1
|
Gilboa E, Gupta V, Muharemagic D, Ham S, Stelekati E, Clark E. KLF2 inhibition expands tumor-resident T cells and enhances tumor immunity. RESEARCH SQUARE 2025:rs.3.rs-5966555. [PMID: 40162209 PMCID: PMC11952643 DOI: 10.21203/rs.3.rs-5966555/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Tissue resident memory CD8+ T cells (Trm) constitute a distinct population of non-circulating memory T cells1-5 vastly exceeding the number of circulating T cells5, and play a pivotal role in protective immunity against pathogens6-8. How to promote the generation of vaccine specific Trm remains an important challenge. Whether Trm contribute also to immune control of tumors or just correlate with an unrelated process linked to clinical outcome has not been unequivocally established9,10, and phenotypic markers such as co-expression of CD69 and CD103 or CD49a integrins commonly used to monitor tumor infiltrating Trm do not unambiguously define this subset. Here we tested the hypothesis that transient downregulation of KLF2, the most conserved feature of Trm ontogeny4,11,12, will promote the differentiation of vaccine activated CD8+ T cells into Trm and enhance antitumor immunity. We show that 4-1BB antibody targeted delivery of a KLF2 siRNA to tumor bearing mice led to the downregulation of KLF2 in vaccine activated CD8+ T cells and the accumulation of phenotypically defined intratumoral CD69+CD103+ and CD69+CD49a+ CD8+ T cells which correlated with enhanced control of tumor growth. This study could serve as the foundation of a broadly applicable and clinically useful way to promote the generation of vaccine specific Trm and provides direct evidence that intratumoral CD8+CD69+CD103+ and CD8+CD69+CD49a+ cells are indeed Trm and that Trm contribute to tumor immunity.
Collapse
|
2
|
Lauver MD, Katz ZE, Markus H, Derosia NM, Jin G, Ayers KN, Butic AB, Bushey K, Abendroth CS, Liu DJ, Lukacher AE. The CXCR6-CXCL16 axis mediates T cell control of polyomavirus infection in the kidney. PLoS Pathog 2025; 21:e1012969. [PMID: 40043065 PMCID: PMC11922244 DOI: 10.1371/journal.ppat.1012969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/19/2025] [Accepted: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
BK polyomavirus (PyV) establishes lifelong asymptomatic infections in the reno-urinary system of most humans. BKPyV-associated nephropathy is the leading infectious cause of kidney allograft loss. Using mouse PyV, a natural murine pathogen that also persists in the kidney, we define a dominant chemokine receptor-chemokine axis that directs T cell infiltration of the kidney. We found that CXCR6 was required for CD4+ and CD8+ T cells to be recruited to and retained in the kidney, respectively. Absence of CXCR6 impaired virus control in the kidney. The soluble form of CXCL16 was increased in kidneys of infected mice and in vivo CXCL16 neutralization reduced numbers of virus-specific CD8+ T cells infiltrating the kidney. In vivo administration of IL-12 upregulated CXCR6 expression on virus-specific CD8+ T cells, improved T cell recruitment to the infected kidney, and reduced virus levels. Notably, T cells in kidney biopsies from PyV-associated nephropathy patients express CXCR6 and transcriptional analysis shows significant upregulation of CXCR6 and CXCL16. These findings demonstrate the importance of the CXCR6-CXCL16 axis in regulating T cell responses in the kidney to PyV infection.
Collapse
MESH Headings
- Animals
- Chemokine CXCL16
- Receptors, CXCR6
- Mice
- Polyomavirus Infections/immunology
- Polyomavirus Infections/metabolism
- Polyomavirus Infections/pathology
- Polyomavirus Infections/virology
- CD8-Positive T-Lymphocytes/immunology
- Humans
- Kidney/immunology
- Kidney/virology
- Kidney/pathology
- Mice, Inbred C57BL
- Chemokines, CXC/immunology
- Chemokines, CXC/metabolism
- Chemokines, CXC/genetics
- Receptors, Scavenger/immunology
- Receptors, Scavenger/metabolism
- Receptors, Scavenger/genetics
- CD4-Positive T-Lymphocytes/immunology
- Tumor Virus Infections/immunology
- Mice, Knockout
- BK Virus/immunology
- Receptors, Virus/immunology
- Receptors, Virus/metabolism
- Receptors, Virus/genetics
- Receptors, Chemokine/immunology
- Receptors, Chemokine/metabolism
- Receptors, Chemokine/genetics
- Kidney Diseases/immunology
- Kidney Diseases/virology
Collapse
Affiliation(s)
- Matthew D. Lauver
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Zoe E. Katz
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Havell Markus
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Nicole M. Derosia
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Ge Jin
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Katelyn N. Ayers
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Arrienne B. Butic
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Kaitlyn Bushey
- Bio X Cell, Inc., Lebanon, New Hampshire, United States of America
| | - Catherine S. Abendroth
- Department of Pathology, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States of America
| | - Dajiang J. Liu
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Aron E. Lukacher
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| |
Collapse
|
3
|
Shafer AM, Kenna E, Golden LAF, Elhossiny AM, Perry KD, Wilkowski J, Yan W, Kaczkofsky B, McGue J, Bresler SC, Courtney AH, Dalman JM, Galban CJ, Jiang W, Espinoza CE, Chugh R, Iyer MK, Frankel TL, Pasca di Magliano M, Dlugosz AA, Angeles CV. An immunocompetent mouse model of liposarcoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.31.634916. [PMID: 40297505 PMCID: PMC12036434 DOI: 10.1101/2025.01.31.634916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Liposarcoma (LPS) is the most prevalent soft tissue sarcoma. The most common biological subtypes are well-differentiated (WDLPS), a low-grade disease that can evolve to high-grade dedifferentiated LPS (DDLPS), with increased rates of recurrence and metastasis and low response rates to chemotherapy and targeted therapies. Preclinical testing of immunotherapeutics for LPS has been held back by the lack of an immunocompetent mouse model. Here, we present a spontaneous immunocompetent LPS mouse model, ACPP, with targeted deletion of Trp53 and Pten in adipocytes to mimic signaling alterations observed in human LPS. Similar to human LPS, tumors arising in ACPP mice produce WDLPS and DDLPS, along with tumors that exhibit both WD and DD components. Murine and human DDLPS tumors possess transcriptional similarities, including increased expression of oncogenes Cdk4 and Hmga2 and reduced expression of the tumor suppressor Cebpa; further, both mouse and human DDLPS exhibit either high or low T cell infiltration. Syngeneic cell lines derived from spontaneous ACPP DDLPS reliably produce tumors following orthotopic injection, each with distinct growth patterns, aggressiveness and tumor infiltrating lymphocyte profiles. These models provide much needed tools to understand the complex immunobiology of LPS and greatly accelerate the pace of preclinical studies to uncover new therapies for patients with this aggressive malignancy.
Collapse
|
4
|
Li J, Xiao C, Li C, He J. Tissue-resident immune cells: from defining characteristics to roles in diseases. Signal Transduct Target Ther 2025; 10:12. [PMID: 39820040 PMCID: PMC11755756 DOI: 10.1038/s41392-024-02050-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/28/2024] [Accepted: 11/04/2024] [Indexed: 01/19/2025] Open
Abstract
Tissue-resident immune cells (TRICs) are a highly heterogeneous and plastic subpopulation of immune cells that reside in lymphoid or peripheral tissues without recirculation. These cells are endowed with notably distinct capabilities, setting them apart from their circulating leukocyte counterparts. Many studies demonstrate their complex roles in both health and disease, involving the regulation of homeostasis, protection, and destruction. The advancement of tissue-resolution technologies, such as single-cell sequencing and spatiotemporal omics, provides deeper insights into the cell morphology, characteristic markers, and dynamic transcriptional profiles of TRICs. Currently, the reported TRIC population includes tissue-resident T cells, tissue-resident memory B (BRM) cells, tissue-resident innate lymphocytes, tissue-resident macrophages, tissue-resident neutrophils (TRNs), and tissue-resident mast cells, but unignorably the existence of TRNs is controversial. Previous studies focus on one of them in specific tissues or diseases, however, the origins, developmental trajectories, and intercellular cross-talks of every TRIC type are not fully summarized. In addition, a systemic overview of TRICs in disease progression and the development of parallel therapeutic strategies is lacking. Here, we describe the development and function characteristics of all TRIC types and their major roles in health and diseases. We shed light on how to harness TRICs to offer new therapeutic targets and present burning questions in this field.
Collapse
Affiliation(s)
- Jia Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
5
|
Xie D, Lu G, Mai G, Guo Q, Xu G. Tissue-resident memory T cells in diseases and therapeutic strategies. MedComm (Beijing) 2025; 6:e70053. [PMID: 39802636 PMCID: PMC11725047 DOI: 10.1002/mco2.70053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
Tissue-resident memory T (TRM) cells are crucial components of the immune system that provide rapid, localized responses to recurrent pathogens at mucosal and epithelial barriers. Unlike circulating memory T cells, TRM cells are located within peripheral tissues, and they play vital roles in antiviral, antibacterial, and antitumor immunity. Their unique retention and activation mechanisms, including interactions with local epithelial cells and the expression of adhesion molecules, enable their persistence and immediate functionality in diverse tissues. Recent advances have revealed their important roles in chronic inflammation, autoimmunity, and cancer, illuminating both their protective and their pathogenic potential. This review synthesizes current knowledge on TRM cells' molecular signatures, maintenance pathways, and functional dynamics across different tissues. We also explore the interactions of TRM cells with other immune cells, such as B cells, macrophages, and dendritic cells, highlighting the complex network that underpins the efficacy of TRM cells in immune surveillance and response. Understanding the nuanced regulation of TRM cells is essential for developing targeted therapeutic strategies, including vaccines and immunotherapies, to enhance their protective roles while mitigating adverse effects. Insights into TRM cells' biology hold promise for innovative treatments for infectious diseases, cancer, and autoimmune conditions.
Collapse
Affiliation(s)
- Daoyuan Xie
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Guanting Lu
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Gang Mai
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Qiuyan Guo
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di Herbs, Artemisinin Research Center, Institute of Chinese Materia MedicaAcademy of Chinese Medical SciencesBeijingChina
| | - Guofeng Xu
- Inflammation & Allergic Diseases Research UnitThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| |
Collapse
|
6
|
Fritsch N, Aparicio-Soto M, Curato C, Riedel F, Thierse HJ, Luch A, Siewert K. Chemical-Specific T Cell Tests Aim to Bridge a Gap in Skin Sensitization Evaluation. TOXICS 2024; 12:802. [PMID: 39590982 PMCID: PMC11598016 DOI: 10.3390/toxics12110802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/30/2024] [Accepted: 11/02/2024] [Indexed: 11/28/2024]
Abstract
T cell activation is the final key event (KE4) in the adverse outcome pathway (AOP) of skin sensitization. However, validated new approach methodologies (NAMs) for evaluating this step are missing. Accordingly, chemicals that activate an unusually high frequency of T cells, as does the most prevalent metal allergen nickel, are not yet identified in a regulatory context. T cell reactivity to chemical sensitizers might be especially relevant in real-life scenarios, where skin injury, co-exposure to irritants in chemical mixtures, or infections may trigger the heterologous innate immune stimulation necessary to induce adaptive T cell responses. Additionally, cross-reactivity, which underlies cross-allergies, can only be assessed by T cell tests. To date, several experimental T cell tests are available that use primary naïve and memory CD4+ and CD8+ T cells from human blood. These include priming and lymphocyte proliferation tests and, most recently, activation-induced marker (AIM) assays. All approaches are challenged by chemical-mediated toxicity, inefficient or unknown generation of T cell epitopes, and a low throughput. Here, we summarize solutions and strategies to confirm in vitro T cell signals. Broader application and standardization are necessary to possibly define chemical applicability domains and to strengthen the role of T cell tests in regulatory risk assessment.
Collapse
Affiliation(s)
- Nele Fritsch
- German Federal Institute for Risk Assessment (BfR), Department of Chemical and Product Safety, Dermatotoxicology Study Centre, 10589 Berlin, Germany; (N.F.); (C.C.); (F.R.)
- Institute of Biotechnology, Technical University of Berlin, 10115 Berlin, Germany
| | - Marina Aparicio-Soto
- German Federal Institute for Risk Assessment (BfR), Department of Chemical and Product Safety, Dermatotoxicology Study Centre, 10589 Berlin, Germany; (N.F.); (C.C.); (F.R.)
| | - Caterina Curato
- German Federal Institute for Risk Assessment (BfR), Department of Chemical and Product Safety, Dermatotoxicology Study Centre, 10589 Berlin, Germany; (N.F.); (C.C.); (F.R.)
| | - Franziska Riedel
- German Federal Institute for Risk Assessment (BfR), Department of Chemical and Product Safety, Dermatotoxicology Study Centre, 10589 Berlin, Germany; (N.F.); (C.C.); (F.R.)
| | - Hermann-Josef Thierse
- German Federal Institute for Risk Assessment (BfR), Department of Chemical and Product Safety, Dermatotoxicology Study Centre, 10589 Berlin, Germany; (N.F.); (C.C.); (F.R.)
| | - Andreas Luch
- German Federal Institute for Risk Assessment (BfR), Department of Chemical and Product Safety, Dermatotoxicology Study Centre, 10589 Berlin, Germany; (N.F.); (C.C.); (F.R.)
- Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Katherina Siewert
- German Federal Institute for Risk Assessment (BfR), Department of Chemical and Product Safety, Dermatotoxicology Study Centre, 10589 Berlin, Germany; (N.F.); (C.C.); (F.R.)
| |
Collapse
|
7
|
Iijima N. The emerging role of effector functions exerted by tissue-resident memory T cells. OXFORD OPEN IMMUNOLOGY 2024; 5:iqae006. [PMID: 39193473 PMCID: PMC11213632 DOI: 10.1093/oxfimm/iqae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/14/2024] [Accepted: 06/04/2024] [Indexed: 08/29/2024] Open
Abstract
The magnitude of the effector functions of memory T cells determines the consequences of the protection against invading pathogens and tumor development or the pathogenesis of autoimmune and allergic diseases. Tissue-resident memory T cells (TRM cells) are unique T-cell populations that persist in tissues for long periods awaiting re-encounter with their cognate antigen. Although TRM cell reactivation primarily requires the presentation of cognate antigens, recent evidence has shown that, in addition to the conventional concept, TRM cells can be reactivated without the presentation of cognate antigens. Non-cognate TRM cell activation is triggered by cross-reactive antigens or by several combinations of cytokines, including interleukin (IL)-2, IL-7, IL-12, IL-15 and IL-18. The activation mode of TRM cells reinforces their cytotoxic activity and promotes the secretion of effector cytokines (such as interferon-gamma and tumor necrosis factor-alpha). This review highlights the key features of TRM cell maintenance and reactivation and discusses the importance of effector functions that TRM cells exert upon being presented with cognate and/or non-cognate antigens, as well as cytokines secreted by TRM and non-TRM cells within the tissue microenvironment.
Collapse
Affiliation(s)
- Norifumi Iijima
- Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition (NIBN), Ibaraki, Osaka, Japan
| |
Collapse
|
8
|
Hsu J, Kim S, Anandasabapathy N. Vaccinia Virus: Mechanisms Supporting Immune Evasion and Successful Long-Term Protective Immunity. Viruses 2024; 16:870. [PMID: 38932162 PMCID: PMC11209207 DOI: 10.3390/v16060870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/13/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Vaccinia virus is the most successful vaccine in human history and functions as a protective vaccine against smallpox and monkeypox, highlighting the importance of ongoing research into vaccinia due to its genetic similarity to other emergent poxviruses. Moreover, vaccinia's ability to accommodate large genetic insertions makes it promising for vaccine development and potential therapeutic applications, such as oncolytic agents. Thus, understanding how superior immunity is generated by vaccinia is crucial for designing other effective and safe vaccine strategies. During vaccinia inoculation by scarification, the skin serves as a primary site for the virus-host interaction, with various cell types playing distinct roles. During this process, hematopoietic cells undergo abortive infections, while non-hematopoietic cells support the full viral life cycle. This differential permissiveness to viral replication influences subsequent innate and adaptive immune responses. Dendritic cells (DCs), key immune sentinels in peripheral tissues such as skin, are pivotal in generating T cell memory during vaccinia immunization. DCs residing in the skin capture viral antigens and migrate to the draining lymph nodes (dLN), where they undergo maturation and present processed antigens to T cells. Notably, CD8+ T cells are particularly significant in viral clearance and the establishment of long-term protective immunity. Here, we will discuss vaccinia virus, its continued relevance to public health, and viral strategies permissive to immune escape. We will also discuss key events and populations leading to long-term protective immunity and remaining key gaps.
Collapse
Affiliation(s)
- Joy Hsu
- Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
- Department of Dermatology, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10021, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
- Englander Institute of Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Suyon Kim
- Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA;
| | - Niroshana Anandasabapathy
- Department of Dermatology, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10021, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
- Englander Institute of Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
9
|
G Popova P, Chen SP, Liao S, Sadarangani M, Blakney AK. Clinical perspective on topical vaccination strategies. Adv Drug Deliv Rev 2024; 208:115292. [PMID: 38522725 DOI: 10.1016/j.addr.2024.115292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/01/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024]
Abstract
Vaccination is one of the most successful measures in modern medicine to combat diseases, especially infectious diseases, and saves millions of lives every year. Vaccine design and development remains critical and involves many aspects, including the choice of platform, antigen, adjuvant, and route of administration. Topical vaccination, defined herein as the introduction of a vaccine to any of the three layers of the human skin, has attracted interest in recent years as an alternative vaccination approach to the conventional intramuscular administration because of its potential to be needle-free and induce a superior immune response against pathogens. In this review, we describe recent progress in developing topical vaccines, highlight progress in the development of delivery technologies for topical vaccines, discuss potential factors that might impact the topical vaccine efficacy, and provide an overview of the current clinical landscape of topical vaccines.
Collapse
Affiliation(s)
- Petya G Popova
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, British Columbia V6T 2B9, Canada; Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, British Columbia V6T 1Z4, Canada
| | - Sunny P Chen
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, British Columbia V6T 2B9, Canada; Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, British Columbia V6T 1Z4, Canada
| | - Suiyang Liao
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, British Columbia V6T 2B9, Canada; Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, British Columbia V6T 1Z4, Canada; Life Science Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia V6T 1Z3, Canada
| | - Manish Sadarangani
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, 950 West 28th Ave, Vancouver, British Columbia V5Z 4H4, Canada; Department of Pediatrics, University of British Columbia, 4480 Oak St, Vancouver, BC V6H 0B3, Canada
| | - Anna K Blakney
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, British Columbia V6T 2B9, Canada; Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, British Columbia V6T 1Z4, Canada.
| |
Collapse
|
10
|
Pei L, Hickman HD. T Cell Surveillance during Cutaneous Viral Infections. Viruses 2024; 16:679. [PMID: 38793562 PMCID: PMC11126121 DOI: 10.3390/v16050679] [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: 03/28/2024] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 05/26/2024] Open
Abstract
The skin is a complex tissue that provides a strong physical barrier against invading pathogens. Despite this, many viruses can access the skin and successfully replicate in either the epidermal keratinocytes or dermal immune cells. In this review, we provide an overview of the antiviral T cell biology responding to cutaneous viral infections and how these responses differ depending on the cellular targets of infection. Much of our mechanistic understanding of T cell surveillance of cutaneous infection has been gained from murine models of poxvirus and herpesvirus infection. However, we also discuss other viral infections, including flaviviruses and papillomaviruses, in which the cutaneous T cell response has been less extensively studied. In addition to the mechanisms of successful T cell control of cutaneous viral infection, we highlight knowledge gaps and future directions with possible impact on human health.
Collapse
Affiliation(s)
| | - Heather D. Hickman
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA;
| |
Collapse
|
11
|
Sheikhlary S, Lopez DH, Moghimi S, Sun B. Recent Findings on Therapeutic Cancer Vaccines: An Updated Review. Biomolecules 2024; 14:503. [PMID: 38672519 PMCID: PMC11048403 DOI: 10.3390/biom14040503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/06/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Cancer remains one of the global leading causes of death and various vaccines have been developed over the years against it, including cell-based, nucleic acid-based, and viral-based cancer vaccines. Although many vaccines have been effective in in vivo and clinical studies and some have been FDA-approved, there are major limitations to overcome: (1) developing one universal vaccine for a specific cancer is difficult, as tumors with different antigens are different for different individuals, (2) the tumor antigens may be similar to the body's own antigens, and (3) there is the possibility of cancer recurrence. Therefore, developing personalized cancer vaccines with the ability to distinguish between the tumor and the body's antigens is indispensable. This paper provides a comprehensive review of different types of cancer vaccines and highlights important factors necessary for developing efficient cancer vaccines. Moreover, the application of other technologies in cancer therapy is discussed. Finally, several insights and conclusions are presented, such as the possibility of using cold plasma and cancer stem cells in developing future cancer vaccines, to tackle the major limitations in the cancer vaccine developmental process.
Collapse
Affiliation(s)
- Sara Sheikhlary
- Department of Biomedical Engineering, College of Engineering, The University of Arizona, Tucson, AZ 85721, USA
| | - David Humberto Lopez
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| | - Sophia Moghimi
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| | - Bo Sun
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| |
Collapse
|
12
|
Reider IE, Lin E, Krouse TE, Parekh NJ, Nelson AM, Norbury CC. γδ T Cells Mediate a Requisite Portion of a Wound Healing Response Triggered by Cutaneous Poxvirus Infection. Viruses 2024; 16:425. [PMID: 38543790 PMCID: PMC10975054 DOI: 10.3390/v16030425] [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: 02/06/2024] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 04/01/2024] Open
Abstract
Infection at barrier sites, e.g., skin, activates local immune defenses that limit pathogen spread, while preserving tissue integrity. Phenotypically distinct γδ T cell populations reside in skin, where they shape immunity to cutaneous infection prior to onset of an adaptive immune response by conventional αβ CD4+ (TCD4+) and CD8+ (TCD8+) T cells. To examine the mechanisms used by γδ T cells to control cutaneous virus replication and tissue pathology, we examined γδ T cells after infection with vaccinia virus (VACV). Resident γδ T cells expanded and combined with recruited γδ T cells to control pathology after VACV infection. However, γδ T cells did not play a role in control of local virus replication or blockade of systemic virus spread. We identified a unique wound healing signature that has features common to, but also features that antagonize, the sterile cutaneous wound healing response. Tissue repair generally occurs after clearance of a pathogen, but viral wound healing started prior to the peak of virus replication in the skin. γδ T cells contributed to wound healing through induction of multiple cytokines/growth factors required for efficient wound closure. Therefore, γδ T cells modulate the wound healing response following cutaneous virus infection, maintaining skin barrier function to prevent secondary bacterial infection.
Collapse
Affiliation(s)
- Irene E. Reider
- Department of Microbiology & Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Eugene Lin
- Department of Microbiology & Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Tracy E. Krouse
- Department of Microbiology & Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Nikhil J. Parekh
- Department of Microbiology & Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Amanda M. Nelson
- Department of Dermatology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Christopher C. Norbury
- Department of Microbiology & Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
13
|
Li M, Yang L, Wang C, Cui M, Wen Z, Liao Z, Han Z, Zhao Y, Lang B, Chen H, Qian J, Shu Y, Zeng X, Sun C. Rapid Induction of Long-Lasting Systemic and Mucosal Immunity via Thermostable Microneedle-Mediated Chitosan Oligosaccharide-Encapsulated DNA Nanoparticles. ACS NANO 2023; 17:24200-24217. [PMID: 37991848 DOI: 10.1021/acsnano.3c09521] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Most existing vaccines, delivered by intramuscular injection (IM), are typically associated with stringent storage requirements under cold-chain distribution and professional administration by medical personnel and often result in the induction of weak mucosal immunity. In this context, we reported a microneedle (MN) patch to deliver chitosan oligosaccharide (COS)-encapsulated DNA vaccines (DNA@COS) encoding spike and nucleocapsid proteins of SARS-CoV-2 as a vaccination technology. Compared with IM immunization, intradermal administration via the MN-mediated DNA vaccine effectively induces a comparable level of neutralizing antibody against SARS-CoV-2 variants. Surprisingly, we found that MN-mediated intradermal immunization elicited superior systemic and mucosal T cell immunity with enhanced magnitude, polyfunctionality, and persistence. Importantly, the DNA@COS nanoparticle vaccine loaded in an MN patch can be stored at room temperature for at least 1 month without a significant decrease of its immunogenicity. Mechanically, our strategy enhanced dendritic cell maturation and antiviral immunity by activating the cGAS-STING-mediated IFN signaling pathway. In conclusion, this work provides valuable insights for the rapid development of an easy-to-administer and thermostable technology for mucosal vaccines.
Collapse
Affiliation(s)
- Minchao Li
- School of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, China
| | - Li Yang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Congcong Wang
- School of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, China
| | - Mingting Cui
- School of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, China
| | - Ziyu Wen
- School of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, China
| | - Zhiheng Liao
- School of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, China
| | - Zirong Han
- School of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, China
| | - Yangguo Zhao
- School of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, China
| | - Bing Lang
- School of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, China
| | - Hongzhong Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Jun Qian
- School of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, China
| | - Yuelong Shu
- School of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, China
- Key Laboratory of Pathogen Infection Prevention and Control (MOE), State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 102629, P.R. China
| | - Xiaowei Zeng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Caijun Sun
- School of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen university), Ministry of Education, Guangzhou 514400, China
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University; Guangdong 518107, China
| |
Collapse
|
14
|
Edwards C, Shah SA, Gebhardt T, Jewell CM. Exploiting Unique Features of Microneedles to Modulate Immunity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2302410. [PMID: 37380199 PMCID: PMC10753036 DOI: 10.1002/adma.202302410] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/01/2023] [Indexed: 06/30/2023]
Abstract
Microneedle arrays (MNAs) are small patches containing hundreds of short projections that deliver signals directly to dermal layers without causing pain. These technologies are of special interest for immunotherapy and vaccine delivery because they directly target immune cells concentrated in the skin. The targeting abilities of MNAs result in efficient immune responses-often more protective or therapeutic-compared to conventional needle delivery. MNAs also offer logistical benefits, such as self-administration and transportation without refrigeration. Thus, numerous preclinical and clinical studies are exploring these technologies. Here the unique advantages of MNA, as well as critical challenges-such as manufacturing and sterility issues-the field faces to enable widespread deployment are discussed. How MNA design parameters can be exploited for controlled release of vaccines and immunotherapies, and the application to preclinical models of infection, cancer, autoimmunity, and allergies are explained. Specific strategies are also discussed to reduce off-target effects compared to conventional vaccine delivery routes, and novel chemical and manufacturing controls that enable cargo stability in MNAs across flexible intervals and temperatures. Clinical research using MNAs is then examined. Drawbacks of MNAs and the implications, and emerging opportunities to exploit MNAs for immune engineering and clinical use are concluded.
Collapse
Affiliation(s)
- Camilla Edwards
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Shrey A Shah
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Thomas Gebhardt
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, VIC, 3000, Australia
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
- US Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD, 21201, USA
- Robert E. Fischell Institute for Biomedical Devices, College Park, MD, 20742, USA
- Department of Microbiology and Immunology, University of Maryland Medical School, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, 21201, USA
| |
Collapse
|
15
|
Smith NP, Yan Y, Pan Y, Williams JB, Manakongtreecheep K, Pant S, Zhao J, Tian T, Pan T, Stingley C, Wu K, Zhang J, Kley AL, Sorger PK, Villani AC, Kupper TS. Resident memory T cell development is associated with AP-1 transcription factor upregulation across anatomical niches. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.29.560006. [PMID: 37873428 PMCID: PMC10592877 DOI: 10.1101/2023.09.29.560006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Tissue-resident memory T (T RM ) cells play a central role in immune responses to pathogens across all barrier tissues after infection. However, the underlying mechanisms that drive T RM differentiation and priming for their recall effector function remains unclear. In this study, we leveraged both newly generated and publicly available single-cell RNA-sequencing (scRNAseq) data generated across 10 developmental time points to define features of CD8 T RM across both skin and small-intestine intraepithelial lymphocytes (siIEL). We employed linear modeling to capture temporally-associated gene programs that increase their expression levels in T cell subsets transitioning from an effector to a memory T cell state. In addition to capturing tissue-specific gene programs, we defined a consensus T RM signature of 60 genes across skin and siIEL that can effectively distinguish T RM from circulating T cell populations, providing a more specific T RM signature than what was previously generated by comparing bulk T RM to naïve or non-tissue resident memory populations. This updated T RM signature included the AP-1 transcription factor family members Fos, Fosb and Fosl2 . Moreover, ATACseq analysis detected an enrichment of AP-1-specific motifs at open chromatin sites in mature T RM . CyCIF tissue imaging detected nuclear co-localization of AP-1 members Fosb and Junb in resting CD8 T RM >100 days post-infection. Taken together, these results reveal a critical role of AP-1 transcription factor members in T RM biology and suggests a novel mechanism for rapid reactivation of resting T RM in tissue upon antigen encounter.
Collapse
|
16
|
Rotrosen E, Kupper TS. Assessing the generation of tissue resident memory T cells by vaccines. Nat Rev Immunol 2023; 23:655-665. [PMID: 37002288 PMCID: PMC10064963 DOI: 10.1038/s41577-023-00853-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2023] [Indexed: 04/03/2023]
Abstract
Vaccines have been a hugely successful public health intervention, virtually eliminating many once common diseases of childhood. However, they have had less success in controlling endemic pathogens including Mycobacterium tuberculosis, herpesviruses and HIV. A focus on vaccine-mediated generation of neutralizing antibodies, which has been a successful approach for some pathogens, has been complicated by the emergence of escape variants, which has been seen for pathogens such as influenza viruses and SARS-CoV-2, as well as for HIV-1. We discuss how vaccination strategies aimed at generating a broad and robust T cell response may offer superior protection against pathogens, particularly those that have been observed to mutate rapidly. In particular, we consider here how a focus on generating resident memory T cells may be uniquely effective for providing immunity to pathogens that typically infect (or become reactivated in) the skin, respiratory mucosa or other barrier tissues.
Collapse
Affiliation(s)
- Elizabeth Rotrosen
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA, USA
- Boston University School of Medicine, Boston, MA, USA
| | - Thomas S Kupper
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
17
|
Yu Y, Wang J, Wu MX. Microneedle-Mediated Immunization Promotes Lung CD8+ T-Cell Immunity. J Invest Dermatol 2023; 143:1983-1992.e3. [PMID: 37044258 PMCID: PMC10524108 DOI: 10.1016/j.jid.2023.03.1672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 03/09/2023] [Accepted: 03/23/2023] [Indexed: 04/14/2023]
Abstract
Microneedle array has proven more efficient in stimulating humoral immunity than intramuscular vaccination. However, its effectiveness in inducing pulmonary CD8+ T cells remains elusive, which is essential to the frontline defense against pulmonary viral infections such as influenza and COVID-19 viruses. The current investigation reveals that superior CD8+ T-cell responses are elicited by immunization with a microneedle array over intradermal or intramuscular immunization using the model antigen ovalbumin, irrespective of whether or not the antigen is provided in the lung. Mechanistically, microneedle array-mediated immunization targeted the epidermal layer and stimulated predominantly Langerhans cells, resulting in increased expression of α4β1 adhesion molecules on the CD8+ T-cell surface, which may play a role in T-cell homing to the lung, whereas CD8+ T cells induced by intramuscular immunization did not express the adhesion molecule sufficiently. CD8+ T cells with a lung-homing propensity were also seen after intradermal vaccination, yet to a much lesser extent. Accordingly, microneedle array immunization provided stronger protection against influenza viral infection than intradermal or intramuscular immunization. The observations offer insights into a strong cross-talk between epidermal immunization and lung immunity and are valuable for designing and delivering vaccines against respiratory viral infections.
Collapse
Affiliation(s)
- Yang Yu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Dermatology, Harvard Medical School, Boston, Massachusetts, USA
| | - Ji Wang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Dermatology, Harvard Medical School, Boston, Massachusetts, USA; The first affiliated Hospital, Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
| | - Mei X Wu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Dermatology, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
18
|
Strobl J, Haniffa M. Functional heterogeneity of human skin-resident memory T cells in health and disease. Immunol Rev 2023; 316:104-119. [PMID: 37144705 PMCID: PMC10952320 DOI: 10.1111/imr.13213] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/11/2023] [Accepted: 04/15/2023] [Indexed: 05/06/2023]
Abstract
The human skin is populated by a diverse pool of memory T cells, which can act rapidly in response to pathogens and cancer antigens. Tissue-resident memory T cells (TRM ) have been implicated in range of allergic, autoimmune and inflammatory skin diseases. Clonal expansion of cells with TRM properties is also known to contribute to cutaneous T-cell lymphoma. Here, we review the heterogeneous phenotypes, transcriptional programs, and effector functions of skin TRM . We summarize recent studies on TRM formation, longevity, plasticity, and retrograde migration and contextualize the findings to skin TRM and their role in maintaining skin homeostasis and altered functions in skin disease.
Collapse
Affiliation(s)
- Johanna Strobl
- Department of DermatologyMedical University of ViennaViennaAustria
- CeMM Research Center for Molecular MedicineViennaAustria
| | - Muzlifah Haniffa
- Wellcome Sanger InstituteCambridgeUK
- Department of Dermatology and NIHR Newcastle Biomedical Research CentreNewcastle Hospitals NHS Foundation TrustNewcastle upon TyneUK
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| |
Collapse
|
19
|
Shafaati M, Zandi M. Human monkeypox (hMPXV) re-emergence: Host immunity status and current vaccines landscape. J Med Virol 2023; 95:e28251. [PMID: 36271768 DOI: 10.1002/jmv.28251] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/28/2022] [Accepted: 10/19/2022] [Indexed: 01/11/2023]
Abstract
Monkeypox virus is a member of the Orthopoxvirus genus and the Poxviridae family. Orthopoxviruses are among the most intricate animal viruses. The pathogenicity of human monkeypox infection has been emphasized in response to its recent emergence in non-endemic countries and the threat of bioterrorism. It is always necessary to take appropriate precautions in exposure to emerging or re-emerging infections. Here, we focus on the current state of the human monkeypox infection outbreak, research & development of immune responses, and clinical interventions to prevent and treat the human monkeypox virus and other human poxviruses.
Collapse
Affiliation(s)
- Maryam Shafaati
- Department of Microbiology, Faculty of Science, Jahrom Branch, Islamic Azad University, Jahrom, Iran
- Occupational Sleep Research, Baharloo Hospital, Tehran University of Medical Science, Tehran, Iran
| | - Milad Zandi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Gao A, Zhao W, Wu R, Su R, Jin R, Luo J, Gao C, Li X, Wang C. Tissue-resident memory T cells: The key frontier in local synovitis memory of rheumatoid arthritis. J Autoimmun 2022; 133:102950. [PMID: 36356551 DOI: 10.1016/j.jaut.2022.102950] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/27/2022] [Accepted: 10/29/2022] [Indexed: 11/09/2022]
Abstract
Rheumatoid arthritis (RA) is a highly disabling, systemic autoimmune disease. It presents a remarkable tendency to recur, which renders it almost impossible for patients to live without drugs. Under such circumstances, many patients have to suffer the pain of recurrent attacks as well as the side effects of long-term medication. Current therapies for RA are primarily systemic treatments without targeting the problem that RA is more likely to recur locally. Emerging studies suggest the existence of a mechanism mediating local memory during RA, which is closely related to the persistent residence of tissue-resident memory T cells (TRM). TRM, one of the memory T cell subsets, reside in tissues providing immediate immune protection but driving recurrent local inflammation on the other hand. The heterogeneity among synovial TRM is unclear, with the dominated CD8+ TRM observed in inflamed synovium of RA patients coming into focus. Besides local arthritis relapse, TRM may also contribute to extra-articular organ involvement in RA due to their migration potential. Future integration of single-cell RNA sequencing (scRNA-seq) with spatial transcriptomics to explore the gene expression patterns of TRM in both temporal dimension and spatial dimension may help us identify specific therapeutic targets. Targeting synovial TRM to suppress local arthritis flares while using systemic therapies to prevent extra-articular organ involvement may provide a new perspective to address RA recurrence.
Collapse
Affiliation(s)
- Anqi Gao
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory for Immunomicroecology, Shanxi, China
| | - Wenpeng Zhao
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory for Immunomicroecology, Shanxi, China
| | - Ruihe Wu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory for Immunomicroecology, Shanxi, China
| | - Rui Su
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory for Immunomicroecology, Shanxi, China
| | - Ruqing Jin
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory for Immunomicroecology, Shanxi, China
| | - Jing Luo
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory for Immunomicroecology, Shanxi, China
| | - Chong Gao
- Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital/Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaofeng Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory for Immunomicroecology, Shanxi, China
| | - Caihong Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory for Immunomicroecology, Shanxi, China.
| |
Collapse
|
21
|
Chopra A, Gupta A. Skin as an immune organ and the site of biomimetic, non-invasive vaccination. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2022. [DOI: 10.1016/j.medntd.2022.100196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
22
|
Balmert SC, Ghozloujeh ZG, Carey CD, Williams LH, Zhang J, Shahi P, Amer M, Sumpter TL, Erdos G, Korkmaz E, Falo LD. A microarray patch SARS-CoV-2 vaccine induces sustained antibody responses and polyfunctional cellular immunity. iScience 2022; 25:105045. [PMID: 36062075 PMCID: PMC9425707 DOI: 10.1016/j.isci.2022.105045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 04/19/2022] [Accepted: 08/25/2022] [Indexed: 12/01/2022] Open
Abstract
Sustainable global immunization campaigns against COVID-19 and other emerging infectious diseases require effective, broadly deployable vaccines. Here, we report a dissolvable microarray patch (MAP) SARS-CoV-2 vaccine that targets the immunoresponsive skin microenvironment, enabling efficacious needle-free immunization. Multicomponent MAPs delivering both SARS-CoV-2 S1 subunit antigen and the TLR3 agonist Poly(I:C) induce robust antibody and cellular immune responses systemically and in the respiratory mucosa. MAP vaccine-induced antibodies bind S1 and the SARS-CoV-2 receptor-binding domain, efficiently neutralize the virus, and persist at high levels for more than a year. The MAP platform reduces systemic toxicity of the delivered adjuvant and maintains vaccine stability without refrigeration. When applied to human skin, MAP vaccines activate skin-derived migratory antigen-presenting cells, supporting the feasibility of human translation. Ultimately, this shelf-stable MAP vaccine improves immunogenicity and safety compared to traditional intramuscular vaccines and offers an attractive alternative for global immunization efforts against a range of infectious pathogens.
Collapse
Affiliation(s)
- Stephen C. Balmert
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | - Cara Donahue Carey
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Li’an H. Williams
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jiying Zhang
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Preeti Shahi
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Maher Amer
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Tina L. Sumpter
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Geza Erdos
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Emrullah Korkmaz
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15261, USA
| | - Louis D. Falo
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15261, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- The McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
23
|
Yang N, Garcia A, Meyer C, Tuschl T, Merghoub T, Wolchok JD, Deng L. Heat-inactivated modified vaccinia virus Ankara boosts Th1 cellular and humoral immunity as a vaccine adjuvant. NPJ Vaccines 2022; 7:120. [PMID: 36261460 PMCID: PMC9580433 DOI: 10.1038/s41541-022-00542-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 09/26/2022] [Indexed: 12/02/2022] Open
Abstract
Protein or peptide-based subunit vaccines have generated excitement and renewed interest in combating human cancer or COVID-19 outbreak. One major concern for subunit vaccine application is the weak immune responses induced by protein or peptides. Developing novel and effective vaccine adjuvants are critical for the success of subunit vaccines. Here we explored the potential of heat-inactivated MVA (heat-iMVA) as a vaccine adjuvant. Heat-iMVA dramatically enhances T cell responses and antibodies responses, mainly toward Th1 immune responses when combined with protein or peptide-based immunogen. The adjuvant effect of Heat-iMVA is stronger than live MVA and is dependent on the cGAS/STING-mediated cytosolic DNA-sensing pathway. In a therapeutic vaccination model based on tumor neoantigen peptide vaccine, Heat-iMVA significantly extended the survival and delayed tumor growth. When combined with SARS-CoV-2 spike protein, Heat-iMVA induced more robust spike-specific antibody production and more potent neutralization antibodies. Our results support that Heat-iMVA can be developed as a safe and potent vaccine adjuvant for subunit vaccines against cancer or SARS-CoV-2.
Collapse
Affiliation(s)
- Ning Yang
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Aitor Garcia
- Laboratory of RNA Molecular Biology, The Rockefeller University, New York, NY, 10065, USA
| | - Cindy Meyer
- Laboratory of RNA Molecular Biology, The Rockefeller University, New York, NY, 10065, USA
| | - Thomas Tuschl
- Laboratory of RNA Molecular Biology, The Rockefeller University, New York, NY, 10065, USA
| | - Taha Merghoub
- Immuno-oncology service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Jedd D Wolchok
- Immuno-oncology service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Liang Deng
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
- Immuno-oncology service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
24
|
Shchelkunov SN, Sergeev AA, Titova KA, Pyankov SA, Starostina E, Borgoyakova MB, Kisakova LA, Kisakov DN, Karpenko LI, Yakubitskiy SN. Comparison of the Effectiveness of Transepidemal and Intradermal Immunization of Mice with the Vacinia Virus. Acta Naturae 2022; 14:111-118. [PMID: 36694907 PMCID: PMC9844093 DOI: 10.32607/actanaturae.11857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 11/30/2022] [Indexed: 01/22/2023] Open
Abstract
The spread of the monkeypox virus infection among humans in many countries outside of Africa, which started in 2022, is now drawing the attention of the medical and scientific communities to the fact that immunization against this infection is sorely needed. According to current guidelines, immunization of people with the first-generation smallpox vaccine based on the vaccinia virus (VACV) LIVP strain, which is licensed in Russia, should be performed via transepidermal inoculation (skin scarification, s.s.). However, the long past experience of using this vaccination technique suggests that it does not ensure virus inoculation into patients' skin with enough reliability. The procedure of intradermal (i.d.) injection of a vaccine can be an alternative to s.s. inoculation. The effectiveness of i.d. vaccination can depend on the virus injection site on the body. Therefore, the aim of this study was to compare the development of the humoral and cellular immune responses in BALB/c mice immunized with the LIVP VACV strain, which was administered either by s.s. inoculation or i.d. injection into the same tail region of the animal. A virus dose of 105 pfu was used in both cases. ELISA of serum samples revealed no significant difference in the dynamics and level of production of VACV-specific IgM and IgG after i.d. or s.s. vaccination. A ELISpot analysis of splenocytes from the vaccinated mice showed that i.d. administration of VACV LIVP to mice induces a significantly greater T-cell immune response compared to s.s. inoculation. In order to assess the protective potency, on day 45 post immunization, mice were intranasally infected with lethal doses of either the cowpox virus (CPXV) or the ectromelia virus (ECTV), which is evolutionarily distant from the VACV and CPXV. Both vaccination techniques ensured complete protection of mice against infection with the CPXV. However, when mice were infected with a highly virulent strain of ECTV, 50% survived in the i.d. immunized group, whereas only 17% survived in the s.s. immunized group. It appears, therefore, that i.d. injection of the VACV can elicit a more potent protective immunity against orthopoxviruses compared to the conventional s.s. technique.
Collapse
Affiliation(s)
- S. N. Shchelkunov
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russia
| | - A. A. Sergeev
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russia
| | - K. A. Titova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russia
| | - S. A. Pyankov
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russia
| | - E.V. Starostina
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russia
| | - M. B. Borgoyakova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russia
| | - L. A. Kisakova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russia
| | - D. N. Kisakov
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russia
| | - L. I. Karpenko
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russia
| | - S. N. Yakubitskiy
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo, Novosibirsk region, 630559 Russia
| |
Collapse
|
25
|
Sakamoto A, Osawa H, Hashimoto H, Mizuno T, Hasyim AA, Abe YI, Okahashi Y, Ogawa R, Iyori M, Shida H, Yoshida S. A replication-competent smallpox vaccine LC16m8Δ-based COVID-19 vaccine. Emerg Microbes Infect 2022; 11:2359-2370. [PMID: 36069348 PMCID: PMC9527789 DOI: 10.1080/22221751.2022.2122580] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Viral vectors are a potent vaccine platform for inducing humoral and T-cell immune responses. Among the various viral vectors, replication-competent ones are less commonly used for coronavirus disease 2019 (COVID-19) vaccine development compared with replication-deficient ones. Here, we show the availability of a smallpox vaccine LC16m8Δ (m8Δ) as a replication-competent viral vector for a COVID-19 vaccine. M8Δ is a genetically stable variant of the licensed and highly effective Japanese smallpox vaccine LC16m8. Here, we generated two m8Δ recombinants: one harbouring a gene cassette encoding the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S) glycoprotein, named m8Δ-SARS2(P7.5-S)-HA; and one encoding the S protein with a highly polybasic motif at the S1/S2 cleavage site, named m8Δ-SARS2(P7.5-SHN)-HA. M8Δ-SARS2(P7.5-S)-HA induced S-specific antibodies in mice that persisted for at least six weeks after a homologous boost immunization. All eight analysed serum samples displayed neutralizing activity against an S-pseudotyped virus at a level similar to that of serum samples from patients with COVID-19, and more than half (5/8) also had neutralizing activity against the Delta/B.1.617.2 variant of concern. Importantly, most serum samples also neutralized the infectious SARS-CoV-2 Wuhan and Delta/B.1.617.2 strains. In contrast, immunization with m8Δ-SARS2(P7.5-SHN)-HA elicited significantly lower antibody titres, and the induced antibodies had less neutralizing activity. Regarding T-cell immunity, both m8Δ recombinants elicited S-specific multifunctional CD8+ and CD4+ T-cell responses even after just a primary immunization. Thus, m8Δ provides an alternative method for developing a novel COVID-19 vaccine.
Collapse
Affiliation(s)
- Akihiko Sakamoto
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Ishikawa, Japan
| | - Hiroaki Osawa
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Ishikawa, Japan
| | - Hinata Hashimoto
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Ishikawa, Japan
| | - Tetsushi Mizuno
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Ishikawa, Japan.,Department of Global Infectious Diseases, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Ammar A Hasyim
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Ishikawa, Japan
| | - Yu-Ichi Abe
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Ishikawa, Japan
| | - Yuto Okahashi
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Ishikawa, Japan
| | - Ryohei Ogawa
- Department of Radiology, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - Mitsuhiro Iyori
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Ishikawa, Japan
| | - Hisatoshi Shida
- Division of Molecular Virology, Institute of Immunological Science, Hokkaido University, Sapporo, Japan
| | - Shigeto Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Ishikawa, Japan
| |
Collapse
|
26
|
Yenyuwadee S, Sanchez-Trincado Lopez JL, Shah R, Rosato PC, Boussiotis VA. The evolving role of tissue-resident memory T cells in infections and cancer. SCIENCE ADVANCES 2022; 8:eabo5871. [PMID: 35977028 PMCID: PMC9385156 DOI: 10.1126/sciadv.abo5871] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 07/05/2022] [Indexed: 06/12/2023]
Abstract
Resident memory T cells (TRM) form a distinct type of T memory cells that stably resides in tissues. TRM form an integral part of the immune sensing network and have the ability to control local immune homeostasis and participate in immune responses mediated by pathogens, cancer, and possibly autoantigens during autoimmunity. TRM express residence gene signatures, functional properties of both memory and effector cells, and remarkable plasticity. TRM have a well-established role in pathogen immunity, whereas their role in antitumor immune responses and immunotherapy is currently evolving. As TRM form the most abundant T memory cell population in nonlymphoid tissues, they are attractive targets for therapeutic exploitation. Here, we provide a concise review of the development and physiological role of CD8+ TRM, their involvement in diseases, and their potential therapeutic exploitation.
Collapse
Affiliation(s)
- Sasitorn Yenyuwadee
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Department of Dermatology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jose Luis Sanchez-Trincado Lopez
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Laboratory of Immunomedicine, School of Medicine, Complutense University of Madrid, Ave Complutense S/N, 28040 Madrid, Spain
| | - Rushil Shah
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Cornell University, Ithaca, NY 14850 , USA
| | - Pamela C Rosato
- The Geisel School of Medicine at Dartmouth, Lebanon, NH 03755, USA
| | - Vassiliki A Boussiotis
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
27
|
Chandrasekar SS, Phanse Y, Riel M, Hildebrand RE, Hanafy M, Osorio JE, Abdelgayed SS, Talaat AM. Systemic Neutralizing Antibodies and Local Immune Responses Are Critical for the Control of SARS-CoV-2. Viruses 2022; 14:v14061262. [PMID: 35746733 PMCID: PMC9227431 DOI: 10.3390/v14061262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 02/01/2023] Open
Abstract
Antibody measurements are primarily used to evaluate experimental and approved COVID-19 vaccines, which is unilateral considering our immune responses’ complex nature. Previously, we showed that nanoparticle plasmid DNA adjuvant system, QAC, and MVA based vaccines were immunogenic against SARS-CoV-2. Here, we report on the protective efficacy of systemic humoral and mucosal cell-mediated immune responses in transgenic mice models against SARS-CoV-2 following nanoparticle immunization. Parenteral, intramuscular administration of QAC-based plasmid DNA vaccine-encoding SARS-CoV-2 S and N led to the induction of significant serum neutralizing humoral responses, which reduced viral burden in the lungs and prevented viral dissemination to the brain. In contrast, the mucosal, intranasal administration of a heterologous vaccine elicited significant mucosal cell-mediated immune responses in the lungs that limited lung viral replication. The presented results demonstrate that serum neutralizing humoral and local lung T-cell immune responses are critical for the control of SARS-CoV-2 replication.
Collapse
Affiliation(s)
- Shaswath S. Chandrasekar
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (M.R.); (R.E.H.); (M.H.); (J.E.O.)
| | | | - Mariah Riel
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (M.R.); (R.E.H.); (M.H.); (J.E.O.)
| | - Rachel E. Hildebrand
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (M.R.); (R.E.H.); (M.H.); (J.E.O.)
| | - Mostafa Hanafy
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (M.R.); (R.E.H.); (M.H.); (J.E.O.)
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Jorge E. Osorio
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (M.R.); (R.E.H.); (M.H.); (J.E.O.)
- Colombia Wisconsin One Health Consortium, Universidad Nacional Medellín, Calle 75#79a 5, Colombia
| | - Sherein S. Abdelgayed
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt;
| | - Adel M. Talaat
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (M.R.); (R.E.H.); (M.H.); (J.E.O.)
- Pan Genome Systems, Madison, WI 53719, USA;
- Correspondence: ; Tel.: +1-608-262-2861
| |
Collapse
|
28
|
Coffey JW, van der Burg NMD, Rananakomol T, Ng HI, Fernando GJP, Kendall MAF. An Ultrahigh‐Density Microneedle Array for Skin Vaccination: Inducing Epidermal Cell Death by Increasing Microneedle Density Enhances Total IgG and IgG1 Immune Responses. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202100151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Jacob W. Coffey
- The Delivery of Drugs and Genes Group (D2G) Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia QLD 4072 Australia
- Department of Chemical Engineering David H. Koch Institute for Integrative Cancer Research Massachusetts Institute of Technology Cambridge MA 02139 USA
- Division of Gastroenterology Brigham and Women's Hospital Harvard Medical School Boston MA 02115 USA
- Department of Microbiology and Immunology Peter Doherty Institute for Infection and Immunology University of Melbourne Melbourne VIC 3000 Australia
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology The University of Queensland St Lucia QLD 4072 Australia
| | - Nicole M. D. van der Burg
- The Delivery of Drugs and Genes Group (D2G) Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia QLD 4072 Australia
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology The University of Queensland St Lucia QLD 4072 Australia
| | - Thippayawan Rananakomol
- The Delivery of Drugs and Genes Group (D2G) Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia QLD 4072 Australia
| | - Hwee-Ing Ng
- The Delivery of Drugs and Genes Group (D2G) Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia QLD 4072 Australia
| | - Germain J. P. Fernando
- The Delivery of Drugs and Genes Group (D2G) Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia QLD 4072 Australia
- The University of Queensland School of Chemistry and Molecular Biosciences Brisbane QLD 4072 Australia
- Vaxxas Pty Translational Research Institute Woolloongabba QLD 4102 Australia
| | - Mark A. F. Kendall
- The Delivery of Drugs and Genes Group (D2G) Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia QLD 4072 Australia
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology The University of Queensland St Lucia QLD 4072 Australia
- The University of Queensland School of Chemistry and Molecular Biosciences Brisbane QLD 4072 Australia
| |
Collapse
|
29
|
Doan TA, Forward T, Tamburini BAJ. Trafficking and retention of protein antigens across systems and immune cell types. Cell Mol Life Sci 2022; 79:275. [PMID: 35505125 PMCID: PMC9063628 DOI: 10.1007/s00018-022-04303-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/01/2022] [Accepted: 04/12/2022] [Indexed: 12/05/2022]
Abstract
In response to infection or vaccination, the immune system initially responds non-specifically to the foreign insult (innate) and then develops a specific response to the foreign antigen (adaptive). The programming of the immune response is shaped by the dispersal and delivery of antigens. The antigen size, innate immune activation and location of the insult all determine how antigens are handled. In this review we outline which specific cell types are required for antigen trafficking, which processes require active compared to passive transport, the ability of specific cell types to retain antigens and the viruses (human immunodeficiency virus, influenza and Sendai virus, vesicular stomatitis virus, vaccinia virus) and pattern recognition receptor activation that can initiate antigen retention. Both where the protein antigen is localized and how long it remains are critically important in shaping protective immune responses. Therefore, understanding antigen trafficking and retention is necessary to understand the type and magnitude of the immune response and essential for the development of novel vaccine and therapeutic targets.
Collapse
Affiliation(s)
- Thu A Doan
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, USA.,Immunology Graduate Program, University of Colorado School of Medicine, Aurora, USA
| | - Tadg Forward
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, USA
| | - Beth A Jirón Tamburini
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, USA. .,Immunology Graduate Program, University of Colorado School of Medicine, Aurora, USA. .,Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
30
|
Churchill MJ, du Bois H, Heim TA, Mudianto T, Steele MM, Nolz JC, Lund AW. Infection-induced lymphatic zippering restricts fluid transport and viral dissemination from skin. J Exp Med 2022; 219:e20211830. [PMID: 35353138 PMCID: PMC8972184 DOI: 10.1084/jem.20211830] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 01/19/2022] [Accepted: 03/14/2022] [Indexed: 01/13/2023] Open
Abstract
Lymphatic vessels are often considered passive conduits that flush antigenic material, pathogens, and cells to draining lymph nodes. Recent evidence, however, suggests that lymphatic vessels actively regulate diverse processes from antigen transport to leukocyte trafficking and dietary lipid absorption. Here we tested the hypothesis that infection-induced changes in lymphatic transport actively contribute to innate host defense. We demonstrate that cutaneous vaccinia virus infection by scarification activates dermal lymphatic capillary junction tightening (zippering) and lymph node lymphangiogenesis, which are associated with reduced fluid transport and cutaneous viral sequestration. Lymphatic-specific deletion of VEGFR2 prevented infection-induced lymphatic capillary zippering, increased fluid flux out of tissue, and allowed lymphatic dissemination of virus. Further, a reduction in dendritic cell migration to lymph nodes in the absence of lymphatic VEGFR2 associated with reduced antiviral CD8+ T cell expansion. These data indicate that VEGFR2-driven lymphatic remodeling is a context-dependent, active mechanism of innate host defense that limits viral dissemination and facilitates protective, antiviral CD8+ T cell responses.
Collapse
Affiliation(s)
- Madeline J. Churchill
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| | - Haley du Bois
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY
| | - Taylor A. Heim
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY
| | - Tenny Mudianto
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY
| | - Maria M. Steele
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY
| | - Jeffrey C. Nolz
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| | - Amanda W. Lund
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY
| |
Collapse
|
31
|
Yim SG, Hwang YH, An S, Seong KY, Kim SY, Kim S, Lee H, Lee KO, Kim MY, Kim D, Kim YJ, Yang SY. Low-Temperature Multiple Micro-Dispensing on Microneedles for Accurate Transcutaneous Smallpox Vaccination. Vaccines (Basel) 2022; 10:vaccines10040561. [PMID: 35455310 PMCID: PMC9024753 DOI: 10.3390/vaccines10040561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/29/2022] [Accepted: 04/01/2022] [Indexed: 12/10/2022] Open
Abstract
Smallpox is an acute contagious disease caused by the variola virus. According to WHO guidelines, the smallpox vaccine is administrated by scarification into the epidermis using a bifurcated needle moistened with a vaccine solution. However, this invasive vaccination method involving multiple skin punctures requires a special technique to inoculate, as well as a cold chain for storage and distribution of vaccine solutions containing a live virus. Here, we report a transcutaneous smallpox vaccination using a live vaccinia-coated microneedle (MN) patch prepared by a low-temperature multiple nanoliter-level dispensing system, enabling accurate transdermal delivery of live vaccines and maintenance of bioactivity. The live vaccinia in hyaluronic acid (HA) solutions was selectively coated on the solid MN tips, and the coating amount of the vaccine was precisely controlled through a programmed multiple dispensing process with high accuracy under low temperature conditions (2–8 °C) for smallpox vaccination. Inoculation of mice (BALB/C mouse) with the MN patch coated with the second-generation smallpox vaccine increased the neutralizing antibody titer and T cell immune response. Interestingly, the live vaccine-coated MN patch maintained viral titers at −20 °C for 4 weeks and elevated temperature (37 °C) for 1 week, highlighting improved storage stability of the live virus formulated into coated MN patches. This coated MN platform using contact dispensing technique provides a simple and effective method for smallpox vaccination.
Collapse
Affiliation(s)
- Sang-Gu Yim
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea; (S.-G.Y.); (S.A.); (K.-Y.S.); (H.L.)
| | - Yun-Ho Hwang
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju 28159, Korea; (Y.-H.H.); (S.Y.K.); (M.-Y.K.); (D.K.)
| | - Seonyeong An
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea; (S.-G.Y.); (S.A.); (K.-Y.S.); (H.L.)
| | - Keum-Yong Seong
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea; (S.-G.Y.); (S.A.); (K.-Y.S.); (H.L.)
| | - Seo-Yeon Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju 28159, Korea; (Y.-H.H.); (S.Y.K.); (M.-Y.K.); (D.K.)
| | - Semin Kim
- SNVIA Co., Ltd., Hyowon Industry-Cooperation Building, Busan 46241, Korea; (S.K.); (K.-O.L.)
| | - Hyeseon Lee
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea; (S.-G.Y.); (S.A.); (K.-Y.S.); (H.L.)
| | - Kang-Oh Lee
- SNVIA Co., Ltd., Hyowon Industry-Cooperation Building, Busan 46241, Korea; (S.K.); (K.-O.L.)
| | - Mi-Young Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju 28159, Korea; (Y.-H.H.); (S.Y.K.); (M.-Y.K.); (D.K.)
| | - Dokeun Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju 28159, Korea; (Y.-H.H.); (S.Y.K.); (M.-Y.K.); (D.K.)
| | - You-Jin Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju 28159, Korea; (Y.-H.H.); (S.Y.K.); (M.-Y.K.); (D.K.)
- Correspondence: (Y.-J.K.); (S.-Y.Y.)
| | - Seung-Yun Yang
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea; (S.-G.Y.); (S.A.); (K.-Y.S.); (H.L.)
- Correspondence: (Y.-J.K.); (S.-Y.Y.)
| |
Collapse
|
32
|
Yu Y, Lian Z, Cui Y. The OH system: A panorama view of the PPV-host interaction. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2022; 98:105220. [PMID: 35066165 DOI: 10.1016/j.meegid.2022.105220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 11/19/2021] [Accepted: 01/17/2022] [Indexed: 06/14/2023]
Abstract
Poxviruses are a family of specialized cytoplasm-parasitic DNA viruses that replicate and assembly in virus factory. In Parapoxvirus (PPV) genus, with the orf virus (ORFV) as a representative species of this genus, their behaviors are significantly different from that of Orthopoxvirus, and the plots of viral practical solutions for evading host immunity are intricate and fascinating, particularly to anti-host and host's antiviral mechanisms. In order to protect the virus factory from immune elimination caused by infection, PPVs attempt to interfere with multiple stress levels of host, mainly by modulating innate immunity response (IIR) and adaptive immunity response (AIR). Given that temporarily constructed by virus infection, ORFV-HOST (OH) system accompanied by viral strategies is carefully managed in the virus factory, thus directing many life-critical events once undergoing the IIR and AIR. Evolutionarily, to reduce the risk of system destruction, ORFV have evolved into a mild-looking mode to avoid overstimulation. Moreover, the current version of development also focus on recognizing and hijacking more than eight antiviral security mechanisms of host cells, such as the 2',5'-oligoadenylate synthetase (OAS)/RNase L and PKR systems, the ubiquitin protease system (UPS), and so on. In summary, this review assessed inescapable pathways as mentioned above, through which viruses compete with their hosts strategically. The OH system provides a panoramic view and a powerful platform for us to study the PPV-Host interaction, as well as the corresponding implications on a great application potential in anti-virus design.
Collapse
Affiliation(s)
- Yongzhong Yu
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China.
| | - Zhengxing Lian
- College of Animal Science and Technology, China Agricultural University, Beijing 100039, PR China
| | - Yudong Cui
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| |
Collapse
|
33
|
CD8 T Cell Vaccines and a Cytomegalovirus-Based Vector Approach. Life (Basel) 2021; 11:life11101097. [PMID: 34685468 PMCID: PMC8538937 DOI: 10.3390/life11101097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 11/17/2022] Open
Abstract
The twentieth century witnessed a huge expansion in the number of vaccines used with great success in combating diseases, especially the ones caused by viral and bacterial pathogens. Despite this, several major public health threats, such as HIV, tuberculosis, malaria, and cancer, still pose an enormous humanitarian and economic burden. As vaccines based on the induction of protective, neutralizing antibodies have not managed to effectively combat these diseases, in recent decades, the focus has increasingly shifted towards the cellular immune response. There is substantial evidence demonstrating CD8 T cells as key players in the protection not only against many viral and bacterial pathogens, but also in the fight against neoplastic cells. Here, we present arguments for CD8 T cells to be considered as promising candidates for vaccine targeting. We discuss the heterogeneity of CD8 T cell populations and their contribution in the protection of the host. We also outline several strategies of using a common human pathogen, cytomegalovirus, as a vaccine vector since accumulated data strongly suggest it represents a promising approach to the development of novel vaccines against both pathogens and tumors.
Collapse
|
34
|
Riedel F, Aparicio-Soto M, Curato C, Thierse HJ, Siewert K, Luch A. Immunological Mechanisms of Metal Allergies and the Nickel-Specific TCR-pMHC Interface. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:10867. [PMID: 34682608 PMCID: PMC8535423 DOI: 10.3390/ijerph182010867] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/03/2021] [Accepted: 10/11/2021] [Indexed: 12/19/2022]
Abstract
Besides having physiological functions and general toxic effects, many metal ions can cause allergic reactions in humans. We here review the immune events involved in the mediation of metal allergies. We focus on nickel (Ni), cobalt (Co) and palladium (Pd), because these allergens are among the most prevalent sensitizers (Ni, Co) and immediate neighbors in the periodic table of the chemical elements. Co-sensitization between Ni and the other two metals is frequent while the knowledge on a possible immunological cross-reactivity using in vivo and in vitro approaches remains limited. At the center of an allergic reaction lies the capability of a metal allergen to form T cell epitopes that are recognized by specific T cell receptors (TCR). Technological advances such as activation-induced marker assays and TCR high-throughput sequencing recently provided new insights into the interaction of Ni2+ with the αβ TCR-peptide-major histocompatibility complex (pMHC) interface. Ni2+ functionally binds to the TCR gene segment TRAV9-2 or a histidine in the complementarity determining region 3 (CDR3), the main antigen binding region. Thus, we overview known, newly identified and hypothesized mechanisms of metal-specific T cell activation and discuss current knowledge on cross-reactivity.
Collapse
Affiliation(s)
- Franziska Riedel
- Department for Chemicals and Product Safety, Federal Institute for Risk Assessment, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany; (M.A.-S.); (C.C.); (H.-J.T.); (K.S.); (A.L.)
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2, 14195 Berlin, Germany
| | - Marina Aparicio-Soto
- Department for Chemicals and Product Safety, Federal Institute for Risk Assessment, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany; (M.A.-S.); (C.C.); (H.-J.T.); (K.S.); (A.L.)
| | - Caterina Curato
- Department for Chemicals and Product Safety, Federal Institute for Risk Assessment, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany; (M.A.-S.); (C.C.); (H.-J.T.); (K.S.); (A.L.)
| | - Hermann-Josef Thierse
- Department for Chemicals and Product Safety, Federal Institute for Risk Assessment, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany; (M.A.-S.); (C.C.); (H.-J.T.); (K.S.); (A.L.)
| | - Katherina Siewert
- Department for Chemicals and Product Safety, Federal Institute for Risk Assessment, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany; (M.A.-S.); (C.C.); (H.-J.T.); (K.S.); (A.L.)
| | - Andreas Luch
- Department for Chemicals and Product Safety, Federal Institute for Risk Assessment, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany; (M.A.-S.); (C.C.); (H.-J.T.); (K.S.); (A.L.)
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2, 14195 Berlin, Germany
| |
Collapse
|
35
|
Park SL, Mackay LK. Decoding Tissue-Residency: Programming and Potential of Frontline Memory T Cells. Cold Spring Harb Perspect Biol 2021; 13:a037960. [PMID: 33753406 PMCID: PMC8485744 DOI: 10.1101/cshperspect.a037960] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Memory T-cell responses are partitioned between the blood, secondary lymphoid organs, and nonlymphoid tissues. Tissue-resident memory T (Trm) cells are a population of immune cells that remain permanently in tissues without recirculating in blood. These nonrecirculating cells serve as a principal node in the anamnestic response to invading pathogens and developing malignancies. Here, we contemplate how T-cell tissue residency is defined and shapes protective immunity in the steady state and in the context of disease. We review the properties and heterogeneity of Trm cells, highlight the critical roles these cells play in maintaining tissue homeostasis and eliciting immune pathology, and explore how they might be exploited to treat disease.
Collapse
Affiliation(s)
- Simone L Park
- Department of Microbiology & Immunology at The Peter Doherty Institute for Infection & Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Laura K Mackay
- Department of Microbiology & Immunology at The Peter Doherty Institute for Infection & Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| |
Collapse
|
36
|
Schunkert EM, Shah PN, Divito SJ. Skin Resident Memory T Cells May Play Critical Role in Delayed-Type Drug Hypersensitivity Reactions. Front Immunol 2021; 12:654190. [PMID: 34497600 PMCID: PMC8419326 DOI: 10.3389/fimmu.2021.654190] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 08/06/2021] [Indexed: 12/31/2022] Open
Abstract
Delayed-type drug hypersensitivity reactions (dtDHR) are immune-mediated reactions with skin and visceral manifestations ranging from mild to severe. Clinical care is negatively impacted by a limited understanding of disease pathogenesis. Though T cells are believed to orchestrate disease, the type of T cell and the location and mechanism of T cell activation remain unknown. Resident memory T cells (TRM) are a unique T cell population potentially well situated to act as key mediators in disease pathogenesis, but significant obstacles to defining, identifying, and testing TRM in dtDHR preclude definitive conclusions at this time. Deeper mechanistic interrogation to address these unanswered questions is necessary, as involvement of TRM in disease has significant implications for prediction, diagnosis, and treatment of disease.
Collapse
|
37
|
Emmanuel T, Mistegård J, Bregnhøj A, Johansen C, Iversen L. Tissue-Resident Memory T Cells in Skin Diseases: A Systematic Review. Int J Mol Sci 2021; 22:ijms22169004. [PMID: 34445713 PMCID: PMC8396505 DOI: 10.3390/ijms22169004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/09/2021] [Accepted: 08/17/2021] [Indexed: 02/06/2023] Open
Abstract
In health, the non-recirculating nature and long-term persistence of tissue-resident memory T cells (TRMs) in tissues protects against invading pathogens. In disease, pathogenic TRMs contribute to the recurring traits of many skin diseases. We aimed to conduct a systematic literature review on the current understanding of the role of TRMs in skin diseases and identify gaps as well as future research paths. EMBASE, PubMed, SCOPUS, Web of Science, Clinicaltrials.gov and WHO Trials Registry were searched systematically for relevant studies from their inception to October 2020. Included studies were reviewed independently by two authors. This study was conducted in accordance with the PRISMA-S guidelines. This protocol was registered with the PROSPERO database (ref: CRD42020206416). We identified 96 studies meeting the inclusion criteria. TRMs have mostly been investigated in murine skin and in relation to infectious skin diseases. Pathogenic TRMs have been characterized in various skin diseases including psoriasis, vitiligo and cutaneous T-cell lymphoma. Studies are needed to discover biomarkers that may delineate TRMs poised for pathogenic activity in skin diseases and establish to which extent TRMs are contingent on the local skin microenvironment. Additionally, future studies may investigate the effects of current treatments on the persistence of pathogenic TRMs in human skin.
Collapse
|
38
|
Bertschi NL, Bazzini C, Schlapbach C. The Concept of Pathogenic TH2 Cells: Collegium Internationale Allergologicum Update 2021. Int Arch Allergy Immunol 2021; 182:365-380. [PMID: 33845475 DOI: 10.1159/000515144] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/09/2021] [Indexed: 11/19/2022] Open
Abstract
T helper (TH) cells have evolved into distinct subsets that mediate specific immune responses to protect the host against a myriad of infectious and noninfectious challenges. However, if dysregulated, TH-cell subsets can cause inflammatory disease. Emerging evidence now suggests that human allergic disease is caused by a distinct subpopulation of pathogenic TH2 cells. Pathogenic TH2 cells from different type-2-driven diseases share a core phenotype and show overlapping functional attributes. The unique differentiation requirements, activating signals, and metabolic characteristics of pathogenic TH2 cells are just being discovered. A better knowledge of this particular TH2 cell population will enable the specific targeting of disease-driving pathways in allergy. In this review, we introduce a rational for classifying TH cells into distinct subsets, discuss the current knowledge on pathogenic TH2 cells, and summarize their involvement in allergic diseases.
Collapse
Affiliation(s)
- Nicole L Bertschi
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Cecilia Bazzini
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Christoph Schlapbach
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
39
|
Korkmaz E, Balmert SC, Sumpter TL, Carey CD, Erdos G, Falo LD. Microarray patches enable the development of skin-targeted vaccines against COVID-19. Adv Drug Deliv Rev 2021; 171:164-186. [PMID: 33539853 PMCID: PMC8060128 DOI: 10.1016/j.addr.2021.01.022] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/10/2021] [Accepted: 01/27/2021] [Indexed: 12/13/2022]
Abstract
The COVID-19 pandemic is a serious threat to global health and the global economy. The ongoing race to develop a safe and efficacious vaccine to prevent infection by SARS-CoV-2, the causative agent for COVID-19, highlights the importance of vaccination to combat infectious pathogens. The highly accessible cutaneous microenvironment is an ideal target for vaccination since the skin harbors a high density of antigen-presenting cells and immune accessory cells with broad innate immune functions. Microarray patches (MAPs) are an attractive intracutaneous biocargo delivery system that enables safe, reproducible, and controlled administration of vaccine components (antigens, with or without adjuvants) to defined skin microenvironments. This review describes the structure of the SARS-CoV-2 virus and relevant antigenic targets for vaccination, summarizes key concepts of skin immunobiology in the context of prophylactic immunization, and presents an overview of MAP-mediated cutaneous vaccine delivery. Concluding remarks on MAP-based skin immunization are provided to contribute to the rational development of safe and effective MAP-delivered vaccines against emerging infectious diseases, including COVID-19.
Collapse
Affiliation(s)
- Emrullah Korkmaz
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Stephen C Balmert
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Tina L Sumpter
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Cara Donahue Carey
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Geza Erdos
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Louis D Falo
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| |
Collapse
|
40
|
Choi IJ, Cha HR, Hwang SJ, Baek SK, Lee JM, Choi SO. Live Vaccinia Virus-Coated Microneedle Array Patches for Smallpox Vaccination and Stockpiling. Pharmaceutics 2021; 13:209. [PMID: 33546332 PMCID: PMC7913550 DOI: 10.3390/pharmaceutics13020209] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/28/2021] [Accepted: 01/30/2021] [Indexed: 12/01/2022] Open
Abstract
Although smallpox has been eradicated globally, the potential use of the smallpox virus in bioterrorism indicates the importance of stockpiling smallpox vaccines. Considering the advantages of microneedle-based vaccination over conventional needle injections, in this study, we examined the feasibility of microneedle-based smallpox vaccination as an alternative approach for stockpiling smallpox vaccines. We prepared polylactic acid (PLA) microneedle array patches by micromolding and loaded a second-generation smallpox vaccine on the microneedle tips via dip coating. We evaluated the effect of excipients and drying conditions on vaccine stability in vitro and examined immune responses in female BALB/c mice by measuring neutralizing antibodies and interferon (IFN)-γ-secreting cells. Approximately 40% of the virus titer was reduced during the vaccine-coating process, with or without excipients. At -20 °C, the smallpox vaccine coated on the microneedles was stable up to 6 months. Compared to natural evaporation, vacuum drying was more efficient in improving the smallpox vaccine stability. Microneedle-based vaccination of the mice elicited neutralizing antibodies beginning 3 weeks after immunization; the levels were maintained for 12 weeks. It significantly increased IFN-γ-secreting cells 12 weeks after priming, indicating the induction of cellular immune responses. The smallpox-vaccine-coated microneedles could serve as an alternative delivery system for vaccination and stockpiling.
Collapse
Affiliation(s)
- In-Jeong Choi
- QuadMedicine R&D Centre, QuadMedicine, Inc., Seongnam 13209, Korea; (I.-J.C.); (S.-K.B.)
| | - Hye-Ran Cha
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea; (H.-R.C.); (S.J.H.)
| | - Su Jin Hwang
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea; (H.-R.C.); (S.J.H.)
| | - Seung-Ki Baek
- QuadMedicine R&D Centre, QuadMedicine, Inc., Seongnam 13209, Korea; (I.-J.C.); (S.-K.B.)
| | - Jae Myun Lee
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea; (H.-R.C.); (S.J.H.)
| | - Seong-O Choi
- QuadMedicine R&D Centre, QuadMedicine, Inc., Seongnam 13209, Korea; (I.-J.C.); (S.-K.B.)
| |
Collapse
|
41
|
Tokura Y, Phadungsaksawasdi P, Kurihara K, Fujiyama T, Honda T. Pathophysiology of Skin Resident Memory T Cells. Front Immunol 2021; 11:618897. [PMID: 33633737 PMCID: PMC7901930 DOI: 10.3389/fimmu.2020.618897] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/21/2020] [Indexed: 12/11/2022] Open
Abstract
Tissue resident memory T (TRM) cells reside in peripheral, non-lymphoid tissues such as the skin, where they act as alarm-sensor cells or cytotoxic cells. Physiologically, skin TRM cells persist for a long term and can be reactivated upon reinfection with the same antigen, thus serving as peripheral sentinels in the immune surveillance network. CD8+CD69+CD103+ TRM cells are the well-characterized subtype that develops in the epidermis. The local mediators such as interleukin (IL)-15 and transforming growth factor (TGF)-β are required for the formation of long-lived TRM cell population in skin. Skin TRM cells engage virus-infected cells, proliferate in situ in response to local antigens and do not migrate out of the epidermis. Secondary TRM cell populations are derived from pre-existing TRM cells and newly recruited TRM precursors from the circulation. In addition to microbial pathogens, topical application of chemical allergen to skin causes delayed-type hypersensitivity and amplifies the number of antigen-specific CD8+ TRM cells at challenged site. Skin TRM cells are also involved in the pathological conditions, including vitiligo, psoriasis, fixed drug eruption and cutaneous T-cell lymphoma (CTCL). The functions of these TRM cells seem to be different, depending on each pathology. Psoriasis plaques are seen in a recurrent manner especially at the originally affected sites. Upon stimulation of the skin of psoriasis patients, the CD8+CD103+CD49a- TRM cells in the epidermis seem to be reactivated and initiate IL-17A production. Meanwhile, autoreactive CD8+CD103+CD49a+ TRM cells secreting interferon-γ are present in lesional vitiligo skin. Fixed drug eruption is another disease where skin TRM cells evoke its characteristic clinical appearance upon administration of a causative drug. Intraepidermal CD8+ TRM cells with an effector-memory phenotype resident in the skin lesions of fixed drug eruption play a major contributing role in the development of localized tissue damage. CTCL develops primarily in the skin by a clonal expansion of a transformed TRM cells. CD8+ CTCL with the pagetoid epidermotropic histology is considered to originate from epidermal CD8+ TRM cells. This review will discuss the current understanding of skin TRM biology and their contribution to skin homeostasis and diseases.
Collapse
Affiliation(s)
- Yoshiki Tokura
- Department of Dermatology, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Department of Cellular & Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | | | - Kazuo Kurihara
- Department of Dermatology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Toshiharu Fujiyama
- Department of Dermatology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tetsuya Honda
- Department of Dermatology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
42
|
Korkmaz E, Balmert SC, Carey CD, Erdos G, Falo LD. Emerging skin-targeted drug delivery strategies to engineer immunity: A focus on infectious diseases. Expert Opin Drug Deliv 2021; 18:151-167. [PMID: 32924651 PMCID: PMC9355143 DOI: 10.1080/17425247.2021.1823964] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Infectious pathogens are global disrupters. Progress in biomedical science and technology has expanded the public health arsenal against infectious diseases. Specifically, vaccination has reduced the burden of infectious pathogens. Engineering systemic immunity by harnessing the cutaneous immune network has been particularly attractive since the skin is an easily accessible immune-responsive organ. Recent advances in skin-targeted drug delivery strategies have enabled safe, patient-friendly, and controlled deployment of vaccines to cutaneous microenvironments for inducing long-lived pathogen-specific immunity to mitigate infectious diseases, including COVID-19. AREAS COVERED This review briefly discusses the basics of cutaneous immunomodulation and provides a concise overview of emerging skin-targeted drug delivery systems that enable safe, minimally invasive, and effective intracutaneous administration of vaccines for engineering systemic immune responses to combat infectious diseases. EXPERT OPINION In-situ engineering of the cutaneous microenvironment using emerging skin-targeted vaccine delivery systems offers remarkable potential to develop diverse immunization strategies against pathogens. Mechanistic studies with standard correlates of vaccine efficacy will be important to compare innovative intracutaneous drug delivery strategies to each other and to existing clinical approaches. Cost-benefit analyses will be necessary for developing effective commercialization strategies. Significant involvement of industry and/or government will be imperative for successfully bringing novel skin-targeted vaccine delivery methods to market for their widespread use.
Collapse
Affiliation(s)
- Emrullah Korkmaz
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephen C. Balmert
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Cara Donahue Carey
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Geza Erdos
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Louis D. Falo
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA,UPMC Hillman Cancer Center, Pittsburgh, PA, USA,Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA,The McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
43
|
Sun Z, Kim JH, Kim SH, Kim HR, Zhang K, Pan Y, Ko MK, Kim BM, Chu H, Lee HR, Kim HL, Kim JH, Fu X, Hyun YM, Yun KN, Kim JY, Lee DW, Song SY, Lin CP, Clark RA, Lee KH, Kupper TS, Park CO. Skin-resident natural killer T cells participate in cutaneous allergic inflammation in atopic dermatitis. J Allergy Clin Immunol 2021; 147:1764-1777. [PMID: 33516870 DOI: 10.1016/j.jaci.2020.11.049] [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: 02/03/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Natural killer T (NKT) cells are unconventional T cells that bridge innate and adaptive immunity. NKT cells have been implicated in the development of atopic dermatitis (AD). OBJECTIVE We aimed to investigate the role of NKT cells in AD development, especially in skin. METHODS Global proteomic and transcriptomic analyses were performed by using skin and blood from human healthy-controls and patients with AD. Levels of CXCR4 and CXCL12 expression in skin NKT cells were analyzed in human AD and mouse AD models. By using parabiosis and intravital imaging, the role of skin CXCR4+ NKT cells was further evaluated in models of mice with AD by using CXCR4-conditionally deficient or CXCL12 transgenic mice. RESULTS CXCR4 and its cognate ligand CXCL12 were significantly upregulated in the skin of humans with AD by global transcriptomic and proteomic analyses. CXCR4+ NKT cells were enriched in AD skin, and their levels were consistently elevated in our models of mice with AD. Allergen-induced NKT cells participate in cutaneous allergic inflammation. Similar to tissue-resident memory T cells, the predominant skin NKT cells were CXCR4+ and CD69+. Skin-resident NKT cells uniquely expressed CXCR4, unlike NKT cells in the liver, spleen, and lymph nodes. Skin fibroblasts were the main source of CXCL12. CXCR4+ NKT cells preferentially trafficked to CXCL12-rich areas, forming an enriched CXCR4+ tissue-resident NKT cells/CXCL12+ cell cluster that developed in acute and chronic allergic inflammation in our models of mice with AD. CONCLUSIONS CXCR4+ tissue-resident NKT cells may form a niche that contributes to AD, in which CXCL12 is highly expressed.
Collapse
Affiliation(s)
- ZhengWang Sun
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hye Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea; Brain Korea 21 Project, Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Seo Hyeong Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea; Brain Korea 21 Project, Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Ran Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - KeLun Zhang
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea; Brain Korea 21 Project, Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Youdong Pan
- Department of Dermatology and Harvard Skin Disease Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Min Kyung Ko
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Bo Mi Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Howard Chu
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Hee Ra Lee
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Li Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea; Brain Korea 21 Project, Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hyung Kim
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Xiujun Fu
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Young-Min Hyun
- Brain Korea 21 Project, Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, Korea; Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
| | - Ki Na Yun
- Biomedical Omics Group, Korea Basic Science Institute, Ochang, Korea
| | - Jin Young Kim
- Biomedical Omics Group, Korea Basic Science Institute, Ochang, Korea
| | - Dong Won Lee
- Department of Plastic and Reconstructive Surgery, Institute for Human Tissue Restoration, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Yong Song
- Department of Plastic and Reconstructive Surgery, Institute for Human Tissue Restoration, Yonsei University College of Medicine, Seoul, Korea
| | - Charles P Lin
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Rachael A Clark
- Department of Dermatology and Harvard Skin Disease Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Kwang Hoon Lee
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea; Brain Korea 21 Project, Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, Korea.
| | - Thomas S Kupper
- Department of Dermatology and Harvard Skin Disease Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass.
| | - Chang Ook Park
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea; Brain Korea 21 Project, Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, Korea; Department of Dermatology and Harvard Skin Disease Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass.
| |
Collapse
|
44
|
Salerno-Gonçalves R, Chen WH, Mulligan MJ, Frey SE, Stapleton JT, Keitel WA, Bailey J, Sendra E, Hill H, Johnson RA, Sztein MB. Vaccine-related major cutaneous reaction size correlates with cellular-mediated immune responses after tularaemia immunisation. Clin Transl Immunology 2021; 10:e1239. [PMID: 33505681 PMCID: PMC7814273 DOI: 10.1002/cti2.1239] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 01/31/2023] Open
Abstract
Objectives Francisella tularensis, the causative agent of tularaemia, is an exceptionally infectious bacterium, potentially fatal for humans if left untreated and with the potential to be developed as a bioweapon. Both natural infection and live-attenuated vaccine strain (LVS) confer good protection against tularaemia. LVS vaccination is traditionally administered by scarification, and the formation of a cutaneous reaction or take at the vaccination site is recognised as a clinical correlate of protection. Although previous studies have suggested that high antibody titres following vaccination might serve as a useful surrogate marker, the immunological correlates of protection remain unknown. Methods We investigated the host T-cell-mediated immune (T-CMI) responses elicited following immunisation with LVS vaccine formulated by the DynPort Vaccine Company (DVC-LVS) or the United States Army Medical Research Institute of Infectious Diseases (USAMRIID-LVS). We compared T-CMI responses prompted by these vaccines and correlated them with take size. Results We found that both LVS vaccines elicited similar T-CMI responses. Interestingly, take size associated with the T cells' ability to proliferate, secrete IFN-γ and mobilise degranulation, suggesting that these responses play an essential role in tularaemia protection. Conclusions These results renew the appreciation for vaccination through the scarification as a prime route of inoculation to target pathogens driving specific T-CMI responses and provide further evidence that T-CMI plays a role in protection from tularaemia.
Collapse
Affiliation(s)
- Rosangela Salerno-Gonçalves
- Center for Vaccine Development and Global Health (CVD) University of Maryland School of Medicine Baltimore MD USA
| | - Wilbur H Chen
- Center for Vaccine Development and Global Health (CVD) University of Maryland School of Medicine Baltimore MD USA
| | - Mark J Mulligan
- The Hope Clinic of the Emory Vaccine Center Department of Medicine Division of Infectious Diseases Emory University School of Medicine Decatur GA USA.,Present address: NYU Langone Vaccine Center NYU Grossman School of Medicine Alexandria Center for Life Sciences (West Tower) New York NY USA
| | - Sharon E Frey
- Division of Infectious Diseases, Allergy and Immunology Saint Louis University School of Medicine St. Louis MO USA
| | - Jack T Stapleton
- Iowa City Veterans Administration and the University of Iowa Iowa City IA USA
| | - Wendy A Keitel
- Departments of Molecular Virology and Microbiology, and Medicine Baylor College of Medicine Houston TX USA
| | - Jason Bailey
- Emmes Rockville MD USA.,Present address: Armed Forces Health Surveillance Branch, Integrated Biosurveillance Silver Spring MD USA
| | | | | | - Robert A Johnson
- Biomedical Advanced Research and Development Authority Department of Health and Human Services ASPR Washington DC USA
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health (CVD) University of Maryland School of Medicine Baltimore MD USA.,Program in Oncology University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center Baltimore MD USA
| |
Collapse
|
45
|
Epicutaneous immunization with modified vaccinia Ankara viral vectors generates superior T cell immunity against a respiratory viral challenge. NPJ Vaccines 2021; 6:1. [PMID: 33398010 PMCID: PMC7782760 DOI: 10.1038/s41541-020-00265-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022] Open
Abstract
Modified Vaccinia Ankara (MVA) was recently approved as a smallpox vaccine. Variola is transmitted by respiratory droplets and MVA immunization by skin scarification (s.s.) protected mice far more effectively against lethal respiratory challenge with vaccinia virus (VACV) than any other route of delivery, and at lower doses. Comparisons of s.s. with intradermal, subcutaneous, or intramuscular routes showed that MVAOVA s.s.-generated T cells were both more abundant and transcriptionally unique. MVAOVA s.s. produced greater numbers of lung Ova-specific CD8+ TRM and was superior in protecting mice against lethal VACVOVA respiratory challenge. Nearly as many lung TRM were generated with MVAOVA s.s. immunization compared to intra-tracheal immunization with MVAOVA and both routes vaccination protected mice against lethal pulmonary challenge with VACVOVA. Strikingly, MVAOVA s.s.-generated effector T cells exhibited overlapping gene transcriptional profiles to those generated via intra-tracheal immunization. Overall, our data suggest that heterologous MVA vectors immunized via s.s. are uniquely well-suited as vaccine vectors for respiratory pathogens, which may be relevant to COVID-19. In addition, MVA delivered via s.s. could represent a more effective dose-sparing smallpox vaccine.
Collapse
|
46
|
Fang H, Li Q, Wang G. The role of T cells in pemphigus vulgaris and bullous pemphigoid. Autoimmun Rev 2020; 19:102661. [DOI: 10.1016/j.autrev.2020.102661] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 04/03/2020] [Indexed: 12/28/2022]
|
47
|
Manh DH, Weiss LN, Thuong NV, Mizukami S, Dumre SP, Luong QC, Thanh LC, Thang CM, Huu PT, Phuc LH, Nhung CTH, Mai NT, Truong NQ, Ngu VTT, Quoc DK, Ha TTN, Ton T, An TV, Halhouli O, Quynh LN, Kamel MG, Karbwang J, Huong VTQ, Huy NT, Hirayama K. Kinetics of CD4 + T Helper and CD8 + Effector T Cell Responses in Acute Dengue Patients. Front Immunol 2020; 11:1980. [PMID: 33072068 PMCID: PMC7542683 DOI: 10.3389/fimmu.2020.01980] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 07/22/2020] [Indexed: 12/13/2022] Open
Abstract
Background: The protective or pathogenic role of T lymphocytes during the acute phase of dengue virus (DENV) infection has not been fully understood despite its importance in immunity and vaccine development. Objectives: This study aimed to clarify the kinetics of T lymphocyte subsets during the clinical course of acute dengue patients. Study design: In this hospital-based cohort study, 59 eligible Vietnamese dengue patients were recruited and admitted. They were investigated and monitored for T cell subsets and a panel of clinical and laboratory parameters every day until discharged and at post-discharge from the hospital. Results: We described for the first time the kinetics of T cell response during the clinical course of DENV infection. Severe cases showed significantly lower levels of effector CD8+ T cells compared to mild cases at day −1 (p = 0.017) and day 0 (p = 0.033) of defervescence. After defervescence, these cell counts in severe cases increased rapidly to equalize with the levels of mild cases. Our results also showed a decline in total CD4+ T, Th1, Th1/17 cells during febrile phase of dengue patients compared to normal controls or convalescent phase. On the other hand, Th2 cells increased during DENV infection until convalescent phase. Cytokines such as interferon-γ, IL-12p70, IL-5, IL-23, IL-17A showed tendency to decrease on day 0 and 1 compared with convalescence and only IL-5 showed significance indicating the production during acute phase was not systemic. Conclusion: With a rigorous study design, we uncovered the kinetics of T cells in natural DENV infection. Decreased number of effector CD8+ T cells in the early phase of infection and subsequent increment after defervescence day probably associated with the T cell migration in DENV infection.
Collapse
Affiliation(s)
- Dao Huy Manh
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.,Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Lan Nguyen Weiss
- Department of Immunology and Microbiology, Pasteur Institute, Ho Chi Minh City, Vietnam
| | - Nguyen Van Thuong
- Department of Immunology and Microbiology, Pasteur Institute, Ho Chi Minh City, Vietnam
| | - Shusaku Mizukami
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.,Department of Clinical Product Development, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Shyam Prakash Dumre
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Quang Chan Luong
- National Program for Dengue Control, Pasteur Institute, Ho Chi Minh City, Vietnam
| | - Le Chi Thanh
- HIV Laboratory, Pasteur Institute, Ho Chi Minh City, Vietnam
| | - Cao Minh Thang
- Department of Immunology and Microbiology, Pasteur Institute, Ho Chi Minh City, Vietnam
| | | | | | - Cao Thi Hong Nhung
- Department of Immunology and Microbiology, Pasteur Institute, Ho Chi Minh City, Vietnam
| | - Nguyen Thi Mai
- Department of Immunology and Microbiology, Pasteur Institute, Ho Chi Minh City, Vietnam
| | - Nguyen Quang Truong
- Department of Immunology and Microbiology, Pasteur Institute, Ho Chi Minh City, Vietnam
| | - Vu Thien Thu Ngu
- Department of Immunology and Microbiology, Pasteur Institute, Ho Chi Minh City, Vietnam
| | - Do Kien Quoc
- National Program for Dengue Control, Pasteur Institute, Ho Chi Minh City, Vietnam
| | - Tran Thi Ngoc Ha
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Tran Ton
- HIV Laboratory, Pasteur Institute, Ho Chi Minh City, Vietnam
| | - Tran Van An
- Nguyen Dinh Chieu Hospital, Ben Tre, Vietnam
| | - Oday Halhouli
- Faculty of Medicine, The University of Jordan, Amman, Jordan.,Online Research Club (www.onlineresearchclub.org/), Nagasaki, Japan
| | - Le Nhat Quynh
- Online Research Club (www.onlineresearchclub.org/), Nagasaki, Japan.,Hue University of Medicine and Pharmacy, Hue, Vietnam
| | - Mohamed Gomaa Kamel
- Online Research Club (www.onlineresearchclub.org/), Nagasaki, Japan.,Faculty of Medicine, Minia University, Minya, Egypt
| | - Juntra Karbwang
- Department of Clinical Product Development, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Vu Thi Que Huong
- Department of Immunology and Microbiology, Pasteur Institute, Ho Chi Minh City, Vietnam
| | - Nguyen Tien Huy
- Department of Clinical Product Development, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.,Online Research Club (www.onlineresearchclub.org/), Nagasaki, Japan
| | - Kenji Hirayama
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.,Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
48
|
Dhodapkar MV, Dhodapkar KM. Tissue-resident memory-like T cells in tumor immunity: Clinical implications. Semin Immunol 2020; 49:101415. [PMID: 33011063 DOI: 10.1016/j.smim.2020.101415] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/06/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022]
Abstract
Tissue-resident memory (TRM) T cells are distinct population of non-circulating lymphocytes that play an important role in mediating regional immunity. TRM- like cells have now been identified as a component of tumor-infiltrating lymphocytes in several human tumors and correlate with outcome and response to immunotherapy. TRM cells have also been shown to mediate anti-tumor immunity in murine models. Biology of TRM cells has several implications for clinical cancer immunotherapy. Here we discuss newer insights into the biology of TRM T cells and discuss their implications for understanding the heterogeneity of immune microenvironment in tumors as well as improving the efficacy of cancer vaccines, immune-checkpoint blockade and adoptive cellular therapies in the clinic.
Collapse
Affiliation(s)
- Madhav V Dhodapkar
- Department of Hematology / Medical Oncology, Emory University, Atlanta, GA, United States; Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Kavita M Dhodapkar
- Winship Cancer Institute, Emory University, Atlanta, GA, United States; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, Atlanta, GA, United States.
| |
Collapse
|
49
|
Wang T, Shen Y, Luyten S, Yang Y, Jiang X. Tissue-resident memory CD8+ T cells in cancer immunology and immunotherapy. Pharmacol Res 2020; 159:104876. [DOI: 10.1016/j.phrs.2020.104876] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023]
|
50
|
Th1 concomitant immune response mediated by IFN-γ protects against sand fly delivered Leishmania infection: Implications for vaccine design. Cytokine 2020; 147:155247. [PMID: 32873468 DOI: 10.1016/j.cyto.2020.155247] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/26/2020] [Accepted: 08/08/2020] [Indexed: 02/07/2023]
Abstract
Leishmaniasis is an unresolved global health problem with a high socio-economic impact. Data generated in mouse models has revealed that the Th1 response, with IL-12, IFN-γ, TNF-α, and IL-2 as prominent cytokines, predominantly controls the disease progression. Premised on these findings, all examined vaccine formulations have been aimed at generating a long-lived memory Th1 response. However, all vaccine formulations with the exception of live Leishmania inoculation (leishmanization) have failed to sufficiently protect against sand fly delivered infection. It has been recently unraveled that sand fly dependent factors may compromise pre-existing Th1 memory. Further scrutinizing the immune response after leishmanization has uncovered the prominent role of early (within hours) and robust IFN-γ production (Th1 concomitant immunity) in controlling the sand fly delivered secondary infection. The response is dependent upon parasite persistence and subclinical ongoing primary infection. The immune correlates of concomitant immunity (Resident Memory T cells and Effector T subsets) mitigate the early effects of sand fly delivered infection and help to control the disease. In this review, we have described the early events after sand fly challenge and the role of Th1 concomitant immunity in the protective immune response in leishmanized resistant mouse model, although leishmanization is under debate for human use. Undoubtedly, the lessons we learn from leishmanization must be further implemented in alternative vaccine approaches.
Collapse
|