1
|
Guerrero GG, Madrid-Marina V, Martínez-Romero A, Torres-Poveda K, Favela-Hernández JM. Host-Pathogen Interaction Interface: Promising Candidate Targets for Vaccine-Induced Protective and Memory Immune Responses. Vaccines (Basel) 2025; 13:418. [PMID: 40333316 PMCID: PMC12031405 DOI: 10.3390/vaccines13040418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 05/09/2025] Open
Abstract
Vaccine formulations are a successful strategy against pathogen transmission because vaccine candidates induce effective and long-lasting memory immune responses (B and CD4+ T cells) at systemic and mucosal sites. Extracellular vesicles of lipoproteins, bioactive compounds from plants and invertebrates (sponges) encapsulated in liposomes, and glycoproteins can target these sites. The vaccine candidates developed can mimic microbial pathogens in a way that successfully links the innate and adaptive immune responses. In addition, vaccines plus adjuvants promote and maintain an inflammatory response. In this review, we aimed to identify the host-pathogen interface as a rich source of candidate targets for vaccine-induced protective and long-lasting memory immune responses.
Collapse
Affiliation(s)
- Gloria G. Guerrero
- Unidad Académica de Ciencias Biológicas, Universidad Autónoma de Zacatecas, Zacatecas 98600, Zac., Mexico
| | - Vicente Madrid-Marina
- Centro de Investigación en Enfermedades infecciosas (CISEI), Instituto Nacional de Salud Pública (INSP), Cuernavaca 62100, Mor., Mexico
| | - Aurora Martínez-Romero
- Facultad de Química, Universidad Juárez del Estado de Durango, Gómez Palacio 34100, Dgo., Mexico
| | - Kirvis Torres-Poveda
- Centro de Investigación en Enfermedades infecciosas (CISEI), Instituto Nacional de Salud Pública (INSP), Cuernavaca 62100, Mor., Mexico
- Secretaria de Ciencia, Humanidades y Tecnologías (SECIHTI), Instituto Nacional de Salud Pública, Cuernavaca 62100, Mor., Mexico
| | - Juan Manuel Favela-Hernández
- Facultad de Química, Universidad Juárez del Estado de Durango, Gómez Palacio 34100, Dgo., Mexico
- Instituto Multidisciplinario de Ciencias “Avicena”, Torreón 27250, Coah., Mexico
| |
Collapse
|
2
|
Ansari A, Sachan S, Ahuja J, Venkadesan S, Nikam B, Kumar V, Jain S, Singh BP, Coshic P, Sikka K, Wig N, Sette A, Weiskopf D, Mohanty D, Soneja M, Gupta N. Distinct features of a peripheral T helper subset that drives the B cell response in dengue virus infection. Cell Rep 2025; 44:115366. [PMID: 40073863 PMCID: PMC12032839 DOI: 10.1016/j.celrep.2025.115366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 11/28/2024] [Accepted: 02/06/2025] [Indexed: 03/14/2025] Open
Abstract
Dengue-virus-induced humoral immunity can increase the risk of severe disease, but the factors influencing this response are poorly understood. Here, we investigate the contribution of CD4+ T cells to B cell responses in human dengue infection. We identify a dominant peripheral PD-1+ T cell subset that accumulates in severe patients and could induce B cell differentiation via interleukin-21 (IL-21)-related pathway. Single-cell analyses reveal heterogeneity within PD-1+ cells, demonstrating the coexistence of subsets with "helper" (IL-21+) or "cytotoxic" characteristics. The IL-21+ subset displays a distinct clonotypic and transcriptomic signature compared to follicular helper T cells and persists as a memory in lymph nodes. Notably, we show that the IL-21+ subset seems to majorly drive the extrafollicular B cell responses in dengue. Our study establishes the peripheral IL-21+ subset as a potential determinant of the humoral response to dengue virus infection. These findings provide important insights into the T-cell-dependent regulation of humoral responses and can inform the design of effective dengue vaccines.
Collapse
Affiliation(s)
- Asgar Ansari
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Shilpa Sachan
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Jatin Ahuja
- Department of Medicine, All India Institute of Medical Sciences, New Delhi 110029, India
| | | | - Bhushan Nikam
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Vinod Kumar
- Department of Medicine, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Shweta Jain
- Department of Medicine, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Bhanu Pratap Singh
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Poonam Coshic
- Department of Transfusion Medicine, AIIMS, New Delhi 110029, India
| | - Kapil Sikka
- Department of Otorhinolaryngology, Head and Neck Surgery, AIIMS, New Delhi 110029, India
| | - Naveet Wig
- Department of Medicine, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Daniela Weiskopf
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Debasisa Mohanty
- Bioinformatics Center, National Institute of Immunology, New Delhi 110067, India
| | - Manish Soneja
- Department of Medicine, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Nimesh Gupta
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi 110067, India.
| |
Collapse
|
3
|
Mohamed AO, Boone DT, Ferry SL, Peck MC, Santos AM, Soderholm HE, Wittling MC, Paulos C, Turk MJ, Huang YH. CD4 T cell depletion increases memory differentiation of endogenous and CAR T cells and enhances the efficacy of Super2 and IL-33-armored CAR T cells against solid tumors. J Immunother Cancer 2025; 13:e009994. [PMID: 39933839 PMCID: PMC11815418 DOI: 10.1136/jitc-2024-009994] [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: 07/01/2024] [Accepted: 01/23/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Responsiveness to chimeric antigen receptor (CAR) T cell therapy correlates with CAR T cell expansion and persistence in vivo. Multiple strategies improve persistence by increasing stem-like properties or sustaining CAR T cell activity with combination therapies. Here, we describe the intrinsic ability of CAR T cells to differentiate into memory T cells, the effect of cytokine armoring, and neoadjuvant CD4 depletion therapy on CAR and tumor-specific endogenous memory T cells. METHODS TRP1-specific or NKG2D CAR T cells alone or with Super2+IL-33 (S233) armoring and/or CD4 depletion were evaluated in immunocompetent B16F10 melanoma or MC38 colon cell carcinoma models without preconditioning. We characterized CAR and endogenous tumor-specific memory T cell precursors, establishment of circulating (TCIRC) and resident (TRM) memory T cell subsets, and ability to protect against secondary tumors. RESULTS TRP1-specific or NKG2D CAR T cells had no effect on primary tumor growth in immunocompetent mice unless they were combined with S233 armoring or CD4 depletion. Unarmored CAR T cells expressed a stem-like phenotype in the tumor-draining lymph node and differentiated into CAR TCIRC memory cells in lymphoid organs and CAR TRM cells in the skin. In contrast, S233-armored CAR T cells exhibited an activated effector phenotype and differentiated inefficiently into CAR effector and central memory T cells. Combining CD4 therapy with unarmored CAR T cells increased CAR TCIRC and TRM memory T cells. Either CD4 depletion therapy or S233-armored CAR T cells induced activation of tumor-specific endogenous T cells that differentiated into both TCIRC and TRM memory T cells. CD4 depletion and S233-armored CAR T cell combination therapy synergized to increase endogenous memory T cells. CONCLUSIONS Unarmored TRP-1-specific or NKG2D CAR T cells have intrinsic stem-like properties and differentiate into memory T cell subsets but are non-protective against primary or secondary tumors. S233 cytokine armoring alone or with CD4 depletion improved effector responses but limited CAR memory T cell generation. S233-armored CAR T cells or CD4 depletion therapy induced endogenous tumor-specific TCIRC and TRM T cells, but the combination potentiated endogenous memory T cell generation and resulted in improved protection against B16F10 rechallenge.
Collapse
Affiliation(s)
- Asmaa O Mohamed
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - David Tyler Boone
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Shannon L Ferry
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Melanie C Peck
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Alicia M Santos
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Haille E Soderholm
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | | | - Chrystal Paulos
- Department of Surgery, Emory University, Atlanta, Georgia, USA
- Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Mary Jo Turk
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
- Dartmouth Cancer Center, Lebanon, New Hampshire, USA
| | - Yina H Huang
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
- Dartmouth Cancer Center, Lebanon, New Hampshire, USA
- Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, New Hampshire, USA
| |
Collapse
|
4
|
Singer M, Husseiny MI. Immunological Considerations for the Development of an Effective Herpes Vaccine. Microorganisms 2024; 12:1846. [PMID: 39338520 PMCID: PMC11434158 DOI: 10.3390/microorganisms12091846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/27/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
Research is underway to develop a vaccine to prevent and cure infection from herpes simplex virus (HSV). It emphasizes the critical need for immunization to address public health issues and the shortcomings of existing treatment options. Furthermore, studies on the HSV vaccine advance the field of immunology and vaccine creation, which may help in the battle against other viral illnesses. The current lack of such a vaccine is, in part, due to herpes viral latency in sensory ganglions. Current vaccines rely on tissue-resident memory CD8+ T cells, which are known to provide protection against subsequent HSV reinfection and reactivation without correlating with other immune subsets. For that reason, there is no effective vaccine that can provide protection against latent or recurrent herpes infection. This review focuses on conventional methods for evaluating the efficacy of a herpes vaccine using differential CD8+ T cells and important unaccounted immune aspects for designing an effective vaccine against herpes.
Collapse
Affiliation(s)
- Mahmoud Singer
- School of Medicine, University of California Irvine, Irvine, CA 92617, USA
| | - Mohamed I. Husseiny
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
5
|
Shen A, Garrett A, Chao CC, Liu D, Cheng C, Wang Z, Qian C, Zhu Y, Mai J, Jiang C. A comprehensive meta-analysis of tissue resident memory T cells and their roles in shaping immune microenvironment and patient prognosis in non-small cell lung cancer. Front Immunol 2024; 15:1416751. [PMID: 39040095 PMCID: PMC11260734 DOI: 10.3389/fimmu.2024.1416751] [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: 04/13/2024] [Accepted: 06/20/2024] [Indexed: 07/24/2024] Open
Abstract
Tissue-resident memory T cells (TRM) are a specialized subset of long-lived memory T cells that reside in peripheral tissues. However, the impact of TRM-related immunosurveillance on the tumor-immune microenvironment (TIME) and tumor progression across various non-small-cell lung cancer (NSCLC) patient populations is yet to be elucidated. Our comprehensive analysis of multiple independent single-cell and bulk RNA-seq datasets of patient NSCLC samples generated reliable, unique TRM signatures, through which we inferred the abundance of TRM in NSCLC. We discovered that TRM abundance is consistently positively correlated with CD4+ T helper 1 cells, M1 macrophages, and resting dendritic cells in the TIME. In addition, TRM signatures are strongly associated with immune checkpoint and stimulatory genes and the prognosis of NSCLC patients. A TRM-based machine learning model to predict patient survival was validated and an 18-gene risk score was further developed to effectively stratify patients into low-risk and high-risk categories, wherein patients with high-risk scores had significantly lower overall survival than patients with low-risk. The prognostic value of the risk score was independently validated by the Cancer Genome Atlas Program (TCGA) dataset and multiple independent NSCLC patient datasets. Notably, low-risk NSCLC patients with higher TRM infiltration exhibited enhanced T-cell immunity, nature killer cell activation, and other TIME immune responses related pathways, indicating a more active immune profile benefitting from immunotherapy. However, the TRM signature revealed low TRM abundance and a lack of prognostic association among lung squamous cell carcinoma patients in contrast to adenocarcinoma, indicating that the two NSCLC subtypes are driven by distinct TIMEs. Altogether, this study provides valuable insights into the complex interactions between TRM and TIME and their impact on NSCLC patient prognosis. The development of a simplified 18-gene risk score provides a practical prognostic marker for risk stratification.
Collapse
Affiliation(s)
- Aidan Shen
- Department of Precision Medicine, Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States
| | - Aliesha Garrett
- Department of Precision Medicine, Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States
| | - Cheng-Chi Chao
- Department of Pipeline Development, Biomap, Inc., San Francisco, CA, United States
| | - Dongliang Liu
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Chao Cheng
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Zhaohui Wang
- Department of Precision Medicine, Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States
| | - Chen Qian
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Yangzhi Zhu
- Department of Precision Medicine, Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States
| | - Junhua Mai
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States
| | - Chongming Jiang
- Department of Precision Medicine, Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States
| |
Collapse
|
6
|
Di Domenico M, Serretiello E, Smimmo A, Vieira e Silva FF, Raimondi SA, Pascariello C, Marino MM, Lo Muzio L, Caponio VCA, Cantore S, Ballini A. Monitoring of Immune Memory by Phenotypical Lymphocyte Subsets Identikit: An Observational Study in a Blood Donors' Cohort. J Pers Med 2024; 14:733. [PMID: 39063987 PMCID: PMC11277854 DOI: 10.3390/jpm14070733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
The cross-talk between the innate and adaptive immune response represents the first defense weapon against the threat of pathogens. Substantial evidence has shown a relationship between immune phenotype lymphocytes and COVID-19 disease severity and/or implication in susceptibility to SARS-CoV-2 infection. Recently, belonging to ABO blood groups has been investigated as a correlation factor to COVID-19 disease. This pilot study investigated lymphocyte typing in a cohort of blood donors to understand the underlying mechanism in SARS-CoV-2 infection linked to the blood group. The study cohort consisted of 20-64-year-old subjects, without comorbidities, from both sexes, who were COVID-19 vaccinated with previous or no infection history. Whole blood samples, collected at A.O.R.N. Sant'Anna and San Sebastiano Hospital (Campania Region), were processed by multiparametric cytofluorimetric assay, to characterize CD4+ helper and CD8+ cytotoxic T cell CD3+ subpopulations. The CD45RA, CCR7, CD27, CD28, CD57 and PD-1 markers were investigated to delineate the peripheral T-cell maturation stages. Differences were detected in ABO blood types in CD3+, CD4+ gated on CD3+, CD8+ and CD8+ gated on CD3+ percentage. These results contribute to identifying a memory cell "identikit" profile in COVID-19 disease, thus leading to a useful tool in precision medicine.
Collapse
Affiliation(s)
- Marina Di Domenico
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.D.D.); (E.S.); (A.S.); (F.F.V.e.S.); (M.M.M.); (A.B.)
| | - Enrica Serretiello
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.D.D.); (E.S.); (A.S.); (F.F.V.e.S.); (M.M.M.); (A.B.)
- Clinical Pathology and Microbiology Unit, San Giovanni di Dio and Ruggi D’Aragona University Hospital, 84131 Salerno, Italy
| | - Annafrancesca Smimmo
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.D.D.); (E.S.); (A.S.); (F.F.V.e.S.); (M.M.M.); (A.B.)
| | - Fábio França Vieira e Silva
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.D.D.); (E.S.); (A.S.); (F.F.V.e.S.); (M.M.M.); (A.B.)
| | - Sonia Anna Raimondi
- Azienda Ospedaliera “Sant’Anna e San Sebastiano”, 81100 Caserta, Italy; (S.A.R.); (C.P.)
| | - Caterina Pascariello
- Azienda Ospedaliera “Sant’Anna e San Sebastiano”, 81100 Caserta, Italy; (S.A.R.); (C.P.)
| | - Maria Michela Marino
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.D.D.); (E.S.); (A.S.); (F.F.V.e.S.); (M.M.M.); (A.B.)
| | - Lorenzo Lo Muzio
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy; (L.L.M.); (V.C.A.C.)
| | - Vito Carlo Alberto Caponio
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy; (L.L.M.); (V.C.A.C.)
| | - Stefania Cantore
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.D.D.); (E.S.); (A.S.); (F.F.V.e.S.); (M.M.M.); (A.B.)
| | - Andrea Ballini
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.D.D.); (E.S.); (A.S.); (F.F.V.e.S.); (M.M.M.); (A.B.)
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy; (L.L.M.); (V.C.A.C.)
| |
Collapse
|
7
|
Miyahara A, Umeki A, Sato K, Nomura T, Yamamoto H, Miyasaka T, Tanno D, Matsumoto I, Zong T, Kagesawa T, Oniyama A, Kawamura K, Yuan X, Yokoyama R, Kitai Y, Kanno E, Tanno H, Hara H, Yamasaki S, Saijo S, Iwakura Y, Ishii K, Kawakami K. Innate phase production of IFN-γ by memory and effector T cells expressing early activation marker CD69 during infection with Cryptococcus deneoformans in the lungs. Infect Immun 2024; 92:e0002424. [PMID: 38700335 PMCID: PMC11237684 DOI: 10.1128/iai.00024-24] [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: 01/18/2024] [Accepted: 04/09/2024] [Indexed: 05/05/2024] Open
Abstract
Cryptococcus deneoformans is a yeast-type fungus that causes fatal meningoencephalitis in immunocompromised patients and evades phagocytic cell elimination through an escape mechanism. Memory T (Tm) cells play a central role in preventing the reactivation of this fungal pathogen. Among these cells, tissue-resident memory T (TRM) cells quickly respond to locally invaded pathogens. This study analyzes the kinetics of effector T (Teff) cells and Tm cells in the lungs after cryptococcal infection. Emphasis is placed on the kinetics and cytokine expression of TRM cells in the early phase of infection. CD4+ Tm cells exhibited a rapid increase by day 3, peaked at day 7, and then either maintained their levels or exhibited a slight decrease until day 56. In contrast, CD8+ Tm cells reached their peak on day 3 and thereafter decreased up to day 56 post-infection. These Tm cells were predominantly composed of CD69+ TRM cells and CD69+ CD103+ TRM cells. Disruption of the CARD9 gene resulted in reduced accumulation of these TRM cells and diminished interferon (IFN) -γ expression in TRM cells. TRM cells were derived from T cells with T cell receptors non-specific to ovalbumin in OT-II mice during cryptococcal infection. In addition, TRM cells exhibited varied behavior in different tissues. These results underscore the importance of T cells, which produce IFN-γ in the lungs during the early stage of infection, in providing early protection against cryptococcal infection through CARD9 signaling.
Collapse
Grants
- 18H02851, 21H02965 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 19K17920, 21K16314 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- JP19jm0210073, JP20jm0210073, JP21jm0210073 Japan Agency for Medical Research and Development (AMED)
- ID-014 MSD Life Science Foundation, Public Interest Incorporated Foundation (SD Life Science Foundation)
- 20-02, 21-04 medical mycology research center, chiba university
Collapse
Affiliation(s)
- Anna Miyahara
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Aya Umeki
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Ko Sato
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Toshiki Nomura
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hideki Yamamoto
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Tomomitsu Miyasaka
- Center for Medical Education, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Daiki Tanno
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Ikumi Matsumoto
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Tong Zong
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Takafumi Kagesawa
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Akiho Oniyama
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Kotone Kawamura
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Xiaoliang Yuan
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Rin Yokoyama
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yuki Kitai
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Emi Kanno
- Department of Translational Science for Nursing, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hiromasa Tanno
- Department of Translational Science for Nursing, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hiromitsu Hara
- Department of Immunology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, Japan
| | - Shinobu Saijo
- Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, Japan
| | - Yoichiro Iwakura
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Keiko Ishii
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Kazuyoshi Kawakami
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
8
|
Zhang J, Yao Z. Immune cell trafficking: a novel perspective on the gut-skin axis. Inflamm Regen 2024; 44:21. [PMID: 38654394 DOI: 10.1186/s41232-024-00334-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024] Open
Abstract
Immune cell trafficking, an essential mechanism for maintaining immunological homeostasis and mounting effective responses to infections, operates under a stringent regulatory framework. Recent advances have shed light on the perturbation of cell migration patterns, highlighting how such disturbances can propagate inflammatory diseases from their origin to distal organs. This review collates and discusses current evidence that demonstrates atypical communication between the gut and skin, which are conventionally viewed as distinct immunological spheres, in the milieu of inflammation. We focus on the aberrant, reciprocal translocation of immune cells along the gut-skin axis as a pivotal factor linking intestinal and dermatological inflammatory conditions. Recognizing that the translation of these findings into clinical practices is nascent, we suggest that therapeutic strategies aimed at modulating the axis may offer substantial benefits in mitigating the widespread impact of inflammatory diseases.
Collapse
Affiliation(s)
- Jiayan Zhang
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhirong Yao
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
9
|
Jiang C, Chao CC, Li J, Ge X, Shen A, Jucaud V, Cheng C, Shen X. Tissue-resident memory T cell signatures from single-cell analysis associated with better melanoma prognosis. iScience 2024; 27:109277. [PMID: 38455971 PMCID: PMC10918229 DOI: 10.1016/j.isci.2024.109277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/05/2024] [Accepted: 02/15/2024] [Indexed: 03/09/2024] Open
Abstract
Tissue-resident memory T cells (TRM) are a specialized T cell population residing in peripheral tissues. The presence and potential impact of TRM in the tumor immune microenvironment (TIME) remain to be elucidated. Here, we systematically investigated the relationship between TRM and melanoma TIME based on multiple clinical single-cell RNA-seq datasets and developed signatures indicative of TRM infiltration. TRM infiltration is associated with longer overall survival and abundance of T cells, NK cells, M1 macrophages, and memory B cells in the TIME. A 22-gene TRM-derived risk score was further developed to effectively classify patients into low- and high-risk categories, distinguishing overall survival and immune activation, particularly in T cell-mediated responses. Altogether, our analysis suggests that TRM abundance is associated with melanoma TIME activation and patient survival, and the TRM-based machine learning model can potentially predict prognosis in melanoma patients.
Collapse
Affiliation(s)
- Chongming Jiang
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Cheng-Chi Chao
- Department of Pipeline Development, Biomap, Inc, San Francisco, CA, USA
| | - Jianrong Li
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Xin Ge
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Aidan Shen
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Chao Cheng
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Xiling Shen
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
- Xilis, Inc., Durham, NC 27713, USA
| |
Collapse
|
10
|
Xu H, Zhou R, Chen Z. Tissue-Resident Memory T Cell: Ontogenetic Cellular Mechanism and Clinical Translation. Clin Exp Immunol 2023; 214:249-259. [PMID: 37586053 PMCID: PMC10719502 DOI: 10.1093/cei/uxad090] [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: 03/08/2023] [Revised: 06/22/2023] [Accepted: 08/15/2023] [Indexed: 08/18/2023] Open
Abstract
Mounting evidence has indicated the essential role of tissue-resident memory T (TRM) cells for frontline protection against viral infection and for cancer immune surveillance (Mueller SN, Mackay LK. Tissue-resident memory T cells: local specialists in immune defense. Nat Rev Immunol 2016, 16, 79-89. doi:10.1038/nri.2015.3.). TRM cells are transcriptionally, phenotypically, and functionally distinct from circulating memory T (Tcirm) cells. It is necessary to understand the unique ontogenetic mechanism, migratory regulation, and biological function of TRM cells. In this review, we discuss recent insights into cellular mechanisms and discrete responsiveness in different tissue microenvironments underlying TRM cell development. We also emphasize the translational potential of TRM cells by focusing on their establishment in association with improved protection in mucosal tissues against various types of diseases and effective strategies for eliciting TRM cells in both pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Haoran Xu
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong; Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong; Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Runhong Zhou
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong; Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong; Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Zhiwei Chen
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong; Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong; Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- State Key Laboratory for Emerging Infectious Diseases, University of Hong Kong; Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| |
Collapse
|
11
|
Feng M, Liu X, Hao X, Ren Y, Dong G, Tian J, Wang Y, Du L, Wang Y, Wang C. Fatty Acids Support the Fitness and Functionality of Tumor-Resident CD8+ T Cells by Maintaining SCML4 Expression. Cancer Res 2023; 83:3368-3384. [PMID: 37610617 DOI: 10.1158/0008-5472.can-23-0287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 06/08/2023] [Accepted: 08/21/2023] [Indexed: 08/24/2023]
Abstract
CD8+ tissue-resident memory T (Trm) cells and tumor-infiltrating lymphocytes (TIL) regulate tumor immunity and immune surveillance. Characterization of Trm cells and TILs could help identify potential strategies to boost antitumor immunity. Here, we found that the transcription factor SCML4 was required for the progression and polyfunctionality of Trm cells and was associated with a better prognosis in patients with cancer. Moreover, SCML4 maintained multiple functions of TILs. Increased expression of SCML4 in CD8+ cells significantly reduced the growth of multiple types of tumors in mice, while deletion of SCML4 reduced antitumor immunity and promoted CD8+ T-cell exhaustion. Mechanistically, SCML4 recruited the HBO1-BRPF2-ING4 complex to reprogram the expression of T cell-specific genes, thereby enhancing the survival and effector functions of Trm cells and TILs. SCML4 expression was promoted by fatty acid metabolism through mTOR-IRF4-PRDM1 signaling, and fatty acid metabolism-induced epigenetic modifications that promoted tissue-resident and multifunctional gene expression in Trm cells and TILs. SCML4 increased the therapeutic effect of anti-PD-1 treatment by elevating the expression of effector molecules in TILs and inhibiting the apoptosis of TILs, which could be further enhanced by adding an inhibitor of H3K14ac deacetylation. These results provide a mechanistic perspective of functional regulation of tumor-localized Trm cells and TILs and identify an important activation target for tumor immunotherapy. SIGNIFICANCE SCML4 upregulation in CD8+ Trm cells and tumor-infiltrating lymphocytes induced by fatty acid metabolism enhances antitumor immune responses, providing an immunometabolic axis to target for cancer treatment. See related commentary by Chakraborty et al., p. 3321.
Collapse
Affiliation(s)
- Maoxiao Feng
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Clinical Laboratory, Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiaoyan Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiaodong Hao
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yidan Ren
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Guoying Dong
- Department of Anatomy and Key Laboratory of Experimental Teratology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jie Tian
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yuli Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yunshan Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
12
|
Ibidapo-Obe O, Bruns T. Tissue-resident and innate-like T cells in patients with advanced chronic liver disease. JHEP Rep 2023; 5:100812. [PMID: 37691689 PMCID: PMC10485156 DOI: 10.1016/j.jhepr.2023.100812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 09/12/2023] Open
Abstract
Chronic liver disease results from the orchestrated interplay of components of innate and adaptive immunity in response to liver tissue damage. Recruitment, positioning, and activation of immune cells can contribute to hepatic cell death, inflammation, and fibrogenesis. With disease progression and increasing portal pressure, repeated translocation of bacterial components from the intestinal lumen through the epithelial and vascular barriers leads to persistent mucosal, hepatic, and systemic inflammation which contributes to tissue damage, immune dysfunction, and microbial infection. It is increasingly recognised that innate-like and adaptive T-cell subsets located in the liver, mucosal surfaces, and body cavities play a critical role in the progression of advanced liver disease and inflammatory complications of cirrhosis. Mucosal-associated invariant T cells, natural killer T cells, γδ T cells, and tissue-resident memory T cells in the gut, liver, and ascitic fluid share certain characteristic features, which include that they recognise microbial products, tissue alarmins, cytokines, and stress ligands in tissues, and perform effector functions in chronic liver disease. This review highlights recent advances in the comprehension of human tissue-resident and unconventional T-cell populations and discusses the mechanisms by which they contribute to inflammation, fibrosis, immunosuppression, and antimicrobial surveillance in patients with cirrhosis. Understanding the complex interactions of immune cells in different compartments and their contribution to disease progression will provide further insights for effective diagnostic interventions and novel immunomodulatory strategies in patients with advanced chronic liver disease.
Collapse
Affiliation(s)
- Oluwatomi Ibidapo-Obe
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Tony Bruns
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
13
|
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
|
14
|
Davé V, Richert-Spuhler LE, Arkatkar T, Warrier L, Pholsena T, Johnston C, Schiffer JT, Prlic M, Lund JM. Recurrent infection transiently expands human tissue T cells while maintaining long-term homeostasis. J Exp Med 2023; 220:e20210692. [PMID: 37314481 PMCID: PMC10267593 DOI: 10.1084/jem.20210692] [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: 03/29/2021] [Revised: 03/13/2023] [Accepted: 05/15/2023] [Indexed: 06/15/2023] Open
Abstract
Chronic viral infections are known to lead to T cell exhaustion or dysfunction. However, it remains unclear if antigen exposure episodes from periodic viral reactivation, such as herpes simplex virus type-2 (HSV-2) recrudescence, are sufficient to induce T cell dysfunction, particularly in the context of a tissue-specific localized, rather than a systemic, infection. We designed and implemented a stringent clinical surveillance protocol to longitudinally track both viral shedding and in situ tissue immune responses in a cohort of HSV+ volunteers that agreed to avoid using anti-viral therapy for the course of this study. Comparing lesion to control skin biopsies, we found that tissue T cells expanded immediately after reactivation, and then returned numerically and phenotypically to steady state. T cell responses appeared to be driven at least in part by migration of circulating T cells to the infected tissue. Our data indicate that tissue T cells are stably maintained in response to HSV reactivation, resembling a series of acute recall responses.
Collapse
Affiliation(s)
- Veronica Davé
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Global Health, Graduate Program in Pathobiology, University of Washington, Seattle, WA, USA
| | - Laura E. Richert-Spuhler
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Tanvi Arkatkar
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Global Health, Graduate Program in Pathobiology, University of Washington, Seattle, WA, USA
| | - Lakshmi Warrier
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Global Health, Graduate Program in Pathobiology, University of Washington, Seattle, WA, USA
| | | | - Christine Johnston
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Joshua T. Schiffer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Martin Prlic
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Global Health, Graduate Program in Pathobiology, University of Washington, Seattle, WA, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Jennifer M. Lund
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Global Health, Graduate Program in Pathobiology, University of Washington, Seattle, WA, USA
| |
Collapse
|
15
|
Li L, Tian Y. The role of metabolic reprogramming of tumor-associated macrophages in shaping the immunosuppressive tumor microenvironment. Biomed Pharmacother 2023; 161:114504. [PMID: 37002579 DOI: 10.1016/j.biopha.2023.114504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/04/2023] [Accepted: 03/07/2023] [Indexed: 03/15/2023] Open
Abstract
Macrophages are potent immune effector cells in innate immunity and exert dual-effects in the tumor microenvironment (TME). Tumor-associated macrophages (TAMs) make up a significant portion of TME immune cells. Similar to M1/M2 macrophages, TAMs are also highly plastic, and their functions are regulated by cytokines, chemokines and other factors in the TME. The metabolic changes in TAMs are significantly associated with polarization towards a protumour or antitumour phenotype. The metabolites generated via TAM metabolic reprogramming in turn promote tumor progression and immune tolerance. In this review, we explore the metabolic reprogramming of TAMs in terms of energy, amino acid and fatty acid metabolism and the potential roles of these changes in immune suppression.
Collapse
Affiliation(s)
- Lunxu Li
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
16
|
Zhang N, Yao H, Zhang Z, Li Z, Chen X, Zhao Y, Ju R, He J, Pan H, Liu X, Lv Y. Ongoing involvers and promising therapeutic targets of hepatic fibrosis: The hepatic immune microenvironment. Front Immunol 2023; 14:1131588. [PMID: 36875101 PMCID: PMC9978172 DOI: 10.3389/fimmu.2023.1131588] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 02/02/2023] [Indexed: 02/18/2023] Open
Abstract
Hepatic fibrosis is often secondary to chronic inflammatory liver injury. During the development of hepatic fibrosis, the damaged hepatocytes and activated hepatic stellate cells (HSCs) caused by the pathogenic injury could secrete a variety of cytokines and chemokines, which will chemotactic innate and adaptive immune cells of liver tissue and peripheral circulation infiltrating into the injury site, mediating the immune response against injury and promoting tissue reparation. However, the continuous release of persistent injurious stimulus-induced inflammatory cytokines will promote HSCs-mediated fibrous tissue hyperproliferation and excessive repair, which will cause hepatic fibrosis development and progression to cirrhosis even liver cancer. And the activated HSCs can secrete various cytokines and chemokines, which directly interact with immune cells and actively participate in liver disease progression. Therefore, analyzing the changes in local immune homeostasis caused by immune response under different pathological states will greatly enrich our understanding of liver diseases' reversal, chronicity, progression, and even deterioration of liver cancer. In this review, we summarized the critical components of the hepatic immune microenvironment (HIME), different sub-type immune cells, and their released cytokines, according to their effect on the development of progression of hepatic fibrosis. And we also reviewed and analyzed the specific changes and the related mechanisms of the immune microenvironment in different chronic liver diseases.Moreover, we retrospectively analyzed whether the progression of hepatic fibrosis could be alleviated by modulating the HIME.We aimed to elucidate the pathogenesis of hepatic fibrosis and provide the possibility for exploring the therapeutic targets for hepatic fibrosis.
Collapse
Affiliation(s)
- Nana Zhang
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Huimin Yao
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Zhixuan Zhang
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Zhuoqun Li
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xue Chen
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yan Zhao
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ran Ju
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jiayi He
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Heli Pan
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaoli Liu
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yi Lv
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
17
|
Lim YS, Shin H. CD8 tissue-resident memory T cells bridge the gap between humoral and cell-mediated immunity. Mucosal Immunol 2023; 16:3-4. [PMID: 36640865 DOI: 10.1016/j.mucimm.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/13/2023]
Abstract
Tissue-resident memory T (TRM) cells exert antiviral effects in situ using a variety of cell-intrinsic mechanisms. In this issue of Mucosal Immunology, Rosato and colleagues demonstrate that mucosal CD8 TRM cells can also facilitate the extravasation of antibodies with neutralizing capacities by modulating transcytotic receptors and vascular permeability, thus revealing a new mechanism by which TRM cells modulate humoral responses and protect the host from incoming pathogens.
Collapse
Affiliation(s)
- Ying Shiang Lim
- Department of Medicine/Division of Infectious Disease, Washington University School of Medicine, St Louis, Missouri, USA
| | - Haina Shin
- Department of Medicine/Division of Infectious Disease, Washington University School of Medicine, St Louis, Missouri, USA.
| |
Collapse
|
18
|
Wiarda JE, Loving CL. Intraepithelial lymphocytes in the pig intestine: T cell and innate lymphoid cell contributions to intestinal barrier immunity. Front Immunol 2022; 13:1048708. [PMID: 36569897 PMCID: PMC9772029 DOI: 10.3389/fimmu.2022.1048708] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
Abstract
Intraepithelial lymphocytes (IELs) include T cells and innate lymphoid cells that are important mediators of intestinal immunity and barrier defense, yet most knowledge of IELs is derived from the study of humans and rodent models. Pigs are an important global food source and promising biomedical model, yet relatively little is known about IELs in the porcine intestine, especially during formative ages of intestinal development. Due to the biological significance of IELs, global importance of pig health, and potential of early life events to influence IELs, we collate current knowledge of porcine IEL functional and phenotypic maturation in the context of the developing intestinal tract and outline areas where further research is needed. Based on available findings, we formulate probable implications of IELs on intestinal and overall health outcomes and highlight key findings in relation to human IELs to emphasize potential applicability of pigs as a biomedical model for intestinal IEL research. Review of current literature suggests the study of porcine intestinal IELs as an exciting research frontier with dual application for betterment of animal and human health.
Collapse
Affiliation(s)
- Jayne E. Wiarda
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, United States,Immunobiology Graduate Program, Iowa State University, Ames, IA, United States,Department of Veterinary Microbiology and Preventative Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Crystal L. Loving
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, United States,Immunobiology Graduate Program, Iowa State University, Ames, IA, United States,*Correspondence: Crystal L. Loving,
| |
Collapse
|
19
|
Sun M, Shi W, Wu Y, He Z, Sun J, Cai S, Luo Q. Immunogenic Nanovesicle-Tandem-Augmented Chemoimmunotherapy via Efficient Cancer-Homing Delivery and Optimized Ordinal-Interval Regime. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 10:e2205247. [PMID: 36453573 PMCID: PMC9811449 DOI: 10.1002/advs.202205247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/27/2022] [Indexed: 06/17/2023]
Abstract
The strategy of combining immune checkpoint inhibitors (ICIs) with anthracycline is recommended by clinical guidelines for the standard-of-care treatment of triple-negative breast cancer (TNBC). Nevertheless, several fundamental clinical principles are yet to be elucidated to achieve a great therapeutic effect, including cancer-homing delivery efficiency and ordinal-interval regime. Tumor-derived extracellular vesicles (TDEVs), as vectors for intratumoral intercellular communication, can encapsulate therapeutic agents and home tumors. However, PD-L1 overexpression in TDEVs leads to systemic immunosuppression during in vivo circulation, ultimately inhibiting intratumoral T activity. In this study, CRISPR/Cas9-edited Pd-l1KO TDEV-fusogenic anthracycline doxorubicin (DOX) liposomes with high drug encapsulation (97%) are fabricated, which homologously deliver DOX to breast cancer cells to intensify the immunogenic response and induce PD-L1 overexpression in the tumor. By setting the stage for sensitizing tumors to ICIs, sequential treatment with disulfide-linked PD1-cross-anchored TDEVs nanogels at one-day interval could sustainably release PD1 in the tumor, triggering a high proportion of effector T cell-mediated destruction of orthotopic and metastatic tumors without off-target side effects in the 4T1-bearing TNBC mouse model. Such a TDEV-tandem-augmented chemoimmunotherapeutic strategy with efficient cancer-homing delivery capacity and optimized ordinal-interval regime provides a solid foundation for developing chemoimmunotherapeutic formulations for TNBC therapy at the clinical level.
Collapse
Affiliation(s)
- Mengchi Sun
- School of PharmacyShenyang Pharmaceutical UniversityShenyangLiaoning110016P. R. China
| | - Wen Shi
- Department of PharmacyThe First Hospital of China Medical UniversityShenyangLiaoning110001P. R. China
| | - Yuxia Wu
- Department of PharmacyThe First Hospital of China Medical UniversityShenyangLiaoning110001P. R. China
| | - Zhonggui He
- Wuya College of InnovationShenyang Pharmaceutical UniversityShenyangLiaoning110016P. R. China
| | - Jin Sun
- Wuya College of InnovationShenyang Pharmaceutical UniversityShenyangLiaoning110016P. R. China
| | - Shuang Cai
- Department of PharmacyThe First Hospital of China Medical UniversityShenyangLiaoning110001P. R. China
| | - Qiuhua Luo
- Department of PharmacyThe First Hospital of China Medical UniversityShenyangLiaoning110001P. R. China
| |
Collapse
|
20
|
Suryadevara N, Kumar A, Ye X, Rogers M, Williams JV, Wilson JT, Karijolich J, Joyce S. A molecular signature of lung-resident CD8 + T cells elicited by subunit vaccination. Sci Rep 2022; 12:19101. [PMID: 36351985 PMCID: PMC9645351 DOI: 10.1038/s41598-022-21620-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 09/29/2022] [Indexed: 11/10/2022] Open
Abstract
Natural infection as well as vaccination with live or attenuated viruses elicit tissue resident, CD8+ memory T cell (Trm) response. Trm cells so elicited act quickly upon reencounter with the priming agent to protect the host. These Trm cells express a unique molecular signature driven by the master regulators-Runx3 and Hobit. We previously reported that intranasal instillation of a subunit vaccine in a prime boost vaccination regimen installed quick-acting, CD8+ Trm cells in the lungs that protected against lethal vaccinia virus challenge. It remains unexplored whether CD8+ Trm responses so elicited are driven by a similar molecular signature as those elicited by microbes in a real infection or by live, attenuated pathogens in conventional vaccination. We found that distinct molecular signatures distinguished subunit vaccine-elicited lung interstitial CD8+ Trm cells from subunit vaccine-elicited CD8+ effector memory and splenic memory T cells. Nonetheless, the transcriptome signature of subunit vaccine elicited CD8+ Trm resembled those elicited by virus infection or vaccination. Clues to the basis of tissue residence and function of vaccine specific CD8+ Trm cells were found in transcripts that code for chemokines and chemokine receptors, purinergic receptors, and adhesins when compared to CD8+ effector and splenic memory T cells. Our findings inform the utility of protein-based subunit vaccination for installing CD8+ Trm cells in the lungs to protect against respiratory infectious diseases that plague humankind.
Collapse
Affiliation(s)
- Naveenchandra Suryadevara
- Department of Veterans Affairs, Tennessee Valley Healthcare Center, Nashville, TN, 37212, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Amrendra Kumar
- Department of Veterans Affairs, Tennessee Valley Healthcare Center, Nashville, TN, 37212, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Xiang Ye
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Meredith Rogers
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Paediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA
| | - John V Williams
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Paediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA
- Institute for Infection, Immunity, and Inflammation in Children (i4Kids), Pittsburgh, PA, 15224, USA
| | - John T Wilson
- Department of Chemical and Biomolecular Engineering and Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - John Karijolich
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Sebastian Joyce
- Department of Veterans Affairs, Tennessee Valley Healthcare Center, Nashville, TN, 37212, USA.
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| |
Collapse
|
21
|
Differential Homing Receptor Profiles of Lymphocytes Induced by Attenuated versus Live Plasmodium falciparum Sporozoites. Vaccines (Basel) 2022; 10:vaccines10101768. [PMID: 36298634 PMCID: PMC9611729 DOI: 10.3390/vaccines10101768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/02/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022] Open
Abstract
The onset of an adaptive immune response provides the signals required for differentiation of antigen-specific lymphocytes into effector cells and imprinting of these cells for re-circulation to the most appropriate anatomical site (i.e., homing). Lymphocyte homing is governed by the expression of tissue-specific lymphocyte homing receptors that bind to unique tissue-specific ligands on endothelial cells. In this study, a whole-parasite malaria vaccine (radiation-attenuated sporozoites (RAS)) was used as a model system to establish homing receptor signatures induced by the parasite delivered through mosquito bite to provide a benchmark of desirable homing receptors for malaria vaccine developers. This immunization regimen resulted in the priming of antigen-specific B cells and CD8+ T cells for homing primarily to the skin and T/B cell compartments of secondary lymphoid organs. Infection with live sporozoites, however, triggers the upregulation of homing receptor for the liver and the skin, demonstrating that there is a difference in the signal provided by attenuated vs. live sporozoites. This is the first report on imprinting of homing routes by Plasmodium sporozoites and, surprisingly, it also points to additional, yet to be identified, signals provided by live parasites that prime lymphocytes for homing to the liver. The data also demonstrate the utility of this method for assessing the potential of vaccine formulations to direct antigen-specific lymphocytes to the most relevant anatomical site, thus potentially impacting vaccine efficacy.
Collapse
|
22
|
Li Y, You Z, Tang R, Ma X. Tissue-resident memory T cells in chronic liver diseases: Phenotype, development and function. Front Immunol 2022; 13:967055. [PMID: 36172356 PMCID: PMC9511135 DOI: 10.3389/fimmu.2022.967055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/22/2022] [Indexed: 12/02/2022] Open
Abstract
Tissue-resident memory (TRM) T cells are a unique subset of memory T cells that are critical for the first line of defense against pathogens or antigens in peripheral non-lymphoid tissues such as liver, gut, and skin. Generally, TRM cells are well adapted to the local environment in a tissue-specific manner and typically do not circulate but persist in tissues, distinguishing them from other memory T cell lineages. There is strong evidence that liver TRM cells provide a robust adaptive immune response to potential threats. Indeed, the potent effector function of hepatic TRM cells makes it essential for chronic liver diseases, including viral and parasite infection, autoimmune liver diseases (AILD), nonalcoholic fatty liver disease (NAFLD), hepatocellular carcinoma (HCC) and liver transplantation. Manipulation of hepatic TRM cells might provide novel promising strategies for precision immunotherapy of chronic liver diseases. Here, we provide insights into the phenotype of hepatic TRM cells through surface markers, transcriptional profiles and effector functions, discuss the development of hepatic TRM cells in terms of cellular origin and factors affecting their development, analyze the role of hepatic TRM cells in chronic liver diseases, as well as share our perspectives on the current status of hepatic TRM cell research.
Collapse
|
23
|
Borges ÁH, Follmann F, Dietrich J. Chlamydia trachomatis vaccine development - a view on the current challenges and how to move forward. Expert Rev Vaccines 2022; 21:1555-1567. [PMID: 36004386 DOI: 10.1080/14760584.2022.2117694] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Chlamydia trachomatis is the most common sexually transmitted bacterial pathogen in the world. A licensed vaccine is not yet available, but the first vaccines have entered clinical trials. AREAS COVERED : We describe the progress that has been made in our understanding of the type of immunity that a protective vaccine should induce, and the challenges that vaccine developers face. We also focus on the clinical development of a chlamydia vaccine. The first chlamydia vaccine candidate has now been tested in a clinical phase-I trial, and another phase-I trial is currently running. We discuss what it will take to continue this development and what future trial setups could look like. EXPERT OPINION The chlamydia field is coming of age and the first phase I clinical trial of a C. trachomatis vaccine has been successfully completed. We expect and hope that this will motivate various stakeholders to support further development of chlamydia vaccines in humans.
Collapse
Affiliation(s)
- Álvaro H Borges
- Statens Serum Institut, Department of Infectious Diseases Immunology, Kobenhavn, 2300 Denmark
| | | | - Jes Dietrich
- Statens Serum Institut, Department of Infectious Diseases Immunology, Kobenhavn, 2300 Denmark
| |
Collapse
|
24
|
Gary EN, Tursi NJ, Warner B, Parzych EM, Ali AR, Frase D, Moffat E, Embury-Hyatt C, Smith TRF, Broderick KE, Humeau L, Kobasa D, Patel A, Kulp DW, Weiner DB. Mucosal chemokine adjuvant enhances synDNA vaccine-mediated responses to SARS-CoV-2 and provides heterologous protection in vivo. Cell Rep Med 2022; 3:100693. [PMID: 35839767 PMCID: PMC9237025 DOI: 10.1016/j.xcrm.2022.100693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/16/2022] [Accepted: 06/23/2022] [Indexed: 11/28/2022]
Abstract
The global coronavirus disease 2019 (COVID-19) pandemic has claimed more than 5 million lives. Emerging variants of concern (VOCs) continually challenge viral control. Directing vaccine-induced humoral and cell-mediated responses to mucosal surfaces may enhance vaccine efficacy. Here we investigate the immunogenicity and protective efficacy of optimized synthetic DNA plasmids encoding wild-type severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein (pS) co-formulated with the plasmid-encoded mucosal chemokine cutaneous T cell-attracting chemokine (pCTACK; CCL27). pCTACK-co-immunized animals exhibit increased spike-specific antibodies at the mucosal surface and increased frequencies of interferon gamma (IFNγ)+ CD8+ T cells in the respiratory mucosa. pCTACK co-immunization confers 100% protection from heterologous Delta VOC challenge. This study shows that mucosal chemokine adjuvants can direct vaccine-induced responses to specific immunological sites and have significant effects on heterologous challenge. Further study of this unique chemokine-adjuvanted vaccine approach in the context of SARS-CoV-2 vaccines is likely important.
Collapse
Affiliation(s)
- Ebony N Gary
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Nicholas J Tursi
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bryce Warner
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Elizabeth M Parzych
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Ali R Ali
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Drew Frase
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Estella Moffat
- National Center for Foreign Animal Disease (NCFAD), Canadian Food Inspection Agency, Winnipeg, MB, Canada
| | - Carissa Embury-Hyatt
- National Center for Foreign Animal Disease (NCFAD), Canadian Food Inspection Agency, Winnipeg, MB, Canada
| | | | | | | | - Darwyn Kobasa
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada; Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Ami Patel
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Daniel W Kulp
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - David B Weiner
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA.
| |
Collapse
|
25
|
Koyaman-Nasu R, Wang Y, Hasegawa I, Endo Y, Nakayama T, Kimura MY. The cellular and molecular basis of CD69 function in anti-tumor immunity. Int Immunol 2022; 34:555-561. [PMID: 35689672 DOI: 10.1093/intimm/dxac024] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/09/2022] [Indexed: 12/16/2022] Open
Abstract
Cancer immunotherapy utilizes our immune system to attack cancer cells and is an extremely promising strategy for cancer treatment. Although immune-checkpoint blockade, such as anti-PD-1 antibody (Ab), has demonstrated significant enhancement of anti-tumor immunity and has induced notable clinical outcomes, its response rates remain low, and adverse effects are always a matter of concern; therefore, new targets for cancer immunotherapy are always desired. In this situation, new concepts are needed to fuel the investigation of new target molecules for cancer immunotherapy. We propose that CD69 is one such target molecule. CD69 is known to be an activation marker of leukocytes and is also considered a crucial regulator of various immune responses through its interacting proteins. CD69 promotes T cell retention in lymphoid tissues via sphingosine-1-phosphate receptor 1 (S1P1) internalization and also plays roles in the pathogenesis of inflammatory disorders through interacting with its functional ligands Myl9/12 (myosin light chains 9, 12a and 12b). In anti-tumor immunity, CD69 is known to be expressed on T cells in the tumor microenvironment (TME) and tumor-draining lymph nodes (TDLNs). We revealed that CD69 negatively regulates the effector function of intratumoral T cells and importantly controls the 'exhaustion' of CD8 T cells. In addition, we and others showed that either CD69 deficiency or the administration of anti-CD69 monoclonal antibody enhances anti-tumor immunity. Thus, CD69 is an attractive target for cancer immunotherapy.
Collapse
Affiliation(s)
- Ryo Koyaman-Nasu
- Department of Experimental Immunology, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Yangsong Wang
- Department of Experimental Immunology, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Ichita Hasegawa
- Department of Experimental Immunology, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Yukihiro Endo
- Department of Experimental Immunology, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan.,AMED-CREST, AMED, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Motoko Y Kimura
- Department of Experimental Immunology, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| |
Collapse
|
26
|
Szczęsny G, Kopec M, Politis DJ, Kowalewski ZL, Łazarski A, Szolc T. A Review on Biomaterials for Orthopaedic Surgery and Traumatology: From Past to Present. MATERIALS 2022; 15:ma15103622. [PMID: 35629649 PMCID: PMC9145924 DOI: 10.3390/ma15103622] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 02/04/2023]
Abstract
The principal features essential for the success of an orthopaedic implant are its shape, dimensional accuracy, and adequate mechanical properties. Unlike other manufactured products, chemical stability and toxicity are of increased importance due to the need for biocompatibility over an implants life which could span several years. Thus, the combination of mechanical and biological properties determines the clinical usefulness of biomaterials in orthopaedic and musculoskeletal trauma surgery. Materials commonly used for these applications include stainless steel, cobalt-chromium and titanium alloys, ceramics, polyethylene, and poly(methyl methacrylate) (PMMA) bone cement. This study reviews the properties of commonly used materials and the advantages and disadvantages of each, with special emphasis on the sensitivity, toxicity, irritancy, and possible mutagenic and teratogenic capabilities. In addition, the production and final finishing processes of implants are discussed. Finally, potential directions for future implant development are discussed, with an emphasis on developing advanced personalised implants, according to a patient’s stature and physical requirements.
Collapse
Affiliation(s)
- Grzegorz Szczęsny
- Department of Orthopaedic Surgery and Traumatology, Medical University, 4 Lindleya Str., 02-005 Warsaw, Poland; (G.S.); (A.Ł.)
| | - Mateusz Kopec
- Institute of Fundamental Technological Research, Polish Academy of Sciences, 5B Pawińskiego Str., 02-106 Warsaw, Poland; (Z.L.K.); (T.S.)
- Correspondence:
| | - Denis J. Politis
- Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 20537, Cyprus;
| | - Zbigniew L. Kowalewski
- Institute of Fundamental Technological Research, Polish Academy of Sciences, 5B Pawińskiego Str., 02-106 Warsaw, Poland; (Z.L.K.); (T.S.)
| | - Adam Łazarski
- Department of Orthopaedic Surgery and Traumatology, Medical University, 4 Lindleya Str., 02-005 Warsaw, Poland; (G.S.); (A.Ł.)
| | - Tomasz Szolc
- Institute of Fundamental Technological Research, Polish Academy of Sciences, 5B Pawińskiego Str., 02-106 Warsaw, Poland; (Z.L.K.); (T.S.)
| |
Collapse
|
27
|
Li L, Tang W, Zhang Y, Jia M, Wang L, Li Q, Han Q, Peng X, Xie Y, Wu J, Wang Z, Zhen J, Wang X, Liu M, Sun Y, Zhang C, Yi F. Targeting tissue-resident memory CD8 + T cells in the kidney is a potential therapeutic strategy to ameliorate podocyte injury and glomerulosclerosis. Mol Ther 2022; 30:2746-2759. [PMID: 35514086 PMCID: PMC9372318 DOI: 10.1016/j.ymthe.2022.04.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/21/2022] [Accepted: 04/29/2022] [Indexed: 12/01/2022] Open
Abstract
Although tissue-resident memory T (TRM) cells, a recently identified non-circulating memory T cell population, play a crucial role in mediating local immune responses and protect against pathogens upon local reinfection, the composition, effector function, and specificity of TRM cells in the kidney and their relevance for chronic kidney disease remain unknown. In this study, we found that renal tissue displayed high abundance of tissue-resident lymphocytes and the proportion of CD8+ TRM cells was significantly increased in the kidney from patients and mice with focal segmental glomerulosclerosis (FSGS), diabetic kidney disease (DKD) and lupus nephritis (LN). Mechanistically, IL-15 significantly promoted CD8+ TRM cell formation and activation, thereby promoting podocyte injury and glomerulosclerosis. Interestingly, Sparsentan, the dual angiotensin II (Ang II) receptor and endothelin Type A receptor antagonist, can also reduce TRM cell responses by intervening IL-15 signaling, exploring its new pharmacological functions. Mechanistically, Sparsentan inhibited Ang II or endothelin-1 (ET-1)-mediated IL-15 signaling, thereby further regulating renal CD8+ TRM cell fates. Collectively, our studies provide direct evidence for the pivotal role of renal CD8+ TRM cells in podocyte injury, and further strengthen that targeting TRM cells represents a novel therapeutic strategy for patients with glomerular diseases.
Collapse
Affiliation(s)
- Liang Li
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Wei Tang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Yan Zhang
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Meng Jia
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Limei Wang
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China, 250012
| | - Quanxin Li
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Qingsheng Han
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Xiuping Peng
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Yusheng Xie
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Jichao Wu
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Ziying Wang
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Junhui Zhen
- Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Xiaojie Wang
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Min Liu
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Yu Sun
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China, 430022
| | - Fan Yi
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012.
| |
Collapse
|
28
|
Ionizable Lipid Nanoparticles Enhanced the Synergistic Adjuvant Effect of CpG ODNs and QS21 in a Varicella Zoster Virus Glycoprotein E Subunit Vaccine. Pharmaceutics 2022; 14:pharmaceutics14050973. [PMID: 35631559 PMCID: PMC9143440 DOI: 10.3390/pharmaceutics14050973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 12/10/2022] Open
Abstract
Varicella zoster virus (VZV) causes two diseases: varicella upon primary infection and herpes zoster when latent viruses in the sensory ganglia reactivate. While varicella vaccines depend on humoral immunity to prevent VZV infection, cell-mediated immunity (CMI), which plays a therapeutic role in the control or elimination of reactivated VZV in infected cells, is decisive for zoster vaccine efficacy. As one of the most abundant glycoproteins of VZV, conserved glycoprotein E (gE) is essential for viral replication and transmission between ganglion cells, thus making it an ideal target subunit vaccine antigen; gE has been successfully used in the herpes zoster vaccine ShingrixTM on the market. In this report, we found that ionizable lipid nanoparticles (LNPs) approved by the Food and Drug Administration (FDA) as vectors for coronavirus disease 2019 (COVID-19) mRNA vaccines could enhance the synergistic adjuvant effect of CpG oligodeoxynucleotides (CpG ODNs) and QS21 on VZV-gE, affecting both humoral immunity and CMI. Vaccines made with these LNPs showed promise as varicella vaccines without a potential risk of herpes zoster, which identifies them as a novel type of herpes zoster vaccine similar to ShingrixTM. All of the components in this LNP-CpG-QS21 adjuvant system were proven to be safe after mass vaccination, and the high proportion of cholesterol contained in the LNPs was helpful for limiting the cytotoxicity induced by QS21, which may lead to the development of a novel herpes zoster subunit vaccine for clinical application.
Collapse
|
29
|
Koelle DM, Dong L, Jing L, Laing KJ, Zhu J, Jin L, Selke S, Wald A, Varon D, Huang ML, Johnston C, Corey L, Posavad CM. HSV-2-Specific Human Female Reproductive Tract Tissue Resident Memory T Cells Recognize Diverse HSV Antigens. Front Immunol 2022; 13:867962. [PMID: 35432373 PMCID: PMC9009524 DOI: 10.3389/fimmu.2022.867962] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/07/2022] [Indexed: 01/05/2023] Open
Abstract
Antigen-specific TRM persist and protect against skin or female reproductive tract (FRT) HSV infection. As the pathogenesis of HSV differs between humans and model organisms, we focus on humans with well-characterized recurrent genital HSV-2 infection. Human CD8+ TRM persisting at sites of healed human HSV-2 lesions have an activated phenotype but it is unclear if TRM can be cultivated in vitro. We recovered HSV-specific TRM from genital skin and ectocervix biopsies, obtained after recovery from recurrent genital HSV-2, using ex vivo activation by viral antigen. Up to several percent of local T cells were HSV-reactive ex vivo. CD4 and CD8 T cell lines were up to 50% HSV-2-specific after sorting-based enrichment. CD8 TRM displayed HLA-restricted reactivity to specific HSV-2 peptides with high functional avidities. Reactivity to defined peptides persisted locally over several month and was quite subject-specific. CD4 TRM derived from biopsies, and from an extended set of cervical cytobrush specimens, also recognized diverse HSV-2 antigens and peptides. Overall we found that HSV-2-specific TRM are abundant in the FRT between episodes of recurrent genital herpes and maintain competency for expansion. Mucosal sites are accessible for clinical monitoring during immune interventions such as therapeutic vaccination.
Collapse
Affiliation(s)
- David M. Koelle
- Department of Medicine, University of Washington, Seattle, WA, United States
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Translational Research, Benaroya Research Institute, Seattle, WA, United States
| | - Lichun Dong
- Department of Medicine, University of Washington, Seattle, WA, United States
| | - Lichen Jing
- Department of Medicine, University of Washington, Seattle, WA, United States
| | - Kerry J. Laing
- Department of Medicine, University of Washington, Seattle, WA, United States
| | - Jia Zhu
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Lei Jin
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Stacy Selke
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Anna Wald
- Department of Medicine, University of Washington, Seattle, WA, United States
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
- Department of Epidemiology, University of Washington, Seattle, WA, United States
| | - Dana Varon
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Meei-Li Huang
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Christine Johnston
- Department of Medicine, University of Washington, Seattle, WA, United States
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Lawrence Corey
- Department of Medicine, University of Washington, Seattle, WA, United States
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Christine M. Posavad
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| |
Collapse
|
30
|
Fernández-Gallego N, Sánchez-Madrid F, Cibrian D. Role of AHR Ligands in Skin Homeostasis and Cutaneous Inflammation. Cells 2021; 10:cells10113176. [PMID: 34831399 PMCID: PMC8622815 DOI: 10.3390/cells10113176] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 02/07/2023] Open
Abstract
Aryl hydrocarbon receptor (AHR) is an important regulator of skin barrier function. It also controls immune-mediated skin responses. The AHR modulates various physiological functions by acting as a sensor that mediates environment–cell interactions, particularly during immune and inflammatory responses. Diverse experimental systems have been used to assess the AHR’s role in skin inflammation, including in vitro assays of keratinocyte stimulation and murine models of psoriasis and atopic dermatitis. Similar approaches have addressed the role of AHR ligands, e.g., TCDD, FICZ, and microbiota-derived metabolites, in skin homeostasis and pathology. Tapinarof is a novel AHR-modulating agent that inhibits skin inflammation and enhances skin barrier function. The topical application of tapinarof is being evaluated in clinical trials to treat psoriasis and atopic dermatitis. In the present review, we summarize the effects of natural and synthetic AHR ligands in keratinocytes and inflammatory cells, and their relevance in normal skin homeostasis and cutaneous inflammatory diseases.
Collapse
Affiliation(s)
- Nieves Fernández-Gallego
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), 28006 Madrid, Spain;
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Francisco Sánchez-Madrid
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), 28006 Madrid, Spain;
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (F.S.-M.); (D.C.)
| | - Danay Cibrian
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), 28006 Madrid, Spain;
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (F.S.-M.); (D.C.)
| |
Collapse
|
31
|
Balaji S, Cholan PK, Victor DJ. An emphasis of T-cell subsets as regulators of periodontal health and disease. J Clin Transl Res 2021; 7:648-656. [PMID: 34778595 PMCID: PMC8580519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 02/25/2021] [Accepted: 07/25/2021] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND The pathogenesis of complex diseases like periodontitis is moderated by the balance in immune inflammatory responses. T-lymphocytes are immune cells that descend from the bone marrow. Furthermore, they develop in the thymus playing an indispensable role in adaptive immune responses. The periodontal microenvironment allows differentiation of various groups of T-lymphocytes such as CD4+ (Th1/Th2/Th17/Treg/Tfh/Th9/T22), CD8+ cells, gamma-delta (γd) T cells, or memory cells based on the current regional cytokine milieu to secrete distinct cytokines and other molecules required for resolution of inflammation or result in progression of the disease based on interactions among various cells. AIM The dynamism of T-lymphocytes in the immunopathogenesis of periodontal diseases resulting in tissue destruction is established but the mechanisms of immunoregulation that underpins periodontal disease progression are cumbersome. This review aims to understand the distinct types of T cells and their effector functions with their portrayal in periodontal disease. RELEVANCE FOR PATIENTS This review gives valuable insights on the possibility of predicting periodontal disease progression, on the management and its prognosis by evaluating specific cytokines of destructive T-cell phenotype, and on the future perspectives of therapeutic modalities including ways of modulating host immune and inflammatory responses to establish periodontal homeostasis and areas of research.
Collapse
Affiliation(s)
- Saranya Balaji
- Department of Periodontics, SRM Dental College, Chennai, Tamil Nadu, India
| | - Priyanka K. Cholan
- Department of Periodontics, SRM Dental College, Chennai, Tamil Nadu, India
| | | |
Collapse
|
32
|
Assessment of the Tissue Resident Memory Cells in Lesional Skin of Patients with Psoriasis and in Healthy Skin of Healthy Volunteers. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182111251. [PMID: 34769769 PMCID: PMC8582754 DOI: 10.3390/ijerph182111251] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND In the course of plaque psoriasis, tissue resident memory cells (TRM) are responsible for the phenomenon of "immune memory" of lesions, i.e., the appearance of recurrences of lesions in the same location, as well as Koebner phenomenon. We present results determining the location and amount of TRM in psoriatic lesions in patients suffering from plaque psoriasis, as well as an analysis of the relationship between TRM markers expression and the duration and severity of the disease. METHODS TRM markers (CD4, CD8, CD103, CD69, CD49, CXCR6) and tissue expression of cytokines (IL-17, IL-22) in the lesional psoriatic skin of 32 patients compared with 10 healthy skin samples were evaluated by immunohistochemistry. RESULTS The presence of TRM markers in both the epidermis and skin with psoriatic eruptions was demonstrated in much higher amounts compared with the skin of healthy volunteers. A significant positive relationship was demonstrated between the expression of TRM markers in patients with plaque psoriasis and the duration of skin lesions. There was no relationship between the amount of TRM and the severity of plaque psoriasis. CONCLUSIONS A thorough understanding of the mechanisms responsible for the development and relapse of plaque psoriasis may contribute to the implementation of more effective therapies.
Collapse
|
33
|
Ichiki Y, Ueno M, Yanagi S, Kanasaki Y, Goto H, Fukuyama T, Mikami S, Nakanishi K, Ishida T. An analysis of the immunological tumor microenvironment of primary tumors and regional lymph nodes in squamous cell lung cancer. Transl Lung Cancer Res 2021; 10:3520-3537. [PMID: 34584854 PMCID: PMC8435388 DOI: 10.21037/tlcr-21-479] [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: 06/10/2021] [Accepted: 08/19/2021] [Indexed: 12/25/2022]
Abstract
Background Various immune cells that play a central role in antitumor immunity accumulate in primary tumors and regional lymph nodes. Such cellular accumulation and the molecular expression were analyzed to elucidate the immunological tumor microenvironment. Methods Fifty squamous cell lung cancer patients with complete resection were included. Resected specimens from primary lung tumors and regional lymph nodes were immunostained for immune-related molecules, such as CD8, CD103, major histocompatibility complex (MHC) class I, and programmed cell death protein ligand-1 (PD-L1), and the relationship between the prognosis and clinicopathological factors was retrospectively analyzed. Results Tumor-infiltrating lymphocytes and CD8+ lymphocytes, intratumoral and intrastromal CD103+ lymphocytes, tumor diameter, pathological T and N factors, and pathological stage were significant prognostic factors for the disease-specific survival (DSS) in a univariate analysis. In a multivariate analysis, intratumoral and intrastromal CD103+ lymphocytes and pathological T and N factors were independent prognostic factors of the DSS. Significant concordance was found between the PD-L1 expression of primary tumors and metastatic lymph nodes as well as among tumor-infiltrating lymphocytes, CD8+ lymphocytes and CD103+ lymphocytes. Infiltration of CD103+ lymphocytes into the tumor was significantly correlated with an increased PD-L1 expression of cancer cells in both primary tumors and reginal lymph node metastases. Both the intratumoral infiltration of CD103+ lymphocytes and PD-L1 expression of cancer cells were significantly higher in lymph node metastases than in primary tumors. Conclusions CD103+ lymphocyte infiltration in the primary tumor was shown to be strongly involved in the prognosis.
Collapse
Affiliation(s)
- Yoshinobu Ichiki
- Department of General Thoracic Surgery, National Hospital Organization, Saitama Hospital, Wako, Japan.,Second Department of Surgery, University of Occupational and Environmental Health, School of Medicine, Kitakyushu, Japan
| | - Mari Ueno
- Department of Diagnostic Pathology, National Hospital Organization, Saitama Hospital, Wako, Japan
| | - Shinya Yanagi
- Department of Diagnostic Pathology, National Hospital Organization, Saitama Hospital, Wako, Japan
| | - Yoshiro Kanasaki
- Department of General Thoracic Surgery, National Hospital Organization, Saitama Hospital, Wako, Japan
| | - Hidenori Goto
- Department of General Thoracic Surgery, National Hospital Organization, Saitama Hospital, Wako, Japan
| | - Takashi Fukuyama
- Division of Biomedical Research, Kitasato University Medical Center, Kitamoto, Japan
| | - Shuji Mikami
- Department of Diagnostic Pathology, National Hospital Organization, Saitama Hospital, Wako, Japan
| | - Kozo Nakanishi
- Department of General Thoracic Surgery, National Hospital Organization, Saitama Hospital, Wako, Japan
| | - Tsuyoshi Ishida
- Department of Diagnostic Pathology, National Hospital Organization, Saitama Hospital, Wako, Japan
| |
Collapse
|
34
|
Shukla A, Cano-Mejia J, Andricovich J, Burga RA, Sweeney EE, Fernandes R. An Engineered Prussian Blue Nanoparticles-based Nanoimmunotherapy Elicits Robust and Persistent Immunological Memory in a TH-MYCN Neuroblastoma Model. ADVANCED NANOBIOMED RESEARCH 2021; 1. [PMID: 34435194 DOI: 10.1002/anbr.202100021] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A combination therapy using Prussian blue nanoparticles (PBNP) as photothermal therapy (PTT) agents coated with CpG oligodeoxynucleotides, an immunologic adjuvant, as a nanoimmunotherapy (CpG-PBNP-PTT) for neuroblastoma (NB) is described. NB driven by MYCN amplification confers high risk and correlates with a dismal prognosis, accounting for the majority of NB-related mortality. The efficacy of the CpG-PBNP-PTT nanoimmunotherapy in a clinically relevant, TH-MYCN murine NB model (9464D) overexpressing MYCN is tested. When administered to 9464D NB cells in vitro, CpG-PBNP-PTT triggers thermal dose-dependent immunogenic cell death and tumor cell priming for immune recognition in vitro, measured by the expression of specific costimulatory and antigen-presenting molecules. In vivo, intratumorally administered CpG-PBNP-PTT generates complete tumor regression and significantly higher long-term survival compared to controls. Furthermore, CpG-PBNP-PTT-treated mice reject tumor rechallenge. Ex vivo studies confirm these therapeutic responses result from the generation of robust T cell-mediated immunological memory. Consequently, in a synchronous 9464D tumor model, CpG-PBNP-PTT induces complete tumor regression on the treated flank and significantly slows tumor progression on the untreated flank, improving animal survival. These findings demonstrate that localized administration of the CpG-PBNP-PTT nanoimmunotherapy drives potent systemic T cell responses in solid tumors such as NB and therefore has therapeutic implications for NB.
Collapse
Affiliation(s)
- Anshi Shukla
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Science and Engineering Hall 8 Floor, Washington, DC 20052, USA
| | - Juliana Cano-Mejia
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Science and Engineering Hall 8 Floor, Washington, DC 20052, USA
| | - Jaclyn Andricovich
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Science and Engineering Hall 8 Floor, Washington, DC 20052, USA.,The Institute for Biomedical Sciences, The George Washington University,2300 Eye Street NW, Ross Hall Room 561, Washington, DC 20037, USA
| | - Rachel A Burga
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Science and Engineering Hall 8 Floor, Washington, DC 20052, USA.,The Institute for Biomedical Sciences, The George Washington University,2300 Eye Street NW, Ross Hall Room 561, Washington, DC 20037, USA
| | - Elizabeth E Sweeney
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Science and Engineering Hall 8 Floor, Washington, DC 20052, USA
| | - Rohan Fernandes
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Science and Engineering Hall 8 Floor, Washington, DC 20052, USA
| |
Collapse
|
35
|
Intranasal Nanoparticle Vaccination Elicits a Persistent, Polyfunctional CD4 T Cell Response in the Murine Lung Specific for a Highly Conserved Influenza Virus Antigen That Is Sufficient To Mediate Protection from Influenza Virus Challenge. J Virol 2021; 95:e0084121. [PMID: 34076479 DOI: 10.1128/jvi.00841-21] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Lung-localized CD4 T cells play a critical role in the control of influenza virus infection and can provide broadly protective immunity. However, current influenza vaccination strategies primarily target influenza hemagglutinin (HA) and are administered peripherally to induce neutralizing antibodies. We have used an intranasal vaccination strategy targeting the highly conserved influenza nucleoprotein (NP) to elicit broadly protective lung-localized CD4 T cell responses. The vaccine platform consists of a self-assembling nanolipoprotein particle (NLP) linked to NP with an adjuvant. We have evaluated the functionality, in vivo localization, and persistence of the T cells elicited. Our study revealed that intranasal vaccination elicits a polyfunctional subset of lung-localized CD4 T cells that persist long term. A subset of these lung CD4 T cells localize to the airway, where they can act as early responders following encounter with cognate antigen. Polyfunctional CD4 T cells isolated from airway and lung tissue produce significantly more effector cytokines IFN-γ and TNF-α, as well as cytotoxic functionality. When adoptively transferred to naive recipients, CD4 T cells from NLP:NP-immunized lung were sufficient to mediate 100% survival from lethal challenge with H1N1 influenza virus. IMPORTANCE Exploiting new, more efficacious strategies to potentiate influenza virus-specific immune responses is important, particularly for at-risk populations. We have demonstrated the promise of direct intranasal protein vaccination to establish long-lived immunity in the lung with CD4 T cells that possess features and positioning in the lung that are associated with both immediate and long-term immunity, as well as demonstrating direct protective potential.
Collapse
|
36
|
The human memory T cell compartment changes across tissues of the female reproductive tract. Mucosal Immunol 2021; 14:862-872. [PMID: 33953338 PMCID: PMC8225572 DOI: 10.1038/s41385-021-00406-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 04/12/2021] [Accepted: 04/14/2021] [Indexed: 02/04/2023]
Abstract
Memory CD4 T cells in tissues fulfill numerous functions that are critical for local immune homeostasis and protection against pathogens. Previous studies have highlighted the phenotypic and functional heterogeneity of circulating and tissue-resident memory CD4 T cells across different human tissues such as skin, lung, liver, and colon. Comparatively little is known in regard to memory CD4 T cells across tissues of the female reproductive tract (FRT). We examined CD4 T cells in donor-matched vaginal, ecto- and endocervical tissues, which differ in mucosal structure and exposure to external environmental stimuli. We hypothesized that this could be reflected by tissue-specific differences in the memory CD4 T cell compartment. We found differences in CD4 subset distribution across these tissues. Specifically, CD69+CD103+ CD4 T cells were significantly more abundant in vaginal than cervical tissues. In contrast, the transcriptional profiles of CD4 subsets were fairly conserved across FRT tissues. CD69+CD103+ CD4 T cells showed a TH17 bias independent of tissue niche. Our data suggest that FRT tissues affect T cell subset distribution but have limited effects on the transcriptome of each subset. We discuss the implications for barrier immunity in the FRT.
Collapse
|
37
|
Gu HJ, Song S, Roh JY, Jung Y, Kim HJ. Expression pattern of tissue-resident memory T cells in cutaneous lupus erythematosus. Lupus 2021; 30:1427-1437. [PMID: 34013817 DOI: 10.1177/09612033211017218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Tissue resident memory T cells (TRMs) persist long-term in peripheral tissues without recirculation, triggering an immediate protective inflammatory state upon the re-recognition of the antigen. Despite evidence incriminating the dysregulation of TRMs in autoimmune diseases, few studies have examined their expression in cutaneous lupus erythematosus (CLE). OBJECTIVES We aimed to examine whether there are differences among TRM populations in CLE depending on different clinical conditions, such as the CLE subtype or association with systemic lupus erythematosus, and to determine the effect of type I interferon (IFN) on the development of TRMs in CLE. METHODS CLE disease activity was evaluated using the Cutaneous Lupus Erythematosus Disease Area and Severity Index. The expression of the TRM markers CD69 and CD103 in CLE lesions was evaluated by immunofluorescence. Flow cytometry was performed on peripheral blood mononuclear cells after IFNα treatment. RESULTS The number of TRMs expressing either CD69 or CD103 was significantly higher in CLE lesions than in control skin; however, it was not significantly different between discoid lupus erythematosus and subacute CLE, or dependent on the presence of concomitant systemic lupus. Lesional severity was not correlated with an increase in TRMs in CLE. IFNα treatment induced a conspicuous increase in CD69 expression in skin-homing T cells, more profoundly in CD4+ T cells than in CD8+ T cells. CONCLUSIONS Skin TRMs, either CD69 or CD103-positive cells, showed increased levels in the lesional skin of CLE, and IFNα increased the expression of CD69 in T cells.
Collapse
Affiliation(s)
- Hyeon-Jung Gu
- Department of Health Science and Technology, Gachon Advanced Institute for Health Science & Technology, Gachon University, Incheon, Korea
| | - Shinyoung Song
- Department of Dermatology, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Joo Young Roh
- Department of Dermatology, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - YunJae Jung
- Department of Health Science and Technology, Gachon Advanced Institute for Health Science & Technology, Gachon University, Incheon, Korea.,Department of Microbiology, Gachon University College of Medicine, Incheon, Korea
| | - Hee Joo Kim
- Department of Dermatology, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| |
Collapse
|
38
|
Gehlhausen JR, Iwasaki A. B cells join T cell clusters in the host response to recurrent herpes simplex virus 2 infection. J Clin Invest 2021; 131:148300. [PMID: 33938452 DOI: 10.1172/jci148300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Recurrent genital herpes lesions are infiltrated by various leukocytes, yet the role of B cell subsets in this process is unknown. In this issue of the JCI, Ford et al. describe the presence and antibody-secreting role of local B cell populations in herpes simplex virus 2 (HSV-2) recurrent lesions. The authors analyzed a comprehensive array of sequential skin biopsy specimens from HSV-2-infected patients over time and at various stages of infection. Using immunofluorescence and in situ hybridization, the authors show the presence of rare IgD+ naive B cells and IgG-expressing antibody-secreting cells (ASCs) in recurrent HSV-2 lesions embedded in CD4+ T cell-rich dermal immune infiltrates, levels of which transiently increase during lesion reactivation and healing. Notably, local increases in HSV-2-specific antibodies in recurrent lesions were detected, whereas serum HSV-2 antibody levels remained stable. Future research is needed to understand the precise role of these tissue-visiting B cells in disease resolution.
Collapse
Affiliation(s)
| | - Akiko Iwasaki
- Department of Dermatology and.,Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Molecular Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA.,Howard Hughes Medical Institute
| |
Collapse
|
39
|
Booth JS, Goldberg E, Patil SA, Barnes RS, Greenwald BD, Sztein MB. Age-dependency of terminal ileum tissue resident memory T cell responsiveness profiles to S. Typhi following oral Ty21a immunization in humans. Immun Ageing 2021; 18:19. [PMID: 33874975 PMCID: PMC8053564 DOI: 10.1186/s12979-021-00227-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 03/16/2021] [Indexed: 01/02/2023]
Abstract
BACKGROUND The impact of aging on the immune system is unequivocal and results in an altered immune status termed immunosenescence. In humans, the mechanisms of immunosenescence have been examined almost exclusively in blood. However, most immune cells are present in tissue compartments and exhibit differential cell (e.g., memory T cells -TM) subset distributions. Thus, it is crucial to understand immunosenescence in tissues, especially those that are exposed to pathogens (e.g., intestine). Using a human model of oral live attenuated typhoid vaccine, Ty21a, we investigated the effect of aging on terminal ileum (TI) tissue resident memory T (TRM) cells. TRM provide immediate adaptive effector immune responsiveness at the infection site. However, it is unknown whether aging impacts TRM S. Typhi-responsive cells at the site of infection (e.g., TI). Here, we determined the effect of aging on the induction of TI S. Typhi-responsive TRM subsets elicited by Ty21a immunization. RESULTS We observed that aging impacts the frequencies of TI-lamina propria mononuclear cells (LPMC) TM and TRM in both Ty21a-vaccinated and control groups. In unvaccinated volunteers, the frequencies of LPMC CD103- CD4+ TRM displayed a positive correlation with age whilst the CD4/CD8 ratio in LPMC displayed a negative correlation with age. We observed that elderly volunteers have weaker S. Typhi-specific mucosal immune responses following Ty21a immunization compared to adults. For example, CD103+ CD4+ TRM showed reduced IL-17A production, while CD103- CD4+ TRM exhibited lower levels of IL-17A and IL-2 in the elderly than in adults following Ty21a immunization. Similar results were observed in LPMC CD8+ TRM and CD103- CD8+ T cell subsets. A comparison of multifunctional (MF) profiles of both CD4+ and CD8+ TRM subsets between elderly and adults also showed significant differences in the quality and quantity of elicited single (S) and MF responses. CONCLUSIONS Aging influences tissue resident TM S. Typhi-specific responses in the terminal ileum following oral Ty21a-immunization. This study is the first to provide insights in the generation of local vaccine-specific responses in the elderly population and highlights the importance of evaluating tissue immune responses in the context of infection and aging. TRIAL REGISTRATION This study was approved by the Institutional Review Board and registered on ClinicalTrials.gov (identifier NCT03970304 , Registered 29 May 2019 - Retrospectively registered).
Collapse
Affiliation(s)
- Jayaum S Booth
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Eric Goldberg
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Seema A Patil
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Robin S Barnes
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Bruce D Greenwald
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
- Program in Oncology, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, 21201, USA.
| |
Collapse
|
40
|
D Helble J, N Starnbach M. T cell responses to Chlamydia. Pathog Dis 2021; 79:6164867. [PMID: 33693620 DOI: 10.1093/femspd/ftab014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/07/2021] [Indexed: 12/27/2022] Open
Abstract
Chlamydia trachomatis is the most commonly reported sexually transmitted infection in the United States. The high prevalence of infection and lack of a vaccine indicate a critical knowledge gap surrounding the host's response to infection and how to effectively generate protective immunity. The immune response to C. trachomatis is complex, with cells of the adaptive immune system playing a crucial role in bacterial clearance. Here, we discuss the CD4+ and CD8+ T cell response to Chlamydia, the importance of antigen specificity and the role of memory T cells during the recall response. Ultimately, a deeper understanding of protective immune responses is necessary to develop a vaccine that prevents the inflammatory diseases associated with Chlamydia infection.
Collapse
Affiliation(s)
- Jennifer D Helble
- Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
41
|
Stolley JM, Johnston TS, Soerens AG, Beura LK, Rosato PC, Joag V, Wijeyesinghe SP, Langlois RA, Osum KC, Mitchell JS, Masopust D. Retrograde migration supplies resident memory T cells to lung-draining LN after influenza infection. J Exp Med 2021; 217:151876. [PMID: 32568362 PMCID: PMC7398169 DOI: 10.1084/jem.20192197] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/20/2020] [Accepted: 05/12/2020] [Indexed: 01/06/2023] Open
Abstract
Numerous observations indicate that resident memory T cells (TRM) undergo unusually rapid attrition within the lung. Here we demonstrate that contraction of lung CD8+ T cell responses after influenza infection is contemporized with egress of CD69+/CD103+ CD8+ T cells to the draining mediastinal LN via the lymphatic vessels, which we term retrograde migration. Cells within the draining LN retained canonical markers of lung TRM, including CD103 and CD69, lacked Ly6C expression (also a feature of lung TRM), maintained granzyme B expression, and did not equilibrate among immunized parabiotic mice. Investigations of bystander infection or removal of the TCR from established memory cells revealed that the induction of the TRM phenotype was dependent on antigen recognition; however, maintenance was independent. Thus, local lung infection induces CD8+ T cells with a TRM phenotype that nevertheless undergo retrograde migration, yet remain durably committed to the residency program within the draining LN, where they provide longer-lived regional memory while chronicling previous upstream antigen experiences.
Collapse
Affiliation(s)
- J Michael Stolley
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Timothy S Johnston
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Andrew G Soerens
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Lalit K Beura
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN.,Department of Microbiology and Immunology, Brown University, Providence, RI
| | - Pamela C Rosato
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Vineet Joag
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Sathi P Wijeyesinghe
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Ryan A Langlois
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Kevin C Osum
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Jason S Mitchell
- Center for Immunology, University of Minnesota, Minneapolis, MN.,Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - David Masopust
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN.,Center for Immunology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
42
|
Bartsch LM, Damasio MPS, Subudhi S, Drescher HK. Tissue-Resident Memory T Cells in the Liver-Unique Characteristics of Local Specialists. Cells 2020; 9:cells9112457. [PMID: 33187162 PMCID: PMC7696520 DOI: 10.3390/cells9112457] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/02/2020] [Accepted: 11/06/2020] [Indexed: 12/23/2022] Open
Abstract
T cells play an important role to build up an effective immune response and are essential in the eradication of pathogens. To establish a long-lasting protection even after a re-challenge with the same pathogen, some T cells differentiate into memory T cells. Recently, a certain subpopulation of memory T cells at different tissue-sites of infection was detected-tissue-resident memory T cells (TRM cells). These cells can patrol in the tissue in order to encounter their cognate antigen to establish an effective protection against secondary infection. The liver as an immunogenic organ is exposed to a variety of pathogens entering the liver through the systemic blood circulation or via the portal vein from the gut. It could be shown that intrahepatic TRM cells can reside within the liver tissue for several years. Interestingly, hepatic TRM cell differentiation requires a distinct cytokine milieu. In addition, TRM cells express specific surface markers and transcription factors, which allow their identification delimited from their circulating counterparts. It could be demonstrated that liver TRM cells play a particular role in many liver diseases such as hepatitis B and C infection, non-alcoholic fatty liver disease and even play a role in the development of hepatocellular carcinoma and in building long-lasting immune responses after vaccination. A better understanding of intrahepatic TRM cells is critical to understand the pathophysiology of many liver diseases and to identify new potential drug targets for the development of novel treatment strategies.
Collapse
Affiliation(s)
- Lea M. Bartsch
- Correspondence: (L.M.B.); (H.K.D.); Tel.: +1-(617)-724-7515 (L.M.B. & H.K.D.)
| | | | | | - Hannah K. Drescher
- Correspondence: (L.M.B.); (H.K.D.); Tel.: +1-(617)-724-7515 (L.M.B. & H.K.D.)
| |
Collapse
|
43
|
Dabee S, Mkhize NN, Jaspan HB, Lewis D, Gumbi PP, Passmore JAS. Initiation of Antiretroviral Therapy Differentially Influences Genital and Systemic Immune Activation in HIV-Infected Women. AIDS Res Hum Retroviruses 2020; 36:821-830. [PMID: 32524856 DOI: 10.1089/aid.2019.0268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Antiretroviral therapy (ART) has significantly improved the quality of life of HIV-infected individuals: reducing plasma viremia, restoring CD4+ T cell numbers, and correcting imbalances in blood memory T cell subsets. While ART improves immune correlates at mucosal sites, including the lower female genital tract (FGT), ART initiation has been associated with reactivation of common FGT infections. We investigated the effect of ART on immune activation and inflammation in the genital tract. We measured blood and genital T cell activation, proliferation, and immunosenescence (CD38, HLADR, Ki67, CD127, and CD57), and cytokine levels in women on ART for ∼7 years (cross-sectional analysis) or initiating ART (immediately before and 1 month after). Effector memory T cells predominated in blood and FGT during chronic infection, irrespective of ART status. In women initiating ART, 1 month was insufficient for T cell reconstitution, or alterations in T cell subset distribution, despite both plasma and genital viral loads decreasing to undetectable levels in most participants. Initiating ART was accompanied by a decline in plasma IP-10 that correlated with decreased blood CD38 expression in blood (p = .0204) but not in the FGT. The reduction in plasma (but not genital) cytokine levels due to ART initiation was dependent on their concentrations before treatment. While T cell activation decreased significantly in blood (CD4: p = .032; CD8: p = .0137), activation levels remained similar in the genital tract despite 1 month of treatment. Overall, the decrease in cellular activation and inflammation seen in blood with ART initiation was not evident in the FGT.
Collapse
Affiliation(s)
- Smritee Dabee
- Division of Medical Virology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
| | | | - Heather B. Jaspan
- Division of Medical Virology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Seattle Childrens Hospital, Seattle, Washington, USA
| | - David Lewis
- Western Sydney Sexual Health Centre, Parramatta, Australia
- Westmead Clinical School and Centre for Infectious Diseases and Microbiology & Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, Australia
| | - Pamela P. Gumbi
- Department of Biochemistry, University of KwaZulu Natal, Pietermaritzburg, South Africa
| | - Jo-Ann S. Passmore
- Division of Medical Virology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- NRF-DST Centre of Excellence in HIV Prevention, Centre for the AIDS Programme of Research in South Africa, University of KwaZulu Natal, Durban, South Africa
- National Health Laboratory Service, Cape Town, South Africa
| |
Collapse
|
44
|
Mannie MD, DeOca KB, Bastian AG, Moorman CD. Tolerogenic vaccines: Targeting the antigenic and cytokine niches of FOXP3 + regulatory T cells. Cell Immunol 2020; 355:104173. [PMID: 32712270 PMCID: PMC7444458 DOI: 10.1016/j.cellimm.2020.104173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023]
Abstract
FOXP3+ regulatory T cells (Tregs) constitute a critical barrier that enforces tolerance to both the self-peptidome and the extended-self peptidome to ensure tissue-specific resistance to autoimmune, allergic, and other inflammatory disorders. Here, we review intuitive models regarding how T cell antigen receptor (TCR) specificity and antigen recognition efficiency shape the Treg and conventional T cell (Tcon) repertoires to adaptively regulate T cell maintenance, tissue-residency, phenotypic stability, and immune function in peripheral tissues. Three zones of TCR recognition efficiency are considered, including Tcon recognition of specific low-efficiency self MHC-ligands, Treg recognition of intermediate-efficiency agonistic self MHC-ligands, and Tcon recognition of cross-reactive high-efficiency agonistic foreign MHC-ligands. These respective zones of TCR recognition efficiency are key to understanding how tissue-resident immune networks integrate the antigenic complexity of local environments to provide adaptive decisions setting the balance of suppressive and immunogenic responses. Importantly, deficiencies in the Treg repertoire appear to be an important cause of chronic inflammatory disease. Deficiencies may include global deficiencies in Treg numbers or function, subtle 'holes in the Treg repertoire' in tissue-resident Treg populations, or simply Treg insufficiencies that are unable to counter an overwhelming molecular mimicry stimulus. Tolerogenic vaccination and Treg-based immunotherapy are two therapeutic modalities meant to restore dominance of Treg networks to reverse chronic inflammatory disease. Studies of these therapeutic modalities in a preclinical setting have provided insight into the Treg niche, including the concept that intermediate-efficiency TCR signaling, high IFN-β concentrations, and low IL-2 concentrations favor Treg responses and active dominant mechanisms of immune tolerance. Overall, the purpose here is to assimilate new and established concepts regarding how cognate TCR specificity of the Treg repertoire and the contingent cytokine networks provide a foundation for understanding Treg suppressive strategy.
Collapse
Affiliation(s)
- Mark D Mannie
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States.
| | - Kayla B DeOca
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| | - Alexander G Bastian
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| | - Cody D Moorman
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| |
Collapse
|
45
|
Editorial of Harnessing the Power of T Cells: The Promising Hope for a Universal Influenza Vaccine. Vaccines (Basel) 2020; 8:vaccines8030376. [PMID: 32664485 PMCID: PMC7565606 DOI: 10.3390/vaccines8030376] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022] Open
|
46
|
Nakamura Y, Mimuro H, Kunisawa J, Furusawa Y, Takahashi D, Fujimura Y, Kaisho T, Kiyono H, Hase K. Microfold cell-dependent antigen transport alleviates infectious colitis by inducing antigen-specific cellular immunity. Mucosal Immunol 2020; 13:679-690. [PMID: 32042052 DOI: 10.1038/s41385-020-0263-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 12/31/2019] [Accepted: 01/13/2020] [Indexed: 02/04/2023]
Abstract
Infectious colitis is one of the most common health issues worldwide. Microfold (M) cells actively transport luminal antigens to gut-associated lymphoid tissue to induce IgA responses; however, it remains unknown whether M cells contribute to the induction of cellular immune responses. Here we report that M cell-dependent antigen transport plays a critical role in the induction of Th1, Th17, and Th22 responses against gut commensals in the steady state. The establishment of commensal-specific cellular immunity was a prerequisite for preventing bacterial dissemination during enteropathogenic Citrobacter rodentium infection. Therefore, M cell-null mice developed severe colitis with increased bacterial dissemination. This abnormality was associated with mucosal barrier dysfunction. These observations suggest that antigen transport by M cells may help maintain gut immune homeostasis by eliciting antigen-specific cellular immune responses.
Collapse
Affiliation(s)
- Yutaka Nakamura
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, 105-0011, Japan.,Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Hitomi Mimuro
- Division of Bacteriology, International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo (IMSUT), 108-8639, Tokyo, Japan.,Division of Infectious Diseases, Research Institute of Microbial Diseases (RIMD), Osaka University, Osaka, 565-0871, Japan
| | - Jun Kunisawa
- International Research and Development Center for Mucosal Vaccine, The Institute of Medical Science, The University of Tokyo (IMSUT), 108-8639, Tokyo, Japan.,Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, 567-0085, Japan
| | - Yukihiro Furusawa
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, 105-0011, Japan.,Department of Liberal Arts and Sciences, Toyama Prefectural University, Toyama, 939-0398, Japan
| | - Daisuke Takahashi
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, 105-0011, Japan
| | - Yumiko Fujimura
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, 105-0011, Japan
| | - Tsuneyasu Kaisho
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, 641-8509, Japan
| | - Hiroshi Kiyono
- International Research and Development Center for Mucosal Vaccine, The Institute of Medical Science, The University of Tokyo (IMSUT), 108-8639, Tokyo, Japan.,Department of Mucosal Immunology, The University of Tokyo Distinguished Professor Unit, International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo (IMSUT), Tokyo, 108-8639, Japan.,Division of Gastroenterology, Department of Medicine, School of Medicine and Chiba University-UCSD Center for Mucosal Immunology, Allergy and Vaccines (cMAV), University of California, San Diego, CA, 92093, USA.,Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-0856, Japan
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, 105-0011, Japan. .,International Research and Development Center for Mucosal Vaccine, The Institute of Medical Science, The University of Tokyo (IMSUT), 108-8639, Tokyo, Japan.
| |
Collapse
|
47
|
Misumi I, Starmer J, Uchimura T, Beck MA, Magnuson T, Whitmire JK. Obesity Expands a Distinct Population of T Cells in Adipose Tissue and Increases Vulnerability to Infection. Cell Rep 2020; 27:514-524.e5. [PMID: 30970254 PMCID: PMC6652206 DOI: 10.1016/j.celrep.2019.03.030] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 08/16/2018] [Accepted: 03/08/2019] [Indexed: 02/07/2023] Open
Abstract
Obesity in humans is associated with poorer health outcomes after infections compared with non-obese individuals. Here, we examined the effects of white adipose tissue and obesity on T cell responses to viral infection in mice. We show that lymphocytic choriomeningitis virus (LCMV) grows to high titer in adipose tissue. Virus-specific T cells enter the adipose tissue to resolve infection but then remain as a memory population distinct from memory T cells in lymphoid tissues. Memory T cells in adipose tissue are abundant in lean mice, and diet-induced obesity further increases memory T cell number in adipose tissue and spleen. Upon re-challenge infection, memory T cells rapidly cause severe pathogenesis, leading to increases in lipase levels, calcification of adipose tissue, pancreatitis, and reduced survival in obese mice but not lean mice. Thus, obesity leads to a unique form of viral pathogenesis involving memory T cell-dependent adipocyte destruction and damage to other tissues. Obesity is associated with increased morbidity and mortality after viral infections. Using a mouse model of obesity, Misumi et al. identify a distinct population of memory T cells in white adipose tissue and a memory cell-dependent pathogenic response to infection that leads to acute fat necrosis, pancreatitis, and lethality.
Collapse
Affiliation(s)
- Ichiro Misumi
- Department of Genetics, UNC-Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Joshua Starmer
- Department of Genetics, UNC-Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Toru Uchimura
- Department of Genetics, UNC-Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Melinda A Beck
- Department of Nutrition, UNC-Chapel Hill Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - Terry Magnuson
- Department of Genetics, UNC-Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Jason K Whitmire
- Department of Genetics, UNC-Chapel Hill School of Medicine, Chapel Hill, NC, USA; Department of Microbiology & Immunology, UNC-Chapel Hill School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
48
|
Abstract
The impact of spaceflight on the immune system has been investigated extensively during spaceflight missions and in model experiments conducted on Earth. Data suggest that the spaceflight environment may affect the development of acquired immunity, and immune responses. Herein we summarize and discuss the influence of the spaceflight environment on acquired immunity. Bone marrow and the thymus, two major primary lymphoid organs, are evidently affected by gravitational change during spaceflight. Changes in the microenvironments of these organs impair lymphopoiesis, and thereby may indirectly impinge on acquired immunity. Acquired immune responses may also be disturbed by gravitational fluctuation, stressors, and space radiation both directly and in a stress hormone-dependent manner. These changes may affect acquired immune responses to pathogens, allergens, and tumors.
Collapse
|
49
|
Dobson HE, Dias LDS, Kohn EM, Fites S, Wiesner DL, Dileepan T, Kujoth GC, Abraham A, Ostroff GR, Klein BS, Wüthrich M. Antigen discovery unveils resident memory and migratory cell roles in antifungal resistance. Mucosal Immunol 2020; 13:518-529. [PMID: 31900406 PMCID: PMC7183437 DOI: 10.1038/s41385-019-0244-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 10/03/2019] [Accepted: 11/27/2019] [Indexed: 02/04/2023]
Abstract
Priming at the site of natural infection typically elicits a protective T cell response against subsequent pathogen encounter. Here, we report the identification of a novel fungal antigen that we harnessed for mucosal vaccination and tetramer generation to test whether we can elicit protective, antigen-specific tissue-resident memory (Trm) CD4+ T cells in the lung parenchyma. In contrast to expectations, CD69+, CXCR3+, CD103- Trm cells failed to protect against a lethal pulmonary fungal infection. Surprisingly, systemic vaccination induced a population of tetramer+ CD4+ T cells enriched within the pulmonary vasculature, and expressing CXCR3 and CX3CR1, that migrated to the lung tissue upon challenge and efficiently protected mice against infection. Mucosal vaccine priming of Trm may not reliably protect against mucosal pathogens.
Collapse
Affiliation(s)
- Hannah E Dobson
- Departments of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Lucas Dos Santos Dias
- Departments of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Elaine M Kohn
- Departments of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Scott Fites
- Departments of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Darin L Wiesner
- Departments of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Thamotharampillai Dileepan
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Gregory C Kujoth
- Departments of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Ambily Abraham
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Gary R Ostroff
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Bruce S Klein
- Departments of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Departments of Internal Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Departments of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Marcel Wüthrich
- Departments of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
50
|
Fonseca R, Beura LK, Quarnstrom CF, Ghoneim HE, Fan Y, Zebley CC, Scott MC, Fares-Frederickson NJ, Wijeyesinghe S, Thompson EA, Borges da Silva H, Vezys V, Youngblood B, Masopust D. Developmental plasticity allows outside-in immune responses by resident memory T cells. Nat Immunol 2020; 21:412-421. [PMID: 32066954 PMCID: PMC7096285 DOI: 10.1038/s41590-020-0607-7] [Citation(s) in RCA: 202] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 01/17/2020] [Indexed: 01/02/2023]
Abstract
Central memory T (TCM) cells patrol lymph nodes and perform conventional memory responses on restimulation: proliferation, migration and differentiation into diverse T cell subsets while also self-renewing. Resident memory T (TRM) cells are parked within single organs, share properties with terminal effectors and contribute to rapid host protection. We observed that reactivated TRM cells rejoined the circulating pool. Epigenetic analyses revealed that TRM cells align closely with conventional memory T cell populations, bearing little resemblance to recently activated effectors. Fully differentiated TRM cells isolated from small intestine epithelium exhibited the potential to differentiate into TCM cells, effector memory T cells and TRM cells on recall. Ex-TRM cells, former intestinal TRM cells that rejoined the circulating pool, heritably maintained a predilection for homing back to their tissue of origin on subsequent reactivation and a heightened capacity to redifferentiate into TRM cells. Thus, TRM cells can rejoin the circulation but are advantaged to re-form local TRM when called on.
Collapse
Affiliation(s)
- Raissa Fonseca
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
- Department of Microbiology and Immunology, University of Melbourne and the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Lalit K Beura
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Clare F Quarnstrom
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Hazem E Ghoneim
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
- Department of Microbial Infection and Immunity, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Yiping Fan
- Department of Computational Biology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Caitlin C Zebley
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Milcah C Scott
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Nancy J Fares-Frederickson
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Sathi Wijeyesinghe
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Emily A Thompson
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Henrique Borges da Silva
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Vaiva Vezys
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Benjamin Youngblood
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - David Masopust
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA.
| |
Collapse
|