC-derived cytokines and growth factors. This review also discusses future research directions and the potential of targeting ILCs for the treatment of inflammation-associated bone disorders.
Collapse
Affiliation(s)
- Zuoxing Wu
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cells, State Key Laboratory of Cellular Stress Biology, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Xixi Lin
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cells, State Key Laboratory of Cellular Stress Biology, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Guixin Yuan
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cells, State Key Laboratory of Cellular Stress Biology, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Na Li
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cells, State Key Laboratory of Cellular Stress Biology, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Ren Xu
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cells, State Key Laboratory of Cellular Stress Biology, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
6
|
Cai L, Lv Y, Yan Q, Guo W. Cytokines: The links between bone and the immune system. Injury 2024; 55:111203. [PMID: 38043143 DOI: 10.1016/j.injury.2023.111203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 11/05/2023] [Accepted: 11/12/2023] [Indexed: 12/05/2023]
Abstract
Osteoporosis results from an imbalance in a highly balanced physiological process called bone remodeling, in which osteoclast-mediated bone resorption and osteoblast-mediated bone formation play important roles. Osteoimmunology is a newly discovered interdisciplinary research field that focuses on the relationship between bone and the immune system. Specifically, bone and the immune system interact through cytokines, immune cells secrete cytokines, and cytokines finely regulate bone metabolism by mediating the differentiation and activity of osteoclasts and osteoblasts. Therefore, understanding the influence of cytokines on bone metabolism is conducive for the development of novel targeted drugs against immune-related bone diseases. This review summarizes the pathophysiological functions of various common cytokines in bone and discusses the potential clinical value of multiple cytokines in immune-mediated bone diseases.
Collapse
Affiliation(s)
- Liping Cai
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, China; Department of Endocrinology, Rheumatology and Immunology, Anyang People's Hospital, Anyang, Henan 455000, China
| | - You Lv
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Qihui Yan
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Weiying Guo
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
7
|
Chi Z, Yang H, Liu J. Study on the combined toxicity of DEHP and lead on the blood system of rats. CHEMOSPHERE 2024; 349:140908. [PMID: 38072204 DOI: 10.1016/j.chemosphere.2023.140908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/01/2023] [Accepted: 12/03/2023] [Indexed: 01/10/2024]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is a commonly used phthalate ester compound, while lead is a persistent and bioaccumulative heavy metal. Both can be exposed to the body through a variety of ways, which may have an impact on the blood system. In this study, we examined the impact of co-exposure to DEHP (0, 10, 100 mg/kg) and Pb (0, 5, 50 mg/kg) on the blood system of male SD rats. The study revealed that continuous exposure to DEHP and Pb for 20 days resulted in a decrease in leukocytes and lymphocytes, while an increase in neutrophils and monocytes. Co-exposure led to a significant decrease in the spleen coefficients. Furthermore, the combined exposure could increase the ratio of bone marrow cells in G1 phase, and decrease the ratio of cells in S phase and G2 phase. Cytokine testing showed that combined exposure affects the secretion of hematopoietic factors and may cause bone marrow cell apoptosis. Single or combined exposure to DEHP and Pb can cause oxidative stress in serum and bone marrow. Overall, these results indicate that the co-exposure of DEHP and Pb adversely affected the blood system of rats, mainly due to the induction of oxidative stress and ultimately affects the secretion of cytokines. The combined effect of the two substances is primarily antagonistic. These results have important implications for the risk assessment of combined pollution and provide valuable theoretical guidance.
Collapse
Affiliation(s)
- Zhenxing Chi
- School of Marine Science and Technology, Harbin Institute of Technology, Weihai, 264209, China; Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Hangzhou, 310015, China.
| | - Hanfeng Yang
- School of Marine Science and Technology, Harbin Institute of Technology, Weihai, 264209, China
| | - Jia Liu
- School of Marine Science and Technology, Harbin Institute of Technology, Weihai, 264209, China
| |
Collapse
|
8
|
Talreja J, Peng C, Samavati L. MIF modulates p38/ERK phosphorylation via MKP-1 induction in sarcoidosis. iScience 2024; 27:108746. [PMID: 38299032 PMCID: PMC10829885 DOI: 10.1016/j.isci.2023.108746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 09/21/2023] [Accepted: 12/12/2023] [Indexed: 02/02/2024] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a versatile cytokine that influences a variety of cellular processes important for immune regulation and tissue homeostasis. Sarcoidosis is a granulomatous disease characterized by extensive local inflammation and increased T helper cell mediated cytokines. We have shown that MIF has a modulatory role in cytokine networks in sarcoidosis. We investigated the effect of exogenous MIF on sarcoidosis alveolar macrophages (AMs), CD14+ monocytes and peripheral blood mononuclear cells (PBMCs). Our results showed that MIF negatively regulates the increased MAPKs (pp38 and pERK1/2) activation by inducing Mitogen-activated protein kinase phosphatase (MKP)-1. We found that MIF decreased IL-6 and IL-1β production, increased the percentage of regulatory T-cells (Tregs), and induced IL-1R antagonist (IL-1RA) and IL-10 production. Thus, the results of our study suggest that exogenous MIF modulates MAPK activation by inducing MKP-1and Tregs as well as IL-10 and IL-1RA, and hence plays a modulatory role in immune activation in sarcoidosis.
Collapse
Affiliation(s)
- Jaya Talreja
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Wayne State University, School of Medicine and Detroit Medical Center, Detroit, MI 48201, USA
| | - Changya Peng
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Wayne State University, School of Medicine and Detroit Medical Center, Detroit, MI 48201, USA
| | - Lobelia Samavati
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Wayne State University, School of Medicine and Detroit Medical Center, Detroit, MI 48201, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI 48201, USA
| |
Collapse
|
9
|
Rožmanić C, Lisnić B, Pribanić Matešić M, Mihalić A, Hiršl L, Park E, Lesac Brizić A, Indenbirken D, Viduka I, Šantić M, Adler B, Yokoyama WM, Krmpotić A, Juranić Lisnić V, Jonjić S, Brizić I. Perinatal murine cytomegalovirus infection reshapes the transcriptional profile and functionality of NK cells. Nat Commun 2023; 14:6412. [PMID: 37828009 PMCID: PMC10570381 DOI: 10.1038/s41467-023-42182-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 10/02/2023] [Indexed: 10/14/2023] Open
Abstract
Infections in early life can elicit substantially different immune responses and pathogenesis than infections in adulthood. Here, we investigate the consequences of murine cytomegalovirus infection in newborn mice on NK cells. We show that infection severely compromised NK cell maturation and functionality in newborns. This effect was not due to compromised virus control. Inflammatory responses to infection dysregulated the expression of major transcription factors governing NK cell fate, such as Eomes, resulting in impaired NK cell function. Most prominently, NK cells from perinatally infected mice have a diminished ability to produce IFN-γ due to the downregulation of long non-coding RNA Ifng-as1 expression. Moreover, the bone marrow's capacity to efficiently generate new NK cells is reduced, explaining the prolonged negative effects of perinatal infection on NK cells. This study demonstrates that viral infections in early life can profoundly impact NK cell biology, including long-lasting impairment in NK cell functionality.
Collapse
Affiliation(s)
- Carmen Rožmanić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Berislav Lisnić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | | | - Andrea Mihalić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Lea Hiršl
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Eugene Park
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ana Lesac Brizić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Daniela Indenbirken
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Ina Viduka
- Department of Microbiology and Parasitology, University of Rijeka, Faculty of Medicine, Rijeka, Croatia
| | - Marina Šantić
- Department of Microbiology and Parasitology, University of Rijeka, Faculty of Medicine, Rijeka, Croatia
| | - Barbara Adler
- Max von Pettenkofer Institute & Gene Center, Virology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Wayne M Yokoyama
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Astrid Krmpotić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Vanda Juranić Lisnić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Stipan Jonjić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.
| | - Ilija Brizić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.
| |
Collapse
|
10
|
Joshi JC, Joshi B, Zhang C, Banerjee S, Vellingiri V, Raghunathrao VAB, Zhang L, Amin R, Song Y, Mehta D. RGS2 is an innate immune checkpoint for TLR4 and Gαq-mediated IFNγ generation and lung injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.22.559016. [PMID: 37790514 PMCID: PMC10542520 DOI: 10.1101/2023.09.22.559016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
IFNγ, a type II interferon secreted by immune cells, augments tissue responses to injury following pathogenic infections leading to lethal acute lung injury (ALI). Alveolar macrophages (AM) abundantly express Toll-like receptor-4 and represent the primary cell type of the innate immune system in the lungs. A fundamental question remains whether AM generation of IFNg leads to uncontrolled innate response and perpetuated lung injury. LPS induced a sustained increase in IFNg levels and unresolvable inflammatory lung injury in the mice lacking RGS2 but not in RGS2 null chimeric mice receiving WT bone marrow or receiving the RGS2 gene in AM. Thus, indicating RGS2 serves as a gatekeeper of IFNg levels in AM and thereby lung's innate immune response. RGS2 functioned by forming a complex with TLR4 shielding Gaq from inducing IFNg generation and AM inflammatory signaling. Thus, inhibition of Gaq blocked IFNg generation and subverted AM transcriptome from being inflammatory to reparative type in RGS2 null mice, resolving lung injury. Highlights RGS2 levels are inversely correlated with IFNγ in ARDS patient's AM.RGS2 in alveolar macrophages regulate the inflammatory lung injury.During pathogenic insult RGS2 functioned by forming a complex with TLR4 shielding Gαq from inducing IFNγ generation and AM inflammatory signaling. eToc Blurb Authors demonstrate an essential role of RGS2 in macrophages in airspace to promoting anti-inflammatory function of alveolar macrophages in lung injury. The authors provided new insight into the dynamic control of innate immune response by Gαq and RGS2 axis to prevent ALI.
Collapse
|
11
|
Dimitroglou Y, Aggeli C, Theofilis P, Tsioufis P, Oikonomou E, Chasikidis C, Tsioufis K, Tousoulis D. Novel Anti-Inflammatory Therapies in Coronary Artery Disease and Acute Coronary Syndromes. Life (Basel) 2023; 13:1669. [PMID: 37629526 PMCID: PMC10455741 DOI: 10.3390/life13081669] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/27/2023] [Accepted: 07/30/2023] [Indexed: 08/27/2023] Open
Abstract
Evidence suggests that inflammation plays an important role in atherosclerosis and the consequent clinical presentation, including stable coronary artery disease (CAD) and acute coronary syndromes (ACS). The most essential elements are cytokines, proteins with hormone-like properties that are produced by the immune cells, endothelial cells, platelets, fibroblasts, and some stromal cells. Interleukins (IL-1β and IL-6), chemokines, interferon-γ (IFN-γ), and tumor necrosis factor-alpha (TNF-α) are the cytokines commonly associated with endothelial dysfunction, vascular inflammation, and atherosclerosis. These molecules can be targeted by commonly used therapeutic substances or selective molecules that exert targeted anti-inflammatory actions. The most significant anti-inflammatory therapies are aspirin, statins, colchicine, IL-1β inhibitors, and IL-6 inhibitors, along with novel therapies such as TNF-α inhibitors and IL-1 receptor antagonists. Aspirin and statins are well-established therapies for atherosclerosis and CAD and their pleiotropic and anti-inflammatory actions contribute to their efficacy and favorable profile. Colchicine may also be considered in high-risk patients if recurrent ACS episodes occur when on optimal medical therapy according to the most recent guidelines. Recent randomized studies have also shown that therapies specifically targeting inflammatory interleukins and inflammation can reduce the risk for cardiovascular events, but these therapies are yet to be fully implemented in clinical practice. Preclinical research is also intense, targeting various inflammatory mediators that are believed to be implicated in CAD, namely repeated transfers of the soluble mutant of IFN-γ receptors, NLRP3 inflammasome inhibitors, IL-10 delivery by nanocarriers, chemokine modulatory treatments, and reacting oxygen species (ROS) targeting nanoparticles. Such approaches, although intriguing and promising, ought to be tested in clinical settings before safe conclusions can be drawn. Although the link between inflammation and atherosclerosis is significant, further studies are needed in order to elucidate this association and improve outcomes in patients with CAD.
Collapse
Affiliation(s)
- Yannis Dimitroglou
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| | - Constantina Aggeli
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| | - Panagiotis Theofilis
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| | - Panagiotis Tsioufis
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| | - Evangelos Oikonomou
- Third Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, University of Athens Medical School, 11527 Athens, Greece;
| | - Christos Chasikidis
- Department of Cardiology, General Hospital of Corinth, 20100 Corinth, Greece;
| | - Konstantinos Tsioufis
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| | - Dimitris Tousoulis
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| |
Collapse
|
12
|
Zamora R, Forsberg JA, Shah AM, Unselt D, Grey S, Lisboa FA, Billiar TR, Schobel SA, Potter BK, Elster EA, Vodovotz Y. Central role for neurally dysregulated IL-17A in dynamic networks of systemic and local inflammation in combat casualties. Sci Rep 2023; 13:6618. [PMID: 37095162 PMCID: PMC10126120 DOI: 10.1038/s41598-023-33623-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 04/15/2023] [Indexed: 04/26/2023] Open
Abstract
Dynamic Network Analysis (DyNA) and Dynamic Hypergraphs (DyHyp) were used to define protein-level inflammatory networks at the local (wound effluent) and systemic circulation (serum) levels from 140 active-duty, injured service members (59 with TBI and 81 non-TBI). Interleukin (IL)-17A was the only biomarker elevated significantly in both serum and effluent in TBI vs. non-TBI casualties, and the mediator with the most DyNA connections in TBI wounds. DyNA combining serum and effluent data to define cross-compartment correlations suggested that IL-17A bridges local and systemic circulation at late time points. DyHyp suggested that systemic IL-17A upregulation in TBI patients was associated with tumor necrosis factor-α, while IL-17A downregulation in non-TBI patients was associated with interferon-γ. Correlation analysis suggested differential upregulation of pathogenic Th17 cells, non-pathogenic Th17 cells, and memory/effector T cells. This was associated with reduced procalcitonin in both effluent and serum of TBI patients, in support of an antibacterial effect of Th17 cells in TBI patients. Dysregulation of Th17 responses following TBI may drive cross-compartment inflammation following combat injury, counteracting wound infection at the cost of elevated systemic inflammation.
Collapse
Affiliation(s)
- Ruben Zamora
- Department of Surgery, University of Pittsburgh, W944 Starzl Biomedical Sciences Tower, 200 Lothrop St., Pittsburgh, PA, 15213, USA
- Center for Inflammation and Regeneration Modeling, McGowan Institute for Regenerative Medicine, Pittsburgh, PA, 15219, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Jonathan A Forsberg
- Department of Surgery, Uniformed Services University of Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, 20814, USA
| | - Ashti M Shah
- Department of Surgery, University of Pittsburgh, W944 Starzl Biomedical Sciences Tower, 200 Lothrop St., Pittsburgh, PA, 15213, USA
| | - Desiree Unselt
- Department of Surgery, Uniformed Services University of Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, 20814, USA
- Surgical Critical Care Initiative (SC2i), Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA
- The Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Scott Grey
- Department of Surgery, Uniformed Services University of Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, 20814, USA
- Surgical Critical Care Initiative (SC2i), Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA
- The Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Felipe A Lisboa
- Department of Surgery, Uniformed Services University of Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, 20814, USA
- Surgical Critical Care Initiative (SC2i), Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA
- The Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, W944 Starzl Biomedical Sciences Tower, 200 Lothrop St., Pittsburgh, PA, 15213, USA
- Center for Inflammation and Regeneration Modeling, McGowan Institute for Regenerative Medicine, Pittsburgh, PA, 15219, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Seth A Schobel
- Department of Surgery, Uniformed Services University of Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, 20814, USA
- Surgical Critical Care Initiative (SC2i), Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA
- The Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Benjamin K Potter
- Department of Surgery, Uniformed Services University of Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, 20814, USA
- Surgical Critical Care Initiative (SC2i), Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA
| | - Eric A Elster
- Department of Surgery, Uniformed Services University of Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, 20814, USA
- Surgical Critical Care Initiative (SC2i), Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA
| | - Yoram Vodovotz
- Department of Surgery, University of Pittsburgh, W944 Starzl Biomedical Sciences Tower, 200 Lothrop St., Pittsburgh, PA, 15213, USA.
- Center for Inflammation and Regeneration Modeling, McGowan Institute for Regenerative Medicine, Pittsburgh, PA, 15219, USA.
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
13
|
Qudus MS, Tian M, Sirajuddin S, Liu S, Afaq U, Wali M, Liu J, Pan P, Luo Z, Zhang Q, Yang G, Wan P, Li Y, Wu J. The roles of critical pro-inflammatory cytokines in the drive of cytokine storm during SARS-CoV-2 infection. J Med Virol 2023; 95:e28751. [PMID: 37185833 DOI: 10.1002/jmv.28751] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/17/2023] [Accepted: 04/07/2023] [Indexed: 05/17/2023]
Abstract
In patients with severe COVID-19, acute respiratory distress syndrome (ARDS), multiple organ dysfunction syndrome (MODS), and even mortality can result from cytokine storm, which is a hyperinflammatory medical condition caused by the excessive and uncontrolled release of pro-inflammatory cytokines. High levels of numerous crucial pro-inflammatory cytokines, such as interleukin-1 (IL-1), IL-2, IL-6, tumor necrosis factor-α, interferon (IFN)-γ, IFN-induced protein 10 kDa, granulocyte-macrophage colony-stimulating factor, monocyte chemoattractant protein-1, and IL-10 and so on, have been found in severe COVID-19. They participate in cascade amplification pathways of pro-inflammatory responses through complex inflammatory networks. Here, we review the involvements of these critical inflammatory cytokines in SARS-CoV-2 infection and discuss their potential roles in triggering or regulating cytokine storm, which can help to understand the pathogenesis of severe COVID-19. So far, there is rarely effective therapeutic strategy for patients with cytokine storm besides using glucocorticoids, which is proved to result in fatal side effects. Clarifying the roles of key involved cytokines in the complex inflammatory network of cytokine storm will help to develop an ideal therapeutic intervention, such as neutralizing antibody of certain cytokine or inhibitor of some inflammatory signal pathways.
Collapse
Affiliation(s)
- Muhammad Suhaib Qudus
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Mingfu Tian
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Summan Sirajuddin
- Department of Health and Biological Sciences, Abasyn University, Peshawar, Pakistan
| | - Siyu Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Uzair Afaq
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Muneeba Wali
- Department of Allied Health Sciences, CECOS University of IT and Emerging Sciences, Peshawar, Pakistan
| | - Jinbiao Liu
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Pan Pan
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Zhen Luo
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Qiwei Zhang
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Ge Yang
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Pin Wan
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Yongkui Li
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Jianguo Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- Foshan Institute of Medical Microbiology, Foshan, China
| |
Collapse
|
14
|
Gholami S, Korosec CS, Farhang-Sardroodi S, Dick DW, Craig M, Ghaemi MS, Ooi HK, Heffernan JM. A mathematical model of protein subunits COVID-19 vaccines. Math Biosci 2023; 358:108970. [PMID: 36773843 PMCID: PMC9911981 DOI: 10.1016/j.mbs.2023.108970] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 01/21/2023] [Accepted: 01/22/2023] [Indexed: 02/12/2023]
Abstract
We consider a general mathematical model for protein subunit vaccine with a focus on the MF59-adjuvanted spike glycoprotein-clamp vaccine for SARS-CoV-2, and use the model to study immunological outcomes in the humoral and cell-mediated arms of the immune response from vaccination. The mathematical model is fit to vaccine clinical trial data. We elucidate the role of Interferon-γ and Interleukin-4 in stimulating the immune response of the host. Model results, and results from a sensitivity analysis, show that a balance between the TH1 and TH2 arms of the immune response is struck, with the TH1 response being dominant. The model predicts that two-doses of the vaccine at 28 days apart will result in approximately 85% humoral immunity loss relative to peak immunity approximately 6 months post dose 1.
Collapse
Affiliation(s)
- Samaneh Gholami
- Modelling Infection & Immunity Lab, Centre for Disease Modelling, Mathematics & Statistics, York University, Toronto, Ontario, Canada.
| | - Chapin S Korosec
- Modelling Infection & Immunity Lab, Centre for Disease Modelling, Mathematics & Statistics, York University, Toronto, Ontario, Canada
| | - Suzan Farhang-Sardroodi
- Department of Mathematics, University of Manitoba, Winnipeg, Manitoba, Canada; Modelling Infection & Immunity Lab, Centre for Disease Modelling, Mathematics & Statistics, York University, Toronto, Ontario, Canada
| | - David W Dick
- Modelling Infection & Immunity Lab, Centre for Disease Modelling, Mathematics & Statistics, York University, Toronto, Ontario, Canada
| | - Morgan Craig
- Sainte-Justine University Hospital Research Centre and Department of Mathematics and Statistics, Université de Montréal, Montreal, Quebec, Canada
| | - Mohammad Sajjad Ghaemi
- Digital Technologies Research Centre, National Research Council Canada, Toronto, ON, Canada
| | - Hsu Kiang Ooi
- Digital Technologies Research Centre, National Research Council Canada, Toronto, ON, Canada
| | - Jane M Heffernan
- Modelling Infection & Immunity Lab, Centre for Disease Modelling, Mathematics & Statistics, York University, Toronto, Ontario, Canada
| |
Collapse
|
15
|
Lee YJ, Yassa C, Park SH, Song SW, Jung WH, Lee YW, Kang H, Kim JE. Interactions between Malassezia and New Therapeutic Agents in Atopic Dermatitis Affecting Skin Barrier and Inflammation in Recombinant Human Epidermis Model. Int J Mol Sci 2023; 24:ijms24076171. [PMID: 37047166 PMCID: PMC10094540 DOI: 10.3390/ijms24076171] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Several studies have reported the pathogenic role of Malassezia in atopic dermatitis (AD); the significance of Malassezia’s influence on AD needs to be further investigated. Dupilumab, a monoclonal antibody to anti-Interleukin (IL) 4Rα, and ruxolitinib, a Janus kinase (JAK)1/2 inhibitor, are the first approved biologics and inhibitors widely used for AD treatment. In this study, we aimed to investigate how Malassezia Restricta (M. restricta) affects the skin barrier and inflammation in AD and interacts with the AD therapeutic agents ruxolitinib and anti-IL4Rα. To induce an in vitro AD model, a reconstructed human epidermis (RHE) was treated with IL-4 and IL-13. M. restricta was inoculated on the surface of RHE, and anti-IL4Rα or ruxolitinib was supplemented to model treated AD lesions. Histological and molecular analyses were performed. Skin barrier and ceramide-related molecules were downregulated by M. restricta and reverted by anti-IL4Rα and ruxolitinib. Antimicrobial peptides, VEGF, Th2-related, and JAK/STAT pathway molecules were upregulated by M. restricta and suppressed by anti-IL4Rα and ruxolitinib. These findings show that M. restricta aggravated skin barrier function and Th2 inflammation and decreased the efficacy of anti-IL4Rα and ruxolitinib.
Collapse
|
16
|
Habanjar O, Bingula R, Decombat C, Diab-Assaf M, Caldefie-Chezet F, Delort L. Crosstalk of Inflammatory Cytokines within the Breast Tumor Microenvironment. Int J Mol Sci 2023; 24:4002. [PMID: 36835413 PMCID: PMC9964711 DOI: 10.3390/ijms24044002] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Several immune and immunocompetent cells, including dendritic cells, macrophages, adipocytes, natural killer cells, T cells, and B cells, are significantly correlated with the complex discipline of oncology. Cytotoxic innate and adaptive immune cells can block tumor proliferation, and others can prevent the immune system from rejecting malignant cells and provide a favorable environment for tumor progression. These cells communicate with the microenvironment through cytokines, a chemical messenger, in an endocrine, paracrine, or autocrine manner. These cytokines play an important role in health and disease, particularly in host immune responses to infection and inflammation. They include chemokines, interleukins (ILs), adipokines, interferons, colony-stimulating factors (CSFs), and tumor necrosis factor (TNF), which are produced by a wide range of cells, including immune cells, such as macrophages, B-cells, T-cells, and mast cells, as well as endothelial cells, fibroblasts, a variety of stromal cells, and some cancer cells. Cytokines play a crucial role in cancer and cancer-related inflammation, with direct and indirect effects on tumor antagonistic or tumor promoting functions. They have been extensively researched as immunostimulatory mediators to promote the generation, migration and recruitment of immune cells that contribute to an effective antitumor immune response or pro-tumor microenvironment. Thus, in many cancers such as breast cancer, cytokines including leptin, IL-1B, IL-6, IL-8, IL-23, IL-17, and IL-10 stimulate while others including IL-2, IL-12, and IFN-γ, inhibit cancer proliferation and/or invasion and enhance the body's anti-tumor defense. Indeed, the multifactorial functions of cytokines in tumorigenesis will advance our understanding of cytokine crosstalk pathways in the tumor microenvironment, such as JAK/STAT, PI3K, AKT, Rac, MAPK, NF-κB, JunB, cFos, and mTOR, which are involved in angiogenesis, cancer proliferation and metastasis. Accordingly, targeting and blocking tumor-promoting cytokines or activating and amplifying tumor-inhibiting cytokines are considered cancer-directed therapies. Here, we focus on the role of the inflammatory cytokine system in pro- and anti-tumor immune responses, discuss cytokine pathways involved in immune responses to cancer and some anti-cancer therapeutic applications.
Collapse
Affiliation(s)
- Ola Habanjar
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Rea Bingula
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Caroline Decombat
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Mona Diab-Assaf
- Equipe Tumorigénèse Pharmacologie Moléculaire et Anticancéreuse, Faculté des Sciences II, Université Libanaise Fanar, Beyrouth 1500, Lebanon
| | - Florence Caldefie-Chezet
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Laetitia Delort
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| |
Collapse
|
17
|
Karbalaeimahdi M, Farajnia S, Bargahi N, Ghadiri-Moghaddam F, Rasouli Jazi HR, Bakhtiari N, Ghasemali S, Zarghami N. The Role of Interferons in Long Covid Infection. J Interferon Cytokine Res 2023; 43:65-76. [PMID: 36795973 DOI: 10.1089/jir.2022.0193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Abstract
Although the new generation of vaccines and anti-COVID-19 treatment regimens facilitated the management of acute COVID-19 infections, concerns about post-COVID-19 syndrome or Long Covid are rising. This issue can increase the incidence and morbidity of diseases such as diabetes, and cardiovascular, and lung infections, especially among patients suffering from neurodegenerative disease, cardiac arrhythmias, and ischemia. There are numerous risk factors that cause COVID-19 patients to experience post-COVID-19 syndrome. Three potential causes attributed to this disorder include immune dysregulation, viral persistence, and autoimmunity. Interferons (IFNs) are crucial in all aspects of post-COVID-19 syndrome etiology. In this review, we discuss the critical and double-edged role of IFNs in post-COVID-19 syndrome and how innovative biomedical approaches that target IFNs can reduce the occurrence of Long Covid infection.
Collapse
Affiliation(s)
- Mohammad Karbalaeimahdi
- Department of Medical Biotechnology, School of Advanced Medical Sciences, Tabriz, Iran.,Biotechnology Research Center, Tabriz, Iran
| | - Safar Farajnia
- Biotechnology Research Center, Tabriz, Iran.,Drug Applied Research Center, Tabriz, Iran
| | | | - Farzaneh Ghadiri-Moghaddam
- Drug Applied Research Center, Tabriz, Iran.,Department of Biology, Faculty of Science, Azarbaijan Shahid Madani University, Tabriz, Iran
| | | | | | | | - Nosratollah Zarghami
- Department of Medical Biochemistry, Faculty of Medicine, Istanbul Aydin University, Istanbul, Turkey.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
18
|
Ramirez-Gonzalez JB, Morales-BuenRostro LE, Garcia-Covarrubias L, Pacheco-Domínguez RL, Durazo-Arvizu R, Cuevas-Medina EN, Furuzawa-Carballeda J, Osorio-Juárez RA, Correa-Rotter R, Rincón-Pedrero R, Alberú-Gómez J, López Cervantes M. Assessment of the Relationship Between Inflammation and Glomerular Filtration Rate. Can J Kidney Health Dis 2023; 10:20543581221132748. [PMID: 36700057 PMCID: PMC9869199 DOI: 10.1177/20543581221132748] [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: 05/02/2022] [Accepted: 08/22/2022] [Indexed: 01/20/2023] Open
Abstract
Background Chronic kidney disease (CKD) is a global health problem. As it progresses to end stages, renal replacement therapy is required but ultimately, the best treatment is transplantation. Decreased renal function has been associated with an inflammatory state associated to primary CKD and in kidney transplant recipients (KTRs). Objective To establish how the serum concentrations of some cytokines, such as interleukin (IL)-2, IL-8, IL-22, IL-17α, interferon-gamma, IL-4, and transforming growth factor-β, correlate with various CKD stages. Methods One hundred and forty-one KTRs between the ages of 18 and 75 years were included in the study. We also included 112 live kidney donors, 37 CKD PGCKD+3, and 76 GPhealthy. Participants were grouped according to their glomerular filtration rate (GFR) and their circulating cytokine levels, previously quantified by ELISA. Results By linear regression analysis, we established the relation of each cytokine with the GFR. Transforming growth factor-β correlated positively with the GFR in the study population, except in healthy individuals. A negative correlation of IL-8 and IL-17α and GFR was found in all cases. Conclusions Whether these cytokines (IL-8 and IL-17α) could be used as inflammatory biomarkers indicating CKD progression, regardless of the type of population, remains to be prospectively determined.
Collapse
Affiliation(s)
- J. B. Ramirez-Gonzalez
- Faculty of Medicine, Department of Public Health, Universidad Nacional Autónoma de México, Ciudad de Mexico, México
| | - L. E. Morales-BuenRostro
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de Mexico, México
| | - L. Garcia-Covarrubias
- Department of Transplantation, Hospital General de México Dr. Eduardo Liceaga, Ciudad de México, México
| | - R. L. Pacheco-Domínguez
- Faculty of Medicine, Department of Public Health, Universidad Nacional Autónoma de México, Ciudad de Mexico, México
| | - R. Durazo-Arvizu
- Department of Public Health Sciences, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - E. N. Cuevas-Medina
- Department of Cardiology, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | - J. Furuzawa-Carballeda
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de Mexico, México
| | - R. A. Osorio-Juárez
- Faculty of Medicine, Department of Public Health, Universidad Nacional Autónoma de México, Ciudad de Mexico, México
| | - R. Correa-Rotter
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de Mexico, México
| | - R. Rincón-Pedrero
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de Mexico, México
| | - Josefina Alberú-Gómez
- Department of Transplantation, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de Mexico, México,Josefina Alberú-Gómez, Department of Transplantation, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga # 15, Colonia Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México CP 14080, México.
| | - Malaquías López Cervantes
- Faculty of Medicine, Department of Public Health, Universidad Nacional Autónoma de México, Ciudad de Mexico, México,Malaquías López Cervantes, Faculty of Medicine, Department of Public Health, Universidad Nacional Autónoma de México, Av. Universidad Nacional Autónoma de México #3000, Colonica Universidad Nacional Autonoma de México, CU, Delegación Coyoacán, Ciudad de México, CP 04510, México. ;
| |
Collapse
|
19
|
Tsioufis P, Theofilis P, Tsioufis K, Tousoulis D. The Impact of Cytokines in Coronary Atherosclerotic Plaque: Current Therapeutic Approaches. Int J Mol Sci 2022; 23:ijms232415937. [PMID: 36555579 PMCID: PMC9788180 DOI: 10.3390/ijms232415937] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Coronary atherosclerosis is a chronic pathological process that involves inflammation together with endothelial dysfunction and lipoprotein dysregulation. Experimental studies during the past decades have established the role of inflammatory cytokines in coronary artery disease, namely interleukins (ILs), tumor necrosis factor (TNF)-α, interferon-γ, and chemokines. Moreover, their value as biomarkers in disease development and progression further enhance the validity of this interaction. Recently, cytokine-targeted treatment approaches have emerged as potential tools in the management of atherosclerotic disease. IL-1β, based on the results of the CANTOS trial, remains the most validated option in reducing the residual cardiovascular risk. Along the same line, colchicine was also proven efficacious in preventing major adverse cardiovascular events in large clinical trials of patients with acute and chronic coronary syndrome. Other commercially available agents targeting IL-6 (tocilizumab), TNF-α (etanercept, adalimumab, infliximab), or IL-1 receptor antagonist (anakinra) have mostly been assessed in the setting of other inflammatory diseases and further testing in atherosclerosis is required. In the future, potential targeting of the NLRP3 inflammasome, anti-inflammatory IL-10, or atherogenic chemokines could represent appealing options, provided that patient safety is proven to be of no concern.
Collapse
|
20
|
Korosec CS, Farhang-Sardroodi S, Dick DW, Gholami S, Ghaemi MS, Moyles IR, Craig M, Ooi HK, Heffernan JM. Long-term durability of immune responses to the BNT162b2 and mRNA-1273 vaccines based on dosage, age and sex. Sci Rep 2022; 12:21232. [PMID: 36481777 PMCID: PMC9732004 DOI: 10.1038/s41598-022-25134-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022] Open
Abstract
The lipid nanoparticle (LNP)-formulated mRNA vaccines BNT162b2 and mRNA-1273 are a widely adopted multi vaccination public health strategy to manage the COVID-19 pandemic. Clinical trial data has described the immunogenicity of the vaccine, albeit within a limited study time frame. Here, we use a within-host mathematical model for LNP-formulated mRNA vaccines, informed by available clinical trial data from 2020 to September 2021, to project a longer term understanding of immunity as a function of vaccine type, dosage amount, age, and sex. We estimate that two standard doses of either mRNA-1273 or BNT162b2, with dosage times separated by the company-mandated intervals, results in individuals losing more than 99% humoral immunity relative to peak immunity by 8 months following the second dose. We predict that within an 8 month period following dose two (corresponding to the original CDC time-frame for administration of a third dose), there exists a period of time longer than 1 month where an individual has lost more than 99% humoral immunity relative to peak immunity, regardless of which vaccine was administered. We further find that age has a strong influence in maintaining humoral immunity; by 8 months following dose two we predict that individuals aged 18-55 have a four-fold humoral advantage compared to aged 56-70 and 70+ individuals. We find that sex has little effect on the immune response and long-term IgG counts. Finally, we find that humoral immunity generated from two low doses of mRNA-1273 decays at a substantially slower rate relative to peak immunity gained compared to two standard doses of either mRNA-1273 or BNT162b2. Our predictions highlight the importance of the recommended third booster dose in order to maintain elevated levels of antibodies.
Collapse
Affiliation(s)
- Chapin S Korosec
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
| | - Suzan Farhang-Sardroodi
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
- Department of Mathematics, University of Manitoba, 186 Dysart Road, Winnipeg, MB, R3T 2N2, Canada
| | - David W Dick
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
| | - Sameneh Gholami
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
| | - Mohammad Sajjad Ghaemi
- Digital Technologies Research Centre, National Research Council Canada, 222 College Street, Toronto, ON, M5T 3J1, Canada
| | - Iain R Moyles
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
| | - Morgan Craig
- Department of Mathematics and Statistics, Université de Montréal & Sainte-Justine University Hospital Research Centre, 3175, ch. Côte Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Hsu Kiang Ooi
- Digital Technologies Research Centre, National Research Council Canada, 222 College Street, Toronto, ON, M5T 3J1, Canada
| | - Jane M Heffernan
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
| |
Collapse
|
21
|
Gauthier T, Chen W. IFN-γ and TGF-β, Crucial Players in Immune Responses: A Tribute to Howard Young. J Interferon Cytokine Res 2022; 42:643-654. [PMID: 36516375 PMCID: PMC9917322 DOI: 10.1089/jir.2022.0132] [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: 06/07/2022] [Accepted: 06/18/2022] [Indexed: 12/15/2022] Open
Abstract
Interferon gamma (IFN-γ) and transforming growth factor beta (TGF-β), both pleiotropic cytokines, have been long studied and described as critical mediators of the immune response, notably in T cells. One of the investigators who made seminal and critical discoveries in the field of IFN-γ biology is Dr. Howard Young. In this review, we provide an overview of the biology of IFN-γ as well as its role in cancer and autoimmunity with an emphasis on Dr. Young's critical work in the field. We also describe how Dr. Young's work influenced our own research studying the role of TGF-β in the modulation of immune responses.
Collapse
Affiliation(s)
- Thierry Gauthier
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health, Bethesda, Maryland, USA
| | - WanJun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
22
|
Influence of SARS-COV-2 Infection on Cytokine Production by Mitogen-Stimulated Peripheral Blood Mononuclear Cells and Neutrophils in COVID-19 Intensive Care Unit Patients. Microorganisms 2022; 10:microorganisms10112194. [PMID: 36363785 PMCID: PMC9695671 DOI: 10.3390/microorganisms10112194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/30/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
We sought to investigate the influence of SARS-CoV-2 infection on the cytokine profiles of peripheral blood mononuclear cells (PBMCs) and neutrophils from coronavirus disease 2019 (COVID-19) intensive care unit (ICU) patients. Neutrophils and PBMCs were separated and stimulated with the mitogen phytohemagglutinin. Culture supernatants of mitogen-stimulated PBMCs and neutrophils from 88 COVID-19 ICU patients and 88 healthy controls were evaluated for levels of granulocyte-macrophage colony-stimulating factor (GM-CSF), interferon (IFN)-α, IFN-γ, interleukin (IL)-2, -4, -5, -6, -9, -10, -12, -17A, and tumor necrosis factor (TNF)-α using anti-cytokine antibody MACSPlex capture beads. Cytokine profiles of PBMCs showed significantly lower levels of GM-CSF, IFN-γ, IL-6, IL-9, IL-10, IL-17A, and TNF-α (p < 0.0001) in COVID-19 ICU patients. In contrast, COVID-19 ICU patients showed higher median levels of IL-2 (p < 0.001) and IL-5 (p < 0.01) by PBMCs. As for neutrophils, COVID-19 ICU patients showed significantly lower levels of GM-CSF, IFN-γ, IL-2, IL-4, IL-5, IL-6, IL-9, IL-10, IL-17A, IL-12, TNF-α (p < 0.0001), and IFN-α (p < 0.01). T-helper (Th)1:Th2 cytokine ratios revealed lower inflammatory cytokine for PBMCs and neutrophils in COVID-19 ICU patients. Cytokine production profiles and Th1:Th2 cytokine ratios suggest that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has an immunomodulatory effect on PBMCs and neutrophils. This study also suggests that the increased levels of several cytokines in the serum are not sourced from PBMCs and neutrophils.
Collapse
|
23
|
Mohr E, Hinnenthal T, Gryzik S, Hoang Y, Lischke T, Retzlaff J, Mekonnen A, Paul F, Valleriani A, Radbruch A, Vera J, Baumgrass R. Bin-based visualization of cytokine-co-expression patterns of IL-10-producing CD4 T cell subsets. Eur J Immunol 2022; 52:1684-1687. [PMID: 36067024 DOI: 10.1002/eji.202249829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 12/14/2022]
Affiliation(s)
- Elodie Mohr
- German Rheumatism Research Center, A Leibniz Institute, Berlin, Germany
| | - Timo Hinnenthal
- German Rheumatism Research Center, A Leibniz Institute, Berlin, Germany
| | - Stefanie Gryzik
- German Rheumatism Research Center, A Leibniz Institute, Berlin, Germany
| | - Yen Hoang
- German Rheumatism Research Center, A Leibniz Institute, Berlin, Germany
| | - Timo Lischke
- German Rheumatism Research Center, A Leibniz Institute, Berlin, Germany
| | - Jimmy Retzlaff
- Systems Tumour Immunology, Friedrich-Alexander-University of Erlangen-Nürnberg, Universitätsklinikum, Erlangen, Germany
| | - Ariana Mekonnen
- German Rheumatism Research Center, A Leibniz Institute, Berlin, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité, Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Angelo Valleriani
- Max Planck Institute of Colloids and Interfaces, Biomaterials Department, Potsdam, Germany
| | - Andreas Radbruch
- German Rheumatism Research Center, A Leibniz Institute, Berlin, Germany.,Charité, Campus Berlin Mitte, Berlin, Germany
| | - Julio Vera
- Systems Tumour Immunology, Friedrich-Alexander-University of Erlangen-Nürnberg, Universitätsklinikum, Erlangen, Germany
| | - Ria Baumgrass
- German Rheumatism Research Center, A Leibniz Institute, Berlin, Germany.,University of Potsdam, Potsdam, Germany
| |
Collapse
|
24
|
Miladinova E, Lilkova E, Krachmarova E, Malinova K, Petkov P, Ilieva N, Nacheva G, Litov L. Heparan Sulfate Facilitates Binding of hIFN γ to Its Cell-Surface Receptor hIFNGR1. Int J Mol Sci 2022; 23:ijms23169415. [PMID: 36012678 PMCID: PMC9408938 DOI: 10.3390/ijms23169415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/12/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
Human interferon-gamma (hIFNγ) is a crucial signaling molecule with an important role in the initialization and development of the immune response of the host. However, its aberrant activity is also associated with the progression of a multitude of autoimmune and other diseases, which determines the need for effective inhibitors of its activity. The development of such treatments requires proper understanding of the interaction of hIFNγ to its cell-surface receptor hIFNGR1. Currently, there is no comprehensive model of the mechanism of this binding process. Here, we employ molecular dynamics simulations to study on a microscopic level the process of hIFNγ–hIFNGR1 complex formation in different scenarios. We find that the two molecules alone fail to form a stable complex, but the presence of heparan-sulfate-like oligosaccharides largely facilitates the process by both demobilizing the highly flexible C-termini of the cytokine and assisting in the proper positioning of its globule between the receptor subunits. An antiproliferative-activity assay on cells depleted from cell-surface heparan sulfate (HS) sulfation together with the phosphorylation levels of the signal transducer and activator of transcription STAT1 confirms qualitatively the simulation-based multistage complex-formation model. Our results reveal the key role of HS and its proteoglycans in all processes involving hIFNγ signalling.
Collapse
Affiliation(s)
- Elisaveta Miladinova
- Faculty of Physics, Sofia University “St. Kliment Ohridski”, 5 James Bourchier Blvd., 1164 Sofia, Bulgaria
| | - Elena Lilkova
- Institute of Information and Communication Technologies, Bulgarian Academy of Sciences, 2 Acad. G. Bonchev Str., 1113 Sofia, Bulgaria
- Correspondence: (E.L.); (E.K.)
| | - Elena Krachmarova
- Institute of Molecular Biology “Roumen Tsanev”, Bulgarian Academy of Sciences, 21 Acad. G. Bonchev Str., 1113 Sofia, Bulgaria
- Correspondence: (E.L.); (E.K.)
| | - Kristina Malinova
- Institute of Molecular Biology “Roumen Tsanev”, Bulgarian Academy of Sciences, 21 Acad. G. Bonchev Str., 1113 Sofia, Bulgaria
| | - Peicho Petkov
- Faculty of Physics, Sofia University “St. Kliment Ohridski”, 5 James Bourchier Blvd., 1164 Sofia, Bulgaria
| | - Nevena Ilieva
- Institute of Information and Communication Technologies, Bulgarian Academy of Sciences, 2 Acad. G. Bonchev Str., 1113 Sofia, Bulgaria
| | - Genoveva Nacheva
- Institute of Molecular Biology “Roumen Tsanev”, Bulgarian Academy of Sciences, 21 Acad. G. Bonchev Str., 1113 Sofia, Bulgaria
| | - Leandar Litov
- Faculty of Physics, Sofia University “St. Kliment Ohridski”, 5 James Bourchier Blvd., 1164 Sofia, Bulgaria
| |
Collapse
|
25
|
Coutant F, Bachet R, Pin JJ, Alonzo M, Miossec P. Monoclonal antibodies from B cells of patients with anti-MDA5 antibody-positive dermatomyositis directly stimulate interferon gamma production. J Autoimmun 2022; 130:102831. [DOI: 10.1016/j.jaut.2022.102831] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/09/2022] [Accepted: 04/09/2022] [Indexed: 01/10/2023]
|
26
|
Shen H, He J, Ling X, Liu C, Wang Y, Zhang X, He X, Yang H, Chen M, Shi Q. Wheel-Running Exercise Protects Ovariectomized Mice from Bone Loss via IFN- γ-Mediated Suppression of the NF- κB and MAPK Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2030818. [PMID: 35602099 PMCID: PMC9117016 DOI: 10.1155/2022/2030818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 04/08/2022] [Indexed: 11/25/2022]
Abstract
Physical exercise is recommended as a preventative approach for osteoporosis; however, the effect of physical exercise on bone mass remains controversial. Additionally, the immune regulation of physical exercise on bone mass remains unclear. To determine whether wheel-running (WR) exercise contributes to improving bone mineral density (BMD) and investigate the involved immune mechanism, ovariectomized (OVX) and sham-operated mice were treated with 8 weeks of WR exercise. The distal femurs of the mice were sequentially scanned, reconstructed, and analyzed using microcomputed tomography and related software to assess BMD and bone microarchitecture. Flow cytometry assays were applied to investigate alterations in immune cells and inflammatory cytokines. In vitro, osteoclast differentiation was conducted to determine the effect of IFN-γ on osteoclastogenesis and the underlying mechanism. As a result, trabecular parameters were decreased in the OVX mice compared with the sham group. However, WR exercise significantly improved the deterioration in the bone microarchitecture of the OVX mice with an increase of 60.00% in BMD, 55.18% in bone volume, 66.67% in trabecular number, 32.52% in trabecular thickness, and a decrease of 19.44% in trabecular separation. Similarly, WR exercise increased the proportion of CD8+ T cells from 7.26 ± 1.71% to 10.23 ± 1.35% in the spleen and from 1.62 ± 0.54% to 2.38 ± 0.43% in the bone marrow of the OVX mice (P < 0.05). The expression of IFN-γ was also increased in the OVX + WR mice compared with the OVX mice (1.65 ± 0.45% vs. 2.26 ± 0.34%, P < 0.05). In vitro studies demonstrated an inhibitory effect of IFN-γ on osteoclastogenesis in a dose- and time-dependent manner. Meanwhile, the classical NF-κB and MAPK pathways were found to be critical in IFN-γ-mediated inhibition of osteoclast differentiation. In conclusion, our study discovered that WR exercise rescued bone loss in the OVX mice in an IFN-γ-mediated immunomodulatory manner. After WR exercise, IFN-γ expression was restored by activated CD8+ T cells, consequently leading to the inhibition of osteoclastogenesis and the recovery from bone loss through the NF-κB and MAPK pathways.
Collapse
Affiliation(s)
- Hao Shen
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Orthopedics Institute of Soochow University, Medical College of Soochow University, No. 899, Pinghai Road, Suzhou, Jiangsu 215006, China
| | - Jiaheng He
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Orthopedics Institute of Soochow University, Medical College of Soochow University, No. 899, Pinghai Road, Suzhou, Jiangsu 215006, China
| | - Xuwei Ling
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Orthopedics Institute of Soochow University, Medical College of Soochow University, No. 899, Pinghai Road, Suzhou, Jiangsu 215006, China
| | - Chang Liu
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Orthopedics Institute of Soochow University, Medical College of Soochow University, No. 899, Pinghai Road, Suzhou, Jiangsu 215006, China
| | - Yi Wang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Orthopedics Institute of Soochow University, Medical College of Soochow University, No. 899, Pinghai Road, Suzhou, Jiangsu 215006, China
| | - Xiongjinfu Zhang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Orthopedics Institute of Soochow University, Medical College of Soochow University, No. 899, Pinghai Road, Suzhou, Jiangsu 215006, China
| | - Xu He
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Orthopedics Institute of Soochow University, Medical College of Soochow University, No. 899, Pinghai Road, Suzhou, Jiangsu 215006, China
| | - Huilin Yang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Orthopedics Institute of Soochow University, Medical College of Soochow University, No. 899, Pinghai Road, Suzhou, Jiangsu 215006, China
| | - Mimi Chen
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Orthopedics Institute of Soochow University, Medical College of Soochow University, No. 899, Pinghai Road, Suzhou, Jiangsu 215006, China
| | - Qin Shi
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Orthopedics Institute of Soochow University, Medical College of Soochow University, No. 899, Pinghai Road, Suzhou, Jiangsu 215006, China
| |
Collapse
|
27
|
Packialakshmi B, Hira S, Lund K, Zhang AH, Halterman J, Feng Y, Scott DW, Lees JR, Zhou X. NFAT5 contributes to the pathogenesis of experimental autoimmune encephalomyelitis (EAE) and decrease of T regulatory cells in female mice. Cell Immunol 2022; 375:104515. [DOI: 10.1016/j.cellimm.2022.104515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 11/03/2022]
|
28
|
Interferon-γ induces interleukin-6 production by neutrophils via the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway. BMC Res Notes 2021; 14:447. [PMID: 34895310 PMCID: PMC8666078 DOI: 10.1186/s13104-021-05860-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
Objective Interferon-gamma (IFN-γ) is overexpressed in rheumatoid synovium and thought to be involved in the pathogenesis of rheumatoid arthritis (RA). In this study, we examined our hypothesis that IFN-γ activates innate immune cells and upregulates inflammatory cytokines. Peripheral blood neutrophils were stimulated with IFN-γ in the presence or absence of Janus kinase (JAK) inhibitors. Interleukin-6 (IL-6) mRNA and protein expression were analyzed using real-time polymerase chain reaction (PCR) method and enzyme-linked immunosorbent assay. Protein phosphorylation of JAKs or STAT1 was assessed by Western blot using phospho-specific antibodies. Results IFN-γ stimulation induces IL-6 expression in protein and mRNA levels in human neutrophils. Furthermore, IFN-γ stimulation induces JAK1/JAK2 phosphorylation and downstream signal transducer and activator of transcription (STAT) 1 phosphorylation in human neutrophils. Although all JAKi, blocked IFN-γ-induced JAK1.2/STAT1 phosphorylation at higher concentrations (100 nM), baricitinib most efficiently inhibited IFN-γ-induced JAK1.2/STAT1 phosphorylation at lower concentrations (≤ 25 nM). Among these JAKi, baricitinib was the most potent regulator for IFN-γ-induced IL-6 production in human neutrophils. Our data indicate that IFN-γ upregulates IL-6 production via the JAK1/2-STAT1 pathway in human innate immune cells. Furthermore, this IFN-γ-mediated IL-6 induction via JAK/STAT was downregulated by JAKi. Supplementary Information The online version contains supplementary material available at 10.1186/s13104-021-05860-w.
Collapse
|
29
|
Association between senescence of T cells and disease activity in patients with systemic lupus erythematosus. Reumatologia 2021; 59:292-301. [PMID: 34819703 PMCID: PMC8609380 DOI: 10.5114/reum.2021.110318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 10/05/2021] [Indexed: 01/10/2023] Open
Abstract
Objectives Systemic lupus erythematosus (SLE) patients are predisposed to chronic immune activation, leading to accelerated immunosenescence. The aging of the immune system causes the T cells to express several senescence markers such as CD57 and KLRG1, which produce pro-inflammatory cytokine interferon γ (IFN-γ). Immunosenescence was associated with high morbidity and mortality in other diseases. This research was conducted to prove the association between senescent T cells and SLE disease activity. Material and methods This research was an observational cross-sectional study on 53 women aged 16–45 years diagnosed with SLE based on SLICC 2012 criteria. All subjects were recorded for demographic and clinical data, and their SLE disease activity index (SLEDAI) score was measured to evaluate disease activity. Active disease was defined as SLEDAI score ≥ 3. The CD57 antigen and KLRG1 expression on CD4+ and CD8+ T cells were calculated from peripheral blood mononuclear cells (PBMC) by flow cytometry. Interferon γ was measured from serum using ELISA. The comparison was done using the Mann-Whitney U test, and correlation was tested using the Spearman test. Associations between variables were calculated using linear regression models. Results Systemic lupus erythematosus patients with active disease had markedly higher CD4+KLRG1+ (3.1 [1.3–5.5]% vs. 0.3 [0.1–0.5]%), CD8+CD57+ (11.6 ±7.1% vs. 2.4 ±2.0%, p = 0.000), and CD8+KLRG1+ T cell percentages (13.7 ±7.5% vs. 0.3 ±0.1%, p = 0.000), and IFN- γ levels (208.9 [148.3–233.8] vs. 146.7 [130.2–210.8] pg/ml, p = 0.048), compared to the inactive patients. Positive correlation and association was found between the CD8+CD57+ and CD8+KLRG1+ percentages with the SLEDAI score (p = 0.007 and p = 0.007, for the linear regression analysis, respectively). Conclusions Systemic lupus erythematosus patients showed significantly higher senescence T cell markers compared to controls, and the increase of T cell senescence, especially in the CD8 compartment, has some association with increased disease activity in patients with SLE.
Collapse
|
30
|
Farhang-Sardroodi S, Korosec CS, Gholami S, Craig M, Moyles IR, Ghaemi MS, Ooi HK, Heffernan JM. Analysis of Host Immunological Response of Adenovirus-Based COVID-19 Vaccines. Vaccines (Basel) 2021; 9:861. [PMID: 34451985 PMCID: PMC8402548 DOI: 10.3390/vaccines9080861] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 12/18/2022] Open
Abstract
During the SARS-CoV-2 global pandemic, several vaccines, including mRNA and adenovirus vector approaches, have received emergency or full approval. However, supply chain logistics have hampered global vaccine delivery, which is impacting mass vaccination strategies. Recent studies have identified different strategies for vaccine dose administration so that supply constraints issues are diminished. These include increasing the time between consecutive doses in a two-dose vaccine regimen and reducing the dosage of the second dose. We consider both of these strategies in a mathematical modeling study of a non-replicating viral vector adenovirus vaccine in this work. We investigate the impact of different prime-boost strategies by quantifying their effects on immunological outcomes based on simple system of ordinary differential equations. The boost dose is administered either at a standard dose (SD) of 1000 or at a low dose (LD) of 500 or 250 vaccine particles. Results show dose-dependent immune response activity. Our model predictions show that by stretching the prime-boost interval to 18 or 20, in an SD/SD or SD/LD regimen, the minimum promoted antibody (Nab) response will be comparable with the neutralizing antibody level measured in COVID-19 recovered patients. Results also show that the minimum stimulated antibody in SD/SD regimen is identical with the high level observed in clinical trial data. We conclude that an SD/LD regimen may provide protective capacity, which will allow for conservation of vaccine doses.
Collapse
Affiliation(s)
- Suzan Farhang-Sardroodi
- Modelling Infection and Immunity Lab, Mathematics Statistics, York University, Toronto, ON M3J 1P3, Canada; (C.S.K.); (S.G.)
- Centre for Disease Modelling (CDM), Mathematics Statistics, York University, Toronto, ON M3J 1P3, Canada;
| | - Chapin S. Korosec
- Modelling Infection and Immunity Lab, Mathematics Statistics, York University, Toronto, ON M3J 1P3, Canada; (C.S.K.); (S.G.)
- Centre for Disease Modelling (CDM), Mathematics Statistics, York University, Toronto, ON M3J 1P3, Canada;
| | - Samaneh Gholami
- Modelling Infection and Immunity Lab, Mathematics Statistics, York University, Toronto, ON M3J 1P3, Canada; (C.S.K.); (S.G.)
- Centre for Disease Modelling (CDM), Mathematics Statistics, York University, Toronto, ON M3J 1P3, Canada;
| | - Morgan Craig
- Sainte-Justine University Hospital Research Centre and Department of Mathematics and Statistics, Université de Montréal, Montreal, QC H3T 1J4, Canada;
| | - Iain R. Moyles
- Centre for Disease Modelling (CDM), Mathematics Statistics, York University, Toronto, ON M3J 1P3, Canada;
| | - Mohammad Sajjad Ghaemi
- Digital Technologies Research Centre, National Research Council Canada, Toronto, ON C1A 4P3, Canada; (M.S.G.); (H.K.O.)
| | - Hsu Kiang Ooi
- Digital Technologies Research Centre, National Research Council Canada, Toronto, ON C1A 4P3, Canada; (M.S.G.); (H.K.O.)
| | - Jane M. Heffernan
- Modelling Infection and Immunity Lab, Mathematics Statistics, York University, Toronto, ON M3J 1P3, Canada; (C.S.K.); (S.G.)
- Centre for Disease Modelling (CDM), Mathematics Statistics, York University, Toronto, ON M3J 1P3, Canada;
| |
Collapse
|
31
|
Inflamm-Aging-Related Cytokines of IL-17 and IFN- γ Accelerate Osteoclastogenesis and Periodontal Destruction. J Immunol Res 2021; 2021:9919024. [PMID: 34395635 PMCID: PMC8357511 DOI: 10.1155/2021/9919024] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/23/2021] [Accepted: 07/14/2021] [Indexed: 11/18/2022] Open
Abstract
Periodontal disease (PD), as an age-related disease, prevalent in middle-aged and elderly population, is characterized as inflammatory periodontal tissue loss, including gingival inflammation and alveolar bone resorption. However, the definite mechanism of aging-related inflammation in PD pathology needs further investigation. Our study is aimed at exploring the effect of inflamm-aging-related cytokines of interleukin-17 (IL-17) and interferon-γ (IFN-γ) on osteoclastogenesis in vitro and periodontal destruction in vivo. For receptor activator of nuclear factor-κB ligand- (RANKL-) primed bone marrow macrophages (BMMs), IL-17 and IFN-γ enhanced osteoclastogenesis, with the expression of osteoclastogenic mRNA (TRAP, c-Fos, MMP-9, Ctsk, and NFATc1) and protein (c-Fos and MMP-9) upregulated. Ligament-induced rat models were established to investigate the role of IL-17 and IFN-γ on experimental periodontitis. Both IL-17 and IFN-γ could enhance the local inflammation in gingival tissues. Although there might be an antagonistic interaction between IL-17 and IFN-γ, IL-17 and IFN-γ could facilitate alveolar bone loss and osteoclast differentiation.
Collapse
|
32
|
Zaiatz Bittencourt V, Jones F, Tosetto M, Doherty GA, Ryan EJ. Dysregulation of Metabolic Pathways in Circulating Natural Killer Cells Isolated from Inflammatory Bowel Disease Patients. J Crohns Colitis 2021; 15:1316-1325. [PMID: 33460436 PMCID: PMC8328302 DOI: 10.1093/ecco-jcc/jjab014] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIMS Inflammatory bowel diseases [IBD], comprising Crohn's disease [CD] and ulcerative colitis [UC], are chronic conditions characterized by severe dysregulation of innate and adaptive immunity resulting in the destruction of the intestinal mucosa. Natural killer [NK] cells play a pivotal role in the dynamic interaction between the innate and adaptive immune response. There is an increasing appreciation for the key role immunometabolism plays in the regulation of NK cell function, yet little remains known about the metabolic profile, cytokine secretion, and killing capacity of human NK cells during active IBD. METHODS Peripheral blood mononuclear cells were isolated from peripheral blood of patients with moderate to severely active IBD and healthy controls. NK cells were stained with a combination of cell surface receptors, intracellular cytokines, and proteins and analyzed by flow cytometry. For measurements of NK cell cytotoxicity, the calcein-AM release assay was performed. The metabolic profile was analyzed by an extracellular flux analyzer. RESULTS NK cells from IBD patients produce large quantities of pro-inflammatory cytokines, IL-17A and TNF-α ex vivo, but have limited killing capability. Furthermore, patient NK cells have reduced mitochondrial mass and oxidative phosphorylation. mTORC1, an important cell and metabolic regulator, demonstrated limited activity in both freshly isolated cells and cytokine-stimulated cells. CONCLUSIONS Our results demonstrate that circulating NK cells of IBD patients have an unbalanced metabolic profile, with faulty mitochondria and reduced capacity to kill. These aberrations in NK cell metabolism may contribute to defective killing and thus the secondary infections and increased risk of cancer observed in IBD patients.
Collapse
Affiliation(s)
- Vanessa Zaiatz Bittencourt
- Centre for Colorectal Disease, St Vincent’s University Hospital, School of Medicine, University College Dublin, Dublin, Ireland
| | - Fiona Jones
- Centre for Colorectal Disease, St Vincent’s University Hospital, School of Medicine, University College Dublin, Dublin, Ireland
| | - Miriam Tosetto
- Centre for Colorectal Disease, St Vincent’s University Hospital, School of Medicine, University College Dublin, Dublin, Ireland
| | - Glen A Doherty
- Centre for Colorectal Disease, St Vincent’s University Hospital, School of Medicine, University College Dublin, Dublin, Ireland
| | - Elizabeth J Ryan
- Centre for Colorectal Disease, St Vincent’s University Hospital, School of Medicine, University College Dublin, Dublin, Ireland
- Department of Biological Sciences, Health Research Institute, University of Limerick, Limerick, Ireland
| |
Collapse
|
33
|
Luostarinen S, Hämäläinen M, Hatano N, Muraki K, Moilanen E. The inflammatory regulation of TRPA1 expression in human A549 lung epithelial cells. Pulm Pharmacol Ther 2021; 70:102059. [PMID: 34302984 DOI: 10.1016/j.pupt.2021.102059] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/06/2021] [Accepted: 07/17/2021] [Indexed: 10/20/2022]
Abstract
Transient receptor potential ankyrin-1 (TRPA1) is an ion channel mediating pain and cough signals in sensory neurons. We and others have shown that TRPA1 is also expressed in some non-neuronal cells and supports inflammatory responses. To address the pathogenesis and to uncover potential targets for pharmacotherapy in inflammatory lung diseases, we set out to study the expression of TRPA1 in human A549 lung epithelial cells under inflammatory conditions. TRPA1 expression was determined by RT-qPCR and Western blotting at a mRNA and protein level, respectively and its function was studied by Fluo 3-AM intracellular Ca2+ measurement in A549 lung epithelial cells. TRPA1 promoter activity was assessed by reporter gene assay. TRPA1 expression was very low in A549 cells in the absence of inflammatory stimuli. Tumor necrosis factor-α (TNF-α) significantly increased TRPA1 expression and a synergy was found between TNF-α, interleukin-1β (IL-1β) and interferon-γ (IFN-γ). Reporter gene experiments indicate that the combination of TNF-α and IL-1β increases TRPA1 promoter activity while the effect of IFN-γ seems to be non-transcriptional. Interestingly, the glucocorticoid dexamethasone downregulated TRPA1 expression in A549 cells by reducing TRPA1 mRNA stability in a transcription-dependent manner. Furthermore, pharmacological blockade of TRPA1 reduced the production of the pro-inflammatory cytokine IL-8. In conclusion, TRPA1 was found to be expressed and functional in human A549 lung epithelial cells under inflammatory conditions. The anti-inflammatory steroid dexamethasone reduced TRPA1 expression through post-transcriptional mechanisms. The results reveal TRPA1 as a potential mediator and drug target in inflammatory lung conditions.
Collapse
Affiliation(s)
- Samu Luostarinen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Mari Hämäläinen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Noriyuki Hatano
- Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya, Japan
| | - Katsuhiko Muraki
- Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya, Japan
| | - Eeva Moilanen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland.
| |
Collapse
|
34
|
Hirawat R, Saifi MA, Godugu C. Targeting inflammatory cytokine storm to fight against COVID-19 associated severe complications. Life Sci 2021; 267:118923. [PMID: 33358906 PMCID: PMC7831473 DOI: 10.1016/j.lfs.2020.118923] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 12/09/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023]
Abstract
Such testing and trying time probably never seen before in the human history. The novel coronavirus disease abbreviated as COVID-19 is the ongoing health crisis which entered into human life in late December 2019. The ease of transmission between humans and the undetectability in early stage makes COVID-19 frightening and unprecedented. The disease is characterised by pneumonia progressing to breathing difficulty, acute respiratory distress syndrome (ARDS) and multi-organ failure. Clinical studies suggest excessive release of inflammatory mediators leads to cytokine storm, a phenomenon which appears to be potentially life-threatening in COVID-19. Across the globe, when the world authorities are grappling to contain the virus, our review provides a glimpse on structure, pathophysiology of the virus and further sheds light on various clinical complications associated with the disease in order to open up/raise new horizons to explore various possible theoretical targets for COVID-19. The review also portrays a question and debates: Can targeting cytokine storm can be a feasible approach to combat COVID-19?
Collapse
Affiliation(s)
- Rishabh Hirawat
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Mohd Aslam Saifi
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India.
| |
Collapse
|
35
|
Impact of cytokines levels and interleukin 6 (-634 C>G) polymorphism on clinical outcomes in patients with sepsis. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
36
|
Ríos-Ríos WDJ, Sosa-Luis SA, Torres-Aguilar H. T Cells Subsets in the Immunopathology and Treatment of Sjogren's Syndrome. Biomolecules 2020; 10:E1539. [PMID: 33187265 PMCID: PMC7698113 DOI: 10.3390/biom10111539] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/06/2020] [Accepted: 11/08/2020] [Indexed: 02/06/2023] Open
Abstract
Sjogren´s syndrome (SS) is an autoimmune disease whose pathogenesis is characterized by an exacerbated T cell infiltration in exocrine glands, markedly associated to the inflammatory and detrimental features as well as the disease progression. Several helper T cell subsets sequentially converge at different stages of the ailment, becoming involved in specific pathologic roles. Initially, their activated phenotype endows them with high migratory properties and increased pro-inflammatory cytokine secretion in target tissues. Later, the accumulation of immunomodulatory T cells-derived factors, such as IL-17, IFN-γ, or IL-21, preserve the inflammatory environment. These effects favor strong B cell activation, instigating an extrafollicular antibody response in ectopic lymphoid structures mediated by T follicular helper cells (Tfh) and leading to disease progression. Additionally, the memory effector phenotype of CD8+ T cells present in SS patients suggests that the presence of auto-antigen restricted CD8+ T cells might trigger time-dependent and specific immune responses. Regarding the protective roles of traditional regulatory T cells (Treg), uncertain evidence shows decrease or invariable numbers of circulating and infiltrating cells. Nevertheless, an emerging Treg subset named follicular regulatory T cells (Tfr) seems to play a critical protective role owing to their deficiency that enhances SS development. In this review, the authors summarize the current knowledge of T cells subsets contribution to the SS immunopathology, focusing on the cellular and biomolecular properties allowing them to infiltrate and to harm target tissues, and that simultaneously make them key therapeutic targets for SS treatment.
Collapse
Affiliation(s)
- William de Jesús Ríos-Ríos
- Department of Clinical Immunology Research of Biochemical Sciences Faculty, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca City 68120, Mexico;
| | - Sorely Adelina Sosa-Luis
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico;
| | - Honorio Torres-Aguilar
- Department of Clinical Immunology Research of Biochemical Sciences Faculty, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca City 68120, Mexico;
| |
Collapse
|
37
|
Casella G, Rasouli J, Thome R, Descamps HC, Vattikonda A, Ishikawa L, Boehm A, Hwang D, Zhang W, Xiao D, Park J, Zhang GX, Alvarez JI, Rostami A, Ciric B. Interferon-γ/Interleukin-27 Axis Induces Programmed Death Ligand 1 Expression in Monocyte-Derived Dendritic Cells and Restores Immune Tolerance in Central Nervous System Autoimmunity. Front Immunol 2020; 11:576752. [PMID: 33193372 PMCID: PMC7649367 DOI: 10.3389/fimmu.2020.576752] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/02/2020] [Indexed: 12/03/2022] Open
Abstract
Antigen (Ag)-specific tolerance induction by intravenous (i. v.) injection of high-dose auto-Ags has been explored for therapy of autoimmune diseases, including multiple sclerosis (MS). It is thought that the advantage of such Ag-specific therapy over non-specific immunomodulatory treatments would be selective suppression of a pathogenic immune response without impairing systemic immunity, thus avoiding adverse effects of immunosuppression. Auto-Ag i.v. tolerance induction has been extensively studied in experimental autoimmune encephalomyelitis (EAE), an animal model of MS, and limited clinical trials demonstrated that it is safe and beneficial to a subset of MS patients. Nonetheless, the mechanisms of i.v. tolerance induction are incompletely understood, hampering the development of better approaches and their clinical application. Here, we describe a pathway whereby auto-Ag i.v. injected into mice with ongoing clinical EAE induces interferon-gamma (IFN-γ) secretion by auto-Ag-specific CD4+ T cells, triggering interleukin (IL)-27 production by conventional dendritic cells type 1 (cDC1). IL-27 then, via signal transducer and activator of transcription 3 activation, induces programmed death ligand 1 (PD-L1) expression by monocyte-derived dendritic cells (moDCs) in the central nervous system of mice with EAE. PD-L1 interaction with programmed cell death protein 1 on pathogenic CD4+ T cells leads to their apoptosis/anergy, resulting in disease amelioration. These findings identify a key role of the IFN-γ/IL-27/PD-L1 axis, involving T cells/cDC1/moDCs in the induction of i.v. tolerance.
Collapse
Affiliation(s)
- Giacomo Casella
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Javad Rasouli
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Rodolfo Thome
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Hélène C Descamps
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Asrita Vattikonda
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Larissa Ishikawa
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Alexandra Boehm
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Daniel Hwang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Weifeng Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Dan Xiao
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Jeongho Park
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States.,College of Veterinary Medicine & Institute of Veterinary Science, Kangwon National University, Chuncheon, South Korea
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Jorge I Alvarez
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Bogoljub Ciric
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
38
|
Enteroviral Pathogenesis of Type 1 Diabetes: The Role of Natural Killer Cells. Microorganisms 2020; 8:microorganisms8070989. [PMID: 32630332 PMCID: PMC7409131 DOI: 10.3390/microorganisms8070989] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/27/2020] [Accepted: 06/28/2020] [Indexed: 12/16/2022] Open
Abstract
Enteroviruses, especially group B coxsackieviruses (CV-B), have been associated with the development of chronic diseases such as type 1 diabetes (T1D). The pathological mechanisms that trigger virus-induced autoimmunity against islet antigens in T1D are not fully elucidated. Animal and human studies suggest that NK cells response to CV-B infection play a crucial role in the enteroviral pathogenesis of T1D. Indeed, CV-B-infected cells can escape from cytotoxic T cells recognition and destruction by inhibition of cell surface expression of HLA class I antigen through non-structural viral proteins, but they can nevertheless be killed by NK cells. Cytolytic activity of NK cells towards pancreatic beta cells persistently-infected with CV-B has been reported and defective viral clearance by NK cells of patients with T1D has been suggested as a mechanism leading to persistence of CV-B and triggering autoimmunity reported in these patients. The knowledge about host antiviral defense against CV-B infection is not only crucial to understand the susceptibility to virus-induced T1D but could also contribute to the design of new preventive or therapeutic approaches for individuals at risk for T1D or newly diagnosed patients.
Collapse
|
39
|
SARS-CoV-2 infection: The role of cytokines in COVID-19 disease. Cytokine Growth Factor Rev 2020; 54:62-75. [PMID: 32513566 PMCID: PMC7265853 DOI: 10.1016/j.cytogfr.2020.06.001] [Citation(s) in RCA: 742] [Impact Index Per Article: 148.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 05/19/2020] [Accepted: 06/01/2020] [Indexed: 02/06/2023]
Abstract
A wide range of cytokines are involved in the development of COVID-19 disease. Some of these biomolecules are related to the progression and even to the prognosis of the infection. Findings on the role of cytokine storm associated with SARS-CoV-2 infection can be useful in order to manage this highly virulent disease.
COVID-19 disease, caused by infection with SARS-CoV-2, is related to a series of physiopathological mechanisms that mobilize a wide variety of biomolecules, mainly immunological in nature. In the most severe cases, the prognosis can be markedly worsened by the hyperproduction of mainly proinflammatory cytokines, such as IL-1, IL-6, IL-12, IFN-γ, and TNF-α, preferentially targeting lung tissue. This study reviews published data on alterations in the expression of different cytokines in patients with COVID-19 who require admission to an intensive care unit. Data on the implication of cytokines in this disease and their effect on outcomes will support the design of more effective approaches to the management of COVID-19.
Collapse
|
40
|
Kato M. New insights into IFN-γ in rheumatoid arthritis: role in the era of JAK inhibitors. Immunol Med 2020; 43:72-78. [PMID: 32338187 DOI: 10.1080/25785826.2020.1751908] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 03/30/2020] [Indexed: 02/08/2023] Open
Abstract
The treatment of rheumatoid arthritis (RA) is now entering a new era, the era of Janus kinase (JAK) inhibitors. JAK inhibitors target multiple cytokines including IL-6 and exhibit a beneficial treatment effect in patients with RA and inadequate response to conventional synthetic or biologic disease-modifying anti-rheumatic drugs. Since the treatment effect of JAK inhibitors is promising even for patients refractory to anti-IL-6 therapy, it needs to be considered how multiple cytokines play roles in the pathogenesis of RA. It is also worth noting that an increased risk of herpes zoster is specifically related to the use of JAK inhibitors. Among cytokines targeted by JAK inhibitors, the current review focuses on IFN-γ, particularly on its role in synovial biology, autoimmunity, bone metabolism, pain, and varicella zoster virus infection. Recent studies provided new insights into IFN-γ in the pathogenesis of RA, which may account for the efficacy of JAK inhibitors.
Collapse
Affiliation(s)
- Masaru Kato
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
41
|
Majai GE, Gogolák P, Tóth M, Hodrea J, Horváth D, Fésüs L, Rajnavölgyi É, Bácsi A. Autologous apoptotic neutrophils inhibit inflammatory cytokine secretion by human dendritic cells, but enhance Th1 responses. FEBS Open Bio 2020; 10:1492-1502. [PMID: 32473089 PMCID: PMC7396436 DOI: 10.1002/2211-5463.12904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 05/26/2020] [Indexed: 01/13/2023] Open
Abstract
Neutrophils represent the most abundant cell type in peripheral blood and exhibit a remarkably brief (6–8 h) half‐life in circulation. The fundamental role of these professional phagocytes has been established in acute inflammation, based on their potential to both initiate and receive inflammatory signals. Furthermore, neutrophils also take part in maintaining chronic inflammatory processes, such as in various autoimmune diseases. Here, we demonstrate that human autologous apoptotic neutrophils are readily engulfed by immature monocyte‐derived dendritic cells (moDCs) with similar efficiency as allogeneic apoptotic neutrophils [Majai G et al. (2010) J Leukoc Biol 88, 981–991]. Interestingly, in contrast to the allogeneic system, exposure of moDCs to autologous apoptotic neutrophils inhibits LPS + IFN‐γ‐induced production of inflammatory cytokines in a phagocytosis‐independent manner. Autologous apoptotic neutrophil‐primed DCs are able to modulate T‐cell responses by inducing the generation of IFN‐γ‐secreting cells while hampering that of IL‐17A‐producing cells. Our observations indicate that capture of autologous apoptotic neutrophils by immature DCs may impede further neutrophil‐mediated phagocytosis and tissue damage, and allow increased clearance of dying cells by macrophages.
Collapse
Affiliation(s)
- Gyöngyike Emese Majai
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Hungary
| | - Péter Gogolák
- Department of Immunology, Faculty of Medicine, University of Debrecen, Hungary
| | - Márta Tóth
- Department of Immunology, Faculty of Medicine, University of Debrecen, Hungary.,Doctoral School of Molecular Cellular and Immune Biology, University of Debrecen, Hungary
| | - Judit Hodrea
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Hungary
| | - Dorottya Horváth
- Department of Immunology, Faculty of Medicine, University of Debrecen, Hungary.,Doctoral School of Molecular Medicine, University of Debrecen, Hungary
| | - László Fésüs
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Hungary
| | - Éva Rajnavölgyi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Hungary
| | - Attila Bácsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Hungary
| |
Collapse
|
42
|
Lefeuvre CMJ, Payet CA, Fayet OM, Maillard S, Truffault F, Bondet V, Duffy D, de Montpreville V, Ghigna MR, Fadel E, Mansuet-Lupo A, Alifano M, Validire P, Gossot D, Behin A, Eymard B, Berrih-Aknin S, Le Panse R. Risk factors associated with myasthenia gravis in thymoma patients: The potential role of thymic germinal centers. J Autoimmun 2020; 106:102337. [DOI: 10.1016/j.jaut.2019.102337] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/21/2019] [Accepted: 09/23/2019] [Indexed: 12/17/2022]
|
43
|
Xu K, Jin L. The role of heparin/heparan sulphate in the IFN-γ-led Arena. Biochimie 2019; 170:1-9. [PMID: 31794784 DOI: 10.1016/j.biochi.2019.11.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 11/26/2019] [Indexed: 02/08/2023]
Abstract
IFN-γ (Interferon-gamma) is a pleiotropic cytokine. It is often involved in a variety of physiological processes by binding to the cell surface transmembrane receptor (IFN-γR) to initiate a series of signalling pathways that transmit external signals from cell surface receptors to the cell nucleus. Heparan sulphate (HS), a highly sulphated linear polysaccharide, is ubiquitous on the mammalian cell surface and extracellular matrix. Electrostatic interactions can be generated between the highly sulphated HS region and specific basic amino acid residues in the IFN-γ structure, thereby detaining IFN-γ on the cell surface, and the concentration of IFN-γ on the cell surface is thus, changed. IFN-γ retained on the cell surface will optimize the binding of IFN-γ to the transmembrane receptor resulting in high efficiency signalling. Heparin is a glycosaminoglycan with a structure similar to HS. The structural similarity provides a basis for modelling exogenous heparin dependence for interference with IFN-γ function. This model can be summarized as follows: First, the competitive binding effect; heparin bound to cytokines by competing with membrane-associated HS, causes a decrease in cytokine concentration on the cell surface. Second, the principle of priority occupancy; heparin can occupy the receptor binding site on cytokines, partially preventing the IFN-γ-IFN-γR interaction. These two models interfere with IFN-γ signal transmission. To decipher the mechanism by which heparin influences IFN-γ activity, studies of the structure-activity relationship are in progress. This paper summarizes research progress on the IFN-γ signalling pathway, heparin interference with IFN-γ activity and the structure-activity relationship between heparin and IFN-γ.
Collapse
Affiliation(s)
- Kening Xu
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, 266000, Shandong, PR China
| | - Lan Jin
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, 266000, Shandong, PR China.
| |
Collapse
|
44
|
Petermann F, Pękowska A, Johnson CA, Jankovic D, Shih HY, Jiang K, Hudson WH, Brooks SR, Sun HW, Villarino AV, Yao C, Singleton K, Akondy RS, Kanno Y, Sher A, Casellas R, Ahmed R, O'Shea JJ. The Magnitude of IFN-γ Responses Is Fine-Tuned by DNA Architecture and the Non-coding Transcript of Ifng-as1. Mol Cell 2019; 75:1229-1242.e5. [PMID: 31377117 PMCID: PMC6754279 DOI: 10.1016/j.molcel.2019.06.025] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 04/05/2019] [Accepted: 06/18/2019] [Indexed: 12/24/2022]
Abstract
Interferon gamma (IFN-γ), critical for host defense and tumor surveillance, requires tight control of its expression. Multiple cis-regulatory elements exist around Ifng along with a non-coding transcript, Ifng-as1 (also termed NeST). Here, we describe two genetic models generated to dissect the molecular functions of this locus and its RNA product. DNA deletion within the Ifng-as1 locus disrupted chromatin organization of the extended Ifng locus, impaired Ifng response, and compromised host defense. Insertion of a polyA signal ablated the Ifng-as1 full-length transcript and impaired host defense, while allowing proper chromatin structure. Transient knockdown of Ifng-as1 also reduced IFN-γ production. In humans, discordant expression of IFNG and IFNG-AS1 was evident in memory T cells, with high expression of this long non-coding RNA (lncRNA) and low expression of the cytokine. These results establish Ifng-as1 as an important regulator of Ifng expression, as a DNA element and transcribed RNA, involved in dynamic and cell state-specific responses to infection.
Collapse
Affiliation(s)
- Franziska Petermann
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, NIAMS, NIH, Bethesda, MD 20892, USA
| | | | - Catrina A Johnson
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, NIAMS, NIH, Bethesda, MD 20892, USA
| | - Dragana Jankovic
- Immunobiology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Han-Yu Shih
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, NIAMS, NIH, Bethesda, MD 20892, USA
| | - Kan Jiang
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, NIAMS, NIH, Bethesda, MD 20892, USA
| | - William H Hudson
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Stephen R Brooks
- Biodata Mining and Discovery Section, NIAMS, NIH, Bethesda, MD 20892, USA
| | - Hong-Wei Sun
- Biodata Mining and Discovery Section, NIAMS, NIH, Bethesda, MD 20892, USA
| | - Alejandro V Villarino
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, NIAMS, NIH, Bethesda, MD 20892, USA
| | - Chen Yao
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, NIAMS, NIH, Bethesda, MD 20892, USA
| | - Kentner Singleton
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, NIAMS, NIH, Bethesda, MD 20892, USA
| | - Rama S Akondy
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Yuka Kanno
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, NIAMS, NIH, Bethesda, MD 20892, USA.
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Rafael Casellas
- Lymphocyte Nuclear Biology, NIAMS, NIH, Bethesda, MD 20892, USA
| | - Rafi Ahmed
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - John J O'Shea
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, NIAMS, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
45
|
Sharkey DJ, Glynn DJ, Schjenken JE, Tremellen KP, Robertson SA. Interferon-gamma inhibits seminal plasma induction of colony-stimulating factor 2 in mouse and human reproductive tract epithelial cells. Biol Reprod 2019; 99:514-526. [PMID: 29596569 DOI: 10.1093/biolre/ioy071] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 03/26/2018] [Indexed: 12/26/2022] Open
Abstract
Seminal fluid interacts with the female reproductive tract to initiate a permissive immune response that facilitates embryo implantation and pregnancy success. The immune-regulatory cytokine interferon-γ (IFNG), which can be elevated in seminal plasma, is associated with reduced fertility. Here, we investigated how IFNG influences the female immune response to seminal fluid. In human Ect1 cervical epithelial cells, IFNG added at physiologically relevant concentrations substantially impaired seminal plasma-induced synthesis of key cytokines colony-stimulating factor 2 (CSF2) and interleukin-6 (IL6). Seminal fluid-induced CSF2 synthesis was also suppressed in the uterus of mice in vivo, when IFNG was delivered transcervically 12 h after mating. Transforming growth factor B1 (TGFB1) is the major seminal fluid signaling factor which elicits CSF2 induction, and IFNG exhibited potent dose-dependent suppression of CSF2 synthesis induced by TGFB1 in murine uterine epithelial cells in vitro. Similarly, IFNG suppressed TGFB1-mediated CSF2 induction in Ect1 cells and human primary cervical epithelial cells; however, IL6 regulation by IFNG was independent of TGFB1. Quantitative PCR confirmed that CSF2 regulation by IFNG in Ect1 cells occurs at the gene transcription level, secondary to IFNG suppression of TGFBR2 encoding TGFB receptor 2. Conversely, TGFB1 suppressed IFNG receptor 1 and 2 genes IFNGR1 and IFNGR2. These data identify IFNG as a potent inhibitor of the TGFB-mediated seminal fluid interaction with relevant reproductive tract epithelia in mice and human. These findings raise the prospect that IFNG in the male partner's seminal fluid impairs immune adaptation for pregnancy following coitus in women.
Collapse
Affiliation(s)
- David J Sharkey
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Danielle J Glynn
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - John E Schjenken
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Kelton P Tremellen
- Repromed Pty Ltd, Dulwich, South Australia, Australia.,School of Pharmacy and Medical Sciences, University of South Australia, South Australia, Australia
| | - Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
46
|
Chung KF. IFN-γ: A Driver of Cough Hypersensitivity Pathways in Chronic Cough? Am J Respir Crit Care Med 2019; 198:827-828. [PMID: 29758165 DOI: 10.1164/rccm.201804-0740ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Kian Fan Chung
- 1 National Heart and Lung Institute Imperial College London London, United Kingdom and.,2 Royal Brompton and Harefield NHS Trust London, United Kingdom
| |
Collapse
|
47
|
Weyand CM, Watanabe R, Zhang H, Akiyama M, Berry GJ, Goronzy JJ. Cytokines, growth factors and proteases in medium and large vessel vasculitis. Clin Immunol 2019; 206:33-41. [PMID: 30772599 PMCID: PMC6693995 DOI: 10.1016/j.clim.2019.02.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/13/2019] [Accepted: 02/13/2019] [Indexed: 02/06/2023]
Abstract
Giant cell arteritis and Takayasu arteritis are autoimmune vasculitides that cause aneurysm formation and tissue infarction. Extravascular inflammation consists of an intense acute phase response. Deeper understanding of pathogenic events in the vessel wall has highlighted the loss of tissue protective mechanisms, the intrusion of immune cells into "forbidden territory", and the autonomy of self-renewing vasculitic infiltrates. Adventitial vasa vasora critically control vessel wall access and drive differentiation of tissue-invasive T cells. Selected T cells establish tissue residency and build autonomous, self-sufficient inflammatory lesions. Pathogenic effector T cells intrude and survive due to failed immune checkpoint inhibition. Vasculitis-sustaining T cells and macrophages provide a broad portfolio of effector functions, involving heterogeneous populations of pro-inflammatory T cells and diverse macrophage subsets that ultimately induce wall capillarization and intimal hyperplasia. Redirecting diagnostic and therapeutic strategies from control of extravascular inflammatory markers to suppression of vascular inflammation will improve disease management.
Collapse
Affiliation(s)
- Cornelia M Weyand
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, USA.
| | - Ryu Watanabe
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, USA
| | - Hui Zhang
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, USA
| | - Mitsuhiro Akiyama
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, USA
| | - Gerald J Berry
- Department of Pathology, Stanford University School of Medicine, USA
| | - Jörg J Goronzy
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, USA
| |
Collapse
|
48
|
Christiansen RJ, Münch HJ, Bonefeld CM, Thyssen JP, Sloth JJ, Geisler C, Søballe K, Jellesen MS, Jakobsen SS. Cytokine Profile in Patients with Aseptic Loosening of Total Hip Replacements and Its Relation to Metal Release and Metal Allergy. J Clin Med 2019; 8:jcm8081259. [PMID: 31434199 PMCID: PMC6723430 DOI: 10.3390/jcm8081259] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/06/2019] [Accepted: 08/09/2019] [Indexed: 12/13/2022] Open
Abstract
Metal release from total hip replacements (THRs) is associated with aseptic loosening (AL). It has been proposed that the underlying immunological response is caused by a delayed type IV hypersensitivity-like reaction to metals, i.e., metal allergy. The purpose of this study was to investigate the immunological response in patients with AL in relation to metal release and the prevalence of metal allergy. THR patients undergoing revision surgery due to AL or mechanical implant failures were included in the study along with a control group consisting of primary THR patients. Comprehensive cytokine analyses were performed on serum and periimplant tissue samples along with metal analysis using inductive coupled plasma mass spectrometry (ICP-MS). Patient patch testing was done with a series of metals related to orthopedic implant. A distinct cytokine profile was found in the periimplant tissue of patients with AL. Significantly increased levels of the proinflammatory cytokines IL-1β, IL-2, IL-8, IFN-γ and TNF-α, but also the anti-inflammatory IL-10 were detected. A general increase of metal concentrations in the periimplant tissue was observed in both revision groups, while Cr was significantly increased in patient serum with AL. No difference in the prevalence of metal sensitivity was established by patch testing. Increased levels of IL-1β, IL-8, and TNF-α point to an innate immune response. However, the presence of IL-2 and IFN-γ indicates additional involvement of T cell-mediated response in patients with AL, although this could not be detected by patch testing.
Collapse
Affiliation(s)
- Rune J Christiansen
- Department of Mechanical Engineering, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark.
- Department of Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| | - Henrik J Münch
- Institute of Clinical Medicine-Orthopedic Surgery, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Charlotte M Bonefeld
- Department of Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Jacob P Thyssen
- Institute of Clinical Medicine, Copenhagen University, Gentofte Hospital, DK-2900 Hellerup, Denmark
| | - Jens J Sloth
- National Food Institute, Research Group on Nanobio Science, Technical University of Denmark, DK-2860 Søborg, Denmark
| | - Carsten Geisler
- Department of Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Kjeld Søballe
- Institute of Clinical Medicine-Orthopedic Surgery, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Morten S Jellesen
- Department of Mechanical Engineering, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - Stig S Jakobsen
- Institute of Clinical Medicine-Orthopedic Surgery, Aarhus University, DK-8000 Aarhus C, Denmark.
| |
Collapse
|
49
|
Ibarra A, Mendieta-Arbesú E, Suarez-Meade P, García-Vences E, Martiñón S, Rodriguez-Barrera R, Lomelí J, Flores-Romero A, Silva-García R, Buzoianu-Anguiano V, Borlongan CV, Frydman TD. Motor Recovery after Chronic Spinal Cord Transection in Rats: A Proof-of-Concept Study Evaluating a Combined Strategy. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2019; 18:52-62. [DOI: 10.2174/1871527317666181105101756] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/20/2018] [Accepted: 10/29/2018] [Indexed: 12/14/2022]
Abstract
Background:
The chronic phase of Spinal Cord (SC) injury is characterized by the presence
of a hostile microenvironment that causes low activity and a progressive decline in neurological function;
this phase is non-compatible with regeneration. Several treatment strategies have been investigated
in chronic SC injury with no satisfactory results. OBJECTIVE- In this proof-of-concept study,
we designed a combination therapy (Comb Tx) consisting of surgical glial scar removal plus scar inhibition,
accompanied with implantation of mesenchymal stem cells (MSC), and immunization with
neural-derived peptides (INDP).
Methods:
This study was divided into three subsets, all in which Sprague Dawley rats were subjected
to a complete SC transection. Sixty days after injury, animals were randomly allocated into two groups
for therapeutic intervention: control group and animals receiving the Comb-Tx. Sixty-three days after
treatment we carried out experiments analyzing motor recovery, presence of somatosensory evoked
potentials, neural regeneration-related genes, and histological evaluation of serotoninergic fibers.
Results:
Comb-Tx induced a significant locomotor and electrophysiological recovery. An increase in the
expression of regeneration-associated genes and the percentage of 5-HT+ fibers was noted at the caudal
stump of the SC of animals receiving the Comb-Tx. There was a significant correlation of locomotor recovery
with positive electrophysiological activity, expression of GAP43, and percentage of 5-HT+ fibers.
Conclusion:
Comb-Tx promotes motor and electrophysiological recovery in the chronic phase of SC
injury subsequent to a complete transection. Likewise, it is capable of inducing the permissive microenvironment
to promote axonal regeneration.
Collapse
Affiliation(s)
- Antonio Ibarra
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| | - Erika Mendieta-Arbesú
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| | - Paola Suarez-Meade
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| | - Elisa García-Vences
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| | | | - Roxana Rodriguez-Barrera
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| | - Joel Lomelí
- Instituto Politecnico Nacional, Escuela Superior de Medicina, Ciudad de Mexico, Mexico
| | - Adrian Flores-Romero
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| | | | | | - Cesar V. Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, United States
| | - Tamara D. Frydman
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| |
Collapse
|
50
|
Yang L, Zhang S, Wang G. Keratin 17 in disease pathogenesis: from cancer to dermatoses. J Pathol 2018; 247:158-165. [PMID: 30306595 DOI: 10.1002/path.5178] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/17/2018] [Accepted: 10/02/2018] [Indexed: 12/16/2022]
Abstract
Keratin 17 (K17) is a type I intermediate filament mainly expressed in the basal cells of epithelia. As a multifaceted cytoskeletal protein, K17 regulates a myriad of biological processes, including cell proliferation and growth, skin inflammation and hair follicle cycling. Aberrant overexpression of K17 is found in various diseases ranging from psoriasis to malignancies such as breast, cervical, oral squamous and gastric carcinomas. Moreover, genetic mutation in KRT17 is related to tissue-specific diseases, represented by steatocystoma multiplex and pachyonychia congenita. In this review, we summarize our findings concerning the regulatory mechanisms of K17 overexpression in psoriasis and compare them to the literature relating to other diseases. We discuss data that proinflammatory cytokines, including interleukin-17 (IL-17), IL-22, interferon-gamma (IFN-γ), transforming growth factor-beta (TGF-β) and transcription factors glioma-associated oncogene homolog 1/2 (Gli1/2), Nrf2 and p53 can regulate K17 by transcriptional and translational control. Moreover, post-translational modification, including phosphorylation and ubiquitination, is involved in the regulation of K17 stability and biological functions. We therefore review the current understanding of the K17 regulatory mechanism and its pathogenic role in diseases from dermatoses to cancer. Prospects for anti-K17 therapy in diagnosis, prognosis and disease treatment are also discussed. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Luting Yang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR, China
| | - Shaolong Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR, China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR, China
| |
Collapse
|