1
|
Moon CY, Belabed M, Park MD, Mattiuz R, Puleston D, Merad M. Dendritic cell maturation in cancer. Nat Rev Cancer 2025; 25:225-248. [PMID: 39920276 PMCID: PMC11954679 DOI: 10.1038/s41568-024-00787-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 02/09/2025]
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that are present at low abundance in the circulation and tissues; they serve as crucial immune sentinels by continually sampling their environment, migrating to secondary lymphoid organs and shaping adaptive immune responses through antigen presentation. Owing to their ability to orchestrate tolerogenic or immunogenic responses to a specific antigen, DCs have a pivotal role in antitumour immunity and the response to immune checkpoint blockade and other immunotherapeutic approaches. The multifaceted functions of DCs are acquired through a complex, multistage process called maturation. Although the role of inflammatory triggers in driving DC maturation was established decades ago, less is known about DC maturation in non-inflammatory contexts, such as during homeostasis and in cancer. The advent of single-cell technologies has enabled an unbiased, high-dimensional characterization of various DC states, including mature DCs. This approach has clarified the molecular programmes associated with DC maturation and also revealed how cancers exploit these pathways to subvert immune surveillance. In this Review, we discuss the mechanisms by which cancer disrupts DC maturation and highlight emerging therapeutic opportunities to modulate DC states. These insights could inform the development of DC-centric immunotherapies, expanding the arsenal of strategies to enhance antitumour immunity.
Collapse
Affiliation(s)
- Chang Yoon Moon
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meriem Belabed
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D Park
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raphaël Mattiuz
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Puleston
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
2
|
Ma Y, Shao J, Liu W, Gao S, Zhou G, Qi X, Chang H. Molecular Mechanism of VSV-Vectored ASFV Vaccine Activating Immune Response in DCs. Vet Sci 2025; 12:36. [PMID: 39852910 PMCID: PMC11769090 DOI: 10.3390/vetsci12010036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
The vesicular stomatitis virus (VSV)-vectored African swine fever virus (ASFV) vaccine can induce efficient immune response, but the potential mechanism remains unsolved. In order to investigate the efficacy of recombinant viruses (VSV-p35, VSV-p72)-mediated dendritic cells (DCs) maturation and the mechanism of inducing T-cell immune response, the functional effects of recombinant viruses on DC activation and target antigens presentation were explored in this study. The results showed that surface-marked molecules (CD80, CD86, CD40, and MHC-II) and secreted cytokines (IL-4, TNF-α, IFN-γ) were highly expressed in the recombinant virus-infected DCs. In addition, the co-culture results of recombinant virus-treated DCs with naive T cells showed that the Th1- and Th17-type responses were effectively activated. Taken together, the study indicated that the VSV-vectored ASFV vaccine activated the maturation of DCs and the Th1- and Th17-type immune response, which provided a theoretical basis for the development of novel ASF vaccines.
Collapse
Affiliation(s)
- Yunyun Ma
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730030, China; (Y.M.); (J.S.); (W.L.); (S.G.); (G.Z.)
| | - Junjun Shao
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730030, China; (Y.M.); (J.S.); (W.L.); (S.G.); (G.Z.)
| | - Wei Liu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730030, China; (Y.M.); (J.S.); (W.L.); (S.G.); (G.Z.)
| | - Shandian Gao
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730030, China; (Y.M.); (J.S.); (W.L.); (S.G.); (G.Z.)
| | - Guangqing Zhou
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730030, China; (Y.M.); (J.S.); (W.L.); (S.G.); (G.Z.)
| | - Xuefeng Qi
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Huiyun Chang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730030, China; (Y.M.); (J.S.); (W.L.); (S.G.); (G.Z.)
| |
Collapse
|
3
|
Zhao J, Zhang H, Zhao Y, Lin Z, Lin F, Wang Z, Mo Q, Lu G, Zhao G, Wang G. Exploratory Research for HIF-1α Overexpression Tumor Antigen in the Activation of Dendritic Cells and the Potent Anti-Tumor Immune Response. Cancer Manag Res 2024; 16:1813-1822. [PMID: 39713567 PMCID: PMC11662640 DOI: 10.2147/cmar.s482363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 11/24/2024] [Indexed: 12/24/2024] Open
Abstract
Background Tumor-specific antigens play an important role in dendritic cell (DC)-based immunotherapy. The acquisition of tumor-specific antigens, which are essential for DC-based immunotherapy, poses a significant challenge. This study aimed to explore the efficacy of hypoxia inducible factor-1α (HIF-1α) overexpression tumor antigens in DC-based immunotherapy. Methods An HIF-1α over-expression cell line was constructed to prepare HIF-1α overexpression tumor antigens. The expression of CD14, CD40, CD80, CD86, and HLA-DR on the surface of dendritic cells derived from monocytes was assessed using flow cytometry after stimulation with tumor antigens enriched in HIF-1α. T cell proliferation was analyzed by CFSE division following incubation with mature DCs. The apoptotic tumor cells were detected through annexin V/PI staining following coculture with dendritic cells (DCs) stimulated by HIF-1α enriched antigens. The detection of damage-associated molecular pattern molecules (DAMPs) HMGB1 and calreticulin (CALR) was performed using Western blotting. Results The results demonstrated that HIF-1α-enriched tumor antigens significantly upregulated the expression of CD40, CD80, CD86, and HLA-DR in DCs compared to normal tumor antigens. Furthermore, co-incubation with HIF-1α-enriched tumor antigen-activated DCs enhanced T cell proliferation and stimulated the T cell-mediated cytotoxicity. Notably, the expression of DAMPs, such as HMGB1 and CALR, was elevated in HIF-1α-enriched tumor antigens. Conclusion Our findings demonstrate that tumor antigens enriched with HIF-1α may encompass tumor-specific antigens capable of stimulating DC activation, thereby enhancing T cell proliferation and cytotoxicity. These results provide support for the further advancement of HIF-1α enriched tumor antigens in preclinical and clinical investigations pertaining to tumor treatment.
Collapse
Affiliation(s)
- Jinjin Zhao
- Clinical Laboratory, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
- Key Laboratory of Nano-Drug Delivery System Construction and Application in Xinxiang City, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Haiguang Zhang
- Department of Gynecology, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Yilin Zhao
- Department of Cardiology, the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Zhiqiang Lin
- Clinical Laboratory, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Fei Lin
- Department of Cardiology, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
- Cardiovascular Repair Engineering Technology Research Center, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Zhiyin Wang
- Clinical Laboratory, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Qingjiang Mo
- Clinical Laboratory, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Guangjian Lu
- Clinical Laboratory, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Guoan Zhao
- Department of Cardiology, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
- Cardiovascular Repair Engineering Technology Research Center, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Guoqiang Wang
- Clinical Laboratory, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| |
Collapse
|
4
|
Busselaar J, Sijbranda M, Borst J. The importance of type I interferon in orchestrating the cytotoxic T-cell response to cancer. Immunol Lett 2024; 270:106938. [PMID: 39490629 DOI: 10.1016/j.imlet.2024.106938] [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: 08/30/2024] [Revised: 10/22/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
Both type I interferon (IFN-I) and CD4+ T-cell help are required to generate effective CD8+ T-cell responses to cancer. We here outline based on existing literature how IFN-I signaling and CD4+ T-cell help are connected. Both impact on the functional state of dendritic cells (DCs), particularly conventional (c)DC1. The cDC1s are critical for crosspresentation of cell-associated antigens and for delivery of CD4+ T-cell help for cytotoxic T-lymphocyte (CTL) effector and memory differentiation. In infection, production of IFN-I is prompted by pathogen-associated molecular patterns (PAMPs), while in cancer it relies on danger-associated molecular patterns (DAMPs). IFN-I production by tumor cells and pDCs in the tumor micro-environment (TME) is often limited. IFN-I signals increase the ability of migratory cDC1s and cDC2s to transport tumor antigens to tumor-draining lymph nodes (tdLNs). IFN-I also enables cDC1s to form and sustain the platform for help delivery by stimulating the production of chemokines that attract CD4+ and CD8+ T cells. IFN-I promotes delivery of help in concert with CD40 signals by additive and synergistic impact on cross-presentation and provision of critical costimulatory and cytokine signals for CTL effector and memory differentiation. The scenario of CD4+ T-cell help therefore depends on IFN-I signaling. This scenario can play out in tdLNs as well as in the TME, thereby contributing to the cancer immunity cycle. The collective observations may explain why both IFN-I and CD4+ T-cell help signatures in the TME correlate with good prognosis and response to PD-1 targeting immunotherapy in human cancer. They also may explain why a variety of tumor types in which IFN-I signaling is attenuated, remain devoid of functional CTLs.
Collapse
Affiliation(s)
- Julia Busselaar
- Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Merel Sijbranda
- Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Jannie Borst
- Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
| |
Collapse
|
5
|
Allard CC, Salti S, Mourad W, Hassan GS. Implications of CD154 and Its Receptors in the Pathogenesis and Treatment of Systemic Lupus Erythematosus. Cells 2024; 13:1621. [PMID: 39404385 PMCID: PMC11482534 DOI: 10.3390/cells13191621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024] Open
Abstract
CD154, also known as CD40 ligand, is a costimulatory molecule involved in humoral and adaptive immune responses upon pairing with its classical receptor, CD40. The CD154/CD40 dyad is a key participant in the pathogenesis of many autoimmune diseases, including systemic lupus erythematosus (SLE). In SLE, the major cells at play, T and B lymphocytes, are shown to overexpress CD154 and CD40, respectively. Subsequently, these cells and other CD40-positive cells engage in numerous effector functions contributing to SLE development. With the recent identification of additional receptors for CD154, all belonging to the integrin family, the role of CD154 in SLE is more complex and calls for deeper investigation into its biological significance. Many therapeutic strategies directed against the CD154/CD40 couple have been deployed for the treatment of SLE and proved efficient in animal models and human studies. However, the incidence of thromboembolic complications in patients treated with these anti-CD154/CD40 antibodies halted their further clinical assessments and called for another class of therapies targeting these molecules. Second-generation antibodies directed against CD154 or CD40 are showing promising results in the advanced stages of clinical testing. Our review presents a thorough description of CD154 and its receptors, CD40 and the integrin family members in SLE pathogenesis. All these elements of the CD154 system represent important therapeutic targets for the treatment of SLE.
Collapse
Affiliation(s)
| | | | - Walid Mourad
- Laboratoire d’Immunologie Cellulaire et Moléculaire, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Rue Saint-Denis, Tour Viger, Montréal, QC H2X 0A9, Canada; (C.C.A.); (S.S.)
| | - Ghada S. Hassan
- Laboratoire d’Immunologie Cellulaire et Moléculaire, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Rue Saint-Denis, Tour Viger, Montréal, QC H2X 0A9, Canada; (C.C.A.); (S.S.)
| |
Collapse
|
6
|
Kirkpatrick C, Lu YCW. Deciphering CD4 + T cell-mediated responses against cancer. Mol Carcinog 2024; 63:1209-1220. [PMID: 38725218 PMCID: PMC11166516 DOI: 10.1002/mc.23730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/05/2024] [Indexed: 05/15/2024]
Abstract
It's been long thought that CD8+ cytotoxic T cells play a major role in T cell-mediated antitumor responses, whereas CD4+ T cells merely provide some assistance to CD8+ T cells as the "helpers." In recent years, numerous studies support the notion that CD4+ T cells play an indispensable role in antitumor responses. Here, we summarize and discuss the current knowledge regarding the roles of CD4+ T cells in antitumor responses and immunotherapy, with a focus on the molecular and cellular mechanisms behind these observations. These new insights on CD4+ T cells may pave the way to further optimize cancer immunotherapy.
Collapse
Affiliation(s)
- Catherine Kirkpatrick
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Yong-Chen William Lu
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
7
|
Caudell DL, Dugan GO, Babitzki G, Schubert C, Braendli-Baiocco A, Wasserman K, Acona G, Stern M, Passioukov A, Cline JM, Charo J. Systemic immune response to a CD40 agonist antibody in nonhuman primates. J Leukoc Biol 2024; 115:1084-1093. [PMID: 38372596 PMCID: PMC11626834 DOI: 10.1093/jleuko/qiae031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 10/13/2023] [Accepted: 11/30/2023] [Indexed: 02/20/2024] Open
Abstract
The cell surface molecule CD40 is a member of the tumor necrosis factor receptor superfamily and is broadly expressed by immune cells including B cells, dendritic cells, and monocytes, as well as other normal cells and some malignant cells. CD40 is constitutively expressed on antigen-presenting cells, and ligation promotes functional maturation, leading to an increase in antigen presentation and cytokine production, and a subsequent increase in the activation of antigen-specific T cells. It is postulated that CD40 agonists can mediate both T cell-dependent and T cell-independent immune mechanisms of tumor regression in mice and patients. In addition, it is believed that CD40 activation also promotes apoptotic death of tumor cells and that the presence of the molecule on the surface of cancer cells is an important factor in the generation of tumor-specific T cell responses that contribute to tumor cell elimination. Notably, CD40 agonistic therapies were evaluated in patients with solid tumors and hematologic malignancies with reported success as a single agent. Preclinical studies have shown that subcutaneous administration of CD40 agonistic antibodies reduces systemic toxicity and elicits a stronger and localized pharmacodynamic response. Two independent studies in cynomolgus macaque (Macaca fascicularis) were performed to further evaluate potentially immunotoxicological effects associated with drug-induced adverse events seen in human subjects. Studies conducted in monkeys showed that when selicrelumab is administered at doses currently used in clinical trial patients, via subcutaneous injection, it is safe and effective at stimulating a systemic immune response.
Collapse
Affiliation(s)
- David L. Caudell
- Section on Comparative Medicine, Department of Pathology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, United States
| | - Gregory O. Dugan
- Section on Comparative Medicine, Department of Pathology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, United States
| | - Galina Babitzki
- Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Diagnostics GmbH, F. Hoffmann-La Roche AG, Staffelseestrasse 2-8, 81477 Munich, Germany
| | - Christine Schubert
- Pharmaceutical Research and Early Development, Pharmaceutical Science, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Annamaria Braendli-Baiocco
- Pharmaceutical Research and Early Development, Pharmaceutical Science, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Ken Wasserman
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University School of Medicine, 3900 Reservoir Rd NW #337, Washington, DC 20007, United States
| | - Gonzalo Acona
- Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Zurich, F. Hoffmann-La Roche AG, Wagistrasse 10, 8952 Schlieren, Zurich, Switzerland
| | - Martin Stern
- Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Alexandre Passioukov
- Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Zurich, F. Hoffmann-La Roche AG, Wagistrasse 10, 8952 Schlieren, Zurich, Switzerland
| | - J. Mark Cline
- Section on Comparative Medicine, Department of Pathology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, United States
| | - Jehad Charo
- Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Zurich, F. Hoffmann-La Roche AG, Wagistrasse 10, 8952 Schlieren, Zurich, Switzerland
| |
Collapse
|
8
|
Zhu Z, Peng Q, Duan X, Li J. Interleukin-12: Structure, Function, and Its Impact in Colorectal Cancer. J Interferon Cytokine Res 2024; 44:158-169. [PMID: 38498032 DOI: 10.1089/jir.2023.0190] [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: 03/19/2024] Open
Abstract
Interleukin 12 (IL-12) is a heterodimer consisting of 2 subunits, p35 and p40, with unique associations and interacting functions with its family members. IL-12 is one of the most important cytokines regulating the immune system response and is integral to adaptive immunity. IL-12 has shown marked therapeutic potential in a variety of tumor types. This review therefore summarizes the characteristics of IL-12 and its application in tumor treatment, focusing on its antitumor effects in colorectal cancer (CRC) and potential radiosensitization mechanisms. We aim to provide a current reference for IL-12 and other potential CRC treatment strategies.
Collapse
Affiliation(s)
- Ziwei Zhu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Qian Peng
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Xingmei Duan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine University of Electronic Science and Technology of China, Chengdu, People's Republic of. China
| | - Jie Li
- School of Medicine, Southwest Medical University of China, Luzhou, People's Republic of China
- Department of Radiotherapy, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| |
Collapse
|
9
|
Sobral MC, Mooney DJ. Materials-Based Approaches for Cancer Vaccination. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:179-187. [PMID: 38166245 DOI: 10.4049/jimmunol.2300482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/27/2023] [Indexed: 01/04/2024]
Abstract
Therapeutic cancer vaccines offer the promise of stimulating the immune system to specifically eradicate tumor cells and establish long-term memory to prevent tumor recurrence. However, despite showing benign safety profiles and the ability to generate Ag-specific cellular responses, cancer vaccines have been hampered by modest clinical efficacy. Lessons learned from these studies have led to the emergence of innovative materials-based strategies that aim to boost the clinical activity of cancer vaccines. In this Brief Review, we provide an overview of the key elements needed for an effective vaccine-induced antitumor response, categorize current approaches to therapeutic cancer vaccination, and explore recent advances in materials-based strategies to potentiate cancer vaccines.
Collapse
Affiliation(s)
- Miguel C Sobral
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA; and Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA; and Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| |
Collapse
|
10
|
Luri-Rey C, Gomis G, Glez-Vaz J, Manzanal A, Martinez Riaño A, Rodriguez Ruiz ME, Teijeira A, Melero I. Cytotoxicity as a form of immunogenic cell death leading to efficient tumor antigen cross-priming. Immunol Rev 2024; 321:143-151. [PMID: 37822051 DOI: 10.1111/imr.13281] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Antigen cross-priming of CD8+ T cells is a critical process necessary for the effective expansion and activation of CD8+ T cells endowed with the ability to recognize and destroy tumor cells. The cross-presentation of tumor antigens to cross-prime CD8+ T cells is mainly mediated, if not only, by a subset of professional antigen-presenting cells termed type-1 conventional dendritic cells (cDC1). The demise of malignant cells can be immunogenic if it occurs in the context of premortem stress. These ways of dying are termed immunogenic cell death (ICD) and are associated with biochemical features favoring cDC1 for the efficient cross-priming of tumor antigens. Immunosurveillance and the success of immunotherapies heavily rely on the ability of cytotoxic immune cells, primarily CD8+ T cells and NK cells, to detect and eliminate tumor cells through mechanisms collectively known as cytotoxicity. Recent studies have revealed the significance of NK- and CTL-mediated cytotoxicity as a prominent form of immunogenic cell death, resulting in mechanisms that promote and sustain antigen-specific immune responses. This review focuses on the mechanisms underlying the cross-presentation of antigens released during tumor cell killing by cytotoxic immune cells, with an emphasis on the role of cDC1 cells. Indeed, cDC1s are instrumental in the effectiveness of most immunotherapies, underscoring the significance of tumor antigen cross-priming in contexts of immunogenic cell death. The notion of the potent immunogenicity of cell death resulting from NK or cytotoxic T lymphocyte (CTL)-mediated cytotoxicity has far-reaching implications for cancer immunotherapy.
Collapse
Affiliation(s)
- Carlos Luri-Rey
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | - Gabriel Gomis
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | - Javier Glez-Vaz
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | - Almudena Manzanal
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | - Ana Martinez Riaño
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | | | - Alvaro Teijeira
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Department of Oncology, Clinica Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Department of Oncology, Clinica Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Pharmacy, University "G. D'Annunzio" Chieti-Pescara, Chieti, Italy
| |
Collapse
|
11
|
Matsuda M, Mochizuki S. Control of A/D type CpG-ODN aggregates to a suitable size for induction of strong immunostimulant activity. Biochem Biophys Rep 2023; 36:101573. [PMID: 37954170 PMCID: PMC10633530 DOI: 10.1016/j.bbrep.2023.101573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/14/2023] Open
Abstract
Among several types of CpG-ODNs, A/D-type CpG-ODNs have potent adjuvant activity to induce Th-1 immune responses, but exhibit a propensity to aggregate. For the clinical application of A/D-type CpG-ODNs, it is necessary to control such aggregation and obtain a comprehensive understanding of the relationship between their structure and the immune responses. This study revealed that a representative A/D-type CpG ODN, D35, adopted a single-stranded structure in water, while it assembled into aggregates in response to Na+ ions. From polyacrylamide gel electrophoresis and circular dichroism analyses, D35 adopted a homodimeric form (duplex) via palindromic sequences in low-Na+-concentration conditions (10-50 mM NaCl). After replacement of the solution with PBS, quadruplexes began to form in a manner coordinated by Na+, resulting in large aggregates. The duplexes and small aggregates prepared in 50 mM NaCl showed not only high cellular uptake but also high affinity to Toll-like receptor 9 (TLR9) proteins, leading to the production of a large amount of interferon-α for peripheral blood mononuclear cells. The much larger aggregates prepared in 100 mM NaCl were incorporated into cells at a high level, but showed a low ability to induce cytokine production. This suggests that the large aggregates have difficulty inducing TLR9 dimerization, resulting in loss of the stimulation of the cells. We thus succeeded in inducing adequate innate immunity in vitro by controlling and adjusting the formation of D35 aggregates. Therefore, the findings in this study for D35 ODNs could be a vital research foundation for in vivo applications.
Collapse
Affiliation(s)
- Miyu Matsuda
- Department of Chemistry and Biochemistry, The University of Kitakyushu, 1-1 Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka, 808-0135, Japan
| | - Shinichi Mochizuki
- Department of Chemistry and Biochemistry, The University of Kitakyushu, 1-1 Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka, 808-0135, Japan
| |
Collapse
|
12
|
Zaini A, Dalit L, Sheikh AA, Zhang Y, Thiele D, Runting J, Rodrigues G, Ng J, Bramhall M, Scheer S, Hailes L, Groom JR, Good-Jacobson KL, Zaph C. Heterogeneous Tfh cell populations that develop during enteric helminth infection predict the quality of type 2 protective response. Mucosal Immunol 2023; 16:642-657. [PMID: 37392971 DOI: 10.1016/j.mucimm.2023.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 06/19/2023] [Accepted: 06/24/2023] [Indexed: 07/03/2023]
Abstract
T follicular helper (Tfh) cells are an important component of germinal center (GC)-mediated humoral immunity. Yet, how a chronic type 1 versus protective type 2 helminth infection modulates Tfh-GC responses remains poorly understood. Here, we employ the helminth Trichuris muris model and demonstrate that Tfh cell phenotypes and GC are differentially regulated in acute versus chronic infection. The latter failed to induce Tfh-GC B cell responses, with Tfh cells expressing Τ-bet and interferon-γ. In contrast, interleukin-4-producing Tfh cells dominate responses to an acute, resolving infection. Heightened expression and increased chromatin accessibility of T helper (Th)1- and Th2 cell-associated genes are observed in chronic and acute induced Tfh cells, respectively. Blockade of the Th1 cell response by T-cell-intrinsic T-bet deletion promoted Tfh cell expansion during chronic infection, pointing to a correlation between a robust Tfh cell response and protective immunity to parasites. Finally, blockade of Tfh-GC interactions impaired type 2 immunity, revealing the critical protective role of GC-dependent Th2-like Tfh cell responses during acute infection. Collectively, these results provide new insights into the protective roles of Tfh-GC responses and identify distinct transcriptional and epigenetic features of Tfh cells that emerge during resolving or chronic T. muris infection.
Collapse
Affiliation(s)
- Aidil Zaini
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Biochemistry and Molecular Biology, Clayton, Australia; Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, Australia
| | - Lennard Dalit
- Division of Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Amania A Sheikh
- Division of Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Yan Zhang
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Biochemistry and Molecular Biology, Clayton, Australia
| | - Daniel Thiele
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Microbiology, Monash University, Clayton, Australia
| | - Jessica Runting
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Biochemistry and Molecular Biology, Clayton, Australia
| | - Grace Rodrigues
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Biochemistry and Molecular Biology, Clayton, Australia
| | - Judy Ng
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Biochemistry and Molecular Biology, Clayton, Australia
| | - Michael Bramhall
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Biochemistry and Molecular Biology, Clayton, Australia; Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Australia
| | - Sebastian Scheer
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Biochemistry and Molecular Biology, Clayton, Australia
| | - Lauren Hailes
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Biochemistry and Molecular Biology, Clayton, Australia
| | - Joanna R Groom
- Division of Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Kim L Good-Jacobson
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Biochemistry and Molecular Biology, Clayton, Australia.
| | - Colby Zaph
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Biochemistry and Molecular Biology, Clayton, Australia.
| |
Collapse
|
13
|
Lei X, Wang Y, Broens C, Borst J, Xiao Y. Immune checkpoints targeting dendritic cells for antibody-based modulation in cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 382:145-179. [PMID: 38225102 DOI: 10.1016/bs.ircmb.2023.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Dendritic cells (DC) are professional antigen-presenting cells which link innate to adaptive immunity. DC play a central role in regulating antitumor T-cell responses in both tumor-draining lymph nodes (TDLN) and the tumor microenvironment (TME). They modulate effector T-cell responses via immune checkpoint proteins (ICPs) that can be either stimulatory or inhibitory. Functions of DC are often impaired by the suppressive TME leading to tumor immune escape. Therefore, better understanding of the mechanisms of action of ICPs expressed by (tumor-infiltrating) DC will lead to potential new treatment strategies. Genetic manipulation and high-dimensional analyses have provided insight in the interactions between DC and T-cells in TDLN and the TME upon ICP targeting. In this review, we discuss (tumor-infiltrating) DC lineage cells and tumor tissue specific "mature" DC states and their gene signatures in relation to anti-tumor immunity. We also review a number of ICPs expressed by DC regarding their functions in phagocytosis, DC activation, or inhibition and outline position in, or promise for clinical trials in cancer immunotherapy. Collectively, we highlight the critical role of DC and their exact status in the TME for the induction and propagation of T-cell immunity to cancer.
Collapse
Affiliation(s)
- Xin Lei
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Yizhi Wang
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Chayenne Broens
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Jannie Borst
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Yanling Xiao
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
14
|
Dickerson LK, Carter JA, Kohli K, Pillarisetty VG. Emerging interleukin targets in the tumour microenvironment: implications for the treatment of gastrointestinal tumours. Gut 2023; 72:1592-1606. [PMID: 37258094 DOI: 10.1136/gutjnl-2023-329650] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/15/2023] [Indexed: 06/02/2023]
Abstract
The effectiveness of antitumour immunity is dependent on intricate cytokine networks. Interleukins (ILs) are important mediators of complex interactions within the tumour microenvironment, including regulation of tumour-infiltrating lymphocyte proliferation, differentiation, migration and activation. Our evolving and increasingly nuanced understanding of the cell type-specific and heterogeneous effects of IL signalling has presented unique opportunities to fine-tune elaborate IL networks and engineer new targeted immunotherapeutics. In this review, we provide a primer for clinicians on the challenges and potential of IL-based treatment. We specifically detail the roles of IL-2, IL-10, IL-12 and IL-15 in shaping the tumour-immune landscape of gastrointestinal malignancies, paying particular attention to promising preclinical findings, early-stage clinical research and innovative therapeutic approaches that may properly place ILs to the forefront of immunotherapy regimens.
Collapse
Affiliation(s)
| | - Jason A Carter
- Hepatopancreatobiliary Surgery, University of Washington, Seattle, Washington, USA
| | - Karan Kohli
- Hepatopancreatobiliary Surgery, University of Washington, Seattle, Washington, USA
- Flatiron Bio, Palo Alto, California, USA
| | - Venu G Pillarisetty
- Hepatopancreatobiliary Surgery, University of Washington, Seattle, Washington, USA
| |
Collapse
|
15
|
Sun L, Su Y, Jiao A, Wang X, Zhang B. T cells in health and disease. Signal Transduct Target Ther 2023; 8:235. [PMID: 37332039 PMCID: PMC10277291 DOI: 10.1038/s41392-023-01471-y] [Citation(s) in RCA: 266] [Impact Index Per Article: 133.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 06/20/2023] Open
Abstract
T cells are crucial for immune functions to maintain health and prevent disease. T cell development occurs in a stepwise process in the thymus and mainly generates CD4+ and CD8+ T cell subsets. Upon antigen stimulation, naïve T cells differentiate into CD4+ helper and CD8+ cytotoxic effector and memory cells, mediating direct killing, diverse immune regulatory function, and long-term protection. In response to acute and chronic infections and tumors, T cells adopt distinct differentiation trajectories and develop into a range of heterogeneous populations with various phenotype, differentiation potential, and functionality under precise and elaborate regulations of transcriptional and epigenetic programs. Abnormal T-cell immunity can initiate and promote the pathogenesis of autoimmune diseases. In this review, we summarize the current understanding of T cell development, CD4+ and CD8+ T cell classification, and differentiation in physiological settings. We further elaborate the heterogeneity, differentiation, functionality, and regulation network of CD4+ and CD8+ T cells in infectious disease, chronic infection and tumor, and autoimmune disease, highlighting the exhausted CD8+ T cell differentiation trajectory, CD4+ T cell helper function, T cell contributions to immunotherapy and autoimmune pathogenesis. We also discuss the development and function of γδ T cells in tissue surveillance, infection, and tumor immunity. Finally, we summarized current T-cell-based immunotherapies in both cancer and autoimmune diseases, with an emphasis on their clinical applications. A better understanding of T cell immunity provides insight into developing novel prophylactic and therapeutic strategies in human diseases.
Collapse
Affiliation(s)
- Lina Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Yanhong Su
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Anjun Jiao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Xin Wang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China.
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China.
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China.
| |
Collapse
|
16
|
Gressier E, Schulte-Schrepping J, Petrov L, Brumhard S, Stubbemann P, Hiller A, Obermayer B, Spitzer J, Kostevc T, Whitney PG, Bachem A, Odainic A, van de Sandt C, Nguyen THO, Ashhurst T, Wilson K, Oates CVL, Gearing LJ, Meischel T, Hochheiser K, Greyer M, Clarke M, Kreutzenbeck M, Gabriel SS, Kastenmüller W, Kurts C, Londrigan SL, Kallies A, Kedzierska K, Hertzog PJ, Latz E, Chen YCE, Radford KJ, Chopin M, Schroeder J, Kurth F, Gebhardt T, Sander LE, Sawitzki B, Schultze JL, Schmidt SV, Bedoui S. CD4 + T cell calibration of antigen-presenting cells optimizes antiviral CD8 + T cell immunity. Nat Immunol 2023; 24:979-990. [PMID: 37188942 DOI: 10.1038/s41590-023-01517-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 04/13/2023] [Indexed: 05/17/2023]
Abstract
Antiviral CD8+ T cell immunity depends on the integration of various contextual cues, but how antigen-presenting cells (APCs) consolidate these signals for decoding by T cells remains unclear. Here, we describe gradual interferon-α/interferon-β (IFNα/β)-induced transcriptional adaptations that endow APCs with the capacity to rapidly activate the transcriptional regulators p65, IRF1 and FOS after CD4+ T cell-mediated CD40 stimulation. While these responses operate through broadly used signaling components, they induce a unique set of co-stimulatory molecules and soluble mediators that cannot be elicited by IFNα/β or CD40 alone. These responses are critical for the acquisition of antiviral CD8+ T cell effector function, and their activity in APCs from individuals infected with severe acute respiratory syndrome coronavirus 2 correlates with milder disease. These observations uncover a sequential integration process whereby APCs rely on CD4+ T cells to select the innate circuits that guide antiviral CD8+ T cell responses.
Collapse
Affiliation(s)
- Elise Gressier
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia.
| | - Jonas Schulte-Schrepping
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Lev Petrov
- Translational Immunology, Berlin Institute of Health (BIH) & Charité University Medicine, Berlin, Germany
| | - Sophia Brumhard
- Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Paula Stubbemann
- Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Anna Hiller
- Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Benedikt Obermayer
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Core Unit Bioinformatics, Berlin, Germany
| | - Jasper Spitzer
- Institute of Innate Immunity, University of Bonn, Bonn, Germany
| | - Tomislav Kostevc
- Translational Immunology, Berlin Institute of Health (BIH) & Charité University Medicine, Berlin, Germany
| | - Paul G Whitney
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Annabell Bachem
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Alexandru Odainic
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
- Institute of Innate Immunity, University of Bonn, Bonn, Germany
| | - Carolien van de Sandt
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Thi H O Nguyen
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Thomas Ashhurst
- Sydney Cytometry Core Research Facility, Charles Perkins Centre, Centenary Institute and University of Sydney, Sydney, New South Wales, Australia
| | - Kayla Wilson
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Clare V L Oates
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Linden J Gearing
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Tina Meischel
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Katharina Hochheiser
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Marie Greyer
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Michele Clarke
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | | | - Sarah S Gabriel
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Wolfgang Kastenmüller
- Würzburg Institute of Systems Immunology, Max Planck Research Group, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Christian Kurts
- Institute of Experimental Immunology, University of Bonn, Bonn, Germany
| | - Sarah L Londrigan
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Axel Kallies
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Paul J Hertzog
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Eicke Latz
- Institute of Innate Immunity, University of Bonn, Bonn, Germany
| | - Yu-Chen E Chen
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Kristen J Radford
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Michael Chopin
- Department of Biochemistry, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Jan Schroeder
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Florian Kurth
- Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas Gebhardt
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Leif E Sander
- Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Birgit Sawitzki
- Translational Immunology, Berlin Institute of Health (BIH) & Charité University Medicine, Berlin, Germany
| | - Joachim L Schultze
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE and University of Bonn, Bonn, Germany
| | | | - Sammy Bedoui
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia.
- Institute of Experimental Immunology, University of Bonn, Bonn, Germany.
| |
Collapse
|
17
|
Bourque J, Hawiger D. Activation, Amplification, and Ablation as Dynamic Mechanisms of Dendritic Cell Maturation. BIOLOGY 2023; 12:biology12050716. [PMID: 37237529 DOI: 10.3390/biology12050716] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/07/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023]
Abstract
T cell responses to cognate antigens crucially depend on the specific functionality of dendritic cells (DCs) activated in a process referred to as maturation. Maturation was initially described as alterations of the functional status of DCs in direct response to multiple extrinsic innate signals derived from foreign organisms. More recent studies, conducted mainly in mice, revealed an intricate network of intrinsic signals dependent on cytokines and various immunomodulatory pathways facilitating communication between individual DCs and other cells for the orchestration of specific maturation outcomes. These signals selectively amplify the initial activation of DCs mediated by innate factors and dynamically shape DC functionalities by ablating DCs with specific functions. Here, we discuss the effects of the initial activation of DCs that crucially includes the production of cytokine intermediaries to collectively achieve amplification of the maturation process and further precise sculpting of the functional landscapes among DCs. By emphasizing the interconnectedness of the intracellular and intercellular mechanisms, we reveal activation, amplification, and ablation as the mechanistically integrated components of the DC maturation process.
Collapse
Affiliation(s)
- Jessica Bourque
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Daniel Hawiger
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| |
Collapse
|
18
|
Jin SM, Yoo YJ, Shin HS, Kim S, Lee SN, Lee CH, Kim H, Kim JE, Bae YS, Hong J, Noh YW, Lim YT. A nanoadjuvant that dynamically coordinates innate immune stimuli activation enhances cancer immunotherapy and reduces immune cell exhaustion. NATURE NANOTECHNOLOGY 2023; 18:390-402. [PMID: 36635335 DOI: 10.1038/s41565-022-01296-w] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/15/2022] [Indexed: 06/17/2023]
Abstract
Although conventional innate immune stimuli contribute to immune activation, they induce exhausted immune cells, resulting in suboptimal cancer immunotherapy. Here we suggest a kinetically activating nanoadjuvant (K-nanoadjuvant) that can dynamically integrate two waves of innate immune stimuli, resulting in effective antitumour immunity without immune cell exhaustion. The combinatorial code of K-nanoadjuvant is optimized in terms of the order, duration and time window between spatiotemporally activating Toll-like receptor 7/8 agonist and other Toll-like receptor agonists. K-nanoadjuvant induces effector/non-exhausted dendritic cells that programme the magnitude and persistence of interleukin-12 secretion, generate effector/non-exhausted CD8+ T cells, and activate natural killer cells. Treatment with K-nanoadjuvant as a monotherapy or in combination therapy with anti-PD-L1 or liposomes (doxorubicin) results in strong antitumour immunity in murine models, with minimal systemic toxicity, providing a strategy for synchronous and dynamic tailoring of innate immunity for enhanced cancer immunotherapy.
Collapse
Affiliation(s)
- Seung Mo Jin
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yeon Jeong Yoo
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hong Sik Shin
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sohyun Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sang Nam Lee
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Republic of Korea
| | - Chang Hoon Lee
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hyunji Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jung-Eun Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yong-Soo Bae
- Department of Biological Sciences, Science Research Center (SRC) for Immune Research on Non-lymphoid Organ (CIRNO), Department of Biological Science, Sungkyunkwan University, Suwon, Republic of Korea
| | - JungHyub Hong
- Department of Biological Sciences, Science Research Center (SRC) for Immune Research on Non-lymphoid Organ (CIRNO), Department of Biological Science, Sungkyunkwan University, Suwon, Republic of Korea
| | - Young-Woock Noh
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongju, Republic of Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
19
|
CD4 + T cells in cancer. NATURE CANCER 2023; 4:317-329. [PMID: 36894637 DOI: 10.1038/s43018-023-00521-2] [Citation(s) in RCA: 205] [Impact Index Per Article: 102.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 01/20/2023] [Indexed: 03/11/2023]
Abstract
Cancer immunology and immunotherapy are driving forces of research and development in oncology, mostly focusing on CD8+ T cells and the tumor microenvironment. Recent progress highlights the importance of CD4+ T cells, corresponding to the long-known fact that CD4+ T cells are central players and coordinators of innate and antigen-specific immune responses. Moreover, they have now been recognized as anti-tumor effector cells in their own right. Here we review the current status of CD4+ T cells in cancer, which hold great promise for improving knowledge and therapies in cancer.
Collapse
|
20
|
MHC-dressing on dendritic cells: Boosting anti-tumor immunity via unconventional tumor antigen presentation. Semin Immunol 2023; 66:101710. [PMID: 36640616 DOI: 10.1016/j.smim.2023.101710] [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: 08/26/2022] [Revised: 12/21/2022] [Accepted: 01/07/2023] [Indexed: 01/15/2023]
Abstract
Dendritic cells are crucial for anti-tumor immune responses due to their ability to activate cytotoxic effector CD8+ T cells. Canonically, in anti-tumor immunity, dendritic cells activate CD8+ T cells in a process termed cross-presentation. Recent studies have demonstrated that another type of antigen presentation, MHC-dressing, also serves to activate CD8+ T cells against tumor cell-derived antigens. Understanding MHC-dressing's specific contributions to anti-tumor immunity can open up novel therapeutic avenues. In this review, we summarize the early studies that identified MHC-dressing as a relevant antigen presentation pathway before diving into a deeper discussion of the biology of MHC-dressing, focusing in particular on which dendritic cell subsets are most capable of performing MHC-dressing and how MHC-dressing compares to other forms of antigen presentation. We conclude by discussing the implications MHC-dressing has for anti-tumor immunity.
Collapse
|
21
|
Kawabe T. Homeostasis and immunological function of self-driven memory-phenotype CD4 + T lymphocytes. Immunol Med 2023; 46:1-8. [PMID: 36218322 DOI: 10.1080/25785826.2022.2129370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
CD4+ T lymphocytes play an essential role in adaptive immune responses. In pathogen infection, naïve CD4+ T cells that strongly respond to foreign antigens robustly proliferate to differentiate into effector/memory cells, contributing to elimination of the pathogen concerned. In addition to this conventional T cell activation pathway, naïve T cells can also weakly respond to self antigens in the periphery to spontaneously acquire a memory phenotype through homeostatic proliferation in steady state. Such 'memory-phenotype' (MP) CD4+ T lymphocytes are distinguishable from foreign antigen-specific memory cells in terms of marker expression. Once generated, MP cells are maintained by rapid proliferation while differentiating into the T-bet+ 'MP1' subset, with the latter response promoted by IL-12 homeostatically produced by type 1 dendritic cells. Importantly, MP1 cells possess innate immune function; they can produce IFN-γ in response to IL-12 and IL-18 to contribute to host defense against pathogens. Similarly, the presence of RORγt+ 'MP17' and Gata3hi 'MP2' cells as well as their potential immune functions have been proposed. In this review, I will discuss our current understanding on the unique mechanisms of generation, maintenance, and differentiation of MP CD4+ T lymphocytes as well as their functional significance in various disease conditions.
Collapse
Affiliation(s)
- Takeshi Kawabe
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
22
|
de Mey W, Locy H, De Ridder K, De Schrijver P, Autaers D, Lakdimi A, Esprit A, Franceschini L, Thielemans K, Verdonck M, Breckpot K. An mRNA mix redirects dendritic cells towards an antiviral program, inducing anticancer cytotoxic stem cell and central memory CD8 + T cells. Front Immunol 2023; 14:1111523. [PMID: 36860873 PMCID: PMC9969480 DOI: 10.3389/fimmu.2023.1111523] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/26/2023] [Indexed: 02/16/2023] Open
Abstract
Dendritic cell (DC)-maturation stimuli determine the potency of these antigen-presenting cells and, therefore, the quality of the T-cell response. Here we describe that the maturation of DCs via TriMix mRNA, encoding CD40 ligand, a constitutively active variant of toll-like receptor 4 and the co-stimulatory molecule CD70, enables an antibacterial transcriptional program. Besides, we further show that the DCs are redirected into an antiviral transcriptional program when CD70 mRNA in TriMix is replaced with mRNA encoding interferon-gamma and a decoy interleukin-10 receptor alpha, forming a four-component mixture referred to as TetraMix mRNA. The resulting TetraMixDCs show a high potential to induce tumor antigen-specific T cells within bulk CD8+ T cells. Tumor-specific antigens (TSAs) are emerging and attractive targets for cancer immunotherapy. As T-cell receptors recognizing TSAs are predominantly present on naive CD8+ T cells (TN), we further addressed the activation of tumor antigen-specific T cells when CD8+ TN cells are stimulated by TriMixDCs or TetraMixDCs. In both conditions, the stimulation resulted in a shift from CD8+ TN cells into tumor antigen-specific stem cell-like memory, effector memory and central memory T cells with cytotoxic capacity. These findings suggest that TetraMix mRNA, and the antiviral maturation program it induces in DCs, triggers an antitumor immune reaction in cancer patients.
Collapse
|
23
|
Chen CH, Weng TH, Huang HH, Huang LY, Huang KY, Chen PR, Yeh KY, Huang CT, Chien YT, Chuang PY, Lin YL, Tsai NM, Liu SJ, Su YC, Weng SL, Liao KW. A flexible liposomal polymer complex as a platform of specific and regulable immune regulation for individual cancer immunotherapy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2023; 42:29. [PMID: 36691089 PMCID: PMC9869520 DOI: 10.1186/s13046-023-02601-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/16/2023] [Indexed: 01/24/2023]
Abstract
BACKGROUND The applicability and therapeutic efficacy of specific personalized immunotherapy for cancer patients is limited by the genetic diversity of the host or the tumor. Side-effects such as immune-related adverse events (IRAEs) derived from the administration of immunotherapy have also been observed. Therefore, regulatory immunotherapy is required for cancer patients and should be developed. METHODS The cationic lipo-PEG-PEI complex (LPPC) can stably and irreplaceably adsorb various proteins on its surface without covalent linkage, and the bound proteins maintain their original functions. In this study, LPPC was developed as an immunoregulatory platform for personalized immunotherapy for tumors to address the barriers related to the heterogenetic characteristics of MHC molecules or tumor associated antigens (TAAs) in the patient population. Here, the immune-suppressive and highly metastatic melanoma, B16F10 cells were used to examine the effects of this platform. Adsorption of anti-CD3 antibodies, HLA-A2/peptide, or dendritic cells' membrane proteins (MP) could flexibly provide pan-T-cell responses, specific Th1 responses, or specific Th1 and Th2 responses, depending on the host needs. Furthermore, with regulatory antibodies, the immuno-LPPC complex properly mediated immune responses by adsorbing positive or negative antibodies, such as anti-CD28 or anti-CTLA4 antibodies. RESULTS The results clearly showed that treatment with LPPC/MP/CD28 complexes activated specific Th1 and Th2 responses, including cytokine release, CTL and prevented T-cell apoptosis. Moreover, LPPC/MP/CD28 complexes could eliminate metastatic B16F10 melanoma cells in the lung more efficiently than LPPC/MP. Interestingly, the melanoma resistance of mice treated with LPPC/MP/CD28 complexes would be reversed to susceptible after administration with LPPC/MP/CTLA4 complexes. NGS data revealed that LPPC/MP/CD28 complexes could enhance the gene expression of cytokine and chemokine pathways to strengthen immune activation than LPPC/MP, and that LPPC/MP/CTLA4 could abolish the LPPC/MP complex-mediated gene expression back to un-treatment. CONCLUSIONS Overall, we proved a convenient and flexible immunotherapy platform for developing personalized cancer therapy.
Collapse
Affiliation(s)
- Chia-Hung Chen
- grid.413593.90000 0004 0573 007XDepartment of Medical Research, Hsinchu MacKay Memorial Hospital, Hsinchu City, 30071 Taiwan
| | - Tzu-Han Weng
- grid.413593.90000 0004 0573 007XDepartment of Dermatology, MacKay Memorial Hospital, Taipei City, 10449 Taiwan
| | - Hsiao-Hsuan Huang
- grid.260539.b0000 0001 2059 7017Industrial Development Graduate Program of College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan
| | - Ling-Ya Huang
- grid.260539.b0000 0001 2059 7017Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan
| | - Kai-Yao Huang
- grid.413593.90000 0004 0573 007XDepartment of Medical Research, Hsinchu MacKay Memorial Hospital, Hsinchu City, 30071 Taiwan ,grid.452449.a0000 0004 1762 5613Department of Medicine, MacKay Medical College, 25245 New Taipei City, Taiwan
| | - Pin-Rong Chen
- grid.260539.b0000 0001 2059 7017Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan
| | - Kuang-Yu Yeh
- grid.260539.b0000 0001 2059 7017Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan
| | - Chi-Ting Huang
- grid.260539.b0000 0001 2059 7017Department of Biological Science and Technology, National Yang Ming Chiao Tung University, 30068 Hsinchu City, Taiwan
| | - Yu-Tzu Chien
- grid.260539.b0000 0001 2059 7017Department of Biological Science and Technology, National Yang Ming Chiao Tung University, 30068 Hsinchu City, Taiwan
| | - Po-Ya Chuang
- grid.260539.b0000 0001 2059 7017Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan
| | - Yu-Ling Lin
- grid.28665.3f0000 0001 2287 1366Agricultural Biotechnology Research Center, Academia Sinica, Taipei, 11529 Taiwan
| | - Nu-Man Tsai
- grid.411641.70000 0004 0532 2041Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City, 40201 Taiwan ,grid.411645.30000 0004 0638 9256Department of Pathology and Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City, 40201 Taiwan
| | - Shih-Jen Liu
- grid.59784.370000000406229172National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, 350401 Miaoli, Taiwan
| | - Yu-Cheng Su
- grid.260539.b0000 0001 2059 7017Department of Biological Science and Technology, National Yang Ming Chiao Tung University, 30068 Hsinchu City, Taiwan
| | - Shun-Long Weng
- grid.452449.a0000 0004 1762 5613Department of Medicine, MacKay Medical College, 25245 New Taipei City, Taiwan ,grid.413593.90000 0004 0573 007XDepartment of Obstetrics and Gynecology, Hsinchu MacKay Memorial Hospital, Hsinchu City, 30071 Taiwan ,grid.507991.30000 0004 0639 3191MacKay Junior College of Medicine, Nursing and Management, Taipei City, 11260 Taiwan
| | - Kuang-Wen Liao
- grid.260539.b0000 0001 2059 7017Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan ,grid.260539.b0000 0001 2059 7017Department of Biological Science and Technology, National Yang Ming Chiao Tung University, 30068 Hsinchu City, Taiwan ,grid.412019.f0000 0000 9476 5696Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City, 80708 Taiwan ,grid.412019.f0000 0000 9476 5696College of Dental Medicine, Kaohsiung Medical University School of Dentistry, Kaohsiung City, 80708 Taiwan ,grid.64523.360000 0004 0532 3255Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan City, 70101 Taiwan ,grid.260539.b0000 0001 2059 7017Center for Intelligent Drug Systems and Smart Bio-Devices, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan
| |
Collapse
|
24
|
Cohn IS, Henrickson SE, Striepen B, Hunter CA. Immunity to Cryptosporidium: Lessons from Acquired and Primary Immunodeficiencies. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2261-2268. [PMID: 36469846 PMCID: PMC9731348 DOI: 10.4049/jimmunol.2200512] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/30/2022] [Indexed: 01/04/2023]
Abstract
Cryptosporidium is a ubiquitous protozoan parasite that infects gut epithelial cells and causes self-limited diarrhea in immunocompetent individuals. However, in immunocompromised hosts with global defects in T cell function, this infection can result in chronic, life-threatening disease. In addition, there is a subset of individuals with primary immunodeficiencies associated with increased risk for life-threatening cryptosporidiosis. These patients highlight MHC class II expression, CD40-CD40L interactions, NF-κB signaling, and IL-21 as key host factors required for resistance to this enteric pathogen. Understanding which immune deficiencies do (or do not) lead to increased risk for severe Cryptosporidium may reveal mechanisms of parasite restriction and aid in the identification of novel strategies to manage this common pathogen in immunocompetent and deficient hosts.
Collapse
Affiliation(s)
- Ian S. Cohn
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah E. Henrickson
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA
- Division of Allergy Immunology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
25
|
Wu R, Murphy KM. DCs at the center of help: Origins and evolution of the three-cell-type hypothesis. J Exp Med 2022; 219:e20211519. [PMID: 35543702 PMCID: PMC9098650 DOI: 10.1084/jem.20211519] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 11/06/2022] Open
Abstract
Last year was the 10th anniversary of Ralph Steinman's Nobel Prize awarded for his discovery of dendritic cells (DCs), while next year brings the 50th anniversary of that discovery. Current models of anti-viral and anti-tumor immunity rest solidly on Steinman's discovery of DCs, but also rely on two seemingly unrelated phenomena, also reported in the mid-1970s: the discoveries of "help" for cytolytic T cell responses by Cantor and Boyse in 1974 and "cross-priming" by Bevan in 1976. Decades of subsequent work, controversy, and conceptual changes have gradually merged these three discoveries into current models of cell-mediated immunity against viruses and tumors.
Collapse
Affiliation(s)
- Renee Wu
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO
| | - Kenneth M. Murphy
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO
| |
Collapse
|
26
|
Prasit KK, Ferrer-Font L, Burn OK, Anderson RJ, Compton BJ, Schmidt AJ, Mayer JU, Chen CJJ, Dasyam N, Ritchie DS, Godfrey DI, Mattarollo SR, Dundar PR, Painter GF, Hermans IF. Intratumoural administration of an NKT cell agonist with CpG promotes NKT cell infiltration associated with an enhanced antitumour response and abscopal effect. Oncoimmunology 2022; 11:2081009. [PMID: 35712122 PMCID: PMC9196710 DOI: 10.1080/2162402x.2022.2081009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Intratumoural administration of unmethylated cytosine-phosphate-guanine motifs (CpG) to stimulate toll-like receptor (TLR)-9 has been shown to induce tumour regression in preclinical studies and some efficacy in the clinic. Because activated natural killer T (NKT) cells can cooperate with pattern-recognition via TLRs to improve adaptive immune responses, we assessed the impact of combining a repeated dosing regimen of intratumoural CpG with a single intratumoural dose of the NKT cell agonist α-galactosylceramide (α-GalCer). The combination was superior to CpG alone at inducing regression of established tumours in several murine tumour models, primarily mediated by CD8+ T cells. An antitumour effect on distant untreated tumours (abscopal effect) was reliant on sustained activity of NKT cells and was associated with infiltration of KLRG1+ NKT cells in tumours and draining lymph nodes at both injected and untreated distant sites. Cytometric analysis pointed to increased exposure to type I interferon (IFN) affecting many immune cell types in the tumour and lymphoid organs. Accordingly, antitumour activity was lost in animals in which dendritic cells (DCs) were incapable of signaling through the type I IFN receptor. Studies in conditional ablation models showed that conventional type 1 DCs and plasmacytoid DCs were required for the response. In tumour models where the combined treatment was less effective, the addition of tumour-antigen derived peptide, preferably conjugated to α-GalCer, significantly enhanced the antitumour response. The combination of TLR ligation, NKT cell agonism, and peptide delivery could therefore be adapted to induce responses to both known and unknown antigens.
Collapse
Affiliation(s)
- Kef K Prasit
- Malaghan Institute of Medical Research, Wellington, New Zealand.,Maurice Wilkins Centre, Auckland, New Zealand
| | - Laura Ferrer-Font
- Maurice Wilkins Centre, Auckland, New Zealand.,Hugh Green Cytometry Centre, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Olivia K Burn
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Regan J Anderson
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Benjamin J Compton
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Alfonso J Schmidt
- Hugh Green Cytometry Centre, Malaghan Institute of Medical Research, Wellington, New Zealand
| | | | - Chun-Jen J Chen
- Maurice Wilkins Centre, Auckland, New Zealand.,School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | | | - David S Ritchie
- Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia.,University of Melbourne, Melbourne, Australia
| | - Dale I Godfrey
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Australia
| | - Stephen R Mattarollo
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - P Rod Dundar
- Maurice Wilkins Centre, Auckland, New Zealand.,School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Gavin F Painter
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Ian F Hermans
- Malaghan Institute of Medical Research, Wellington, New Zealand.,Maurice Wilkins Centre, Auckland, New Zealand
| |
Collapse
|
27
|
Kawabe T, Ciucci T, Kim KS, Tayama S, Kawajiri A, Suzuki T, Tanaka R, Ishii N, Jankovic D, Zhu J, Sprent J, Bosselut R, Sher A. Redefining the Foreign Antigen and Self-Driven Memory CD4 + T-Cell Compartments via Transcriptomic, Phenotypic, and Functional Analyses. Front Immunol 2022; 13:870542. [PMID: 35707543 PMCID: PMC9190281 DOI: 10.3389/fimmu.2022.870542] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/29/2022] [Indexed: 01/03/2023] Open
Abstract
Under steady-state conditions, conventional CD4+ T lymphocytes are classically divided into naïve (CD44lo CD62Lhi) and memory (CD44hi CD62Llo) cell compartments. While the latter population is presumed to comprise a mixture of distinct subpopulations of explicit foreign antigen (Ag)-specific “authentic” memory and foreign Ag-independent memory-phenotype (MP) cells, phenotypic markers differentially expressed in these two cell types have yet to be identified. Moreover, while MP cells themselves have been previously described as heterogeneous, it is unknown whether they consist of distinct subsets defined by marker expression. In this study, we demonstrate using combined single-cell RNA sequencing and flow cytometric approaches that self-driven MP CD4+ T lymphocytes are divided into CD127hi Sca1lo, CD127hi Sca1hi, CD127lo Sca1hi, and CD127lo Sca1lo subpopulations that are Bcl2lo, while foreign Ag-specific memory cells are CD127hi Sca1hi Bcl2hi. We further show that among the four MP subsets, CD127hi Sca1hi lymphocytes represent the most mature and cell division-experienced subpopulation derived from peripheral naïve precursors. Finally, we provide evidence arguing that this MP subpopulation exerts the highest responsiveness to Th1-differentiating cytokines and can induce colitis. Together, our findings define MP CD4+ T lymphocytes as a unique, self-driven population consisting of distinct subsets that differ from conventional foreign Ag-specific memory cells in marker expression and establish functional relevance for the mature subset of CD127hi Sca1hi MP cells.
Collapse
Affiliation(s)
- Takeshi Kawabe
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan.,Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Thomas Ciucci
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.,David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester, Rochester, NY, United States
| | - Kwang Soon Kim
- Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, South Korea
| | - Shunichi Tayama
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akihisa Kawajiri
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takumi Suzuki
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Riou Tanaka
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Naoto Ishii
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Dragana Jankovic
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Jinfang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Jonathan Sprent
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Rémy Bosselut
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
28
|
Reactivation of latent tuberculosis with TNF inhibitors: critical role of the beta 2 chain of the IL-12 receptor. Cell Mol Immunol 2021; 18:1644-1651. [PMID: 34021269 PMCID: PMC8245521 DOI: 10.1038/s41423-021-00694-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/26/2021] [Indexed: 02/04/2023] Open
Abstract
Tumor necrosis factor (TNF) inhibitors have improved a lot the treatment of numerous diseases, with the well-known example of rheumatoid arthritis (RA). In the early 2000s, postmarketing data quickly revealed an alarming number of severe tuberculosis (TB) under such treatment. These findings were consistent with previous results in mice where TNF is essential for lymph node formation and granuloma organization. The effects of TNF inhibition on RA synovium structure are very similar to those on granuloma, with changes in cellular interactions, cytokine, and chemokine production. In addition to the role of TNF in granuloma, the interleukin (IL)-12/interferon (IFN)-γ pathway is required for an efficient host defense against TB. Primary and secondary immunodeficiencies affecting this pathway lead to severe bacillus Calmette-Guérin (BCG) reaction or full TB. Any chronic inflammation as in RA induces a systemic Th1 defect that predisposes to TB through specific downregulation of the IL-12Rß2 chain. When TNF inhibitors are initiated, this transiently increases this risk of TB, through effects on cellular interactions in a latent TB granuloma. At a later stage, when a better control disease activity is obtained, the risk of TB is reduced but not abrogated. Given the clear benefit from TNF inhibition, latent TB infection screening at baseline is essential for an optimal safety.
Collapse
|
29
|
Cabeza-Cabrerizo M, Cardoso A, Minutti CM, Pereira da Costa M, Reis E Sousa C. Dendritic Cells Revisited. Annu Rev Immunol 2021; 39:131-166. [PMID: 33481643 DOI: 10.1146/annurev-immunol-061020-053707] [Citation(s) in RCA: 417] [Impact Index Per Article: 104.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dendritic cells (DCs) possess the ability to integrate information about their environment and communicate it to other leukocytes, shaping adaptive and innate immunity. Over the years, a variety of cell types have been called DCs on the basis of phenotypic and functional attributes. Here, we refocus attention on conventional DCs (cDCs), a discrete cell lineage by ontogenetic and gene expression criteria that best corresponds to the cells originally described in the 1970s. We summarize current knowledge of mouse and human cDC subsets and describe their hematopoietic development and their phenotypic and functional attributes. We hope that our effort to review the basic features of cDC biology and distinguish cDCs from related cell types brings to the fore the remarkable properties of this cell type while shedding some light on the seemingly inordinate complexity of the DC field.
Collapse
Affiliation(s)
- Mar Cabeza-Cabrerizo
- Immunobiology Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom;
| | - Ana Cardoso
- Immunobiology Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom;
| | - Carlos M Minutti
- Immunobiology Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom;
| | | | - Caetano Reis E Sousa
- Immunobiology Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom;
| |
Collapse
|
30
|
Dermal IRF4+ dendritic cells and monocytes license CD4+ T helper cells to distinct cytokine profiles. Nat Commun 2020; 11:5637. [PMID: 33159073 PMCID: PMC7647995 DOI: 10.1038/s41467-020-19463-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 10/11/2020] [Indexed: 12/14/2022] Open
Abstract
Antigen (Ag)-presenting cells (APC) instruct CD4+ helper T (Th) cell responses, but it is unclear whether different APC subsets contribute uniquely in determining Th differentiation in pathogen-specific settings. Here, we use skin-relevant, fluorescently-labeled bacterial, helminth or fungal pathogens to track and characterize the APC populations that drive Th responses in vivo. All pathogens are taken up by a population of IRF4+ dermal migratory dendritic cells (migDC2) that similarly upregulate surface co-stimulatory molecules but express pathogen-specific cytokine and chemokine transcripts. Depletion of migDC2 reduces the amount of Ag in lymph node and the development of IFNγ, IL-4 and IL-17A responses without gain of other cytokine responses. Ag+ monocytes are an essential source of IL-12 for both innate and adaptive IFNγ production, and inhibit follicular Th cell development. Our results thus suggest that Th cell differentiation does not require specialized APC subsets, but is driven by inducible and pathogen-specific transcriptional programs in Ag+ migDC2 and monocytes.
Collapse
|
31
|
Fusion of Dendritic Cells Activating Rv2299c Protein Enhances the Protective Immunity of Ag85B-ESAT6 Vaccine Candidate against Tuberculosis. Pathogens 2020; 9:pathogens9110865. [PMID: 33105734 PMCID: PMC7690420 DOI: 10.3390/pathogens9110865] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/16/2020] [Accepted: 10/20/2020] [Indexed: 12/21/2022] Open
Abstract
In Mycobacterium tuberculosis infection, naïve T cells that encounter mycobacterial antigens through dendritic cells (DCs) induce various CD4+ T-cell responses; therefore, appropriate DC activation is the key for protective immunity against tuberculosis. We previously found that Rv2299c-matured DCs induce Th1 differentiation with bactericidal activity. In this study, to prove that Rv2299c could enhance the protective immunity of other vaccine candidates comprising T-cell-stimulating antigens, Ag85B-ESAT6, a well-known vaccine candidate, was selected as a fusion partner of Rv2299c. Recombinant Rv2299c-Ag85B-ESAT6 protein induced DC maturation and activation. Furthermore, fusion of Rv2299c enhanced the protective efficacy of the Ag85B-ESAT6 vaccine in a mouse model and significantly higher production of TNF-α and IL-2 was detected in the lungs, spleen, and lymph nodes of the group immunized with the Rv2299c-fused protein than with Ag85B-ESAT6. In addition, fusion of Rv2299c enhanced the Ag85B-ESAT6-mediated expansion of multifunctional CD4+ T cells. These data suggested that the DC-activating protein Rv2299c may potentiate the protective immunity of the vaccine candidate comprising T cell antigens.
Collapse
|
32
|
Sun T, Nguyen A, Gommerman JL. Dendritic Cell Subsets in Intestinal Immunity and Inflammation. THE JOURNAL OF IMMUNOLOGY 2020; 204:1075-1083. [PMID: 32071090 DOI: 10.4049/jimmunol.1900710] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 10/11/2019] [Indexed: 12/21/2022]
Abstract
The mammalian intestine is a complex environment that is constantly exposed to Ags derived from food, microbiota, and metabolites. Intestinal dendritic cells (DC) have the responsibility of establishing oral tolerance against these Ags while initiating immune responses against mucosal pathogens. We now know that DC are a heterogeneous population of innate immune cells composed of classical and monocyte-derived DC, Langerhans cells, and plasmacytoid DC. In the intestine, DC are found in organized lymphoid tissues, such as the mesenteric lymph nodes and Peyer's patches, as well as in the lamina propria. In this Brief Review, we review recent work that describes a division of labor between and collaboration among gut DC subsets in the context of intestinal homeostasis and inflammation. Understanding relationships between DC subtypes and their biological functions will rationalize oral vaccine design and will provide insights into treatments that quiet pathological intestinal inflammation.
Collapse
Affiliation(s)
- Tian Sun
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S1A8, Canada
| | - Albert Nguyen
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S1A8, Canada
| | - Jennifer L Gommerman
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S1A8, Canada
| |
Collapse
|
33
|
Requirements for the differentiation of innate T-bet high memory-phenotype CD4 + T lymphocytes under steady state. Nat Commun 2020; 11:3366. [PMID: 32632165 PMCID: PMC7338451 DOI: 10.1038/s41467-020-17136-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 06/11/2020] [Indexed: 12/13/2022] Open
Abstract
CD4+ T lymphocytes consist of naïve, antigen-specific memory, and memory-phenotype (MP) cell compartments at homeostasis. We recently showed that MP cells exert innate-like effector function during host defense, but whether MP CD4+ T cells are functionally heterogeneous and, if so, what signals specify the differentiation of MP cell subpopulations under homeostatic conditions is still unclear. Here we characterize MP lymphocytes as consisting of T-bethigh, T-betlow, and T-bet− subsets, with innate, Th1-like effector activity exclusively associated with T-bethigh cells. We further show that the latter population depends on IL-12 produced by CD8α+ type 1 dendritic cells (DC1) for its differentiation. Finally, our data demonstrate that this tonic IL-12 production requires TLR-MyD88 signaling independent of foreign agonists, and is further enhanced by CD40-CD40L interactions between DC1 and CD4+ T lymphocytes. We propose that optimal differentiation of T-bethigh MP lymphocytes at homeostasis is driven by self-recognition signals at both the DC and Tcell levels. CD4+ T cells contain a T-bethigh memory-phenotype (MP) population with innate-like functions. Here the authors characterize the requirements for their differentiation at homeostasis and identify a function for IL-12 that is tonically produced by type 1 dendritic cells in an MyD88- and CD40-dependent, but foreign PAMP-independent manner.
Collapse
|
34
|
Byrne-Hoffman CN, Deng W, McGrath O, Wang P, Rojanasakul Y, Klinke DJ. Interleukin-12 elicits a non-canonical response in B16 melanoma cells to enhance survival. Cell Commun Signal 2020; 18:78. [PMID: 32450888 PMCID: PMC7249691 DOI: 10.1186/s12964-020-00547-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/06/2020] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Oncogenesis rewires signaling networks to confer a fitness advantage to malignant cells. For instance, the B16F0 melanoma cell model creates a cytokine sink for Interleukin-12 (IL-12) to deprive neighboring cells of this important anti-tumor immune signal. While a cytokine sink provides an indirect fitness advantage, does IL-12 provide an intrinsic advantage to B16F0 cells? METHODS Acute in vitro viability assays were used to compare the cytotoxic effect of imatinib on a melanoma cell line of spontaneous origin (B16F0) with a normal melanocyte cell line (Melan-A) in the presence of IL-12. The results were analyzed using a mathematical model coupled with a Markov Chain Monte Carlo approach to obtain a posterior distribution in the parameters that quantified the biological effect of imatinib and IL-12. Intracellular signaling responses to IL-12 were compared using flow cytometry in 2D6 cells, a cell model for canonical signaling, and B16F0 cells, where potential non-canonical signaling occurs. Bayes Factors were used to select among competing signaling mechanisms that were formulated as mathematical models. Analysis of single cell RNAseq data from human melanoma patients was used to explore generalizability. RESULTS Functionally, IL-12 enhanced the survival of B16F0 cells but not normal Melan-A melanocytes that were challenged with a cytotoxic agent. Interestingly, the ratio of IL-12 receptor components (IL12RB2:IL12RB1) was increased in B16F0 cells. A similar pattern was observed in human melanoma. To identify a mechanism, we assayed the phosphorylation of proteins involved in canonical IL-12 signaling, STAT4, and cell survival, Akt. In contrast to T cells that exhibited a canonical response to IL-12 by phosphorylating STAT4, IL-12 stimulation of B16F0 cells predominantly phosphorylated Akt. Mechanistically, the differential response in B16F0 cells is explained by both ligand-dependent and ligand-independent aspects to initiate PI3K-AKT signaling upon IL12RB2 homodimerization. Namely, IL-12 promotes IL12RB2 homodimerization with low affinity and IL12RB2 overexpression promotes homodimerization via molecular crowding on the plasma membrane. CONCLUSIONS The data suggest that B16F0 cells shifted the intracellular response to IL-12 from engaging immune surveillance to favoring cell survival. Identifying how signaling networks are rewired in model systems of spontaneous origin can inspire therapeutic strategies in humans. Interleukin-12 is a key cytokine that promotes anti-tumor immunity, as it is secreted by antigen presenting cells to activate Natural Killer cells and T cells present within the tumor microenvironment. Thinking of cancer as an evolutionary process implies that an immunosuppressive tumor microenvironment could arise during oncogenesis by interfering with endogenous anti-tumor immune signals, like IL-12. Previously, we found that B16F0 cells, a cell line derived from a spontaneous melanoma, interrupts this secreted heterocellular signal by sequestering IL-12, which provides an indirect fitness advantage. Normally, IL-12 signals via a receptor comprised of two components, IL12RB1 and IL12RB2, that are expressed in a 1:1 ratio and activates STAT4 as a downstream effector. Here, we report that B16F0 cells gain an intrinsic advantage by rewiring the canonical response to IL-12 to instead initiate PI3K-AKT signaling, which promotes cell survival. The data suggest a model where overexpressing one component of the IL-12 receptor, IL12RB2, enables melanoma cells to shift the functional response via both IL-12-mediated and molecular crowding-based IL12RB2 homodimerization. To explore the generalizability of these results, we also found that the expression of IL12RB2:IL12RB1 is similarly skewed in human melanoma based on transcriptional profiles of melanoma cells and tumor-infiltrating lymphocytes. Additional file 6: Video abstract. (MP4 600 kb).
Collapse
Affiliation(s)
- Christina N Byrne-Hoffman
- Department of Pharmaceutical Sciences; West Virginia University, 1 Medical Center Drive, Morgantown, 26506, WV, US
| | - Wentao Deng
- Department of Microbiology, Immunology, and Cell Biology; West Virginia University, 1 Medical Center Drive, Morgantown, 26506, WV, US
| | - Owen McGrath
- Department of Chemical and Biomedical Engineering; West Virginia University, 395 Evansdale Drive, Morgantown, 26506, WV, US
| | - Peng Wang
- Department of Pharmaceutical Sciences; West Virginia University, 1 Medical Center Drive, Morgantown, 26506, WV, US
| | - Yon Rojanasakul
- Department of Pharmaceutical Sciences; West Virginia University, 1 Medical Center Drive, Morgantown, 26506, WV, US
| | - David J Klinke
- Department of Microbiology, Immunology, and Cell Biology; West Virginia University, 1 Medical Center Drive, Morgantown, 26506, WV, US. .,Department of Chemical and Biomedical Engineering; West Virginia University, 395 Evansdale Drive, Morgantown, 26506, WV, US. .,WVU Cancer Institute; West Virginia University, 1 Medical Center Drive, Morgantown, 26506, WV, US.
| |
Collapse
|
35
|
Hilligan KL, Ronchese F. Antigen presentation by dendritic cells and their instruction of CD4+ T helper cell responses. Cell Mol Immunol 2020; 17:587-599. [PMID: 32433540 DOI: 10.1038/s41423-020-0465-0] [Citation(s) in RCA: 232] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/10/2020] [Indexed: 12/20/2022] Open
Abstract
Dendritic cells are powerful antigen-presenting cells that are essential for the priming of T cell responses. In addition to providing T-cell-receptor ligands and co-stimulatory molecules for naive T cell activation and expansion, dendritic cells are thought to also provide signals for the differentiation of CD4+ T cells into effector T cell populations. The mechanisms by which dendritic cells are able to adapt and respond to the great variety of infectious stimuli they are confronted with, and prime an appropriate CD4+ T cell response, are only partly understood. It is known that in the steady-state dendritic cells are highly heterogenous both in phenotype and transcriptional profile, and that this variability is dependent on developmental lineage, maturation stage, and the tissue environment in which dendritic cells are located. Exposure to infectious agents interfaces with this pre-existing heterogeneity by providing ligands for pattern-recognition and toll-like receptors that are variably expressed on different dendritic cell subsets, and elicit production of cytokines and chemokines to support innate cell activation and drive T cell differentiation. Here we review current information on dendritic cell biology, their heterogeneity, and the properties of different dendritic cell subsets. We then consider the signals required for the development of different types of Th immune responses, and the cellular and molecular evidence implicating different subsets of dendritic cells in providing such signals. We outline how dendritic cell subsets tailor their response according to the infectious agent, and how such transcriptional plasticity enables them to drive different types of immune responses.
Collapse
Affiliation(s)
- Kerry L Hilligan
- Malaghan Institute of Medical Research, Wellington, 6012, New Zealand.,Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Franca Ronchese
- Malaghan Institute of Medical Research, Wellington, 6012, New Zealand.
| |
Collapse
|
36
|
Abstract
Alzheimer’s disease (AD) is a chronic neurodegenerative disease characterized by the accumulation of amyloid plaques and neurofibrillary tangles in the brain. The AD pathophysiology entails chronic inflammation involving innate immune cells including microglia, astrocytes, and other peripheral blood cells. Inflammatory mediators such as cytokines and complements are also linked to AD pathogenesis. Despite increasing evidence supporting the association between abnormal inflammation and AD, no well-established inflammatory biomarkers are currently available for AD. Since many reports have shown that abnormal inflammation precedes the outbreak of the disease, non-invasive and readily available peripheral inflammatory biomarkers should be considered as possible biomarkers for early diagnosis of AD. In this mini-review, we introduce the peripheral biomarker candidates related to abnormal inflammation in AD and discuss their possible molecular mechanisms. Furthermore, we also summarize the current state of inflammatory biomarker research in clinical practice and molecular diagnostics. We believe this review will provide new insights into biomarker candidates for the early diagnosis of AD with systemic relevance to inflammation during AD pathogenesis.
Collapse
Affiliation(s)
- Jong-Chan Park
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Korea
| | - Sun-Ho Han
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Korea
| | - Inhee Mook-Jung
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Korea
| |
Collapse
|
37
|
Connick K, Lalor R, Murphy A, O'Neill SM, El Shanawany EE. Sarcocystis fusiformis whole cyst antigen activates pro-inflammatory dendritic cells. J Parasit Dis 2020; 44:186-193. [PMID: 32174724 DOI: 10.1007/s12639-019-01181-9] [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: 08/17/2019] [Accepted: 11/18/2019] [Indexed: 11/30/2022] Open
Abstract
Sarcocystis fusiformis is a coccidian tissue parasite that causes infection in buffalo in countries such an Egypt, China, Iraq and Iran, resulting in significant economic losses to the agricultural industry annually. There is a lack of studies examining host-parasite interactions at the level of the immune response and the present study investigates the interaction between S. fusiformis whole cyst antigens (SFWCA) and dendritic cells (DCs), cells critical to the activation of adaptive immunity. In this study bone marrow derived DCs (BMDCs) were phenotyped following treatment with SFWCA by measuring cell viability, cytokine secretion, and cell surface marker expression. While SFWCA exhibited cytotoxic effects on BMDCs at higher concentrations, lower concentrations of SFWCA activated pro-inflammatory DCs that significantly secreted interleukin (IL)-12p40, tumor necrosis factor alpha, IL-6 and IL-10. These cells also displayed enhanced expression of TLR4, CD80, CD86 and MHC II on their surface, which is indicative of full DCs maturation. Moreover, SFWCA significantly attenuated the capacity of BMDCs to suppress Th2 associated cytokines, notably IL-5 and IL-13, while simultaneously exhibiting no effects on the secretion of interferon (IFN)-γ, IL-2, IL-17, and IL-10. In conclusion, this is the first study to provide fundamental insight into the activation of DCs by SFWCA, providing us with some awareness into the interaction of the Sarcosystis parasite with its host. The pro-inflammatory inducing ability of this antigen is in keeping with studies performed in other protozoan parasites and therefore understanding these interactions is important in the development of future therapeutic strategies.
Collapse
Affiliation(s)
- K Connick
- 1School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - R Lalor
- 1School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - A Murphy
- 1School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - S M O'Neill
- 1School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Eman E El Shanawany
- 2Department of Parasitology and Animal Diseases, National Research Centre, El Bhooth St, Dokki, Post Pox 12622, Giza, Egypt
| |
Collapse
|
38
|
Shirley JL, Keeler GD, Sherman A, Zolotukhin I, Markusic DM, Hoffman BE, Morel LM, Wallet MA, Terhorst C, Herzog RW. Type I IFN Sensing by cDCs and CD4 + T Cell Help Are Both Requisite for Cross-Priming of AAV Capsid-Specific CD8 + T Cells. Mol Ther 2020; 28:758-770. [PMID: 31780366 PMCID: PMC7054715 DOI: 10.1016/j.ymthe.2019.11.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 12/23/2022] Open
Abstract
Adeno-associated virus (AAV) vectors are widely used in clinical gene therapy to correct genetic disease by in vivo gene transfer. Although the vectors are useful, in part because of their limited immunogenicity, immune responses directed at vector components have complicated applications in humans. These include, for instance, innate immune sensing of vector components by plasmacytoid dendritic cells (pDCs), which sense the vector DNA genome via Toll-like receptor 9. Adaptive immune responses employ antigen presentation by conventional dendritic cells (cDCs), which leads to cross-priming of capsid-specific CD8+ T cells. In this study, we sought to determine the mechanisms that promote licensing of cDCs, which is requisite for CD8+ T cell activation. Blockage of type 1 interferon (T1 IFN) signaling by monoclonal antibody therapy prevented cross-priming. Furthermore, experiments in cell-type-restricted knockout mice showed a specific requirement for the receptor for T1 IFN (IFNaR) in cDCs. In contrast, natural killer (NK) cells are not needed, indicating a direct rather than indirect effect of T1 IFN on cDCs. In addition, co-stimulation by CD4+ T cells via CD40-CD40L was required for cross-priming, and blockage of co-stimulation but not of T1 IFN additionally reduced antibody formation against capsid. These mechanistic insights inform the development of targeted immune interventions.
Collapse
Affiliation(s)
- Jamie L Shirley
- Department Pediatrics, University of Florida, Gainesville, FL, USA
| | | | | | - Irene Zolotukhin
- Department Pediatrics, University of Florida, Gainesville, FL, USA
| | - David M Markusic
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Brad E Hoffman
- Department Pediatrics, University of Florida, Gainesville, FL, USA
| | - Laurence M Morel
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Mark A Wallet
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA, USA
| | - Roland W Herzog
- Department Pediatrics, University of Florida, Gainesville, FL, USA; Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
39
|
Bourque J, Hawiger D. Immunomodulatory Bonds of the Partnership between Dendritic Cells and T Cells. Crit Rev Immunol 2019; 38:379-401. [PMID: 30792568 DOI: 10.1615/critrevimmunol.2018026790] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
By acquiring, processing, and presenting both foreign and self-antigens, dendritic cells (DCs) initiate T cell activation that is shaped through the immunomodulatory functions of a variety of cell-membrane-bound molecules including BTLA-HVEM, CD40-CD40L, CTLA-4-CD80/CD86, CD70-CD27, ICOS-ICOS-L, OX40-OX40L, and PD-L1-PD-1, as well as several key cytokines and enzymes such as interleukin-6 (IL-6), IL-12, IL-23, IL-27, transforming growth factor-beta 1 (TGF-β1), retinaldehyde dehydrogenase (Raldh), and indoleamine 2,3-dioxygenase (IDO). Some of these distinct immunomodulatory signals are mediated by specific subsets of DCs, therefore contributing to the functional specialization of DCs in the priming and regulation of immune responses. In addition to responding to the DC-mediated signals, T cells can reciprocally modulate the immunomodulatory capacities of DCs, further refining immune responses. Here, we review recent studies, particularly in experimental mouse systems, that have delineated the integrated mechanisms of crucial immunomodulatory pathways that enable specific populations of DCs and T cells to work intimately together as single functional units that are indispensable for the maintenance of immune homeostasis.
Collapse
Affiliation(s)
- Jessica Bourque
- Department of Molecular Microbiology and Immunology, St. Louis University School of Medicine, St. Louis, MO, USA
| | - Daniel Hawiger
- Department of Molecular Microbiology and Immunology, St. Louis University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
40
|
Abstract
Cancer immunotherapy aims to promote the activity of cytotoxic T lymphocytes (CTLs) within a tumour, assist the priming of tumour-specific CTLs in lymphoid organs and establish efficient and durable antitumour immunity. During priming, help signals are relayed from CD4+ T cells to CD8+ T cells by specific dendritic cells to optimize the magnitude and quality of the CTL response. In this Review, we highlight the cellular dynamics and membrane receptors that mediate CD4+ T cell help and the molecular mechanisms of the enhanced antitumour activity of CTLs. We outline how deficient CD4+ T cell help reduces the response of CTLs and how maximizing CD4+ T cell help can improve outcomes in cancer immunotherapy strategies.
Collapse
|
41
|
Poncet AF, Blanchard N, Marion S. Toxoplasma and Dendritic Cells: An Intimate Relationship That Deserves Further Scrutiny. Trends Parasitol 2019; 35:870-886. [PMID: 31492624 DOI: 10.1016/j.pt.2019.08.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/01/2019] [Accepted: 08/04/2019] [Indexed: 02/07/2023]
Abstract
Toxoplasma gondii (Tg), an obligate intracellular parasite of the phylum Apicomplexa, infects a wide range of animals, including humans. A hallmark of Tg infection is the subversion of host responses, which is thought to favor parasite persistence and propagation to new hosts. Recently, a variety of parasite-secreted modulatory effectors have been uncovered in fibroblasts and macrophages, but the specific interplay between Tg and dendritic cells (DCs) is just beginning to emerge. In this review, we summarize the current knowledge on Tg-DC interactions, including innate recognition, cytokine production, and antigen presentation, and discuss open questions regarding how Tg-secreted effectors may shape DC functions to perturb innate and adaptive immunity.
Collapse
Affiliation(s)
- Anaïs F Poncet
- Centre d'Infection et d'Immunité de Lille, Université de Lille, Inserm U1019, CNRS UMR 8204, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Nicolas Blanchard
- Centre de Physiopathologie Toulouse Purpan (CPTP), Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France. @inserm.fr
| | - Sabrina Marion
- Centre d'Infection et d'Immunité de Lille, Université de Lille, Inserm U1019, CNRS UMR 8204, CHU Lille, Institut Pasteur de Lille, Lille, France. @pasteur-lille.fr
| |
Collapse
|
42
|
Lin B, Dutta B, Fraser IDC. Systematic Investigation of Multi-TLR Sensing Identifies Regulators of Sustained Gene Activation in Macrophages. Cell Syst 2019; 5:25-37.e3. [PMID: 28750197 DOI: 10.1016/j.cels.2017.06.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 04/26/2017] [Accepted: 06/22/2017] [Indexed: 12/22/2022]
Abstract
A typical pathogen presents a combination of Toll-like receptor (TLR) ligands during infection. Although individual TLR pathways have been well characterized, the nature of this "combinatorial code" in pathogen sensing remains unclear. Here, we conducted a comprehensive transcriptomic analysis of primary macrophages stimulated with all possible pairwise combinations of four different TLR ligands to understand the requirements, kinetics, and outcome of combined pathway engagement. We find that signal integration between TLR pathways leads to non-additive responses for a subset of immune mediators with sustained expression (>6 hr) properties and T cell polarizing function. To identify the underlying regulators, we conducted a focused RNAi screen and identified four genes-Helz2, Phf11d, Sertad3, and Zscan12-which preferentially affect the late phase response of TLR-induced immune effector expression. This study reveals key molecular details of how contemporaneous signaling through multiple TLRs, as would often be the case with pathogen infection, produce biological outcomes distinct from the single ligands typically used to characterize TLR pathways.
Collapse
Affiliation(s)
- Bin Lin
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bhaskar Dutta
- Bioinformatics Group, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Iain D C Fraser
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
43
|
Gramlich R, Aliahmadi E, Peiser M. In Vitro Induction of T Helper 17 Cells by Synergistic Activation of Human Monocyte-Derived Langerhans Cell-Like Cells with Bacterial Agonists. Int J Mol Sci 2019; 20:ijms20061367. [PMID: 30893757 PMCID: PMC6471444 DOI: 10.3390/ijms20061367] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/05/2019] [Accepted: 03/08/2019] [Indexed: 01/02/2023] Open
Abstract
In the case of epidermal barrier disruption, pathogens encounter skin-resident Langerhans cells (LCs) and are recognized by pathogen recognition receptors such as Toll-like receptors (TLRs). As the majority of microorganisms exhibit more than one TLR ligand, the mechanisms of subsequent T cell differentiation are complex and far from clear. In this study, we investigated combinatory effects on Th cell polarization by bacterial cell wall compounds peptidoglycan (PGN) and lipopolysaccharide (LPS) and by bacterial nucleic acid (DNA). Expression of maturation markers CD40, CD80, HLA-DR and CCR7 and the release of IL-1β, IL-6 and IL-23 was strongly enhanced by simultaneous exposure to PGN, LPS and DNA in LCs. As all these factors were potential Th17 driving cytokines, we investigated the potency of combinatory TLR stimuli to induce Th17 cells via LC activation. High amounts of IL-17A and IL-22, key cytokines of Th17 cells, were detected. By intracellular costaining of IL-17+T cells, IL-22− (Th17) and IL-22+ (immature Th17) cells were identified. Interestingly, one population of LPS stimulated cells skewed into IL-9+Th cells, and LPS synergized with PGN while inducing high IL-22. In conclusion, our data indicates that when mediated by a fine-tuned signal integration via LCs, bacterial TLR agonists synergize and induce Th17 differentiation.
Collapse
Affiliation(s)
- Robert Gramlich
- Institute of Molecular Biology and Bioinformatics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany.
- Center for Anesthesiology and Intensive Care Medicine, Department of intensive Care, Medical Center Hamburg-Eppendorf (UKE), Martinistrasse 52, 20246 Hamburg, Germany.
| | - Ehsan Aliahmadi
- Institute of Molecular Biology and Bioinformatics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany.
- Department of Surgery and Vascular Surgery, Franziskus Krankenhaus Berlin, University Hospital of Charité Universitätsmedizin Berlin, Budapester Straße 15-19, 10787 Berlin, Germany.
| | - Matthias Peiser
- Institute of Molecular Biology and Bioinformatics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany.
- Department of Pesticides Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Strasse 8-10, 10589 Berlin, Germany.
| |
Collapse
|
44
|
CECHIM GIOVANA, CHIES JOSÉA. In vitro generation of human monocyte-derived dendritic cells methodological aspects in a comprehensive review. ACTA ACUST UNITED AC 2019. [DOI: 10.1590/0001-3765201920190310] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
45
|
Shinde P, Bharat V, Rodriguez-Oquendo A, Zhou B, Vella AT. Understanding how combinatorial targeting of TLRs and TNFR family costimulatory members promote enhanced T cell responses. Expert Opin Biol Ther 2018; 18:1073-1083. [PMID: 30169979 DOI: 10.1080/14712598.2018.1518422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 08/28/2018] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Due to the ability of pathogen-associated molecular patters and tumor necrosis factor receptor (TNFR) family costimulatory agonists to boost T cell responses, studies have combined Toll-like receptor (TLR) ligands with TNFR family costimulatory receptor agonists to induce impressive and long-lasting T cell responses. Although some studies have determined how these combinatorial vaccines promote enhanced T cell responses, much remains unknown about the mechanism used by these combinations to promote synergistic T cell responses - especially in settings of infectious diseases or cancer. AREAS COVERED In this review, we look in detail at the signaling pathways induced by combinatorial targeting of TLR and TNFR family costimulatory members that help them promote synergistic T cell responses. Understanding this can greatly aid the development of novel vaccine regimens that promote cellular immune responses, which is essential for treating certain infectious diseases and cancer. EXPERT OPINION Vaccines against some infectious diseases as well as therapeutic cancer vaccines require cellular immunity. Therefore, we evaluate here how signaling pathways induced by TLR ligand and costimulatory agonist combinations promote enhanced T cell responses during immunization with model antigens, viral pathogens, or tumor antigens. Once pathways that drive these combinatorial vaccines to boost T cell activation are identified, they can be incorporated in vaccines designed to target pathogens or cancer.
Collapse
Affiliation(s)
- Paurvi Shinde
- a Bloodworks Northwest Research Institute , Seattle , WA , USA
| | - Vinita Bharat
- b Department of Neurosurgery , Stanford University School of Medicine , Stanford , CA , USA
| | | | - Beiyan Zhou
- d Department of Immunology, UConn School of Medicine , UConn Health , Farmington , CT , USA
| | - Anthony T Vella
- d Department of Immunology, UConn School of Medicine , UConn Health , Farmington , CT , USA
| |
Collapse
|
46
|
Uribe-Herranz M, Bittinger K, Rafail S, Guedan S, Pierini S, Tanes C, Ganetsky A, Morgan MA, Gill S, Tanyi JL, Bushman FD, June CH, Facciabene A. Gut microbiota modulates adoptive cell therapy via CD8α dendritic cells and IL-12. JCI Insight 2018; 3:94952. [PMID: 29467322 DOI: 10.1172/jci.insight.94952] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 01/18/2018] [Indexed: 12/30/2022] Open
Abstract
Adoptive T cell therapy (ACT) is a promising new modality for malignancies. Here, we report that adoptive T cell efficacy in tumor-bearing mice is significantly affected by differences in the native composition of the gut microbiome or treatment with antibiotics, or by heterologous fecal transfer. Depletion of bacteria with vancomycin decreased the rate of tumor growth in mice from The Jackson Laboratory receiving ACT, whereas treatment with neomycin and metronidazole had no effect, indicating the role of specific bacteria in host response. Vancomycin treatment induced an increase in systemic CD8α+ DCs, which sustained systemic adoptively transferred antitumor T cells in an IL-12-dependent manner. In subjects undergoing allogeneic hematopoietic cell transplantation, we found that oral vancomycin also increased IL-12 levels. Collectively, our findings demonstrate an important role played by the gut microbiota in the antitumor effectiveness of ACT and suggest potentially new avenues to improve response to ACT by altering the gut microbiota.
Collapse
Affiliation(s)
- Mireia Uribe-Herranz
- Ovarian Cancer Research Center, and.,Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kyle Bittinger
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | - Sonia Guedan
- Abramson Cancer Center and the Department of Pathology and Laboratory Medicine, Perelman School of Medicine
| | - Stefano Pierini
- Ovarian Cancer Research Center, and.,Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ceylan Tanes
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | | | - Saar Gill
- Division of Hematology Oncology, Department of Medicine, and
| | | | - Frederic D Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Carl H June
- Abramson Cancer Center and the Department of Pathology and Laboratory Medicine, Perelman School of Medicine
| | - Andrea Facciabene
- Ovarian Cancer Research Center, and.,Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
47
|
Peron G, de Lima Thomaz L, Camargo da Rosa L, Thomé R, Cardoso Verinaud LM. Modulation of dendritic cell by pathogen antigens: Where do we stand? Immunol Lett 2018; 196:91-102. [PMID: 29427742 DOI: 10.1016/j.imlet.2018.02.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/29/2018] [Accepted: 02/01/2018] [Indexed: 12/24/2022]
Abstract
Dendritic cells (DCs) are essential players in the activation of T cells and in the development of adaptive immune response towards invading pathogens. Upon antigen (Ag) recognition of Pathogen Associated Molecular Patterns (PAMPs) by their receptors (PRRs), DCs are activated and acquire an inflammatory profile. DCs have the ability to direct the profile of helper T (Th) cells towards Th1, Th2, Th17, Th9 and regulatory (Treg) cells. Each subset of Th cells presents a unique gene expression signature and is endowed with the ability to conduct or suppress effector cells in inflammation. Pathogens target DCs during infection. Many studies demonstrated that antigens and molecules derived from pathogens have the ability to dampen DC maturation and activation, leading these cells to a permissive state or tolerogenic profile (tolDCs). Although tolDCs may represent a hindrance in infection control, they could be positively used to modulate inflammatory disorders, such as autoimmune diseases. In this review, we focus on discussing findings that use pathogen-antigen modulated DCs and tolDCs in prophylactics and therapeutics approaches for vaccination against infectious diseases or inflammatory disorders.
Collapse
Affiliation(s)
- Gabriela Peron
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, Campinas, SP, Brazil.
| | - Livia de Lima Thomaz
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, Campinas, SP, Brazil
| | - Larissa Camargo da Rosa
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, Campinas, SP, Brazil
| | - Rodolfo Thomé
- Department of Neurology, Thomas Jefferson University, Philadelphia, USA
| | - Liana Maria Cardoso Verinaud
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, Campinas, SP, Brazil
| |
Collapse
|
48
|
Issaranggun Na Ayuthaya B, Everts V, Pavasant P. The immunopathogenic and immunomodulatory effects of interleukin-12 in periodontal disease. Eur J Oral Sci 2018; 126:75-83. [PMID: 29411897 DOI: 10.1111/eos.12405] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Interleukin 12 (IL-12) is an inflammatory cytokine that promotes the response of the immune system. This cytokine has been implicated as a potent stimulator of several diseases characterized by inflammatory-induced bone destruction, such as rheumatoid arthritis and periodontitis. Yet, the exact role of IL-12 in the development and progress of periodontitis has not been clarified. Several studies have demonstrated a positive correlation between the level of IL-12 and the severity of periodontal destruction. Deletion of IL-12 in mice with periodontitis significantly suppressed the level of bone destruction. Interestingly, next to a role in modulating the pathogenesis, IL-12 also has immunological-regulatory properties. This cytokine induces expression of immunosuppressive molecules, such as indoleamine-pyrrole 2,3-dioxygenase (IDO). Thus, these findings suggest both negative and positive influences of IL-12 in periodontal disease. It is currently proposed that the diversity of action of cytokines is a molecular key which regulates biological development and homeostasis. Accordingly, the actions of IL-12 might be one of the mechanisms that regulate homeostasis of periodontal tissue during and following inflammation. Therefore, this article aims to review both destructive and protective functionalities of IL-12 with an emphasis on periodontal disease.
Collapse
Affiliation(s)
- Benjar Issaranggun Na Ayuthaya
- Department of Pharmacology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Research Unit of Mineralized Tissue, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Vincent Everts
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, the Netherlands
| | - Prasit Pavasant
- Research Unit of Mineralized Tissue, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
49
|
Tachibana M. The Immunosuppressive Function of Myeloid-derived Suppressor Cells Is Regulated by the HMGB1-TLR4 Axis. YAKUGAKU ZASSHI 2018; 138:143-148. [DOI: 10.1248/yakushi.17-00158] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Masashi Tachibana
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University
- Global Center for Medical Engineering and Informatics, Osaka University
| |
Collapse
|
50
|
Oh J, Wu N, Barczak AJ, Barbeau R, Erle DJ, Shin JS. CD40 Mediates Maturation of Thymic Dendritic Cells Driven by Self-Reactive CD4 + Thymocytes and Supports Development of Natural Regulatory T Cells. THE JOURNAL OF IMMUNOLOGY 2018; 200:1399-1412. [PMID: 29321275 DOI: 10.4049/jimmunol.1700768] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 12/13/2017] [Indexed: 01/22/2023]
Abstract
Thymic dendritic cells (tDCs) play an important role in central tolerance by eliminating self-reactive thymocytes or differentiating them to regulatory T (Treg) cells. However, the molecular and cellular mechanisms underlying these functions are not completely understood. We found that mouse tDCs undergo maturation following cognate interaction with self-reactive CD4+ thymocytes and that this maturation is dependent on CD40 signaling. Ablation of CD40 expression in tDCs resulted in a significant reduction in the number of Treg cells in association with a significant reduction in the number of mature tDCs. In addition, CD40-deficient DCs failed to fully mature upon cognate interaction with CD4+ thymocytes in vitro and failed to differentiate them into Treg cells to a sufficient number. These findings suggest that tDCs mature and potentiate Treg cell development in feedback response to self-reactive CD4+ thymocytes.
Collapse
Affiliation(s)
- Jaehak Oh
- Department of Microbiology and Immunology, Sandler Asthma Basic Research Center, University of California San Francisco, San Francisco, CA 94143; and
| | - Nan Wu
- Department of Microbiology and Immunology, Sandler Asthma Basic Research Center, University of California San Francisco, San Francisco, CA 94143; and
| | - Andrea J Barczak
- Department of Medicine, Sandler Asthma Basic Research Center, University of California San Francisco, San Francisco, CA 94143
| | - Rebecca Barbeau
- Department of Medicine, Sandler Asthma Basic Research Center, University of California San Francisco, San Francisco, CA 94143
| | - David J Erle
- Department of Medicine, Sandler Asthma Basic Research Center, University of California San Francisco, San Francisco, CA 94143
| | - Jeoung-Sook Shin
- Department of Microbiology and Immunology, Sandler Asthma Basic Research Center, University of California San Francisco, San Francisco, CA 94143; and
| |
Collapse
|