1
|
Amer H, Flanagan KL, Kampan NC, Itsiopoulos C, Scott CL, Kartikasari AER, Plebanski M. Interleukin-6 Is a Crucial Factor in Shaping the Inflammatory Tumor Microenvironment in Ovarian Cancer and Determining Its Hot or Cold Nature with Diagnostic and Prognostic Utilities. Cancers (Basel) 2025; 17:1691. [PMID: 40427188 PMCID: PMC12109964 DOI: 10.3390/cancers17101691] [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: 03/17/2025] [Revised: 05/05/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Ovarian cancer (OC) remains the leading cause of cancer-related deaths among women, often diagnosed at advanced stages due to the lack of effective early diagnostic procedures. To reduce the high mortality rates in OC, reliable biomarkers are urgently needed, especially to detect OC at its earliest stage, predict specific drug responses, and monitor patients. The cytokine interleukin-6 (IL6) is associated with low survival rates, treatment resistance, and recurrence. In this review, we summarize the role of IL6 in inflammation and how IL6 contributes to ovarian tumorigenesis within the tumor microenvironment, influencing whether the tumor is subsequently classified as "hot" or "cold". We further dissect the molecular and cellular mechanisms through which IL6 production and downstream signaling are regulated, to enhance our understanding of its involvement in OC development, as well as OC resistance to treatment. We highlight the potential of IL6 to be used as a reliable diagnostic biomarker to help detect OC at its earliest stage, and as a part of predictive and prognostic signatures to improve OC management. We further discuss ways to leverage artificial intelligence and machine learning to integrate IL6 into diverse biomarker-based strategies.
Collapse
Affiliation(s)
- Hina Amer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia
| | - Katie L. Flanagan
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia
- School of Medicine and Health Sciences, University of Tasmania, Launceston, TAS 7250, Australia
- Tasmanian Vaccine Trial Centre, Clifford Craig Foundation, Launceston General Hospital, Launceston, TAS 7250, Australia
| | - Nirmala C. Kampan
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Catherine Itsiopoulos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia
| | - Clare L. Scott
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Faculty of Medicine, Dentistry, and Health Sciences, The University of Melbourne, Parkville, VIC 3052, Australia
- The Royal Women’s Hospital, Parkville, VIC 3052, Australia
| | | | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia
| |
Collapse
|
2
|
Wu M, Sapin-Minet A, Gaucher C. Heparin, an active excipient to carry biosignal molecules: Applications in tissue engineering - A review. Int J Biol Macromol 2025; 312:143959. [PMID: 40334894 DOI: 10.1016/j.ijbiomac.2025.143959] [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: 02/07/2025] [Revised: 04/29/2025] [Accepted: 05/04/2025] [Indexed: 05/09/2025]
Abstract
Drug repositioning refers to new medical application exploration for existing drugs. Heparins, beyond their well-known anticoagulant properties widely used in clinics, present the capacity to carry biosignal molecules that is responsible for other properties such as anti-inflammatory, angiogenesis. Thus, heparins interaction with different biosignal molecules such as cytokines and growth factors have recently drawn attention and have promoted heparin repositioning as an active excipient with useful applications as drug-delivery systems and biomaterial-based tissue engineering scaffolds. Indeed, biomaterial heparinization can further help in their formulation such as in self-assembled heparin-based hydrogels or nanoparticles, and improve their biocompatibility. Moreover, the capacity of heparin to carry biosignal molecules enables the direct functionalization of heparinized biomaterial for tissue engineering. Both heparin characteristics namely the biosignal molecule carrying and biomaterial heparinization are reviewed here along their combination for biomaterial functionalization in tissue engineering applications.
Collapse
Affiliation(s)
- Meiling Wu
- Université de Lorraine, CITHEFOR, F-54000 Nancy, France
| | | | | |
Collapse
|
3
|
Eltayeb A, Redwan EM. T-cell immunobiology and cytokine storm of COVID-19. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:1-30. [PMID: 40246342 DOI: 10.1016/bs.pmbts.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The 2019 coronavirus illness (COVID 2019) first manifests as a newly identified pneumonia and may quickly escalate to acute respiratory distress syndrome, which has caused a global pandemic. Except for individualized supportive care, no curative therapy has been steadfastly advised for COVID-19 up until this point. T cells and virus-specific T lymphocytes are required to guard against viral infection, particularly COVID-19. Delayed immunological reconstitution (IR) and cytokine storm (CS) continue to be significant barriers to COVID-19 cure. While severe COVID-19 patients who survived the disease had considerable lymphopenia and increased neutrophils, especially in the elderly, their T cell numbers gradually recovered. Exhausted T lymphocytes and elevated levels of pro-inflammatory cytokines, including IL6, IL10, IL2, and IL17, are observed in peripheral blood and the lungs. It implies that while convalescent plasma, IL-6 blocking, mesenchymal stem cells, and corticosteroids might decrease CS, Thymosin α1 and adaptive COVID-19-specific T cells could enhance IR. There is an urgent need for more clinical research in this area throughout the world to open the door to COVID-19 treatment in the future.
Collapse
Affiliation(s)
- Ahmed Eltayeb
- Biological Science Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Elrashdy M Redwan
- Biological Science Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
4
|
Phan AT, Aunins E, Cruz-Morales E, Dwivedi G, Bunkofske M, Eberhard JN, Aldridge DL, Said H, Banda O, Tam Y, Christian DA, Vonderheide RH, Kedl RM, Weissman D, Alameh MG, Hunter CA. The type I IFN-IL-27 axis promotes mRNA vaccine-induced CD8 + T cell responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.16.633383. [PMID: 39896632 PMCID: PMC11785111 DOI: 10.1101/2025.01.16.633383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The ability of lipid nanoparticle (LNP)-delivered mRNA vaccines to induce type I IFNs is critical to promote CD8 + T cell responses. The studies presented here indicate that immunization with nucleoside modified mRNA-LNP vaccines drives myeloid cell expression of the cytokine IL-27, which acts on antigen-specific CD8 + T cells to sustain T cell expansion. In vitro and in vivo studies revealed that type I IFN signaling is necessary for mRNA-LNP-induced IL-27 production, that immunization failed in IL-27 KO mice, and that immunization of IFNAR1-deficient mice with mRNA-LNP particles that also encode IL-27 mRNA restored antigen-specific CD8 + T cell responses. In addition, IL-27 mRNA-LNPs served as an adjuvant that improved cytolytic CD8 + T cell responses and the therapeutic efficacy of mRNA-LNPs to drive anti-pathogen and anti-tumor immunity. These studies highlight the central role of IL-27 in mRNA-LNP induced CD8 + T cell responses and the ability of this cytokine to augment the functionality of the CD8 + T cell response for prophylactic or therapeutic immunization.
Collapse
|
5
|
Yang YL, Zhang RR, Pang JY, Xing JH, Guo TK, Shi CW, Yang GL, Huang HB, Jiang YL, Wang JZ, Cao X, Wang N, Zeng Y, Yang WT, Yao JY, Wang CF. Immunoprotective effect of recombinant Lactobacillus plantarum expressing largemouth bass virus MCP on largemouth bass. FISH & SHELLFISH IMMUNOLOGY 2024; 155:109986. [PMID: 39461397 DOI: 10.1016/j.fsi.2024.109986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 09/26/2024] [Accepted: 10/24/2024] [Indexed: 10/29/2024]
Abstract
Largemouth bass virus (LMBV) is an infectious pathogen that causes high mortality rates in largemouth bass, and outbreaks of this virus can significantly harm the aquaculture industry. Currently, no vaccine has been developed that can effectively prevent the transmission of LMBV. In this study, we constructed a recombinant Lactobacillus plantarum (L. plantarum) strain capable of expressing the MCP gene of LMBV and displaying this protein on its surface; then, we evaluated the immunoprotective effect of this recombinant bacterium on largemouth bass. Western blotting, immunofluorescence, and flow cytometry confirmed that MCP was successfully expressed and anchored on the surfaces of NC8 cells. Immunization of largemouth bass with NC8-pSIP409-pgsA'-MCP via the oral feeding route induced CD4, CD8, IL-1β, and IL-6 gene expression. In addition, NC8-pSIP409-pgsA'-MCP at different CFUs increased the survival of largemouth bass after LMBV infection; in particular, NC8-pSIP409-pgsA'-MCP (109 CFU) resulted in approximately 30 % survival. NC8-pSIP409-pgsA'-MCP immunization alleviated the pathological changes in the liver and spleen, exerting a more advantageous protective effect. These data suggest that the recombinant L. plantarum strain NC8-pSIP409-pgsA'-MCP can increase the resistance of largemouth bass to LMBV infection and that this strain is a promising candidate oral vaccine for the prevention of LMBV infection.
Collapse
Affiliation(s)
- Yong-Lei Yang
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Rong-Rong Zhang
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Jing-Yi Pang
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Jun-Hong Xing
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Tian-Kui Guo
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chun-Wei Shi
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Gui-Lian Yang
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Hai-Bin Huang
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yan-Long Jiang
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Jian-Zhong Wang
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Xin Cao
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Nan Wang
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yan Zeng
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Wen-Tao Yang
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China.
| | - Jia-Yun Yao
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China; Agriculture Ministry Key Laboratory of Healthy Freshwater Aquaculture, Key Laboratory of Fish Health and Nutrition of Zhejiang Province, Zhejiang Institute of Freshwater Fisheries, Huzhou, China.
| | - Chun-Feng Wang
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China.
| |
Collapse
|
6
|
Ding JQ, Zhang JQ, Zhao SJ, Jiang DB, Lu JR, Yang SY, Wang J, Sun YJ, Huang YN, Hu CC, Zhang XY, Zhang JX, Liu TY, Han CY, Qiao XP, Guo J, Zhao C, Yang K. Follicular CD8 + T cells promote immunoglobulin production and demyelination in multiple sclerosis and a murine model. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167303. [PMID: 38878831 DOI: 10.1016/j.bbadis.2024.167303] [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: 01/15/2024] [Revised: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 06/18/2024]
Abstract
Emerging evidence underscores the importance of CD8+ T cells in the pathogenesis of multiple sclerosis (MS), but the precise mechanisms remain ambiguous. This study intends to elucidate the involvement of a novel subset of follicular CD8+ T cells (CD8+CXCR5+ T) in MS and an experimental autoimmune encephalomyelitis (EAE) murine model. The expansion of CD8+CXCR5+ T cells was observed in both MS patients and EAE mice during the acute phase. In relapsing MS patients, higher frequencies of circulating CD8+CXCR5+ T cells were positively correlated with new gadolinium-enhancement lesions in the central nervous system (CNS). In EAE mice, frequencies of CD8+CXCR5+ T cells were also positively correlated with clinical scores. These cells were found to infiltrate into ectopic lymphoid-like structures in the spinal cords during the peak of the disease. Furthermore, CD8+CXCR5+ T cells, exhibiting high expression levels of ICOS, CD40L, IL-21, and IL-6, were shown to facilitate B cell activation and differentiation through a synergistic interaction between CD40L and IL-21. Transferring CD8+CXCR5+ T cells into naïve mice confirmed their ability to enhance the production of anti-MOG35-55 antibodies and contribute to the disease progression. Consequently, CD8+CXCR5+ T cells may play a role in CNS demyelination through heightening humoral immune responses.
Collapse
Affiliation(s)
- Jia-Qi Ding
- Department of Immunology, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Shaanxi, China; Department of Neurology, Tangdu Hospital, Air Force Medical University (the Fourth Military Medical University), Shaanxi, China
| | - Jun-Qi Zhang
- Department of Immunology, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Shaanxi, China
| | - Si-Jia Zhao
- Department of Neurology, Tangdu Hospital, Air Force Medical University (the Fourth Military Medical University), Shaanxi, China
| | - Dong-Bo Jiang
- Department of Immunology, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Shaanxi, China
| | - Jia-Rui Lu
- Department of Neurology, Tangdu Hospital, Air Force Medical University (the Fourth Military Medical University), Shaanxi, China
| | - Shu-Ya Yang
- Department of Immunology, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Shaanxi, China
| | - Jing Wang
- Department of Immunology, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Shaanxi, China
| | - Yuan-Jie Sun
- Department of Immunology, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Shaanxi, China
| | - Yi-Nan Huang
- Department of Emergency, the Second Affiliated Hospital (Xixian New District Central Hospital), Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Chen-Chen Hu
- Department of Immunology, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Shaanxi, China
| | - Xi-Yang Zhang
- Department of Immunology, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Shaanxi, China
| | - Jia-Xing Zhang
- Department of Immunology, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Shaanxi, China
| | - Tian-Yue Liu
- Department of Immunology, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Shaanxi, China
| | - Chen-Ying Han
- Department of Immunology, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Shaanxi, China
| | - Xu-Peng Qiao
- Department of Immunology, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Shaanxi, China
| | - Jun Guo
- Department of Neurology, Tangdu Hospital, Air Force Medical University (the Fourth Military Medical University), Shaanxi, China.
| | - Cong Zhao
- Department of Neurology, Air Force Medical Center of PLA, Beijing, China.
| | - Kun Yang
- Department of Immunology, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Shaanxi, China.
| |
Collapse
|
7
|
Setiawan E, Putra A, Nabih DI, Ovaditya SZ, Rizaldy R. Mesenchymal stem cells suppress inflammation by downregulating interleukin-6 expression in intestinal perforation animal model. Ann Med Surg (Lond) 2024; 86:5776-5783. [PMID: 39359817 PMCID: PMC11444626 DOI: 10.1097/ms9.0000000000002395] [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: 04/19/2024] [Accepted: 07/01/2024] [Indexed: 10/04/2024] Open
Abstract
Introduction Intestinal perforation has significant fatality due to sepsis contamination and prolonged inflammation. Studies showed that mesenchymal stem cells (MSCs) secreted cytokines and growth factors to reduce inflammation. This study aims to reveal the role of MSCs in controlling inflammation in intestinal perforation wound healing by measuring interleukin-6 (IL-6) and leukocytes in injured tissue. Materials and methods A total of 48 rat models with a 10-mm longitudinal incision at the small intestine were divided into four groups: sham, control, Treatment group 1 (T1) injected with MSC doses of 1.5×106 cells and Treatment group 2 (T2) with 3×106 cells. IL-6 expressions were determined using western blot analysis, whereas the leukocyte infiltrations were assessed using the histopathological examination. All variables were evaluated on day 3 and 7. Results Leukocyte infiltration is significantly lower in T1 and T2 compared to control group in day 3 and 7 (P<0.05), while there were no differences between the two treatment groups. The expression of IL-6 was found to be significantly lower in the T1 and T2 groups compared to the control group on days 3 and 7 (P<0.05), with no significant differences observed between the two treatment groups. Conclusion MSCs administration in rats with intestinal perforation reduced inflammation by controlling leukocyte infiltration and IL-6 expression.
Collapse
Affiliation(s)
- Eko Setiawan
- Department of Surgery, Faculty of Medicine, Universitas Islam Sultan Agung, Semarang
| | - Agung Putra
- Stem Cell and Cancer Research (SCCR) Laboratory, Faculty of Medicine, Universitas Islam Sultan Agung, Semarang
- Department of Pathological Anatomy, Faculty of Medicine, Universitas Islam Sultan Agung, Semarang
- Department of Postgraduate Biomedical Science, Faculty of Medicine, Universitas Islam Sultan Agung, Semarang
| | - Dimas Irfan Nabih
- Faculty of Medicine, Universitas Islam Sultan Agung, Semarang, Indonesia
| | | | - Rheza Rizaldy
- Faculty of Medicine, Universitas Islam Sultan Agung, Semarang, Indonesia
| |
Collapse
|
8
|
Summer M, Ali S, Fiaz U, Hussain T, Khan RRM, Fiaz H. Revealing the molecular mechanisms in wound healing and the effects of different physiological factors including diabetes, age, and stress. J Mol Histol 2024; 55:637-654. [PMID: 39120834 DOI: 10.1007/s10735-024-10223-3] [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: 02/29/2024] [Accepted: 07/05/2024] [Indexed: 08/10/2024]
Abstract
Wounds are the common fates in various microbial infections and physical damages including accidents, surgery, and burns. In response, a healthy body with a potent immune system heals that particular site within optimal time by following the coagulation, inflammation, proliferation, and remodeling phenomenon. However, certain malfunctions in the body due to various diseases particularly diabetes and other physiological factors like age, stress, etc., prolong the process of wound healing through various mechanisms including the Akt, Polyol, and Hexosamine pathways. The current review thoroughly explains the wound types, normal wound healing mechanisms, and the immune system's role. Moreover, the mechanistic role of diabetes is also elaborated comprehensively.
Collapse
Affiliation(s)
- Muhammad Summer
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, GC University Lahore, Lahore, 54000, Pakistan.
| | - Shaukat Ali
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, GC University Lahore, Lahore, 54000, Pakistan.
| | - Umaima Fiaz
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, GC University Lahore, Lahore, 54000, Pakistan
| | - Tauqeer Hussain
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, GC University Lahore, Lahore, 54000, Pakistan
| | | | - Hashim Fiaz
- Ammer-ud-Din Medical College, Lahore, 54000, Pakistan
| |
Collapse
|
9
|
Póvoa da Costa F, de Sarges KML, da Silva R, dos Santos EF, do Nascimento MH, Rodrigues AM, Cantanhede MHD, Rodrigues FBB, Viana MDNDSDA, Leite MDM, de Oliveira CF, Neves PFMD, Pereira Neto GDS, de Brito MTFM, da Silva ALS, Henriques DF, Quaresma JAS, Falcão LFM, Queiroz MAF, Vallinoto IMVC, Vallinoto ACR, Viana GMR, dos Santos EJM. Genetic, Clinical, Epidemiological, and Immunological Profiling of IgG Response Duration after SARS-CoV-2 Infection. Int J Mol Sci 2024; 25:8740. [PMID: 39201427 PMCID: PMC11354850 DOI: 10.3390/ijms25168740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
The IgG response against SARS-CoV-2 infection can persist for over six months (long response; LR). However, among 30% of those infected, the duration can be as short as three months or less (short response; SR). The present study assembled serological data on the anti-SARS-CoV-2 IgG response duration of two previous studies and integrated these results with the plasmatic cytokine levels and genetic profile of 10 immune-relevant SNPs that were also previously published, along with the plasmatic total IgG, IgA, and IgM levels, allowing for the genetic, clinical, immunological, and epidemiological aspects of the post-COVID-19 IgG response duration to be understood. The SR was associated with previous mild acute COVID-19 and with an SNP (rs2228145) in IL6R related to low gene expression. Additionally, among the SR subgroup, no statistically significant Spearman correlations were observed between the plasma levels of IL-17A and the Th17 regulatory cytokines IFN-γ (rs = 0.2399; p = 0.1043), IL-4 (rs = 0.0273; p = 0.8554), and IL-2 (rs = 0.2204; p = 0.1365), while among the LR subgroup, weaker but statistically significant Spearman correlations were observed between the plasma levels of IL-17A and IFN-γ (rs = 0.3873; p = 0.0016), IL-4 (rs = 0.2671; p = 0.0328), and IL-2 (rs = 0.3959; p = 0.0012). These results suggest that the Th17 response mediated by the IL-6 pathway has a role in the prolonged IgG response to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Flávia Póvoa da Costa
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Kevin Matheus Lima de Sarges
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Rosilene da Silva
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Erika Ferreira dos Santos
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Matheus Holanda do Nascimento
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
| | - Alice Maciel Rodrigues
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
| | - Marcos Henrique Damasceno Cantanhede
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Fabíola Brasil Barbosa Rodrigues
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Maria de Nazaré do Socorro de Almeida Viana
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Mauro de Meira Leite
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Camille Ferreira de Oliveira
- Section of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Health Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 67000-000, Pará, Brazil; (C.F.d.O.); (D.F.H.)
| | - Pablo Fabiano Moura das Neves
- Center of Biological and Health Sciences, State University of Pará, Belém 66050-540, Pará, Brazil; (P.F.M.d.N.); (J.A.S.Q.); (L.F.M.F.)
| | - Gabriel dos Santos Pereira Neto
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil
| | - Mioni Thieli Figueiredo Magalhães de Brito
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
| | - Andréa Luciana Soares da Silva
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
| | - Daniele Freitas Henriques
- Section of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Health Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 67000-000, Pará, Brazil; (C.F.d.O.); (D.F.H.)
| | - Juarez Antônio Simões Quaresma
- Center of Biological and Health Sciences, State University of Pará, Belém 66050-540, Pará, Brazil; (P.F.M.d.N.); (J.A.S.Q.); (L.F.M.F.)
| | - Luiz Fábio Magno Falcão
- Center of Biological and Health Sciences, State University of Pará, Belém 66050-540, Pará, Brazil; (P.F.M.d.N.); (J.A.S.Q.); (L.F.M.F.)
| | - Maria Alice Freitas Queiroz
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil
| | - Izaura Maria Vieira Cayres Vallinoto
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil
| | - Antonio Carlos Rosário Vallinoto
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil
| | - Giselle Maria Rachid Viana
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
- Malaria Basic Research Laboratory, Parasitology Section, Evandro Chagas Institute, Health Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 67000-000, Pará, Brazil
| | - Eduardo José Melo dos Santos
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| |
Collapse
|
10
|
Gunasekara N, Clauss D, Bloch W. Effects of Exercise-Induced Changes in Myokine Expression on the Tumor Microenvironment. Sports Med Int Open 2024; 8:a22831663. [PMID: 38933599 PMCID: PMC11204211 DOI: 10.1055/a-2283-1663] [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: 10/10/2023] [Accepted: 04/26/2024] [Indexed: 06/28/2024] Open
Abstract
In this narrative review, we summarize the direct and indirect effects that myokines have on the tumor microenvironment. We took studies of various cancer types and species into account. Systematic reviews and meta-analyses that matched the search terms were also considered. We searched databases for six months. As a narrative approach was chosen, no data was analyzed or reanalyzed. The goal of this narrative review is to create an overview on the topic to identify research gaps and answer the questions as to whether myokine expression may be relevant in cancer research in regard to the tumor microenvironment. Six commonly known myokines were chosen. We found strong links between the influence exercise has on interleukin-6, oncostatin M, secreted protein acidic and rich in cysteine, and irisin in the context of tumor progression and inhibition via interactions with the tumor microenvironment. It became clear that the effects of myokines on the tumor microenvironment can vary and contribute to disease progression or regression. Interactions among myokines and immune cells must also be considered and require further investigation. To date, no study has shown a clear connection, while multiple studies suggest further investigation of the topic, similar to the effects of exercise on myokine expression.
Collapse
Affiliation(s)
- Nadira Gunasekara
- Institute of Cardiology and Sports Medicine, German Sport University
Cologne, Cologne, Germany
| | - Dorothea Clauss
- Institute of Cardiology and Sports Medicine, German Sport University
Cologne, Cologne, Germany
| | - Wilhelm Bloch
- Institute of Cardiology and Sports Medicine, German Sport University
Cologne, Cologne, Germany
| |
Collapse
|
11
|
Ragab AAY, Doyle MF, Chen J, Fang Y, Lunetta KL, Murabito JM. Immune cell phenotypes and mortality in the Framingham Heart Study. Immun Ageing 2024; 21:37. [PMID: 38867269 PMCID: PMC11167945 DOI: 10.1186/s12979-024-00431-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/23/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Global life expectancy is rising, with the 60 + age group projected to hit 2 billion by 2050. Aging impacts the immune system. A notable marker of immune system aging is the presence of Aging-Related Immune Cell Phenotypes (ARIPs). Despite their importance, links between immune cell phenotypes including ARIPs and mortality are underexplored. We prospectively investigated 16 different immune cell phenotypes using flow cytometry and IL-6 in relation to survival outcome among dementia-free Framingham Heart Study (FHS) offspring cohort participants who attended the seventh exam (1998-2001). RESULTS Among 996 participants (mean age 62 years, range 40 to 88 years, 52% female), the 19-year survival rate was 65%. Adjusting for age, sex, and cytomegalovirus (CMV) serostatus, higher CD4/CD8 and Tc17/CD8 + Treg ratios were significantly associated with lower all-cause mortality (HR: 0.86 [0.76-0.96], 0.84 [0.74-0.94], respectively), while higher CD8 regulatory cell levels (CD8 + CD25 + FoxP3 +) were associated with increased all-cause mortality risk (HR = 1.17, [1.03-1.32]). Elevated IL-6 levels correlated with higher all-cause, cardiovascular, and non-cardiovascular mortality (HR = 1.43 [1.26-1.62], 1.70 [1.31-2.21], and 1.36 [1.18-1.57], respectively). However, after adjusting for cardiovascular risk factors and prevalent cancer alongside age, sex, and CMV, immune cell phenotypes were no longer associated with mortality in our cohort. Nonetheless, IL-6 remained significantly associated with all-cause and cardiovascular mortality (HRs: 1.3 [1.13-1.49], 1.5 [1.12-1.99], respectively). CONCLUSIONS In 19-year follow-up, higher Tc17/CD8 + Treg and CD4/CD8 ratios were associated with lower all-cause mortality, while the CD8 + CD25 + FoxP3 + (CD8 + Treg) phenotype showed increased risk. Elevated IL-6 levels consistently correlated with amplified mortality risks. These findings highlight the links between immune phenotypes and mortality, suggesting implications for future research and clinical considerations.
Collapse
Affiliation(s)
- Ahmed A Y Ragab
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA.
| | - Margaret F Doyle
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, VT, USA
| | - Jiachen Chen
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Yuan Fang
- Binghamton University, State University of New York, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - Kathryn L Lunetta
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Joanne M Murabito
- Framingham Heart Study, National Heart, Lung, and Blood Institute and Boston University Chobanian & Avedisian School of Medicine, Framingham, MA, USA.
- Department of Medicine, Section of General Internal Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA.
| |
Collapse
|
12
|
Islam R, Heyer J, Figura M, Wang X, Nie X, Nathaniel B, Indumathy S, Hartmann K, Pleuger C, Fijak M, Kliesch S, Dittmar F, Pilatz A, Wagenlehner F, Hedger M, Loveland B, Hotaling JH, Guo J, Loveland KL, Schuppe HC, Fietz D. T cells in testicular germ cell tumors: new evidence of fundamental contributions by rare subsets. Br J Cancer 2024; 130:1893-1903. [PMID: 38649788 PMCID: PMC11183042 DOI: 10.1038/s41416-024-02669-9] [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: 10/03/2023] [Accepted: 03/21/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Immune cell infiltration is heterogeneous but common in testicular germ cell tumors (TGCT) and pre-invasive germ cell neoplasia in situ (GCNIS). Tumor-infiltrating T cells including regulatory T (Treg) and follicular helper T (Tfh) cells are found in other cancer entities, but their contributions to TGCT are unknown. METHODS Human testis specimens from independent patient cohorts were analyzed using immunohistochemistry, flow cytometry and single-cell RNA sequencing (scRNA-seq) with special emphasis on delineating T cell subtypes. RESULTS Profound changes in immune cell composition within TGCT, shifting from macrophages in normal testes to T cells plus B and dendritic cells in TGCT, were documented. In most samples (96%), the CD4+ T cell frequency exceeded that of CD8+ cells, with decreasing numbers from central to peripheral tumor areas, and to tumor-free, contralateral testes. T cells including Treg and Tfh were most abundant in seminoma compared to mixed tumors and embryonal carcinoma. CONCLUSION Despite considerable heterogeneity between patients, T cell subtypes form a key part of the TGCT microenvironment. The novel finding of rare Treg and Tfh cells in human testis suggests their involvement in TGCT pathobiology, with implications for understanding tumor progression, to assess patients' prognosis, and as putative targets for personalized immunotherapy.
Collapse
Affiliation(s)
- Rashidul Islam
- Dept. of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
- Department of Developmental Pathology, Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Jannis Heyer
- Dept. of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
- Dept. of Urology, Pediatric Urology and Andrology, Justus Liebig University, Giessen, Germany
| | - Miriam Figura
- Dept. of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
- Dept. of Urology, Pediatric Urology and Andrology, Justus Liebig University, Giessen, Germany
| | - Xiaoyan Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute of Stem Cell and Regenerative Medicine, Beijing, China
| | - Xichen Nie
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Benedict Nathaniel
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Sivanjah Indumathy
- Dept. of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Katja Hartmann
- Dept. of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
| | - Christiane Pleuger
- Hessian Centre of Reproductive Medicine, Justus-Liebig-University, Giessen, Germany
- Institute of Anatomy and Cell Biology, Justus Liebig University, Giessen, Germany
| | - Monika Fijak
- Hessian Centre of Reproductive Medicine, Justus-Liebig-University, Giessen, Germany
- Institute of Anatomy and Cell Biology, Justus Liebig University, Giessen, Germany
| | - Sabine Kliesch
- Centre of Reproductive Medicine and Andrology, University of Muenster, Muenster, Germany
| | - Florian Dittmar
- Dept. of Urology, Pediatric Urology and Andrology, Justus Liebig University, Giessen, Germany
| | - Adrian Pilatz
- Dept. of Urology, Pediatric Urology and Andrology, Justus Liebig University, Giessen, Germany
- Hessian Centre of Reproductive Medicine, Justus-Liebig-University, Giessen, Germany
| | - Florian Wagenlehner
- Dept. of Urology, Pediatric Urology and Andrology, Justus Liebig University, Giessen, Germany
- Hessian Centre of Reproductive Medicine, Justus-Liebig-University, Giessen, Germany
| | - Mark Hedger
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | | | - James H Hotaling
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Jingtao Guo
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute of Stem Cell and Regenerative Medicine, Beijing, China
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Kate L Loveland
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Hans-Christian Schuppe
- Dept. of Urology, Pediatric Urology and Andrology, Justus Liebig University, Giessen, Germany
- Hessian Centre of Reproductive Medicine, Justus-Liebig-University, Giessen, Germany
| | - Daniela Fietz
- Dept. of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany.
- Hessian Centre of Reproductive Medicine, Justus-Liebig-University, Giessen, Germany.
| |
Collapse
|
13
|
Baskerville R, Castell L, Bermon S. Sports and Immunity, from the recreational to the elite athlete. Infect Dis Now 2024; 54:104893. [PMID: 38531477 DOI: 10.1016/j.idnow.2024.104893] [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: 03/12/2024] [Accepted: 03/22/2024] [Indexed: 03/28/2024]
Abstract
The pivotal role of the immune system in physical activity is well-established. While interactions are complex, they tend to constitute discrete immune responses. Moderate intensity exercise causes leukocytosis with a mild anti-inflammatory cytokine profile and immunoenhancement. Above a threshold of intensity, lactate-mediated IL-6 release causes a proinflammatory state followed by a depressed inflammatory state, which stimulates immune adaptation and longer term cardiometabolic enhancement. Exercise-related immune responses are modulated by sex, age and immunonutrition. At all ability levels, these factors collectively affect the immune balance between enhancement or overload and dysfunction. Excessive training, mental stress or insufficient recovery risks immune cell exhaustion and hypothalamic pituitary axis (HPA) stress responses causing immunodepression with negative impacts on performance or general health. Participation in sport provides additional immune benefits in terms of ensuring regularity, social inclusion, mental well-being and healthier life choices in terms of diet and reduced smoking and alcohol, thereby consolidating healthy lifestyles and longer term health. Significant differences exist between recreational and professional athletes in terms of inherent characteristics, training resilience and additional stresses arising from competition schedules, travel-related infections and stress. Exercise immunology examines the central role of immunity in exercise physiology and straddles multiple disciplines ranging from neuroendocrinology to nutrition and genetics, with the aim of guiding athletes to train optimally and safely. This review provides a brief outline of the main interactions of immunity and exercise, some influencing factors, and current guidance on maintaining immune health.
Collapse
Affiliation(s)
| | - Linda Castell
- Green Templeton College, University of Oxford, Oxford, UK
| | - Stéphane Bermon
- World Athletics Health and Science Department, Monaco and LAMHESS, University Côte d'Azur, Nice, France
| |
Collapse
|
14
|
Zhang Y, Zhao J, Jin Q, Zhuang L. Transcriptomic Analyses and Experimental Validation Identified Immune-Related lncRNA-mRNA Pair MIR210HG- BPIFC Regulating the Progression of Hypertrophic Cardiomyopathy. Int J Mol Sci 2024; 25:2816. [PMID: 38474063 DOI: 10.3390/ijms25052816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/19/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a disease in which the myocardium of the heart becomes asymmetrically thickened, malformed, disordered, and loses its normal structure and function. Recent studies have demonstrated the significant involvement of inflammatory responses in HCM. However, the precise role of immune-related long non-coding RNAs (lncRNAs) in the pathogenesis of HCM remains unclear. In this study, we performed a comprehensive analysis of immune-related lncRNAs in HCM. First, transcriptomic RNA-Seq data from both HCM patients and healthy individuals (GSE180313) were reanalyzed thoroughly. Key HCM-related modules were identified using weighted gene co-expression network analysis (WGCNA). A screening for immune-related lncRNAs was conducted within the key modules using immune-related mRNA co-expression analysis. Based on lncRNA-mRNA pairs that exhibit shared regulatory microRNAs (miRNAs), we constructed a competing endogenous RNA (ceRNA) network, comprising 9 lncRNAs and 17 mRNAs that were significantly correlated. Among the 26 lncRNA-mRNA pairs, only the MIR210HG-BPIFC pair was verified by another HCM dataset (GSE130036) and the isoprenaline (ISO)-induced HCM cell model. Furthermore, knockdown of MIR210HG increased the regulatory miRNAs and decreased the mRNA expression of BPIFC correspondingly in AC16 cells. Additionally, the analysis of immune cell infiltration indicated that the MIR210HG-BPIFC pair was potentially involved in the infiltration of naïve CD4+ T cells and CD8+ T cells. Together, our findings indicate that the decreased expression of the lncRNA-mRNA pair MIR210HG-BPIFC was significantly correlated with the pathogenesis of the disease and may be involved in the immune cell infiltration in the mechanism of HCM.
Collapse
Affiliation(s)
- Yuan Zhang
- Institute of Genetics and Reproduction, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Jiuxiao Zhao
- Institute of Genetics and Reproduction, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiao Jin
- Institute of Genetics and Reproduction, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lenan Zhuang
- Institute of Genetics and Reproduction, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou 310016, China
| |
Collapse
|
15
|
Ge H, Yang Q, Lyu S, Du Z, Liu X, Shang X, Xu M, Liu J, Zhang T. Egg White Peptides Accelerating the Wound Healing Process Through Modulating the PI3K-AKT Pathway: A Joint Analysis of Transcriptomic and Proteomic. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:4100-4115. [PMID: 38373195 DOI: 10.1021/acs.jafc.3c08466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
Wound healing is a multiphase process with a complex repair mechanism; trauma-repairing products with safety and high efficiency have a great market demand. Egg white peptides (EWP) have various physiological regulatory functions and have been proven efficient in ameliorating skin damage. However, their underlying regulation mechanism has not been revealed. This study further evaluated the EWP ameliorating mechanism by conducting a full-thickness skin wound model. Results demonstrated that EWP administration significantly inhibited the expression of pro-inflammatory and shortened the inflammatory phase. Besides, EWP can accelerate the secretion of growth factors (PDGF, VEGF, and TGF-β1) in skin tissue, significantly increasing the regeneration of granulation tissue and endothelium in the proliferation phase, thereby promoting wound healing. After 400 mg/kg EWP interventions for 13 days postoperation, the wound healing rate reached 90%. The combination of transcriptomic and proteomic analyses demonstrated the ameliorating efficiency effects of EWP on wound healing. EWP mainly participates in the functional network with the PI3K-AKT signaling pathway as the core to accelerate wound healing. These findings suggest a promising EWP-based strategy for accelerating wound healing.
Collapse
Affiliation(s)
- Huifang Ge
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei 230036, P. R. China
| | - Qi Yang
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, P. R. China
| | - Siwen Lyu
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, P. R. China
| | - Zhiyang Du
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, P. R. China
| | - Xuanting Liu
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, P. R. China
| | - Xiaomin Shang
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, P. R. China
| | - Menglei Xu
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, P. R. China
| | - Jingbo Liu
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, P. R. China
| | - Ting Zhang
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun 130062, P. R. China
| |
Collapse
|
16
|
Catarino JDS, de Oliveira RF, Silva MV, Sales-Campos H, de Vito FB, da Silva DAA, Naves LL, Oliveira CJF, Rodrigues DBR, Rodrigues V. Genetic variation of FcγRIIa induces higher uptake of Leishmania infantum and modulates cytokine production by adherent mononuclear cells in vitro. Front Immunol 2024; 15:1343602. [PMID: 38455048 PMCID: PMC10917923 DOI: 10.3389/fimmu.2024.1343602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/01/2024] [Indexed: 03/09/2024] Open
Abstract
Introduction Single nucleotide variations (SNVs) are specific genetic variations that commonly occur in a population and often do not manifest phenotypically. However, depending on their location and the type of nucleotide exchanged, an SNV can alter or inhibit the function of the gene in which it occurs. Immunoglobulin G (IgG) receptor genes have exhibited several polymorphisms, including rs1801274, which is found in the FcgRIIa gene. The replacement of A with T results in a Histidine (H) to Arginine (R) substitution, altering the affinity of the IgG receptor for IgG subtypes and C-reactive protein (CRP). In this study, we analyzed rs1801274 and its functional implications concerning L. Infantum uptake and cytokine production. Methods We genotyped 201 individuals from an endemic area for visceral leishmaniasis to assess the presence of rs1801274 using Taqman probes for a candidate gene study. Additionally, we included seventy individuals from a non-endemic area for a functional study. Subsequently, we isolated and cultivated one-week adherent mononuclear cells (AMCs) derived from the peripheral blood of participants residing in the non-endemic region in the presence of L. infantum promastigotes, with and without antigen-specific IgG and/or CRP. We analyzed the rate of phagocytosis and the production of nitric oxide (NO), tumor necrosis factor (TNF)-a, interleukin (IL)-10, IL-12 p70, IL-1b, IL- 6, and IL-8 in the culture supernatants. Results and discussion In participants from the endemic region, the A/G (H/R isoform) heterozygous genotype was significantly associated with susceptibility to the disease. Furthermore, SNVs induced a change in the phagocytosis rate in an opsonin-dependent manner. Opsonization with IgG increased the production of IL-10, TNF-a, and IL-6 in AMCs with the H/R isoform, followed by a decrease in NO production. The results presented here suggest that the rs1801274 polymorphism is linked to a higher susceptibility to visceral leishmaniasis.
Collapse
Affiliation(s)
- Jonatas da Silva Catarino
- Laboratory of Immunology, Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Rafael Faria de Oliveira
- Laboratory of Immunology, Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Marcos Vinicius Silva
- Laboratory of Immunology, Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Helioswilton Sales-Campos
- Laboratory of Immunology, Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
- Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, GO, Brazil
| | - Fernanda Bernadelli de Vito
- Laboratory of Immunology, Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Djalma Alexandre Alves da Silva
- Laboratory of Immunology, Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Lucila Langoni Naves
- Laboratory of Immunology, Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Carlo José Freire Oliveira
- Laboratory of Immunology, Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Denise Bertulucci Rocha Rodrigues
- Laboratory of Immunology, Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Virmondes Rodrigues
- Laboratory of Immunology, Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
- National Institute of Neuroimmuno Modulation, Rio de Janeiro, Brazil
| |
Collapse
|
17
|
Hu Y, Dong Z, Liu K. Unraveling the complexity of STAT3 in cancer: molecular understanding and drug discovery. J Exp Clin Cancer Res 2024; 43:23. [PMID: 38245798 PMCID: PMC10799433 DOI: 10.1186/s13046-024-02949-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/08/2024] [Indexed: 01/22/2024] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a transcriptional factor involved in almost all cancer hallmark features including tumor proliferation, metastasis, angiogenesis, immunosuppression, tumor inflammation, metabolism reprogramming, drug resistance, cancer stemness. Therefore, STAT3 has become a promising therapeutic target in a wide range of cancers. This review focuses on the up-to-date knowledge of STAT3 signaling in cancer. We summarize both the positive and negative modulators of STAT3 together with the cancer hallmarks involving activities regulated by STAT3 and highlight its extremely sophisticated regulation on immunosuppression in tumor microenvironment and metabolic reprogramming. Direct and indirect inhibitors of STAT3 in preclinical and clinical studies also have been summarized and discussed. Additionally, we highlight and propose new strategies of targeting STAT3 and STAT3-based combinations with established chemotherapy, targeted therapy, immunotherapy and combination therapy. These efforts may provide new perspectives for STAT3-based target therapy in cancer.
Collapse
Affiliation(s)
- Yamei Hu
- Tianjian Laboratory for Advanced Biomedical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
- Medical Research Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zigang Dong
- Tianjian Laboratory for Advanced Biomedical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China.
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China.
| | - Kangdong Liu
- Tianjian Laboratory for Advanced Biomedical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China.
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan, China.
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, China.
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China.
| |
Collapse
|
18
|
Ragab AAY, Doyle MF, Chen J, Fang Y, Lunetta KL, Murabito JM. Aging-Related Immune Cell Phenotypes and Mortality in the Framingham Heart Study. RESEARCH SQUARE 2023:rs.3.rs-3773986. [PMID: 38234796 PMCID: PMC10793514 DOI: 10.21203/rs.3.rs-3773986/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Background The global increase in human life expectancy is evident. The total number of individuals aged 60 or above is anticipated to reach 2 billion by 2050. Aging, an inherently complex process, manifests prominently in the changes observed in the immune system. A notable marker of immune system aging is the presence of Aging-Related Immune Cell Phenotypes (ARIPs). Despite their significance, the connections between various ARIPs and mortality have not been thoroughly investigated. We prospectively investigated 16 different ARIPs using flow cytometry, namely, CD4/CD8 ratio, Granzyme B + CD8/Granyzme B + CD4, TN/TM = Tn / (Teff + Tem + Tcm) for TN/TM CD4 + and TN/TM CD8 + ratios, Th17/CD4 + Treg, Tc17/CD8 + Treg, Th17, Tc17, CD4 + Temra, CD8 + Temra, CD4 + CD25 + FoxP3+ (CD4 + Treg), CD8 + CD25 + FoxP3+ (CD8 + Treg) CD4 + CD27-, CD4 + CD28-CD27-, CD8 + CD27-, CD8 + CD28-CD27- and IL-6 in relation to survival outcome among dementia-free Framingham Heart Study (FHS) offspring cohort participants who attended the seventh exam (1998-2001). Results Among 996 participants (mean age 62 years, range 40 to 88 years, 52% female), the survival rate was 65% during 19 years of follow-up. For the model adjusting for age, sex, and cytomegalovirus (CMV) serostatus, higher CD4/CD8 and Tc17/CD8 + Treg ratios were significantly associated with lower all-cause mortality (HR:0.86 [0.76-0.96], 0.84 [0.74-0.94], respectively) and higher CD8 regulatory cell levels (CD8 + CD25 + FoxP3+) were associated with higher all-cause mortality (HR = 1.17, [1.03-1.32]). Higher IL-6 levels were associated with higher all-cause, cardiovascular, and non-cardiovascular mortality (HR = 1.43 [1.26-1.62], 1.70 [1.31-2.21], and 1.36 [1.18-1.57], respectively).
Collapse
Affiliation(s)
| | | | | | - Yuan Fang
- Binghamton University, State University of New York
| | | | | |
Collapse
|
19
|
Grebenciucova E, VanHaerents S. Interleukin 6: at the interface of human health and disease. Front Immunol 2023; 14:1255533. [PMID: 37841263 PMCID: PMC10569068 DOI: 10.3389/fimmu.2023.1255533] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 09/11/2023] [Indexed: 10/17/2023] Open
Abstract
Interleukin 6 (IL-6) is a pleiotropic cytokine executing a diverse number of functions, ranging from its effects on acute phase reactant pathways, B and T lymphocytes, blood brain barrier permeability, synovial inflammation, hematopoiesis, and embryonic development. This cytokine empowers the transition between innate and adaptive immune responses and helps recruit macrophages and lymphocytes to the sites of injury or infection. Given that IL-6 is involved both in the immune homeostasis and pathogenesis of several autoimmune diseases, research into therapeutic modulation of IL-6 axis resulted in the approval of a number of effective treatments for several autoimmune disorders like neuromyelitis optica spectrum disorder (NMOSD), rheumatoid arthritis, juvenile idiopathic arthritis, polyarticular juvenile idiopathic arthritis, giant cell arteritis (GCA), and cytokine release syndrome, associated with SARS-CoV2 pneumonia. This review discusses downstream inflammatory pathways of IL-6 expression and therapeutic applications of IL-6 blockade, currently investigated for the treatment of several other autoimmune conditions such as autoimmune encephalitis, autoimmune epilepsy, as well as myelin oligodendrocyte glycoprotein associated demyelination (MOGAD). This review further highlights the need for clinical trials to evaluate IL-6 blockade in disorders such neuropsychiatric lupus erythematosus (SLE), sarcoidosis and Behcet's.
Collapse
Affiliation(s)
- Elena Grebenciucova
- Feinberg School of Medicine, Department of Neurology, Northwestern University, Chicago, IL, United States
| | | |
Collapse
|
20
|
Nesovic LD, Roach CJ, Joshi G, Gill HS. Delivery of gold nanoparticle-conjugated M2e influenza vaccine in mice using coated microneedles. Biomater Sci 2023; 11:5859-5871. [PMID: 37455612 DOI: 10.1039/d3bm00305a] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
As a prospective influenza vaccination platform, a microneedle patch offers advantages such as self-administration and reduction of needle-phobia-associated vaccination avoidance. In an effort to design a broadly protective influenza vaccine we have previously developed a vaccine formulation containing the highly conserved ectodomain sequence of the M2 influenza protein (M2e) attached to the surface of gold nanoparticles (AuNPs) with CpG as a soluble adjuvant (AuNP-M2e + sCpG). Our previous studies have used the intranasal route for vaccination and demonstrated broad protection from this vaccine. Here we asked the question whether the same formulation can be effective when administered to mice using microneedles. We demonstrate that the microneedles can be coated with AuNP-M2e + sCpG formulation, and the AuNPs from the coating can be readily resuspended without aggregation. The AuNPs were delivered with high efficiency into murine skin, and the AuNPs cleared the skin within 12 h of microneedle treatment. After vaccination, strong M2e-specific humoral and cellular responses were stimulated, and the vaccinated mice were 100% protected following a lethal challenge with influenza A/PR/8/34 (H1N1).
Collapse
Affiliation(s)
- Lazar D Nesovic
- Department of Chemical Engineering Texas Tech University, 8th and Canton, Lubbock, Texas 79409, USA.
| | - Carsen J Roach
- Department of Chemical Engineering Texas Tech University, 8th and Canton, Lubbock, Texas 79409, USA.
| | - Gaurav Joshi
- Department of Chemical Engineering Texas Tech University, 8th and Canton, Lubbock, Texas 79409, USA.
| | - Harvinder Singh Gill
- Department of Chemical Engineering Texas Tech University, 8th and Canton, Lubbock, Texas 79409, USA.
| |
Collapse
|
21
|
Chen M, Venturi V, Munier CML. Dissecting the Protective Effect of CD8 + T Cells in Response to SARS-CoV-2 mRNA Vaccination and the Potential Link with Lymph Node CD8 + T Cells. BIOLOGY 2023; 12:1035. [PMID: 37508464 PMCID: PMC10376827 DOI: 10.3390/biology12071035] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/04/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023]
Abstract
SARS-CoV-2 vaccines have played a crucial role in effectively reducing COVID-19 disease severity, with a new generation of vaccines that use messenger RNA (mRNA) technology being administered globally. Neutralizing antibodies have featured as the heroes of vaccine-induced immunity. However, vaccine-elicited CD8+ T cells may have a significant impact on the early protective effects of the mRNA vaccine, which are evident 12 days after initial vaccination. Vaccine-induced CD8+ T cells have been shown to respond to multiple epitopes of SARS-CoV-2 and exhibit polyfunctionality in the periphery at the early stage, even when neutralizing antibodies are scarce. Furthermore, SARS-CoV-2 mRNA vaccines induce diverse subsets of memory CD8+ T cells that persist for more than six months following vaccination. However, the protective role of CD8+ T cells in response to the SARS-CoV-2 mRNA vaccines remains a topic of debate. In addition, our understanding of CD8+ T cells in response to vaccination in the lymph nodes, where they first encounter antigen, is still limited. This review delves into the current knowledge regarding the protective role of polyfunctional CD8+ T cells in controlling the virus, the response to SARS-CoV-2 mRNA vaccines, and the contribution to supporting B cell activity and promoting immune protection in the lymph nodes.
Collapse
Affiliation(s)
- Mengfei Chen
- The Kirby Institute, UNSW, Sydney, NSW 2052, Australia
| | | | | |
Collapse
|
22
|
Al-Rasheed M, Ball C, Parthiban S, Ganapathy K. Evaluation of protection and immunity induced by infectious bronchitis vaccines administered by oculonasal, spray or gel routes in commercial broiler chicks. Vaccine 2023:S0264-410X(23)00642-4. [PMID: 37316407 DOI: 10.1016/j.vaccine.2023.05.073] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/16/2023]
Abstract
Broiler chicks' responses following combined IBV live attenuated Massachusetts and 793B strains through gel, spray or oculonasal (ON) vaccination routes were cross-compared. Subsequently, the responses following IBV M41 challenge of the unvaccinated and vaccinated groups were also assessed. Post-vaccination humoral and mucosal immune responses, alongside viral load kinetics in swabs and tissues, were determined using commercial ELISA assays, monoclonal antibody-based IgG and IgA ELISA assays and qRT-PCR respectively. After challenged with IBV-M41 strain, humoral and mucosal immune responses, ciliary protection, viral load kinetics, and immune gene mRNA transcriptions between the three vaccination methods were examined and compared. Findings showed that post-vaccinal humoral and mucosal immune responses were similar in all three vaccination methods. Post vaccinal viral load kinetics is influenced by method of administration. The viral load peaked in the ON group within the tissues and the OP/CL swabs in the first and third weeks respectively. Following M41 challenge, ciliary protection and mucosal immune responses were not influenced by vaccination methods as all three methods offered equal ciliary protection. Immune gene mRNA transcriptions varied by vaccination methods. Significant up-regulation of MDA5, TLR3, IL-6, IFN-α and IFN-β genes were recorded for ON method. For both spray and gel methods, significant up-regulation of only MDA5 and IL-6 genes were noted. The spray and gel-based vaccination methods gave equivalent levels of ciliary protection and mucosal immunity to M41 virulent challenge comparable to those provided by the ON vaccination. Analysis of viral load and patterns of immune gene transcription of the vaccinated-challenged groups revealed high similarity between turbinate and choanal cleft tissues compared to HG and trachea. With regards to immune gene mRNA transcription, for all the vaccinated-challenged groups, similar results were found except for IFN-α, IFN-β and TLR3, which were up-regulated only in ON compared to gel and spray vaccination methods.
Collapse
Affiliation(s)
- Mohammed Al-Rasheed
- Institute of Infection and Global Health, University of Liverpool, Cheshire, UK; College of Veterinary Medicine, King Faisal University, Al-Ahsa, Saudi Arabia; Avian Research Center, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Christopher Ball
- Institute of Infection and Global Health, University of Liverpool, Cheshire, UK
| | - Sivamurthy Parthiban
- Institute of Infection and Global Health, University of Liverpool, Cheshire, UK; Department of Animal Biotechnology, Madras Veterinary College, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | - Kannan Ganapathy
- Institute of Infection and Global Health, University of Liverpool, Cheshire, UK.
| |
Collapse
|
23
|
Bachus H, McLaughlin E, Lewis C, Papillion AM, Benveniste EN, Hill DD, Rosenberg AF, Ballesteros-Tato A, León B. IL-6 prevents Th2 cell polarization by promoting SOCS3-dependent suppression of IL-2 signaling. Cell Mol Immunol 2023; 20:651-665. [PMID: 37046042 PMCID: PMC10229632 DOI: 10.1038/s41423-023-01012-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 03/23/2023] [Indexed: 04/14/2023] Open
Abstract
Defective interleukin-6 (IL-6) signaling has been associated with Th2 bias and elevated IgE levels. However, the underlying mechanism by which IL-6 prevents the development of Th2-driven diseases remains unknown. Using a model of house dust mite (HDM)-induced Th2 cell differentiation and allergic airway inflammation, we showed that IL-6 signaling in allergen-specific T cells was required to prevent Th2 cell differentiation and the subsequent IgE response and allergic inflammation. Th2 cell lineage commitment required strong sustained IL-2 signaling. We found that IL-6 turned off IL-2 signaling during early T-cell activation and thus inhibited Th2 priming. Mechanistically, IL-6-driven inhibition of IL-2 signaling in responding T cells was mediated by upregulation of Suppressor Of Cytokine Signaling 3 (SOCS3). This mechanism could be mimicked by pharmacological Janus Kinase-1 (JAK1) inhibition. Collectively, our results identify an unrecognized mechanism that prevents the development of unwanted Th2 cell responses and associated diseases and outline potential preventive interventions.
Collapse
Affiliation(s)
- Holly Bachus
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Erin McLaughlin
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Crystal Lewis
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Amber M Papillion
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
- Alexion Pharmaceuticals, Inc., New Haven, CT, USA
| | - Etty N Benveniste
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dave Durell Hill
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alexander F Rosenberg
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
- Informatics Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - André Ballesteros-Tato
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Beatriz León
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
24
|
Fraticelli S, Lucioni M, Neri G, Marchiori D, Cristinelli C, Merli M, Monaco R, Borra T, Lazzaro A, Uccella S, Arcaini L, Paulli M. T-Cells Subsets in Castleman Disease: Analysis of 28 Cases Including Unicentric, Multicentric and HHV8-Related Clinical Forms. Int J Mol Sci 2023; 24:ijms24097813. [PMID: 37175521 PMCID: PMC10178230 DOI: 10.3390/ijms24097813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/14/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Castleman disease (CD) is a rare lymphoproliferative disorder that includes various clinico-pathological subtypes. According to clinical course, CD is divided into unicentric CD (UCD) and multicentric CD (MCD). MCD is further distinguished based on the etiological driver in herpes virus-8-related MCD (that can occur in the setting of HIV); in MCD associated with POEMS syndrome (polyneuropathy, organomegaly, endocrinopathy, monoclonal protein, and skin changes); and idiopathic MCD (iMCD). The latter can also be divided in iMCD-TAFRO (thrombocytopenia, anasarca, fever, myelofibrosis, organomegaly) and iMCD not otherwise specified. To date, CD pathogenesis is still uncertain, but CD may represent the histological and clinical result of heterogeneous pathomechanisms. Transcriptome investigations in CD lymph nodes have documented the expression and up-regulation of different cytokines; furthermore, few recent studies have shown alterations of different T-cell subsets in CD patients, suggesting a possible role of the nodal microenvironment in CD development. On this basis, our study aimed to investigate the distribution of T-cell subsets in the clinico-pathological spectrum of CD. We evaluated the CD4/CD8 ratio and the number of T-regulatory (T-reg) FOXP3+ cells in 28 CD cases. In total, 32% of cases showed a decreased CD4/CD8 ratio due to increased CD8+ T-cells, including both UCD, iMCD, and HHV8+ MCD cases. The T-reg subset analysis revealed a statistically significant (p < 0.0001) lower mean number of FOXP3+ T-reg cells in CD cases when compared with non-specific reactive lymph nodes. We did not find statistically significant differences in T-reg numbers between the different CD subtypes. These findings may suggest that alterations in T-cell subpopulations that can lead to disruption of immune system control may contribute to the numerous changes in different cellular compartments that characterize CD.
Collapse
Affiliation(s)
- Sara Fraticelli
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Marco Lucioni
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
- Pathology Unit, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy
| | - Giuseppe Neri
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Deborah Marchiori
- Pathology Unit, Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | | | - Michele Merli
- Division of Hematology, Ospedale di Circolo e Fondazione Macchi, 21100 Varese, Italy
| | - Rodolfo Monaco
- Pathology Unit, Ospedale Guglielmo da Saliceto, 29121 Piacenza, Italy
| | - Tiziana Borra
- Department of Pathology, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy
| | - Antonio Lazzaro
- Division of Hematology and Bone Marrow Transplant Center, Ospedale Guglielmo da Saliceto, 29121 Piacenza, Italy
| | - Silvia Uccella
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
- Pathology Service, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Luca Arcaini
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
- Division of Hematology, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy
| | - Marco Paulli
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
- Pathology Unit, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy
| |
Collapse
|
25
|
Sun Q, Zhao X, Li R, Liu D, Pan B, Xie B, Chi X, Cai D, Wei P, Xu W, Wei K, Zhao Z, Fu Y, Ni L, Dong C. STAT3 regulates CD8+ T cell differentiation and functions in cancer and acute infection. J Exp Med 2023; 220:e20220686. [PMID: 36688918 PMCID: PMC9884582 DOI: 10.1084/jem.20220686] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 11/05/2022] [Accepted: 01/04/2023] [Indexed: 01/24/2023] Open
Abstract
In cancer, persistent antigens drive CD8+ T cell differentiation into exhausted progenitor (Texprog) and terminally exhausted (Texterm) cells. However, how the extrinsic and intrinsic regulatory mechanisms cooperate during this process still remains not well understood. Here, we found that STAT3 signaling plays essential roles in promoting intratumor Texterm cell development by enhancing their effector functions and survival, which results in better tumor control. In tumor microenvironments, STAT3 is predominantly activated by IL-10 and IL-21, but not IL-6. Besides, STAT3 also plays critical roles in the development and function of terminally differentiated effector CD8+ T cells in acute infection. Mechanistically, STAT3 transcriptionally promotes the expression of effector function-related genes, while it suppresses those expressed by the progenitor Tex subset. Moreover, STAT3 functions in collaboration with BATF and IRF4 to mediate chromatin activation at the effector gene loci. Thus, we have elucidated the roles of STAT3 signaling in terminally differentiated CD8+ T cell development, especially in cancer, which benefits the development of more effective immunotherapies against tumors.
Collapse
Affiliation(s)
- Qinli Sun
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Xiaohong Zhao
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Ruifeng Li
- Institute for Immunology, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Dingfeng Liu
- Department of Gynaecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai, China
| | - Birui Pan
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Bowen Xie
- Institute for Immunology, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Xinxin Chi
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Dongli Cai
- Department of Gynaecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai, China
| | - Peng Wei
- Institute for Immunology, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Wei Xu
- Institute for Immunology, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Kun Wei
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Zixuan Zhao
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Yujie Fu
- Institute for Immunology, Tsinghua University, Beijing, China
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai, China
| | - Ling Ni
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Chen Dong
- Institute for Immunology, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai, China
- Research Unit of Immune Regulation and Immune Diseases of Chinese Academy of Medical Sciences, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai, China
| |
Collapse
|
26
|
Zeng Z, Maiti A, Herbrich S, Cai T, Cavazos A, Manzella T, Ma H, Hayes K, Matthews J, DiNardo C, Daver N, Konopleva M. Triple combination targeting methyltransferase, BCL-2, and PD-1 facilitates antileukemia responses in acute myeloid leukemia. Cancer 2023; 129:531-540. [PMID: 36477735 PMCID: PMC11884106 DOI: 10.1002/cncr.34566] [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: 05/31/2022] [Revised: 09/04/2022] [Accepted: 09/23/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND A recent breakthrough therapy combining the BCL-2 inhibitor venetoclax with hypomethylating agents (HMAs) targeting DNA methyltransferase has improved outcomes for patients with acute myeloid leukemia (AML), but the responses and long-term survival in older/unfit patients and in patients with relapsed/refractory AML remain suboptimal. Recent studies showed that inhibition of BCL-2 or DNA methyltransferase modulates AML T-cell immunity. METHODS By using flow cytometry and time-of-flight mass cytometry, the authors examined the effects of the HMA decitabine combined with the BCL-2 inhibitor venetoclax (DAC/VEN therapy) on leukemia cells and T cells in patients with AML who received DAC/VEN therapy in a clinical trial. The authors investigated the response of programmed cell death protein 1 (PD-1) inhibition in the DAC/VEN-treated samples in vitro and investigated the triple combination of PD-1 inhibition with HMA/venetoclax in the trial patients who had AML. RESULTS DAC/VEN therapy effectively targeted leukemia cells and upregulated the expression of the immune checkpoint-inhibitory receptor PD-1 in T cells while preserving CD4-positive and CD8-positive memory T cells in a subset of patients with AML who were tested. In vitro PD-1 inhibition potentiated the antileukemia response in DAC/VEN-treated AML samples. The combined use of azacitidine, venetoclax, and nivolumab eliminated circulating blasts and leukemia stem cells/progenitor cells and expanded the percentage of CD8-positive memory T cells in an illustrative patient with relapsed AML who responded to the regimen in an ongoing clinical trial. CONCLUSIONS Immunomodulation by targeting PD-1 enhances the therapeutic effect of combining an HMA and venetoclax in patients with AML.
Collapse
Affiliation(s)
- Zhihong Zeng
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Abhishek Maiti
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shelley Herbrich
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tianyu Cai
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Antonio Cavazos
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Taylor Manzella
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Helen Ma
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kala Hayes
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jairo Matthews
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Courtney DiNardo
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naval Daver
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marina Konopleva
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
27
|
Bonney EA. A Framework for Understanding Maternal Immunity. Immunol Allergy Clin North Am 2023; 43:e1-e20. [PMID: 37179052 PMCID: PMC10484232 DOI: 10.1016/j.iac.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
This is an alternative and controversial framing of the data relevant to maternal immunity. It argues for a departure from classical theory to view, interrogate and interpret existing data.
Collapse
Affiliation(s)
- Elizabeth A Bonney
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Vermont Robert Larner College of Medicine, Given Building, Room C246, 89 Beaumont Avenue, Burlington, VT 05405, USA.
| |
Collapse
|
28
|
Yang H, Zhang P, Wang Q, Cheng K, Zhao Y. The research development of STAT3 in hepatic ischemia-reperfusion injury. Front Immunol 2023; 14:1066222. [PMID: 36761734 PMCID: PMC9902876 DOI: 10.3389/fimmu.2023.1066222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/10/2023] [Indexed: 01/25/2023] Open
Abstract
Ischemia-reperfusion injury (IRI) is a common complication of surgery, which can cause rapid deterioration of the liver function, increase the risk of graft rejection, and seriously affect the prognosis of patients. The signal transducer and activator of transcription 3 (STAT3) protein has been implicated in pathogenesis of IRI. STAT3 influences the mitochondria through multiple pathways and is also involved in apoptosis and other forms of programmed cell death. STAT3 is associated with Janus kinase (JAK), phosphoinositide-3 kinase (PI3K), and heme oxygenase-1 (HO-1) in liver IRI. The STAT3 pathway plays a dual role in IRI as it can also regulate lipid metabolism which may have potential for treating IRI fatty liver. In this review, we summarize research on the function of STAT3 in liver IRI to provide references for its application in the clinic.
Collapse
Affiliation(s)
| | | | | | | | - Yujun Zhao
- Engineering and Technology Research Center for Transplantation Medicine of National Health Comission, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
29
|
Huseni MA, Wang L, Klementowicz JE, Yuen K, Breart B, Orr C, Liu LF, Li Y, Gupta V, Li C, Rishipathak D, Peng J, Şenbabaoǧlu Y, Modrusan Z, Keerthivasan S, Madireddi S, Chen YJ, Fraser EJ, Leng N, Hamidi H, Koeppen H, Ziai J, Hashimoto K, Fassò M, Williams P, McDermott DF, Rosenberg JE, Powles T, Emens LA, Hegde PS, Mellman I, Turley SJ, Wilson MS, Mariathasan S, Molinero L, Merchant M, West NR. CD8 + T cell-intrinsic IL-6 signaling promotes resistance to anti-PD-L1 immunotherapy. Cell Rep Med 2023; 4:100878. [PMID: 36599350 PMCID: PMC9873827 DOI: 10.1016/j.xcrm.2022.100878] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 10/14/2022] [Accepted: 12/08/2022] [Indexed: 01/05/2023]
Abstract
Although immune checkpoint inhibitors (ICIs) are established as effective cancer therapies, overcoming therapeutic resistance remains a critical challenge. Here we identify interleukin 6 (IL-6) as a correlate of poor response to atezolizumab (anti-PD-L1) in large clinical trials of advanced kidney, breast, and bladder cancers. In pre-clinical models, combined blockade of PD-L1 and the IL-6 receptor (IL6R) causes synergistic regression of large established tumors and substantially improves anti-tumor CD8+ cytotoxic T lymphocyte (CTL) responses compared with anti-PD-L1 alone. Circulating CTLs from cancer patients with high plasma IL-6 display a repressed functional profile based on single-cell RNA sequencing, and IL-6-STAT3 signaling inhibits classical cytotoxic differentiation of CTLs in vitro. In tumor-bearing mice, CTL-specific IL6R deficiency is sufficient to improve anti-PD-L1 activity. Thus, based on both clinical and experimental evidence, agents targeting IL-6 signaling are plausible partners for combination with ICIs in cancer patients.
Collapse
Affiliation(s)
| | - Lifen Wang
- Genentech, South San Francisco, CA 94080, USA
| | | | - Kobe Yuen
- Genentech, South San Francisco, CA 94080, USA
| | | | | | - Li-Fen Liu
- Genentech, South San Francisco, CA 94080, USA
| | - Yijin Li
- Genentech, South San Francisco, CA 94080, USA
| | | | - Congfen Li
- Genentech, South San Francisco, CA 94080, USA
| | | | - Jing Peng
- Genentech, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | - Ning Leng
- Genentech, South San Francisco, CA 94080, USA
| | | | | | - James Ziai
- Genentech, South San Francisco, CA 94080, USA
| | | | | | | | | | - Jonathan E Rosenberg
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Thomas Powles
- Barts Experimental Cancer Medicine Centre, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Leisha A Emens
- University of Pittsburgh Medical Center, Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | | | - Ira Mellman
- Genentech, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Bonifácio LP, Ramacciotti E, Agati LB, Vilar FC, Silva ACTD, Louzada Júnior P, Fonseca BALD, Souza HCCD, Oliveira CCCD, Aguiar VCR, Quadros CADA, Dusilek C, Itinose K, Risson R, Ferreira LRR, Lopes RD, Kallas EG, Bellissimo-Rodrigues F. Efficacy and safety of Ixekizumab vs. low-dose IL-2 vs. Colchicine vs. standard of care in the treatment of patients hospitalized with moderate-to-critical COVID-19: A pilot randomized clinical trial (STRUCK: Survival Trial Using Cytokine Inhibitors). Rev Soc Bras Med Trop 2023; 56:e0565. [PMID: 37075454 PMCID: PMC10109354 DOI: 10.1590/0037-8682-0565-2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 03/08/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND Cases of coronavirus disease 2019 (COVID-19) requiring hospitalization continue to appear in vulnerable populations, highlighting the importance of novel treatments. The hyperinflammatory response underlies the severity of the disease, and targeting this pathway may be useful. Herein, we tested whether immunomodulation focusing on interleukin (IL)-6, IL-17, and IL-2, could improve the clinical outcomes of patients admitted with COVID-19. METHODS This multicenter, open-label, prospective, randomized controlled trial was conducted in Brazil. Sixty hospitalized patients with moderate-to-critical COVID-19 received in addition to standard of care (SOC): IL-17 inhibitor (ixekizumab 80 mg SC/week) 1 dose every 4 weeks; low-dose IL-2 (1.5 million IU per day) for 7 days or until discharge; or indirect IL-6 inhibitor (colchicine) orally (0.5 mg) every 8 hours for 3 days, followed by 4 weeks at 0.5 mg 2x/day; or SOC alone. The primary outcome was accessed in the "per protocol" population as the proportion of patients with clinical improvement, defined as a decrease greater or equal to two points on the World Health Organization's (WHO) seven-category ordinal scale by day 28. RESULTS All treatments were safe, and the efficacy outcomes did not differ significantly from those of SOC. Interestingly, in the colchicine group, all participants had an improvement of greater or equal to two points on the WHO seven-category ordinal scale and no deaths or patient deterioration were observed. CONCLUSIONS Ixekizumab, colchicine, and IL-2 were demonstrated to be safe but ineffective for COVID-19 treatment. These results must be interpreted cautiously because of the limited sample size.
Collapse
Affiliation(s)
- Lívia Pimenta Bonifácio
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Ribeirão Preto, SP, Brasil
| | - Eduardo Ramacciotti
- Science Valley Research Institute, São Paulo, SP, Brasil
- Grupo Leforte, Hospital e Maternidade Christóvão da Gama, Santo André, SP, Brasil
| | | | | | | | - Paulo Louzada Júnior
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Ribeirão Preto, SP, Brasil
| | | | | | | | - Valéria Cristina Resende Aguiar
- Science Valley Research Institute, São Paulo, SP, Brasil
- Grupo Leforte, Hospital e Maternidade Christóvão da Gama, Santo André, SP, Brasil
| | | | | | | | | | | | - Renato Delascio Lopes
- Brazilian Clinical Research Institute, São Paulo, SP, Brasil
- Duke University Medical Center - Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Esper Georges Kallas
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Moléstias Infecciosas e Parasitárias, São Paulo, SP, Brasil
| | | |
Collapse
|
31
|
Soliman AM, Barreda DR. Acute Inflammation in Tissue Healing. Int J Mol Sci 2022; 24:ijms24010641. [PMID: 36614083 PMCID: PMC9820461 DOI: 10.3390/ijms24010641] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/19/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
There are well-established links between acute inflammation and successful tissue repair across evolution. Innate immune reactions contribute significantly to pathogen clearance and activation of subsequent reparative events. A network of molecular and cellular regulators supports antimicrobial and tissue repair functions throughout the healing process. A delicate balance must be achieved between protection and the potential for collateral tissue damage associated with overt inflammation. In this review, we summarize the contributions of key cellular and molecular components to the acute inflammatory process and the effective and timely transition toward activation of tissue repair mechanisms. We further discuss how the disruption of inflammatory responses ultimately results in chronic non-healing injuries.
Collapse
Affiliation(s)
- Amro M. Soliman
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Daniel R. Barreda
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Correspondence: ; Tel.: +1-(780)492-0375
| |
Collapse
|
32
|
Lv Y, Ricard L, Gaugler B, Huang H, Ye Y. Biology and clinical relevance of follicular cytotoxic T cells. Front Immunol 2022; 13:1036616. [PMID: 36591286 PMCID: PMC9794565 DOI: 10.3389/fimmu.2022.1036616] [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: 09/04/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Follicular cytotoxic T (Tfc) cells are a newly identified subset of CD8+ T cells enriched in B cell follicles and their surroundings, which integrate multiple functions such as killing, memory, supporting and regulation. Tfc cells share similarities with follicular helper T (Tfh) cells, conventional cytotoxic CD8+ T (Tc cells)cells and follicular regulatory T (Tfr) cells, while they express distinct transcription factors, phenotype, and perform different functions. With the participation of cytokines and cell-cell interactions, Tfc cells modulate Tfh cells and B cells and play an essential role in regulating the humoral immunity. Furthermore, Tfc cells have been found to change in their frequencies and functions during the occurrence and progression of chronic infections, immune-mediated diseases and cancers. Strategies targeting Tfc cells are under investigations, bringing novel insights into control of these diseases. We summarize the characteristics of Tfc cells, and introduce the roles and potential targeting modalities of Tfc cells in different diseases.
Collapse
Affiliation(s)
- Yuqi Lv
- Bone Marrow Transplantation Center, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China,Liangzhu Laboratory of Zhejiang University Medical Center, Hangzhou, Zhejiang, China,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China,Zhejiang Province Stem Cell and Cellular Immunotherapy Engineering Laboratory, Hangzhou, Zhejiang, China
| | - Laure Ricard
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France,AP-HP, Hôpital Saint-Antoine, Service d’Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, Paris, France
| | - Béatrice Gaugler
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France,AP-HP, Hôpital Saint-Antoine, Service d’Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, Paris, France
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China,Liangzhu Laboratory of Zhejiang University Medical Center, Hangzhou, Zhejiang, China,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China,Zhejiang Province Stem Cell and Cellular Immunotherapy Engineering Laboratory, Hangzhou, Zhejiang, China,*Correspondence: Yishan Ye, ; He Huang,
| | - Yishan Ye
- Bone Marrow Transplantation Center, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China,Liangzhu Laboratory of Zhejiang University Medical Center, Hangzhou, Zhejiang, China,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China,Zhejiang Province Stem Cell and Cellular Immunotherapy Engineering Laboratory, Hangzhou, Zhejiang, China,*Correspondence: Yishan Ye, ; He Huang,
| |
Collapse
|
33
|
Alomari N, Totonchy J. IL-21 signaling promotes the establishment of KSHV infection in human tonsil lymphocytes by increasing differentiation and targeting of plasma cells. Front Immunol 2022; 13:1010274. [PMID: 36569889 PMCID: PMC9769966 DOI: 10.3389/fimmu.2022.1010274] [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: 08/02/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Introduction Factors influencing Kaposi's sarcoma-associated herpesvirus (KSHV) transmission and the early stages of KSHV infection in the human immune system remain poorly characterized. KSHV is known to extensively manipulate the host immune system and the cytokine milieu, and cytokines are known to influence the progression of KSHV-associated diseases. Our previous work identified the early targeting of plasma cells for KSHV infection. In this study, we examine whether IL-21, a cytokine known to profoundly influence plasma cell fate, influences the early stages of KSHV infection in B lymphocytes. Methods Using our unique model of ex vivo KSHV infection in tonsil lymphocytes, we investigate the influence of IL-21 supplementation, IL-21 neutralization, the distribution of IL-21 receptor on B cell subsets and IL-21 secreting T cell subsets on the establishment of KSHV infection in human B cells. Results We show that IL-21 signaling promotes KSHV infection by promoting both total plasma cell numbers and increasing KSHV infection in plasma cells as early as 3 days post-infection. We further demonstrate that the synergistic effect of KSHV infection and IL-21 treatment on plasma cell frequencies is due to differentiation of new plasma cells from naïve B cell precursors. We examine T cells secreting IL-21 in our tonsil specimens and determine that IL-21 producing CD8+ central memory T cells are correlated with plasma cell frequencies and KSHV targeting of plasma cells. Discussion These results demonstrate the novel finding that differentiation of new plasma cells is involved in the early stages of KSHV infection in B cells, and that IL-21 signaling can potentiate this effect thereby increasing the overall magnitude of KSHV infection at early timepoints. These results suggest that IL-21 signaling represents a host-level susceptibility factor for the establishment of KSHV infection.
Collapse
|
34
|
Chandran S, Tang Q. Impact of interleukin-6 on T cells in kidney transplant recipients. Am J Transplant 2022; 22 Suppl 4:18-27. [PMID: 36453710 DOI: 10.1111/ajt.17209] [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: 09/05/2022] [Accepted: 09/23/2022] [Indexed: 12/02/2022]
Abstract
Interleukin-6 (IL-6), a multifunctional proinflammatory cytokine, plays a key role in T cell activation, survival, and differentiation. Acting as a switch that induces the differentiation of naïve T cells into Th17 cells and inhibits their development into regulatory T cells, IL-6 promotes rejection and abrogates tolerance. Therapies that target IL-6 signaling include antibodies to IL-6 and the IL-6 receptor and inhibitors of janus kinases; several of these therapeutics have demonstrated robust clinical efficacy in autoimmune and inflammatory diseases. Clinical trials of IL-6 inhibition in kidney transplantation have focused primarily on its effects on B cells, plasma cells, and HLA antibodies. In this review, we summarize the impact of IL-6 on T cells in experimental models of transplant and describe the effects of IL-6 inhibition on the T cell compartment in kidney transplant recipients.
Collapse
Affiliation(s)
- Sindhu Chandran
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Qizhi Tang
- Department of Surgery, Diabetes Center, Gladstone-UCSF Institute of Genome Immunology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
35
|
4,7-Didehydro-neophysalin B Protects Rat Lung Epithelial Cells against Hydrogen Peroxide-Induced Oxidative Damage through Nrf2-Mediated Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4189083. [PMID: 36132230 PMCID: PMC9484967 DOI: 10.1155/2022/4189083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 06/29/2022] [Accepted: 07/19/2022] [Indexed: 12/14/2022]
Abstract
The administration of 4,7-didehydro-neophysalin B is expected to be a promising strategy for mitigating oxidative stress in respiratory diseases. This study was aimed at investigating the efficacy of 4,7-didehydro-neophysalin B for apoptosis resistance of rat lung epithelial cells (RLE-6TN) to oxidative stress and evaluating its underlying mechanism of action. The RLE-6TN cells treated with hydrogen peroxide (H2O2) were divided into five groups, and 4,7-didehydro-neophysalin B was administered into it. To evaluate its mechanism of action, the expression of oxidative stress and apoptotic proteins was investigated. 4,7-Didehydro-neophysalin B significantly inhibited H2O2-induced RLE-6TN cell damage. It also activated the Nrf2 signaling pathway which was evident from the increased transcription of antioxidant responsive of KLF9, NQO1, Keap-1, and HO-1. Nrf2 was found to be a potential target of 4,7-didehydro-neophysalin B. The protein levels of Bcl-2 and Bcl-xL were increased while Bax and p53 were decreased significantly. Flow cytometry showed that 4,7-didehydro-neophysalin B protected RLE-6TN cells from apoptosis and has improved the oxidative damage. This study provided a promising evidence that 4,7-didehydro-neophysalin B can be a therapeutic option for oxidative stress in respiratory diseases.
Collapse
|
36
|
Khaledi M, Sameni F, Yahyazade S, Radandish M, Owlia P, Bagheri N, Afkhami H, Mahjoor M, Esmaelpour Z, Kohansal M, Aghaei F. COVID-19 and the potential of Janus family kinase (JAK) pathway inhibition: A novel treatment strategy. Front Med (Lausanne) 2022; 9:961027. [PMID: 36111104 PMCID: PMC9469902 DOI: 10.3389/fmed.2022.961027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Recent evidence proposed that the severity of the coronavirus disease 2019 (COVID-19) in patients is a consequence of cytokine storm, characterized by increased IL-1β, IL-6, IL-18, TNF-α, and IFN-γ. Hence, managing the cytokine storm by drugs has been suggested for the treatment of patients with severe COVID-19. Several of the proinflammatory cytokines involved in the pathogenesis of COVID-19 infection recruit a distinct intracellular signaling pathway mediated by JAKs. Consequently, JAK inhibitors, including baricitinib, pacritinib, ruxolitinib, and tofacitinib, may represent an effective therapeutic strategy for controlling the JAK to treat COVID-19. This study indicates the mechanism of cytokine storm and JAK/STAT pathway in COVID-19 as well as the medications used for JAK/STAT inhibitors.
Collapse
Affiliation(s)
- Mansoor Khaledi
- Department of Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Fatemeh Sameni
- Department of Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Sheida Yahyazade
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maedeh Radandish
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parviz Owlia
- Molecular Microbiology Research Center, Faculty of Medicine, Shahed University, Tehran, Iran
- *Correspondence: Parviz Owlia ;
| | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Nader Bagheri
| | | | - Mohamad Mahjoor
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Esmaelpour
- Reference Laboratory for Bovine Tuberculosis, Razi Vaccine and Serum Research Institute, Karaj, Iran
| | - Maryam Kohansal
- Department of Medical Biotechnology, Fasa University of Medical Sciences, Fasa, Iran
| | - Farzad Aghaei
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
37
|
Sayın Ekinci N, Darbaş Ş, Uçar F. CXCR5+CD8+ Follicular Cytotoxic T Cell Biology and Its Relationship with Diseases. TURKISH JOURNAL OF IMMUNOLOGY 2022. [DOI: 10.4274/tji.galenos.2022.04796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
38
|
Aliyu M, Zohora FT, Anka AU, Ali K, Maleknia S, Saffarioun M, Azizi G. Interleukin-6 cytokine: An overview of the immune regulation, immune dysregulation, and therapeutic approach. Int Immunopharmacol 2022; 111:109130. [PMID: 35969896 DOI: 10.1016/j.intimp.2022.109130] [Citation(s) in RCA: 134] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/26/2022] [Accepted: 08/03/2022] [Indexed: 12/19/2022]
Abstract
Several studies have shown that interleukin 6 (IL-6) is a multifunctional cytokine with both pro-inflammatory and anti-inflammatory activity, depending on the immune response context. Macrophages are among several cells that secrete IL-6, which they express upon activation by antigens, subsequently inducing fever and production of acute-phase proteins from the liver. Moreover, IL-6 induces the final maturation of B cells into memory B cells and plasma cells as well as an adaptive role for short-term energy allocation. Activation of IL-6 receptors results in the intracellular activation of the JAK/STAT pathway with resultant production of inflammatory cytokines. Several mechanisms-controlled IL-6 expression, but aberrant production was shown to be crucial in the pathogenesis of many diseases, which include autoimmune and chronic inflammatory diseases. IL-6 in combination with transforming growth factor β (TGF-β) induced differentiation of naïve T cells to Th17 cells, which is the cornerstone in autoimmune diseases. Recently, IL-6 secretion was shown to form the backbone of hypercytokinemia seen in the Coronavirus disease 2019 (COVID-19)-associated hyperinflammation and multiorgan failure. There are two classes of approved IL-6 inhibitors: anti-IL-6 receptor monoclonal antibodies (e.g., tocilizumab) and anti-IL-6 monoclonal antibodies (i.e., siltuximab). These drugs have been evaluated in patients with rheumatoid arthritis, juvenile idiopathic arthritis, cytokine release syndrome, and COVID-19 who have systemic inflammation. JAK/STAT pathway blockers were also successfully used in dampening IL-6 signal transduction. A better understanding of different mechanisms that modulate IL-6 expression will provide the much-needed solution with excellent safety and efficacy profiles for the treatment of autoimmune and inflammatory diseases in which IL-6 derives their pathogenesis.
Collapse
Affiliation(s)
- Mansur Aliyu
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, International Campus, TUMS-IC, Tehran, Iran; Department of Medical Microbiology, Faculty of Clinical Science, College of Health Sciences, Bayero University, Kano, Nigeria
| | - Fatema Tuz Zohora
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Malaysia
| | - Abubakar Umar Anka
- Department of Medical Laboratory Science, College of Medical Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Kashif Ali
- Department of Pharmacy Abdul Wali, Khan University Mardan, Pakistan
| | - Shayan Maleknia
- Biopharmaceutical Research Center, AryoGen Pharmed Inc., Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Saffarioun
- Biopharmaceutical Research Center, AryoGen Pharmed Inc., Alborz University of Medical Sciences, Karaj, Iran
| | - Gholamreza Azizi
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
39
|
Groza Y, Jemelkova J, Kafkova LR, Maly P, Raska M. IL-6 and its role in IgA nephropathy development. Cytokine Growth Factor Rev 2022; 66:1-14. [PMID: 35527168 DOI: 10.1016/j.cytogfr.2022.04.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 04/05/2022] [Indexed: 02/07/2023]
Abstract
IL-6 is considered one of the well characterized cytokines exhibiting homeostatic, pro- and anti-inflammatory activities, depending on the receptor variant and the induced intracellular cis- or trans-signaling responses. IL-6-activated pathways are involved in the regulation of cell proliferation, survival, differentiation, and cell metabolism changes. Deviations in IL-6 levels or abnormal response to IL-6 signaling are associated with several autoimmune diseases including IgA nephropathy (IgAN), one of most frequent primary glomerulonephritis worldwide. IgAN is associated with increased plasma concentration of IL-6 and increased plasma concentration of aberrantly galactosylated IgA1 immunoglobulin (Gd-IgA1). Gd-IgA1 is specifically recognized by autoantibodies, leading to the formation of circulating immune complexes (CIC) with nephritogenic potential, since CIC deposited in the glomerular mesangium induce mesangial cells proliferation and glomerular injury. Infection of the upper respiratory or digestive tract enhances IL-6 production and in IgAN patients is often followed by the macroscopic hematuria. This review recapitulates general aspects of IL-6 signaling and summarizes experimental evidences about IL-6 involvement in the etiopathogenesis of IgA nephropathy through the production of Gd-IgA1 and regulation of mesangial cell proliferation.
Collapse
Affiliation(s)
- Yaroslava Groza
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Center, Prumyslova 595, Vestec 252 50, Czech Republic
| | - Jana Jemelkova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, Olomouc 779 00, Czech Republic
| | - Leona Raskova Kafkova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, Olomouc 779 00, Czech Republic.
| | - Petr Maly
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Center, Prumyslova 595, Vestec 252 50, Czech Republic
| | - Milan Raska
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, Olomouc 779 00, Czech Republic.
| |
Collapse
|
40
|
Cusimano GM, Gary EN, Bell MR, Warner BM, Connors J, Tursi NJ, Ali AR, Zhang S, Canziani G, Taramangalam B, Gordon EA, Chaiken IM, Wootton SK, Smith T, Ramos S, Kobasa D, Weiner DB, Kutzler MA, Haddad EK. Improved Durability to SARS-CoV-2 Vaccine Immunity following Coimmunization with Molecular Adjuvant Adenosine Deaminase-1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:118-127. [PMID: 35750334 PMCID: PMC9246991 DOI: 10.4049/jimmunol.2200056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/26/2022] [Indexed: 06/03/2023]
Abstract
Although severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines have demonstrated strong immunogenicity and protection against severe disease, concerns about the duration and breadth of these responses remain. In this study, we show that codelivery of plasmid-encoded adenosine deaminase-1 (pADA) with SARS-CoV-2 spike glycoprotein DNA enhances immune memory and durability in vivo. Coimmunized mice displayed increased spike-specific IgG of higher affinity and neutralizing capacity as compared with plasmid-encoded spike-only-immunized animals. Importantly, pADA significantly improved the longevity of these enhanced responses in vivo. This coincided with durable increases in frequencies of plasmablasts, receptor-binding domain-specific memory B cells, and SARS-CoV-2-specific T follicular helper cells. Increased spike-specific T cell polyfunctionality was also observed. Notably, animals coimmunized with pADA had significantly reduced viral loads compared with their nonadjuvanted counterparts in a SARS-CoV-2 infection model. These data suggest that pADA enhances immune memory and durability and supports further translational studies.
Collapse
Affiliation(s)
- Gina M Cusimano
- Department of Medicine, Drexel University College of Medicine, Philadelphia, PA
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA
| | - Ebony N Gary
- The Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA
| | - Matthew R Bell
- Department of Medicine, Drexel University College of Medicine, Philadelphia, PA
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA
| | - Bryce M Warner
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Jennifer Connors
- Department of Medicine, Drexel University College of Medicine, Philadelphia, PA
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA
| | - Nicholas J Tursi
- The Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA
| | - Ali R Ali
- The Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA
| | - Shiyu Zhang
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA
| | - Gabriela Canziani
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA
| | | | - Emma A Gordon
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA
| | - Irwin M Chaiken
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA
| | - Sarah K Wootton
- Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | | | | | - Darwyn Kobasa
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - David B Weiner
- The Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA
| | - Michele A Kutzler
- Department of Medicine, Drexel University College of Medicine, Philadelphia, PA;
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA
| | - Elias K Haddad
- Department of Medicine, Drexel University College of Medicine, Philadelphia, PA;
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA
| |
Collapse
|
41
|
Activated-memory T cells influence naïve T cell fate: a noncytotoxic function of human CD8 T cells. Commun Biol 2022; 5:634. [PMID: 35768564 PMCID: PMC9243096 DOI: 10.1038/s42003-022-03596-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 06/15/2022] [Indexed: 01/26/2023] Open
Abstract
T cells are endowed with the capacity to sense their environment including other T cells around them. They do so to set their numbers and activation thresholds. This form of regulation has been well-studied within a given T cell population - i.e., within the naïve or memory pool; however, less is known about the cross-talk between T cell subsets. Here, we tested whether memory T cells interact with and influence surrounding naïve T cells. We report that human naïve CD8 T cells (TN) undergo phenotypic and transcriptional changes in the presence of autologous activated-memory CD8 T cells (TMem). Following in vitro co-culture with activated central memory cells (TCM), ~3% of the TN acquired activation/memory canonical markers (CD45RO and CD95) in an MHC-I dependent-fashion. Using scRNA-seq, we also observed that ~3% of the TN acquired an activated/memory signature, while ~84% developed a unique activated transcriptional profile hybrid between naïve and activated memory. Pseudotime trajectory analysis provided further evidence that TN with an activated/memory or hybrid phenotype were derived from TN. Our data reveal a non-cytotoxic function of TMem with potential to activate autologous TN into the activated/memory pool. These findings may have implications for host-protection and autoimmunity that arises after vaccination, infection or transplantation.
Collapse
|
42
|
Huse C, Anstensrud AK, Michelsen AE, Ueland T, Broch K, Woxholt S, Yang K, Sharma K, Tøllefsen IM, Bendz B, Amundsen BH, Damås JK, Berg ES, Bjørkelund E, Quiles-Jiménez A, Bjerkeli V, Bendz C, Kleveland O, Stensaeth KH, Opdahl A, Kløw NE, Andersen GØ, Wiseth R, Halvorsen B, Gullestad L, Seljeflot I, Aukrust P, Osnes L, Dahl TB. Interleukin-6 inhibition in ST-elevation myocardial infarction: Immune cell profile in the randomised ASSAIL-MI trial. EBioMedicine 2022; 80:104013. [PMID: 35504178 PMCID: PMC9079006 DOI: 10.1016/j.ebiom.2022.104013] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/02/2022] [Accepted: 04/05/2022] [Indexed: 12/31/2022] Open
Abstract
Background We recently showed that interleukin (IL)-6 inhibition by tocilizumab improves myocardial salvage in ST-elevation myocardial infarction (STEMI). However, the mechanisms for this effect are not clear. Methods In this exploratory sub-study of the ASSAIL-MI trial, we examined leukocyte differential counts and their relation to myocardial salvage and peak troponin T (TnT) in STEMI patients randomised to tocilizumab (n = 101) or placebo (n = 98). We performed RNA-sequencing on whole blood (n = 40) and T cells (n = 20). B and T cell subpopulations were examined by flow cytometry (n = 69). Findings (i) STEMI patients had higher neutrophil counts at hospitalisation compared with stable angina patients. (ii) After percutaneous coronary intervention there was a gradual decline in neutrophils, which was significantly more pronounced in the tocilizumab group. (iii) The decrease in neutrophils in the tocilizumab group was associated with improved myocardial salvage and lower peak TnT. (iv) RNA-sequencing suggested that neutrophil function was also attenuated by tocilizumab. (v) B and T cell sub-populations changed only minimally after STEMI with minor effects of tocilizumab, supported as well by RNA-sequencing analyses of T cells. (vi) However, a low CD8+ count was associated with improved myocardial salvage in patients admitted to the hospital > 3 h after symptom onset. Interpretation Tocilizumab induced a rapid reduction in neutrophils and seemed to attenuate neutrophil function in STEMI patients potentially related to the beneficial effects of tocilizumab on myocardial salvage. Funding South-Eastern Norway Regional Health Authority (Nos. 2019067, 2017084), the Central Norway Regional Health Authority and Norwegian Research Council (No. 283867).
Collapse
Affiliation(s)
- Camilla Huse
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anne Kristine Anstensrud
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Annika E Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; K. G. Jebsen Thrombosis Research and Expertise Centre (TREC), The Arctic University of Norway, Tromsø, Norway
| | - Kaspar Broch
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway; K. G. Jebsen Cardiac Research Centre and Centre for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Sindre Woxholt
- Clinic of Cardiology, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Kuan Yang
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kapil Sharma
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | | | - Bjørn Bendz
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Brage Høyem Amundsen
- Clinic of Cardiology, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Jan Kristian Damås
- Department of Infectious Disease, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway; Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Erlend Sturle Berg
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | | | - Ana Quiles-Jiménez
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Vigdis Bjerkeli
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Christina Bendz
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Ola Kleveland
- Clinic of Cardiology, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Knut Haakon Stensaeth
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway; Department of Radiology and Nuclear Medicine, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Anders Opdahl
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Nils-Einar Kløw
- Department of Radiology, Oslo University Hospital Ullevål, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Geir Øystein Andersen
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway; Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Rune Wiseth
- Clinic of Cardiology, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lars Gullestad
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway; K. G. Jebsen Cardiac Research Centre and Centre for Heart Failure Research, University of Oslo, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ingebjørg Seljeflot
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Pål Aukrust
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Rheumatology, Dermatology and Infectious Disease, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Liv Osnes
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Tuva B Dahl
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital, Norway.
| |
Collapse
|
43
|
Akama-Garren EH, Carroll MC. T Cell Help in the Autoreactive Germinal Center. Scand J Immunol 2022; 95:e13192. [PMID: 35587582 DOI: 10.1111/sji.13192] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 11/29/2022]
Abstract
The germinal center serves as a site of B cell selection and affinity maturation, critical processes for productive adaptive immunity. In autoimmune disease tolerance is broken in the germinal center reaction, leading to production of autoreactive B cells that may propagate disease. Follicular T cells are crucial regulators of this process, providing signals necessary for B cell survival in the germinal center. Here we review the emerging roles of follicular T cells in the autoreactive germinal center. Recent advances in immunological techniques have allowed study of the gene expression profiles and repertoire of follicular T cells at unprecedented resolution. These studies provide insight into the potential role follicular T cells play in preventing or facilitating germinal center loss of tolerance. Improved understanding of the mechanisms of T cell help in autoreactive germinal centers provides novel therapeutic targets for diseases of germinal center dysfunction.
Collapse
Affiliation(s)
- Elliot H Akama-Garren
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,Harvard-MIT Health Sciences and Technology, Harvard Medical School, Boston, MA, USA
| | - Michael C Carroll
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
44
|
Tvedt THA, Rose-John S, Tsykunova G, Ahmed AB, Gedde-Dahl T, Ersvær E, Bruserud Ø. IL-6 Responsiveness of CD4+ and CD8+ T Cells after Allogeneic Stem Cell Transplantation Differs between Patients and Is Associated with Previous Acute Graft versus Host Disease and Pretransplant Antithymocyte Globulin Therapy. J Clin Med 2022; 11:jcm11092530. [PMID: 35566660 PMCID: PMC9104003 DOI: 10.3390/jcm11092530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/24/2022] [Accepted: 04/25/2022] [Indexed: 11/22/2022] Open
Abstract
Graft-versus-host disease (GVHD), one of the most common and serious complications after allogeneic stem cell transplantation, is mediated by allocative T cells. IL-6 mediates both pro- and anti-inflammatory effects and modulates T cell response through classical signaling and trans-signaling. We investigated the effects on the mTOR and JAK/STAT pathways after various types of IL-6 signaling for circulating T cells were derived from 31 allotransplant recipients 90 days post-transplant. Cells were stimulated with IL-6 alone, hyper-IL-6 (trans-signaling), IL-6+IL-6 receptor (IL-6R; classical + trans-signaling) and IL-6+IL-6R+soluble gp130-Fc (classical signaling), and flow cytometry was used to investigate the effects on phosphorylation of AKT (Thr308), mTOR (Ser2442), STAT3 (Ser727) and STAT3 (Tyr705). CD3+CD4+ and CD3+C8+ T cells responded to classical and trans IL-6 stimulation with increased STAT3 (Tyr705) phosphorylation; these responses were generally stronger for CD3+CD4+ cells. STAT3 (Tyr705) responses were stronger for patients with previous acute GVHD; CD3+CD4+ cells from GVHD patients showed an additional STAT3 (Ser727) response, whereas patients without acute GVHD showed additional mTOR (Ser2448) responses. Furthermore, treatment with antithymocyte globulin as a part of GVHD prophylaxis was associated with generally weaker STAT3 (Tyr705) responses and altered STAT3 (Ser727) responsiveness of CD3+CD4+ cells together with increased mTOR (Ser2448) responses for the CD3+CD8+ cells. Thus, early post-transplant CD3+CD4+ and CD3+ CD8+ T cell subsets differ in their IL-6 responsiveness; this responsiveness is modulated by antithymocyte globulin and differs between patients with and without previous acute GVHD. These observations suggest that allotransplant recipients will be heterogeneous with regard to the effects of post-transplant IL-6 targeting.
Collapse
Affiliation(s)
- Tor Henrik Anderson Tvedt
- Department of Hematology, University of Oslo, 0424 Oslo, Norway;
- Section for Hematology, Institute of Clinical Science, University of Bergen, 5007 Bergen, Norway;
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (G.T.); (A.B.A.)
- Correspondence: Correspondence:
| | - Stefan Rose-John
- Institute of Biochemistry, Kiel University, Olshausenstrasse 40, 24118 Kiel, Germany;
| | - Galina Tsykunova
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (G.T.); (A.B.A.)
| | - Aymen Bushra Ahmed
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (G.T.); (A.B.A.)
| | - Tobias Gedde-Dahl
- Department of Hematology, University of Oslo, 0424 Oslo, Norway;
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
| | - Elisabeth Ersvær
- Department of Biomedical Laboratory Scientist Education, Western Norway University of Applied Sciences, 5063 Bergen, Norway;
| | - Øystein Bruserud
- Section for Hematology, Institute of Clinical Science, University of Bergen, 5007 Bergen, Norway;
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (G.T.); (A.B.A.)
| |
Collapse
|
45
|
Mayberry CL, Logan NA, Wilson JJ, Chang CH. Providing a Helping Hand: Metabolic Regulation of T Follicular Helper Cells and Their Association With Disease. Front Immunol 2022; 13:864949. [PMID: 35493515 PMCID: PMC9047778 DOI: 10.3389/fimmu.2022.864949] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 03/25/2022] [Indexed: 01/02/2023] Open
Abstract
T follicular helper (Tfh) cells provide support to B cells upon arrival in the germinal center, and thus are critical for the generation of a robust adaptive immune response. Tfh express specific transcription factors and cellular receptors including Bcl6, CXCR5, PD-1, and ICOS, which are critical for homing and overall function. Generally, the induction of an immune response is tightly regulated. However, deviation during this process can result in harmful autoimmunity or the inability to successfully clear pathogens. Recently, it has been shown that Tfh differentiation, activation, and proliferation may be linked with the cellular metabolic state. In this review we will highlight recent discoveries in Tfh differentiation and explore how these cells contribute to functional immunity in disease, including autoimmune-related disorders, cancer, and of particular emphasis, during infection.
Collapse
Affiliation(s)
| | | | | | - Chih-Hao Chang
- The Jackson Laboratory, Bar Harbor, ME, United States
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
- *Correspondence: Chih-Hao Chang,
| |
Collapse
|
46
|
García-Juárez M, Camacho-Morales A. Defining the role of anti- and pro-inflammatory outcomes of Interleukin-6 in mental health. Neuroscience 2022; 492:32-46. [DOI: 10.1016/j.neuroscience.2022.03.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/03/2022] [Accepted: 03/16/2022] [Indexed: 01/03/2023]
|
47
|
Souza-Moreira L, Tan Y, Wang Y, Wang JP, Salkhordeh M, Virgo J, Florian M, Murray AB, Watpool I, McIntyre L, English S, Stewart DJ, Mei SH. Poly(I:C) Enhances Mesenchymal Stem Cell Control of Myeloid Cells From COVID-19 Patients. iScience 2022; 25:104188. [PMID: 35402859 PMCID: PMC8975597 DOI: 10.1016/j.isci.2022.104188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/25/2022] [Accepted: 03/30/2022] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are being studied for the treatment of COVID-19-associated critical illness, due to their immunomodulatory properties. Here, we hypothesized that viral mimic-priming improves MSCs’ abilities to rebalance the dysregulated immune responses in COVID-19. Transcriptome analysis of poly(I:C)-primed MSCs (pIC-MSCs) showed upregulation of pathways in antiviral and immunomodulatory responses. Together with increased expression of antiviral proteins such as MX1, IFITM3, and OAS1, these changes translated to greater effector functions in regulating monocytes and granulocytes while further enhancing MSCs’ ability to block SARS-CoV-2 pseudovirus entry into epithelial cells. Most importantly, the addition of pIC-MSCs to COVID-19 patient whole blood significantly reduced inflammatory neutrophils and increased M2 monocytes while enhancing their phagocytic effector function. We reveal for the first time that MSCs can be primed by Toll-like receptor 3 agonist to improve their ability to rebalance the dysregulated immune responses seen in severe SARS-CoV-2 infection. Viral stimuli mimic enhances MSC’s antiviral and immunomodulatory molecular profile pIC-primed MSCs inhibit SARS-CoV2 pseudovirus entry into epithelial cells pIC-MSCs recalibrate dysregulated cellular response in COVID patient’s whole blood
Collapse
|
48
|
Džopalić T, Tomić S, Bekić M, Vučević D, Mihajlović D, Eraković M, Čolić M. Ex vivo study of IL-6 expression and function in immune cell subsets from human periapical lesions. Int Endod J 2022; 55:480-494. [PMID: 35150455 DOI: 10.1111/iej.13704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 01/28/2022] [Accepted: 02/05/2022] [Indexed: 11/29/2022]
Abstract
AIM Even though IL-6 is a key inflammatory cytokine in periapical lesions (PLs), its function in stable periapical disease is unknown. Therefore, the aim of this study was to investigate following: first, the ex vivo production of IL-6 by clinically different PLs; next, subsets of immune cells expressing IL-6 in PLs according to their inflammatory status and finally, modulatory effect of IL-6 on T-cell cytokine production in cell cultures. METHODOLOGY Inflammatory cells were isolated from a total of 95 human PLs. Detection of IL-6+ cells within the myeloid and lymphoid populations was performed by multicolour flow cytometry. ELISA and FlowCytomix Microbeads Assay were used to measure cytokine levels in culture supernatants. To study the role of IL-6 in PLs, mononuclear cells (MNC) from symptomatic (Sy) or asymptomatic (Asy) PLs were treated with IL-6 or Tocilizumab, an IL-6R blocking antibody. The differences between groups were tested by unpaired t-test, Mann-Whitney or Friedman tests. RESULTS The levels of IL-6 in PL MNC culture supernatants were significantly higher compared to total PL cells and PL granulocytes (p<0.001). MNC from Sy PLs produced significantly hihger levels of IL-6 than MNC from Asy PLs (p<0.001). Flow cytometry analysis showed that NKT cells, CD8+ T cells and M2 macrophages (MØ), were dominant IL-6+ cells, in contrast to CD4+ T cells. However, CD8+ and CD4+ T cells contributed the most to IL-6 production. IL-6hi producing MNC cultures had higher levels of Th1 (IFN-γ), Th17 (IL-17A), Tfh (IL-21) and RANKL, whereas Th2 (IL-4) and Tregs cytokines (IL-10, TGF-β) were lower, compared to IL-6low producing cultures. Exogenous IL-6 stimulated 17A, IL-21 and RANKL, independently of PL activation status, but decreased IFN-γ, IL-4 and IL-33 levels in IL-6hi producing cultures. Tocilizumab increased IL-10 and TGF-β in IL-6low producing cultures. All differences were p<0.05. CONCLUSIONS Most immune cells from Sy PLs expressed higher levels of IL-6 compared to Asy PLs, which correlated with IL-6 production in culture. Analysis of cytokines suggested a dominant pro-inflammatory T-cell response and osteodestructive function of IL-6 in PLs judging by Th17/Tfh cell activation, Tregs inhibition and increased RANKL/OPG ratio. Excessive IL-6 production decreased Th1/Th2 responses.
Collapse
Affiliation(s)
- T Džopalić
- University of Niš, Medical Faculty, Niš, Serbia.,University of Defense in Belgrade, Medical Faculty of the Military Medical Academy, Belgrade, Serbia
| | - S Tomić
- University of Belgrade, Institute for the Application of Nuclear Energy, Belgrade, Serbia
| | - M Bekić
- University of Belgrade, Institute for the Application of Nuclear Energy, Belgrade, Serbia
| | - D Vučević
- University of Defense in Belgrade, Medical Faculty of the Military Medical Academy, Belgrade, Serbia
| | - D Mihajlović
- University of Defense in Belgrade, Medical Faculty of the Military Medical Academy, Belgrade, Serbia
| | - M Eraković
- Clinic for Stomatology, Military Medical Academy, Belgrade, Serbia
| | - M Čolić
- University of Belgrade, Institute for the Application of Nuclear Energy, Belgrade, Serbia.,University of East Sarajevo, Medical Faculty Foča, Foča R. Srpska Bosnia and Herzegovina
| |
Collapse
|
49
|
Qin T, Guo E, Lu F, Fu Y, Liu S, Xiao R, Wu X, Liu C, He C, Wang Z, Qin X, Hu D, You L, Li F, Li X, Huang X, Ma D, Xu X, Yang B, Fan J. Impact of chemotherapy and immunotherapy on the composition and function of immune cells in COVID-19 convalescent with gynecological tumors. Aging (Albany NY) 2021; 13:24943-24962. [PMID: 34862879 PMCID: PMC8714165 DOI: 10.18632/aging.203739] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/22/2021] [Indexed: 01/08/2023]
Abstract
Ongoing pandemic and potential resurgence of Coronavirus disease 2019 (COVID-19) has prompted urgent efforts to investigate the immunological memory of convalescent patients, especially in patients with active cancers. Here we performed single-cell RNA sequencing in peripheral blood samples of 3 healthy donors (HDs), 4 COVID-19 patients (Covs) and 4 COVID-19 patients with active gynecological tumor (TCs) pre- and post- anti-tumor treatment. All Covs patients had recovered from their acute infection. Interestingly, the molecular features of PBMCs in TCs are similar to that in Covs, suggesting that convalescent COVID-19 with gynecologic tumors do not have major immunological changes and may be protected against reinfection similar to COVID-19 patients without tumors. Moreover, the chemotherapy given to these patients mainly caused neutropenia, while having little effect on the proportion and functional phenotype of T and B cells, and T cell clonal expansion. Notably, anti-PD-L1 treatment massively increased cytotoxic scores of NK cells, and T cells, and facilitated clonal expansion of T cells in these patients. It is likely that T cells could protect patients from SARS-CoV-2 virus reinfection and anti-PD-L1 treatment can enhance the anti-viral activity of the T cells.
Collapse
Affiliation(s)
- Tianyu Qin
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ensong Guo
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Funian Lu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yu Fu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Si Liu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rourou Xiao
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xue Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chen Liu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chao He
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zizhuo Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xu Qin
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dianxing Hu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lixin You
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fuxia Li
- Department of Gynecology, Foshan Women and Children’s Hospital Affiliated to Southern Medical University, Foshan 528000, China
| | - Xi Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Cell, Development and Cancer Biology, Oregon Health and Sciences University, Portland, OR 97201, USA
| | - Xiaoyuan Huang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ding Ma
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoyan Xu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bin Yang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Junpeng Fan
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
50
|
Lacorcia M, Bhattacharjee S, Laubhahn K, Alhamdan F, Ram M, Muschaweckh A, Potaczek DP, Kosinska A, Garn H, Protzer U, Renz H, Prazeres da Costa C. Fetomaternal immune cross talk modifies T-cell priming through sustained changes to DC function. J Allergy Clin Immunol 2021; 148:843-857.e6. [PMID: 33684437 DOI: 10.1016/j.jaci.2021.02.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 02/18/2021] [Accepted: 02/26/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Prenatal exposure to infections can modify immune development. These environmental disturbances during early life potentially alter the incidence of inflammatory disorders as well as priming of immune responses. Infection with the helminth Schistosoma mansoni is widely studied for its ability to alter immune responsiveness and is associated with variations in coinfection, allergy, and vaccine efficacy in endemic populations. OBJECTIVE Exposure to maternal schistosomiasis during early life, even without transmission of infection, can result in priming effects on offspring immune responses to bystander antigenic challenges as related to allergic responsiveness and vaccination, with this article seeking to further clarify the effects and underlying immunologic imprinting. METHODS Here, we have combined a model of chronic maternal schistosomiasis infection with a thorough analysis of subsequent offspring immune responses to allergy and vaccination models, including viral challenge and steady-state changes to immune cell compartments. RESULTS We have demonstrated that maternal schistosomiasis alters CD4+ responses during allergic sensitization and challenge in a skewed IL-4/B-cell-dominant response to antigenic challenge associated with limited inflammatory response. Beyond that, we have uncovered previously unidentified alterations to CD8+ T-cell responses during immunization that are dependent on vaccine formulation and have functional impact on the efficacy of vaccination against viral infection in a murine hepatitis B virus model. CONCLUSION In addition to steady-state modifications to CD4+ T-cell polarization and B-cell priming, we have traced these modified CD8+ responses to an altered dendritic cell phenotype sustained into adulthood, providing evidence for complex priming effects imparted by infection via fetomaternal cross talk.
Collapse
Affiliation(s)
- Matthew Lacorcia
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
| | - Sonakshi Bhattacharjee
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
| | - Kristina Laubhahn
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany; Pediatric Allergology, Department of Pediatrics, Dr von Hauner Children's Hospital, University Hospital, Ludwig Maximilian University Munich, Munich, Germany; German Center for Lung Research, Ludwig Maximilian University Munich, Munich, Germany
| | - Fahd Alhamdan
- Biochemical Pharmacological Center, Translational Inflammation Division & Core Facility for Single Cell Multiomics, Philipps University Marburg, Marburg, Germany
| | - Marija Ram
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
| | - Andreas Muschaweckh
- Department of Neurology, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Daniel P Potaczek
- Biochemical Pharmacological Center, Translational Inflammation Division & Core Facility for Single Cell Multiomics, Philipps University Marburg, Marburg, Germany
| | - Anna Kosinska
- Institute for Virology Technical University of Munich, Munich, Germany
| | - Holger Garn
- Biochemical Pharmacological Center, Translational Inflammation Division & Core Facility for Single Cell Multiomics, Philipps University Marburg, Marburg, Germany
| | - Ulrike Protzer
- Institute for Virology Technical University of Munich, Munich, Germany
| | - Harald Renz
- Biochemical Pharmacological Center, Translational Inflammation Division & Core Facility for Single Cell Multiomics, Philipps University Marburg, Marburg, Germany
| | - Clarissa Prazeres da Costa
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany.
| |
Collapse
|