51
|
Gopinath S, Lu P, Iwasaki A. Cutting Edge: The Use of Topical Aminoglycosides as an Effective Pull in "Prime and Pull" Vaccine Strategy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:1703-1707. [PMID: 32122994 PMCID: PMC11694766 DOI: 10.4049/jimmunol.1900462] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 02/01/2020] [Indexed: 12/12/2022]
Abstract
The presence of tissue-resident memory T cells at barrier tissues is critical for long-lasting protective immune responses. Previous work has shown that tissue-resident memory T cells can be established by "pulling" virus-specific effector T cells from circulation to the genital mucosa via topical vaginal application of chemokines in mice. Once established, these cells protect hosts against genital herpes infection. We recently showed that vaginal application of aminoglycoside antibiotics induces robust activation of the IFN signaling pathway, including upregulation of chemokine expression within the tissue in mice. In this study, we show that a single topical application of neomycin, an inexpensive and vaginally nontoxic antibiotic, is sufficient to pull CD8 T cells to the vaginal mucosa and provide protection against genital herpes infection in mice.
Collapse
Affiliation(s)
- Smita Gopinath
- Howard Hughes Medical Institute, Chevy Chase, MD 20815
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519; and
| | - Peiwen Lu
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519; and
| | - Akiko Iwasaki
- Howard Hughes Medical Institute, Chevy Chase, MD 20815;
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519; and
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06519
| |
Collapse
|
52
|
Deli F, Romano F, Gualini G, Mariani GM, Sala I, Veneziano F, Bertero L, Cassoni P, Aimetti M. Resident memory T cells: Possible players in periodontal disease recurrence. J Periodontal Res 2020; 55:324-330. [DOI: 10.1111/jre.12709] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 09/12/2019] [Accepted: 10/10/2019] [Indexed: 01/12/2023]
Affiliation(s)
- Federico Deli
- Department of Surgical Sciences, C.I.R. Dental School Section of Periodontology University of Turin Turin Italy
| | - Federica Romano
- Department of Surgical Sciences, C.I.R. Dental School Section of Periodontology University of Turin Turin Italy
| | - Giacomo Gualini
- Department of Surgical Sciences, C.I.R. Dental School Section of Periodontology University of Turin Turin Italy
| | - Giulia Maria Mariani
- Department of Surgical Sciences, C.I.R. Dental School Section of Periodontology University of Turin Turin Italy
| | - Irene Sala
- Department of Surgical Sciences, C.I.R. Dental School Section of Periodontology University of Turin Turin Italy
| | - Francesca Veneziano
- Department of Medical Sciences Pathology Unit University of Turin Turin Italy
| | - Luca Bertero
- Department of Medical Sciences Pathology Unit University of Turin Turin Italy
| | - Paola Cassoni
- Department of Medical Sciences Pathology Unit University of Turin Turin Italy
| | - Mario Aimetti
- Department of Surgical Sciences, C.I.R. Dental School Section of Periodontology University of Turin Turin Italy
| |
Collapse
|
53
|
E Silva RDF, de Oliveira BC, da Silva AA, Brelaz de Castro MCA, Ferreira LFGR, Hernandes MZ, de Brito MEF, de-Melo-Neto OP, Rezende AM, Pereira VRA. Immunogenicity of Potential CD4 + and CD8 + T Cell Epitopes Derived From the Proteome of Leishmania braziliensis. Front Immunol 2020; 10:3145. [PMID: 32117204 PMCID: PMC7033680 DOI: 10.3389/fimmu.2019.03145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 12/27/2019] [Indexed: 11/14/2022] Open
Abstract
Background: A safe and effective vaccine against human leishmaniasis still requires the identification of better antigens for immunization and adequate models to evaluate the immune response. To support vaccine development, this work tested the immunogenicity of 10 different peptides derived from the proteome of Leishmania braziliensis, which were selected by their in silico affinity to MHC complexes. Research design and Methods: Comparative cell proliferation assays were performed by culturing, in the presence of each peptide, PBMC cells from subclinical subjects (SC), cutaneous leishmaniasis patients with active disease (AD), post-treatment (PT) individuals, and healthy controls. Culture supernatants were then used for Th1, Th2, and Th17 cytokine measurements. Cells from selected PT samples were also used to assess the expression, by T cells, of the T-bet Th1 transcription factor. Results: A robust cell proliferation was observed for the SC group, for all the tested peptides. The levels of Th1 cytokines were peptide-dependent and had substantial variations between groups, where, for instance, IFN-γ and TNF levels were some of the highest, particularly on PT cultures, when compared to IL-2. On the other hand, Th2 cytokines displayed much less variation. IL-6 was the most abundant among all the evaluated cytokines while IL-4 and IL-10 could be found at much lower concentrations. IL-17 was also detected with variations in SC and AD groups. T-bet was up-regulated in CD4+ and CD8+ T cells from the PT group after stimulation with all peptides. Conclusions: The peptide epitopes can differentially stimulate cells from SC, AD, and PT individuals, leading to distinct immune responses.
Collapse
Affiliation(s)
- Rafael de Freitas E Silva
- Department of Natural Sciences, Universidade de Pernambuco, Garanhuns, Brazil.,Department of Immunology, Fundação Oswaldo Cruz, Recife, Brazil
| | | | | | - Maria Carolina Accioly Brelaz de Castro
- Department of Immunology, Fundação Oswaldo Cruz, Recife, Brazil.,Parasitology Laboratory, Universidade Federal de Pernambuco, Vitória de Santo Antão, Brazil
| | | | | | | | | | | | | |
Collapse
|
54
|
Riding RL, Harris JE. The Role of Memory CD8 + T Cells in Vitiligo. THE JOURNAL OF IMMUNOLOGY 2020; 203:11-19. [PMID: 31209143 DOI: 10.4049/jimmunol.1900027] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/05/2019] [Indexed: 12/31/2022]
Abstract
Vitiligo is an autoimmune skin disease mediated by autoreactive CD8+ T cells that destroy the pigment-producing cells of the epidermis, melanocytes, leading to areas of depigmentation. Patients with vitiligo require lifelong treatment to regain and maintain their pigment. Clinical observations uncovered the importance of autoimmune memory in vitiligo because cessation of treatment frequently led to relapse of disease at the site of previous lesions. A subset of memory T cells known as CD8+ resident memory T cells (TRM) are long-lived, nonmigratory memory cells that persist in most nonlymphoid tissues, including the skin. Recent reports describe the presence of CD8+ TRM in lesional vitiligo patient skin and suggest their role as active players in disease maintenance. In this review, we will discuss the role of skin CD8+ TRM in maintaining disease in vitiligo and the opportunity to target this population to induce a long-lasting reversal of disease.
Collapse
Affiliation(s)
- Rebecca L Riding
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA 01605
| | - John E Harris
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA 01605
| |
Collapse
|
55
|
Memory CD4 + T Cells in Immunity and Autoimmune Diseases. Cells 2020; 9:cells9030531. [PMID: 32106536 PMCID: PMC7140455 DOI: 10.3390/cells9030531] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/20/2020] [Accepted: 02/20/2020] [Indexed: 12/26/2022] Open
Abstract
CD4+ T helper (Th) cells play central roles in immunity in health and disease. While much is known about the effector function of Th cells in combating pathogens and promoting autoimmune diseases, the roles and biology of memory CD4+ Th cells are complex and less well understood. In human autoimmune diseases such as multiple sclerosis (MS), there is a critical need to better understand the function and biology of memory T cells. In this review article we summarize current concepts in the field of CD4+ T cell memory, including natural history, developmental pathways, subsets, and functions. Furthermore, we discuss advancements in the field of the newly-described CD4+ tissue-resident memory T cells and of CD4+ memory T cells in autoimmune diseases, two major areas of important unresolved questions in need of answering to advance new vaccine design and development of novel treatments for CD4+ T cell-mediated autoimmune diseases.
Collapse
|
56
|
Poh CM, Chan YH, Ng LFP. Role of T Cells in Chikungunya Virus Infection and Utilizing Their Potential in Anti-Viral Immunity. Front Immunol 2020; 11:287. [PMID: 32153590 PMCID: PMC7046835 DOI: 10.3389/fimmu.2020.00287] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 02/05/2020] [Indexed: 11/17/2022] Open
Abstract
Chikungunya virus (CHIKV) is an arthropod-borne alphavirus that causes hallmark debilitating polyarthralgia, fever, and rash in patients. T cell-mediated immunity, especially CD4+ T cells, are known to participate in the pathogenic role of CHIKV immunopathology. The other T cell subsets, notably CD8+, NKT, and gamma-delta (γδ) T cells, can also contribute to protective immunity, but their effect is not actuated during the natural course of infection. This review serves to consolidate and discuss the multifaceted roles of these T cell subsets during acute and chronic phases of CHIKV infection, and highlight gaps in the current literature. Importantly, the unique characteristics of skin-resident memory T cells are outlined to propose novel prophylactic strategies that utilize their properties to provide adequate, lasting protection.
Collapse
Affiliation(s)
- Chek Meng Poh
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Yi-Hao Chan
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Lisa F P Ng
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore.,National University of Singapore Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
57
|
Le Tortorec A, Matusali G, Mahé D, Aubry F, Mazaud-Guittot S, Houzet L, Dejucq-Rainsford N. From Ancient to Emerging Infections: The Odyssey of Viruses in the Male Genital Tract. Physiol Rev 2020; 100:1349-1414. [PMID: 32031468 DOI: 10.1152/physrev.00021.2019] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The male genital tract (MGT) is the target of a number of viral infections that can have deleterious consequences at the individual, offspring, and population levels. These consequences include infertility, cancers of male organs, transmission to the embryo/fetal development abnormalities, and sexual dissemination of major viral pathogens such as human immunodeficiency virus (HIV) and hepatitis B virus. Lately, two emerging viruses, Zika and Ebola, have additionally revealed that the human MGT can constitute a reservoir for viruses cleared from peripheral circulation by the immune system, leading to their sexual transmission by cured men. This represents a concern for future epidemics and further underlines the need for a better understanding of the interplay between viruses and the MGT. We review here how viruses, from ancient viruses that integrated the germline during evolution through old viruses (e.g., papillomaviruses originating from Neanderthals) and more modern sexually transmitted infections (e.g., simian zoonotic HIV) to emerging viruses (e.g., Ebola and Zika) take advantage of genital tract colonization for horizontal dissemination, viral persistence, vertical transmission, and endogenization. The MGT immune responses to viruses and the impact of these infections are discussed. We summarize the latest data regarding the sources of viruses in semen and the complex role of this body fluid in sexual transmission. Finally, we introduce key animal findings that are relevant for our understanding of viral infection and persistence in the human MGT and suggest future research directions.
Collapse
Affiliation(s)
- Anna Le Tortorec
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Giulia Matusali
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Dominique Mahé
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Florence Aubry
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Séverine Mazaud-Guittot
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Laurent Houzet
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Nathalie Dejucq-Rainsford
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| |
Collapse
|
58
|
Cho HS, Ha S, Shin HM, Reboldi A, Hall JA, Huh JR, Usherwood EJ, Berg LJ. CD8 + T Cells Require ITK-Mediated TCR Signaling for Migration to the Intestine. Immunohorizons 2020; 4:57-71. [PMID: 32034085 PMCID: PMC7521019 DOI: 10.4049/immunohorizons.1900093] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 01/12/2020] [Indexed: 12/21/2022] Open
Abstract
The Tec kinase IL-2–inducible T cell kinase (ITK) regulates the expression of TCR-induced genes. Itk−/− T cell responses are impaired but not absent. ITK inhibition prevented colitis disease progression and impaired T cell migration to the colon in mice. To examine the function of ITK in T cell migration to the intestine, we examined the number of gut T cells in Itk−/− mice and then evaluated their expression of gut-homing receptors. Combined with in vitro murine T cell stimulation and in vivo migration assay using congenic B6 mice, we demonstrated an essential role for ITK in T cell migration to the intestine in mice. Reconstitution of Itk−/− mouse CD8+ T cells with IFN regulatory factor 4 restored gut-homing properties, providing mechanistic insight into the function of ITK-mediated signaling in CD8+ T cell migration to the intestinal mucosa in mice.
Collapse
Affiliation(s)
- Hyoung-Soo Cho
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Soyoung Ha
- Department of Immunology, Harvard Medical School, Boston, MA 02115
| | - Hyun Mu Shin
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Andrea Reboldi
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Jason A Hall
- The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016; and
| | - Jun R Huh
- Department of Immunology, Harvard Medical School, Boston, MA 02115
| | - Edward J Usherwood
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| | - Leslie J Berg
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605;
| |
Collapse
|
59
|
Owczarczyk-Saczonek A, Krajewska-Włodarczyk M, Kasprowicz-Furmańczyk M, Placek W. Immunological Memory of Psoriatic Lesions. Int J Mol Sci 2020; 21:ijms21020625. [PMID: 31963581 PMCID: PMC7014148 DOI: 10.3390/ijms21020625] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 01/09/2023] Open
Abstract
The natural course of psoriasis is the appearance of new lesions in the place of previous ones, which disappeared after a successful therapy. Recent studies of psoriasis etiopathogenesis showed that after psoriatic plaques have disappeared, in healthy skin we can still find a trace of inflammation in the form of tissue resident memory cells (TRM). They are originally responsible for protection against viral and bacterial infections in non-lymphatic tissues. In psoriatic inflammation, they are characterized by heterogeneity depending on their origin. CD8+ T cells TRM are abundantly present in psoriatic epidermis, while CD4+ TRM preferentially populate the dermis. In psoriasis, epidermal CD8+ TRM cells express CLA, CCR6, CD103 and IL-23R antigen and produce IL-17A during ex vivo stimulation. However, CD4+ CD103+ TRM can also colonize the epidermis and produce IL-22 during stimulation. Besides T cells, Th22 and epidermal DCs proved that epidermal cells in healed skin were still present and functioning after several years of disease remission. It explains the clinical phenomenon of the tendency of psoriatic lesions to relapse in the same location and it allows to develop new therapeutic strategies in the future.
Collapse
Affiliation(s)
- Agnieszka Owczarczyk-Saczonek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, The University of Warmia and Mazury, Al. Wojska Polskiego 30, 10-229 Olsztyn, Poland; (M.K.-F.); (W.P.)
- Correspondence: ; Tel.: +48-89-678-6670; Fax: +48-89-678-6675
| | - Magdalena Krajewska-Włodarczyk
- Department of Rheumatology, Municipal Hospital in Olsztyn, 10-229 Olsztyn, Poland;
- Department of Internal Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland
| | - Marta Kasprowicz-Furmańczyk
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, The University of Warmia and Mazury, Al. Wojska Polskiego 30, 10-229 Olsztyn, Poland; (M.K.-F.); (W.P.)
| | - Waldemar Placek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, The University of Warmia and Mazury, Al. Wojska Polskiego 30, 10-229 Olsztyn, Poland; (M.K.-F.); (W.P.)
| |
Collapse
|
60
|
Abstract
The skin is the outermost organ of the body and is exposed to many kinds of external pathogens. To manage this, the skin contains multiple types of immune cells. To achieve sufficient induction of cutaneous adaptive immune responses, the antigen presentation/recognition in the skin is an essential process. Recent studies have expanded our knowledge of how T cells survey their cognate antigens in the skin. In addition, the formation of a lymphoid cluster, named inducible skin-associated lymphoid tissue (iSALT), has been reported during skin inflammation. Although iSALT may not be classified as a typical tertiary lymphoid organ, it provides specific antigen presentation sites in the skin. In this article, we provide an overview of the antigen presentation mechanism in the skin, with a focus on the development of iSALT and its function.
Collapse
Affiliation(s)
- Gyohei Egawa
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
61
|
Generation of protective pneumococcal-specific nasal resident memory CD4 + T cells via parenteral immunization. Mucosal Immunol 2020; 13:172-182. [PMID: 31659300 PMCID: PMC6917870 DOI: 10.1038/s41385-019-0218-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 09/23/2019] [Accepted: 10/04/2019] [Indexed: 02/08/2023]
Abstract
The generation of tissue-resident memory T cells (TRM) is an essential aspect of immunity at mucosal surfaces, and it has been suggested that preferential generation of TRM is one of the principal advantages of mucosally administered vaccines. We have previously shown that antigen-specific, IL-17-producing CD4+ T cells can provide capsular antibody-independent protection against nasal carriage of Streptococcus pneumoniae; but whether pneumococcus-responsive TRM are localized within the nasal mucosa and are sufficient for protection from carriage has not been determined. Here, we show that intranasal administration of live or killed pneumococci to mice generates pneumococcus-responsive IL-17A-producing CD4+ mucosal TRM. Furthermore, we show that these cells are sufficient to mediate long-lived, neutrophil-dependent protection against subsequent pneumococcal nasal challenge. Unexpectedly, and in contrast with the prevailing paradigm, we found that parenteral administration of killed pneumococci also generates protective IL-17A+CD4+ TRM in the nasal mucosa. These results demonstrate a critical and sufficient role of TRM in prevention of pneumococcal colonization, and further that these cells can be generated by parenteral immunization. Our findings therefore have important implications regarding the generation of immune protection at mucosal surfaces by vaccination.
Collapse
|
62
|
A role for the CCR5-CCL5 interaction in the preferential migration of HSV-2-specific effector cells to the vaginal mucosa upon nasal immunization. Mucosal Immunol 2019; 12:1391-1403. [PMID: 31551493 DOI: 10.1038/s41385-019-0203-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 08/09/2019] [Accepted: 08/25/2019] [Indexed: 02/04/2023]
Abstract
Our current study focused on elucidating the role of specific chemokine-receptor interactions in antigen (Ag)-specific immune cell migration from nasal to genital mucosal tissues. This cellular migration is critical to induce effective Ag-specific immune responses against sexually transmitted genital infections. In this study, nasal immunization with live attenuated HSV-2 TK- induced the upregulation of CCR5 expression in effector immune cells, including CD4+ T cells, in Ag-priming sites and vaginal tissue. The CCR5 ligands CCL3, CCL4, and CCL5 all showed upregulated expression in vaginal tissue; in particular, CCL5 expression was highly enhanced in the stromal cells of vaginal tissue after nasal immunization. Intravaginal blockade of CCL5 by using neutralizing antibody diminished the number of HSV-2-specific effector cells in the vagina. Furthermore, loss of CCR5, a receptor for CCL5, impaired the migration of nasally primed Ag-specific effector cells from the airway to vagina. Effector cells adoptively transferred from CCR5-deficient mice failed to migrate into vaginal tissue, consequently increasing recipient mice's susceptibility to HSV-2 vaginal infection. These results indicate that the CCR5-CCL5 chemokine pathway is required for the migration and retention of nasally primed Ag-specific effector cells in vagina for providing protective immunity against HSV-2 infection.
Collapse
|
63
|
Knight FC, Gilchuk P, Kumar A, Becker KW, Sevimli S, Jacobson ME, Suryadevara N, Wang-Bishop L, Boyd KL, Crowe JE, Joyce S, Wilson JT. Mucosal Immunization with a pH-Responsive Nanoparticle Vaccine Induces Protective CD8 + Lung-Resident Memory T Cells. ACS NANO 2019; 13:10939-10960. [PMID: 31553872 PMCID: PMC6832804 DOI: 10.1021/acsnano.9b00326] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Tissue-resident memory T cells (TRM) patrol nonlymphoid organs and provide superior protection against pathogens that commonly infect mucosal and barrier tissues, such as the lungs, intestine, liver, and skin. Thus, there is a need for vaccine technologies that can induce a robust, protective TRM response in these tissues. Nanoparticle (NP) vaccines offer important advantages over conventional vaccines; however, there has been minimal investigation into the design of NP-based vaccines for eliciting TRM responses. Here, we describe a pH-responsive polymeric nanoparticle vaccine for generating antigen-specific CD8+ TRM cells in the lungs. With a single intranasal dose, the NP vaccine elicited airway- and lung-resident CD8+ TRM cells and protected against respiratory virus challenge in both sublethal (vaccinia) and lethal (influenza) infection models for up to 9 weeks after immunization. In elucidating the contribution of material properties to the resulting TRM response, we found that the pH-responsive activity of the carrier was important, as a structurally analogous non-pH-responsive control carrier elicited significantly fewer lung-resident CD8+ T cells. We also demonstrated that dual-delivery of protein antigen and nucleic acid adjuvant on the same NP substantially enhanced the magnitude, functionality, and longevity of the antigen-specific CD8+ TRM response in the lungs. Compared to administration of soluble antigen and adjuvant, the NP also mediated retention of vaccine cargo in pulmonary antigen-presenting cells (APCs), enhanced APC activation, and increased production of TRM-related cytokines. Overall, these data suggest a promising vaccine platform technology for rapid generation of protective CD8+ TRM cells in the lungs.
Collapse
Affiliation(s)
- Frances C. Knight
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Pavlo Gilchuk
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Amrendra Kumar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Kyle W. Becker
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Sema Sevimli
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Max E. Jacobson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Naveenchandra Suryadevara
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Lihong Wang-Bishop
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Kelli L. Boyd
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - James E. Crowe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Chemical and Physical Biology Program, Vanderbilt University, Nashville, TN 37235, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sebastian Joyce
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - John T. Wilson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Corresponding Author:
| |
Collapse
|
64
|
Griss J, Bauer W, Wagner C, Simon M, Chen M, Grabmeier-Pfistershammer K, Maurer-Granofszky M, Roka F, Penz T, Bock C, Zhang G, Herlyn M, Glatz K, Läubli H, Mertz KD, Petzelbauer P, Wiesner T, Hartl M, Pickl WF, Somasundaram R, Steinberger P, Wagner SN. B cells sustain inflammation and predict response to immune checkpoint blockade in human melanoma. Nat Commun 2019; 10:4186. [PMID: 31519915 PMCID: PMC6744450 DOI: 10.1038/s41467-019-12160-2] [Citation(s) in RCA: 264] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 08/22/2019] [Indexed: 01/01/2023] Open
Abstract
Tumor associated inflammation predicts response to immune checkpoint blockade in human melanoma. Current theories on regulation of inflammation center on anti-tumor T cell responses. Here we show that tumor associated B cells are vital to melanoma associated inflammation. Human B cells express pro- and anti-inflammatory factors and differentiate into plasmablast-like cells when exposed to autologous melanoma secretomes in vitro. This plasmablast-like phenotype can be reconciled in human melanomas where plasmablast-like cells also express T cell-recruiting chemokines CCL3, CCL4, CCL5. Depletion of B cells in melanoma patients by anti-CD20 immunotherapy decreases tumor associated inflammation and CD8+ T cell numbers. Plasmablast-like cells also increase PD-1+ T cell activation through anti-PD-1 blockade in vitro and their frequency in pretherapy melanomas predicts response and survival to immune checkpoint blockade. Tumor associated B cells therefore orchestrate and sustain melanoma inflammation and may represent a predictor for survival and response to immune checkpoint blockade therapy.
Collapse
Affiliation(s)
- Johannes Griss
- Department of Dermatology, Medical University of Vienna, 1090, Vienna, Austria.
- EMBL-European Bioinformatics Institute, Wellcome Trust Genome Campus, CB10 1SD Hinxton, Cambridge, UK.
| | - Wolfgang Bauer
- Department of Dermatology, Medical University of Vienna, 1090, Vienna, Austria
| | - Christine Wagner
- Department of Dermatology, Medical University of Vienna, 1090, Vienna, Austria
| | - Martin Simon
- Department of Dermatology, Medical University of Vienna, 1090, Vienna, Austria
| | - Minyi Chen
- Department of Dermatology, Medical University of Vienna, 1090, Vienna, Austria
| | - Katharina Grabmeier-Pfistershammer
- Department of Dermatology, Medical University of Vienna, 1090, Vienna, Austria
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Margarita Maurer-Granofszky
- Department of Dermatology, Medical University of Vienna, 1090, Vienna, Austria
- Children's Cancer Research Institute, 1090, Vienna, Austria
| | - Florian Roka
- Department of Dermatology, Medical University of Vienna, 1090, Vienna, Austria
| | - Thomas Penz
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Gao Zhang
- Molecular & Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA, 19104-4265, USA
- Department of Neurosurgery & The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, 27710, USA
| | - Meenhard Herlyn
- Molecular & Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA, 19104-4265, USA
| | - Katharina Glatz
- Institute of Pathology, University Hospital Basel, 4031, Basel, Switzerland
| | - Heinz Läubli
- Division of Medical Oncology, University Hospital Basel, 4031, Basel, Switzerland
| | - Kirsten D Mertz
- Institute of Pathology, Cantonal Hospital Baselland, 4410, Liestal, Switzerland
| | - Peter Petzelbauer
- Department of Dermatology, Medical University of Vienna, 1090, Vienna, Austria
| | - Thomas Wiesner
- Department of Dermatology, Medical University of Vienna, 1090, Vienna, Austria
| | - Markus Hartl
- Mass Spectrometry Facility, Max F. Perutz Laboratories (MFPL), University of Vienna, Vienna BioCenter (VBC), 1030, Vienna, Austria
| | - Winfried F Pickl
- Division of Cellular Immunology and Immunohematology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Rajasekharan Somasundaram
- Molecular & Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA, 19104-4265, USA
| | - Peter Steinberger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Stephan N Wagner
- Department of Dermatology, Medical University of Vienna, 1090, Vienna, Austria.
| |
Collapse
|
65
|
Mita Y, Kimura MY, Hayashizaki K, Koyama-Nasu R, Ito T, Motohashi S, Okamoto Y, Nakayama T. Crucial role of CD69 in anti-tumor immunity through regulating the exhaustion of tumor-infiltrating T cells. Int Immunol 2019; 30:559-567. [PMID: 30085193 DOI: 10.1093/intimm/dxy050] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/04/2018] [Indexed: 12/20/2022] Open
Abstract
The introduction of immune checkpoint inhibitors in cancer treatment highlights the negative regulation of anti-tumor immunity, such as effector T-cell exhaustion in the tumor microenvironment. However, the mechanisms underlying the induction and prevention of T-cell exhaustion remain largely unknown. We found that CD69, a type II glycoprotein known to regulate inflammation through T-cell migration and retention in tissues, plays an important role in inducing the exhaustion of tumor-infiltrating T cells. Cd69-/- mice showed reduced tumor growth and metastasis in a 4T1-luc2 murine breast cancer model, in which increased numbers of tumor-infiltrating lymphocytes, relatively little T-cell exhaustion, and enhanced IFNγ production were observed. Anti-CD69 monoclonal antibody treatment attenuated the T-cell exhaustion and tumor progression in tumor-bearing mice. These findings highlight a novel role of CD69 in controlling the tumor immune escape mediated by T-cell exhaustion and indicate that CD69 is a novel target for cancer immunotherapy.
Collapse
Affiliation(s)
- Yukiyoshi Mita
- Department of Immunology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba, Japan.,Department of Otorhinolaryngology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba, Japan
| | - Motoko Y Kimura
- Department of Immunology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba, Japan.,Department of Medical Immunology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba, Japan
| | - Koji Hayashizaki
- Department of Immunology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba, Japan
| | - Ryo Koyama-Nasu
- Department of Immunology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba, Japan
| | - Toshihiro Ito
- Department of Immunology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba, Japan
| | - Shinichiro Motohashi
- Department of Medical Immunology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba, Japan
| | - Yoshitaka Okamoto
- Department of Otorhinolaryngology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba, Japan
| |
Collapse
|
66
|
Pattacini L, Woodward Davis A, Czartoski J, Mair F, Presnell S, Hughes SM, Hyrien O, Lentz GM, Kirby AC, Fialkow MF, Hladik F, Prlic M, Lund JM. A pro-inflammatory CD8+ T-cell subset patrols the cervicovaginal tract. Mucosal Immunol 2019; 12:1118-1129. [PMID: 31312028 PMCID: PMC6717561 DOI: 10.1038/s41385-019-0186-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/11/2019] [Accepted: 06/14/2019] [Indexed: 02/04/2023]
Abstract
The immune system of the cervicovaginal tract (CVT) must balance immunosurveillance and active immunity against pathogens with maintenance of tolerance to resident microbiota and to fetal and partner antigens for reproductive purposes. Thus, we predicted that CVT immunity is characterized by distinctive features compared to blood and other tissue compartments. Indeed, we found that CVT CD8+ T-cells had unique transcriptional profiles, particularly in their cytokine signature, compared to that reported for CD8+ T-cells in other tissue sites. Among these CVT CD8+ T-cells, we identified a CD69- CD103- subset that was characterized by reduced migration in response to tissue-exit signals and higher pro-inflammatory potential as compared to their blood counterpart. These inflammatory mucosal CD8+ T-cells (Tim) were increased in frequency in the CVT of individuals with chronic infection, pointing to a potential role in perpetuating inflammation. Our findings highlight the specialized nature of immunity within the CVT and identify Tim cells as potential therapeutic targets to tame tissue inflammation upon chronic infection.
Collapse
Affiliation(s)
- Laura Pattacini
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, U.S.A
| | - Amanda Woodward Davis
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, U.S.A
| | - Julie Czartoski
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, U.S.A
| | - Florian Mair
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, U.S.A
| | - Scott Presnell
- System Immunology Division, Benaroya Research Institute, Seattle, WA, U.S.A
| | - Sean M. Hughes
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, U.S.A
| | - Ollivier Hyrien
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, U.S.A.,Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, U.S.A
| | - Gretchen M. Lentz
- Departments of Obstetrics and Gynecology, and Medicine, University of Washington, Seattle, WA, U.S.A
| | - Anna C. Kirby
- Departments of Obstetrics and Gynecology, and Medicine, University of Washington, Seattle, WA, U.S.A
| | - Michael F. Fialkow
- Departments of Obstetrics and Gynecology, and Medicine, University of Washington, Seattle, WA, U.S.A
| | - Florian Hladik
- Departments of Obstetrics and Gynecology, and Medicine, University of Washington, Seattle, WA, U.S.A
| | - Martin Prlic
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, U.S.A
| | - Jennifer M. Lund
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, U.S.A.,Department of Global Health, University of Washington, Seattle, WA, U.S.A
| |
Collapse
|
67
|
Garaud S, Buisseret L, Solinas C, Gu-Trantien C, de Wind A, Van den Eynden G, Naveaux C, Lodewyckx JN, Boisson A, Duvillier H, Craciun L, Ameye L, Veys I, Paesmans M, Larsimont D, Piccart-Gebhart M, Willard-Gallo K. Tumor infiltrating B-cells signal functional humoral immune responses in breast cancer. JCI Insight 2019; 5:129641. [PMID: 31408436 DOI: 10.1172/jci.insight.129641] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Tumor-infiltrating B-cells (TIL-B) in breast cancer (BC) have previously been associated with improved clinical outcomes; however, their role(s) in tumor immunity is not currently well known. This study confirms and extends the correlation between higher TIL-B densities and positive outcomes through an analysis of HER2-positive and triple-negative BC patients from the BIG 02-98 clinical trial (10yr mean follow-up). Fresh tissue analyses identify an increase in TIL-B density in untreated primary BC compared to normal breast tissues, which is associated with global, CD4+ and CD8+ TIL, higher tumor grades, higher proliferation and hormone receptor negativity. All B-cell differentiation stages are detectable but significant increases in memory TIL-B are consistently present. BC with higher infiltrates are specifically characterized by germinal center TIL-B, which in turn are correlated with TFH TIL and antibody-secreting TIL-B principally located in tertiary lymphoid structures. Some TIL-B also interact directly with tumor cells. Functional analyses reveal TIL-B are responsive to BCR stimulation ex vivo, express activation markers and produce cytokines and immunoglobulins despite reduced expression of the antigen-presenting molecules HLA-DR and CD40. Overall, these data support the concept that ongoing humoral immune responses are generated by TIL-B and help to generate effective anti-tumor immunity at the tumor site.
Collapse
Affiliation(s)
| | | | | | | | - Alexandre de Wind
- Department of Pathology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Gert Van den Eynden
- Department of Pathology, GZA Ziekenhuizen, Sint-Augustinus campus, Wilrijk, Belgium
| | | | | | | | | | - Ligia Craciun
- Department of Pathology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | - Denis Larsimont
- Department of Pathology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Martine Piccart-Gebhart
- Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | | |
Collapse
|
68
|
Life-long control of cytomegalovirus (CMV) by T resident memory cells in the adipose tissue results in inflammation and hyperglycemia. PLoS Pathog 2019; 15:e1007890. [PMID: 31220189 PMCID: PMC6605679 DOI: 10.1371/journal.ppat.1007890] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 07/02/2019] [Accepted: 06/03/2019] [Indexed: 12/13/2022] Open
Abstract
Cytomegalovirus (CMV) is a ubiquitous herpesvirus infecting most of the world’s population. CMV has been rigorously investigated for its impact on lifelong immunity and potential complications arising from lifelong infection. A rigorous adaptive immune response mounts during progression of CMV infection from acute to latent states. CD8 T cells, in large part, drive this response and have very clearly been demonstrated to take up residence in the salivary gland and lungs of infected mice during latency. However, the role of tissue resident CD8 T cells as an ongoing defense mechanism against CMV has not been studied in other anatomical locations. Therefore, we sought to identify additional locations of anti-CMV T cell residency and the physiological consequences of such a response. Through RT-qPCR we found that mouse CMV (mCMV) infected the visceral adipose tissue and that this resulted in an expansion of leukocytes in situ. We further found, through flow cytometry, that adipose tissue became enriched in cytotoxic CD8 T cells that are specific for mCMV antigens from day 7 post infection through the lifespan of an infected animal (> 450 days post infection) and that carry markers of tissue residence. Furthermore, we found that inflammatory cytokines are elevated alongside the expansion of CD8 T cells. Finally, we show a correlation between the inflammatory state of adipose tissue in response to mCMV infection and the development of hyperglycemia in mice. Overall, this study identifies adipose tissue as a location of viral infection leading to a sustained and lifelong adaptive immune response mediated by CD8 T cells that correlates with hyperglycemia. These data potentially provide a mechanistic link between metabolic syndrome and chronic infection. Mouse cytomegalovirus (mCMV) infection results in initial systemic viremia that is thereafter controlled by the adaptive immune system. Control is mediated in part by T cells that render the virus undetectable systemically, and latent in specific organs, including the lungs and salivary glands. It remains unclear how latent virus is controlled across tissues given the large pool of systemic mCMV-specific T cells. We explored mCMV control in the adipose tissue, whose cellular constituents are potentially susceptible to infection. We found that mCMV infects the adipose tissue during the acute phase, causing local inflammation and a lifelong mCMV-specific CD8 T cell immune response. The response consisted largely from non-recirculating, tissue-resident T cells. The infected adipose tissue showed signs of metabolic changes, that may potentially predispose the infected host to metabolic dysregulation as evidenced by hyperglycemia. Accumulation and persistence of mCMV specific non-circulating resident CD8 T cells (Trm) in adipose tissue reveal a likely generalized mechanism of mCMV tissue reservoir control by Trm cells and identify the adipose tissue as a persistent mCMV reservoir, with potential implications for metabolic health.
Collapse
|
69
|
Zhang F, Yang T, Ao H, Zhai L, Tan Z, Wang Y, Xing K, Zhao X, Wang Z, Yu Y, Wang C. Novel nucleotide variants in SLA-DOB and CD4 are associated with immune traits in pregnant sows. Gene 2019; 707:22-29. [PMID: 31026568 DOI: 10.1016/j.gene.2019.04.057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/28/2019] [Accepted: 04/19/2019] [Indexed: 11/17/2022]
Abstract
Reinforcing the immunity of pregnant sows can not only improve their own health condition but also increase the survival rate and healthy status of their piglets. This study aims to find single-nucleotide polymorphism (SNP) and molecular markers that are associated with the immune traits of pregnant sows. SLA-DOB and CD4 were selected as candidate genes, and blood samples were randomly collected from pregnant Landrace sows and used to detect T-lymphocyte subsets, interferon alpha, interleukin 6, Toll-like receptor 3, serum antibody immunoglobulin G, and porcine reproductive and respiratory syndrome virus-specific antibody. Then, association analyses were conducted for the polymorphic sites of candidate genes with immune traits. We found 12 mutations in the two genes and conducted an association study with eight of them. Our results indicated that among the eight mutations, SNP1, SNP2, and SNP3 of the SLA-DOB gene and Ins9, SNP10, and SNP11 in the CD4 gene are newly discovered mutations. Except for SNP1, SNP3, and SNP11, the other five SNPs are associated with at least one immune trait tested. Especially, SNP2 and Ins9 are significantly associated with at least one of the T-lymphocyte subgroups and at least one antibody. These novel mutations have potential important effects on the polymorphic loci of the above immune traits in pregnant sows. The results suggest that the SLA-DOB and CD4 genes and their genetic mutations can be considered as important candidate genes and mutations for the immunity of pregnant sows.
Collapse
Affiliation(s)
- Fengxia Zhang
- National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, Beijing, China
| | - Ting Yang
- National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, Beijing, China
| | - Hong Ao
- The State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Liwei Zhai
- National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, Beijing, China
| | - Zhen Tan
- National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, Beijing, China
| | - Yuan Wang
- National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, Beijing, China
| | - Kai Xing
- National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, Beijing, China
| | - Xitong Zhao
- National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, Beijing, China
| | - Zhiquan Wang
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Ying Yu
- National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, Beijing, China..
| | - Chuduan Wang
- National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, Beijing, China..
| |
Collapse
|
70
|
Takamura S, Kohlmeier JE. Establishment and Maintenance of Conventional and Circulation-Driven Lung-Resident Memory CD8 + T Cells Following Respiratory Virus Infections. Front Immunol 2019; 10:733. [PMID: 31024560 PMCID: PMC6459893 DOI: 10.3389/fimmu.2019.00733] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/19/2019] [Indexed: 12/30/2022] Open
Abstract
Antigen-specific CD8+ tissue-resident memory T cells (TRM cells) persist in the lung following resolution of a respiratory virus infection and provide first-line defense against reinfection. In contrast to other memory T cell populations, such as central memory T cells that circulate between lymph and blood, and effector memory T cells (TEM cells) that circulate between blood and peripheral tissues, TRM cells are best defined by their permanent residency in the tissues and their independence from circulatory T cell populations. Consistent with this, we recently demonstrated that CD8+ TRM cells primarily reside within specific niches in the lung (Repair-Associated Memory Depots; RAMD) that normally exclude CD8+ TEM cells. However, it has also been reported that circulating CD8+ TEM cells continuously convert into CD8+ TRM cells in the lung interstitium, helping to sustain TRM numbers. The relative contributions of these two mechanisms of CD8+ TRM cells maintenance in the lung has been the source of vigorous debate. Here we propose a model in which the majority of CD8+ TRM cells are maintained within RAMD (conventional TRM) whereas a small fraction of TRM are derived from circulating CD8+ TEM cells and maintained in the interstitium. The numbers of both types of TRM cells wane over time due to declines in both RAMD availability and the overall number of TEM in the circulation. This model is consistent with most published reports and has important implications for the development of vaccines designed to elicit protective T cell memory in the lung.
Collapse
Affiliation(s)
- Shiki Takamura
- Department of Immunology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Jacob E Kohlmeier
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
71
|
Equine Herpesvirus 1 Bridles T Lymphocytes To Reach Its Target Organs. J Virol 2019; 93:JVI.02098-18. [PMID: 30651370 PMCID: PMC6430527 DOI: 10.1128/jvi.02098-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 01/03/2019] [Indexed: 11/20/2022] Open
Abstract
Equine herpesvirus 1 (EHV1) is an ancestral alphaherpesvirus that is related to herpes simplex virus 1 and causes respiratory, reproductive, and neurological disorders in Equidae. EHV1 is indisputably a master at exploiting leukocytes to reach its target organs, accordingly evading the host immunity. However, the role of T lymphocytes in cell-associated viremia remains poorly understood. Here we show that activated T lymphocytes efficiently become infected and support viral replication despite the presence of protective immunity. We demonstrate a restricted expression of viral proteins on the surfaces of infected T cells, which prevents immune recognition. In addition, we indicate a hampered release of progeny, which results in the accumulation of nucleocapsids in the T cell nucleus. Upon engagement with the target endothelium, late viral proteins orchestrate viral synapse formation and viral transfer to the contact cell. Our findings have significant implications for the understanding of EHV1 pathogenesis, which is essential for developing innovative therapies to prevent the devastating clinical symptoms of infection. Equine herpesvirus 1 (EHV1) replicates in the respiratory epithelium and disseminates through the body via a cell-associated viremia in leukocytes, despite the presence of neutralizing antibodies. “Hijacked” leukocytes, previously identified as monocytic cells and T lymphocytes, transmit EHV1 to endothelial cells of the endometrium or central nervous system, causing reproductive (abortigenic variants) or neurological (neurological variants) disorders. In the present study, we questioned the potential route of EHV1 infection of T lymphocytes and how EHV1 misuses T lymphocytes as a vehicle to reach the endothelium of the target organs in the absence or presence of immune surveillance. Viral replication was evaluated in activated and quiescent primary T lymphocytes, and the results demonstrated increased infection of activated versus quiescent, CD4+ versus CD8+, and blood- versus lymph node-derived T cells. Moreover, primarily infected respiratory epithelial cells and circulating monocytic cells efficiently transferred virions to T lymphocytes in the presence of neutralizing antibodies. Albeit T-lymphocytes express all classes of viral proteins early in infection, the expression of viral glycoproteins on their cell surface was restricted. In addition, the release of viral progeny was hampered, resulting in the accumulation of viral nucleocapsids in the T cell nucleus. During contact of infected T lymphocytes with endothelial cells, a late viral protein(s) orchestrates T cell polarization and synapse formation, followed by anterograde dynein-mediated transport and transfer of viral progeny to the engaged cell. This represents a sophisticated but efficient immune evasion strategy to allow transfer of progeny virus from T lymphocytes to adjacent target cells. These results demonstrate that T lymphocytes are susceptible to EHV1 infection and that cell-cell contact transmits infectious virus to and from T lymphocytes. IMPORTANCE Equine herpesvirus 1 (EHV1) is an ancestral alphaherpesvirus that is related to herpes simplex virus 1 and causes respiratory, reproductive, and neurological disorders in Equidae. EHV1 is indisputably a master at exploiting leukocytes to reach its target organs, accordingly evading the host immunity. However, the role of T lymphocytes in cell-associated viremia remains poorly understood. Here we show that activated T lymphocytes efficiently become infected and support viral replication despite the presence of protective immunity. We demonstrate a restricted expression of viral proteins on the surfaces of infected T cells, which prevents immune recognition. In addition, we indicate a hampered release of progeny, which results in the accumulation of nucleocapsids in the T cell nucleus. Upon engagement with the target endothelium, late viral proteins orchestrate viral synapse formation and viral transfer to the contact cell. Our findings have significant implications for the understanding of EHV1 pathogenesis, which is essential for developing innovative therapies to prevent the devastating clinical symptoms of infection.
Collapse
|
72
|
Mobilization of CD4+ T lymphocytes in inflamed mucosa reduces pain in colitis mice: toward a vaccinal strategy to alleviate inflammatory visceral pain. Pain 2019; 159:331-341. [PMID: 29140925 DOI: 10.1097/j.pain.0000000000001103] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
T lymphocytes play a pivotal role in endogenous regulation of inflammatory visceral pain. The analgesic activity of T lymphocytes is dependent on their production of opioids, a property acquired on antigen activation. Accordingly, we investigated whether an active recruitment of T lymphocytes within inflamed colon mucosa via a local vaccinal strategy may counteract inflammation-induced visceral pain in mice. Mice were immunized against ovalbumin (OVA). One month after immunization, colitis was induced by adding 3% (wt/vol) dextran sulfate sodium into drinking water containing either cognate antigen OVA or control antigen bovine serum albumin for 5 days. Noncolitis OVA-primed mice were used as controls. Visceral sensitivity was then determined by colorectal distension. Oral administration of OVA but not bovine serum albumin significantly reduced dextran sulfate sodium-induced abdominal pain without increasing colitis severity in OVA-primed mice. Analgesia was dependent on local release of enkephalins by effector anti-OVA T lymphocytes infiltrating the inflamed mucosa. The experiments were reproduced with the bacillus Calmette-Guerin vaccine as antigen. Similarly, inflammatory visceral pain was dramatically alleviated in mice vaccinated against bacillus Calmette-Guerin and then locally administered with live Mycobacterium bovis. Together, these results show that the induction of a secondary adaptive immune response against vaccine antigens in inflamed mucosa may constitute a safe noninvasive strategy to relieve from visceral inflammatory pain.
Collapse
|
73
|
Kim Y, Shin Y, Kang GH. Prognostic significance of CD103+ immune cells in solid tumor: a systemic review and meta-analysis. Sci Rep 2019; 9:3808. [PMID: 30846807 PMCID: PMC6405906 DOI: 10.1038/s41598-019-40527-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 02/19/2019] [Indexed: 11/13/2022] Open
Abstract
CD103 is a transmembrane heterodimer complex that mediates cell adhesion, migration, and lymphocyte homing of cell through interaction with E-cadherin. Recently, CD103+ immune cells in human carcinoma has been investigated as a prognostic factor, however, the correlation between CD103+ immune cells and survival are still elusive. Therefore, a meta-analysis was performed to determine the prognostic value of CD103+ immune cells in solid tumor. Studies relevant to the subject was searched from PubMed, Embase, and Web of Science. Ten studies including 2,824 patients were eligible for the analysis. Tumors positive for CD103+ immune cells were associated with favorable overall survival, disease-free survival, and disease-specific survival. Subgroup analysis revealed that assessing CD103+ immune cells in epithelial and total (both epithelial and stromal) areas or using whole slide section were associated with good prognosis. Furthermore, stromal CD103+ immune cells or CD103+ immune cells evaluated by tissue microarrays were not always significantly prognostic. In conclusion, these results show that CD103+ immune cells are associated with prognosis in solid tumor. However, the region of assessment and selection of material for the evaluation could affect the value of CD103 as a prognostic biomarker.
Collapse
Affiliation(s)
- Younghoon Kim
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea.,Department of Pathology, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Yunjoo Shin
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Gyeong Hoon Kang
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea. .,Department of Pathology, Seoul National University College of Medicine, Seoul, 03080, Korea.
| |
Collapse
|
74
|
Schøller AS, Fonnes M, Nazerai L, Christensen JP, Thomsen AR. Local Antigen Encounter Is Essential for Establishing Persistent CD8 + T-Cell Memory in the CNS. Front Immunol 2019; 10:351. [PMID: 30886617 PMCID: PMC6409353 DOI: 10.3389/fimmu.2019.00351] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 02/11/2019] [Indexed: 12/11/2022] Open
Abstract
While the brain is considered an immune-privileged site, the CNS may nevertheless be the focus of immune mediated inflammation in the case of infection and certain autoimmune diseases, e.g., multiple sclerosis. As in other tissues, it has been found that acute T-cell infiltration may be followed by establishment of persistent local T-cell memory. To improve our understanding regarding the regulation of putative tissue resident memory T (Trm) cells in CNS, we devised a new model system for studying the generation of Trm cells in this site. To this purpose, we exploited the fact that the CNS may be a sanctuary for adenoviral infection, and to minimize virus-induced disease, we chose replication-deficient adenoviruses for infection of the CNS. Non-replicating adenoviruses are known to be highly immunogenic, and our studies demonstrate that intracerebral inoculation causes marked local T-cell recruitment, which is followed by persistent infiltration of the CNS parenchyma by antigen specific CD8+ T cells. Phenotypical analysis of CNS infiltrating antigen specific CD8+ T cells was consistent with these cells being Trms. Regarding the long-term stability of the infiltrate, resident CD8+ T cells expressed high levels of the anti-apoptotic molecule Bcl-2 as well as the proliferation marker Ki-67 suggesting that the population is maintained through steady homeostatic proliferation. Functionally, memory CD8+ T cells from CNS matched peripheral memory cells with regard to capacity for ex vivo cytotoxicity and cytokine production. Most importantly, our experiments revealed a key role for local antigen encounter in the establishment of sustained CD8+ T-cell memory in the brain. Inflammation in the absence of cognate antigen only led to limited and transient infiltration by antigen specific CD8+ T cells. Together these results indicate that memory CD8+ T cells residing in the CNS predominantly mirror previous local infections and immune responses to local autoantigens.
Collapse
Affiliation(s)
- Amalie S Schøller
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Masja Fonnes
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Loulieta Nazerai
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Jan P Christensen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Allan R Thomsen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
75
|
Schnabel CL, Babasyan S, Freer H, Wagner B. CXCL10 production in equine monocytes is stimulated by interferon-gamma. Vet Immunol Immunopathol 2019; 207:25-30. [DOI: 10.1016/j.vetimm.2018.11.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 11/21/2018] [Accepted: 11/23/2018] [Indexed: 12/01/2022]
|
76
|
Olson GS, Moore SW, Richter JM, Garber JJ, Bowman BA, Rawlings CA, Flagg M, Corleis B, Kwon DS. Increased frequency of systemic pro-inflammatory Vδ1 + γδ T cells in HIV elite controllers correlates with gut viral load. Sci Rep 2018; 8:16471. [PMID: 30405182 PMCID: PMC6220338 DOI: 10.1038/s41598-018-34576-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 10/17/2018] [Indexed: 12/11/2022] Open
Abstract
γδ T cells predominate in the intestinal mucosa and help maintain gut homeostasis and mucosal immunity. Although HIV infection significantly alters these cells, what drives these perturbations is unclear. Growing evidence suggests that impaired intestinal immune function in HIV leads to chronic immune activation and disease progression. This occurs even in HIV controllers - individuals with undetectable HIV viremia without antiretroviral therapy (ART). We show that Vδ1+ cells, a subset of γδ T cells described as being important in intestinal barrier function, increase in frequency in HIV-infected individuals, including HIV controllers. These cells resemble terminally differentiated effector memory cells, producing the pro-inflammatory cytokines IFNγ, TNFα, and MIP-1β upon stimulation. Importantly, pro-inflammatory Vδ1+ cell frequency correlates with levels of HIV RNA in intestinal tissue but not in plasma. This study supports a model in which local viral replication in the gut in HIV controllers disrupts the phenotype and function of Vδ1+ cells, a cell type involved in the maintenance of epithelial barrier integrity, and may thereby contribute to systemic immune activation and HIV disease progression.
Collapse
Affiliation(s)
- Gregory S Olson
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Sarah W Moore
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - James M Richter
- Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - John J Garber
- Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Brittany A Bowman
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Crystal A Rawlings
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Meaghan Flagg
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Björn Corleis
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Douglas S Kwon
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America.
| |
Collapse
|
77
|
Li G, Teleki C, Wang T. Memory T Cells in Flavivirus Vaccination. Vaccines (Basel) 2018; 6:E73. [PMID: 30340377 PMCID: PMC6313919 DOI: 10.3390/vaccines6040073] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/11/2018] [Accepted: 10/12/2018] [Indexed: 12/30/2022] Open
Abstract
Flaviviruses include many medically important viruses, such as Dengue virus (DENV), Japanese encephalitis (JEV), tick-borne encephalitis (TBEV), West Nile (WNV), yellow fever (YFV), and Zika viruses (ZIKV). Currently, there are licensed human vaccines for DENV, JEV, TBEV and YFV, but not for WNV or ZIKV. Memory T cells play a central role in adaptive immunity and are important for host protection during flavivirus infection. In this review, we discuss recent findings from animal models and clinical trials and provide new insights into the role of memory T cells in host protective immunity upon vaccination with the licensed flavivirus vaccines.
Collapse
Affiliation(s)
- Guangyu Li
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Cody Teleki
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Tian Wang
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Institute for Human Infections & Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
78
|
Mahanonda R, Champaiboon C, Subbalekha K, Sa‐Ard‐Iam N, Yongyuth A, Isaraphithakkul B, Rerkyen P, Charatkulangkun O, Pichyangkul S. Memory T cell subsets in healthy gingiva and periodontitis tissues. J Periodontol 2018; 89:1121-1130. [DOI: 10.1002/jper.17-0674] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 03/16/2018] [Accepted: 04/25/2018] [Indexed: 12/25/2022]
Affiliation(s)
- Rangsini Mahanonda
- Department of PeriodontologyFaculty of DentistryChulalongkorn University Bangkok Thailand
- Immunology LaboratoryFaculty of DentistryChulalongkorn University Bangkok Thailand
- Research Unit for Immunopathological / Clinical Research in Periodontal DiseaseFaculty of DentistryChulalongkorn University Bangkok Thailand
| | | | - Keskanya Subbalekha
- Department of Oral Maxillofacial SurgeryFaculty of DentistryChulalongkorn University Bangkok Thailand
| | - Noppadol Sa‐Ard‐Iam
- Immunology LaboratoryFaculty of DentistryChulalongkorn University Bangkok Thailand
- Research Unit for Immunopathological / Clinical Research in Periodontal DiseaseFaculty of DentistryChulalongkorn University Bangkok Thailand
| | - Arsarn Yongyuth
- Department of PeriodontologyFaculty of DentistryChulalongkorn University Bangkok Thailand
| | | | - Pimprapa Rerkyen
- Immunology LaboratoryFaculty of DentistryChulalongkorn University Bangkok Thailand
- Research Unit for Immunopathological / Clinical Research in Periodontal DiseaseFaculty of DentistryChulalongkorn University Bangkok Thailand
| | - Orawan Charatkulangkun
- Department of PeriodontologyFaculty of DentistryChulalongkorn University Bangkok Thailand
| | - Sathit Pichyangkul
- Department of PeriodontologyFaculty of DentistryChulalongkorn University Bangkok Thailand
| |
Collapse
|
79
|
Laban S, Suwandi JS, van Unen V, Pool J, Wesselius J, Höllt T, Pezzotti N, Vilanova A, Lelieveldt BPF, Roep BO. Heterogeneity of circulating CD8 T-cells specific to islet, neo-antigen and virus in patients with type 1 diabetes mellitus. PLoS One 2018; 13:e0200818. [PMID: 30089176 PMCID: PMC6082515 DOI: 10.1371/journal.pone.0200818] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 07/03/2018] [Indexed: 11/19/2022] Open
Abstract
Auto-reactive CD8 T-cells play an important role in the destruction of pancreatic β-cells resulting in type 1 diabetes (T1D). However, the phenotype of these auto-reactive cytolytic CD8 T-cells has not yet been extensively described. We used high-dimensional mass cytometry to phenotype autoantigen- (pre-proinsulin), neoantigen- (insulin-DRIP) and virus- (cytomegalovirus) reactive CD8 T-cells in peripheral blood mononuclear cells (PBMCs) of T1D patients. A panel of 33 monoclonal antibodies was designed to further characterise these cells at the single-cell level. HLA-A2 class I tetramers were used for the detection of antigen-specific CD8 T-cells. Using a novel Hierarchical Stochastic Neighbor Embedding (HSNE) tool (implemented in Cytosplore), we identified 42 clusters within the CD8 T-cell compartment of three T1D patients and revealed profound heterogeneity between individuals, as each patient displayed a distinct cluster distribution. Single-cell analysis of pre-proinsulin, insulin-DRIP and cytomegalovirus-specific CD8 T-cells showed that the detected specificities were heterogeneous between and within patients. These findings emphasize the challenge to define the obscure nature of auto-reactive CD8 T-cells.
Collapse
Affiliation(s)
- Sandra Laban
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Jessica S. Suwandi
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Vincent van Unen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Jos Pool
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Joris Wesselius
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Thomas Höllt
- Computational Biology Center, Leiden University Medical Center, Leiden, the Netherlands
- Computer Graphics and Visualization, Delft University of Technology, Delft, the Netherlands
| | - Nicola Pezzotti
- Computer Graphics and Visualization, Delft University of Technology, Delft, the Netherlands
| | - Anna Vilanova
- Computer Graphics and Visualization, Delft University of Technology, Delft, the Netherlands
| | | | - Bart O. Roep
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
- Department of Diabetes Immunology, Diabetes & Metabolism Research Institute at the Beckman Research Institute, City of Hope, Duarte, California, United States of America
- * E-mail:
| |
Collapse
|
80
|
Gatault P, Al-Hajj S, Noble J, Chevallier E, Piollet M, Forconi C, Gaudy-Graffin C, Thibault G, Miquelestorena-Standley E, Halimi JM, Büchler M, Lemoine R, Baron C. CMV-infected kidney grafts drive the expansion of blood-borne CMV-specific T cells restricted by shared class I HLA molecules via presentation on donor cells. Am J Transplant 2018; 18:1904-1913. [PMID: 29377506 DOI: 10.1111/ajt.14672] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 01/16/2018] [Accepted: 01/18/2018] [Indexed: 02/06/2023]
Abstract
We aimed to determine the role of cytomegalovirus (CMV)-infected donor cells in the development of a CMV-specific immune response in kidney transplant recipients. We assessed the CMV pp65-specific immune response by using interferon-ɣ ELISPOT and dextramers in peripheral blood mononuclear cells from 115 recipients (D+R- 31, D+R + 44, D-R + 40) late after transplantation (mean 59 ± 42 months). Receiving a kidney from a D+ donor resulted in a higher number of IFN-ɣ-producing anti-CMV T cells (P = .004). This effect disappeared with the absence of shared HLA class I specificities between donors and recipients (P = .430). To confirm the role of donor cells in stimulating the expansion of newly developed CMV-specific CD8+ T cells after transplantation, we compared the number of HLA-A2-restricted CMV-specific CD8+ T cells in primo-infected recipients who received an HLA-A2 or non-HLA-A2 graft. The median of anti-CMV pp65 T cells restricted by HLA-A2 was very low for patients who received a non-HLA-A2 graft vs an HLA-A2 graft (300 [0-14638] vs. 17972 [222-85594] anti-CMV pp65 CD8+ T cells/million CD8+ T cells, P = .001). This adds new evidence that CMV-infected kidney donor cells present CMV peptides and drive an inflation of memory CMV-specific CD8+ T cells, likely because of frequent CMV replications within the graft.
Collapse
Affiliation(s)
- Philippe Gatault
- EA4245 T2I Transplantation Immunologie Inflammation, University of Tours, Tours, France.,Service de Néphrologie et Immunologie Clinique, CHRU de Tours, Tours, France
| | - Sally Al-Hajj
- EA4245 T2I Transplantation Immunologie Inflammation, University of Tours, Tours, France
| | - Johan Noble
- EA4245 T2I Transplantation Immunologie Inflammation, University of Tours, Tours, France.,Service de Néphrologie et Immunologie Clinique, CHRU de Tours, Tours, France
| | - Eloi Chevallier
- EA4245 T2I Transplantation Immunologie Inflammation, University of Tours, Tours, France.,Service de Néphrologie et Immunologie Clinique, CHRU de Tours, Tours, France
| | - Marie Piollet
- EA4245 T2I Transplantation Immunologie Inflammation, University of Tours, Tours, France
| | - Catherine Forconi
- EA4245 T2I Transplantation Immunologie Inflammation, University of Tours, Tours, France
| | | | - Gilles Thibault
- Laboratory of Immunology Tours, CHRU de Tours, Tours, France
| | - Elodie Miquelestorena-Standley
- EA4245 T2I Transplantation Immunologie Inflammation, University of Tours, Tours, France.,Department of Pathology, CHRU de Tours, Tours, France
| | - Jean-Michel Halimi
- EA4245 T2I Transplantation Immunologie Inflammation, University of Tours, Tours, France.,Service de Néphrologie et Immunologie Clinique, CHRU de Tours, Tours, France
| | - Matthias Büchler
- EA4245 T2I Transplantation Immunologie Inflammation, University of Tours, Tours, France.,Service de Néphrologie et Immunologie Clinique, CHRU de Tours, Tours, France
| | - Roxane Lemoine
- EA4245 T2I Transplantation Immunologie Inflammation, University of Tours, Tours, France
| | - Christophe Baron
- EA4245 T2I Transplantation Immunologie Inflammation, University of Tours, Tours, France.,Service de Néphrologie et Immunologie Clinique, CHRU de Tours, Tours, France
| |
Collapse
|
81
|
Ratajczak W, Niedźwiedzka-Rystwej P, Tokarz-Deptuła B, Deptuła W. Immunological memory cells. Cent Eur J Immunol 2018; 43:194-203. [PMID: 30135633 PMCID: PMC6102609 DOI: 10.5114/ceji.2018.77390] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 02/16/2018] [Indexed: 02/03/2023] Open
Abstract
This article reviews immunological memory cells, currently represented by T and B lymphocytes and natural killer (NK) cells, which determine a rapid and effective response against a second encounter with the same antigen. Among T lymphocytes, functions of memory cells are provided by their subsets: central memory, effector memory, tissue-resident memory, regulatory memory and stem memory T cells. Memory T and B lymphocytes have an essential role in the immunity against microbial pathogens but are also involved in autoimmunity and maternal-fetal tolerance. Furthermore, the evidence of immunological memory has been established for NK cells. NK cells can respond to haptens or viruses, which results in generation of antigen-specific memory cells. T, B and NK cells, which have a role in immunological memory, have been characterized phenotypically and functionally. During the secondary immune response, these cells are involved in the reaction against foreign antigens, including pathogens, and take part in autoimmune diseases, but also are crucial to immunological tolerance and vaccine therapy.
Collapse
Affiliation(s)
- Weronika Ratajczak
- Scientific Circle of Microbiologists, Faculty of Biology, University of Szczecin, Szczecin, Poland
| | | | - Beata Tokarz-Deptuła
- Department of Immunology, Faculty of Biology, University of Szczecin, Szczecin, Poland
| | - Wiesław Deptuła
- Department of Microbiology, Faculty of Biology, University of Szczecin, Szczecin, Poland
| |
Collapse
|
82
|
Cantero J, Genescà M. Maximizing the immunological output of the cervicovaginal explant model. J Immunol Methods 2018; 460:26-35. [PMID: 29894750 DOI: 10.1016/j.jim.2018.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/16/2018] [Accepted: 06/06/2018] [Indexed: 12/24/2022]
Abstract
In the field of sexually transmitted infections (STI), the cervicovaginal explant (CVEx) model, not only provides the opportunity to study the different immunological arms present in these tissues under steady state conditions, but also their response against ex vivo infection with relevant pathogens. The methodology associated to the establishment of the HIV infection model in the cervicovaginal tissue was described in detail by Grivel et al. earlier (Grivel and Margolis, 2009). With this model as a foundation, we illustrate different approaches to obtain a large number of immunological readouts from a single piece of tissue, thus maximizing the immunological output obtained. Additionally, we discuss several ideas to study some of the immunological subsets present in this mucosal tissue by enriching them with the addition of distinct chemokines or specifically inducing their activation. Importantly, most of the methodology and concepts proposed here can be applied to study the immune subsets resident in other tissues. In the field of mucosal immunology, the possibility of studying resident immune subsets from tissue explants offers a great opportunity to understand the real players against invading pathogens and localized pathologies. Furthermore, this model allows for addressing the therapeutic benefit of modulating the activity of certain molecules and immune subsets against invading pathogens, which may infer their contribution to pathogen control and direct novel therapeutic interventions.
Collapse
Affiliation(s)
- Jon Cantero
- Department of Infectious Diseases, Hospital Universitari Vall d'Hebrón, Institut de Recerca (VHIR), 119-129 Passeig Vall d'Hebrón, 08035 Barcelona, Spain; Mucosal Immunology Unit, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Can Ruti Campus, Carretera de Can Ruti, camí de les escoles s/n, 08916 Badalona, Spain
| | - Meritxell Genescà
- Department of Infectious Diseases, Hospital Universitari Vall d'Hebrón, Institut de Recerca (VHIR), 119-129 Passeig Vall d'Hebrón, 08035 Barcelona, Spain; Mucosal Immunology Unit, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Can Ruti Campus, Carretera de Can Ruti, camí de les escoles s/n, 08916 Badalona, Spain.
| |
Collapse
|
83
|
Cruz MS, Diamond A, Russell A, Jameson JM. Human αβ and γδ T Cells in Skin Immunity and Disease. Front Immunol 2018; 9:1304. [PMID: 29928283 PMCID: PMC5997830 DOI: 10.3389/fimmu.2018.01304] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/25/2018] [Indexed: 12/14/2022] Open
Abstract
γδ T lymphocytes maintain skin homeostasis by balancing keratinocyte differentiation and proliferation with the destruction of infected or malignant cells. An imbalance in skin-resident T cell function can aggravate skin-related autoimmune diseases, impede tumor eradication, or disrupt proper wound healing. Much of the published work on human skin T cells attributes T cell function in the skin to αβ T cells, while γδ T cells are an often overlooked participant. This review details the roles played by both αβ and γδ T cells in healthy human skin and then focuses on their roles in skin diseases, such as psoriasis and alopecia areata. Understanding the contribution of skin-resident and skin-infiltrating T cell populations and cross-talk with other immune cells is leading to the development of novel therapeutics for patients. However, there is still much to be learned in order to effectively modulate T cell function and maintain healthy skin homeostasis.
Collapse
Affiliation(s)
| | | | | | - Julie Marie Jameson
- Department of Biological Sciences, California State University of San Marcos, San Marcos, CA, United States
| |
Collapse
|
84
|
Takamura S. Niches for the Long-Term Maintenance of Tissue-Resident Memory T Cells. Front Immunol 2018; 9:1214. [PMID: 29904388 PMCID: PMC5990602 DOI: 10.3389/fimmu.2018.01214] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 05/15/2018] [Indexed: 12/13/2022] Open
Abstract
Tissue-resident memory T cells (TRM cells) are a population of immune cells that reside in the lymphoid and non-lymphoid organs without recirculation through the blood. These important cells occupy and utilize unique anatomical and physiological niches that are distinct from those for other memory T cell populations, such as central memory T cells in the secondary lymphoid organs and effector memory T cells that circulate through the tissues. CD8+ TRM cells typically localize in the epithelial layers of barrier tissues where they are optimally positioned to act as sentinels to trigger antigen-specific protection against reinfection. CD4+ TRM cells typically localize below the epithelial layers, such as below the basement membrane, and cluster in lymphoid structures designed to optimize interactions with antigen-presenting cells upon reinfection. A key feature of TRM populations is their ability to be maintained in barrier tissues for prolonged periods of time. For example, skin CD8+ TRM cells displace epidermal niches originally occupied by γδ T cells, thereby enabling their stable persistence for years. It is also clear that the long-term maintenance of TRM cells in different microenvironments is dependent on multiple tissue-specific survival cues, although the specific details are poorly understood. However, not all TRM persist over the long term. Recently, we identified a new spatial niche for the maintenance of CD8+ TRM cells in the lung, which is created at the site of tissue regeneration after injury [termed repair-associated memory depots (RAMD)]. The short-lived nature of RAMD potentially explains the short lifespans of CD8+ TRM cells in this particular tissue. Clearly, a better understanding of the niche-dependent maintenance of TRM cells will be important for the development of vaccines designed to promote barrier immunity. In this review, we discuss recent advances in our understanding of the properties and nature of tissue-specific niches that maintain TRM cells in different tissues.
Collapse
Affiliation(s)
- Shiki Takamura
- Department of Immunology, Faculty of Medicine, Kindai University, Osaka, Japan
| |
Collapse
|
85
|
Wang YG, Long J, Shao DC, Song H. Hyperbaric oxygen inhibits production of CD3+ T cells in the thymus and facilitates malignant glioma cell growth. J Int Med Res 2018; 46:2780-2791. [PMID: 29785863 PMCID: PMC6124287 DOI: 10.1177/0300060518767796] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective Hyperbaric oxygen (HBO) is an emerging complementary alternative medical approach in glioma treatment. However, its mode of action is unknown, so this was investigated in the present study. Methods We constructed an intracranial glioma model of congenic C57BL/6J mice. Glioma growth under HBO stimulation was assessed by bioluminescent imaging and magnetic resonance imaging. Flow cytometry assessed direct effects of HBO on reactive oxygen species (ROS) signaling of transplanted glioma cells and organs, and quantified mature T cells and subgroups in tumors, the brain, and blood. Results HBO promoted the growth of transplanted GL261-Luc glioma in the intracranial glioma mouse model. ROS signaling of glioma cells and brain cells was significantly downregulated under HBO stimulation, but thymus ROS levels were significantly upregulated. CD3+ T cells were significantly downregulated, while both Ti/Th cells (CD3+CD4+) and Ts/Tc cells (CD3+CD8+) were inhibited in tumors of the HBO group. The percentage of regulatory T cells in Ti/Th (CD3+CD4+) cells was elevated in the tumors and thymuses of the HBO group. Conclusion HBO induced ROS signaling in the thymus, inhibited CD3+ T cell generation, and facilitated malignant glioma cell growth in vivo in the intracranial glioma mouse model.
Collapse
Affiliation(s)
- Yong-Gang Wang
- 1 Department of Neurosurgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Jiang Long
- 1 Department of Neurosurgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Dong-Chuan Shao
- 2 Department of Neurosurgery, First People's Hospital of Kunming, Kunming, Yunnan 650032, China
| | - Hai Song
- 1 Department of Neurosurgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| |
Collapse
|
86
|
Strutt TM, Dhume K, Finn CM, Hwang JH, Castonguay C, Swain SL, McKinstry KK. IL-15 supports the generation of protective lung-resident memory CD4 T cells. Mucosal Immunol 2018; 11:668-680. [PMID: 29186108 PMCID: PMC5975122 DOI: 10.1038/mi.2017.101] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 10/17/2017] [Indexed: 02/04/2023]
Abstract
Tissue-resident memory T cells (TRM) provide optimal defense at the sites of infection, but signals regulating their development are unclear, especially for CD4 T cells. Here we identify two distinct pathways that lead to the generation of CD4 TRM in the lungs following influenza infection. The TRM are transcriptionally distinct from conventional memory CD4 T cells and share a gene signature with CD8 TRM. The CD4 TRM are superior cytokine producers compared with conventional memory cells, can protect otherwise naive mice against a lethal influenza challenge, and display functional specialization by inducing enhanced inflammatory responses from dendritic cells compared with conventional memory cells. Finally, we demonstrate than an interleukin (IL)-2-dependent and a novel IL-2-independent but IL-15-dependent pathway support the generation of cohorts of lung TRM.
Collapse
Affiliation(s)
- Tara M. Strutt
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Kunal Dhume
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Caroline M. Finn
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Ji Hae Hwang
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Catherine Castonguay
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Susan L. Swain
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, USA
| | - K. Kai McKinstry
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
87
|
Jeyanathan M, Yao Y, Afkhami S, Smaill F, Xing Z. New Tuberculosis Vaccine Strategies: Taking Aim at Un-Natural Immunity. Trends Immunol 2018; 39:419-433. [DOI: 10.1016/j.it.2018.01.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 01/02/2018] [Accepted: 01/16/2018] [Indexed: 12/13/2022]
|
88
|
Nagy N, Kuipers HF, Marshall PL, Wang E, Kaber G, Bollyky PL. Hyaluronan in immune dysregulation and autoimmune diseases. Matrix Biol 2018; 78-79:292-313. [PMID: 29625181 DOI: 10.1016/j.matbio.2018.03.022] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/10/2018] [Accepted: 03/30/2018] [Indexed: 02/06/2023]
Abstract
The tissue microenvironment contributes to local immunity and to the pathogenesis of autoimmune diseases - a diverse set of conditions characterized by sterile inflammation, immunity against self-antigens, and destruction of tissues. However, the specific factors within the tissue microenvironment that contribute to local immune dysregulation in autoimmunity are poorly understood. One particular tissue component implicated in multiple autoimmune diseases is hyaluronan (HA), an extracellular matrix (ECM) polymer. HA is abundant in settings of chronic inflammation and contributes to lymphocyte activation, polarization, and migration. Here, we first describe what is known about the size, amount, and distribution of HA at sites of autoimmunity and in associated lymphoid structures in type 1 diabetes, multiple sclerosis, and rheumatoid arthritis. Next, we examine the recent literature on HA and its impact on adaptive immunity, particularly in regards to the biology of lymphocytes and Foxp3+ regulatory T-cells (Treg), a T-cell subset that maintains immune tolerance in healthy individuals. We propose that HA accumulation at sites of chronic inflammation creates a permissive environment for autoimmunity, characterized by CD44-mediated inhibition of Treg expansion. Finally, we address potential tools and strategies for targeting HA and its receptor CD44 in chronic inflammation and autoimmunity.
Collapse
Affiliation(s)
- Nadine Nagy
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Hedwich F Kuipers
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Payton L Marshall
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Esther Wang
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Gernot Kaber
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Paul L Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
89
|
Kimura MY, Hayashizaki K, Tokoyoda K, Takamura S, Motohashi S, Nakayama T. Crucial role for CD69 in allergic inflammatory responses: CD69-Myl9 system in the pathogenesis of airway inflammation. Immunol Rev 2018; 278:87-100. [PMID: 28658550 DOI: 10.1111/imr.12559] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
CD69 has been known as an early activation marker of lymphocytes; whereas, recent studies demonstrate that CD69 also has critical functions in immune responses. Early studies using human samples revealed the involvement of CD69 in various inflammatory diseases including asthma. Moreover, murine disease models using Cd69-/- mice and/or anti-CD69 antibody (Ab) treatment have revealed crucial roles for CD69 in inflammatory responses. However, it had not been clear how the CD69 molecule contributes to the pathogenesis of inflammatory diseases. We recently elucidated a novel mechanism, in which the interaction between CD69 and its ligands, myosin light chain 9, 12a and 12b (Myl9/12) play a critical role in the recruitment of activated T cells into the inflammatory lung. In this review, we first summarize CD69 function based on its structure and then introduce the evidence for the involvement of CD69 in human diseases and murine disease models. Then, we will describe how we discovered CD69 ligands, Myl9 and Myl12, and how the CD69-Myl9 system regulates airway inflammation. Finally, we will discuss possible therapeutic usages of the blocking Ab to the CD69-Myl9 system.
Collapse
Affiliation(s)
- Motoko Y Kimura
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Koji Hayashizaki
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Koji Tokoyoda
- Department of Osteoimmunology, German Rheumatism Research Centre (DRFZ) Berlin, Berlin, Germany
| | - Shiki Takamura
- Department of Immunology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Shinichiro Motohashi
- Department of Medical Immunology Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
90
|
Cellular and Humoral Immunity Protect against Vaginal Zika Virus Infection in Mice. J Virol 2018; 92:JVI.00038-18. [PMID: 29343577 PMCID: PMC5972878 DOI: 10.1128/jvi.00038-18] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 01/07/2023] Open
Abstract
Zika virus (ZIKV), which can cause devastating disease in fetuses of infected pregnant women, can be transmitted by mosquito inoculation and sexual routes. Little is known about immune protection against sexually transmitted ZIKV. In this study, we show that previous infection through intravaginal or subcutaneous routes with a contemporary Brazilian strain of ZIKV can protect against subsequent intravaginal challenge with a homologous strain. Both routes of inoculation induced high titers of ZIKV-specific and neutralizing antibody in serum and the vaginal lumen. Virus-specific T cells were recruited to and retained in the female reproductive tract after intravaginal and subcutaneous ZIKV infection. Studies in mice with genetic or acquired deficiencies in B and/or T cells demonstrated that both lymphocyte populations redundantly protect against intravaginal challenge in ZIKV-immune animals. Passive transfer of ZIKV-immune IgG or T cells significantly limited intravaginal infection of naive mice, although antibody more effectively prevented dissemination throughout the reproductive tract. Collectively, our experiments begin to establish the immune correlates of protection against intravaginal ZIKV infection, which should inform vaccination strategies in nonpregnant and pregnant women.IMPORTANCE The recent ZIKV epidemic resulted in devastating outcomes in fetuses and may affect reproductive health. Unlike other flaviviruses, ZIKV can be spread by sexual contact as well as a mosquito vector. While previous studies have identified correlates of protection for mosquito-mediated infection, few have focused on immunity against sexual transmission. As exposure to ZIKV via mosquito bite has likely occurred to many living in areas where ZIKV is endemic, our study addresses whether this route of infection can protect against subsequent sexual exposure. We demonstrate that subcutaneous ZIKV infection can protect against subsequent vaginal infection by generating both local antiviral T cell and antibody responses. Our research begins to define the immune correlates of protection for ZIKV infection in the vagina and provides a foundation for testing ZIKV vaccines against sexual transmission.
Collapse
|
91
|
Jandus C, Usatorre AM, Viganò S, Zhang L, Romero P. The Vast Universe of T Cell Diversity: Subsets of Memory Cells and Their Differentiation. Methods Mol Biol 2018; 1514:1-17. [PMID: 27787788 DOI: 10.1007/978-1-4939-6548-9_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The T cell receptor confers specificity for antigen recognition to T cells. By the first encounter with the cognate antigen, reactive T cells initiate a program of expansion and differentiation that will define not only the ultimate quantity of specific cells that will be generated, but more importantly their quality and functional heterogeneity. Recent achievements using mouse model infection systems have helped to shed light into the complex network of factors that dictate and sustain memory T cell differentiation, ranging from antigen load, TCR signal strength, metabolic fitness, transcriptional programs, and proliferative potential. The different models of memory T cell differentiation are discussed in this chapter, and key phenotypic and functional attributes of memory T cell subsets are presented, both for mouse and human cells. Therapeutic manipulation of memory T cell generation is expected to provide novel unique ways to optimize current immunotherapies, both in infection and cancer.
Collapse
Affiliation(s)
- Camilla Jandus
- Translational Tumor Immunology Group, Ludwig Cancer Research Center, University of Lausanne, Biopole III, CB02, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Amaia Martínez Usatorre
- Translational Tumor Immunology Group, Ludwig Cancer Research Center, University of Lausanne, Biopole III, CB02, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Selena Viganò
- Translational Tumor Immunology Group, Ludwig Cancer Research Center, University of Lausanne, Biopole III, CB02, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Lianjun Zhang
- Translational Tumor Immunology Group, Ludwig Cancer Research Center, University of Lausanne, Biopole III, CB02, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Pedro Romero
- Translational Tumor Immunology Group, Ludwig Cancer Research Center, University of Lausanne, Biopole III, CB02, Chemin des Boveresses 155, 1066, Epalinges, Switzerland.
| |
Collapse
|
92
|
Park YJ, Lee HK. The Role of Skin and Orogenital Microbiota in Protective Immunity and Chronic Immune-Mediated Inflammatory Disease. Front Immunol 2018; 8:1955. [PMID: 29375574 PMCID: PMC5767596 DOI: 10.3389/fimmu.2017.01955] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 12/19/2017] [Indexed: 12/11/2022] Open
Abstract
The skin and orogenital mucosae, which constitute complex protective barriers against infection and injuries, are not only the first to come into contact with pathogens but are also colonized by a set of microorganisms that are essential to maintain a healthy physiological environment. Using 16S ribosomal RNA metagenomic sequencing, scientists recognized that the microorganism colonization has greater diversity and variability than previously assumed. These microorganisms, such as commensal bacteria, affect the host’s immune response against pathogens and modulate chronic inflammatory responses. Previously, a single pathogen was thought to cause a single disease, but current evidence suggests that dysbiosis of the tissue microbiota may underlie the disease status. Dysbiosis results in aberrant immune responses at the surface and furthermore, affects the systemic immune response. Hence, understanding the initial interaction between the barrier surface immune system and local microorganisms is important for understanding the overall systemic effects of the immune response. In this review, we describe current evidence for the basis of the interactions between pathogens, microbiota, and immune cells on surface barriers and offer explanations for how these interactions may lead to chronic inflammatory disorders.
Collapse
Affiliation(s)
- Young Joon Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea.,KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| |
Collapse
|
93
|
Caldeira-Dantas S, Furmanak T, Smith C, Quinn M, Teos LY, Ertel A, Kurup D, Tandon M, Alevizos I, Snyder CM. The Chemokine Receptor CXCR3 Promotes CD8 + T Cell Accumulation in Uninfected Salivary Glands but Is Not Necessary after Murine Cytomegalovirus Infection. THE JOURNAL OF IMMUNOLOGY 2017; 200:1133-1145. [PMID: 29288198 DOI: 10.4049/jimmunol.1701272] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/17/2017] [Indexed: 01/24/2023]
Abstract
Recent work indicates that salivary glands are able to constitutively recruit CD8+ T cells and retain them as tissue-resident memory T cells, independently of local infection, inflammation, or Ag. To understand the mechanisms supporting T cell recruitment to the salivary gland, we compared T cell migration to the salivary gland in mice that were infected or not with murine CMV (MCMV), a herpesvirus that infects the salivary gland and promotes the accumulation of salivary gland tissue-resident memory T cells. We found that acute MCMV infection increased rapid T cell recruitment to the salivary gland but that equal numbers of activated CD8+ T cells eventually accumulated in infected and uninfected glands. T cell recruitment to uninfected salivary glands depended on chemokines and the integrin α4 Several chemokines were expressed in the salivary glands of infected and uninfected mice, and many of these could promote the migration of MCMV-specific T cells in vitro. MCMV infection increased the expression of chemokines that interact with the receptors CXCR3 and CCR5, but neither receptor was needed for T cell recruitment to the salivary gland during MCMV infection. Unexpectedly, however, the chemokine receptor CXCR3 was critical for T cell accumulation in uninfected salivary glands. Together, these data suggest that CXCR3 and the integrin α4 mediate T cell recruitment to uninfected salivary glands but that redundant mechanisms mediate T cell recruitment after MCMV infection.
Collapse
Affiliation(s)
- Sofia Caldeira-Dantas
- Department of Immunology and Microbiology, Thomas Jefferson University, Philadelphia, PA 19107.,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal.,Life and Health Sciences Research Institute (ICVS)/3B's Associate Laboratory, 4710-057 Braga, Portugal
| | - Thomas Furmanak
- Department of Immunology and Microbiology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Corinne Smith
- Department of Immunology and Microbiology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Michael Quinn
- Department of Immunology and Microbiology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Leyla Y Teos
- Sjögren's Syndrome and Salivary Gland Dysfunction Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892; and
| | - Adam Ertel
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Drishya Kurup
- Department of Immunology and Microbiology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Mayank Tandon
- Sjögren's Syndrome and Salivary Gland Dysfunction Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892; and
| | - Ilias Alevizos
- Sjögren's Syndrome and Salivary Gland Dysfunction Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892; and
| | - Christopher M Snyder
- Department of Immunology and Microbiology, Thomas Jefferson University, Philadelphia, PA 19107;
| |
Collapse
|
94
|
Danahy DB, Strother RK, Badovinac VP, Griffith TS. Clinical and Experimental Sepsis Impairs CD8 T-Cell-Mediated Immunity. Crit Rev Immunol 2017; 36:57-74. [PMID: 27480902 DOI: 10.1615/critrevimmunol.2016017098] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Septic patients experience chronic immunosuppression resulting in enhanced susceptibility to infections normally controlled by T cells. Clinical research on septic patients has shown increased apoptosis and reduced total numbers of CD4 and CD8 T cells, suggesting contributing mechanism driving immunosuppression. Experimental models of sepsis, including cecal ligation and puncture, reverse translated this clinical observation to facilitate hypothesis-driven research and allow the use of an array of experimental tools to probe the impact of sepsis on T-cell immunity. In addition to numerical loss, sepsis functionally impairs the antigen-driven proliferative capacity and effector functions of CD4 and CD8 T cells. Sepsis-induced impairments in both the quantity and quality of T cells results in reduced protective capacity and increased susceptibility of mice to new or previously encountered infections. Therefore, the combined efforts of clinical and experimental sepsis research have begun to elucidate the impact of sepsis on T-cell-mediated immunity and potential T-cell-intrinsic and -extrinsic mechanisms driving chronic immunosuppression. Future work will explore the impact of sepsis on the recently appreciated tissue-resident memory (TRM) T cells, which provide robust protection against localized infections, and dendritic cells, which are needed to activate T cells and promote effective T-cell responses.
Collapse
Affiliation(s)
- Derek B Danahy
- Department of Pathology, University of Iowa, Iowa City, IA; Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA
| | | | - Vladimir P Badovinac
- Department of Pathology, Interdisciplinary Program in Immunology, University of Iowa, Iowa City, Iowa
| | - Thomas S Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN; Microbiology, Immunology and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN; Center for Immunology, University of Minnesota, Minneapolis, MN; Minneapolis VA Health Care System, Minneapolis, Minnesota
| |
Collapse
|
95
|
Calenda G, Keawvichit R, Arrode-Brusés G, Pattanapanyasat K, Frank I, Byrareddy SN, Arthos J, Cicala C, Grasperge B, Blanchard JL, Gettie A, Reimann KA, Ansari AA, Martinelli E. Integrin α 4β 7 Blockade Preferentially Impacts CCR6 + Lymphocyte Subsets in Blood and Mucosal Tissues of Naive Rhesus Macaques. THE JOURNAL OF IMMUNOLOGY 2017; 200:810-820. [PMID: 29196458 DOI: 10.4049/jimmunol.1701150] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/31/2017] [Indexed: 01/27/2023]
Abstract
Infusion of a simianized anti-α4β7 mAb (Rh-α4β7) just before and following SIV infection protected rhesus macaques from developing AIDS and partially from vaginal SIV acquisition. Recently, short-term treatment with Rh-α4β7 in combination with cART was found to lead to prolonged viral suppression after withdrawal of all therapeutic interventions. The humanized form of Rh-α4β7, vedolizumab, is a highly effective treatment for inflammatory bowel disease. To clarify the mechanism of action of Rh-α4β7, naive macaques were infused with Rh-α4β7 and sampled in blood and tissues before and after treatment to monitor several immune cell subsets. In blood, Rh-α4β7 increased the CD4+ and CD8+ T cell counts, but not B cell counts, and preferentially increased CCR6+ subsets while decreasing CD103+ and CD69+ lymphocytes. In mucosal tissues, surprisingly, Rh-α4β7 did not impact integrin α4+ cells, but decreased the frequencies of CCR6+ and CD69+ CD4+ T cells and, in the gut, Rh-α4β7 transiently decreased the frequency of memory and IgA+ B cells. In summary, even in the absence of inflammation, Rh-α4β7 impacted selected immune cell subsets in different tissues. These data provide new insights into the mechanisms by which Rh-α4β7 may mediate its effect in SIV-infected macaques with implications for understanding the effect of treatment with vedolizumab in patients with inflammatory bowel disease.
Collapse
Affiliation(s)
- Giulia Calenda
- Center for Biomedical Research, Population Council, New York, NY 10065
| | - Rassamon Keawvichit
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322.,Department of Immunology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | | | - Kovit Pattanapanyasat
- Department of Immunology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Ines Frank
- Center for Biomedical Research, Population Council, New York, NY 10065
| | - Siddappa N Byrareddy
- Department of Pharmacology and Experimental Neurosciences, University of Nebraska Medical Center, Omaha, NE 68198
| | - James Arthos
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Claudia Cicala
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Brooke Grasperge
- Tulane National Primate Research Center, Tulane University, Covington, LA 70433
| | - James L Blanchard
- Tulane National Primate Research Center, Tulane University, Covington, LA 70433
| | - Agegnehu Gettie
- Aaron Diamond AIDS Research Center, Rockefeller University, New York, NY 10016; and
| | - Keith A Reimann
- MassBiologics, University of Massachusetts Medical School, Boston, MA 02126
| | - Aftab A Ansari
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322;
| | - Elena Martinelli
- Center for Biomedical Research, Population Council, New York, NY 10065;
| |
Collapse
|
96
|
Morrow MP, Kraynyak KA, Sylvester AJ, Dallas M, Knoblock D, Boyer JD, Yan J, Vang R, Khan AS, Humeau L, Sardesai NY, Kim JJ, Plotkin S, Weiner DB, Trimble CL, Bagarazzi ML. Clinical and Immunologic Biomarkers for Histologic Regression of High-Grade Cervical Dysplasia and Clearance of HPV16 and HPV18 after Immunotherapy. Clin Cancer Res 2017; 24:276-294. [PMID: 29084917 DOI: 10.1158/1078-0432.ccr-17-2335] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/04/2017] [Accepted: 10/24/2017] [Indexed: 02/06/2023]
Abstract
Purpose: As previously reported, treatment of high-grade cervical dysplasia with VGX-3100 resulted in complete histopathologic regression (CR) concomitant with elimination of HPV16/18 infection in 40.0% of VGX-3100-treated patients compared with only 14.3% in placebo recipients in a randomized phase IIb study. Here, we identify clinical and immunologic characteristics that either predicted or correlated with therapeutic benefit from VGX-3100 to identify parameters that might guide clinical decision-making for this disease.Experimental Design: We analyzed samples taken from cervical swabs, whole blood, and tissue biopsies/resections to determine correlates and predictors of treatment success.Results: At study entry, the presence of preexisting immunosuppressive factors such as FoxP3 and PD-L1 in cervical lesions showed no association with treatment outcome. The combination of HPV typing and cervical cytology following dosing was predictive for both histologic regression and elimination of detectable virus at the efficacy assessment 22 weeks later (negative predictive value 94%). Patients treated with VGX-3100 who had lesion regression had a statistically significant >2-fold increase in CD137+perforin+CD8+ T cells specific for the HPV genotype causing disease. Increases in cervical mucosal CD137+ and CD103+ infiltrates were observed only in treated patients. Perforin+ cell infiltrates were significantly increased >2-fold in cervical tissue only in treated patients who had histologic CR.Conclusions: Quantitative measures associated with an effector immune response to VGX-3100 antigens were associated with lesion regression. Consequently, these analyses indicate that certain immunologic responses associate with successful resolution of HPV-induced premalignancy, with particular emphasis on the upregulation of perforin in the immunotherapy-induced immune response. Clin Cancer Res; 24(2); 276-94. ©2017 AACR.
Collapse
Affiliation(s)
| | | | | | | | | | - Jean D Boyer
- Inovio Pharmaceuticals, Plymouth Meeting, Pennsylvania
| | - Jian Yan
- Inovio Pharmaceuticals, Plymouth Meeting, Pennsylvania
| | - Russell Vang
- Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Amir S Khan
- Inovio Pharmaceuticals, Plymouth Meeting, Pennsylvania
| | | | | | - J Joseph Kim
- Inovio Pharmaceuticals, Plymouth Meeting, Pennsylvania
| | - Stanley Plotkin
- The University of Pennsylvania, Philadelphia, Pennsylvania.,Vaxconsult, Doylestown, Pennsylvania
| | | | | | | |
Collapse
|
97
|
Mitson-Salazar A, Prussin C. Pathogenic Effector Th2 Cells in Allergic Eosinophilic Inflammatory Disease. Front Med (Lausanne) 2017; 4:165. [PMID: 29057225 PMCID: PMC5635264 DOI: 10.3389/fmed.2017.00165] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 09/20/2017] [Indexed: 12/24/2022] Open
Abstract
There is an absolute requirement for Th2 cells in the pathogenesis of allergen-driven eosinophil-rich type 2 inflammation. Although Th2 cells are generally regarded as a homogeneous population, in the past decade there has been increasing evidence for a minority subpopulation of IL-5+ Th2 cells that have enhanced effector function. This IL-5+ Th2 subpopulation has been termed pathogenic effector Th2 (peTh2), as it exhibits greater effector function and disease association than conventional Th2 cells. peTh2 cells have a different expression profile, differentially express transcription factors, and preferentially use specific signaling pathways. As such, peTh2 cells are a potential target in the treatment of allergic eosinophilic inflammation. This review examines peTh2 cells, both in mouse models and human disease, with an emphasis on their role in the pathogenesis of allergic eosinophilic inflammation.
Collapse
Affiliation(s)
- Alyssa Mitson-Salazar
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States
| | | |
Collapse
|
98
|
Condotta SA, Richer MJ. The immune battlefield: The impact of inflammatory cytokines on CD8+ T-cell immunity. PLoS Pathog 2017; 13:e1006618. [PMID: 29073270 PMCID: PMC5658174 DOI: 10.1371/journal.ppat.1006618] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Stephanie A. Condotta
- Department of Microbiology and Immunology, Microbiome and Disease Tolerance Centre, Rosalind and Morris Goodman Cancer Research Centre McGill University, Montreal, Quebec, Canada
| | - Martin J. Richer
- Department of Microbiology and Immunology, Microbiome and Disease Tolerance Centre, Rosalind and Morris Goodman Cancer Research Centre McGill University, Montreal, Quebec, Canada
| |
Collapse
|
99
|
Hoytema van Konijnenburg DP, Reis BS, Pedicord VA, Farache J, Victora GD, Mucida D. Intestinal Epithelial and Intraepithelial T Cell Crosstalk Mediates a Dynamic Response to Infection. Cell 2017; 171:783-794.e13. [PMID: 28942917 DOI: 10.1016/j.cell.2017.08.046] [Citation(s) in RCA: 196] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 08/02/2017] [Accepted: 08/25/2017] [Indexed: 02/07/2023]
Abstract
Intestinal intraepithelial lymphocytes (IELs) are located at the critical interface between the intestinal lumen, which is chronically exposed to food and microbes, and the core of the body. Using high-resolution microscopy techniques and intersectional genetic tools, we investigated the nature of IEL responses to luminal microbes. We observed that TCRγδ IELs exhibit unique microbiota-dependent location and movement patterns in the epithelial compartment. This behavioral pattern quickly changes upon exposure to different enteric pathogens, resulting in increased interepithelial cell (EC) scanning, expression of antimicrobial genes, and glycolysis. Both dynamic and metabolic changes to γδ IEL depend on pathogen sensing by ECs. Direct modulation of glycolysis is sufficient to change γδ IEL behavior and susceptibility to early pathogen invasion. Our results uncover a coordinated EC-IEL response to enteric infections that modulates lymphocyte energy utilization and dynamics and supports maintenance of the intestinal epithelial barrier. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- David P Hoytema van Konijnenburg
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA; Laboratory of Translational Immunology, University Medical Center Utrecht, 3584 CX, Utrecht, the Netherlands
| | - Bernardo S Reis
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA.
| | - Virginia A Pedicord
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Julia Farache
- Departments of Orofacial Sciences and Pediatrics, Institute of Human Genetics and Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Gabriel D Victora
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY 10065, USA
| | - Daniel Mucida
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
100
|
Abstract
Falling between the classical characteristics of innate immune cells and adaptive T and B cells are a group of lymphocytes termed "unconventional." These cells express antigen-specific T or B cell receptors, but behave with innate characteristics. Well-known members of this group include the gamma-delta T cell and the Natural Killer T cell. Recent literature has greatly expanded scientific knowledge of unconventional lymphocytes, but key questions remain unresolved in the field, including why these cells have been maintained concurrently with conventional innate and adaptive immune cells. Here, we summarize current literature that suggests what their unique purposes may be, including specialized functions with the microbiota and in early development. From the consensus literature, we discuss where we see unconventional lymphocytes fit into the logical organization of the complete immune system.
Collapse
Affiliation(s)
- Lesley Pasman
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
| | - Dennis L Kasper
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|