1
|
Manville RW, Yoshimura RF, Yeromin AV, Hogenkamp D, van der Horst J, Zavala A, Chinedu S, Arena G, Lasky E, Fisher M, Tracy CR, Othy S, Jepps TA, Cahalan MD, Abbott GW. Polymodal K + channel modulation contributes to dual analgesic and anti-inflammatory actions of traditional botanical medicines. Commun Biol 2024; 7:1059. [PMID: 39198706 PMCID: PMC11358443 DOI: 10.1038/s42003-024-06752-y] [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: 06/06/2023] [Accepted: 08/19/2024] [Indexed: 09/01/2024] Open
Abstract
Pain and inflammation contribute immeasurably to reduced quality of life, yet modern analgesic and anti-inflammatory therapeutics can cause dependence and side effects. Here, we screened 1444 plant extracts, prepared primarily from native species in California and the United States Virgin Islands, against two voltage-gated K+ channels - T-cell expressed Kv1.3 and nociceptive-neuron expressed Kv7.2/7.3. A subset of extracts both inhibits Kv1.3 and activates Kv7.2/7.3 at hyperpolarized potentials, effects predicted to be anti-inflammatory and analgesic, respectively. Among the top dual hits are witch hazel and fireweed; polymodal modulation of multiple K+ channel types by hydrolysable tannins contributes to their dual anti-inflammatory, analgesic actions. In silico docking and mutagenesis data suggest pore-proximal extracellular linker sequence divergence underlies opposite effects of hydrolysable tannins on different Kv1 isoforms. The findings provide molecular insights into the enduring, widespread medicinal use of witch hazel and fireweed and demonstrate a screening strategy for discovering dual anti-inflammatory, analgesic small molecules.
Collapse
Affiliation(s)
- Rían W Manville
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Ryan F Yoshimura
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Andriy V Yeromin
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Derk Hogenkamp
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Jennifer van der Horst
- Department of Biomedical Sciences, Vascular Biology Group, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Angel Zavala
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Sonia Chinedu
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Grey Arena
- Redwood Creek Vegetation Team, National Park Service, Sausalito, CA, USA
| | - Emma Lasky
- Redwood Creek Vegetation Team, National Park Service, Sausalito, CA, USA
| | - Mark Fisher
- Philip L. Boyd Deep Canyon Desert Research Center, University of California Natural Reserve System, Indian Wells, CA, USA
| | - Christopher R Tracy
- Philip L. Boyd Deep Canyon Desert Research Center, University of California Natural Reserve System, Indian Wells, CA, USA
| | - Shivashankar Othy
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Thomas A Jepps
- Department of Biomedical Sciences, Vascular Biology Group, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Michael D Cahalan
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA.
| |
Collapse
|
2
|
Lin J, Wu Y, Liu G, Cui R, Xu Y. Advances of ultrasound in tumor immunotherapy. Int Immunopharmacol 2024; 134:112233. [PMID: 38735256 DOI: 10.1016/j.intimp.2024.112233] [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: 01/09/2024] [Revised: 04/11/2024] [Accepted: 05/07/2024] [Indexed: 05/14/2024]
Abstract
Immunotherapy has become a revolutionary method for treating tumors, offering new hope to cancer patients worldwide. Immunotherapy strategies such as checkpoint inhibitors, chimeric antigen receptor T-cell (CAR-T) therapy, and cancer vaccines have shown significant potential in clinical trials. Despite the promising results, there are still limitations that impede the overall effectiveness of immunotherapy; the response to immunotherapy is uneven, the response rate of patients is still low, and systemic immune toxicity accompanied with tumor cell immune evasion is common. Ultrasound technology has evolved rapidly in recent years and has become a significant player in tumor immunotherapy. The introductions of high intensity focused ultrasound and ultrasound-stimulated microbubbles have opened doors for new therapeutic strategies in the fight against tumor. This paper explores the revolutionary advancements of ultrasound combined with immunotherapy in this particular field.
Collapse
Affiliation(s)
- Jing Lin
- Department of Ultrasound, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, Zhuhai, PR China.
| | - Yuwei Wu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macao, PR China
| | - Guangde Liu
- Department of Ultrasound, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, Zhuhai, PR China
| | - Rui Cui
- Department of Ultrasonography, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, PR China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, PR China
| | - Youhua Xu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macao, PR China; Macau University of Science and Technology Zhuhai MUST Science and Technology Research Institute, Hengqin, Zhuhai, PR China.
| |
Collapse
|
3
|
Jin S, Guo Y, Wang X. Development of Platinum Complexes for Tumor Chemoimmunotherapy. Chemistry 2024; 30:e202302948. [PMID: 38171804 DOI: 10.1002/chem.202302948] [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: 09/10/2023] [Indexed: 01/05/2024]
Abstract
Platinum complexes are potential antitumor drugs in chemotherapy. Their impact on tumor treatment could be greatly strengthened by combining with immunotherapy. Increasing evidences indicate that the antitumor activity of platinum complexes is not limited to chemical killing effects, but also extends to immunomodulatory actions. This review introduced the general concept of chemoimmunotherapy and summarized the progress of platinum complexes as chemoimmunotherapeutic agents in recent years. Platinum complexes could be developed into inducers of immunogenic cell death, blockers of immune checkpoint, regulators of immune signaling pathway, and modulators of tumor immune microenvironment, etc. The synergy between chemotherapeutic and immunomodulatory effects reinforces the antitumor activity of platinum complexes, and helps them circumvent the drug resistance and systemic toxicity. The exploration of platinum complexes for chemoimmunotherapy may create new opportunities to revive the discovery of metal anticancer drugs.
Collapse
Affiliation(s)
- Suxing Jin
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, P. R. China
| | - Yan Guo
- School of Materials and Chemical Engineering, Henan University of Urban Construction, Pingdingshan, 467036, Henan, P. R. China
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, P. R. China
| |
Collapse
|
4
|
Mortezaee K. Selective targeting or reprogramming of intra-tumoral Tregs. Med Oncol 2024; 41:71. [PMID: 38341821 DOI: 10.1007/s12032-024-02300-0] [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: 11/18/2023] [Accepted: 01/03/2024] [Indexed: 02/13/2024]
Abstract
Regulatory T cells (Tregs) are critical immunosuppressive cells that are frequently present in the tumor microenvironment of solid cancers and enable progression of tumors toward metastasis. The cells expand in response to tumor-associated antigens and are actively involved in bypassing immunotherapy with immune checkpoint inhibitors through integrating numerous environmental signals. A point here is that Tregs are clonally distinct in peripheral blood from tumor area. Currently, an effective and novel task in cancer immunotherapy is to selectively destabilize or deplete intra-tumoral Tregs in order to avoid systemic inflammatory events. Helios is a transcription factor expressed selectively by Tregs and promotes their stabilization, and Trps1 is a master regulator of intra-tumoral Tregs. Anti-CCR8 and the IL-2Rβγ agonist Bempegaldesleukin selectively target intra-tumoral Treg population, with the former approved to not elicit autoimmunity. Disarming Treg-related immunosuppression in tumors through diverting their reprogramming or promoting naïve T cell differentiation into cells with effector immune activating profile is another promising area of research in cancer immunotherapy. Blimp-1 inhibitors and glucocorticoid-induced TNFR-related protein agonists are example approaches that can be used for diverting Treg differentiation into Th1-like CD4+ T cells, thereby powering immunogenicity against cancer. Finally, selective target of intra-tumoral Tregs and their reprogramming into effector T cells is applicable using low-dose chemotherapy, and high-salt and high-tryptophan diet.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
5
|
Wong C, Stoilova I, Gazeau F, Herbeuval JP, Fourniols T. Mesenchymal stromal cell derived extracellular vesicles as a therapeutic tool: immune regulation, MSC priming, and applications to SLE. Front Immunol 2024; 15:1355845. [PMID: 38390327 PMCID: PMC10881725 DOI: 10.3389/fimmu.2024.1355845] [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: 12/14/2023] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by a dysfunction of the immune system. Mesenchymal stromal cell (MSCs) derived extracellular vesicles (EVs) are nanometer-sized particles carrying a diverse range of bioactive molecules, such as proteins, miRNAs, and lipids. Despite the methodological disparities, recent works on MSC-EVs have highlighted their broad immunosuppressive effect, thus driving forwards the potential of MSC-EVs in the treatment of chronic diseases. Nonetheless, their mechanism of action is still unclear, and better understanding is needed for clinical application. Therefore, we describe in this review the diverse range of bioactive molecules mediating their immunomodulatory effect, the techniques and possibilities for enhancing their immune activity, and finally the potential application to SLE.
Collapse
Affiliation(s)
- Christophe Wong
- EVerZom, Paris, France
- Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8601, Université Paris Cité, Paris, France
- Chemistry and Biology, Modeling and Immunology for Therapy (CBMIT), Université Paris Cité, Paris, France
| | - Ivana Stoilova
- Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8601, Université Paris Cité, Paris, France
- Chemistry and Biology, Modeling and Immunology for Therapy (CBMIT), Université Paris Cité, Paris, France
| | - Florence Gazeau
- Matière et Systèmes Complexes (MSC) UMR CNRS 7057, Université Paris Cité, Paris, France
| | - Jean-Philippe Herbeuval
- Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8601, Université Paris Cité, Paris, France
- Chemistry and Biology, Modeling and Immunology for Therapy (CBMIT), Université Paris Cité, Paris, France
| | | |
Collapse
|
6
|
Santry LA, van Vloten JP, AuYeung AWK, Mould RC, Yates JGE, McAusland TM, Petrik JJ, Major PP, Bridle BW, Wootton SK. Recombinant Newcastle disease viruses expressing immunological checkpoint inhibitors induce a pro-inflammatory state and enhance tumor-specific immune responses in two murine models of cancer. Front Microbiol 2024; 15:1325558. [PMID: 38328418 PMCID: PMC10847535 DOI: 10.3389/fmicb.2024.1325558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 01/02/2024] [Indexed: 02/09/2024] Open
Abstract
Introduction Tumor microenvironments are immunosuppressive due to progressive accumulation of mutations in cancer cells that can drive expression of a range of inhibitory ligands and cytokines, and recruitment of immunomodulatory cells, including myeloid-derived suppressor cells (MDSC), tumor-associated macrophages, and regulatory T cells (Tregs). Methods To reverse this immunosuppression, we engineered mesogenic Newcastle disease virus (NDV) to express immunological checkpoint inhibitors anti-cytotoxic T lymphocyte antigen-4 and soluble programmed death protein-1. Results Intratumoral administration of recombinant NDV (rNDV) to mice bearing intradermal B16-F10 melanomas or subcutaneous CT26LacZ colon carcinomas led to significant changes in the tumor-infiltrating lymphocyte profiles. Vectorizing immunological checkpoint inhibitors in NDV increased activation of intratumoral natural killer cells and cytotoxic T cells and decreased Tregs and MDSCs, suggesting induction of a pro-inflammatory state with greater infiltration of activated CD8+ T cells. These notable changes translated to higher ratios of activated effector/suppressor tumor-infiltrating lymphocytes in both cancer models, which is a promising prognostic marker. Whereas all rNDV-treated groups showed evidence of tumor regression and increased survival in the CT26LacZ and B16-F10, only treatment with NDV expressing immunological checkpoint blockades led to complete responses compared to tumors treated with NDV only. Discussion These data demonstrated that NDV expressing immunological checkpoint inhibitors could reverse the immunosuppressive state of tumor microenvironments and enhance tumor-specific T cell responses.
Collapse
Affiliation(s)
- Lisa A. Santry
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Jacob P. van Vloten
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Amanda W. K. AuYeung
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Robert C. Mould
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Jacob G. E. Yates
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Thomas M. McAusland
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - James J. Petrik
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | | | - Byram W. Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Sarah K. Wootton
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
7
|
Spiliopoulou P, Kaur P, Hammett T, Di Conza G, Lahn M. Targeting T regulatory (T reg) cells in immunotherapy-resistant cancers. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:2. [PMID: 38318526 PMCID: PMC10838381 DOI: 10.20517/cdr.2023.46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 12/11/2023] [Accepted: 01/09/2024] [Indexed: 02/07/2024]
Abstract
Primary or secondary (i.e., acquired) resistance is a common occurrence in cancer patients and is often associated with high numbers of T regulatory (Treg) cells (CD4+CD25+FOXP3+). The approval of ipilimumab and the development of similar pharmacological agents targeting cell surface proteins on Treg cells demonstrates that such intervention may overcome resistance in cancer patients. Hence, the clinical development and subsequent approval of Cytotoxic T Lymphocyte Antigen-4 (CTLA-4) targeting agents can serve as a prototype for similar agents. Such new agents aspire to be highly specific and have a reduced toxicity profile while increasing effector T cell function or effector T/T regulatory (Teff/Treg) ratio. While clinical development with large molecules has shown the greatest advancement, small molecule inhibitors that target immunomodulation are increasingly entering early clinical investigation. These new small molecule inhibitors often target specific intracellular signaling pathways [e.g., phosphoinositide-3-kinase delta (PI3K-δ)] that play an important role in regulating the function of Treg cells. This review will summarize the lessons currently applied to develop novel clinical agents that target Treg cells.
Collapse
Affiliation(s)
- Pavlina Spiliopoulou
- Department of Drug Development Program, Phase I Unit, Beatson West of Scotland Cancer Center, Glasgow G12 0YN, UK
- School of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, UK
| | - Paramjit Kaur
- Department of Oncology Clinical Development, iOnctura SA, Geneva 1202, Switzerland
| | - Tracey Hammett
- Department of Oncology Clinical Development, iOnctura SA, Geneva 1202, Switzerland
| | - Giusy Di Conza
- Department of Oncology Clinical Development, iOnctura SA, Geneva 1202, Switzerland
| | - Michael Lahn
- Department of Oncology Clinical Development, iOnctura SA, Geneva 1202, Switzerland
| |
Collapse
|
8
|
Wang X, Geng S, Meng J, Kang N, Liu X, Xu Y, Lyu H, Xu Y, Xu X, Song X, Zhang B, Wang X, Nuerbulati N, Zhang Z, Zhai D, Mao X, Sun R, Wang X, Wang R, Guo J, Chen SW, Zhou X, Xia T, Qi H, Hu X, Shi Y. Foxp3-mediated blockage of ryanodine receptor 2 underlies contact-based suppression by regulatory T cells. J Clin Invest 2023; 133:e163470. [PMID: 38099494 PMCID: PMC10721146 DOI: 10.1172/jci163470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/10/2023] [Indexed: 12/18/2023] Open
Abstract
The suppression mechanism of Tregs remains an intensely investigated topic. As our focus has shifted toward a model centered on indirect inhibition of DCs, a universally applicable effector mechanism controlled by the transcription factor forkhead box P3 (Foxp3) expression has not been found. Here, we report that Foxp3 blocked the transcription of ER Ca2+-release channel ryanodine receptor 2 (RyR2). Reduced RyR2 shut down basal Ca2+ oscillation in Tregs, which reduced m-calpain activities that are needed for T cells to disengage from DCs, suggesting a persistent blockage of DC antigen presentation. RyR2 deficiency rendered the CD4+ T cell pool immune suppressive and caused it to behave in the same manner as Foxp3+ Tregs in viral infection, asthma, hypersensitivity, colitis, and tumor development. In the absence of Foxp3, Ryr2-deficient CD4+ T cells rescued the systemic autoimmunity associated with scurfy mice. Therefore, Foxp3-mediated Ca2+ signaling inhibition may be a central effector mechanism of Treg immune suppression.
Collapse
Affiliation(s)
- Xiaobo Wang
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Medicine, Tsinghua University, Beijing, China
| | - Shuang Geng
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute, University of Calgary, Calgary, Alberta, Canada
| | - Junchen Meng
- Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Medicine, Tsinghua University, Beijing, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, and
| | - Ning Kang
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Medicine, Tsinghua University, Beijing, China
| | - Xinyi Liu
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Medicine, Tsinghua University, Beijing, China
| | - Yanni Xu
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Medicine, Tsinghua University, Beijing, China
| | - Huiyun Lyu
- Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Ying Xu
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Medicine, Tsinghua University, Beijing, China
| | - Xun Xu
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Medicine, Tsinghua University, Beijing, China
| | - Xinrong Song
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Medicine, Tsinghua University, Beijing, China
| | - Bin Zhang
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Medicine, Tsinghua University, Beijing, China
| | - Xin Wang
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Nuerdida Nuerbulati
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Ze Zhang
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Medicine, Tsinghua University, Beijing, China
| | - Di Zhai
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Medicine, Tsinghua University, Beijing, China
| | - Xin Mao
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Medicine, Tsinghua University, Beijing, China
| | - Ruya Sun
- Department of Basic Medical Sciences, School of Medicine, and
| | - Xiaoting Wang
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University and Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu, China
| | - Ruiwu Wang
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Jie Guo
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - S.R. Wayne Chen
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Xuyu Zhou
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Tie Xia
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Medicine, Tsinghua University, Beijing, China
| | - Hai Qi
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- Collaborative Innovation Center for Biotherapy, Tsinghua University, Beijing, China
| | - Xiaoyu Hu
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- Collaborative Innovation Center for Biotherapy, Tsinghua University, Beijing, China
| | - Yan Shi
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Medicine, Tsinghua University, Beijing, China
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute, University of Calgary, Calgary, Alberta, Canada
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- Collaborative Innovation Center for Biotherapy, Tsinghua University, Beijing, China
| |
Collapse
|
9
|
Bei KF, Moshkelgosha S, Liu BJ, Juvet S. Intragraft regulatory T cells in the modern era: what can high-dimensional methods tell us about pathways to allograft acceptance? Front Immunol 2023; 14:1291649. [PMID: 38077395 PMCID: PMC10701590 DOI: 10.3389/fimmu.2023.1291649] [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: 09/09/2023] [Accepted: 10/31/2023] [Indexed: 12/18/2023] Open
Abstract
Replacement of diseased organs with transplanted healthy donor ones remains the best and often only treatment option for end-stage organ disease. Immunosuppressants have decreased the incidence of acute rejection, but long-term survival remains limited. The broad action of current immunosuppressive drugs results in global immune impairment, increasing the risk of cancer and infections. Hence, achievement of allograft tolerance, in which graft function is maintained in the absence of global immunosuppression, has long been the aim of transplant clinicians and scientists. Regulatory T cells (Treg) are a specialized subset of immune cells that control a diverse array of immune responses, can prevent allograft rejection in animals, and have recently been explored in early phase clinical trials as an adoptive cellular therapy in transplant recipients. It has been established that allograft residency by Tregs can promote graft acceptance, but whether intragraft Treg functional diversification and spatial organization contribute to this process is largely unknown. In this review, we will explore what is known regarding the properties of intragraft Tregs during allograft acceptance and rejection. We will summarize recent advances in understanding Treg tissue residency through spatial, transcriptomic and high-dimensional cytometric methods in both animal and human studies. Our discussion will explore properties of intragraft Tregs in mediating operational tolerance to commonly transplanted solid organs. Finally, given recent developments in Treg cellular therapy, we will review emerging knowledge of whether and how these adoptively transferred cells enter allografts in humans. An understanding of the properties of intragraft Tregs will help lay the foundation for future therapies that will promote immune tolerance.
Collapse
Affiliation(s)
- Ke Fan Bei
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Sajad Moshkelgosha
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Bo Jie Liu
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Stephen Juvet
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
10
|
Bousso P, Grandjean CL. Immunomodulation under the lens of real-time in vivo imaging. Eur J Immunol 2023; 53:e2249921. [PMID: 37051691 DOI: 10.1002/eji.202249921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/16/2022] [Accepted: 04/11/2023] [Indexed: 04/14/2023]
Abstract
Modulation of cells and molecules of the immune system not only represents a major opportunity to treat a variety of diseases including infections, cancer, autoimmune, and inflammatory disorders but could also help understand the intricacies of immune responses. A detailed mechanistic understanding of how a specific immune intervention may provide clinical benefit is essential for the rational design of efficient immunomodulators. Visualizing the impact of immunomodulation in real-time and in vivo has emerged as an important approach to achieve this goal. In this review, we aim to illustrate how multiphoton intravital imaging has helped clarify the mode of action of immunomodulatory strategies such as antibodies or cell therapies. We also discuss how optogenetics combined with imaging will further help manipulate and precisely understand immunomodulatory pathways. Combined with other single-cell technologies, in vivo dynamic imaging has therefore a major potential for guiding preclinical development of immunomodulatory drugs.
Collapse
Affiliation(s)
- Philippe Bousso
- Dynamics of Immune Responses Unit, Institut Pasteur, INSERM U1223, Université de Paris Cité, Paris, France
| | - Capucine L Grandjean
- Dynamics of Immune Responses Unit, Institut Pasteur, INSERM U1223, Université de Paris Cité, Paris, France
| |
Collapse
|
11
|
Liang C, He J, Zhao X, Hong J, Ma X, Mao M, Nie W, Wu G, Dong Y, Xu W, Huang L, Xie HY. Monitoring the Cascade of Tumor-specific Immune Response in vivo via Chemoenzymatic Proximity Labeling. Angew Chem Int Ed Engl 2023; 62:e202304838. [PMID: 37650228 DOI: 10.1002/anie.202304838] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/25/2023] [Accepted: 08/30/2023] [Indexed: 09/01/2023]
Abstract
Monitoring the highly dynamic and complex immune response remains a great challenge owing to the lack of reliable and specific approaches. Here, we develop a strategy to monitor the cascade of tumor immune response through the cooperation of pore-forming alginate gel with chemoenzymatic proximity-labeling. A macroporous gel containing tumor-associated antigens, adjuvants, and pro-inflammatory cytokines is utilized to recruit endogenous DCs and enhance their maturation in vivo. The mature DCs are then modified with GDP-fucose-fucosyltransferase (GDP-Fuc-Fuct) via the self-catalysis of fucosyltransferase (Fuct). Following the migration of the obtained Fuct-DCs to the draining lymph nodes (dLNs), the molecular recognition mediated interaction of DCs and T cells leads to the successful decoration of T cells with GDP-Fuc-azide through the Fuct catalyzed proximity-labeling. Therefore, the activated tumor-specific T cells in dLNs and tumors can be identified through bioorthogonal labeling, opening up a new avenue for studying the immune mechanism of tumors in situ.
Collapse
Affiliation(s)
- Chao Liang
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Jiaqi He
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Xin Zhao
- School of Material Science & Engineering, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Jie Hong
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Xianbin Ma
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Mingchuan Mao
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Weidong Nie
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Guanghao Wu
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Yuping Dong
- School of Material Science & Engineering, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Wei Xu
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Lili Huang
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Hai-Yan Xie
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Chemical Biology Center, Peking University, Beijing, 100191, P. R. China
| |
Collapse
|
12
|
Sabatel C, Bureau F. The innate immune brakes of the lung. Front Immunol 2023; 14:1111298. [PMID: 36776895 PMCID: PMC9915150 DOI: 10.3389/fimmu.2023.1111298] [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: 11/29/2022] [Accepted: 01/02/2023] [Indexed: 01/29/2023] Open
Abstract
Respiratory mucosal surfaces are continuously exposed to not only innocuous non-self antigens but also pathogen-associated molecular patterns (PAMPs) originating from environmental or symbiotic microbes. According to either "self/non-self" or "danger" models, this should systematically result in homeostasis breakdown and the development of immune responses directed to inhaled harmless antigens, such as T helper type (Th)2-mediated asthmatic reactions, which is fortunately not the case in most people. This discrepancy implies the existence, in the lung, of regulatory mechanisms that tightly control immune homeostasis. Although such mechanisms have been poorly investigated in comparison to the ones that trigger immune responses, a better understanding of them could be useful in the development of new therapeutic strategies against lung diseases (e.g., asthma). Here, we review current knowledge on innate immune cells that prevent the development of aberrant immune responses in the lung, thereby contributing to mucosal homeostasis.
Collapse
Affiliation(s)
- Catherine Sabatel
- Laboratory of Cellular and Molecular Immunology, GIGA-Research, University of Liège, Liège, Belgium,Faculty of Veterinary Medicine, University of Liège, Liège, Belgium,*Correspondence: Catherine Sabatel,
| | - Fabrice Bureau
- Laboratory of Cellular and Molecular Immunology, GIGA-Research, University of Liège, Liège, Belgium,Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| |
Collapse
|
13
|
Qian H, Beltran AS. Mesoscience in cell biology and cancer research. CANCER INNOVATION 2022; 1:271-284. [PMID: 38089088 PMCID: PMC10686186 DOI: 10.1002/cai2.33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 10/15/2024]
Abstract
Mesoscale characteristics and their interdimensional correlation are the focus of contemporary interdisciplinary research. Mesoscience is a discipline that has the potential to radically update the existing knowledge structure, which differs from the conventional unit-scale and system-scale research models, revealing a previously untouchable area for scientific research. Integrative biology research aims to dissect the complex problems of life systems by conducting comprehensive research and integrating various disciplines from all biological levels of the living organism. However, the mesoscientific issues between different research units are neglected and challenging. Mesoscale research in biology requires the integration of research theories and methods from other disciplines (mathematics, physics, engineering, and even visual imaging) to investigate theoretical and frontier questions of biological processes through experiments, computations, and modeling. We reviewed integrative paradigms and methods for the biological mesoscale problems (focusing on oncology research) and prospected the potential of their multiple dimensions and upcoming challenges. We expect to establish an interactive and collaborative theoretical platform for further expanding the depth and width of our understanding on the nature of biology.
Collapse
Affiliation(s)
- Haili Qian
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Adriana Sujey Beltran
- Department of Pharmacology, University of North Carolina at Chapel HillChapel HillNCUSA
| |
Collapse
|
14
|
Fathi M, Razavi SM, Sojoodi M, Ahmadi A, Ebrahimi F, Namdar A, Hojjat-Farsangi M, Gholamin S, Jadidi-Niaragh F. Targeting the CTLA-4/B7 axes in glioblastoma: preclinical evidence and clinical interventions. Expert Opin Ther Targets 2022; 26:949-961. [PMID: 36527817 DOI: 10.1080/14728222.2022.2160703] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Glioblastoma Multiforme (GBM) is one of the fatal cancers of the Central Nervous System (CNS). A variety of reasons exist for why previous immunotherapy strategies, especially Immune Checkpoint Blockers (ICBs), did not work in treating GBM patients. The cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) is a key immune checkpoint receptor. Its overexpression in cancer and immune cells causes tumor cell progression. CTLA-4 suppresses anti-tumor responses inside the GBM tumor-immune microenvironment. AREAS COVERED It has been attempted to explain the immunobiology of CTLA-4 as well as its interaction with different immune cells and cancer cells that lead to GBM progression. Additionally, CTLA-4 targeting studies have been reviewed and CTLA-4 combination therapy, as a promising therapeutic target and strategy for GBM immunotherapy, is recommended. EXPERT OPINION CTLA-4 could be a possible supplement for future cancer immunotherapies of GBM. However, many challenges remain such as the high toxicity of CTLA-4 blockers, and the unresponsiveness of most patients to immunotherapy. For the future clinical success of CTLA-4 blocker therapy, combination approaches with other targeted treatments would be a potentially effective strategy. Going forward, predictive biomarkers can be used to reduce trial timelines and increase the chance of success.
Collapse
Affiliation(s)
- Mehrdad Fathi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed-Mostafa Razavi
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Mozhdeh Sojoodi
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Armin Ahmadi
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, AL, USA
| | - Farbod Ebrahimi
- Nanoparticle Process Technology, Faculty of Engineering, University of Duisburg-Essen, Duisburg, Germany
| | - Afshin Namdar
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | | | - Sharareh Gholamin
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW The purpose of this narrative review is to give an overview about the effects of multimodal prehabilitation and current existing and prospectively planned studies. The potential efficacy of exercise in the context of prehabilitation ranges from preoperatively improving patients' functional capacity to inducing cellular mechanisms that affect organ perfusion via endothelial regeneration, anti-inflammatory processes and tumour defense. RECENT FINDINGS Current studies show that prehabilitation is capable of reducing certain postoperative complications and length of hospital stay in certain patient populations. These findings are based on small to mid-size trials with large heterogeneity, lacking generalizability and evidence that prehabilitation has positive effects on long term survival. SUMMARY The concept of prehabilitation contains the features, namely preoperative exercise, nutritional intervention and psychological support. Preoperative exercise holds potential molecular effects that can be utilized in the perioperative period in order to improve patients' postoperative outcome. Future multimodal prehabilitation trials must specifically clarify the clinical impact of this concept on patients' quality of life after major cancer surgery and cancer-specific survival.
Collapse
Affiliation(s)
- Tobias Esser
- Institute of Sports and Sports Medicine, TU Dortmund University, Dortmund
| | - Philipp Zimmer
- Institute of Sports and Sports Medicine, TU Dortmund University, Dortmund
| | - Robert Schier
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department for Anaesthesiology and Intensive Care Medicine, Cologne, Germany
| |
Collapse
|
16
|
van Pul KM, Notohardjo JCL, Fransen MF, Koster BD, Stam AGM, Chondronasiou D, Lougheed SM, Bakker J, Kandiah V, van den Tol MP, Jooss K, Vuylsteke RJCLM, van den Eertwegh AJM, de Gruijl TD. Local delivery of low-dose anti–CTLA-4 to the melanoma lymphatic basin leads to systemic T
reg
reduction and effector T cell activation. Sci Immunol 2022; 7:eabn8097. [DOI: 10.1126/sciimmunol.abn8097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Preclinical studies show that locoregional CTLA-4 blockade is equally effective in inducing tumor eradication as systemic delivery, without the added risk of immune-related side effects. This efficacy is related to access of the CTLA-4 blocking antibodies to tumor-draining lymph nodes (TDLNs). Local delivery of anti–CTLA-4 after surgical removal of primary melanoma, before sentinel lymph node biopsy (SLNB), provides a unique setting to clinically assess the role of TDLN in the biological efficacy of locoregional CTLA-4 blockade. Here, we have evaluated the safety, tolerability, and immunomodulatory effects in the SLN and peripheral blood of a single dose of tremelimumab [a fully human immunoglobulin gamma-2 (IgG2) mAb directed against CTLA-4] in a dose range of 2 to 20 mg, injected intradermally at the tumor excision site 1 week before SLNB in 13 patients with early-stage melanoma (phase 1 trial; NCT04274816). Intradermal delivery was safe and well tolerated and induced activation of migratory dendritic cell (DC) subsets in the SLN. It also induced profound and durable decreases in regulatory T cell (T
reg
) frequencies and activation of effector T cells in both SLN and peripheral blood. Moreover, systemic T cell responses against NY-ESO-1 or MART-1 were primed or boosted (
N
= 7), in association with T cell activation and central memory T cell differentiation. These findings indicate that local administration of anti–CTLA-4 may offer a safe and promising adjuvant treatment strategy for patients with early-stage melanoma. Moreover, our data demonstrate a central role for TDLN in the biological efficacy of CTLA-4 blockade and support TDLN-targeted delivery methods.
Collapse
Affiliation(s)
- Kim M. van Pul
- Amsterdam UMC location Vrije Universiteit, Medical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Amsterdam UMC location Vrije Universiteit, Surgical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| | - Jessica C. L. Notohardjo
- Amsterdam UMC location Vrije Universiteit, Medical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| | - Marieke F. Fransen
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam UMC location Vrije Universiteit, Pulmonary Diseases, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
| | - Bas D. Koster
- Amsterdam UMC location Vrije Universiteit, Medical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| | - Anita G. M. Stam
- Amsterdam UMC location Vrije Universiteit, Medical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| | - Dafni Chondronasiou
- Amsterdam UMC location Vrije Universiteit, Medical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| | - Sinéad M. Lougheed
- Amsterdam UMC location Vrije Universiteit, Medical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| | - Joyce Bakker
- Amsterdam UMC location Vrije Universiteit, Medical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| | - Vinitha Kandiah
- Amsterdam UMC location Vrije Universiteit, Medical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| | - M. Petrousjka van den Tol
- Amsterdam UMC location Vrije Universiteit, Surgical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| | | | | | - Alfons J. M. van den Eertwegh
- Amsterdam UMC location Vrije Universiteit, Medical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| | - Tanja D. de Gruijl
- Amsterdam UMC location Vrije Universiteit, Medical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| |
Collapse
|
17
|
Lam AJ, Haque M, Ward-Hartstonge KA, Uday P, Wardell CM, Gillies JK, Speck M, Mojibian M, Klein Geltink RI, Levings MK. PTEN is required for human Treg suppression of costimulation in vitro. Eur J Immunol 2022; 52:1482-1497. [PMID: 35746855 DOI: 10.1002/eji.202249888] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/18/2022] [Accepted: 06/10/2022] [Indexed: 11/07/2022]
Abstract
Regulatory T cell (Treg) therapy is under clinical investigation for the treatment of transplant rejection, autoimmune disease, and graft-versus-host disease. With the advent of genome editing, attention has turned to reinforcing Treg function for therapeutic benefit. A hallmark of Tregs is dampened activation of PI3K-AKT signalling, of which PTEN is a major negative regulator. Loss-of-function studies of PTEN, however, have not conclusively shown a requirement for PTEN in upholding Treg function and stability. Using CRISPR-based genome editing in human Tregs, we show that PTEN ablation does not cause a global defect in Treg function and stability; rather, it selectively blocks their ability to suppress antigen-presenting cells. PTEN-KO Tregs exhibit elevated glycolytic activity, upregulate FOXP3, maintain a Treg phenotype, and have no discernable defects in lineage stability. Functionally, PTEN is dispensable for human Treg-mediated inhibition of T cell activity in vitro and in vivo, but is required for suppression of costimulatory molecule expression by antigen-presenting cells. These data are the first to define a role for a signalling pathway in controlling a subset of human Treg activity. Moreover, they point to the functional necessity of PTEN-regulated PI3K-AKT activity for optimal human Treg function. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Avery J Lam
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Manjurul Haque
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Kirsten A Ward-Hartstonge
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Prakruti Uday
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Christine M Wardell
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Jana K Gillies
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Madeleine Speck
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Majid Mojibian
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Ramon I Klein Geltink
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada.,Department of Molecular Oncology, BC Cancer Research, Vancouver, BC, V5Z 1L3, Canada
| | - Megan K Levings
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, V6T 1Z3, Canada
| |
Collapse
|
18
|
Wan H, Zhou H, Feng Y, Chen Y, Zhu L, Mi Y. Comprehensive Analysis of 29,464 Cancer Cases and 35,858 Controls to Investigate the Effect of the Cytotoxic T-Lymphocyte Antigen 4 Gene rs231775 A/G Polymorphism on Cancer Risk. Front Oncol 2022; 12:878507. [PMID: 35600409 PMCID: PMC9114750 DOI: 10.3389/fonc.2022.878507] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/28/2022] [Indexed: 12/20/2022] Open
Abstract
In our previous studies, we found that the rs231775 polymorphism of cytotoxic T-lymphocyte antigen 4 (CTLA-4) is associated with risks of different cancer types; however, the association remains controversial and ambiguous, so we conducted an in-depth meta-analysis to verify the association. A complete search of the PubMed, Google Scholar, Embase, Chinese databases, and Web of Science was conducted without regard to language limitations, covering all publications since November 20, 2021. The search criteria for cancer susceptibility associated with the polymorphism in the CTLA-4 gene rs231775 resulted in 87 case-control studies with 29,464 cases and 35,858 controls. The association strength was analyzed using odds ratios and 95% confidence intervals. Overall, we found that the CTLA-4 rs231775 polymorphism may reduce cancer risk. A stratified cancer type analysis showed that CTLA-4 rs231775 polymorphism was a risk factor for colorectal cancer and thyroid cancer; on the other hand, it was a protective factor for breast cancer, liver cancer, cervical cancer, bone cancer, head and neck, and pancreatic cancer. We also classified cancer into five systems and observed an increased association with digestive tract cancer, decreased associations with orthopedic tumors, tumors of the urinary system, and gynecological tumors. In the subgroup based on race, decreased relationships were observed in both Asians and Caucasians. The same decreased association was also shown in the analysis of the source of control analysis. Our present study indicates that the CTLA-4 rs231775 polymorphism contributes to cancer development and aggression.
Collapse
Affiliation(s)
- Hongyuan Wan
- Wuxi Medical College, Jiangnan University, Wuxi, China
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Hangsheng Zhou
- Wuxi Medical College, Jiangnan University, Wuxi, China
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yanyan Feng
- Wuxi Medical College, Jiangnan University, Wuxi, China
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yongquan Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Lijie Zhu
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yuanyuan Mi
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
19
|
Prodjinotho UF, Gres V, Henkel F, Lacorcia M, Dandl R, Haslbeck M, Schmidt V, Winkler AS, Sikasunge C, Jakobsson PJ, Henneke P, Esser-von Bieren J, Prazeres da Costa C. Helminthic dehydrogenase drives PGE 2 and IL-10 production in monocytes to potentiate Treg induction. EMBO Rep 2022; 23:e54096. [PMID: 35357743 PMCID: PMC9066053 DOI: 10.15252/embr.202154096] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 03/02/2022] [Accepted: 03/14/2022] [Indexed: 01/03/2023] Open
Abstract
Immunoregulation of inflammatory, infection‐triggered processes in the brain constitutes a central mechanism to control devastating disease manifestations such as epilepsy. Observational studies implicate the viability of Taenia solium cysts as key factor determining severity of neurocysticercosis (NCC), the most common cause of epilepsy, especially in children, in Sub‐Saharan Africa. Viable, in contrast to decaying, cysts mostly remain clinically silent by yet unknown mechanisms, potentially involving Tregs in controlling inflammation. Here, we show that glutamate dehydrogenase from viable cysts instructs tolerogenic monocytes to release IL‐10 and the lipid mediator PGE2. These act in concert, converting naive CD4+ T cells into CD127−CD25hiFoxP3+CTLA‐4+ Tregs, through the G protein‐coupled receptors EP2 and EP4 and the IL‐10 receptor. Moreover, while viable cyst products strongly upregulate IL‐10 and PGE2 transcription in microglia, intravesicular fluid, released during cyst decay, induces pro‐inflammatory microglia and TGF‐β as potential drivers of epilepsy. Inhibition of PGE2 synthesis and IL‐10 signaling prevents Treg induction by viable cyst products. Harnessing the PGE2‐IL‐10 axis and targeting TGF‐ß signaling may offer an important therapeutic strategy in inflammatory epilepsy and NCC.
Collapse
Affiliation(s)
- Ulrich Fabien Prodjinotho
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine, Technical University of Munich (TUM), Munich, Germany.,Center for Global Health, TUM School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Vitka Gres
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Fiona Henkel
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Matthew Lacorcia
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Ramona Dandl
- Department of Chemistry, Technical University Munich (TUM), Garching, Germany
| | - Martin Haslbeck
- Department of Chemistry, Technical University Munich (TUM), Garching, Germany
| | - Veronika Schmidt
- Center for Global Health, TUM School of Medicine, Technical University of Munich (TUM), Munich, Germany.,Department of Neurology, University Hospital, Klinikum rechts der Isar, Technical University Munich (TUM), Munich, Germany.,Center for Global Health, Institute of Health and Society, University of Oslo, Oslo, Norway
| | - Andrea Sylvia Winkler
- Center for Global Health, TUM School of Medicine, Technical University of Munich (TUM), Munich, Germany.,Department of Neurology, University Hospital, Klinikum rechts der Isar, Technical University Munich (TUM), Munich, Germany.,Center for Global Health, Institute of Health and Society, University of Oslo, Oslo, Norway
| | - Chummy Sikasunge
- Department of Paraclinicals, School of Veterinary Medicine, University of Zambia, Lusaka, Zambia
| | - Per-Johan Jakobsson
- Rheumatology Unit, Department of Medicine, Solna, Karolinska University Hospital, Stockholm, Sweden
| | - Philipp Henneke
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Pediatrics and Adolescent Medicine, Medical Center, University of Freiburg, Freiburg, Germany.,Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Julia Esser-von Bieren
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Clarissa Prazeres da Costa
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine, Technical University of Munich (TUM), Munich, Germany.,Center for Global Health, TUM School of Medicine, Technical University of Munich (TUM), Munich, Germany.,German Center for Infection and Research (DZIF), Munich, Germany
| |
Collapse
|
20
|
Gudi RR, Perez N, Karumuthil-Melethil S, Li G, Vasu C. Activation of T cell checkpoint pathways during β-cell antigen presentation by engineered dendritic cells promotes protection from type 1 diabetes. Immunology 2022; 166:341-356. [PMID: 35404483 DOI: 10.1111/imm.13476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/27/2021] [Accepted: 03/28/2022] [Indexed: 11/29/2022] Open
Abstract
Defective immune regulation has been recognized in type 1 diabetes (T1D). Immune regulatory T cell check-point receptors, which are generally upregulated on activated T cells, have been the molecules of attention as therapeutic targets for enhancing immune response in tumor therapy. Here, we show that pancreatic β-cell antigen (BcAg) presentation by engineered tolerogenic dendritic cells (tDCs) that express CTLA4 selective ligand (B7.1wa) or a combination of CTLA4, PD1 and BTLA selective ligands (B7.1wa, PD-L1, and HVEM-CRD1 respectively; multiligand-DCs) causes an increase in regulatory cytokine and T cell (Treg) responses and suppression of the effector T cell function as compared to engineered control-DCs. Non-obese diabetic (NOD) mice treated with BcAg-pulsed CTLA4-ligand-DCs and multiligand-DCs at pre-diabetic and early-hyperglycemic stages showed significantly lower degree of insulitis, higher frequencies of insulin-positive islets, profound delay in, and reversal of, hyperglycemia for a significant duration. Immune cells from the tDC treated mice not only produced lower amounts of IFNγ and higher amounts of IL10 and TGFβ1 upon BcAg challenge, but also failed to induce hyperglycemia upon adoptive transfer. While both CTLA4-ligand-DCs and multiligand-DCs were effective in inducing tolerance, multiligand-DC treatment produced an overall higher suppressive effect on effector T cell function and disease outcome. These studies show that enhanced engagement of T cell checkpoint receptors during BcAg presentation can modulate T cell function and suppress autoimmunity and progression of the disease in T1D.
Collapse
Affiliation(s)
- Radhika R Gudi
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston
| | - Nicolas Perez
- Department of Surgery, College of Medicine, University of Illinois, Chicago, IL
| | | | - Gongbo Li
- Department of Surgery, College of Medicine, University of Illinois, Chicago, IL
| | - Chenthamarakshan Vasu
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston.,Department of Surgery, College of Medicine, University of Illinois, Chicago, IL
| |
Collapse
|
21
|
Dong J, Huang Y, Zhou Z, Sun M. Breaking Immunosuppressive Barriers by Engineered Nanoplatforms for Turning Cold Tumor to Hot. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Jingwen Dong
- State Key Laboratory of Natural Medicines Department of Pharmaceutics China Pharmaceutical University Nanjing 210009 P. R. China
| | - Ying Huang
- State Key Laboratory of Natural Medicines Department of Pharmaceutics China Pharmaceutical University Nanjing 210009 P. R. China
| | - Zhanwei Zhou
- State Key Laboratory of Natural Medicines Department of Pharmaceutics China Pharmaceutical University Nanjing 210009 P. R. China
| | - Minjie Sun
- State Key Laboratory of Natural Medicines Department of Pharmaceutics China Pharmaceutical University Nanjing 210009 P. R. China
| |
Collapse
|
22
|
Jacobelli J, Buser AE, Heiden DL, Friedman RS. Autoimmunity in motion: Mechanisms of immune regulation and destruction revealed by in vivo imaging. Immunol Rev 2022; 306:181-199. [PMID: 34825390 PMCID: PMC9135487 DOI: 10.1111/imr.13043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/06/2021] [Indexed: 11/30/2022]
Abstract
Autoimmunity arises when mechanisms of immune tolerance fail. Here we discuss mechanisms of T cell activation and tolerance and the dynamics of the autoimmune response at the site of disease. Live imaging of autoimmunity provides the ability to analyze immune cell dynamics at the single-cell level within the complex intact environment where disease occurs. These analyses have revealed mechanisms of T cell activation and tolerance in the lymph nodes, mechanisms of T cell entry into sites of autoimmune disease, and mechanisms leading to pathogenesis or protection in the autoimmune lesions. The overarching conclusions point to stable versus transient T cell antigen presenting cell interactions dictating the balance between T cell activation and tolerance, and T cell restimulation as a driver of pathogenesis at the site of autoimmunity. Findings from models of multiple sclerosis and type 1 diabetes are highlighted, however, the results have implications for basic mechanisms of T cell regulation during immune responses, tumor immunity, and autoimmunity.
Collapse
Affiliation(s)
- Jordan Jacobelli
- Barbara Davis Center for Diabetes, Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Alan E. Buser
- Barbara Davis Center for Diabetes, Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Dustin L. Heiden
- Barbara Davis Center for Diabetes, Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Rachel S. Friedman
- Barbara Davis Center for Diabetes, Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
23
|
Wong HS, Germain RN. Mesoscale T cell antigen discrimination emerges from intercellular feedback. Trends Immunol 2021; 42:865-875. [PMID: 34493455 DOI: 10.1016/j.it.2021.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/13/2021] [Accepted: 08/13/2021] [Indexed: 12/14/2022]
Abstract
Mature T cells must distinguish between foreign and self-antigens to promote host defense while limiting autoimmunity. How such discrimination occurs reproducibly has been explored extensively regarding mechanisms regulating initial T cell activation at short time and length scales. Here, we suggest that T cells encounter a higher-level discriminatory boundary post-activation, empowering or constraining their responses over greater spatiotemporal scales. This boundary emerges from coordinated communication among at least three cell types, forming a control system governed by intercellular circuits, including negative feedback from regulatory T cells (Tregs). We propose that the nonlinearities inherent to this system can amplify subtle baseline imbalances in a single cell type's functional state, altering the threshold for productive T cell responses and autoimmune disease risk.
Collapse
Affiliation(s)
- Harikesh S Wong
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1892, USA.
| | - Ronald N Germain
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1892, USA.
| |
Collapse
|
24
|
Treg-expressed CTLA-4 depletes CD80/CD86 by trogocytosis, releasing free PD-L1 on antigen-presenting cells. Proc Natl Acad Sci U S A 2021; 118:2023739118. [PMID: 34301886 DOI: 10.1073/pnas.2023739118] [Citation(s) in RCA: 183] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Foxp3-expressing CD4+CD25+ regulatory T cells (Tregs) constitutively and highly express the immune checkpoint receptor cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), whose Treg-specific deficiency causes severe systemic autoimmunity. As a key mechanism of Treg-mediated suppression, Treg-expressed CTLA-4 down-regulates the expression of CD80/CD86 costimulatory molecules on antigen-presenting cells (APCs). Here, we show that Treg-expressed CTLA-4 facilitated Treg-APC conjugation and immune synapse formation. The immune synapses thus formed provided a stable platform whereby Tregs were able to deplete CD80/CD86 molecules on APCs by extracting them via CTLA-4-dependent trogocytosis. The depletion occurred even with Tregs solely expressing a mutant CTLA-4 form lacking the cytoplasmic portion required for its endocytosis. The CTLA-4-dependent trogocytosis of CD80/CD86 also accelerated in vitro and in vivo passive transfer of other membrane proteins and lipid molecules from APCs to Tregs without their significant reduction on the APC surface. Furthermore, CD80 down-regulation or blockade by Treg-expressed membrane CTLA-4 or soluble CTLA-4-immunoglobulin (CTLA-4-Ig), respectively, disrupted cis-CD80/programmed death ligand-1 (PD-L1) heterodimers and increased free PD-L1 on dendritic cells (DCs), expanding a phenotypically distinct population of CD80lo free PD-L1hi DCs. Thus, Tregs are able to inhibit the T cell stimulatory activity of APCs by reducing their CD80/CD86 expression via CTLA-4-dependent trogocytosis. This CD80/CD86 reduction on APCs is able to exert dual suppressive effects on T cell immune responses by limiting CD80/CD86 costimulation to naïve T cells and by increasing free PD-L1 available for the inhibition of programmed death-1 (PD-1)-expressing effector T cells. Blockade of CTLA-4 and PD-1/PD-L1 in combination may therefore synergistically hinder Treg-mediated immune suppression, thereby effectively enhancing immune responses, including tumor immunity.
Collapse
|
25
|
Jairaman A, Othy S, Dynes JL, Yeromin AV, Zavala A, Greenberg ML, Nourse JL, Holt JR, Cahalan SM, Marangoni F, Parker I, Pathak MM, Cahalan MD. Piezo1 channels restrain regulatory T cells but are dispensable for effector CD4 + T cell responses. SCIENCE ADVANCES 2021; 7:7/28/eabg5859. [PMID: 34233878 PMCID: PMC8262815 DOI: 10.1126/sciadv.abg5859] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/24/2021] [Indexed: 05/03/2023]
Abstract
T lymphocytes encounter complex mechanical cues during an immune response. The mechanosensitive ion channel, Piezo1, drives inflammatory responses to bacterial infections, wound healing, and cancer; however, its role in helper T cell function remains unclear. In an animal model for multiple sclerosis, experimental autoimmune encephalomyelitis (EAE), we found that mice with genetic deletion of Piezo1 in T cells showed diminished disease severity. Unexpectedly, Piezo1 was not essential for lymph node homing, interstitial motility, Ca2+ signaling, T cell proliferation, or differentiation into proinflammatory T helper 1 (TH1) and TH17 subsets. However, Piezo1 deletion in T cells resulted in enhanced transforming growth factor-β (TGFβ) signaling and an expanded pool of regulatory T (Treg) cells. Moreover, mice with deletion of Piezo1 specifically in Treg cells showed significant attenuation of EAE. Our results indicate that Piezo1 selectively restrains Treg cells, without influencing activation events or effector T cell functions.
Collapse
Affiliation(s)
- Amit Jairaman
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
| | - Shivashankar Othy
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
| | - Joseph L Dynes
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
| | - Andriy V Yeromin
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
| | - Angel Zavala
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
| | - Milton L Greenberg
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
| | - Jamison L Nourse
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697, USA
| | - Jesse R Holt
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697, USA
| | - Stuart M Cahalan
- Howard Hughes Medical Institute, Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA
- Vertex Pharmaceuticals, 3215 Merryfield Row, San Diego, CA 92121, USA
| | - Francesco Marangoni
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
| | - Ian Parker
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Medha M Pathak
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697, USA
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
- Center for Complex Systems Biology, University of California, Irvine, CA 92697, USA
| | - Michael D Cahalan
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697-4561, USA.
- Institute for Immunology, University of California, Irvine, CA 92697, USA
| |
Collapse
|
26
|
Yu K, Hammerschmidt SI, Permanyer M, Galla M, Rothe M, Zheng X, Werth K, Martens R, Lueder Y, Janssen A, Friedrichsen M, Bernhardt G, Förster R. Targeted delivery of regulatory macrophages to lymph nodes interferes with T cell priming by preventing the formation of stable immune synapses. Cell Rep 2021; 35:109273. [PMID: 34161766 DOI: 10.1016/j.celrep.2021.109273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 02/05/2021] [Accepted: 05/27/2021] [Indexed: 02/06/2023] Open
Abstract
Immunosuppressive myeloid cells are frequently induced in tumors and attenuate anti-tumor effector functions. In this study, we differentiate immunosuppressive regulatory macrophages (Mregs) from hematopoietic progenitors and test their potential to suppress adaptive immune responses in lymph nodes. Targeted delivery of Mregs to lymph nodes is facilitated by retroviral overexpression of the chemokine receptor CCR7 and intra-lymphatic cell application. Delivery of Mregs completely abolishes the priming of cognate CD8 cells and strongly reduces delayed-type hypersensitivity reactions. Mreg-mediated T cell suppression requires cell-cell contact-regulated nitric oxide production. Two-photon microscopy reveals that nitric oxide produced by Mregs reduces the interaction duration between dendritic cells and T cells. Exposure of activated T cells to nitric oxide strongly reduces their binding to ICAM-1, indicating that nitrosylation of proteins involved in cell adhesion affects synapse formation. Thus, this study identifies a mechanism of myeloid cell-mediated immune suppression and provides an approach for its therapeutic use.
Collapse
Affiliation(s)
- Kai Yu
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | | | - Marc Permanyer
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Melanie Galla
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Michael Rothe
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Xiang Zheng
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Kathrin Werth
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Rieke Martens
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Yvonne Lueder
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Anika Janssen
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | | | - Günter Bernhardt
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany; German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 30625 Hannover, Germany.
| |
Collapse
|
27
|
Le Luyer J, Schull Q, Auffret P, Lopez P, Crusot M, Belliard C, Basset C, Carradec Q, Poulain J, Planes S, Saulnier D. Dual RNAseq highlights the kinetics of skin microbiome and fish host responsiveness to bacterial infection. Anim Microbiome 2021; 3:35. [PMID: 33962693 PMCID: PMC8106148 DOI: 10.1186/s42523-021-00097-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
Background Tenacibaculum maritimum is a fish pathogen known for causing serious damage to a broad range of wild and farmed marine fish populations worldwide. The recently sequenced genome of T. maritimum strain NCIMB 2154T provided unprecedented information on the possible molecular mechanisms involved in the virulence of this species. However, little is known about the dynamic of infection in vivo, and information is lacking on both the intrinsic host response (gene expression) and its associated microbiota. Here, we applied complementary omic approaches, including dual RNAseq and 16S rRNA gene metabarcoding sequencing using Nanopore and short-read Illumina technologies to unravel the host–pathogen interplay in an experimental infection system using the tropical fish Platax orbicularis as model. Results We showed that the infection of the host is characterised by an enhancement of functions associated with antibiotic and glucans catabolism functions but a reduction of sulfate assimilation process in T. maritimum. The fish host concurrently displays a large panel of immune effectors, notably involving innate response and triggering acute inflammatory response. In addition, our results suggest that fish activate an adaptive immune response visible through the stimulation of T-helper cells, Th17, with congruent reduction of Th2 and T-regulatory cells. Fish were, however, largely sensitive to infection, and less than 25% survived after 96 hpi. These surviving fish showed no evidence of stress (cortisol levels) or significant difference in microbiome diversity compared with controls at the same sampling time. The presence of T. maritimum in resistant fish skin and the total absence of any skin lesions suggest that these fish did not escape contact with the pathogen, but rather that some mechanisms prevented pathogens entry. In resistant individuals, we detected up-regulation of specific immune-related genes differentiating resistant individuals from controls at 96 hpi, which suggests a possible genomic basis of resistance, although no genetic variation in coding regions was found. Conclusion Here we focus in detail on the interplay between common fish pathogens and host immune response during experimental infection. We further highlight key actors of defence response, pathogenicity and possible genomic bases of fish resistance to T. maritimum. Supplementary Information The online version contains supplementary material available at 10.1186/s42523-021-00097-1.
Collapse
Affiliation(s)
- J Le Luyer
- Ifremer, IRD, Institut Louis-Malardé, Univ Polynésie Française, EIO, F-98719 Taravao, Tahiti, Polynésie Française.
| | - Q Schull
- Ifremer, IRD, Institut Louis-Malardé, Univ Polynésie Française, EIO, F-98719 Taravao, Tahiti, Polynésie Française.,MARBEC, Univ. Montpellier, Ifremer, IRD, CNRS, F-34200, Sète, France
| | - P Auffret
- Ifremer, IRD, Institut Louis-Malardé, Univ Polynésie Française, EIO, F-98719 Taravao, Tahiti, Polynésie Française
| | - P Lopez
- Ifremer, IRD, Institut Louis-Malardé, Univ Polynésie Française, EIO, F-98719 Taravao, Tahiti, Polynésie Française.,Virologie et Immunologie Moléculaires, Institut National de la Recherche Agronomique, Université Paris-Saclay, Jouy-en-Josas, France
| | - M Crusot
- Ifremer, IRD, Institut Louis-Malardé, Univ Polynésie Française, EIO, F-98719 Taravao, Tahiti, Polynésie Française.,Univ Polynésie française, Ifremer, IRD, Institut Louis-Malardé, EIO, F-98702 Fa, 'a, Tahiti, Polynésie Française
| | - C Belliard
- Ifremer, IRD, Institut Louis-Malardé, Univ Polynésie Française, EIO, F-98719 Taravao, Tahiti, Polynésie Française
| | - C Basset
- Ifremer, IRD, Institut Louis-Malardé, Univ Polynésie Française, EIO, F-98719 Taravao, Tahiti, Polynésie Française
| | - Q Carradec
- Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, Univ Evry, Université Paris-Saclay, 91057, Evry, France
| | - J Poulain
- Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, Univ Evry, Université Paris-Saclay, 91057, Evry, France
| | - S Planes
- PSL Research University: EPHE-UPVD-CNRS, USR 3278 CRIOBE, Moorea, Polynésie Française.,Laboratoire d'Excellence "CORAIL," USR 3278 CNRS-EPHE-UPVD CRIOBE, Perpignan, France
| | - D Saulnier
- Ifremer, IRD, Institut Louis-Malardé, Univ Polynésie Française, EIO, F-98719 Taravao, Tahiti, Polynésie Française
| |
Collapse
|
28
|
Zhai Y, Moosavi R, Chen M. Immune Checkpoints, a Novel Class of Therapeutic Targets for Autoimmune Diseases. Front Immunol 2021; 12:645699. [PMID: 33968036 PMCID: PMC8097144 DOI: 10.3389/fimmu.2021.645699] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/02/2021] [Indexed: 12/14/2022] Open
Abstract
Autoimmune diseases, such as multiple sclerosis and type-1 diabetes, are the outcomes of a failure of immune tolerance. Immune tolerance is sustained through interplays between two inter-dependent clusters of immune activities: immune stimulation and immune regulation. The mechanisms of immune regulation are exploited as therapeutic targets for the treatment of autoimmune diseases. One of these mechanisms is immune checkpoints (ICPs). The roles of ICPs in maintaining immune tolerance and hence suppressing autoimmunity were revealed in animal models and validated by the clinical successes of ICP-targeted therapeutics for autoimmune diseases. Recently, these roles were highlighted by the clinical discovery that the blockade of ICPs causes autoimmune disorders. Given the crucial roles of ICPs in immune tolerance, it is plausible to leverage ICPs as a group of therapeutic targets to restore immune tolerance and treat autoimmune diseases. In this review, we first summarize working mechanisms of ICPs, particularly those that have been utilized for therapeutic development. Then, we recount the agents and approaches that were developed to target ICPs and treat autoimmune disorders. These agents take forms of fusion proteins, antibodies, nucleic acids, and cells. We also review and discuss safety information for these therapeutics. We wrap up this review by providing prospects for the development of ICP-targeting therapeutics. In summary, the ever-increasing studies and results of ICP-targeting of therapeutics underscore their tremendous potential to become a powerful class of medicine for autoimmune diseases.
Collapse
Affiliation(s)
- Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States
| | - Reza Moosavi
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States
| | - Mingnan Chen
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
29
|
Adabor ES. Computational investigations of the immune response to repeated influenza infections and vaccinations. ROYAL SOCIETY OPEN SCIENCE 2021; 8:201433. [PMID: 33959318 PMCID: PMC8074957 DOI: 10.1098/rsos.201433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 03/05/2021] [Indexed: 06/12/2023]
Abstract
Previous studies have shown that repeated influenza vaccination can enhance susceptibility to subsequent infection with a drifted influenza virus strain. This paper seeks to further understanding of the interactions between influenza viruses and specific immune cells that accompany this phenomenon. The paper argues that repeated vaccination increases susceptibility to infection only in the context of a residual immunity induced by prior vaccination or infection. The results of computational analysis indicate that this is a dynamic consequence of interactions between vaccines, influenza viruses and specific immune cells. In particular, mathematical modelling was used to show that in the presence of residual immunity conferred by a vaccine administered in Canada in the 2013-2014 influenza season, the 2014-2015 season vaccine enhanced susceptibility to infection. Such infection enhancement occurs when the 2014-2015 vaccine boosts suppressive T-regulatory cells induced by the 2013-2014 vaccine, decreasing the strength of antibody responses to the infecting strain. Overall, the study suggests probable characteristics of infecting viruses and vaccines that make repeated influenza infections and vaccinations detrimental.
Collapse
Affiliation(s)
- Emmanuel S. Adabor
- Reserch Centre, African Institute for Mathematical Sciences, Cape Town, South Africa
- Department of Mathematical Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
30
|
Fluorine assembly nanocluster breaks the shackles of immunosuppression to turn the cold tumor hot. Proc Natl Acad Sci U S A 2020; 117:32962-32969. [PMID: 33318219 DOI: 10.1073/pnas.2011297117] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Clinical investigations have shown that a nonimmunogenic "cold" tumor is usually accompanied by few immunopositive cells and more immunosuppressive cells in the tumor microenvironment (TME), which is still the bottleneck of immune activation. Here, a fluorine assembly nanocluster was explored to break the shackles of immunosuppression, reawaken the immune system, and turn the cold tumor "hot." Once under laser irradiation, FS@PMPt produces sufficient reactive oxygen species (ROS) to fracture the ROS-sensitive linker, thus releasing the cisplatin conjugated PMPt to penetrate into the tumors and kill the regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). Meanwhile, ROS will induce potent immunogenic cell death (ICD) and further promote the accumulation of dendritic cells (DCs) and T cells, therefore not only increasing the infiltration of immunopositive cells from the outside but also reducing the immunosuppressive cells from the inside to break through the bottleneck of immune activation. The FS@PMPt nanocluster regulates the immune process in TME from negative to positive, from shallow to deep, to turn the cold tumor into a hot tumor and provoke a robust antitumor immune response.
Collapse
|
31
|
Regulatory T cells suppress Th17 cell Ca 2+ signaling in the spinal cord during murine autoimmune neuroinflammation. Proc Natl Acad Sci U S A 2020; 117:20088-20099. [PMID: 32732436 PMCID: PMC7443932 DOI: 10.1073/pnas.2006895117] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
T lymphocyte motility and interaction dynamics with other immune cells are vital determinants of immune responses. Regulatory T (Treg) cells prevent autoimmune disorders by suppressing excessive lymphocyte activity, but how interstitial motility patterns of Treg cells limit neuroinflammation is not well understood. We used two-photon microscopy to elucidate the spatial organization, motility characteristics, and interactions of endogenous Treg and Th17 cells together with antigen-presenting cells (APCs) within the spinal cord leptomeninges in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. Th17 cells arrive before the onset of clinical symptoms, distribute uniformly during the peak, and decline in numbers during later stages of EAE. In contrast, Treg cells arrive after Th17 cells and persist during the chronic phase. Th17 cells meander widely, interact with APCs, and exhibit cytosolic Ca2+ transients and elevated basal Ca2+ levels before the arrival of Treg cells. In contrast, Treg cells adopt a confined, repetitive-scanning motility while contacting APCs. These locally confined but highly motile Treg cells limit Th17 cells from accessing APCs and suppress Th17 cell Ca2+ signaling by a mechanism that is upstream of store-operated Ca2+ entry. Finally, Treg cell depletion increases APC numbers in the spinal cord and exaggerates ongoing neuroinflammation. Our results point to fundamental differences in motility characteristics between Th17 and Treg cells in the inflamed spinal cord and reveal three potential cellular mechanisms by which Treg cells regulate Th17 cell effector functions: reduction of APC density, limiting access of Th17 cells to APCs, and suppression of Th17 Ca2+ signaling.
Collapse
|
32
|
McIntyre LL, Greilach SA, Othy S, Sears-Kraxberger I, Wi B, Ayala-Angulo J, Vu E, Pham Q, Silva J, Dang K, Rezk F, Steward O, Cahalan MD, Lane TE, Walsh CM. Regulatory T cells promote remyelination in the murine experimental autoimmune encephalomyelitis model of multiple sclerosis following human neural stem cell transplant. Neurobiol Dis 2020. [PMID: 32276110 DOI: 10.1016/j.nbd.2020.14868] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic, inflammatory autoimmune disease that affects the central nervous system (CNS) for which there is no cure. In MS, encephalitogenic T cells infiltrate the CNS causing demyelination and neuroinflammation; however, little is known about the role of regulatory T cells (Tregs) in CNS tissue repair. Transplantation of neural stem and progenitor cells (NSCs and NPCs) is a promising therapeutic strategy to promote repair through cell replacement, although recent findings suggest transplanted NSCs also instruct endogenous repair mechanisms. We have recently described that dampened neuroinflammation and increased remyelination is correlated with emergence of Tregs following human NPC transplantation in a murine viral model of immune-mediated demyelination. In the current study we utilized the prototypic murine autoimmune model of demyelination experimental autoimmune encephalomyelitis (EAE) to test the efficacy of hNSC transplantation. Eight-week-old, male EAE mice receiving an intraspinal transplant of hNSCs during the chronic phase of disease displayed remyelination, dampened neuroinflammation, and an increase in CNS CD4+CD25+FoxP3+ regulatory T cells (Tregs). Importantly, ablation of Tregs abrogated histopathological improvement. Tregs are essential for maintenance of T cell homeostasis and prevention of autoimmunity, and an emerging role for Tregs in maintenance of tissue homeostasis through interactions with stem and progenitor cells has recently been suggested. The data presented here provide direct evidence for collaboration between CNS Tregs and hNSCs promoting remyelination.
Collapse
Affiliation(s)
- Laura L McIntyre
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, United States of America.
| | - Scott A Greilach
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, United States of America
| | - Shivashankar Othy
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, 92697, United States of America
| | - Ilse Sears-Kraxberger
- Reeve-Irvine Research Center, Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, CA 92697, United States of America
| | - Brian Wi
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, United States of America
| | - Julio Ayala-Angulo
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, United States of America
| | - Estelle Vu
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, United States of America
| | - Quan Pham
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, United States of America
| | - Jorge Silva
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, United States of America
| | - Kody Dang
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, United States of America
| | - Fady Rezk
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, United States of America
| | - Oswald Steward
- Reeve-Irvine Research Center, Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, CA 92697, United States of America
| | - Michael D Cahalan
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, 92697, United States of America
| | - Thomas E Lane
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, 92697, United States of America
| | - Craig M Walsh
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, United States of America.
| |
Collapse
|
33
|
Regulatory T cells promote remyelination in the murine experimental autoimmune encephalomyelitis model of multiple sclerosis following human neural stem cell transplant. Neurobiol Dis 2020; 140:104868. [PMID: 32276110 DOI: 10.1016/j.nbd.2020.104868] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 03/05/2020] [Accepted: 04/05/2020] [Indexed: 12/30/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic, inflammatory autoimmune disease that affects the central nervous system (CNS) for which there is no cure. In MS, encephalitogenic T cells infiltrate the CNS causing demyelination and neuroinflammation; however, little is known about the role of regulatory T cells (Tregs) in CNS tissue repair. Transplantation of neural stem and progenitor cells (NSCs and NPCs) is a promising therapeutic strategy to promote repair through cell replacement, although recent findings suggest transplanted NSCs also instruct endogenous repair mechanisms. We have recently described that dampened neuroinflammation and increased remyelination is correlated with emergence of Tregs following human NPC transplantation in a murine viral model of immune-mediated demyelination. In the current study we utilized the prototypic murine autoimmune model of demyelination experimental autoimmune encephalomyelitis (EAE) to test the efficacy of hNSC transplantation. Eight-week-old, male EAE mice receiving an intraspinal transplant of hNSCs during the chronic phase of disease displayed remyelination, dampened neuroinflammation, and an increase in CNS CD4+CD25+FoxP3+ regulatory T cells (Tregs). Importantly, ablation of Tregs abrogated histopathological improvement. Tregs are essential for maintenance of T cell homeostasis and prevention of autoimmunity, and an emerging role for Tregs in maintenance of tissue homeostasis through interactions with stem and progenitor cells has recently been suggested. The data presented here provide direct evidence for collaboration between CNS Tregs and hNSCs promoting remyelination.
Collapse
|
34
|
Genetic and Epigenetic Biomarkers of Immune Checkpoint Blockade Response. J Clin Med 2020; 9:jcm9010286. [PMID: 31968651 PMCID: PMC7019273 DOI: 10.3390/jcm9010286] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/09/2020] [Accepted: 01/12/2020] [Indexed: 02/06/2023] Open
Abstract
Checkpoint inhibitor therapy constitutes a promising cancer treatment strategy that targets the immune checkpoints to re-activate silenced T cell cytotoxicity. In recent pivotal trials, immune checkpoint blockade (ICB) demonstrated durable responses and acceptable toxicity, resulting in the regulatory approval of 8 checkpoint inhibitors to date for 15 cancer indications. However, up to ~85% of patients present with innate or acquired resistance to ICB, limiting its clinical utility. Current response biomarker candidates, including DNA mutation and neoantigen load, immune profiles, as well as programmed death-ligand 1 (PD-L1) expression, are only weak predictors of ICB response. Thus, identification of novel, more predictive biomarkers that could identify patients who would benefit from ICB constitutes one of the most important areas of immunotherapy research. Aberrant DNA methylation (5mC) and hydroxymethylation (5hmC) were discovered in multiple cancers, and dynamic changes of the epigenomic landscape have been identified during T cell differentiation and activation. While their role in cancer immunosuppression remains to be elucidated, recent evidence suggests that 5mC and 5hmC may serve as prognostic and predictive biomarkers of ICB-sensitive cancers. In this review, we describe the role of epigenetic phenomena in tumor immunoediting and other immune evasion related processes, provide a comprehensive update of the current status of ICB-response biomarkers, and highlight promising epigenomic biomarker candidates.
Collapse
|
35
|
Van Coillie S, Wiernicki B, Xu J. Molecular and Cellular Functions of CTLA-4. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:7-32. [PMID: 32185705 DOI: 10.1007/978-981-15-3266-5_2] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) is an inhibitory receptor belonging to the CD28 immunoglobulin subfamily, expressed primarily by T-cells. Its ligands, CD80 and CD86, are typically found on the surface of antigen-presenting cells and can either bind CD28 or CTLA-4, resulting in a costimulatory or a co-inhibitory response, respectively. Because of its dampening effect, CTLA-4 is a crucial regulator of T-cell homeostasis and self-tolerance. The mechanisms by which CTLA-4 exerts its inhibitory function can be categorized as either cell-intrinsic (affects the CTLA-4 expressing T-cell) or cell-extrinsic (affects secondary cells). Research from the last decade has shown that CTLA-4 mainly acts in a cell-extrinsic manner via its competition with CD28, CTLA-4-mediated trans-endocytosis of CD80 and CD86, and its direct tolerogenic effects on the interacting cell. Nonetheless, intrinsic CTLA-4 signaling has been implicated in T-cell motility and the regulation of CTLA-4 its subcellular localization amongst others. CTLA-4 is well recognized as a key immune checkpoint and has gained significant momentum as a therapeutic target in the field of autoimmunity and cancer. In this chapter, we describe the role of costimulation in immune response induction as well as the main mechanisms by which CTLA-4 can inhibit this process.
Collapse
Affiliation(s)
- Samya Van Coillie
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Zwijnaarde, 9052, Ghent, Belgium.
| | - Bartosz Wiernicki
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Zwijnaarde, 9052, Ghent, Belgium
| | - Jie Xu
- Institutes of Biomedical Sciences, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
36
|
Abstract
T cell-mediated elimination of malignant cells is one cornerstone of endogenous and therapeutically induced antitumor immunity. Tumors exploit numerous regulatory mechanisms to suppress T cell immunity. Regulatory T cells (T regs) play a crucial role in this process due to their ability to inhibit antitumoral immune responses and they are known to accumulate in various cancer entities. The chemokine CCL22, predominately produced by dendritic cells (DCs), regulates T reg migration via binding to its receptor CCR4. CCL22 controls T cell immunity, both by recruiting T regs to the tumor tissue and by promoting the formation of DC-T reg contacts in the lymph node. Here, we review the current knowledge on the role of CCL22 in cancer immunity. After revising the principal mechanisms of CCL22-induced immune suppression, we address the factors leading to CCL22 expression and ways of targeting this chemokine therapeutically. Therapeutic interventions to the CCL22-CCR4 axis may represent a promising strategy in cancer immunotherapy.
Collapse
|
37
|
Gudi RR, Karumuthil-Melethil S, Perez N, Li G, Vasu C. Engineered Dendritic Cell-Directed Concurrent Activation of Multiple T cell Inhibitory Pathways Induces Robust Immune Tolerance. Sci Rep 2019; 9:12065. [PMID: 31427630 PMCID: PMC6700167 DOI: 10.1038/s41598-019-48464-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/06/2019] [Indexed: 01/07/2023] Open
Abstract
Inhibitory/repressor-receptors are upregulated significantly on activated T cells, and have been the molecules of attention as targets for inducing immune tolerance. Induction of effective antigen specific tolerance depends on concurrent engagement of the TCR and one or more of these inhibitory receptors. Here, we show, for the first time that dendritic cells (DCs) can be efficiently engineered to express multiple T cell inhibitory ligands, and enhanced engagement of T cell inhibitory receptors, upon antigen presentation, by these DCs can induce effective CD4+ T cell tolerance and suppress autoimmunity. Compared to control DCs, antigen presentation by DCs that ectopically express CTLA4, PD1 and BTLA selective ligands (B7.1wa, PD-L1, and HVEM-CRD1 respectively) individually (mono-ligand DCs) or in combination (multi-ligand DCs) causes an inhibition of CD4+ T cell proliferation and pro-inflammatory cytokine response, as well as increase in Foxp3+ Treg frequency and immune regulatory cytokine production. Administration of self-antigen (mouse thyroglobulin; mTg) loaded multi-ligand DCs caused hyporesponsiveness to mTg challenge, suppression of autoantibody production, and amelioration of experimental autoimmune thyroiditis. Overall, this study shows that engineered DC-directed enhanced concurrent activation of multiple T cell coinhibitory pathways is an effective way to induce self-antigen specific T cell tolerance to suppress ongoing autoimmunity.
Collapse
Affiliation(s)
- Radhika R Gudi
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | | | - Nicolas Perez
- Department of Surgery, College of Medicine, University of Illinois, Chicago, IL, 60612, USA
| | - Gongbo Li
- Department of Surgery, College of Medicine, University of Illinois, Chicago, IL, 60612, USA
| | - Chenthamarakshan Vasu
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA.
- Department of Surgery, College of Medicine, University of Illinois, Chicago, IL, 60612, USA.
| |
Collapse
|
38
|
Riazifar M, Mohammadi MR, Pone EJ, Yeri A, Lässer C, Segaliny AI, McIntyre LL, Shelke GV, Hutchins E, Hamamoto A, Calle EN, Crescitelli R, Liao W, Pham V, Yin Y, Jayaraman J, Lakey JRT, Walsh CM, Van Keuren-Jensen K, Lotvall J, Zhao W. Stem Cell-Derived Exosomes as Nanotherapeutics for Autoimmune and Neurodegenerative Disorders. ACS NANO 2019; 13:6670-6688. [PMID: 31117376 PMCID: PMC6880946 DOI: 10.1021/acsnano.9b01004] [Citation(s) in RCA: 357] [Impact Index Per Article: 71.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
To dissect therapeutic mechanisms of transplanted stem cells and develop exosome-based nanotherapeutics in treating autoimmune and neurodegenerative diseases, we assessed the effect of exosomes secreted from human mesenchymal stem cells (MSCs) in treating multiple sclerosis using an experimental autoimmune encephalomyelitis (EAE) mouse model. We found that intravenous administration of exosomes produced by MSCs stimulated by IFNγ (IFNγ-Exo) (i) reduced the mean clinical score of EAE mice compared to PBS control, (ii) reduced demyelination, (iii) decreased neuroinflammation, and (iv) upregulated the number of CD4+CD25+FOXP3+ regulatory T cells (Tregs) within the spinal cords of EAE mice. Co-culture of IFNγ-Exo with activated peripheral blood mononuclear cells (PBMCs) cells in vitro reduced PBMC proliferation and levels of pro-inflammatory Th1 and Th17 cytokines including IL-6, IL-12p70, IL-17AF, and IL-22 yet increased levels of immunosuppressive cytokine indoleamine 2,3-dioxygenase. IFNγ-Exo could also induce Tregs in vitro in a murine splenocyte culture, likely mediated by a third-party accessory cell type. Further, IFNγ-Exo characterization by deep RNA sequencing suggested that IFNγ-Exo contains anti-inflammatory RNAs, where their inactivation partially hindered the exosomes potential to induce Tregs. Furthermore, we found that IFNγ-Exo harbors multiple anti-inflammatory and neuroprotective proteins. These results not only shed light on stem cell therapeutic mechanisms but also provide evidence that MSC-derived exosomes can potentially serve as cell-free therapies in creating a tolerogenic immune response to treat autoimmune and central nervous system disorders.
Collapse
Affiliation(s)
- Milad Riazifar
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - M. Rezaa Mohammadi
- Department of Materials Science and Engineering, University of California, Irvine, Irvine, California 92697, United States
| | - Egest J. Pone
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
- Department of Physiology and Biophysics, Vaccine Research and Development Center, University of California, Irvine, Irvine, California 92697, United States
| | - Ashish Yeri
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona 85004, United States
| | - Cecilia Lässer
- Krefting Research Center, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden
| | - Aude I. Segaliny
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Laura L. McIntyre
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, Irvine, California 92697, United States
| | - Ganesh Vilas Shelke
- Krefting Research Center, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden
- Department of Surgery, Institute of Clinical Sciences, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg 41345, Sweden
| | - Elizabeth Hutchins
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona 85004, United States
| | - Ashley Hamamoto
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Erika N. Calle
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Rossella Crescitelli
- Krefting Research Center, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden
| | - Wenbin Liao
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Victor Pham
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Yanan Yin
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jayapriya Jayaraman
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Jonathan R. T. Lakey
- Department of Surgery, University of California, Irvine, Orange, California 92868, United States
| | - Craig M. Walsh
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, Irvine, California 92697, United States
| | - Kendall Van Keuren-Jensen
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona 85004, United States
| | - Jan Lotvall
- Krefting Research Center, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden
| | - Weian Zhao
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
- Corresponding Author:
| |
Collapse
|
39
|
Tong AA, Forestell B, Murphy DV, Nair A, Allen F, Myers J, Klauschen F, Shen C, Gopal AA, Huang AY, Mandl JN. Regulatory T cells differ from conventional
CD
4
+
T cells in their recirculatory behavior and lymph node transit times. Immunol Cell Biol 2019; 97:787-798. [DOI: 10.1111/imcb.12276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/20/2019] [Accepted: 05/22/2019] [Indexed: 01/01/2023]
Affiliation(s)
- Alexander A Tong
- Department of Pathology Case Western Reserve University School of Medicine Cleveland OH USA
| | - Benjamin Forestell
- Department of Physiology Department of Microbiology and Immunology McGill Research Centre for Complex Traits McGill University Montreal QC Canada
| | - Daniel V Murphy
- Department of Pediatrics Case Western Reserve University School of Medicine Cleveland OH USA
- The Angie Fowler AYA Cancer Institute UH Rainbow Babies & Children's Hospital Cleveland OH USA
| | - Aditya Nair
- Department of Pediatrics Case Western Reserve University School of Medicine Cleveland OH USA
- The Angie Fowler AYA Cancer Institute UH Rainbow Babies & Children's Hospital Cleveland OH USA
| | - Frederick Allen
- Department of Pathology Case Western Reserve University School of Medicine Cleveland OH USA
| | - Jay Myers
- Department of Pediatrics Case Western Reserve University School of Medicine Cleveland OH USA
- The Angie Fowler AYA Cancer Institute UH Rainbow Babies & Children's Hospital Cleveland OH USA
| | | | - Connie Shen
- Department of Physiology Department of Microbiology and Immunology McGill Research Centre for Complex Traits McGill University Montreal QC Canada
| | - Angelica A Gopal
- Department of Physiology Department of Microbiology and Immunology McGill Research Centre for Complex Traits McGill University Montreal QC Canada
| | - Alex Y Huang
- Department of Pathology Case Western Reserve University School of Medicine Cleveland OH USA
- Department of Pediatrics Case Western Reserve University School of Medicine Cleveland OH USA
- The Angie Fowler AYA Cancer Institute UH Rainbow Babies & Children's Hospital Cleveland OH USA
| | - Judith N Mandl
- Department of Physiology Department of Microbiology and Immunology McGill Research Centre for Complex Traits McGill University Montreal QC Canada
| |
Collapse
|
40
|
Lu C, Zanker D, Lock P, Jiang X, Deng J, Duan M, Liu C, Faou P, Hickey MJ, Chen W. Memory regulatory T cells home to the lung and control influenza A virus infection. Immunol Cell Biol 2019; 97:774-786. [DOI: 10.1111/imcb.12271] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/19/2019] [Accepted: 05/20/2019] [Indexed: 11/27/2022]
Affiliation(s)
- Chunni Lu
- La Trobe Institute for Molecular Science School of Molecular Science La Trobe University Bundoora VIC Australia
| | - Damien Zanker
- La Trobe Institute for Molecular Science School of Molecular Science La Trobe University Bundoora VIC Australia
| | - Peter Lock
- La Trobe Institute for Molecular Science School of Molecular Science La Trobe University Bundoora VIC Australia
| | - Xiangrui Jiang
- La Trobe Institute for Molecular Science School of Molecular Science La Trobe University Bundoora VIC Australia
| | - Jieru Deng
- La Trobe Institute for Molecular Science School of Molecular Science La Trobe University Bundoora VIC Australia
| | - Mubing Duan
- La Trobe Institute for Molecular Science School of Molecular Science La Trobe University Bundoora VIC Australia
| | - Chuanxin Liu
- La Trobe Institute for Molecular Science School of Molecular Science La Trobe University Bundoora VIC Australia
| | - Pierre Faou
- La Trobe Institute for Molecular Science School of Molecular Science La Trobe University Bundoora VIC Australia
| | - Michael J Hickey
- Centre for Inflammatory Diseases Department of Medicine Monash Medical Centre Monash University Clayton VIC Australia
| | - Weisan Chen
- La Trobe Institute for Molecular Science School of Molecular Science La Trobe University Bundoora VIC Australia
| |
Collapse
|
41
|
Ovcinnikovs V, Ross EM, Petersone L, Edner NM, Heuts F, Ntavli E, Kogimtzis A, Kennedy A, Wang CJ, Bennett CL, Sansom DM, Walker LSK. CTLA-4-mediated transendocytosis of costimulatory molecules primarily targets migratory dendritic cells. Sci Immunol 2019; 4:eaaw0902. [PMID: 31152091 PMCID: PMC6570622 DOI: 10.1126/sciimmunol.aaw0902] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 04/02/2019] [Indexed: 12/12/2022]
Abstract
CTLA-4 is a critical negative regulator of the immune system and a major target for immunotherapy. However, precisely how it functions in vivo to maintain immune homeostasis is not clear. As a highly endocytic molecule, CTLA-4 can capture costimulatory ligands from opposing cells by a process of transendocytosis (TE). By restricting costimulatory ligand expression in this manner, CTLA-4 controls the CD28-dependent activation of T cells. Regulatory T cells (Tregs) constitutively express CTLA-4 at high levels and, in its absence, show defects in TE and suppressive function. Activated conventional T cells (Tconv) are also capable of CTLA-4-dependent TE; however, the relative use of this mechanism by Tregs and Tconv in vivo remains unclear. Here, we set out to characterize both the perpetrators and cellular targets of CTLA-4 TE in vivo. We found that Tregs showed constitutive cell surface recruitment of CTLA-4 ex vivo and performed TE rapidly after TCR stimulation. Tregs outperformed activated Tconv at TE in vivo, and expression of ICOS marked Tregs with this capability. Using TCR transgenic Tregs that recognize a protein expressed in the pancreas, we showed that the presentation of tissue-derived self-antigen could trigger Tregs to capture costimulatory ligands in vivo. Last, we identified migratory dendritic cells (DCs) as the major target for Treg-based CTLA-4-dependent regulation in the steady state. These data support a model in which CTLA-4 expressed on Tregs dynamically regulates the phenotype of DCs trafficking to lymph nodes from peripheral tissues in an antigen-dependent manner.
Collapse
Affiliation(s)
- Vitalijs Ovcinnikovs
- Institute of Immunity and Transplantation, University College London Division of Infection and Immunity, Royal Free Campus, NW3 2PF London , UK
| | - Ellen M Ross
- Institute of Immunity and Transplantation, University College London Division of Infection and Immunity, Royal Free Campus, NW3 2PF London , UK
| | - Lina Petersone
- Institute of Immunity and Transplantation, University College London Division of Infection and Immunity, Royal Free Campus, NW3 2PF London , UK
| | - Natalie M Edner
- Institute of Immunity and Transplantation, University College London Division of Infection and Immunity, Royal Free Campus, NW3 2PF London , UK
| | - Frank Heuts
- Institute of Immunity and Transplantation, University College London Division of Infection and Immunity, Royal Free Campus, NW3 2PF London , UK
| | - Elisavet Ntavli
- Institute of Immunity and Transplantation, University College London Division of Infection and Immunity, Royal Free Campus, NW3 2PF London , UK
| | - Alexandros Kogimtzis
- Institute of Immunity and Transplantation, University College London Division of Infection and Immunity, Royal Free Campus, NW3 2PF London , UK
| | - Alan Kennedy
- Institute of Immunity and Transplantation, University College London Division of Infection and Immunity, Royal Free Campus, NW3 2PF London , UK
| | - Chun Jing Wang
- Institute of Immunity and Transplantation, University College London Division of Infection and Immunity, Royal Free Campus, NW3 2PF London , UK
| | - Clare L Bennett
- Institute of Immunity and Transplantation, University College London Division of Infection and Immunity, Royal Free Campus, NW3 2PF London , UK
- Department of Haematology, University College London Cancer Institute, Royal Free Campus, NW3 2PF London, UK
| | - David M Sansom
- Institute of Immunity and Transplantation, University College London Division of Infection and Immunity, Royal Free Campus, NW3 2PF London , UK
| | - Lucy S K Walker
- Institute of Immunity and Transplantation, University College London Division of Infection and Immunity, Royal Free Campus, NW3 2PF London , UK.
| |
Collapse
|
42
|
Regulatory T cells limit unconventional memory to preserve the capacity to mount protective CD8 memory responses to pathogens. Proc Natl Acad Sci U S A 2019; 116:9969-9978. [PMID: 31036644 DOI: 10.1073/pnas.1818327116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Immunological memory exists so that following infection an expanded population of pathogen-specific lymphocytes can rapidly and efficiently control infection in the case of reexposure. However, in the case of CD8+ T lymphocytes, a population of unconventional CD44+CD122+ virtual memory T cells (TVM) has been described that possesses many, though not all, features of "true memory" T cells, without the requirement of first encountering cognate antigen. Here, we demonstrate a role for regulatory T cell-mediated restraint of TVM at least in part through limiting IL-15 trans-presentation by CD11b+ dendritic cells. Further, we show that keeping TVM in check ensures development of functional, antigen-specific "true" memory phenotype CD8+ T cells that can assist in pathogen control upon reexposure.
Collapse
|
43
|
Ugur M, Mueller SN. T cell and dendritic cell interactions in lymphoid organs: More than just being in the right place at the right time. Immunol Rev 2019; 289:115-128. [DOI: 10.1111/imr.12753] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 01/31/2019] [Accepted: 02/03/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Milas Ugur
- Department of Microbiology and Immunology The University of Melbourne, The Peter Doherty Institute for Infection and Immunity Melbourne Victoria Australia
| | - Scott N. Mueller
- Department of Microbiology and Immunology The University of Melbourne, The Peter Doherty Institute for Infection and Immunity Melbourne Victoria Australia
- The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne Melbourne Victoria Australia
| |
Collapse
|
44
|
Lingel H, Brunner-Weinzierl MC. CTLA-4 (CD152): A versatile receptor for immune-based therapy. Semin Immunol 2019; 42:101298. [DOI: 10.1016/j.smim.2019.101298] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 08/05/2019] [Indexed: 12/31/2022]
|
45
|
Akkaya B, Oya Y, Akkaya M, Al Souz J, Holstein AH, Kamenyeva O, Kabat J, Matsumura R, Dorward DW, Glass DD, Shevach EM. Regulatory T cells mediate specific suppression by depleting peptide-MHC class II from dendritic cells. Nat Immunol 2019; 20:218-231. [PMID: 30643268 PMCID: PMC6402611 DOI: 10.1038/s41590-018-0280-2] [Citation(s) in RCA: 168] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 11/08/2018] [Indexed: 01/22/2023]
Abstract
T regulatory cells (Tregs) can activate multiple suppressive mechanisms in vitro upon activation via the T cell receptor resulting in antigen-independent suppression. However, it remains unclear whether similar pathways operate in vivo. Here, we found that antigen-specific Tregs activated by dendritic cells (DCs) pulsed with two antigens suppressed Tnaive specific for both cognate and non-cognate antigens in vitro, but only suppressed Tnaive specific for cognate antigen in vivo. Antigen-specific Tregs formed strong interactions with DC resulting in selective inhibition of the binding of Tnaive to cognate antigen, yet allowing bystander Tnaive access. Strong binding resulted in removal of the cognate peptide-MHCII (pMHCII) from the DC surface reducing the capacity of the DC to present antigen. The enhanced binding of Tregs to DC coupled with their capacity to deplete pMHCII represents a novel pathway for Treg-mediated suppression and may be a mechanism by which Tregs maintain immune homeostasis.
Collapse
Affiliation(s)
- Billur Akkaya
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Yoshihiro Oya
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,Department of Rheumatology, Allergy & Clinical Immunology, National Hospital Organization Chiba-East National Hospital, Chiba, Japan
| | - Munir Akkaya
- Laboratory of Immunogenetics National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Jafar Al Souz
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Amanda H Holstein
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Olena Kamenyeva
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Juraj Kabat
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ryutaro Matsumura
- Department of Rheumatology, Allergy & Clinical Immunology, National Hospital Organization Chiba-East National Hospital, Chiba, Japan
| | - David W Dorward
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Labs, Hamilton, MT, USA
| | - Deborah D Glass
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,Rapa Therapeutics, Rockville, MD, USA
| | - Ethan M Shevach
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
46
|
Yang J, Sakai J, Siddiqui S, Lee RC, Ireland DDC, Verthelyi D, Akkoyunlu M. IL-6 Impairs Vaccine Responses in Neonatal Mice. Front Immunol 2018; 9:3049. [PMID: 30619375 PMCID: PMC6307459 DOI: 10.3389/fimmu.2018.03049] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 12/10/2018] [Indexed: 12/24/2022] Open
Abstract
The inability of infants to mount proper follicular helper T (TFH) cell response renders this age group susceptible to infectious diseases. Initial instruction of T cells by antigen presenting cells and subsequent differentiation into TFH cells are controlled by T cell receptor signal strength, co-stimulatory molecules and cytokines such as IL-6 and IL-21. In immunized adults, IL-6 promotes TFH development by increasing the expression of CXCR5 and the TFH master transcription factor, B cell lymphoma 6. Underscoring the importance of IL-6 in TFH generation, we found improved antibody responses accompanied by increased TFH cells and decreased follicular regulatory helper T (TFR) cells, a Foxp3 expressing inhibitory CD4+ T cell occupying the germinal center (GC), when a tetanus toxoid conjugated pneumococcal polysaccharide type 14 vaccine was injected in adult mice together with IL-6. Paradoxically, in neonates IL-6 containing PPS14-TT vaccine suppressed the already impaired TFH development and antibody responses in addition to increasing TFR cell population. Supporting the diminished TFH development, we detected lower frequency of phospho-STAT-3+ TFH in immunized neonatal T cells after IL-6 stimulation than adult cells. Moreover, IL-6 induced more phospho-STAT-3+ TFR in neonatal cells than adult cells. We also measured lower expression of IL-6R on TFH cells and higher expression on TFR cells in neonatal cells than adult cells, a possible explanation for the difference in IL-6 induced signaling in different age groups. Supporting the flow cytometry findings, microscopic examination revealed the localization of Treg cells in the splenic interfollicular niches of immunized adult mice compared to splenic follicles in neonatal mice. In addition to the limitations in the formation of IL-21 producing TFH cells, neonatal mice GC B cells also expressed lower levels of IL-21R in comparison to the adult mice cells. These findings point to diminished IL-6 activity on neonatal TFH cells as an underlying mechanism of the increased TFR: TFH ratio in immunized neonatal mice.
Collapse
Affiliation(s)
- Jiyeon Yang
- Division of Bacterial Allergenic and Parasitic Diseases, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Jiro Sakai
- Division of Bacterial Allergenic and Parasitic Diseases, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Shafiuddin Siddiqui
- Division of Bacterial Allergenic and Parasitic Diseases, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Robert C Lee
- Division of Bacterial Allergenic and Parasitic Diseases, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Derek D C Ireland
- Office of Biotechnology Products, Division of Biotechnology Review and Research III, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Daniela Verthelyi
- Office of Biotechnology Products, Division of Biotechnology Review and Research III, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Mustafa Akkoyunlu
- Division of Bacterial Allergenic and Parasitic Diseases, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
47
|
Miles B, Connick E. Control of the Germinal Center by Follicular Regulatory T Cells During Infection. Front Immunol 2018; 9:2704. [PMID: 30524440 PMCID: PMC6256122 DOI: 10.3389/fimmu.2018.02704] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 11/01/2018] [Indexed: 02/05/2023] Open
Abstract
Follicular regulatory T cells (Tfr) are a unique subset of CD4 T cells that control and impact adaptive immune responses in the lymphoid follicles and germinal centers (GC). Since their relatively recent discovery, several studies have revealed that Tfr interact with other cells within this niche and shape ensuing responses. Recent advances defining the functional and developmental characteristics of Tfr have revealed key characteristics of Tfr differentiation, GC recruitment and retention, and regulatory properties. Further, Tfr shape the GC response and balance tolerance through interactions with Tfh, by modifying Tfh number, diversity and function, as well as with B cells. Mechanisms by which Tfr regulate the GC include cell-to-cell interactions with Tfh and B cells, as well as altering their environment through cytokine production and sequestration. Tfr have been shown to have a diverse T cell receptor (TCR) repertoire and can be specific for immunizing agents, demonstrating a potential role in vaccine development. Due to these important characteristics and functions, Tfr play a major role in immune tolerance, response to infection, and vaccine efficacy.
Collapse
Affiliation(s)
- Brodie Miles
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, United States
| | - Elizabeth Connick
- Division of Infectious Diseases, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
48
|
Araujo Furlan CL, Tosello Boari J, Rodriguez C, Canale FP, Fiocca Vernengo F, Boccardo S, Beccaria CG, Adoue V, Joffre O, Gruppi A, Montes CL, Acosta Rodriguez EV. Limited Foxp3 + Regulatory T Cells Response During Acute Trypanosoma cruzi Infection Is Required to Allow the Emergence of Robust Parasite-Specific CD8 + T Cell Immunity. Front Immunol 2018; 9:2555. [PMID: 30455700 PMCID: PMC6230662 DOI: 10.3389/fimmu.2018.02555] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 10/17/2018] [Indexed: 12/20/2022] Open
Abstract
While it is now acknowledged that CD4+ T cells expressing CD25 and Foxp3 (Treg cells) regulate immune responses and, consequently, influence the pathogenesis of infectious diseases, the regulatory response mediated by Treg cells upon infection by Trypanosoma cruzi was still poorly characterized. In order to understand the role of Treg cells during infection by this protozoan parasite, we determined in time and space the magnitude of the regulatory response and the phenotypic, functional and transcriptional features of the Treg cell population in infected mice. Contrary to the accumulation of Treg cells reported in most chronic infections in mice and humans, experimental T. cruzi infection was characterized by sustained numbers but decreased relative frequency of Treg cells. The reduction in Treg cell frequency resulted from a massive accumulation of effector immune cells, and inversely correlated with the magnitude of the effector immune response as well as with emergence of acute immunopathology. In order to understand the causes underlying the marked reduction in Treg cell frequency, we evaluated the dynamics of the Treg cell population and found a low proliferation rate and limited accrual of peripheral Treg cells during infection. We also observed that Treg cells became activated and acquired a phenotypic and transcriptional profile consistent with suppression of type 1 inflammatory responses. To assess the biological relevance of the relative reduction in Treg cells frequency observed during T. cruzi infection, we transferred in vitro differentiated Treg cells at early moments, when the deregulation of the ratio between regulatory and conventional T cells becomes significant. Intravenous injection of Treg cells dampened parasite-specific CD8+ T cell immunity and affected parasite control in blood and tissues. Altogether, our results show that limited Treg cell response during the acute phase of T. cruzi infection enables the emergence of protective anti-parasite CD8+ T cell immunity and critically influences host resistance.
Collapse
Affiliation(s)
- Cintia L Araujo Furlan
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología, CONICET, Córdoba, Argentina
| | - Jimena Tosello Boari
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología, CONICET, Córdoba, Argentina
| | - Constanza Rodriguez
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología, CONICET, Córdoba, Argentina
| | - Fernando P Canale
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología, CONICET, Córdoba, Argentina
| | - Facundo Fiocca Vernengo
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología, CONICET, Córdoba, Argentina
| | - Santiago Boccardo
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología, CONICET, Córdoba, Argentina
| | - Cristian G Beccaria
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología, CONICET, Córdoba, Argentina
| | - Véronique Adoue
- Institut National de la Santé et de la Recherche Médicale, Toulouse, France.,Centre National de la Recherche Scientifique, Toulouse, France.,Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Olivier Joffre
- Institut National de la Santé et de la Recherche Médicale, Toulouse, France.,Centre National de la Recherche Scientifique, Toulouse, France.,Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Adriana Gruppi
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología, CONICET, Córdoba, Argentina
| | - Carolina L Montes
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología, CONICET, Córdoba, Argentina
| | - Eva V Acosta Rodriguez
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología, CONICET, Córdoba, Argentina
| |
Collapse
|
49
|
Abdulrahman Z, Kortekaas KE, De Vos Van Steenwijk PJ, Van Der Burg SH, Van Poelgeest MIE. The immune microenvironment in vulvar (pre)cancer: review of literature and implications for immunotherapy. Expert Opin Biol Ther 2018; 18:1223-1233. [DOI: 10.1080/14712598.2018.1542426] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Ziena Abdulrahman
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Gynaecology, Leiden University Medical Center, Leiden, The Netherlands
| | - Kim E Kortekaas
- Department of Gynaecology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Sjoerd H Van Der Burg
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
50
|
Khailaie S, Rowshanravan B, Robert PA, Waters E, Halliday N, Badillo Herrera JD, Walker LSK, Sansom DM, Meyer-Hermann M. Characterization of CTLA4 Trafficking and Implications for Its Function. Biophys J 2018; 115:1330-1343. [PMID: 30219287 PMCID: PMC6170599 DOI: 10.1016/j.bpj.2018.08.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 07/06/2018] [Accepted: 08/02/2018] [Indexed: 12/21/2022] Open
Abstract
CTLA4 is an essential negative regulator of T-cell immune responses and a key checkpoint regulating autoimmunity and antitumor responses. Genetic mutations resulting in quantitative defects in the CTLA4 pathway are also associated with the development of immune dysregulation syndromes in humans. It has been proposed that CTLA4 functions to remove its ligands CD80 and CD86 from opposing cells by a process known as transendocytosis. A quantitative characterization of CTLA4 synthesis, endocytosis, degradation, and recycling and how these affect its function is currently lacking. In a combined in vitro and in silico study, we developed a mathematical model and identified these trafficking parameters. Our model predicts optimal ligand removal in an intermediate affinity range. The intracellular CTLA4 pool as well as fast internalization, recovery of free CTLA4 from internalized complexes, and recycling is critical for sustained functionality. CD80-CTLA4 interactions are predicted to dominate over CD86-CTLA4. Implications of these findings in the context of control of antigen-presenting cells by regulatory T cells and of pathologic genetic deficiencies are discussed. The presented mathematical model can be reused in the community beyond these questions to better understand other trafficking receptors and study the impact of CTLA4 targeting drugs.
Collapse
Affiliation(s)
- Sahamoddin Khailaie
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany; Centre for Individualised Infection Medicine, Hannover, Germany
| | - Behzad Rowshanravan
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London, United Kingdom
| | - Philippe A Robert
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Erin Waters
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London, United Kingdom
| | - Neil Halliday
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London, United Kingdom
| | - Jesus David Badillo Herrera
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Lucy S K Walker
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London, United Kingdom
| | - David M Sansom
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London, United Kingdom.
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany; Centre for Individualised Infection Medicine, Hannover, Germany; Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany.
| |
Collapse
|