1
|
Liu J, Zhao Z, Zanni R, Jiang X, Weichselbaum RR, Lin W. Nanoparticle-Mediated Toll-Like Receptor Activation and Dual Immune Checkpoint Downregulation for Potent Cancer Immunotherapy. ACS NANO 2025; 19:8852-8866. [PMID: 40009747 DOI: 10.1021/acsnano.4c16542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Dual blockade of CD47 and PD-L1 immune checkpoints has shown potential in cancer treatment, but its clinical application is hindered by the on-target off-tumor immunotoxicities of monoclonal antibodies. Herein, we report a core-shell nanoparticle, PPA/HG, comprising polyinosinic: polycytidylic acid (PPA) in the core and a cholesterol-conjugated prodrug of 3-(hydroxyolinoyl)glycine (HG) on the shell, for potent cancer immunotherapy. PPA/HG shows a long half-life in the bloodstream to efficiently accumulate in tumors, where PPA/HG rapidly releases HG and PPA. HG inhibits the histone lysine demethylase 3A/c-Myc transduction for effective CD47 and PD-L1 downregulation in cancer cells while PPA activates toll-like receptor 3 in dendritic cells and tumor-associated macrophages to promote dendritic cell maturation and macrophage repolarization. PPA/HG promotes the infiltration and activation of effector T lymphocytes, meanwhile decreasing the population of immunosuppressive regulatory T cells. Systemic administration of PPA/HG significantly inhibits the progression of orthotopic triple-negative breast cancer and pancreatic ductal adenocarcinoma with minimal side effects.
Collapse
Affiliation(s)
- Jing Liu
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, United States
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, University of Chicago, 5758 South Maryland Avenue, Chicago, Illinois 60637, United States
| | - Zhihao Zhao
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, United States
| | - Richard Zanni
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, United States
| | - Xiaomin Jiang
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, United States
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, University of Chicago, 5758 South Maryland Avenue, Chicago, Illinois 60637, United States
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, University of Chicago, 5758 South Maryland Avenue, Chicago, Illinois 60637, United States
| | - Wenbin Lin
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, United States
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, University of Chicago, 5758 South Maryland Avenue, Chicago, Illinois 60637, United States
| |
Collapse
|
2
|
Pirahmadi S, Zargar M, Pourhashem Z, Vand-Rajabpour H, Sani JJ, Yousefi H, Afzali S, Zakeri S, Mehrizi AA. Selection of combination adjuvants for enhanced immunogenicity of a recombinant CelTOS vaccine against Plasmodium falciparum. Biochem Biophys Res Commun 2025; 748:151310. [PMID: 39809136 DOI: 10.1016/j.bbrc.2025.151310] [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: 10/07/2024] [Revised: 12/17/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
Recently, there has been significant interest in developing combination adjuvants to achieve efficient vaccines. However, it remains uncertain which combinations of adjuvants could best enhance the immune response to the recombinant antigen. In the current study, to improve the immunogenicity of Plasmodium falciparum cell traversal protein for ookinetes and sporozoites (PfCelTOS), we tested three different adjuvants: MPL, Poly I:C, and QS-21 alone or in a triple mixture (MPL/Poly I:C/QS-21; MPQ) and a dual mixture (Poly I:C/QS-21; PQ). BALB/c mice were immunized with recombinant PfCelTOS, either alone or combined with MPL, Poly I:C, QS-21, or with dual and triple adjuvant mixtures. Humoral and cellular immune responses were assessed in the various mouse groups, along with the functional activity of anti-PfCelTOS antibodies in oocyst inhibition. The results showed that administering the PfCelTOS antigen with triple or dual adjuvant mixtures significantly increased specific antibody levels, as well as IFN-ɣ and TNF cytokine production (P < 0.05), compared to PfCelTOS alone or combined with single adjuvants. These vaccine adjuvant mixtures also enhanced transmission-reducing activity (TRA), resulting in 76%-84% reductions in oocyst intensity in functional assays. Interestingly, comparable antibody levels and functional inhibitory activity were observed in the groups that received antigen with both dual and triple adjuvants (P > 0.05). The findings indicate that the dual mixture of Poly I:C and QS-21 is the most effective formulation for a vaccine against PfCelTOS. This discovery has important cost and safety implications by minimizing the adjuvants required to achieve an optimal immune response.
Collapse
MESH Headings
- Animals
- Plasmodium falciparum/immunology
- Mice, Inbred BALB C
- Malaria Vaccines/immunology
- Malaria Vaccines/administration & dosage
- Malaria, Falciparum/prevention & control
- Malaria, Falciparum/immunology
- Female
- Mice
- Adjuvants, Immunologic/pharmacology
- Adjuvants, Immunologic/administration & dosage
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Antibodies, Protozoan/immunology
- Protozoan Proteins/immunology
- Adjuvants, Vaccine/pharmacology
- Poly I-C/immunology
- Poly I-C/pharmacology
- Poly I-C/administration & dosage
- Immunogenicity, Vaccine
- Saponins/immunology
- Saponins/pharmacology
- Lipid A/analogs & derivatives
- Lipid A/pharmacology
- Lipid A/immunology
- Lipid A/administration & dosage
Collapse
Affiliation(s)
- Sakineh Pirahmadi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Mostafa Zargar
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Zeinab Pourhashem
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Hediyeh Vand-Rajabpour
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Jafar J Sani
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Hemn Yousefi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Shima Afzali
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Sedigheh Zakeri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Akram Abouie Mehrizi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
3
|
Adams CS, Kim H, Burtner AE, Lee DS, Dobbins C, Criswell C, Coventry B, Tran-Pearson A, Kim HM, King NP. De novo design of protein minibinder agonists of TLR3. Nat Commun 2025; 16:1234. [PMID: 39890776 PMCID: PMC11785957 DOI: 10.1038/s41467-025-56369-w] [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: 04/18/2024] [Accepted: 01/16/2025] [Indexed: 02/03/2025] Open
Abstract
Toll-like Receptor 3 (TLR3) is a pattern recognition receptor that initiates antiviral immune responses upon binding double-stranded RNA (dsRNA). Several nucleic acid-based TLR3 agonists have been explored clinically as vaccine adjuvants in cancer and infectious disease, but present substantial manufacturing and formulation challenges. Here, we use computational protein design to create novel miniproteins that bind to human TLR3 with nanomolar affinities. Cryo-EM structures of two minibinders in complex with TLR3 reveal that they bind the target as designed, although one partially unfolds due to steric competition with a nearby N-linked glycan. Multivalent forms of both minibinders induce NF-κB signaling in TLR3-expressing cell lines, demonstrating that they may have therapeutically relevant biological activity. Our work provides a foundation for the development of specific, stable, and easy-to-formulate protein-based agonists of TLRs and other pattern recognition receptors.
Collapse
Affiliation(s)
- Chloe S Adams
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
| | - Hyojin Kim
- Center for Biomolecular & Cellular Structure, Institute for Basic Science (IBS), Daejeon, 34126, South Korea
| | - Abigail E Burtner
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
| | - Dong Sun Lee
- Center for Biomolecular & Cellular Structure, Institute for Basic Science (IBS), Daejeon, 34126, South Korea
| | - Craig Dobbins
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
| | - Cameron Criswell
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
| | - Brian Coventry
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Adri Tran-Pearson
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
| | - Ho Min Kim
- Center for Biomolecular & Cellular Structure, Institute for Basic Science (IBS), Daejeon, 34126, South Korea.
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, South Korea.
| | - Neil P King
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA.
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
4
|
Zhang Z, Wang N, Lu J, Qu Y, Song Y, Yang X, Wei Z, Zhang Q, Herdewijn P, Chang J, Wang XN, Wang Z. Synthesis and pharmacodynamic evaluation of 2-aminoindole derivatives against influenza A virus in vitro/vivo. Eur J Med Chem 2025; 281:117044. [PMID: 39547081 DOI: 10.1016/j.ejmech.2024.117044] [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: 09/18/2024] [Revised: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Influenza virus is a kind of respiratory pathogen with high morbidity and mortality, which still threatens human health. Existing anti-influenza drugs have various limitations, such as the inability to alleviate body injury and side effects. There remains an urgent need to develop a novel antiviral drug to efficiently inhibit viral infection while avoiding body injury. A series of 2-aminoindole derivatives were synthesized via the TMSOTf-catalyzed reactions of N-arylynamides with sulfilimines and evaluated for their anti-influenza virus activity. The experimental results showed that 2-aminoindole 3h had significant antiviral activity (EC50 = 8.37 ± 0.65 μM) and the lowest cytotoxicity (CC50 = 669.26 ± 11.42 μM) in vitro. 2-Aminoindole 3h could inhibit viral replication by effectively binding to RNA-dependent RNA polymerase (RdRp), and could also directly target host cells to inhibit cytokine storms and apoptosis induced by viral infection, thereby improving host cell survival rate. In addition, viral load and organ injury in the lung tissue of infected mice were effectively reduced by 2-aminoindole 3h with satisfactory biosafety. These findings highlight the potential of a valuable therapeutic option against influenza infection while also laying the foundation for further research and development in this area.
Collapse
Affiliation(s)
- Zhongmou Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery & Development, Zhengzhou University, Zhengzhou, 450001, China
| | - Nanfang Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Pingyuan Laboratory, State Key Laboratory of Antiviral Drugs, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Jiejie Lu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery & Development, Zhengzhou University, Zhengzhou, 450001, China
| | - Ying Qu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery & Development, Zhengzhou University, Zhengzhou, 450001, China
| | - Yihui Song
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xinyu Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zhanyong Wei
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China
| | - Qi Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Piet Herdewijn
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; XNA Platform, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Junbiao Chang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery & Development, Zhengzhou University, Zhengzhou, 450001, China.
| | - Xiao-Na Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Pingyuan Laboratory, State Key Laboratory of Antiviral Drugs, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China.
| | - Zhenya Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery & Development, Zhengzhou University, Zhengzhou, 450001, China; International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China; XNA Platform, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
5
|
Jeon D, Hill E, McNeel DG. Toll-like receptor agonists as cancer vaccine adjuvants. Hum Vaccin Immunother 2024; 20:2297453. [PMID: 38155525 PMCID: PMC10760790 DOI: 10.1080/21645515.2023.2297453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/16/2023] [Indexed: 12/30/2023] Open
Abstract
Cancer immunotherapy has emerged as a promising strategy to treat cancer patients. Among the wide range of immunological approaches, cancer vaccines have been investigated to activate and expand tumor-reactive T cells. However, most cancer vaccines have not shown significant clinical benefit as monotherapies. This is likely due to the antigen targets of vaccines, "self" proteins to which there is tolerance, as well as to the immunosuppressive tumor microenvironment. To help circumvent immune tolerance and generate effective immune responses, adjuvants for cancer vaccines are necessary. One representative adjuvant family is Toll-Like receptor (TLR) agonists, synthetic molecules that stimulate TLRs. TLRs are the largest family of pattern recognition receptors (PRRs) that serve as the sensors of pathogens or cellular damage. They recognize conserved foreign molecules from pathogens or internal molecules from cellular damage and propel innate immune responses. When used with vaccines, activation of TLRs signals an innate damage response that can facilitate the development of a strong adaptive immune response against the target antigen. The ability of TLR agonists to modulate innate immune responses has positioned them to serve as adjuvants for vaccines targeting infectious diseases and cancers. This review provides a summary of various TLRs, including their expression patterns, their functions in the immune system, as well as their ligands and synthetic molecules developed as TLR agonists. In addition, it presents a comprehensive overview of recent strategies employing different TLR agonists as adjuvants in cancer vaccine development, both in pre-clinical models and ongoing clinical trials.
Collapse
Affiliation(s)
- Donghwan Jeon
- Department of Oncology, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Ethan Hill
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Douglas G. McNeel
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| |
Collapse
|
6
|
Velimirov B, Velimirov BA. Immune Responses Elicited by Outer Membrane Vesicles of Gram-Negative Bacteria: Important Players in Vaccine Development. Life (Basel) 2024; 14:1584. [PMID: 39768292 PMCID: PMC11678573 DOI: 10.3390/life14121584] [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: 10/11/2024] [Revised: 11/12/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
The attractiveness of OMVs derived from Gram-negative bacteria lies in the fact that they have two biomembranes sandwiching a peptidoglycan layer. It is well known that the envelope of OMVs consists of the outer bacterial membrane [OM] and not of the inner one [IM] of the source bacterium. This implies that all outer membranous molecules found in the OM act as antigens. However, under specific conditions, some of the inner membrane proteins can be exported into the outer membrane layer and perform as antigens. A key information was that the used purification procedures for OMVs, the induction methods to increase the production of OMVs as well as the specific mutant strains obtained via genetic engineering affect the composition of potential antigens on the surface and in the lumen of the OMVs. The available literature allowed us to list the major antigens that could be defined on OMVs. The functions of the antigens within the source bacterium are discussed for a better understanding of the various available hypotheses on the biogenesis of vesicle formation. Also, the impacts of OMV antigens on the immune system using animal models are assessed. Furthermore, information on the pathways of OMVs entering the host cell is presented. An example of a bacterial infection that causes epidemic diseases, namely via Neisseria meningitidis, is used to demonstrate that OMVs derived from this pathogen elicit protective immune responses when administered as a vaccine. Furthermore, information on OMV vaccines under development is presented. The assembled knowledge allowed us to formulate a number of reasons why OMVs are attractive as vaccine platforms, as their undesirable side effects remain small, and to provide an outlook on the potential use of OMVs as a vaccine platform.
Collapse
Affiliation(s)
- Branko Velimirov
- Division of Microbiology and Molecular Biology, Medical Faculty, Private Sigmund Freud University, Freudplatz 3, 1020 Wien, Austria;
| | | |
Collapse
|
7
|
Gonzales GA, Huang S, Wilkinson L, Nguyen JA, Sikdar S, Piot C, Naumenko V, Rajwani J, Wood CM, Dinh I, Moore M, Cedeño E, McKenna N, Polyak MJ, Amidian S, Ebacher V, Rosin NL, Carneiro MB, Surewaard B, Peters NC, Mody CH, Biernaskie J, Yates RM, Mahoney DJ, Canton J. The pore-forming apolipoprotein APOL7C drives phagosomal rupture and antigen cross-presentation by dendritic cells. Sci Immunol 2024; 9:eadn2168. [PMID: 39485861 DOI: 10.1126/sciimmunol.adn2168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 07/23/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024]
Abstract
Conventional dendritic cells (cDCs) generate protective cytotoxic T lymphocyte (CTL) responses against extracellular pathogens and tumors. This is achieved through a process known as cross-presentation (XP), and, despite its biological importance, the mechanism(s) driving XP remains unclear. Here, we show that a cDC-specific pore-forming protein called apolipoprotein L 7C (APOL7C) is up-regulated in response to innate immune stimuli and is recruited to phagosomes. Association of APOL7C with phagosomes led to phagosomal rupture and escape of engulfed antigens to the cytosol, where they could be processed via the endogenous MHC class I antigen processing pathway. Accordingly, mice deficient in APOL7C did not efficiently prime CD8+ T cells in response to immunization with bead-bound and cell-associated antigens. Together, our data indicate the presence of dedicated apolipoproteins that mediate the delivery of phagocytosed proteins to the cytosol of activated cDCs to facilitate XP.
Collapse
Affiliation(s)
- Gerone A Gonzales
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Song Huang
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Liam Wilkinson
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jenny A Nguyen
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Saif Sikdar
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Research Institute, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, Alberta, Canada
| | - Cécile Piot
- Immunobiology Laboratory, Francis Crick Institute, London, UK
| | - Victor Naumenko
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, Alberta, Canada
- Riddell Centre for Cancer Immunotherapy, Calgary, Alberta, Canada
| | - Jahanara Rajwani
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Research Institute, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, Alberta, Canada
| | - Cassandra M Wood
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Irene Dinh
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Melanie Moore
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Eymi Cedeño
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Neil McKenna
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Maria J Polyak
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Sara Amidian
- Cell Imaging Core, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | | | - Nicole L Rosin
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Matheus B Carneiro
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Bas Surewaard
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Nathan C Peters
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Christopher H Mody
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Jeff Biernaskie
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Robin M Yates
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Douglas J Mahoney
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Research Institute, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, Alberta, Canada
- Riddell Centre for Cancer Immunotherapy, Calgary, Alberta, Canada
| | - Johnathan Canton
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Riddell Centre for Cancer Immunotherapy, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, Calgary, Alberta, Canada
| |
Collapse
|
8
|
Koumantou D, Adiko AC, Bourdely P, Nugue M, Boedec E, El‐Benna J, Monteiro R, Saveanu C, Laffargue M, Wymann MP, Dalod M, Guermonprez P, Saveanu L. Specific Requirement of the p84/p110γ Complex of PI3Kγ for Antibody-Activated, Inducible Cross-Presentation in Murine Type 2 DCs. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401179. [PMID: 39382167 PMCID: PMC11600261 DOI: 10.1002/advs.202401179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 09/05/2024] [Indexed: 10/10/2024]
Abstract
Cross-presentation by MHCI is optimally efficient in type 1 dendritic cells (DC) due to their high capacity for antigen processing. However, through specific pathways, other DCs, such as type 2 DCs and inflammatory DCs (iDCs) can also cross-present antigens. FcγR-mediated uptake by type 2 DC and iDC subsets mediates antibody-dependent cross-presentation and activation of CD8+ T cell responses. Here, an important role for the p84 regulatory subunit of PI3Kγ in mediating efficient cross-presentation of exogenous antigens in otherwise inefficient cross-presenting cells, such as type 2 DCs and GM-CSF-derived iDCs is identified. FcγR-mediated cross-presentation is shown in type 2 and iDCs depend on the enzymatic activity of the p84/p110γ complex of PI3Kγ, which controls the activity of the NADPH oxidase NOX2 and ROS production in murine spleen type 2 DCs and GM-CSF-derived iDCs. In contrast, p84/p110γ is largely dispensable for cross-presentation by type 1 DCs. These findings suggest that PI3Kγ-targeted therapies, currently considered for oncological practice, may interfere with the ability of type 2 DCs and iDCs to cross-present antigens contained in immune complexes.
Collapse
Affiliation(s)
- Despoina Koumantou
- Centre de Recherche sur l'InflammationINSERM UMR1149CNRS EMR8252Faculté de Médecine site BichatUniversité Paris CitéParis75018France
- Laboratoire d'Excellence InflamexUniversité Paris CitéParis75018France
| | - Aimé Cézaire Adiko
- Centre de Recherche sur l'InflammationINSERM UMR1149CNRS EMR8252Faculté de Médecine site BichatUniversité Paris CitéParis75018France
- Laboratoire d'Excellence InflamexUniversité Paris CitéParis75018France
| | - Pierre Bourdely
- Centre de Recherche sur l'InflammationINSERM UMR1149CNRS EMR8252Faculté de Médecine site BichatUniversité Paris CitéParis75018France
- CNRSINSERMInstitut CochinParis75014France
| | - Mathilde Nugue
- Centre de Recherche sur l'InflammationINSERM UMR1149CNRS EMR8252Faculté de Médecine site BichatUniversité Paris CitéParis75018France
- Laboratoire d'Excellence InflamexUniversité Paris CitéParis75018France
| | - Erwan Boedec
- Centre de Recherche sur l'InflammationINSERM UMR1149CNRS EMR8252Faculté de Médecine site BichatUniversité Paris CitéParis75018France
- Laboratoire d'Excellence InflamexUniversité Paris CitéParis75018France
| | - Jamel El‐Benna
- Centre de Recherche sur l'InflammationINSERM UMR1149CNRS EMR8252Faculté de Médecine site BichatUniversité Paris CitéParis75018France
- Laboratoire d'Excellence InflamexUniversité Paris CitéParis75018France
| | - Renato Monteiro
- Centre de Recherche sur l'InflammationINSERM UMR1149CNRS EMR8252Faculté de Médecine site BichatUniversité Paris CitéParis75018France
- Laboratoire d'Excellence InflamexUniversité Paris CitéParis75018France
| | - Cosmin Saveanu
- Institut PasteurRNA Biology of Fungal PathogensUniversité Paris CitéParis75015France
| | | | - Matthias P. Wymann
- Department of BiomedicineUniversity of BaselMattenstrasse 28BaselCH‐4058Switzerland
| | - Marc Dalod
- CNRSINSERMCIMLCentre d'Immunologie de Marseille‐LuminyTuring Center for Living SystemsAix‐Marseille UniversityMarseille13007France
| | - Pierre Guermonprez
- “Dendritic cells and adaptive immunity”Immunology departmentPasteur InstituteParis75015France
- CNRS UMR3738, Département Biologie du Développement et Cellules SouchesInstitut Pasteur, Université Paris Cité25‐28 rue du Docteur RouxParis75015France
| | - Loredana Saveanu
- Centre de Recherche sur l'InflammationINSERM UMR1149CNRS EMR8252Faculté de Médecine site BichatUniversité Paris CitéParis75018France
- Laboratoire d'Excellence InflamexUniversité Paris CitéParis75018France
| |
Collapse
|
9
|
Sun M, Liu Y, Ni X, Tan R, Wang Y, Jiang Y, Ke D, Du H, Guo G, Liu K. Intranasal immunization with poly I:C and CpG ODN adjuvants enhances the protective efficacy against Helicobacter pylori infection in mice. Microbes Infect 2024:105433. [PMID: 39461584 DOI: 10.1016/j.micinf.2024.105433] [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: 05/14/2024] [Revised: 10/17/2024] [Accepted: 10/19/2024] [Indexed: 10/29/2024]
Abstract
Helicobacter pylori (H. pylori) infection is a serious public health issue, and development of vaccines is a desirable preventive strategy for H. pylori. Toll-like receptor (TLR) ligands have shown potential as vaccine adjuvants that induce immune responses, but polyinosinic-polycytidylic acid (poly I:C), a nucleic acid-based TLR9 ligand, is less well studied in H. pylori vaccine research. Here, we evaluated the effects of poly I:C and CpG oligodeoxynucleotide (CpG ODN), a nucleic acid TLR3 ligand, as adjuvants in combination with the H. pylori recombinant proteins LpoB and UreA to protect against H. pylori infection. For analysis of specific immune responses, the levels of specific antibodies and splenic cytokines were measured in the immunized mice. Compared with CpG ODN, poly I:C could induce mucosal sIgA antibody responses and reduce H. pylori colonization. Additionally, the combination of poly I:C and CpG ODN caused greater immunoprotection and significantly reduced gastritis, exerting synergistic effects. Analysis of splenic cytokines revealed that poly I:C mainly triggered a mixed Th1/Th2/Th17 immune response, whereas the combination of CpG ODN and poly I:C induced a Th1/Th17 immune response. Our findings indicated that increased levels of mucosal sIgA antibodies and a robust splenic Th1/Th17 immune response were associated with reduced H. pylori colonization in vaccinated mice. This study identified a potential TLR ligand adjuvant for developing more effective H. pylori vaccines.
Collapse
Affiliation(s)
- Min Sun
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yu Liu
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiumei Ni
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Runqing Tan
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yi Wang
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yajun Jiang
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dingxin Ke
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Han Du
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Gang Guo
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Kaiyun Liu
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
10
|
Glauß S, Neumeyer V, Hanesch L, Marek J, Hartmann N, Wiedemann GM, Altomonte J. A Novel Chimeric Oncolytic Virus Mediates a Multifaceted Cellular Immune Response in a Syngeneic B16 Melanoma Model. Cancers (Basel) 2024; 16:3405. [PMID: 39410025 PMCID: PMC11475060 DOI: 10.3390/cancers16193405] [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: 09/10/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND/OBJECTIVES Oncolytic virotherapy is a promising approach in cancer immunotherapy. We have previously described a recombinant hybrid oncolytic virus (OV), VSV-NDV, which has a favorable safety profile and therapeutic immunogenicity, leading to direct oncolysis, abscopal effects, and prolonged survival in syngeneic in vivo tumor models. While OVs are known to mediate systemic anti-tumor immune responses, the detailed characterization of local and systemic immune responses to fusogenic oncolytic virotherapy remains unexplored. METHODS AND RESULTS We analyzed immune cell compartments in the spleen, blood, tumor-draining lymph nodes (TDLNs), and tumors over the course of VSV-NDV therapy in a bilateral syngeneic melanoma mouse model. Our results revealed significant local infiltration and activation of T lymphocytes in tumors and globally in the blood and spleen. Notably, in vivo CD8+ T cell depletion led to complete abrogation of the tumor response, highlighting the crucial role of T cells in promoting the therapeutic effects of oncolytic VSV-NDV. In vitro co-culture experiments enabled the interrogation of human immune cell responses to VSV-NDV-mediated oncolysis. Human peripheral blood mononuclear cells (PBMCs) were efficiently stimulated by exposure to VSV-NDV-infected cancer cells, which recapitulates the in vivo murine findings. CONCLUSIONS Taken together, these data characterize a broad anti-tumor immune cell response to oncolytic VSV-NDV therapy and suggest that CD8+ T cells play a decisive role in therapeutic outcome, which supports the further development of this chimeric vector as a multimechanistic immunotherapy for solid cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jennifer Altomonte
- Department of Internal Medicine II, Rechts der Isar Hospital, Technical University of Munich, 81675 Munich, Germany; (S.G.); (V.N.); (L.H.); (J.M.); (N.H.); (G.M.W.)
| |
Collapse
|
11
|
Zhang RR, Yang XY, Yang YL, Guo TK, Huang JS, Yang YS, Shi CW, Yang GL, Huang HB, Wang JZ, Jiang YL, Cao X, Wang N, Zeng Y, Yang WT, Wang CF. TLR3/TRIF and MAVS Signaling Is Essential in Regulating Mucosal T Cell Responses during Rotavirus Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1008-1022. [PMID: 39194407 DOI: 10.4049/jimmunol.2300867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 07/30/2024] [Indexed: 08/29/2024]
Abstract
The functions of the natural dsRNA sensors TLR3 (TRIF) and RIG-I (MAVS) are crucial during viral challenge and have not been accurately clarified in adaptive immune responses to rotavirus (RV) infection. In this study, we found that RV infection caused severe pathological damage to the small intestine of TLR3-/- and TRIF-/- mice. Our data found that dendritic cells from TLR3-/- and TRIF-/- mice had impaired Ag presentation to the RV and attenuated initiation of T cells upon viral infection. These attenuated functions resulted in impaired CD4+ T and CD8+ T function in mice lacking TLR3-TRIF signaling postinfection. Additionally, attenuated proliferative capacity of T cells from TLR3-/- and TRIF-/- mice was observed. Subsequently, we observed a significant reduction in the absolute number of memory T cells in the spleen and mesenteric lymph node (MLN) of TRIF-/- recipient mice following RV infection in a bone marrow chimeric model. Furthermore, there was reduced migration of type 2 classical dendritic cells from the intestine to MLNs after RV infection in TLR3-/- and TRIF-/- mice. Notably, RV infection resulted in attenuated killing of spleen and MLN tissues in TRIF-/- and MAVS-/- mice. Finally, we demonstrated that RV infection promoted apoptosis of CD8+ T cells in TRIF-/- and TLR3-/-MAVS-/- mice. Taken together, our findings highlight an important mechanism of TLR3 signaling through TRIF in mucosal T cell responses to RV and lay the foundation for the development of a novel vaccine.
Collapse
Affiliation(s)
- Rong-Rong Zhang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Xue-Yao Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yong-Lei Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Tian-Kui Guo
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Jing-Shu Huang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Ying-Shi Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chun-Wei Shi
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Gui-Lian Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Hai-Bin Huang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Jian-Zhong Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yan-Long Jiang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Xin Cao
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Nan Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yan Zeng
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Wen-Tao Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chun-Feng Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| |
Collapse
|
12
|
Wang F, Tang YS, Cao F, Shou JW, Wong CK, Shaw PC. 3,4,5-tri-O-caffeoylquinic acid attenuates influenza A virus induced inflammation through Toll-like receptor 3/7 activated signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155896. [PMID: 39053250 DOI: 10.1016/j.phymed.2024.155896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/28/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND 3,4,5-tri-O-caffeoylquinic acid (3,4,5-TCQA), a natural polyphenolic acid, has been shown to be effective against influenza A virus (IAV) infection. Although it was found to inhibit the neuraminidase of IAV, it may also perturb other cellular functions, as polyphenolic acids have shown antioxidant, anti-inflammatory and other activities. PURPOSE This study aimed to investigate the effect of 3,4,5-TCQA at a cell level, which is critical for protecting host cell from IAV infection. STUDY DESIGN AND METHODS We explored the effect of 3,4,5-TCQA on H292 cells infected or un-infected with Pr8 IAV. The major genes and related pathway were identified through RNA sequencing. The pathway was confirmed by qRT-PCR and western blot analysis. The anti-inflammatory activity was evaluated using nitric oxide measurement assay. RESULTS We showed that 3,4,5-TCQA downregulated the immune response in H292 cells, and reduced the cytokine production in Pr8-infected cells, through Toll-like receptor (TLR) signaling pathway. In addition, 3,4,5-TCQA showed anti-inflammatory activity in LPS-activated RAW264.7 cells. CONCLUSION Collectively, our results indicated that 3,4,5-TCQA suppressed inflammation caused by IAV infection through TLR3/7 signaling pathway. This provides a new insight into the antiviral mechanism of 3,4,5-TCQA.
Collapse
Affiliation(s)
- Fan Wang
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yun-Sang Tang
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China; The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Fei Cao
- College of Pharmaceutical Sciences, Hebei University, Baoding, China
| | - Jia-Wen Shou
- Li Dak Sum Yip Yio Chin R&D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Chun-Kwok Wong
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants (CUHK) and Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Pang-Chui Shaw
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China; Li Dak Sum Yip Yio Chin R&D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants (CUHK) and Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
13
|
Martínez-Riaño A, Mosteo L, Molero-Glez P, Márquez-Rodas I, Melero I. Double-Stranded RNA to Mimic Viral Infection for Cancer Immunotherapy. Clin Cancer Res 2024; 30:3355-3357. [PMID: 38869441 DOI: 10.1158/1078-0432.ccr-24-0938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/14/2024]
Abstract
The presence of moieties denoting viral infection is crucial to mount powerful cytotoxic T-cell immune responses acting through innate receptors such as Toll-like receptor 3. For cancer immunotherapy, several safe analogues of viral double-stranded RNA are under clinical development following compelling evidence for efficacy in mouse models. See related article by van Eijck et al., p. 3447.
Collapse
Affiliation(s)
- Ana Martínez-Riaño
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | - Laura Mosteo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
| | - Paula Molero-Glez
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | - Iván Márquez-Rodas
- Servicio de Oncología Médica, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
14
|
Kostinova AM, Latysheva EA, Kostinov MP, Akhmatova NK, Skhodova SA, Vlasenko AE, Cherdantsev AP, Soloveva IL, Khrapunova IA, Loktionova MN, Khromova EA, Poddubikov AA. Comparison of Post-Vaccination Cellular Immune Response in Patients with Common Variable Immune Deficiency. Vaccines (Basel) 2024; 12:843. [PMID: 39203969 PMCID: PMC11360582 DOI: 10.3390/vaccines12080843] [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: 06/17/2024] [Revised: 07/14/2024] [Accepted: 07/18/2024] [Indexed: 09/03/2024] Open
Abstract
BACKGROUND The problem of identifying vaccine-specific T-cell responses is still a matter of debate. Currently, there are no universal, clearly defined, agreed upon criteria for assessing the effectiveness of vaccinations and their immunogenicity for the cellular component of immunity, even for healthy people. But for patients with inborn errors of immunity (IEI), especially those with antibody deficiencies, evaluating cellular immunity holds significant importance. AIM To examine the effect of one and two doses of inactivated adjuvanted subunit influenza vaccines on the expression of endosomal Toll-like receptors (TLRs) on the immune cells and the primary lymphocyte subpopulations in patients with common variable immunodeficiency (CVID). MATERIALS AND METHODS During 2018-2019, six CVID patients received one dose of a quadrivalent adjuvanted influenza vaccine; in 2019-2020, nine patients were vaccinated with two doses of a trivalent inactivated influenza vaccine. The proportion of key lymphocyte subpopulations and expression levels of TLRs were analyzed using flow cytometry with monoclonal antibodies. RESULTS No statistically significant alterations in the absolute values of the main lymphocyte subpopulations were observed in CVID patients before or after vaccination with the different immunization protocols. However, after vaccination, a higher expression of TLR3 and TLR9 in granulocytes, monocytes, and lymphocytes was found in those patients who received two vaccine doses rather than one single dose. CONCLUSION This study marks the first instance of using a simultaneous two-dose vaccination, which is associated with an elevated level of TLR expression in the immune cells. Administration of the adjuvanted vaccines in CVID patients appears promising. Further research into their impact on innate immunity and the development of more effective vaccination regimens is warranted.
Collapse
Affiliation(s)
- Aristitsa Mikhailovna Kostinova
- Federal State Autonomous Educational Institution, Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya Str., 8/2, 119991 Moscow, Russia
- National Research Center Institute of Immunology Federal Medical-Biological Agency of Russia, Kashirskoe Shosse, 24, 115478 Moscow, Russia
| | - Elena Alexandrovna Latysheva
- National Research Center Institute of Immunology Federal Medical-Biological Agency of Russia, Kashirskoe Shosse, 24, 115478 Moscow, Russia
- Faculty of Medicine and Biology, Pirogov Russian National Research Medical University, Ostrovitianov Str., 1, 117513 Moscow, Russia
| | - Mikhail Petrovich Kostinov
- Federal State Autonomous Educational Institution, Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya Str., 8/2, 119991 Moscow, Russia
- Federal State Budgetary Scientific Institution «I.I. Mechnikov Research Institute of Vaccines and Sera», Malyi Kazenniy Pereulok, 5a, 105064 Moscow, Russia
| | - Nelly Kimovna Akhmatova
- Federal State Budgetary Scientific Institution «I.I. Mechnikov Research Institute of Vaccines and Sera», Malyi Kazenniy Pereulok, 5a, 105064 Moscow, Russia
| | - Svetlana Anatolyevna Skhodova
- Federal State Budgetary Scientific Institution «I.I. Mechnikov Research Institute of Vaccines and Sera», Malyi Kazenniy Pereulok, 5a, 105064 Moscow, Russia
| | - Anna Egorovna Vlasenko
- Federal State Budgetary Educational Institution, Higher Education “Samara State Medical University” of the Ministry of Healthcare of the Russian Federation, Chapaevskaya Street, 89, 443099 Samara, Russia
| | - Alexander Petrovich Cherdantsev
- Federal State-Funded Educational Institution, Higher Education “Ulyanovsk State University”, Leo Tolstoy Street, 42, 432017 Ulyanovsk, Russia; (A.P.C.)
| | - Irina Leonidovna Soloveva
- Federal State-Funded Educational Institution, Higher Education “Ulyanovsk State University”, Leo Tolstoy Street, 42, 432017 Ulyanovsk, Russia; (A.P.C.)
| | - Isabella Abramovna Khrapunova
- Federal State Autonomous Educational Institution, Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya Str., 8/2, 119991 Moscow, Russia
| | - Marina Nikolaevna Loktionova
- Federal State Autonomous Educational Institution, Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya Str., 8/2, 119991 Moscow, Russia
- Federal Budget Institute of Science “Central Research Institute of Epidemiology” of the Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, st. Novogireevskaya, 3a, 111123 Moscow, Russia
| | - Ekaterina Alexandrovna Khromova
- Federal State Budgetary Scientific Institution «I.I. Mechnikov Research Institute of Vaccines and Sera», Malyi Kazenniy Pereulok, 5a, 105064 Moscow, Russia
| | - Arseniy Alexandrovich Poddubikov
- Federal State Autonomous Educational Institution, Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya Str., 8/2, 119991 Moscow, Russia
| |
Collapse
|
15
|
Hori A, Toyoura S, Fujiwara M, Taniguchi R, Kano Y, Yamano T, Hanayama R, Nakayama M. MHC class I-dressing is mediated via phosphatidylserine recognition and is enhanced by polyI:C. iScience 2024; 27:109704. [PMID: 38680663 PMCID: PMC11046299 DOI: 10.1016/j.isci.2024.109704] [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: 07/20/2023] [Revised: 02/29/2024] [Accepted: 04/06/2024] [Indexed: 05/01/2024] Open
Abstract
In addition to cross-presentation, cross-dressing plays an important role in the induction of CD8+ T cell immunity. In the process of cross-dressing, conventional dendritic cells (DCs) acquire major histocompatibility complex class I (MHCI) from other cells and subsequently prime CD8+ T cells via the pre-formed antigen-MHCI complexes without antigen processing. However, the mechanisms underlying the cross-dressing pathway, as well as the relative contributions of cross-presentation and cross-dressing to CD8+ T cell priming are not fully understood. Here, we demonstrate that DCs rapidly acquire MHCI-containing membrane fragments from dead cells via the phosphatidylserine recognition-dependent mechanism for cross-dressing. The MHCI dressing is enhanced by a TLR3 ligand polyinosinic-polycytidylic acid (polyI:C). Further, polyI:C promotes not only cross-presentation but also cross-dressing in vivo. Taken together, these results suggest that cross-dressing as well as cross-presentation is involved in inflammatory diseases associated with cell death and type I IFN production.
Collapse
Affiliation(s)
- Arisa Hori
- Laboratory of Immunology and Microbiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan
| | - Saori Toyoura
- Laboratory of Immunology and Microbiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan
| | - Miyu Fujiwara
- Laboratory of Immunology and Microbiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan
| | - Ren Taniguchi
- Laboratory of Immunology and Microbiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan
| | - Yasutaka Kano
- Laboratory of Immunology and Microbiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan
| | - Tomoyoshi Yamano
- Department of Immunology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
- WPI Nano Life Science Institute (NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Rikinari Hanayama
- Department of Immunology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
- WPI Nano Life Science Institute (NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Masafumi Nakayama
- Laboratory of Immunology and Microbiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan
- Research Center for Animal Life Science, Shiga University of Medical Sciences, Seta, Tsukinowa-cho, Otsu, Shiga 520-2192, Japan
| |
Collapse
|
16
|
Sohrabi S, Masoumi J, Naseri B, Ghorbaninezhad F, Alipour S, Kazemi T, Ahmadian Heris J, Aghebati Maleki L, Basirjafar P, Zandvakili R, Doustvandi MA, Baradaran B. STATs signaling pathways in dendritic cells: As potential therapeutic targets? Int Rev Immunol 2024; 43:138-159. [PMID: 37886903 DOI: 10.1080/08830185.2023.2274576] [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: 07/25/2023] [Accepted: 10/16/2023] [Indexed: 10/28/2023]
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells (APCs), including heterogenous populations with phenotypic and functional diversity that coordinate bridging innate and adaptive immunity. Signal transducer and activator of transcriptions (STAT) factors as key proteins in cytokine signaling were shown to play distinct roles in the maturation and antigen presentation of DCs and play a pivotal role in modulating immune responses mediated by DCs such as differentiation of T cells to T helper (Th) 1, Th2 or regulatory T (Treg) cells. This review sheds light on the importance of STAT transcription factors' signaling pathways in different subtypes of DCs and highlights their targeting potential usages for improving DC-based immunotherapies for patients who suffer from cancer or diverse autoimmune conditions according to the type of the STAT transcription factor and its specific activating or inhibitory agent.
Collapse
Affiliation(s)
- Sepideh Sohrabi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Masoumi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahar Naseri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Shiva Alipour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Pedram Basirjafar
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Raziyeh Zandvakili
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
Wang K, Huang H, Zhan Q, Ding H, Li Y. Toll-like receptors in health and disease. MedComm (Beijing) 2024; 5:e549. [PMID: 38685971 PMCID: PMC11057423 DOI: 10.1002/mco2.549] [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: 07/31/2023] [Revised: 03/17/2024] [Accepted: 03/26/2024] [Indexed: 05/02/2024] Open
Abstract
Toll-like receptors (TLRs) are inflammatory triggers and belong to a family of pattern recognition receptors (PRRs) that are central to the regulation of host protective adaptive immune responses. Activation of TLRs in innate immune myeloid cells directs lymphocytes to produce the most appropriate effector responses to eliminate infection and maintain homeostasis of the body's internal environment. Inappropriate TLR stimulation can lead to the development of general autoimmune diseases as well as chronic and acute inflammation, and even cancer. Therefore, TLRs are expected to be targets for therapeutic treatment of inflammation-related diseases, autoimmune diseases, microbial infections, and human cancers. This review summarizes the recent discoveries in the molecular and structural biology of TLRs. The role of different TLR signaling pathways in inflammatory diseases, autoimmune diseases such as diabetes, cardiovascular diseases, respiratory diseases, digestive diseases, and even cancers (oral, gastric, breast, colorectal) is highlighted and summarizes new drugs and related clinical treatments in clinical trials, providing an overview of the potential and prospects of TLRs for the treatment of TLR-related diseases.
Collapse
Affiliation(s)
- Kunyu Wang
- Department of Head and Neck Oncology Surgery, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Hanyao Huang
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Qi Zhan
- Department of Head and Neck Oncology Surgery, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Haoran Ding
- Department of Head and Neck Oncology Surgery, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Yi Li
- Department of Head and Neck Oncology Surgery, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
18
|
Longo Y, Mascaraque SM, Andreacchio G, Werner J, Katahira I, De Marchi E, Pegoraro A, Lebbink RJ, Köhrer K, Petzsch P, Tao R, Di Virgilio F, Adinolfi E, Drexler I. The purinergic receptor P2X7 as a modulator of viral vector-mediated antigen cross-presentation. Front Immunol 2024; 15:1360140. [PMID: 38711513 PMCID: PMC11070468 DOI: 10.3389/fimmu.2024.1360140] [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: 12/22/2023] [Accepted: 04/05/2024] [Indexed: 05/08/2024] Open
Abstract
Introduction Modified Vaccinia Virus Ankara (MVA) is a safe vaccine vector inducing long- lasting and potent immune responses. MVA-mediated CD8+T cell responses are optimally induced, if both, direct- and cross-presentation of viral or recombinant antigens by dendritic cells are contributing. Methods To improve the adaptive immune responses, we investigated the role of the purinergic receptor P2X7 (P2RX7) in MVA-infected feeder cells as a modulator of cross-presentation by non-infected dendritic cells. The infected feeder cells serve as source of antigen and provide signals that help to attract dendritic cells for antigen take up and to license these cells for cross-presentation. Results We demonstrate that presence of an active P2RX7 in major histocompatibility complex (MHC) class I (MHCI) mismatched feeder cells significantly enhanced MVA-mediated antigen cross-presentation. This was partly regulated by P2RX7-specific processes, such as the increased availability of extracellular particles as well as the altered cellular energy metabolism by mitochondria in the feeder cells. Furthermore, functional P2RX7 in feeder cells resulted in a delayed but also prolonged antigen expression after infection. Discussion We conclude that a combination of the above mentioned P2RX7-depending processes leads to significantly increased T cell activation via cross- presentation of MVA-derived antigens. To this day, P2RX7 has been mostly investigated in regards to neuroinflammatory diseases and cancer progression. However, we report for the first time the crucial role of P2RX7 for antigen- specific T cell immunity in a viral infection model.
Collapse
Affiliation(s)
- Ylenia Longo
- Institute of Virology, Universitätsklinikum Düsseldorf, Düsselorf, Germany
| | | | | | - Julia Werner
- Institute of Molecular Medicine II, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| | - Ichiro Katahira
- Institute of Molecular Medicine II, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| | - Elena De Marchi
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Anna Pegoraro
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Robert Jan Lebbink
- Institute of Infection Immunity, University of Utrecht, Utrecht, Netherlands
| | - Karl Köhrer
- Biological and Medical Research Center (BMFZ), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Patrick Petzsch
- Biological and Medical Research Center (BMFZ), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Ronny Tao
- Institute of Virology, Universitätsklinikum Düsseldorf, Düsselorf, Germany
| | | | - Elena Adinolfi
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Ingo Drexler
- Institute of Virology, Universitätsklinikum Düsseldorf, Düsselorf, Germany
| |
Collapse
|
19
|
Adams CS, Kim H, Burtner AE, Lee DS, Dobbins C, Criswell C, Coventry B, Kim HM, King NP. De novo design of protein minibinder agonists of TLR3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.17.589973. [PMID: 38659926 PMCID: PMC11042314 DOI: 10.1101/2024.04.17.589973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Toll-like Receptor 3 (TLR3) is a pattern recognition receptor that initiates antiviral immune responses upon binding double-stranded RNA (dsRNA). Several nucleic acid-based TLR3 agonists have been explored clinically as vaccine adjuvants in cancer and infectious disease, but present substantial manufacturing and formulation challenges. Here, we use computational protein design to create novel miniproteins that bind to human TLR3 with nanomolar affinities. Cryo-EM structures of two minibinders in complex with TLR3 reveal that they bind the target as designed, although one partially unfolds due to steric competition with a nearby N-linked glycan. Multimeric forms of both minibinders induce NF-κB signaling in TLR3-expressing cell lines, demonstrating that they may have therapeutically relevant biological activity. Our work provides a foundation for the development of specific, stable, and easy-to-formulate protein-based agonists of TLRs and other pattern recognition receptors.
Collapse
Affiliation(s)
- Chloe S. Adams
- Institute for Protein Design, University of Washington, Seattle, WA, 98195 USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195 USA
| | - Hyojin Kim
- Center for Biomolecular & Cellular Structure, Institute for Basic Science (IBS), Daejeon 34126, South Korea
| | - Abigail E. Burtner
- Institute for Protein Design, University of Washington, Seattle, WA, 98195 USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195 USA
| | - Dong Sun Lee
- Center for Biomolecular & Cellular Structure, Institute for Basic Science (IBS), Daejeon 34126, South Korea
| | - Craig Dobbins
- Institute for Protein Design, University of Washington, Seattle, WA, 98195 USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195 USA
| | - Cameron Criswell
- Institute for Protein Design, University of Washington, Seattle, WA, 98195 USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195 USA
| | - Brian Coventry
- Institute for Protein Design, University of Washington, Seattle, WA, 98195 USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195 USA
| | - Ho Min Kim
- Center for Biomolecular & Cellular Structure, Institute for Basic Science (IBS), Daejeon 34126, South Korea
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, South Korea
| | - Neil P. King
- Institute for Protein Design, University of Washington, Seattle, WA, 98195 USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195 USA
| |
Collapse
|
20
|
Carroll SL, Pasare C, Barton GM. Control of adaptive immunity by pattern recognition receptors. Immunity 2024; 57:632-648. [PMID: 38599163 PMCID: PMC11037560 DOI: 10.1016/j.immuni.2024.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 04/12/2024]
Abstract
One of the most significant conceptual advances in immunology in recent history is the recognition that signals from the innate immune system are required for induction of adaptive immune responses. Two breakthroughs were critical in establishing this paradigm: the identification of dendritic cells (DCs) as the cellular link between innate and adaptive immunity and the discovery of pattern recognition receptors (PRRs) as a molecular link that controls innate immune activation as well as DC function. Here, we recount the key events leading to these discoveries and discuss our current understanding of how PRRs shape adaptive immune responses, both indirectly through control of DC function and directly through control of lymphocyte function. In this context, we provide a conceptual framework for how variation in the signals generated by PRR activation, in DCs or other cell types, can influence T cell differentiation and shape the ensuing adaptive immune response.
Collapse
Affiliation(s)
- Shaina L Carroll
- Division of Immunology & Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | - Chandrashekhar Pasare
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH USA
| | - Gregory M Barton
- Division of Immunology & Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA; Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720 USA.
| |
Collapse
|
21
|
Kawai T, Ikegawa M, Ori D, Akira S. Decoding Toll-like receptors: Recent insights and perspectives in innate immunity. Immunity 2024; 57:649-673. [PMID: 38599164 DOI: 10.1016/j.immuni.2024.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/18/2024] [Accepted: 03/05/2024] [Indexed: 04/12/2024]
Abstract
Toll-like receptors (TLRs) are an evolutionarily conserved family in the innate immune system and are the first line of host defense against microbial pathogens by recognizing pathogen-associated molecular patterns (PAMPs). TLRs, categorized into cell surface and endosomal subfamilies, recognize diverse PAMPs, and structural elucidation of TLRs and PAMP complexes has revealed their intricate mechanisms. TLRs activate common and specific signaling pathways to shape immune responses. Recent studies have shown the importance of post-transcriptional regulation in TLR-mediated inflammatory responses. Despite their protective functions, aberrant responses of TLRs contribute to inflammatory and autoimmune disorders. Understanding the delicate balance between TLR activation and regulatory mechanisms is crucial for deciphering their dual role in immune defense and disease pathogenesis. This review provides an overview of recent insights into the history of TLR discovery, elucidation of TLR ligands and signaling pathways, and their relevance to various diseases.
Collapse
Affiliation(s)
- Taro Kawai
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Nara 630-0192, Japan; Life Science Collaboration Center (LiSCo), Nara Institute of Science and Technology (NAIST), Nara 630-0192, Japan.
| | - Moe Ikegawa
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Nara 630-0192, Japan
| | - Daisuke Ori
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Nara 630-0192, Japan
| | - Shizuo Akira
- Center for Advanced Modalities and DSS (CAMaD), Osaka University, Osaka 565-0871, Japan; Laboratory of Host Defense, Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan; Department of Host Defense, Research Institute for Microbial Diseases (RIMD), Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
22
|
Wang H, Medina R, Ye J, Zhang Y, Chakraborty S, Valenzuela A, Uher O, Hadrava Vanova K, Sun M, Sang X, Park DM, Zenka J, Gilbert MR, Pacak K, Zhuang Z. rWTC-MBTA Vaccine Induces Potent Adaptive Immune Responses Against Glioblastomas via Dynamic Activation of Dendritic Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308280. [PMID: 38298111 PMCID: PMC11005728 DOI: 10.1002/advs.202308280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/19/2023] [Indexed: 02/02/2024]
Abstract
Despite strides in immunotherapy, glioblastoma multiforme (GBM) remains challenging due to low inherent immunogenicity and suppressive tumor microenvironment. Converting "cold" GBMs to "hot" is crucial for immune activation and improved outcomes. This study comprehensively characterized a therapeutic vaccination strategy for preclinical GBM models. The vaccine consists of Mannan-BAM-anchored irradiated whole tumor cells, Toll-like receptor ligands [lipoteichoic acid (LTA), polyinosinic-polycytidylic acid (Poly (I:C)), and resiquimod (R-848)], and anti-CD40 agonistic antibody (rWTC-MBTA). Intracranial GBM models (GL261, SB28 cells) are used to evaluate the vaccine efficacy. A substantial number of vaccinated mice exhibited complete regression of GBM tumors in a T-cell-dependent manner, with no significant toxicity. Long-term tumor-specific immune memory is confirmed upon tumor rechallenge. In the vaccine-draining lymph nodes of the SB28 model, rWTC-MBTA vaccination triggered a major rise in conventional dendritic cell type 1 (cDC1) 12 h post-treatment, followed by an increase in conventional dendritic cell type 2 (cDC2), monocyte-derived dendritic cell (moDC), and plasmacytoid dendritic cell (pDC) on Day 5 and Day 13. Enhanced cytotoxicity of CD4+ and CD8+ T cells in vaccinated mice is verified in co-culture with tumor cells. Analyses of immunosuppressive signals (T-cell exhaustion, myeloid-derived suppressor cells (MDSC), M2 macrophages) in the GBM microenvironment suggest potential combinations with other immunotherapies for enhanced efficacy. In conclusion, the authors findings demonstrate that rWTC-MBTA induces potent and long-term adaptive immune responses against GBM.
Collapse
Affiliation(s)
- Herui Wang
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
- Present address:
Staff Scientist Neuro‐Oncology BranchNational Cancer Institute Center for Cancer ResearchNational Institutes of HealthBuilding 37 Room 100437 Convent Dr.BethesdaMD20892USA
| | - Rogelio Medina
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Juan Ye
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Yaping Zhang
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | | | - Alex Valenzuela
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Ondrej Uher
- Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of Health9000 Rockville PikeBethesdaMD20892USA
| | - Katerina Hadrava Vanova
- Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of Health9000 Rockville PikeBethesdaMD20892USA
| | - Mitchell Sun
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Xueyu Sang
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Deric M. Park
- John Theurer Cancer CenterHUMCHackensack Meridian School of Medicine92 2nd StHackensackNJ07601USA
| | - Jan Zenka
- Department of Medical BiologyFaculty of ScienceUniversity of South BohemiaČeské Budějovice37005Czech Republic
| | - Mark R. Gilbert
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of Health9000 Rockville PikeBethesdaMD20892USA
| | - Zhengping Zhuang
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
- Present address:
Senior Investigator Neuro‐Oncology BranchNational Cancer Institute Center for Cancer ResearchNational Institutes of HealthBuilding 37 Room 100037 Convent DrBethesdaMD20892USA
| |
Collapse
|
23
|
Yau E, Yang L, Chen Y, Umstead TM, Stanley AE, Halstead ES, Gandhi CK, Yewdell JW, Chroneos ZC. SP-R210 isoforms of Myosin18A modulate endosomal sorting and recognition of influenza A virus infection in macrophages. Microbes Infect 2024; 26:105280. [PMID: 38135024 PMCID: PMC10948314 DOI: 10.1016/j.micinf.2023.105280] [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: 05/28/2023] [Revised: 12/12/2023] [Accepted: 12/17/2023] [Indexed: 12/24/2023]
Abstract
Influenza A virus (IAV) infection causes acute and often lethal inflammation in the lung. The role of macrophages in this adverse inflammation is partially understood. The surfactant protein A receptor 210 (SP-R210) consists of two isoforms, a long (L) SP-R210L and a short (S) SP-R210S isoform encoded by alternative splicing of the myosin 18A gene. We reported that disruption of SP-R210L enhances cytosolic and endosomal antiviral response pathways. Here, we report that SP-R210L antagonizes type I interferon β (IFNβ), as depletion of SP-R210L potentiates IFNβ secretion. SP-R210 antibodies enhance and attenuate IFNβ secretion in SP-R210L replete and deficient macrophages, respectively, indicating that SP-R210 isoform stoichiometry alters macrophage function intrinsically. This reciprocal response is coupled to unopposed and restricted expression of viral genes in control and SP-R210L-deficient macrophages, respectively. Human monocytic cells with sub-stoichiometric expression of SP-R210L resist IAV infection, whereas alveolar macrophages with increased abundance of SP-R210L permit viral gene expression similar to murine macrophages. Uptake and membrane binding studies show that lack of SP-R210 isoforms does not impair IAV binding and internalization. Lack of SP-R210L, however, results in macropinocytic retention of the virus that depends on both SP-R210S and interferon-inducible transmembrane protein-3 (IFITM3). Mass spectrometry and Western blot analyses indicate that SP-R210 isoforms modulate differential recruitment of the Rho-family GTPase RAC1 and guanine nucleotide exchange factors. Our study suggests that SP-R210 isoforms modulate RAC-dependent macropinosomal sorting of IAV to discrete endosomal and lysosomal compartments that either permit or prevent endolysosomal escape and inflammatory sensing of viral genomes in macrophages.
Collapse
Affiliation(s)
- Eric Yau
- Department of Pediatrics, Division of Perinatal-Neonatal Medicine, Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Linlin Yang
- Department of Pediatrics, Division of Perinatal-Neonatal Medicine, Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Yan Chen
- Department of Pediatrics, Division of Perinatal-Neonatal Medicine, Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Todd M Umstead
- Department of Pediatrics, Division of Perinatal-Neonatal Medicine, Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Anne E Stanley
- Mass Spectrometry Core, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - E Scott Halstead
- Department of Pediatrics, Division of Perinatal-Neonatal Medicine, Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pediatrics, Division of Pediatric Critical Care Medicine, Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Chintan K Gandhi
- Department of Pediatrics, Division of Perinatal-Neonatal Medicine, Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Jonathan W Yewdell
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Zissis C Chroneos
- Department of Pediatrics, Division of Perinatal-Neonatal Medicine, Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, USA.
| |
Collapse
|
24
|
Liu YG, Jin SW, Zhang SS, Xia TJ, Liao YH, Pan RL, Yan MZ, Chang Q. Interferon lambda in respiratory viral infection: immunomodulatory functions and antiviral effects in epithelium. Front Immunol 2024; 15:1338096. [PMID: 38495892 PMCID: PMC10940417 DOI: 10.3389/fimmu.2024.1338096] [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: 11/14/2023] [Accepted: 02/19/2024] [Indexed: 03/19/2024] Open
Abstract
Type III interferon (IFN-λ), a new member of the IFN family, was initially considered to possess antiviral functions similar to those of type I interferon, both of which are induced via the JAK/STAT pathway. Nevertheless, recent findings demonstrated that IFN-λ exerts a nonredundant antiviral function at the mucosal surface, preferentially produced in epithelial cells in contrast to type I interferon, and its function cannot be replaced by type I interferon. This review summarizes recent studies showing that IFN-λ inhibits the spread of viruses from the cell surface to the body. Further studies have found that the role of IFN-λ is not only limited to the abovementioned functions, but it can also can exert direct and/or indirect effects on immune cells in virus-induced inflammation. This review focuses on the antiviral activity of IFN-λ in the mucosal epithelial cells and its action on immune cells and summarizes the pathways by which IFN-λ exerts its action and differentiates it from other interferons in terms of mechanism. Finally, we conclude that IFN-λ is a potent epidermal antiviral factor that enhances the respiratory mucosal immune response and has excellent therapeutic potential in combating respiratory viral infections.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ming-Zhu Yan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Chang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
25
|
Thomas S, Pak J, Doss-Gollin S, Ryff K, Beijnen E, Pedersen GK, Christensen D, Levy O, van Haren SD. Human In vitro Modeling Identifies Adjuvant Combinations that Unlock Antigen Cross-presentation and Promote T-helper 1 Development in Newborns, Adults and Elders. J Mol Biol 2024; 436:168446. [PMID: 38242283 PMCID: PMC10922990 DOI: 10.1016/j.jmb.2024.168446] [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: 08/31/2023] [Revised: 12/08/2023] [Accepted: 01/11/2024] [Indexed: 01/21/2024]
Abstract
Adjuvants are vaccine components that can boost the type, magnitude, breadth, and durability of an immune response. We have previously demonstrated that certain adjuvant combinations can act synergistically to enhance and shape immunogenicity including promotion of Th1 and cytotoxic T-cell development. These combinations also promoted protective immunity in vulnerable populations such as newborns. In this study, we employed combined antigen-specific human in vitro models to identify adjuvant combinations that could synergistically promote the expansion of vaccine-specific CD4+ cells, induce cross-presentation on MHC class I, resulting in antigen-specific activation of CD8+ cells, and direct the balance of immune response to favor the production of Th1-promoting cytokines. A screen of 78 adjuvant combinations identified several T cell-potentiating adjuvant combinations. Remarkably, a combination of TLR9 and STING agonists (CpG + 2,3-cGAMP) promoted influenza-specific CD4+ and CD8+ T cell activation and selectively favored production of Th1-polarizing cytokines TNF and IL-12p70 over co-regulated cytokines IL-6 and IL-12p40, respectively. Phenotypic reprogramming towards cDC1-type dendritic cells by CpG + 2,3-cGAMP was also observed. Finally, we characterized the molecular mechanism of this adjuvant combination including the ability of 2,3-cGAMP to enhance DC expression of TLR9 and the dependency of antigen-presenting cell activation on the Sec22b protein important to endoplasmic reticulum-Golgi vesicle trafficking. The identification of the adjuvant combination CpG + 2,3-cGAMP may therefore prove key to the future development of vaccines against respiratory viral infections tailored for the functionally distinct immune systems of vulnerable populations such as older adults and newborns.
Collapse
Affiliation(s)
- Sanya Thomas
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Jensen Pak
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Simon Doss-Gollin
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Kevin Ryff
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Elisabeth Beijnen
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Gabriel K Pedersen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark; Department of Immunology and Microbiology, University of Copenhagen, Denmark
| | - Dennis Christensen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA; Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA; Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Simon D van Haren
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
26
|
Rashid F, Xie Z, Li M, Xie Z, Luo S, Xie L. Roles and functions of IAV proteins in host immune evasion. Front Immunol 2023; 14:1323560. [PMID: 38152399 PMCID: PMC10751371 DOI: 10.3389/fimmu.2023.1323560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/30/2023] [Indexed: 12/29/2023] Open
Abstract
Influenza A viruses (IAVs) evade the immune system of the host by several regulatory mechanisms. Their genomes consist of eight single-stranded segments, including nonstructural proteins (NS), basic polymerase 1 (PB1), basic polymerase 2 (PB2), hemagglutinin (HA), acidic polymerase (PA), matrix (M), neuraminidase (NA), and nucleoprotein (NP). Some of these proteins are known to suppress host immune responses. In this review, we discuss the roles, functions and underlying strategies adopted by IAV proteins to escape the host immune system by targeting different proteins in the interferon (IFN) signaling pathway, such as tripartite motif containing 25 (TRIM25), inhibitor of nuclear factor κB kinase (IKK), mitochondrial antiviral signaling protein (MAVS), Janus kinase 1 (JAK1), type I interferon receptor (IFNAR1), interferon regulatory factor 3 (IRF3), IRF7, and nuclear factor-κB (NF-κB). To date, the IAV proteins NS1, NS2, PB1, PB1-F2, PB2, HA, and PA have been well studied in terms of their roles in evading the host immune system. However, the detailed mechanisms of NS3, PB1-N40, PA-N155, PA-N182, PA-X, M42, NA, and NP have not been well studied with respect to their roles in immune evasion. Moreover, we also highlight the future perspectives of research on IAV proteins.
Collapse
Affiliation(s)
- Farooq Rashid
- Department of Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Guangxi Key Laboratory of Veterinary Biotechnology, Nanning, China
- Key Laboratory of China (Guangxi)-ASEAN Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
| | - Zhixun Xie
- Department of Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Guangxi Key Laboratory of Veterinary Biotechnology, Nanning, China
- Key Laboratory of China (Guangxi)-ASEAN Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
| | - Meng Li
- Department of Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Guangxi Key Laboratory of Veterinary Biotechnology, Nanning, China
- Key Laboratory of China (Guangxi)-ASEAN Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
| | - Zhiqin Xie
- Department of Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Guangxi Key Laboratory of Veterinary Biotechnology, Nanning, China
- Key Laboratory of China (Guangxi)-ASEAN Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
| | - Sisi Luo
- Department of Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Guangxi Key Laboratory of Veterinary Biotechnology, Nanning, China
- Key Laboratory of China (Guangxi)-ASEAN Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
| | - Liji Xie
- Department of Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Guangxi Key Laboratory of Veterinary Biotechnology, Nanning, China
- Key Laboratory of China (Guangxi)-ASEAN Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
| |
Collapse
|
27
|
MacNabb BW, Kline J. MHC cross-dressing in antigen presentation. Adv Immunol 2023; 159:115-147. [PMID: 37996206 DOI: 10.1016/bs.ai.2023.07.001] [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] [Indexed: 11/25/2023]
Abstract
Dendritic cells (DCs) orchestrate T cell responses by presenting antigenic peptides on major histocompatibility complex (MHC) and providing costimulation and other instructive signals. Professional antigen presenting cells (APCs), including DCs, are uniquely capable of generating and presenting peptide antigens derived from exogenous proteins. In addition to these canonical cross-presentation and MHC-II presentation pathways, APCs can also display exogenous peptide/MHC (p/MHC) acquired from neighboring cells and extracellular vesicles (EVs). This process, known as MHC cross-dressing, has been implicated in the regulation of T cell responses in a variety of in vivo contexts, including allogeneic solid organ transplantation, tumors, and viral infection. Although the occurrence of MHC cross-dressing has been clearly demonstrated, the importance of this antigen presentation mechanism continues to be elucidated. The contribution of MHC cross-dressing to overall antigen presentation has been obfuscated by the fact that DCs express the same MHC alleles as all other cells in the host, making it difficult to distinguish p/MHC generated within the DC from p/MHC acquired from another cell. As a result, much of what is known about MHC cross-dressing comes from studies using allogeneic organ transplantation and bone marrow chimeric mice, though recent development of mice bearing conditional knockout MHC and β2-microglobulin alleles should facilitate substantial progress in the coming years. In this review, we highlight recent advances in our understanding of MHC cross-dressing and its role in activating T cell responses in various contexts, as well as the experimental insights into the mechanism by which it occurs.
Collapse
Affiliation(s)
- Brendan W MacNabb
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States.
| | - Justin Kline
- Department of Medicine, Committee on Immunology, and Committee on Cancer Biology, University of Chicago, Chicago, IL, United States.
| |
Collapse
|
28
|
Kostinova AM, Latysheva EA, Akhmatova NK, Vlasenko AE, Skhodova SA, Khromova EA, Linok AV, Poddubikov AA, Latysheva TV, Kostinov MP. Expression of Toll-like Receptors on the Immune Cells in Patients with Common Variable Immune Deficiency after Different Schemes of Influenza Vaccination. Viruses 2023; 15:2091. [PMID: 37896869 PMCID: PMC10611272 DOI: 10.3390/v15102091] [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: 09/14/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND for the first time, the effect of one and two doses of adjuvanted influenza vaccines on toll-like receptors (TLRs) in patients with common variable immunodeficiency (CVID) was studied and compared (primary vaccination with one vs. two doses, primary vs. repeated vaccination). MATERIALS AND METHODS Six patients received one dose of quadrivalent adjuvanted influenza vaccine during the 2018-2019 and 2019-2020 influenza seasons, and nine patients with CVID received two doses of trivalent inactivated influenza vaccine during 2019-2020. Expression of TLRs was measured by flow cytometry. RESULTS The expression of toll-like receptors in patients with CVID was noted both with repeated (annual) administration of the influenza vaccine and in most cases was accompanied by an increase in the proportion of granulocytes (TLR3 and TLR9), lymphocytes (TLR3 and TLR8), and monocytes (TLR3 and TLR9). When carried out for the first time as a simultaneous vaccination with two doses it was accompanied by an increase in the proportion of granulocytes, lymphocytes expressing TLR9, and on monocytes-TLR3 and TLR9. CONCLUSION in CVID patients, the use of adjuvanted vaccines is promising, and research on the influence of the innate immunity and more effective regimens should be continued.
Collapse
Affiliation(s)
- Aristitsa Mikhailovna Kostinova
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya Str. 8/2, 119991 Moscow, Russia (A.A.P.); (M.P.K.)
- National Research Center—Institute of Immunology Federal Medical-Biological Agency of Russia, Kashirskoe Shosse, 24, 115478 Moscow, Russia (T.V.L.)
| | - Elena Alexandrovna Latysheva
- National Research Center—Institute of Immunology Federal Medical-Biological Agency of Russia, Kashirskoe Shosse, 24, 115478 Moscow, Russia (T.V.L.)
- Pirogov Russian National Research Medical University, Ostrovitianov Str. 1, 117997 Moscow, Russia
| | - Nelly Kimovna Akhmatova
- Russian Federal State Budgetary Scientific Institution «I.I. Mechnikov Research Institute of Vaccines and Sera», Malyi Kazenniy Pereulok, 5a, 105064 Moscow, Russia (E.A.K.)
| | - Anna Egorovna Vlasenko
- Federal State Budgetary Educational Institution of Higher Education “Samara State Medical University” of the Ministry of Healthcare of the Russian Federation, Chapaevskaya Street, 89, 443099 Samara, Russia
| | - Svetlana Anatolyevna Skhodova
- Russian Federal State Budgetary Scientific Institution «I.I. Mechnikov Research Institute of Vaccines and Sera», Malyi Kazenniy Pereulok, 5a, 105064 Moscow, Russia (E.A.K.)
| | - Ekaterina Alexandrovna Khromova
- Russian Federal State Budgetary Scientific Institution «I.I. Mechnikov Research Institute of Vaccines and Sera», Malyi Kazenniy Pereulok, 5a, 105064 Moscow, Russia (E.A.K.)
| | - Andrey Viktorovich Linok
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya Str. 8/2, 119991 Moscow, Russia (A.A.P.); (M.P.K.)
| | - Arseniy Alexandrovich Poddubikov
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya Str. 8/2, 119991 Moscow, Russia (A.A.P.); (M.P.K.)
| | - Tatyana Vasilievna Latysheva
- National Research Center—Institute of Immunology Federal Medical-Biological Agency of Russia, Kashirskoe Shosse, 24, 115478 Moscow, Russia (T.V.L.)
| | - Mikhail Petrovich Kostinov
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya Str. 8/2, 119991 Moscow, Russia (A.A.P.); (M.P.K.)
- Russian Federal State Budgetary Scientific Institution «I.I. Mechnikov Research Institute of Vaccines and Sera», Malyi Kazenniy Pereulok, 5a, 105064 Moscow, Russia (E.A.K.)
| |
Collapse
|
29
|
Cao LL, Kagan JC. Targeting innate immune pathways for cancer immunotherapy. Immunity 2023; 56:2206-2217. [PMID: 37703879 PMCID: PMC10591974 DOI: 10.1016/j.immuni.2023.07.018] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/28/2023] [Accepted: 07/26/2023] [Indexed: 09/15/2023]
Abstract
The innate immune system is critical for inducing durable and protective T cell responses to infection and has been increasingly recognized as a target for cancer immunotherapy. In this review, we present a framework wherein distinct innate immune signaling pathways activate five key dendritic cell activities that are important for T cell-mediated immunity. We discuss molecular pathways that can agonize these activities and highlight that no single pathway can agonize all activities needed for durable immunity. The immunological distinctions between innate immunotherapy administration to the tumor microenvironment versus administration via vaccination are examined, with particular focus on the strategies that enhance dendritic cell migration, interferon expression, and interleukin-1 family cytokine production. In this context, we argue for the importance of appreciating necessity vs. sufficiency when considering the impact of innate immune signaling in inflammation and protective immunity and offer a conceptual guideline for the development of efficacious cancer immunotherapies.
Collapse
Affiliation(s)
- Longyue L Cao
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Jonathan C Kagan
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
30
|
Abdel-Maksoud EM, Daha AAEF, Taha NM, Lebda MA, Sadek KM, Alshahrani MY, Ahmed AE, Shukry M, Fadl SE, Elfeky M. Effects of ginger extract and/or propolis extract on immune system parameters of vaccinated broilers. Poult Sci 2023; 102:102903. [PMID: 37506621 PMCID: PMC10413198 DOI: 10.1016/j.psj.2023.102903] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/18/2023] [Accepted: 06/24/2023] [Indexed: 07/30/2023] Open
Abstract
Newcastle disease (ND), avian influenza (AI, H5N8), and infectious bronchitis (IB) are important diseases in the poultry industry and cause significant losses. Vaccination is the most practical method for controlling infectious diseases. To reduce vaccination costs and several disorders in poultry farms, using herbal water supplements for immunomodulation with vaccination is critical to improving or preventing some conditions in the poultry industry. However, drinking water supplementation of ginger extract (GE)/propolis extract (PE) alone/in combination may increase broilers' humoral and cellular immunity due to the immunomodulatory effects of ginger and propolis. This protocol aimed to see how GE/PE alone or in combination improved the immunity, immune organ gene expression, and histology of the immune organs of broilers for 35 d after vaccination against NDV, H5N8, IBV, and IBDV. The chicks were dispensed into 5 groups according to GE and/or PE with vaccination. The control group was offered normal drinking water without any supplements or vaccinations. The GE group was supplemented with ginger extract (1 mL/L drinking water) in the drinking water before and after vaccination for 2 and 3 d, respectively. The GE+PE group was supplemented with GE (0.5 mL/L drinking water) and PE (0.5 mL/L drinking water) in the drinking water before and after vaccination for 2 and 3 d, respectively. The PE group was supplemented with propolis extract (1 mL/L drinking water) in the drinking water before and after vaccination for 2 and 3 d, respectively. The fifth group was the vaccinated untreated group. This experiment showed the immunomodulatory properties of GE and/or PE against 3 common diseases, NDV, AI, and IB, in broiler chicken farms for 35 d applied to a vaccination program. Thus, ginger extract and propolis extract supplementation in drinking water increased antibody titer, INF, IL10, and IL2 and TLR3 gene expression in the bursa of Fabricius, thymus, and spleen, respectively, as well as cellular immunity as indicated by increased CD3, CD4, and CD8 in the bursa of Fabricius, thymus, and spleen, respectively, with normal lymphocytes in the medulla of the bursa, thymus, and spleen. In conclusion, propolis extracts alone or with GE improved all of the metrics mentioned above without harming the histology of the immune organs.
Collapse
Affiliation(s)
- Eman M Abdel-Maksoud
- Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 21526, Egypt
| | - Ahmed Abd El Fattah Daha
- Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 21526, Egypt
| | - Nabil M Taha
- Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 21526, Egypt
| | - Mohamed A Lebda
- Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 21526, Egypt
| | - Kadry M Sadek
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt
| | - Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, 9088, Saudi Arabia
| | - Ahmed Ezzat Ahmed
- Biology Department, College of Science, King Khalid University, Abha, 61413, Saudi Arabia
| | - Mustafa Shukry
- Department of Physiology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt.
| | - Sabreen E Fadl
- Biochemistry Department, Faculty of Veterinary Medicine, Matrouh University, Matrouh, 51744 Egypt
| | - Mohamed Elfeky
- Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 21526, Egypt
| |
Collapse
|
31
|
Marek J, Hanesch L, Krabbe T, El Khawanky N, Heidegger S, Altomonte J. Oncolytic virotherapy with chimeric VSV-NDV synergistically supports RIG-I-dependent checkpoint inhibitor immunotherapy. Mol Ther Oncolytics 2023; 30:117-131. [PMID: 37654972 PMCID: PMC10465858 DOI: 10.1016/j.omto.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023] Open
Abstract
Unraveling the complexities of the tumor microenvironment (TME) and its correlation with responsiveness to immunotherapy has become a main focus in overcoming resistance to such treatments. Targeting tumor-intrinsic retinoic acid-inducible gene-I (RIG-I), a sensor for viral RNA, was shown to transform the TME from an immunogenically "cold" state to an inflamed, "hot" lesion, which we demonstrated previously to be a crucial mediator of the efficacy of immune checkpoint inhibition with anti-cytotoxic T lymphocyte-associated protein 4 (CTLA-4). In this study, we focus on the chimeric oncolytic virus vesicular stomatitis virus (VSV)-Newcastle disease virus (NDV), comprised of genetic components of VSV and NDV, and we investigate its utility to support tumor-intrinsic RIG-I-dependent therapy with anti-CTLA-4. Overall, we demonstrate that treatment with VSV-NDV efficiently delays tumor growth and significantly prolongs survival in a murine model of malignant melanoma, which was further enhanced in combination with anti-CTLA-4. Although the direct oncolytic and pro-inflammatory effects of VSV-NDV therapy were independent of RIG-I activation, the synergism with anti-CTLA-4 therapy and associated activation of tumor-specific T cells was critically dependent on active RIG-I signaling in tumor cells. This work highlights the therapeutic value of utilizing an immune-stimulatory oncolytic virus to sensitize tumors to immune checkpoint inhibition.
Collapse
Affiliation(s)
- Janina Marek
- Department of Internal Medicine II, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Lorenz Hanesch
- Department of Internal Medicine II, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Teresa Krabbe
- Department of Internal Medicine II, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Nadia El Khawanky
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - Simon Heidegger
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - Jennifer Altomonte
- Department of Internal Medicine II, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| |
Collapse
|
32
|
Li H, Wang A, Zhang Y, Wei F. Diverse roles of lung macrophages in the immune response to influenza A virus. Front Microbiol 2023; 14:1260543. [PMID: 37779697 PMCID: PMC10534047 DOI: 10.3389/fmicb.2023.1260543] [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: 07/18/2023] [Accepted: 08/16/2023] [Indexed: 10/03/2023] Open
Abstract
Influenza viruses are one of the major causes of human respiratory infections and the newly emerging and re-emerging strains of influenza virus are the cause of seasonal epidemics and occasional pandemics, resulting in a huge threat to global public health systems. As one of the early immune cells can rapidly recognize and respond to influenza viruses in the respiratory, lung macrophages play an important role in controlling the severity of influenza disease by limiting viral replication, modulating the local inflammatory response, and initiating subsequent adaptive immune responses. However, influenza virus reproduction in macrophages is both strain- and macrophage type-dependent, and ineffective replication of some viral strains in mouse macrophages has been observed. This review discusses the function of lung macrophages in influenza virus infection in order to better understand the pathogenesis of the influenza virus.
Collapse
Affiliation(s)
- Haoning Li
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Aoxue Wang
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Yuying Zhang
- School of Biological Science and Technology, University of Jinan, Jinan, China
| | - Fanhua Wei
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| |
Collapse
|
33
|
Ren H, Jia W, Xie Y, Yu M, Chen Y. Adjuvant physiochemistry and advanced nanotechnology for vaccine development. Chem Soc Rev 2023; 52:5172-5254. [PMID: 37462107 DOI: 10.1039/d2cs00848c] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Vaccines comprising innovative adjuvants are rapidly reaching advanced translational stages, such as the authorized nanotechnology adjuvants in mRNA vaccines against COVID-19 worldwide, offering new strategies to effectively combat diseases threatening human health. Adjuvants are vital ingredients in vaccines, which can augment the degree, extensiveness, and longevity of antigen specific immune response. The advances in the modulation of physicochemical properties of nanoplatforms elevate the capability of adjuvants in initiating the innate immune system and adaptive immunity, offering immense potential for developing vaccines against hard-to-target infectious diseases and cancer. In this review, we provide an essential introduction of the basic principles of prophylactic and therapeutic vaccination, key roles of adjuvants in augmenting and shaping immunity to achieve desired outcomes and effectiveness, and the physiochemical properties and action mechanisms of clinically approved adjuvants for humans. We particularly focus on the preclinical and clinical progress of highly immunogenic emerging nanotechnology adjuvants formulated in vaccines for cancer treatment or infectious disease prevention. We deliberate on how the immune system can sense and respond to the physicochemical cues (e.g., chirality, deformability, solubility, topology, and chemical structures) of nanotechnology adjuvants incorporated in the vaccines. Finally, we propose possible strategies to accelerate the clinical implementation of nanotechnology adjuvanted vaccines, such as in-depth elucidation of nano-immuno interactions, antigen identification and optimization by the deployment of high-dimensional multiomics analysis approaches, encouraging close collaborations among scientists from different scientific disciplines and aggressive exploration of novel nanotechnologies.
Collapse
Affiliation(s)
- Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Wencong Jia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Yujie Xie
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|
34
|
Pirillo C, Al Khalidi S, Sims A, Devlin R, Zhao H, Pinto R, Jasim S, Shearer PA, Shergold AL, Donnelly H, Bravo-Blas A, Loney C, Perona-Wright G, Hutchinson E, Roberts EW. Cotransfer of antigen and contextual information harmonizes peripheral and lymph node conventional dendritic cell activation. Sci Immunol 2023; 8:eadg8249. [PMID: 37478193 PMCID: PMC7616026 DOI: 10.1126/sciimmunol.adg8249] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 06/29/2023] [Indexed: 07/23/2023]
Abstract
T cell responses against infections and cancer are directed by conventional dendritic cells (cDCs) in lymph nodes distant from the site of challenge. Migratory cDCs, which travel from the tissue to the lymph node, not only drive initial T cell activation but also transfer antigen to lymph node-resident cDCs. These resident cells have essential roles defining the character of the resulting T cell response; however, it is unknown how they can appropriately process and present antigens to suitably direct responses given their spatial separation. Here, using a novel strain of influenza A and a modified melanoma model, we show that tissue and lymph node cDC activation is harmonized and that this is driven by cotransfer of contextual cues. In the tumor, incomplete cDC activation in the tumor microenvironment is mirrored by lymph node-resident cDCs, whereas during influenza infection, pathogen-associated molecular patterns cotransferred with antigen drive TLR signaling in resident cDCs and their subsequent robust activation. This cotransfer mechanism explains how individual antigens can be handled distinctly by resident cDCs and how signals driving poor tumoral cDC activation further impact the lymph node. Our findings clarify how tissue context dictates antigenic and, consequently, T cell fate in the lymph node.
Collapse
Affiliation(s)
- C Pirillo
- CRUK Beatson Institute, Glasgow, Great Britain
| | | | - A Sims
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Great Britain
| | - R Devlin
- CRUK Beatson Institute, Glasgow, Great Britain
| | - H Zhao
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Great Britain
- Jinan Center for Disease Control and Prevention, Jinan, Shandong 250021, China
| | - R Pinto
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Great Britain
| | - S Jasim
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Great Britain
| | - PA Shearer
- School of Infection and Immunity, University of Glasgow, Glasgow, Great Britain
| | - AL Shergold
- CRUK Beatson Institute, Glasgow, Great Britain
| | - H Donnelly
- School of Cancer Sciences, University of Glasgow, Glasgow, Great Britain
| | | | - C Loney
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Great Britain
| | - G Perona-Wright
- School of Infection and Immunity, University of Glasgow, Glasgow, Great Britain
| | - E Hutchinson
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Great Britain
- School of Infection and Immunity, University of Glasgow, Glasgow, Great Britain
| | - EW Roberts
- CRUK Beatson Institute, Glasgow, Great Britain
- School of Cancer Sciences, University of Glasgow, Glasgow, Great Britain
| |
Collapse
|
35
|
Shoaib A, Javed S, Wahab S, Azmi L, Tabish M, Sultan MH, Abdelsalam K, Alqahtani SS, Ahmad MF. Cellular, Molecular, Pharmacological, and Nano-Formulation Aspects of Thymoquinone-A Potent Natural Antiviral Agent. Molecules 2023; 28:5435. [PMID: 37513307 PMCID: PMC10383476 DOI: 10.3390/molecules28145435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/09/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
The goal of an antiviral agent research is to find an antiviral drug that reduces viral growth without harming healthy cells. Transformations of the virus, new viral strain developments, the resistance of viral pathogens, and side effects are the current challenges in terms of discovering antiviral drugs. The time has come and it is now essential to discover a natural antiviral agent that has the potential to destroy viruses without causing resistance or other unintended side effects. The pharmacological potency of thymoquinone (TQ) against different communicable and non-communicable diseases has been proven by various studies, and TQ is considered to be a safe antiviral substitute. Adjunctive immunomodulatory effects in addition to the antiviral potency of TQ makes it a major compound against viral infection through modulating the production of nitric oxide and reactive oxygen species, decreasing the cytokine storm, and inhibiting endothelial dysfunction. Nevertheless, TQ's low oral bioavailability, short half-life, poor water solubility, and conventional formulation are barriers to achieving its optimal pharmacologic benefits. Nano-formulation proposes numerous ways to overcome these obstacles through a small particle size, a big surface area, and a variety of surface modifications. Nano-based pharmaceutical innovations to combat viral infections using TQ are a promising approach to treating surmounting viral infections.
Collapse
Affiliation(s)
- Ambreen Shoaib
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
- Pharmacy Practice Research Unit (PPRU), College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Shamama Javed
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Lubna Azmi
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, University of Lucknow, Lucknow 226007, India
| | - Mohammad Tabish
- Department of Pharmacology, College of Medicine, Shaqra University, Shaqra 11961, Saudi Arabia
| | - Muhammad H Sultan
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Karim Abdelsalam
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
- Pharmacy Practice Research Unit (PPRU), College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Saad S Alqahtani
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Md Faruque Ahmad
- Department of Clinical Nutrition, College of Applied Medical Sciences, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
36
|
Viscidi RP, Rowley T, Bossis I. Bioengineered Bovine Papillomavirus L1 Protein Virus-like Particle (VLP) Vaccines for Enhanced Induction of CD8 T Cell Responses through Cross-Priming. Int J Mol Sci 2023; 24:9851. [PMID: 37372999 DOI: 10.3390/ijms24129851] [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: 04/27/2023] [Revised: 06/01/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Safe and effective T cell vaccines are needed for the treatment or prevention of cancers as well as infectious agents where vaccines for neutralizing antibodies have performed poorly. Recent research highlights an important role for tissue-resident memory T cells (TRM cells) in protective immunity and the role of a subset of dendritic cells that are capable of cross-priming for the induction of TRM cells. However, efficient vaccine technologies that operate through cross-priming and induce robust CD8+ T cell responses are lacking. We developed a platform technology by genetically engineering the bovine papillomavirus L1 major capsid protein to insert a polyglutamic acid/cysteine motif in place of wild-type amino acids in the HI loop. Virus-like particles (VLPs) are formed by self-assembly in insect cells infected with a recombinant baculovirus. Polyarginine/cysteine-tagged antigens are linked to the VLP by a reversible disulfide bond. The VLP possesses self-adjuvanting properties due to the immunostimulatory activity of papillomavirus VLPs. Polyionic VLP vaccines induce robust CD8+ T cell responses in peripheral blood and tumor tissues. A prostate cancer polyionic VLP vaccine was more efficacious than other vaccines and immunotherapies for the treatment of prostate cancer in a physiologically relevant murine model and successfully treated more advanced diseases than the less efficacious technologies. The immunogenicity of polyionic VLP vaccines is dependent on particle size, reversible linkage of the antigen to the VLP, and an interferon type 1 and Toll-like receptor (TLR)3/7-dependent mechanism.
Collapse
Affiliation(s)
- Raphael P Viscidi
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Treva Rowley
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Ioannis Bossis
- Department of Animal Production, School of Agricultural Sciences, Forestry & Natural Resources, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
37
|
Seya T, Shingai M, Kawakita T, Matsumoto M. Two Modes of Th1 Polarization Induced by Dendritic-Cell-Priming Adjuvant in Vaccination. Cells 2023; 12:1504. [PMID: 37296625 PMCID: PMC10252737 DOI: 10.3390/cells12111504] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/18/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Viral infections are usually accompanied by systemic cytokinemia. Vaccines need not necessarily mimic infection by inducing cytokinemia, but must induce antiviral-acquired immunity. Virus-derived nucleic acids are potential immune-enhancers and particularly good candidates as adjuvants in vaccines in mouse models. The most important nucleic-acid-sensing process involves the dendritic cell (DC) Toll-like receptor (TLR), which participates in the pattern recognition of foreign DNA/RNA structures. Human CD141+ DCs preferentially express TLR3 in endosomes and recognize double-stranded RNA. Antigen cross-presentation occurs preferentially in this subset of DCs (cDCs) via the TLR3-TICAM-1-IRF3 axis. Another subset, plasmacytoid DCs (pDCs), specifically expresses TLR7/9 in endosomes. They then recruit the MyD88 adaptor, and potently induce type I interferon (IFN-I) and proinflammatory cytokines to eliminate the virus. Notably, this inflammation leads to the secondary activation of antigen-presenting cDCs. Hence, the activation of cDCs via nucleic acids involves two modes: (i) with bystander effect of inflammation and (ii) without inflammation. In either case, the acquired immune response finally occurs with Th1 polarity. The level of inflammation and adverse events depend on the TLR repertoire and the mode of response to their agonists in the relevant DC subsets, and could be predicted by assessing the levels of cytokines/chemokines and T cell proliferation in vaccinated subjects. The main differences in the mode of vaccine sought in infectious diseases and cancer are defined by whether it is prophylactic or therapeutic, whether it can deliver sufficient antigens to cDCs, and how it behaves in the microenvironment of the lesion. Adjuvant can be selected on a case-to-case basis.
Collapse
Affiliation(s)
- Tsukasa Seya
- Nebuta Research Institute for Life Sciences, Aomori University, Aomori 030-0943, Japan;
- Department of Vaccine Immunology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
- Division of Vaccine Immunology, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan; (M.S.); (T.K.)
| | - Masashi Shingai
- Division of Vaccine Immunology, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan; (M.S.); (T.K.)
- Division of Biologics Development, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan
- International Collaboration Unit, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo 001-0021, Japan
| | - Tomomi Kawakita
- Division of Vaccine Immunology, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan; (M.S.); (T.K.)
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo 001-0021, Japan
| | - Misako Matsumoto
- Nebuta Research Institute for Life Sciences, Aomori University, Aomori 030-0943, Japan;
- Department of Vaccine Immunology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
- Division of Vaccine Immunology, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan; (M.S.); (T.K.)
| |
Collapse
|
38
|
Cheng H, Chen W, Lin Y, Zhang J, Song X, Zhang D. Signaling pathways involved in the biological functions of dendritic cells and their implications for disease treatment. MOLECULAR BIOMEDICINE 2023; 4:15. [PMID: 37183207 PMCID: PMC10183318 DOI: 10.1186/s43556-023-00125-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 04/02/2023] [Indexed: 05/16/2023] Open
Abstract
The ability of dendritic cells (DCs) to initiate and regulate adaptive immune responses is fundamental for maintaining immune homeostasis upon exposure to self or foreign antigens. The immune regulatory function of DCs is strictly controlled by their distribution as well as by cytokines, chemokines, and transcriptional programming. These factors work in conjunction to determine whether DCs exert an immunosuppressive or immune-activating function. Therefore, understanding the molecular signals involved in DC-dependent immunoregulation is crucial in providing insight into the generation of organismal immunity and revealing potential clinical applications of DCs. Considering the many breakthroughs in DC research in recent years, in this review we focused on three basic lines of research directly related to the biological functions of DCs and summarized new immunotherapeutic strategies involving DCs. First, we reviewed recent findings on DC subsets and identified lineage-restricted transcription factors that guide the development of different DC subsets. Second, we discussed the recognition and processing of antigens by DCs through pattern recognition receptors, endogenous/exogenous pathways, and the presentation of antigens through peptide/major histocompatibility complexes. Third, we reviewed how interactions between DCs and T cells coordinate immune homeostasis in vivo via multiple pathways. Finally, we summarized the application of DC-based immunotherapy for autoimmune diseases and tumors and highlighted potential research prospects for immunotherapy that targets DCs. This review provides a useful resource to better understand the immunomodulatory signals involved in different subsets of DCs and the manipulation of these immune signals can facilitate DC-based immunotherapy.
Collapse
Affiliation(s)
- Hao Cheng
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wenjing Chen
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yubin Lin
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jianan Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiaoshuang Song
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Dunfang Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
39
|
Preet Kaur A, Alice A, Crittenden MR, Gough MJ. The role of dendritic cells in radiation-induced immune responses. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 378:61-104. [PMID: 37438021 DOI: 10.1016/bs.ircmb.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Dendritic cells perform critical functions in bridging innate and adaptive immunity. Their ability to sense adjuvant signals in their environment, migrate on maturation, and cross-present cell-associated antigens enables these cells to carry antigen from tissue sites to lymph nodes, and thereby prime naïve T cells that cannot enter tissues. Despite being an infrequent cell type in tumors, we discuss how dendritic cells impact the immune environment of tumors and their response to cancer therapies. We review how radiation therapy of tumors can impact dendritic cells, through transfer of cell associated antigens to dendritic cells and the release of endogenous adjuvants, resulting in increased antigen presentation in the tumor-draining lymph nodes. We explore how tumor specific factors can result in negative regulation of dendritic cell function in the tumor, and the impact of direct radiation exposure to dendritic cells in the treatment field. These data suggest an important role for dendritic cell subpopulations in activating new T cell responses and boosting existing T cell responses to tumor associated antigens in tumor draining lymph nodes following radiation therapy. It further justifies a focus on the needs of the lymph node T cells to improve systemic anti-immunity following radiation therapy.
Collapse
Affiliation(s)
- Aanchal Preet Kaur
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, United States
| | - Alejandro Alice
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, United States
| | - Marka R Crittenden
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, United States; The Oregon Clinic, Portland, OR, United States
| | - Michael J Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, United States.
| |
Collapse
|
40
|
Stotts C, Corrales-Medina VF, Rayner KJ. Pneumonia-Induced Inflammation, Resolution and Cardiovascular Disease: Causes, Consequences and Clinical Opportunities. Circ Res 2023; 132:751-774. [PMID: 36927184 DOI: 10.1161/circresaha.122.321636] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Pneumonia is inflammation in the lungs, which is usually caused by an infection. The symptoms of pneumonia can vary from mild to life-threatening, where severe illness is often observed in vulnerable populations like children, older adults, and those with preexisting health conditions. Vaccines have greatly reduced the burden of some of the most common causes of pneumonia, and the use of antimicrobials has greatly improved the survival to this infection. However, pneumonia survivors do not return to their preinfection health trajectories but instead experience an accelerated health decline with an increased risk of cardiovascular disease. The mechanisms of this association are not well understood, but a persistent dysregulated inflammatory response post-pneumonia appears to play a central role. It is proposed that the inflammatory response during pneumonia is left unregulated and exacerbates atherosclerotic vascular disease, which ultimately leads to adverse cardiac events such as myocardial infarction. For this reason, there is a need to better understand the inflammatory cross talk between the lungs and the heart during and after pneumonia to develop therapeutics that focus on preventing pneumonia-associated cardiovascular events. This review will provide an overview of the known mechanisms of inflammation triggered during pneumonia and their relevance to the increased cardiovascular risk that follows this infection. We will also discuss opportunities for new clinical approaches leveraging strategies to promote inflammatory resolution pathways as a novel therapeutic target to reduce the risk of cardiac events post-pneumonia.
Collapse
Affiliation(s)
- Cameron Stotts
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada (C.S., K.J.R).,Centre for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada (C.S., V.F.C.-M.).,University of Ottawa Heart Institute, Ottawa, ON, Canada (C.S., K.J.R)
| | - Vicente F Corrales-Medina
- Centre for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada (C.S., V.F.C.-M.).,Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada (V.F.C-M).,Ottawa Hospital Research Institute, Ottawa, ON, Canada (V.F.C.-M)
| | - Katey J Rayner
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada (C.S., K.J.R).,University of Ottawa Heart Institute, Ottawa, ON, Canada (C.S., K.J.R)
| |
Collapse
|
41
|
Wahyuningtyas R, Wu ML, Chung WB, Chaung HC, Chang KT. Toll-like Receptor-Mediated Immunomodulation of Th1-Type Response Stimulated by Recombinant Antigen of Type 2 Porcine Reproductive and Respiratory Syndrome Virus (PRRSV-2). Viruses 2023; 15:v15030775. [PMID: 36992483 PMCID: PMC10057405 DOI: 10.3390/v15030775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/05/2023] [Accepted: 03/14/2023] [Indexed: 03/22/2023] Open
Abstract
PRRSV infects CD163-positive macrophages and skews their polarization toward an M2 phenotype, followed by T-cell inactivation. In our previous study, we found that recombinant protein A1 antigen derived from PRRSV-2 was a potential vaccine or adjuvant for immunization against PRRSV-2 infection due to its ability to repolarize macrophages into M1 subtype, thereby reducing CD163 expression for viral entry and promoting immunomodulation for Th1-type responses, except for stimulating Toll-like receptor (TLR) activation. The aim of our current study was to evaluate the effects of another two recombinant antigens, A3 (ORF6L5) and A4 (NLNsp10L11), for their ability to trigger innate immune responses including TLR activation. We isolated pulmonary alveolar macrophages (PAMs) from 8- to 12-week-old specific pathogen free (SPF) piglets and stimulated them with PRRSV (0.01 MOI and 0.05 MOI) or antigens. We also investigated the T-cell differentiation by immunological synapse activation of PAMs and CD4+ T-cells in the cocultured system. To confirm the infection of PRRSV in PAMs, we checked the expression of TLR3, 7, 8, and 9. Our results showed that the expression of TLR3, 7, and 9 were significantly upregulated in PAMs by A3 antigen induction, similar to the extent of PRRSV infection. Gene profile results showed that A3 repolarizes macrophages into the M1 subtype potently, in parallel with A1, as indicated by significant upregulation of proinflammatory genes (TNF-α, IL-6, IL-1β and IL-12). Upon immunological synapse activation, A3 potentially differentiated CD4 T cells into Th1 cells, determined by the expression of IL-12 and IFN-γ secretion. On the contrary, antigen A4 promoted regulatory T cell (T-reg) differentiation by significant upregulation of IL-10 expression. Finally, we concluded that the PRRSV-2 recombinant protein A3 provided better protection against PRRSV infection, suggested by its capability to reeducate immunosuppressive M2 macrophages into proinflammatory M1 cells. As M1 macrophages are prone to be functional antigen-presenting cells (APCs), they can call for TLR activation and Th1-type immune response within the immunological synapse.
Collapse
Affiliation(s)
- Rika Wahyuningtyas
- Research Centre for Animal Biologics, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
- Department of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
| | - Mei-Li Wu
- Research Centre for Animal Biologics, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
- Department of Food Science, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
| | - Wen-Bin Chung
- Research Centre for Animal Biologics, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
- Department of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
| | - Hso-Chi Chaung
- Research Centre for Animal Biologics, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
- Department of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
- Flow Cytometry Center, Precision Instruments Center, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
- Correspondence: (H.-C.C.); (K.-T.C.)
| | - Ko-Tung Chang
- Research Centre for Animal Biologics, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
- Flow Cytometry Center, Precision Instruments Center, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
- Correspondence: (H.-C.C.); (K.-T.C.)
| |
Collapse
|
42
|
Henry CM, Castellanos CA, Reis E Sousa C. DNGR-1-mediated cross-presentation of dead cell-associated antigens. Semin Immunol 2023; 66:101726. [PMID: 36758378 DOI: 10.1016/j.smim.2023.101726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 02/09/2023]
Abstract
Conventional dendritic cells type 1 (cDC1) are critical for inducing protective CD8+ T cell responses to tumour and viral antigens. In many instances, cDC1 access those antigens in the form of material internalised from dying tumour or virally-infected cells. How cDC1 extract dead cell-associated antigens and cross-present them in the form of peptides bound to MHC class I molecules to CD8+ T cells remains unclear. Here we review the biology of dendritic cell natural killer group receptor-1 (DNGR-1; also known as CLEC9A), a C-type lectin receptor highly expressed on cDC1 that plays a key role in this process. We highlight recent advances that support a function for DNGR-1 signalling in promoting inducible rupture of phagocytic or endocytic compartments containing dead cell debris, thereby making dead cell-associated antigens accessible to the endogenous MHC class I processing and presentation machinery of cDC1. We further review how DNGR-1 detects dead cells, as well as the functions of the receptor in anti-viral and anti-tumour immunity. Finally, we highlight how the study of DNGR-1 has opened new perspectives into cross-presentation, some of which may have applications in immunotherapy of cancer and vaccination against viral diseases.
Collapse
Affiliation(s)
- Conor M Henry
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Carlos A Castellanos
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Caetano Reis E Sousa
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
43
|
Inflammatory cell death: how macrophages sense neighbouring cell infection and damage. Biochem Soc Trans 2023; 51:303-313. [PMID: 36695550 PMCID: PMC9987993 DOI: 10.1042/bst20220807] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/26/2023]
Abstract
Programmed cell death is a critical host defence strategy during viral infection. Neighbouring cells deal with this death in distinct ways depending on how the infected cell dies. While apoptosis is considered immunologically silent, the lytic pathways of necroptosis and pyroptosis trigger inflammatory responses by releasing inflammatory host molecules. All these pathways have been implicated in influenza A virus infection. Here, we review how cells sense neighbouring infection and death and how sensing shapes ensuing inflammatory responses.
Collapse
|
44
|
Ge Y, Sun F, Zhao B, Kong F, Li Z, Kong X. Bacteria derived extracellular vesicles in the pathogenesis and treatment of gastrointestinal tumours. Front Oncol 2023; 12:1103446. [PMID: 36776356 PMCID: PMC9910087 DOI: 10.3389/fonc.2022.1103446] [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: 11/20/2022] [Accepted: 12/28/2022] [Indexed: 01/27/2023] Open
Abstract
Extracellular vesicles are fundamentally significant in the communication between cells. Outer Membrane Vesicles(OMVs) are a special kind of EVs produced by Gram-negative bacteria, which are minute exosome-like particles budding from the outer membrane, which have been found to play essential roles in diverse bacterial life events, including regulation of microbial interactions, pathogenesis promotion, stress responses and biofilm formation. Recently, and more researches have explored the substantial potentials of EVs as natural functional nanoparticles in the bioengineering applications in infectious diseases, cardiovascular diseases, autoimmune diseases and neurological diseases, such as antibacterial therapy, cancer drugs and immunoadjuvants, with several candidates in clinical trials showing promising efficacy. However, due to the poor understanding of sources, membrane structures and biogenesis mechanisms of EVs, progress in clinical applications still remains timid. In this review, we summarize the latest findings of EVs, especially in gastrointestinal tract tumours, to provide a comprehensive introduction of EVs in tumorigenesis and therapeutics.
Collapse
Affiliation(s)
- Yang Ge
- Changhai Clinical Research Unit, Changhai Hospital, Naval Military Medical University, Shanghai, China,Department of Gastroenterology, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Fengyuan Sun
- Changhai Clinical Research Unit, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Bo Zhao
- Changhai Clinical Research Unit, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Fanyang Kong
- Changhai Clinical Research Unit, Changhai Hospital, Naval Military Medical University, Shanghai, China,Department of Gastroenterology, Changhai Hospital, Naval Military Medical University, Shanghai, China,*Correspondence: Xiangyu Kong, ; Zhaoshen Li, ; Fanyang Kong,
| | - Zhaoshen Li
- Changhai Clinical Research Unit, Changhai Hospital, Naval Military Medical University, Shanghai, China,*Correspondence: Xiangyu Kong, ; Zhaoshen Li, ; Fanyang Kong,
| | - Xiangyu Kong
- Changhai Clinical Research Unit, Changhai Hospital, Naval Military Medical University, Shanghai, China,Department of Gastroenterology, Changhai Hospital, Naval Military Medical University, Shanghai, China,National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, China,*Correspondence: Xiangyu Kong, ; Zhaoshen Li, ; Fanyang Kong,
| |
Collapse
|
45
|
Sakref C, Bendriss-Vermare N, Valladeau-Guilemond J. Phenotypes and Functions of Human Dendritic Cell Subsets in the Tumor Microenvironment. Methods Mol Biol 2023; 2618:17-35. [PMID: 36905506 DOI: 10.1007/978-1-0716-2938-3_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Dendritic cells (DCs) play a key role in the antitumor immunity, as they are at the interface of innate and adaptive immunity. This important task can only be performed thanks to the broad range of mechanisms that DCs can perform to activate other immune cells. As DCs are well known for their outstanding capacity to prime and activate T cells through antigen presentation, DCs were intensively investigated during the past decades. Numerous studies have identified new DC subsets, leading to a large variety of subsets commonly separated into cDC1, cDC2, pDCs, mature DCs, Langerhans cells, monocyte-derived DCs, Axl-DCs, and several other subsets. Here, we review the specific phenotypes, functions, and localization within the tumor microenvironment (TME) of human DC subsets thanks to flow cytometry and immunofluorescence but also with the help of high-output technologies such as single-cell RNA sequencing and imaging mass cytometry (IMC).
Collapse
Affiliation(s)
- Candice Sakref
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- LabEx DEVweCAN, Lyon, France
| | - Nathalie Bendriss-Vermare
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- LabEx DEVweCAN, Lyon, France
- Laboratoire d'Immunothérapie des Cancers de Lyon (LICL), Lyon, France
| | - Jenny Valladeau-Guilemond
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France.
- LabEx DEVweCAN, Lyon, France.
| |
Collapse
|
46
|
Xiao Q, Xia Y. Insights into dendritic cell maturation during infection with application of advanced imaging techniques. Front Cell Infect Microbiol 2023; 13:1140765. [PMID: 36936763 PMCID: PMC10018208 DOI: 10.3389/fcimb.2023.1140765] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Dendritic cells (DCs) are crucial for the initiation and regulation of adaptive immune responses. When encountering immune stimulus such as bacterial and viral infection, parasite invasion and dead cell debris, DCs capture antigens, mature, acquire immunostimulatory activity and transmit the immune information to naïve T cells. Then activated cytotoxic CD8+ T cells directly kill the infected cells, while CD4+ T helper cells release cytokines to aid the activity of other immune cells, and help B cells produce antibodies. Thus, detailed insights into the DC maturation process are necessary for us to understand the working principle of immune system, and develop new medical treatments for infection, cancer and autoimmune disease. This review summarizes the DC maturation process, including environment sensing and antigen sampling by resting DCs, antigen processing and presentation on the cell surface, DC migration, DC-T cell interaction and T cell activation. Application of advanced imaging modalities allows visualization of subcellular and molecular processes in a super-high resolution. The spatiotemporal tracking of DCs position and migration reveals dynamics of DC behavior during infection, shedding novel lights on DC biology.
Collapse
Affiliation(s)
- Qi Xiao
- Genetic Engineering Research Center, School of Life Sciences, Chongqing University, Chongqing, China
- Chongqing Engineering Research Center for Fungal Insecticide, Chongqing, China
- Key Laboratory of Gene Function and Regulation Technologies Under Chongqing Municipal Education Commission, Chongqing, China
- *Correspondence: Qi Xiao,
| | - Yuxian Xia
- Genetic Engineering Research Center, School of Life Sciences, Chongqing University, Chongqing, China
- Chongqing Engineering Research Center for Fungal Insecticide, Chongqing, China
- Key Laboratory of Gene Function and Regulation Technologies Under Chongqing Municipal Education Commission, Chongqing, China
| |
Collapse
|
47
|
Duluc D, Sisirak V. Origin, Phenotype, and Function of Mouse Dendritic Cell Subsets. Methods Mol Biol 2023; 2618:3-16. [PMID: 36905505 DOI: 10.1007/978-1-0716-2938-3_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Dendritic cells are cells of hematopoietic origin that are specialized in antigen presentation and instruction of innate and adaptive immune responses. They are a heterogenous group of cells populating lymphoid organs and most tissues. Dendritic cells are commonly separated in three main subsets that differ in their developmental paths, phenotype, and functions. Most studies on dendritic cells were done primarily in mice; therefore, in this chapter, we propose to summarize the current knowledge and recent progress on mouse dendritic cell subsets' development, phenotype, and functions.
Collapse
Affiliation(s)
- Dorothée Duluc
- Université de Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, Bordeaux, France.
| | - Vanja Sisirak
- UMR CNRS 5164 - Immunoconcept, Université de Bordeaux, Bordeaux, France.
| |
Collapse
|
48
|
Teijeira A, Garasa S, Luri-Rey C, de Andrea C, Gato M, Molina C, Kaisho T, Cirella A, Azpilikueta A, Wculek SK, Egea J, Olivera I, Rodriguez I, Rouzaut A, Verkhusha V, Valencia K, Sancho D, Berraondo P, Melero I. Depletion of Conventional Type-1 Dendritic Cells in Established Tumors Suppresses Immunotherapy Efficacy. Cancer Res 2022; 82:4373-4385. [PMID: 36130020 DOI: 10.1158/0008-5472.can-22-1046] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/29/2022] [Accepted: 09/19/2022] [Indexed: 01/24/2023]
Abstract
The ability of conventional type-1 dendritic cells (cDC1) to cross-present tumor antigens to CD8+ T cells is critical for the induction of antitumor CTLs. Mice that are constitutively deficient in cDC1 cells have been reported to fail to respond to immunotherapy strategies based on checkpoint inhibitors. However, further work is needed to clarify the precise time during immunotherapy treatment that cDC1 cells are required for the beneficial effect of treatment. Here, we used a refined XCR1-DTR-Venus transgenic mouse model to acutely deplete cDC1 cells and trace their behavior using intravital microscopy. Diphtheria toxin-mediated cDC1 depletion prior to immunotherapy treatment with anti-PD-1 and/or anti-CD137 immunostimulatory mAbs completely ablated antitumor efficacy. The efficacy of adoptive T-cell therapy was also hampered by prior cDC1 depletion. After the onset of immunotherapy treatment, depletion of cDC1s only moderately reduced the therapeutic efficacy of anti-PD-1 and anti-CD137 mAbs. Intravital microscopy of liver-engrafted tumors revealed changes in the intratumoral behavior of cDC1 cells in mice receiving immunotherapy, and treatment with diphtheria toxin to deplete cDC1s impaired tumor T-cell infiltration and function. These results reveal that the functional integrity of the cDC1 compartment is required at the onset of various immunotherapies to successfully treat established tumors. SIGNIFICANCE These findings reveal the intratumoral behavior of cDC1 dendritic cells in transgenic mouse models and demonstrate that the efficacy of immunotherapy regimens is precluded by elimination of these cells.
Collapse
Affiliation(s)
- Alvaro Teijeira
- Immunology and Immunotherapy Department, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain.,Navarra Institute of Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Madrid, Spain
| | - Saray Garasa
- Immunology and Immunotherapy Department, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain.,Navarra Institute of Health Research (IDISNA), Pamplona, Spain
| | - Carlos Luri-Rey
- Immunology and Immunotherapy Department, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Carlos de Andrea
- Navarra Institute of Health Research (IDISNA), Pamplona, Spain.,Pathology Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Maria Gato
- Immunology and Immunotherapy Department, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Carmen Molina
- Immunology and Immunotherapy Department, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Tsuneyasu Kaisho
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Assunta Cirella
- Immunology and Immunotherapy Department, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Arantza Azpilikueta
- Immunology and Immunotherapy Department, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain.,Navarra Institute of Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Madrid, Spain
| | - Steffanie K Wculek
- Immunobiology Lab, Centro Nacional de Investigación Cardiovasculares (CNIC), Madrid, Spain
| | - Josune Egea
- Immunology and Immunotherapy Department, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Irene Olivera
- Immunology and Immunotherapy Department, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Inmaculada Rodriguez
- Immunology and Immunotherapy Department, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain.,Navarra Institute of Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Madrid, Spain
| | - Ana Rouzaut
- Immunology and Immunotherapy Department, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain.,Navarra Institute of Health Research (IDISNA), Pamplona, Spain
| | - Vladislav Verkhusha
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Karmele Valencia
- Navarra Institute of Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Madrid, Spain.,Oncology Department, CIMA, Universidad de Navarra, Pamplona, Spain
| | - David Sancho
- Immunobiology Lab, Centro Nacional de Investigación Cardiovasculares (CNIC), Madrid, Spain
| | - Pedro Berraondo
- Immunology and Immunotherapy Department, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain.,Navarra Institute of Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Madrid, Spain
| | - Ignacio Melero
- Immunology and Immunotherapy Department, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain.,Navarra Institute of Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Madrid, Spain.,Deparments of Immunology and Oncology, Clinica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
49
|
Co-infection of porcine deltacoronavirus and porcine epidemic diarrhea virus induces early TRAF6-mediated NF-κB and IRF7 signaling pathways through TLRs. Sci Rep 2022; 12:19443. [PMID: 36376395 PMCID: PMC9660140 DOI: 10.1038/s41598-022-24190-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Porcine deltacoronavirus (PDCoV) and porcine epidemic diarrhea virus (PEDV) infect the small intestine and cause swine enteric coronavirus disease. The mucosal innate immune system is the first line of defense against viral infection. The modulatory effect of PDCoV and PEDV coinfection on antiviral signaling cascades of the intestinal mucosa has not been reported. Here, we investigate the gene expression levels of pattern recognition receptors, downstream inflammatory signaling pathway molecules, and associated cytokines on the intestinal mucosa of neonatal piglets either infected with a single- or co-infected with PDCoV and PEDV using real-time PCR. The results demonstrate that single-PEDV regulates the noncanonical NF-κB signaling pathway through RIG-I regulation. In contrast, single-PDCoV and PDCoV/PEDV coinfection regulate proinflammatory and regulatory cytokines through TRAF6-mediated canonical NF-κB and IRF7 signaling pathways through TLRs. Although PDCoV/PEDV coinfection demonstrated an earlier modulatory effect in these signaling pathways, the regulation of proinflammatory and regulatory cytokines was observed simultaneously during single viral infection. These results suggested that PDCoV/PEDV coinfection may have synergistic effects that lead to enhanced viral evasion of the mucosal innate immune response.
Collapse
|
50
|
Lim CS, Jang YH, Lee GY, Han GM, Jeong HJ, Kim JW, Lee JO. TLR3 forms a highly organized cluster when bound to a poly(I:C) RNA ligand. Nat Commun 2022; 13:6876. [PMID: 36371424 PMCID: PMC9653405 DOI: 10.1038/s41467-022-34602-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 10/25/2022] [Indexed: 11/15/2022] Open
Abstract
Toll-like Receptor 3 (TLR3) initiates a potent anti-viral immune response by binding to double-stranded RNA ligands. Previous crystallographic studies showed that TLR3 forms a homodimer when bound to a 46-base pair RNA ligand. However, this short RNA fails to initiate a robust immune response. To obtain structural insights into the length dependency of TLR3 ligands, we determine the cryo-electron microscopy structure of full-length TLR3 in a complex with a synthetic RNA ligand with an average length of ~400 base pairs. In the structure, the dimeric TLR3 units are clustered along the double-stranded RNA helix in a highly organized and cooperative fashion with a uniform inter-dimer spacing of 103 angstroms. The intracellular and transmembrane domains are dispensable for the clustering because their deletion does not interfere with the cluster formation. Our structural observation suggests that ligand-induced clustering of TLR3 dimers triggers the ordered assembly of intracellular signaling adaptors and initiates a robust innate immune response.
Collapse
Affiliation(s)
- Chan Seok Lim
- Department of Life Sciences and POSTECH, Pohang, 37673 Korea
| | - Yoon Ha Jang
- Department of Life Sciences and POSTECH, Pohang, 37673 Korea
| | - Ga Young Lee
- Department of Life Sciences and POSTECH, Pohang, 37673 Korea
| | - Gu Min Han
- Department of Life Sciences and POSTECH, Pohang, 37673 Korea
| | - Hye Jin Jeong
- grid.49100.3c0000 0001 0742 4007Institute of Membrane Proteins, POSTECH, Pohang, 37673 Korea
| | - Ji Won Kim
- Department of Life Sciences and POSTECH, Pohang, 37673 Korea ,grid.49100.3c0000 0001 0742 4007Institute of Membrane Proteins, POSTECH, Pohang, 37673 Korea
| | - Jie-Oh Lee
- Department of Life Sciences and POSTECH, Pohang, 37673 Korea ,grid.49100.3c0000 0001 0742 4007Institute of Membrane Proteins, POSTECH, Pohang, 37673 Korea
| |
Collapse
|