1
|
Liu JY, Qiao YL, Jiao WE, Tao ZZ, Xu S, Chen SM. Changes in Circulating CD44+CD62L- Treg Subsets and CD44-CD62L+ Treg Subsets Reflect the Clinical Status of Patients with Allergic Rhinitis. Int Arch Allergy Immunol 2024:1-13. [PMID: 39226877 DOI: 10.1159/000540536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 07/18/2024] [Indexed: 09/05/2024] Open
Abstract
INTRODUCTION This study clarified the expression changes and clinical significance of CD44+CD62L- Treg and CD44-CD62L+ Treg subsets in the peripheral blood of patients with allergic rhinitis (AR). METHODS The peripheral blood of 39 patients with AR and 42 healthy controls was collected. Clinical data, such as sex, age, IgE titer, allergen screening information and visual analogue scale (VAS) score, were recorded. Changes in serum IL-2, IL-4, IL-6, IL-10, TNF-α, and IFN-γ were detected using the cytometric bead array method. Flow cytometry was used to detect the proportions of Th1, Th2, Th17, TFH, and Th9 cells and the proportions of CD44+CD62L- Treg and CD44-CD62L+ Treg subsets. Correlation analysis was performed between the CD44+CD62L- Treg subsets and the CD44-CD62L+ Treg subsets with clinical indicators (VAS score, total IgE titer), cytokines (IL-2, IL-4, IL-6, IL-10, TNF-α, IFN-γ), and Th1/Th2/Th17/TFH/Th9 cell proportions. RESULTS Compared to the control group, the proportion of total Treg cells and CD44+CD62L- Treg cells in the AR group decreased, and the proportion of CD44-CD62L+ Treg cells increased (p < 0.05). The proportions of CD44+CD62L- Treg cells significantly negatively correlated with Th2 cells (R = -0.5270, p < 0.05) and positively correlated with Treg cytokine IL-10 (R = 0.6447, p < 0.05). In addition, CD44+CD62L- Treg cells negatively correlated with the VAS score (R = -0.4956, p < 0.05), total IgE level (R = -0.4177, p < 0.05) and Th2 cytokine IL-6 level (R = -0.3034, p < 0.05) but positively correlated with the Th1 cytokine IL-2 (R = 0.4331, p < 0.05). In contrast, the proportion of CD44+CD62L- Treg cells significantly positively correlated with the Th2 cells (R = 0.6187, p < 0.05). Moreover, the proportion of CD44-CD62L+ Treg cells positively correlated with the VAS score (R = 0.4060, p < 0.05), total IgE level (R = 0.5224, p < 0.05) and Th2 cytokine IL-4 (R = 0.2647, p < 0.05) and IL-6 levels (R = 0.3824, p < 0.05) but negatively correlated with Th1 cytokine IL-2 (R = -0.3451, p < 0.05) and IL-10 (R = -0.3277, p < 0.05). CONCLUSION A greater proportion of CD44+CD62L- Tregs correlated with better reversal of the Th1/Th2 imbalance and milder clinical symptoms in AR patients. The presence of more CD44-CD62L+ Tregs correlated with a weaker immunosuppressive effect on Th2 cells and more severe clinical symptoms in AR patients. These findings provide new perspectives for the treatment and disease monitoring of AR.
Collapse
Affiliation(s)
- Jia-Yu Liu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yue-Long Qiao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Wo-Er Jiao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ze-Zhang Tao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shan Xu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shi-Ming Chen
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
2
|
Ohishi K, Ishikura A, Nishida S, Abo H, Nakatsukasa H, Kawashima H. Sialyl Lewis X Defines an Activated and Functional Regulatory T Cell Subpopulation in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1627-1638. [PMID: 38639586 DOI: 10.4049/jimmunol.2300349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 03/26/2024] [Indexed: 04/20/2024]
Abstract
Attempts have been made to elucidate the functional markers of regulatory T cells (Tregs), CD4+Foxp3+ T cells with an immunosuppressive function. Sialyl Lewis X (sLex), a tetrasaccharide Ag, is involved in leukocyte trafficking as selectin ligands and is a marker of highly differentiated Tregs in humans. However, the importance of sLex in murine Tregs remains unknown. In this study, we report that sLex defines the activated and functional subset of murine Tregs. The contact hypersensitivity model showed that murine Tregs strongly express sLex upon activation, accompanied by functional Treg marker elevation, such as Foxp3, CD25, CD103, CD39, and granzyme B. RNA sequencing analysis revealed sLex-positive (sLex+) Tregs expressed genes involved in Treg function at a higher level than sLex-negative (sLex-) Tregs. Using an in vitro suppression assay, we found that sLex+ Tregs could more efficiently suppress naive CD4+ T cell proliferation than sLex- Tregs. In the murine contact hypersensitivity elicitation model, the topical sLex+ Treg injection into the ears suppressed ear inflammation more efficiently than that of sLex- Tregs. Our results indicate that sLex could serve as a unique surface marker of activated and functional Tregs with immunosuppressive functions in mice.
Collapse
Affiliation(s)
- Kanae Ohishi
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Asaki Ishikura
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Shogo Nishida
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Hirohito Abo
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Hiroko Nakatsukasa
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Hiroto Kawashima
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| |
Collapse
|
3
|
Hao Q, Kundu S, Shetty S, Tang H. Runx3 Regulates CD8 + T Cell Local Expansion and CD43 Glycosylation in Mice by H1N1 Influenza A Virus Infection. Int J Mol Sci 2024; 25:4220. [PMID: 38673806 PMCID: PMC11050410 DOI: 10.3390/ijms25084220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
We have recently reported that transcription factor Runx3 is required for pulmonary generation of CD8+ cytotoxic T lymphocytes (CTLs) that play a crucial role in the clearance of influenza A virus (IAV). To understand the underlying mechanisms, we determined the effects of Runx3 knockout (KO) on CD8+ T cell local expansion and phenotypes using an inducible general Runx3 KO mouse model. We found that in contrast to the lungs, Runx3 general KO promoted enlargement of lung-draining mediastinal lymph node (mLN) and enhanced CD8+ and CD4+ T cell expansion during H1N1 IAV infection. We further found that Runx3 deficiency greatly inhibited core 2 O-glycosylation of selectin ligand CD43 on activated CD8+ T cells but minimally affected the cell surface expression of CD43, activation markers (CD44 and CD69) and cell adhesion molecules (CD11a and CD54). Runx3 KO had a minor effect on lung effector CD8+ T cell death by IAV infection. Our findings indicate that Runx3 differently regulates CD8+ T cell expansion in mLNs and lungs by H1N1 IAV infection. Runx3 is required for CD43 core 2 O-glycosylation on activated CD8+ T cells, and the involved Runx3 signal pathway may mediate CD8+ T cell phenotype for pulmonary generation of CTLs.
Collapse
Affiliation(s)
| | | | | | - Hua Tang
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA; (Q.H.); (S.K.); (S.S.)
| |
Collapse
|
4
|
Kolahi Azar H, Imanpour A, Rezaee H, Ezzatifar F, Zarei-Behjani Z, Rostami M, Azami M, Behestizadeh N, Rezaei N. Mesenchymal stromal cells and CAR-T cells in regenerative medicine: The homing procedure and their effective parameters. Eur J Haematol 2024; 112:153-173. [PMID: 37254607 DOI: 10.1111/ejh.14014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/24/2023] [Accepted: 04/24/2023] [Indexed: 06/01/2023]
Abstract
Mesenchymal stromal cells (MSCs) and chimeric antigen receptor (CAR)-T cells are two core elements in cell therapy procedures. MSCs have significant immunomodulatory effects that alleviate inflammation in the tissue regeneration process, while administration of specific chemokines and adhesive molecules would primarily facilitate CAR-T cell trafficking into solid tumors. Multiple parameters affect cell homing, including the recipient's age, the number of cell passages, proper cell culture, and the delivery method. In addition, several chemokines are involved in the tumor microenvironment, affecting the homing procedure. This review discusses parameters that improve the efficiency of cell homing and significant cell therapy challenges. Emerging comprehensive mechanistic strategies such as non-systemic and systemic homing that revealed a significant role in cell therapy remodeling were also reviewed. Finally, the primary implications for the development of combination therapies that incorporate both MSCs and CAR-T cells for cancer treatment were discussed.
Collapse
Affiliation(s)
- Hanieh Kolahi Azar
- Department of Pathology, Tabriz University of Medical Sciences, Tabriz, Iran
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Aylar Imanpour
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hanieh Rezaee
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ezzatifar
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Molecular and Cell Biology Research Center, Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zeinab Zarei-Behjani
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, Advanced School of Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammadreza Rostami
- Division of Food Safety and Hygiene, Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Food Science and Nutrition Group (FSAN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahmoud Azami
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Behestizadeh
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
5
|
Gordon H, Wichmann K, Lewis A, Sanders T, Wildemann M, Hoti I, Hornsby E, Kok KB, Silver A, Lindsay JO, Stagg AJ. Human Intestinal Dendritic Cells Can Overcome Retinoic Acid Signaling to Generate Proinflammatory CD4 T Cells with Both Gut and Skin Homing Properties. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:96-106. [PMID: 37955427 DOI: 10.4049/jimmunol.2300340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 09/18/2023] [Indexed: 11/14/2023]
Abstract
Retinoic acid, produced by intestinal dendritic cells (DCs), promotes T cell trafficking to the intestinal mucosa by upregulating α4β7 integrin and inhibiting the generation of cutaneous leukocyte Ag (CLA) required for skin entry. In the present study, we report that activation of human naive CD4 T cells in an APC-free system generates cells expressing α4β7 alone; in contrast, activation by intestinal DCs that produce retinoic acid and induce high levels of α4β7 also results in CLA expression, generating CLA+α4β7+ "dual tropic" cells, with both gut and skin trafficking potential, that also express high levels of α4β1 integrin. DC generation of CLA+α4β7+ T cells is associated with upregulation of FUT7, a fucosyltransferase involved in CLA generation; requires cell contact; and is enhanced by IL-12/IL-23. The blood CD4+ T cell population contains CLA+α4β7+ cells, which are significantly enriched for cells capable of IFN-γ, IL-17, and TNF-α production compared with conventional CLA-α4β7+ cells. Dual tropic lymphocytes are increased in intestinal tissue from patients with Crohn's disease, and single-cell RNA-sequencing analysis identifies a transcriptionally distinct cluster of FUT7-expressing cells present only in inflamed tissue; expression of genes associated with cell proliferation suggests that these cells are undergoing local activation. The expression of multiple trafficking molecules by CLA+α4β7+ T cells can enable their recruitment by alternative pathways to both skin and gut; they may contribute to both intestinal and cutaneous manifestations of inflammatory bowel disease.
Collapse
Affiliation(s)
- Hannah Gordon
- Centre for Immunobiology, Blizard Institute, Faculty of Medicine and dentistry, Barts and The London Medical School, Queen Mary University of London, London, United Kingdom
| | - Katherine Wichmann
- Centre for Immunobiology, Blizard Institute, Faculty of Medicine and dentistry, Barts and The London Medical School, Queen Mary University of London, London, United Kingdom
| | - Amy Lewis
- Centre for Genomics and Child Health, Blizard Institute, Faculty of Medicine and dentistry, Barts and The London Medical School, Queen Mary University of London, London, United Kingdom
| | - Theodore Sanders
- Centre for Immunobiology, Blizard Institute, Faculty of Medicine and dentistry, Barts and The London Medical School, Queen Mary University of London, London, United Kingdom
| | - Martha Wildemann
- Centre for Immunobiology, Blizard Institute, Faculty of Medicine and dentistry, Barts and The London Medical School, Queen Mary University of London, London, United Kingdom
| | - Inva Hoti
- Centre for Immunobiology, Blizard Institute, Faculty of Medicine and dentistry, Barts and The London Medical School, Queen Mary University of London, London, United Kingdom
| | - Eve Hornsby
- Centre for Immunobiology, Blizard Institute, Faculty of Medicine and dentistry, Barts and The London Medical School, Queen Mary University of London, London, United Kingdom
| | - K Bel Kok
- Department of Gastroenterology, Barts Health NHS Trust, London, United Kingdom
| | - Andrew Silver
- Centre for Genomics and Child Health, Blizard Institute, Faculty of Medicine and dentistry, Barts and The London Medical School, Queen Mary University of London, London, United Kingdom
| | - James O Lindsay
- Centre for Immunobiology, Blizard Institute, Faculty of Medicine and dentistry, Barts and The London Medical School, Queen Mary University of London, London, United Kingdom
- Department of Gastroenterology, Barts Health NHS Trust, London, United Kingdom
| | - Andrew J Stagg
- Centre for Immunobiology, Blizard Institute, Faculty of Medicine and dentistry, Barts and The London Medical School, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
6
|
Abdelbary M, Nolz JC. N-linked glycans: an underappreciated key determinant of T cell development, activation, and function. IMMUNOMETABOLISM (COBHAM, SURREY) 2023; 5:e00035. [PMID: 38027254 PMCID: PMC10662610 DOI: 10.1097/in9.0000000000000035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023]
Abstract
N-linked glycosylation is a post-translational modification that results in the decoration of newly synthesized proteins with diverse types of oligosaccharides that originate from the amide group of the amino acid asparagine. The sequential and collective action of multiple glycosidases and glycosyltransferases are responsible for determining the overall size, composition, and location of N-linked glycans that become covalently linked to an asparagine during and after protein translation. A growing body of evidence supports the critical role of N-linked glycan synthesis in regulating many features of T cell biology, including thymocyte development and tolerance, as well as T cell activation and differentiation. Here, we provide an overview of how specific glycosidases and glycosyltransferases contribute to the generation of different types of N-linked glycans and how these post-translational modifications ultimately regulate multiple facets of T cell biology.
Collapse
Affiliation(s)
- Mahmoud Abdelbary
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey C. Nolz
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
7
|
Hong Y, Walling BL, Kim HR, Serratelli WS, Lozada JR, Sailer CJ, Amitrano AM, Lim K, Mongre RK, Kim KD, Capece T, Lomakina EB, Reilly NS, Vo K, Gerber SA, Fan TC, Yu ALT, Oakes PW, Waugh RE, Jun CD, Reagan PM, Kim M. ST3GAL1 and βII-spectrin pathways control CAR T cell migration to target tumors. Nat Immunol 2023; 24:1007-1019. [PMID: 37069398 PMCID: PMC10515092 DOI: 10.1038/s41590-023-01498-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 03/21/2023] [Indexed: 04/19/2023]
Abstract
Adoptive transfer of genetically engineered chimeric antigen receptor (CAR) T cells is becoming a promising treatment option for hematological malignancies. However, T cell immunotherapies have mostly failed in individuals with solid tumors. Here, with a CRISPR-Cas9 pooled library, we performed an in vivo targeted loss-of-function screen and identified ST3 β-galactoside α-2,3-sialyltransferase 1 (ST3GAL1) as a negative regulator of the cancer-specific migration of CAR T cells. Analysis of glycosylated proteins revealed that CD18 is a major effector of ST3GAL1 in activated CD8+ T cells. ST3GAL1-mediated glycosylation induces the spontaneous nonspecific tissue sequestration of T cells by altering lymphocyte function-associated antigen-1 (LFA-1) endocytic recycling. Engineered CAR T cells with enhanced expression of βII-spectrin, a central LFA-1-associated cytoskeleton molecule, reversed ST3GAL1-mediated nonspecific T cell migration and reduced tumor growth in mice by improving tumor-specific homing of CAR T cells. These findings identify the ST3GAL1-βII-spectrin axis as a major cell-intrinsic program for cancer-targeting CAR T cell migration and as a promising strategy for effective T cell immunotherapy.
Collapse
Affiliation(s)
- Yeonsun Hong
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - Brandon L Walling
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - Hye-Ran Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - William S Serratelli
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - John R Lozada
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - Cooper J Sailer
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA
| | - Andrea M Amitrano
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA
| | - Kihong Lim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - Raj Kumar Mongre
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - Kyun-Do Kim
- Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, Korea
| | - Tara Capece
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - Elena B Lomakina
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
| | - Nicholas S Reilly
- Department of Physics and Astronomy, University of Rochester, Rochester, NY, USA
| | - Kevin Vo
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - Scott A Gerber
- Department of Surgery, University of Rochester, Rochester, NY, USA
| | - Tan-Chi Fan
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou and Chang Gung University, Taoyuan, Taiwan
| | - Alice Lin-Tsing Yu
- Department of Pediatrics/Hematology Oncology, University of California in San Diego, San Diego, CA, USA
| | - Patrick W Oakes
- Department of Physics and Astronomy, University of Rochester, Rochester, NY, USA
| | - Richard E Waugh
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
| | - Chang-Duk Jun
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Patrick M Reagan
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
8
|
Harbour JC, Abdelbary M, Schell JB, Fancher SP, McLean JJ, Nappi TJ, Liu S, Nice TJ, Xia Z, Früh K, Nolz JC. T helper 1 effector memory CD4 + T cells protect the skin from poxvirus infection. Cell Rep 2023; 42:112407. [PMID: 37083328 PMCID: PMC10281076 DOI: 10.1016/j.celrep.2023.112407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/15/2023] [Accepted: 04/04/2023] [Indexed: 04/22/2023] Open
Abstract
Poxvirus infections of the skin are a recent emerging public health concern, yet the mechanisms that mediate protective immunity against these viral infections remain largely unknown. Here, we show that T helper 1 (Th1) memory CD4+ T cells are necessary and sufficient to provide complete and broad protection against poxvirus skin infections, whereas memory CD8+ T cells are dispensable. Core 2 O-glycan-synthesizing Th1 effector memory CD4+ T cells rapidly infiltrate the poxvirus-infected skin microenvironment and produce interferon γ (IFNγ) in an antigen-dependent manner, causing global changes in gene expression to promote anti-viral immunity. Keratinocytes express IFN-stimulated genes, upregulate both major histocompatibility complex (MHC) class I and MHC class II antigen presentation in an IFNγ-dependent manner, and require IFNγ receptor (IFNγR) signaling and MHC class II expression for memory CD4+ T cells to protect the skin from poxvirus infection. Thus, Th1 effector memory CD4+ T cells exhibit potent anti-viral activity within the skin, and keratinocytes are the key targets of IFNγ necessary for preventing poxvirus infection of the epidermis.
Collapse
Affiliation(s)
- Jake C Harbour
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Mahmoud Abdelbary
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - John B Schell
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Samantha P Fancher
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Jack J McLean
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Taylen J Nappi
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Susan Liu
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Timothy J Nice
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Zheng Xia
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Klaus Früh
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Jeffrey C Nolz
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA; Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA; Department of Dermatology, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
9
|
Fernandes Â, Azevedo CM, Silva MC, Faria G, Dantas CS, Vicente MM, Pinho SS. Glycans as shapers of tumour microenvironment: A sweet driver of T-cell-mediated anti-tumour immune response. Immunology 2023; 168:217-232. [PMID: 35574724 DOI: 10.1111/imm.13494] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/22/2022] [Indexed: 01/17/2023] Open
Abstract
Essentially all cells are covered with a dense coat of different glycan structures/sugar chains, giving rise to the so-called glycocalyx. Changes in cellular glycosylation are a hallmark of cancer, affecting most of the pathophysiological processes associated with malignant transformation, including tumour immune responses. Glycans are chief macromolecules that define T-cell development, differentiation, fate, activation and signalling. Thus, the diversity of glycans expressed at the surface of T cells constitutes a fundamental molecular interface with the microenvironment by regulating the bilateral interactions between T-cells and cancer cells, fine-tuning the anti-tumour immune response. In this review, we will introduce the power of glycans as orchestrators of T-cell-mediated immune response in physiological conditions and in cancer. We discuss how glycans modulate the glyco-metabolic landscape in the tumour microenvironment, and whether glycans can synergize with immunotherapy as a way of rewiring T-cell effector functions against cancer cells.
Collapse
Affiliation(s)
- Ângela Fernandes
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal
| | - Catarina M Azevedo
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,School of Medicine and Biological Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Mariana C Silva
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,School of Medicine and Biological Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Guilherme Faria
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Carolina S Dantas
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,School of Medicine and Biological Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Manuel M Vicente
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,School of Medicine and Biological Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Salomé S Pinho
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,School of Medicine and Biological Sciences (ICBAS), University of Porto, Porto, Portugal.,Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
10
|
Delafield DG, Miles HN, Liu Y, Ricke WA, Li L. Complementary proteome and glycoproteome access revealed through comparative analysis of reversed phase and porous graphitic carbon chromatography. Anal Bioanal Chem 2022; 414:5461-5472. [PMID: 35137243 PMCID: PMC9246830 DOI: 10.1007/s00216-022-03934-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/04/2022] [Accepted: 01/27/2022] [Indexed: 11/01/2022]
Abstract
Continual developments in instrumental and analytical techniques have aided in establishing rigorous connections between protein glycosylation and human illness. These illnesses, such as various forms of cancer, are often associated with poor prognoses, prompting the need for more comprehensive characterization of the glycoproteome. While innovative instrumental and computational strategies have largely benefited glycoproteomic analyses, less attention is given to benefits gained through alternative, optimized chromatographic techniques. Porous graphitic carbon (PGC) chromatography has gained considerable interest in glycomics research due to its mobile phase flexibility, increased retention of polar analytes, and improved structural elucidation at higher temperatures. PGC has yet to be systematically compared against or in tandem with standard reversed phase liquid chromatography (RPLC) in high-throughput bottom-up glycoproteomic experiments, leaving the potential benefits unexplored. Performing comparative analysis of single and biphasic separation regimes at a range of column temperatures illustrates complementary advantages for each method. PGC separation is shown to selectively retain shorter, more hydrophilic glycopeptide species, imparting higher average charge, and exhibiting greater microheterogeneity coverage for identified glycosites. Additionally, we demonstrate that liquid-phase separation of glycopeptide isomers may be achieved through both single and biphasic PGC separations, providing a means towards facile, multidimensional glycopeptide characterization. Beyond this, we demonstrate how utilization of multiple separation regimes and column temperatures can aid in profiling the glycoproteome in tumorigenic and aggressive prostate cancer cells. RAW MS proteomic and glycoproteomic datasets have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the dataset identifier PXD024196 (10.6019/PXD024196) and PXD024195, respectively.
Collapse
Affiliation(s)
| | - Hannah N. Miles
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705
| | - Yuan Liu
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705
| | - William A. Ricke
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705,George M. O’Brien Urology Research Center of Excellence, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705,Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA. .,School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, 53705-2222, USA.
| |
Collapse
|
11
|
Burnie J, Persaud AT, Thaya L, Liu Q, Miao H, Grabinsky S, Norouzi V, Lusso P, Tang VA, Guzzo C. P-selectin glycoprotein ligand-1 (PSGL-1/CD162) is incorporated into clinical HIV-1 isolates and can mediate virus capture and subsequent transfer to permissive cells. Retrovirology 2022; 19:9. [PMID: 35597982 PMCID: PMC9123692 DOI: 10.1186/s12977-022-00593-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/21/2022] [Indexed: 11/16/2022] Open
Abstract
Background P-selectin glycoprotein ligand-1 (PSGL-1/CD162) has been studied extensively for its role in mediating leukocyte rolling through interactions with its cognate receptor, P-selectin. Recently, PSGL-1 was identified as a novel HIV-1 host restriction factor, particularly when expressed at high levels in the HIV envelope. Importantly, while the potent antiviral activity of PSGL-1 has been clearly demonstrated in various complementary model systems, the breadth of PSGL-1 incorporation across genetically diverse viral isolates and clinical isolates has yet to be described. Additionally, the biological activity of virion-incorporated PSGL-1 has also yet to be shown. Results Herein we assessed the levels of PSGL-1 on viruses produced through transfection with various amounts of PSGL-1 plasmid DNA (0–250 ng), compared to levels of PSGL-1 on viruses produced through infection of T cell lines and primary PBMC. We found that very low levels of PSGL-1 plasmid DNA (< 2.5 ng/well) were necessary to generate virus models that could closely mirror the phenotype of viruses produced via infection of T cells and PBMC. Unique to this study, we show that PSGL-1 is incorporated in a broad range of HIV-1 and SIV isolates and that virions with incorporated PSGL-1 are detectable in plasma from viremic HIV-1-infected individuals, corroborating the relevance of PSGL-1 in natural infection. Additionally, we show that PSGL-1 on viruses can bind its cognate selectin receptors, P-, E-, and L-selectins. Finally, we show viruses with endogenous levels of PSGL-1 can be captured by P-selectin and transferred to HIV-permissive bystander cells, highlighting a novel role for PSGL-1 in HIV-1 infection. Notably, viruses which contained high levels of PSGL-1 were noninfectious in our hands, in line with previous findings reporting the potent antiviral activity of PSGL-1. Conclusions Our results indicate that levels of PSGL-1 incorporation into virions can vary widely among model systems tested, and that careful tailoring of plasmid levels is required to recapitulate physiological systems when using pseudovirus models. Taken together, our data suggest that PSGL-1 may play diverse roles in the physiology of HIV-1 infection, particularly due to the functionally active state of PSGL-1 on virion surfaces and the breadth of PSGL-1 incorporation among a wide range of viral isolates. Supplementary Information The online version contains supplementary material available at 10.1186/s12977-022-00593-5.
Collapse
Affiliation(s)
- Jonathan Burnie
- Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON, Canada.,Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON, Canada
| | - Arvin Tejnarine Persaud
- Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON, Canada.,Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON, Canada
| | - Laxshaginee Thaya
- Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON, Canada.,Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON, Canada
| | - Qingbo Liu
- Viral Pathogenesis Section, Laboratory of Immunoregulation (LIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Huiyi Miao
- Viral Pathogenesis Section, Laboratory of Immunoregulation (LIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Stephen Grabinsky
- Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON, Canada
| | - Vanessa Norouzi
- Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON, Canada
| | - Paolo Lusso
- Viral Pathogenesis Section, Laboratory of Immunoregulation (LIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Vera A Tang
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, Flow Cytometry and Virometry Core Facility, University of Ottawa, Ottawa, ON, Canada
| | - Christina Guzzo
- Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON, Canada. .,Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON, Canada.
| |
Collapse
|
12
|
McCall JL, Varney ME, Rice E, Dziadowicz SA, Hall C, Blethen KE, Hu G, Barnett JB, Martinez I. Prenatal Cadmium Exposure Alters Proliferation in Mouse CD4 + T Cells via LncRNA Snhg7. Front Immunol 2022; 12:720635. [PMID: 35087510 PMCID: PMC8786704 DOI: 10.3389/fimmu.2021.720635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 12/14/2021] [Indexed: 11/13/2022] Open
Abstract
Objective Prenatal cadmium (Cd) exposure leads to immunotoxic phenotypes in the offspring affecting coding and non-coding genes. Recent studies have shown that long non-coding RNAs (lncRNAs) are integral to T cell regulation. Here, we investigated the role of long non-coding RNA small nucleolar RNA host gene 7 (lncSnhg7) in T cell proliferation. Methods RNA sequencing was used to analyze the expression of lncRNAs in splenic CD4+ T cells with and without CD3/CD28 stimulation. Next, T cells isolated from offspring exposed to control or Cd water throughout mating and gestation were analyzed with and without stimulation with anti-CD3/CD28 beads. Quantitative qPCR and western blotting were used to detect RNA and protein levels of specific genes. Overexpression of a miR-34a mimic was achieved using nucleofection. Apoptosis was measured using flow cytometry and luminescence assays. Flow cytometry was also used to measure T cell proliferation in culture. Finally, lncSnhg7 was knocked down in splenic CD4+ T cells with lentivirus to assess its effect on proliferation. Results We identified 23 lncRNAs that were differentially expressed in stimulated versus unstimulated T cells, including lncSnhg7. LncSnhg7 and a downstream protein, GALNT7, are upregulated in T cells from offspring exposed to Cd during gestation. Overexpression of miR-34a, a regulator of lncSnhg7 and GALNT7, suppresses GALNT7 protein levels in primary T cells, but not in a mouse T lymphocyte cell line. The T cells isolated from Cd-exposed offspring exhibit increased proliferation after activation in vitro, but Treg suppression and CD4+ T cell apoptosis are not affected by prenatal Cd exposure. Knockdown on lncSnhg7 inhibits proliferation of CD4+ T cells. Conclusion Prenatal Cd exposure alters the expression of lncRNAs during T cell activation. The induction of lncSnhg7 is enhanced in splenic T cells from Cd offspring resulting in the upregulation of GALNT7 protein and increased proliferation following activation. miR-34a overexpression decreased GALNT7 expression and knockdown of lncSnhg7 inhibited proliferation suggesting that the lncSnhg7/miR-34a/GALNT7 is an important pathway in primary CD4+ T cells. These data highlight the need to understand the consequences of environmental exposures on lncRNA functions in non-cancerous cells as well as the effects in utero.
Collapse
Affiliation(s)
- Jamie L. McCall
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Melinda E. Varney
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Emily Rice
- West Virginia University Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV, United States
| | - Sebastian A. Dziadowicz
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Casey Hall
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Kathryn E. Blethen
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Gangqing Hu
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
- West Virginia University Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV, United States
- Bioinformatics Core, West Virginia University, Morgantown, WV, United States
| | - John B. Barnett
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
- West Virginia University Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV, United States
| | - Ivan Martinez
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States
- West Virginia University Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
13
|
Park SL, Mackay LK. Decoding Tissue-Residency: Programming and Potential of Frontline Memory T Cells. Cold Spring Harb Perspect Biol 2021; 13:a037960. [PMID: 33753406 PMCID: PMC8485744 DOI: 10.1101/cshperspect.a037960] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Memory T-cell responses are partitioned between the blood, secondary lymphoid organs, and nonlymphoid tissues. Tissue-resident memory T (Trm) cells are a population of immune cells that remain permanently in tissues without recirculating in blood. These nonrecirculating cells serve as a principal node in the anamnestic response to invading pathogens and developing malignancies. Here, we contemplate how T-cell tissue residency is defined and shapes protective immunity in the steady state and in the context of disease. We review the properties and heterogeneity of Trm cells, highlight the critical roles these cells play in maintaining tissue homeostasis and eliciting immune pathology, and explore how they might be exploited to treat disease.
Collapse
Affiliation(s)
- Simone L Park
- Department of Microbiology & Immunology at The Peter Doherty Institute for Infection & Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Laura K Mackay
- Department of Microbiology & Immunology at The Peter Doherty Institute for Infection & Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| |
Collapse
|
14
|
Oliveira T, Zhang M, Joo EJ, Abdel-Azim H, Chen CW, Yang L, Chou CH, Qin X, Chen J, Alagesan K, Almeida A, Jacob F, Packer NH, von Itzstein M, Heisterkamp N, Kolarich D. Glycoproteome remodeling in MLL-rearranged B-cell precursor acute lymphoblastic leukemia. Am J Cancer Res 2021; 11:9519-9537. [PMID: 34646384 PMCID: PMC8490503 DOI: 10.7150/thno.65398] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/03/2021] [Indexed: 01/13/2023] Open
Abstract
B-cell precursor acute lymphoblastic leukemia (BCP-ALL) with mixed-lineage leukemia gene rearrangement (MLL-r) is a poor-prognosis subtype for which additional therapeutic targets are urgently needed. Currently no multi-omics data set for primary MLL r patient cells exists that integrates transcriptomics, proteomics and glycomics to gain an inclusive picture of theranostic targets. Methods: We have integrated transcriptomics, proteomics and glycomics to i) obtain the first inclusive picture of primary patient BCP-ALL cells and identify molecular signatures that distinguish leukemic from normal precursor B-cells and ii) better understand the benefits and limitations of the applied technologies to deliver deep molecular sequence data across major cellular biopolymers. Results: MLL-r cells feature an extensive remodeling of their glycocalyx, with increased levels of Core 2-type O-glycans and complex N-glycans as well as significant changes in sialylation and fucosylation. Notably, glycosaminoglycan remodeling from chondroitin sulfate to heparan sulfate was observed. A survival screen, to determine if glycan remodeling enzymes are redundant, identified MGAT1 and NGLY1, essential components of the N-glycosylation/degradation pathway, as highly relevant within this in vitro screening. OGT and OGA, unique enzymes that regulate intracellular O-GlcNAcylation, were also indispensable. Transcriptomics and proteomics further identified Fes and GALNT7-mediated glycosylation as possible therapeutic targets. While there is overall good correlation between transcriptomics and proteomics data, we demonstrate that a systematic combined multi-omics approach delivers important diagnostic information that is missed when applying a single omics technology. Conclusions: Apart from confirming well-known MLL-r BCP-ALL glycoprotein markers, our integrated multi-omics workflow discovered previously unidentified diagnostic/therapeutic protein targets.
Collapse
Affiliation(s)
- Tiago Oliveira
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD, Australia
| | - Mingfeng Zhang
- Department of Systems Biology, Beckman Research Institute City of Hope, Monrovia, CA, USA
| | - Eun Ji Joo
- Department of Systems Biology, Beckman Research Institute City of Hope, Monrovia, CA, USA
| | - Hisham Abdel-Azim
- Division of Hematology/Oncology and Bone Marrow Transplant, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Chun-Wei Chen
- Department of Systems Biology, Beckman Research Institute City of Hope, Monrovia, CA, USA
| | - Lu Yang
- Department of Systems Biology, Beckman Research Institute City of Hope, Monrovia, CA, USA
| | - Chih-Hsing Chou
- Division of Hematology/Oncology and Bone Marrow Transplant, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Xi Qin
- Department of Systems Biology, Beckman Research Institute City of Hope, Monrovia, CA, USA
| | - Jianjun Chen
- Department of Systems Biology, Beckman Research Institute City of Hope, Monrovia, CA, USA
| | - Kathirvel Alagesan
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD, Australia
| | - Andreia Almeida
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD, Australia
| | - Francis Jacob
- Glyco-Oncology, Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Nicolle H Packer
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD, Australia.,Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, NSW, Australia.,ARC Centre of Excellence for Nanoscale BioPhotonics, Griffith University, QLD and Macquarie University, NSW, Australia
| | - Mark von Itzstein
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD, Australia
| | - Nora Heisterkamp
- Department of Systems Biology, Beckman Research Institute City of Hope, Monrovia, CA, USA.,✉ Corresponding authors: Equal contributions of Nora Heisterkamp, E-mail: ; and Daniel Kolarich, E-mail:
| | - Daniel Kolarich
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD, Australia.,ARC Centre of Excellence for Nanoscale BioPhotonics, Griffith University, QLD and Macquarie University, NSW, Australia.,✉ Corresponding authors: Equal contributions of Nora Heisterkamp, E-mail: ; and Daniel Kolarich, E-mail:
| |
Collapse
|
15
|
van Hooren L, Vaccaro A, Ramachandran M, Vazaios K, Libard S, van de Walle T, Georganaki M, Huang H, Pietilä I, Lau J, Ulvmar MH, Karlsson MCI, Zetterling M, Mangsbo SM, Jakola AS, Olsson Bontell T, Smits A, Essand M, Dimberg A. Agonistic CD40 therapy induces tertiary lymphoid structures but impairs responses to checkpoint blockade in glioma. Nat Commun 2021; 12:4127. [PMID: 34226552 PMCID: PMC8257767 DOI: 10.1038/s41467-021-24347-7] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/09/2021] [Indexed: 12/19/2022] Open
Abstract
Gliomas are brain tumors characterized by an immunosuppressive microenvironment. Immunostimulatory agonistic CD40 antibodies (αCD40) are in clinical development for solid tumors, but are yet to be evaluated for glioma. Here, we demonstrate that systemic delivery of αCD40 in preclinical glioma models induces the formation of tertiary lymphoid structures (TLS) in proximity of meningeal tissue. In treatment-naïve glioma patients, the presence of TLS correlates with increased T cell infiltration. However, systemic delivery of αCD40 induces hypofunctional T cells and impairs the response to immune checkpoint inhibitors in pre-clinical glioma models. This is associated with a systemic induction of suppressive CD11b+ B cells post-αCD40 treatment, which accumulate in the tumor microenvironment. Our work unveils the pleiotropic effects of αCD40 therapy in glioma and reveals that immunotherapies can modulate TLS formation in the brain, opening up for future opportunities to regulate the immune response. Agonistic CD40 antibodies (αCD40) have broad immunostimulatory properties, however their efficacy in glioma remains unclear. Here the authors show that αCD40 promotes the formation of tertiary lymphoid structures but does not improve survival and impairs the response to immune checkpoint blockade in murine glioma models.
Collapse
Affiliation(s)
- Luuk van Hooren
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Alessandra Vaccaro
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Mohanraj Ramachandran
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Konstantinos Vazaios
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Sylwia Libard
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.,Department of Pathology, Uppsala University Hospital, Uppsala, Sweden
| | - Tiarne van de Walle
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Maria Georganaki
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Hua Huang
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Ilkka Pietilä
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Joey Lau
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Maria H Ulvmar
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Maria Zetterling
- Department of Neuroscience, Neurology, Uppsala University, Uppsala, Sweden
| | - Sara M Mangsbo
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Asgeir S Jakola
- Department of Neurosurgery, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Thomas Olsson Bontell
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Pathology and Cytology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anja Smits
- Department of Neuroscience, Neurology, Uppsala University, Uppsala, Sweden.,Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Magnus Essand
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Anna Dimberg
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
16
|
Mandula JK, Rodriguez PC. Tumor-related stress regulates functional plasticity of MDSCs. Cell Immunol 2021; 363:104312. [PMID: 33652258 PMCID: PMC8026602 DOI: 10.1016/j.cellimm.2021.104312] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/15/2021] [Accepted: 01/29/2021] [Indexed: 12/15/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) impair protective anti-tumor immunity and remain major obstacles that stymie the effectiveness of promising cancer therapies. Diverse tumor-derived stressors galvanize the differentiation, intra-tumoral expansion, and immunomodulatory function of MDSCs. These tumor-associated 'axes of stress' underwrite the immunosuppressive programming of MDSCs in cancer and contribute to the phenotypic/functional heterogeneity that characterize tumor-MDSCs. This review discusses various tumor-associated axes of stress that direct MDSC development, accumulation, and immunosuppressive function, as well as current strategies aimed at overcoming the detrimental impact of MDSCs in cancer. To better understand the constellation of signals directing MDSC biology, we herein summarize the pivotal roles, signaling mediators, and effects of reactive oxygen/nitrogen species-related stress, chronic inflammatory stress, hypoxia-linked stress, endoplasmic reticulum stress, metabolic stress, and therapy-associated stress on MDSCs. Although therapeutic targeting of these processes remains mostly pre-clinical, intercepting signaling through the axes of stress could overcome MDSC-related immune suppression in tumor-bearing hosts.
Collapse
Affiliation(s)
- Jessica K Mandula
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Paulo C Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| |
Collapse
|
17
|
Krautter F, Iqbal AJ. Glycans and Glycan-Binding Proteins as Regulators and Potential Targets in Leukocyte Recruitment. Front Cell Dev Biol 2021; 9:624082. [PMID: 33614653 PMCID: PMC7890243 DOI: 10.3389/fcell.2021.624082] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/12/2021] [Indexed: 12/19/2022] Open
Abstract
Leukocyte recruitment is a highly controlled cascade of interactions between proteins expressed by the endothelium and circulating leukocytes. The involvement of glycans and glycan-binding proteins in the leukocyte recruitment cascade has been well-characterised. However, our understanding of these interactions and their regulation has expanded substantially in recent years to include novel lectins and regulatory pathways. In this review, we discuss the role of glycans and glycan-binding proteins, mediating the interactions between endothelium and leukocytes both directly and indirectly. We also highlight recent findings of key enzymes involved in glycosylation which affect leukocyte recruitment. Finally, we investigate the potential of glycans and glycan binding proteins as therapeutic targets to modulate leukocyte recruitment and transmigration in inflammation.
Collapse
Affiliation(s)
- Franziska Krautter
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Asif J Iqbal
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
18
|
Duru G, van Egmond M, Heemskerk N. A Window of Opportunity: Targeting Cancer Endothelium to Enhance Immunotherapy. Front Immunol 2020; 11:584723. [PMID: 33262763 PMCID: PMC7686513 DOI: 10.3389/fimmu.2020.584723] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/19/2020] [Indexed: 12/19/2022] Open
Abstract
Vascular abnormalities in tumors have a major impact on the immune microenvironment in tumors. The consequences of abnormal vasculature include increased hypoxia, acidosis, high intra-tumoral fluid pressure, and angiogenesis. This introduces an immunosuppressive microenvironment that alters immune cell maturation, activation, and trafficking, which supports tumor immune evasion and dissemination of tumor cells. Increasing data suggests that cancer endothelium is a major barrier for traveling leukocytes, ranging from a partial blockade resulting in a selective endothelial barrier, to a complete immune infiltration blockade associated with immune exclusion and immune desert cancer phenotypes. Failed immune cell trafficking as well as immunosuppression within the tumor microenvironment limits the efficacy of immunotherapeutic approaches. As such, targeting proteins with key roles in angiogenesis may potentially reduce immunosuppression and might restore infiltration of anti-tumor immune cells, creating a therapeutic window for successful immunotherapy. In this review, we provide a comprehensive overview of established as well as more controversial endothelial pathways that govern selective immune cell trafficking across cancer endothelium. Additionally, we discuss recent insights and strategies that target tumor vasculature in order to increase infiltration of cytotoxic immune cells during the therapeutic window of vascular normalization hereby improving the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Gizem Duru
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection & Immunity, Amsterdam, Netherlands
| | - Marjolein van Egmond
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection & Immunity, Amsterdam, Netherlands
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Surgery, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Niels Heemskerk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection & Immunity, Amsterdam, Netherlands
| |
Collapse
|
19
|
Zeng J, Eljalby M, Aryal RP, Lehoux S, Stavenhagen K, Kudelka MR, Wang Y, Wang J, Ju T, von Andrian UH, Cummings RD. Cosmc controls B cell homing. Nat Commun 2020; 11:3990. [PMID: 32778659 PMCID: PMC7417590 DOI: 10.1038/s41467-020-17765-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 07/17/2020] [Indexed: 12/15/2022] Open
Abstract
The molecular mechanisms regulating lymphocyte homing into lymph nodes are only partly understood. Here, we report that B cell-specific deletion of the X-linked gene, Cosmc, and the consequent decrease of protein O-glycosylation, induces developmental blocks of mouse B cells. After transfer into wild-type recipient, Cosmc-null B cells fail to home to lymph nodes as well as non-lymphoid organs. Enzymatic desialylation of wild-type B cells blocks their migration into lymph nodes, indicating a requirement of sialylated O-glycans for proper trafficking. Mechanistically, Cosmc-deficient B cells have normal rolling and firm arrest on high endothelium venules (HEV), thereby attributing their inefficient trafficking to alterations in the subsequent transendothelial migration step. Finally, Cosmc-null B cells have defective chemokine signaling responses. Our results thus demonstrate that Cosmc and its effects on O-glycosylation are important for controlling B cell homing.
Collapse
Affiliation(s)
- Junwei Zeng
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Mahmoud Eljalby
- Department of Microbiology & Immunobiology, Harvard Medical School, Boston, MA, USA
| | - Rajindra P Aryal
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sylvain Lehoux
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kathrin Stavenhagen
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Matthew R Kudelka
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Biochemistry, Emory University, Atlanta, GA, USA
| | - Yingchun Wang
- Department of Biochemistry, Emory University, Atlanta, GA, USA
| | - Jianmei Wang
- Department of Biochemistry, Emory University, Atlanta, GA, USA
| | - Tongzhong Ju
- Department of Biochemistry, Emory University, Atlanta, GA, USA
- Office of Biotechnology Products, Center for Drug Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Ulrich H von Andrian
- Department of Microbiology & Immunobiology, Harvard Medical School, Boston, MA, USA
- The Ragon Institute of MGH, MIT & Harvard, Cambridge, MA, USA
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
20
|
De Bousser E, Meuris L, Callewaert N, Festjens N. Human T cell glycosylation and implications on immune therapy for cancer. Hum Vaccin Immunother 2020; 16:2374-2388. [PMID: 32186959 PMCID: PMC7644206 DOI: 10.1080/21645515.2020.1730658] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Glycosylation is an important post-translational modification, giving rise to a diverse and abundant repertoire of glycans on the cell surface, collectively known as the glycome. When focusing on immunity, glycans are indispensable in virtually all signaling and cell-cell interactions. More specifically, glycans have been shown to regulate key pathophysiological steps within T cell biology such as T cell development, thymocyte selection, T cell activity and signaling as well as T cell differentiation and proliferation. They are of major importance in determining the interaction of human T cells with tumor cells. In this review, we will describe the role of glycosylation of human T cells in more depth, elaborate on the importance of glycosylation in the interaction of human T cells with tumor cells and discuss the potential of cancer immunotherapies that are based on manipulating the glycome functions at the tumor immune interface.1,2
Collapse
Affiliation(s)
- Elien De Bousser
- VIB-UGent Center for Medical Biotechnology , Ghent, Belgium.,Department of Biochemistry and Microbiology, Ghent University , Ghent, Belgium
| | - Leander Meuris
- VIB-UGent Center for Medical Biotechnology , Ghent, Belgium.,Department of Biochemistry and Microbiology, Ghent University , Ghent, Belgium
| | - Nico Callewaert
- VIB-UGent Center for Medical Biotechnology , Ghent, Belgium.,Department of Biochemistry and Microbiology, Ghent University , Ghent, Belgium
| | - Nele Festjens
- VIB-UGent Center for Medical Biotechnology , Ghent, Belgium.,Department of Biochemistry and Microbiology, Ghent University , Ghent, Belgium
| |
Collapse
|
21
|
Perkey E, Maurice De Sousa D, Carrington L, Chung J, Dils A, Granadier D, Koch U, Radtke F, Ludewig B, Blazar BR, Siebel CW, Brennan TV, Nolz J, Labrecque N, Maillard I. GCNT1-Mediated O-Glycosylation of the Sialomucin CD43 Is a Sensitive Indicator of Notch Signaling in Activated T Cells. THE JOURNAL OF IMMUNOLOGY 2020; 204:1674-1688. [PMID: 32060138 DOI: 10.4049/jimmunol.1901194] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/10/2020] [Indexed: 01/05/2023]
Abstract
Notch signaling is emerging as a critical regulator of T cell activation and function. However, there is no reliable cell surface indicator of Notch signaling across activated T cell subsets. In this study, we show that Notch signals induce upregulated expression of the Gcnt1 glycosyltransferase gene in T cells mediating graft-versus-host disease after allogeneic bone marrow transplantation in mice. To determine if Gcnt1-mediated O-glycosylation could be used as a Notch signaling reporter, we quantified the core-2 O-glycoform of CD43 in multiple T cell subsets during graft-versus-host disease. Pharmacological blockade of Delta-like Notch ligands abrogated core-2 O-glycosylation in a dose-dependent manner after allogeneic bone marrow transplantation, both in donor-derived CD4+ and CD8+ effector T cells and in Foxp3+ regulatory T cells. CD43 core-2 O-glycosylation depended on cell-intrinsic canonical Notch signals and identified CD4+ and CD8+ T cells with high cytokine-producing ability. Gcnt1-deficient T cells still drove lethal alloreactivity, showing that core-2 O-glycosylation predicted, but did not cause, Notch-dependent T cell pathogenicity. Using core-2 O-glycosylation as a marker of Notch signaling, we identified Ccl19-Cre+ fibroblastic stromal cells as critical sources of Delta-like ligands in graft-versus-host responses irrespective of conditioning intensity. Core-2 O-glycosylation also reported Notch signaling in CD8+ T cell responses to dendritic cell immunization, Listeria infection, and viral infection. Thus, we uncovered a role for Notch in controlling core-2 O-glycosylation and identified a cell surface marker to quantify Notch signals in multiple immunological contexts. Our findings will help refine our understanding of the regulation, cellular source, and timing of Notch signals in T cell immunity.
Collapse
Affiliation(s)
- Eric Perkey
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109.,Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Dave Maurice De Sousa
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montreal, Quebec H1T 2M4, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Léolène Carrington
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Jooho Chung
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Alexander Dils
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - David Granadier
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Ute Koch
- École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Freddy Radtke
- École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, 9007 St. Gallen, Switzerland
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455
| | | | | | - Jeffrey Nolz
- Oregon Health and Sciences University, Portland, OR 97239; and
| | - Nathalie Labrecque
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montreal, Quebec H1T 2M4, Canada; .,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3T 1J4, Canada.,Département de Médecine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Ivan Maillard
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104;
| |
Collapse
|
22
|
Nolz JC, Richer MJ. Control of memory CD8 + T cell longevity and effector functions by IL-15. Mol Immunol 2019; 117:180-188. [PMID: 31816491 DOI: 10.1016/j.molimm.2019.11.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/12/2019] [Accepted: 11/27/2019] [Indexed: 12/11/2022]
Abstract
IL-15 is a member of the common gamma chain family of cytokines and plays important roles in regulating several aspects of innate and adaptive immunity. Besides its established role in controlling homeostatic proliferation and survival of memory CD8+ T cells and natural killer cells, recent findings demonstrate that inflammatory IL-15 can also stimulate a variety of effector functions, such as enhanced cytotoxicity, entry into the cell cycle, and trafficking into non-lymphoid tissues. Here, we discuss how IL-15 is critical in regulating many functions of memory CD8+ T cells and how these processes act collectively to ensure optimal protective cellular immunity against re-infections.
Collapse
Affiliation(s)
- Jeffrey C Nolz
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, United States; Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, United States; Department of Radiation Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, United States.
| | - Martin J Richer
- Department of Microbiology & Immunology, McGill University, 712 McIntyre Medical Building, 3655 promenade Sir William Osler, Montreal, Quebec, Canada; Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
23
|
Osborn JF, Hobbs SJ, Mooster JL, Khan TN, Kilgore AM, Harbour JC, Nolz JC. Central memory CD8+ T cells become CD69+ tissue-residents during viral skin infection independent of CD62L-mediated lymph node surveillance. PLoS Pathog 2019; 15:e1007633. [PMID: 30875408 PMCID: PMC6420010 DOI: 10.1371/journal.ppat.1007633] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 02/11/2019] [Indexed: 01/22/2023] Open
Abstract
Memory CD8+ T cells in the circulation rapidly infiltrate non-lymphoid tissues following infection and provide protective immunity in an antigen-specific manner. However, the subsequent fate of memory CD8+ T cells after entering non-lymphoid tissues such as the skin during a secondary infection is largely unknown. Furthermore, because expression of CD62L is often used to identify the central memory (TCM) CD8+ T cell subset, uncoupling the physical requirement for CD62L-mediated lymph node homing versus other functional attributes of TCM CD8+ T cells remains unresolved. Here, we show that in contrast to naïve CD8+ T cells, memory CD8+ T cells traffic into the skin independent of CD62L-mediated lymph node re-activation and provide robust protective immunity against Vaccinia virus (VacV) infection. TCM, but not effector memory (TEM), CD8+ T cells differentiated into functional CD69+/CD103- tissue residents following viral clearance, which was also dependent on local recognition of antigen in the skin microenvironment. Finally, we found that memory CD8+ T cells expressed granzyme B after trafficking into the skin and utilized cytolysis to provide protective immunity against VacV infection. Collectively, these findings demonstrate that TCM CD8+ T cells become cytolytic following rapid infiltration of the skin to protect against viral infection and subsequently differentiate into functional CD69+ tissue-residents.
Collapse
Affiliation(s)
- Jossef F. Osborn
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Samuel J. Hobbs
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Jana L. Mooster
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Tahsin N. Khan
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Augustus M. Kilgore
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Jake C. Harbour
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Jeffrey C. Nolz
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, Oregon, United States of America
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon, United States of America
- Radiation Medicine, Oregon Health & Science University, Portland, Oregon, United States of America
- * E-mail:
| |
Collapse
|
24
|
Effector and Regulatory T Cells Roll at High Shear Stress by Inducible Tether and Sling Formation. Cell Rep 2019; 21:3885-3899. [PMID: 29281835 DOI: 10.1016/j.celrep.2017.11.099] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/04/2017] [Accepted: 11/28/2017] [Indexed: 01/07/2023] Open
Abstract
The adaptive immune response involves T cell differentiation and migration to sites of inflammation. T cell trafficking is initiated by rolling on inflamed endothelium. Tethers and slings, discovered in neutrophils, facilitate cell rolling at high shear stress. Here, we demonstrate that the ability to form tethers and slings during rolling is highly inducible in T helper 1 (Th1), Th17, and regulatory T (Treg) cells but less in Th2 cells. In vivo, endogenous Treg cells rolled stably in cremaster venules at physiological shear stress. Quantitative dynamic footprinting nanoscopy of Th1, Th17, and Treg cells uncovered the formation of multiple tethers per cell. Human Th1 cells also showed tethers and slings. RNA sequencing (RNA-seq) revealed the induction of cell migration and cytoskeletal genes in sling-forming cells. We conclude that differentiated CD4 T cells stabilize rolling by inducible tether and sling formation. These phenotypic changes approximate the adhesion phenotype of neutrophils and support CD4 T cell access to sites of inflammation.
Collapse
|
25
|
Abstract
Females have more robust immune responses than males, well-illustrated by the degree of inflammation elicited during delayed-type hypersensitivity (DTH) responses. Here, we have investigated underlying sex differences that may contribute to differential footpad DTH responses using wildtype and four core genotypes (FCG) mice and popliteal lymphnode cellularity and gene expression. DTH responses in XX and XY FCG females showed no role for almost all genes expressed on sex chromosomes. After then filtering-out genes differentially expressed between XX and XY females, only one gene was sexually differentially expressed in wildtype mice, glycosylation-dependent cell adhesion molecule 1 (Glycam1), expressed 7-fold higher in females. Glycam1 facilitates leukocyte entry through high endothelial venules. Consistent with greater Glycam1 expression, female nodes contained twice as many cells. While females had more memory T cells in their nodes, males had a higher percentage of T regulatory cells. This sexual dimorphism in wildtype animals manifested pre-pubertally, was enhanced post-pubertally, and was eliminated by castration. The formation of male gonads is determined by the expression of Sry. Sry overexpression, which does not affect testosterone levels, produced an exaggerated male phenotype. We conclude that Sry expression through formation of the male gonad indirectly negatively impacts the potential for local inflammation.
Collapse
|
26
|
Peruzzi C, Nascimento S, Gauer B, Nardi J, Sauer E, Göethel G, Cestonaro L, Fão N, Cattani S, Paim C, Souza J, Gnoatto D, Garcia SC. Inflammatory and oxidative stress biomarkers at protein and molecular levels in workers occupationally exposed to crystalline silica. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:1394-1405. [PMID: 30426371 DOI: 10.1007/s11356-018-3693-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 11/05/2018] [Indexed: 06/09/2023]
Abstract
Workers chronically exposed to respirable crystalline silica (CS) are susceptible to adverse health effects like silicosis and lung cancer. This study aimed to investigate potential early peripheral biomarkers of inflammation and oxidative stress in miners. The subjects enrolled in this study were occupationally unexposed workers (OUW, n = 29) and workers exposed to crystalline silica (WECS), composed by miners, which were divided into two subgroups: workers without silicosis (WECS I, n = 39) and workers diagnosed with silicosis, retired from work (WECS II, n = 42). The following biomarkers were evaluated: gene expression of L-selectin, CXCL2, CXCL8 (IL-8), HO-1, and p53; malondialdehyde (MDA) plasma levels and non-protein thiol levels in erythrocytes. Additionally, protein expression of L-selectin was evaluated to confirm our previous findings. The results demonstrated that gene expression of L-selectin was decreased in the WECS I group when compared to the OUW group (p < 0.05). Regarding gene expression of CXCL2, CXCL8 (IL-8), HO-1, and p53, significant fold change decreases were observed in workers exposed to CS in relation to unexposed workers (p < 0.05). The results of L-selectin protein expression in lymphocyte surface corroborated with our previous findings; thus, significant downregulation in the WECS groups was observed compared to OUW group (p < 0.05). The MDA was negatively associated with the gene expression of CXCL-2, CXCL8 (IL-8), and p53 (p < 0.05). The participants with silicosis (WECS II) presented significant increased non-protein thiol levels in relation to other groups (p < 0.05). Taken together, our findings may contribute to help the knowledge about the complex mechanisms involved in the silicosis pathogenesis and in the risk of lung cancer development in workers chronically exposed to respirable CS.
Collapse
Affiliation(s)
- Caroline Peruzzi
- Laboratório de Toxicologia (LATOX), Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, 90610-000, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas (PPGCF), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Sabrina Nascimento
- Laboratório de Toxicologia (LATOX), Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, 90610-000, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas (PPGCF), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Bruna Gauer
- Laboratório de Toxicologia (LATOX), Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, 90610-000, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas (PPGCF), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Jessica Nardi
- Laboratório de Toxicologia (LATOX), Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, 90610-000, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas (PPGCF), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Elisa Sauer
- Laboratório de Toxicologia (LATOX), Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, 90610-000, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas (PPGCF), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Gabriela Göethel
- Laboratório de Toxicologia (LATOX), Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, 90610-000, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas (PPGCF), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Larissa Cestonaro
- Laboratório de Toxicologia (LATOX), Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, 90610-000, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas (PPGCF), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Nuryan Fão
- Laboratório de Toxicologia (LATOX), Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, 90610-000, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas (PPGCF), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Shanda Cattani
- Laboratório de Toxicologia (LATOX), Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, 90610-000, Brazil
| | - Cristiane Paim
- FUNDACENTRO, Fundação Jorge Duprat e Figueiredo, Porto Alegre, RS, Brazil
| | - Jorge Souza
- Unidade Regional de Saúde do Trabalhador (UREST), Ametista do Sul, RS, Brazil
| | - Daniela Gnoatto
- Unidade Regional de Saúde do Trabalhador (UREST), Ametista do Sul, RS, Brazil
| | - Solange Cristina Garcia
- Laboratório de Toxicologia (LATOX), Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, 90610-000, Brazil.
- Programa de Pós-Graduação em Ciências Farmacêuticas (PPGCF), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| |
Collapse
|
27
|
Giovannone N, Antonopoulos A, Liang J, Geddes Sweeney J, Kudelka MR, King SL, Lee GS, Cummings RD, Dell A, Barthel SR, Widlund HR, Haslam SM, Dimitroff CJ. Human B Cell Differentiation Is Characterized by Progressive Remodeling of O-Linked Glycans. Front Immunol 2018; 9:2857. [PMID: 30619255 PMCID: PMC6302748 DOI: 10.3389/fimmu.2018.02857] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/20/2018] [Indexed: 12/16/2022] Open
Abstract
Germinal centers (GC) are microanatomical niches where B cells proliferate, undergo antibody affinity maturation, and differentiate to long-lived memory B cells and antibody-secreting plasma cells. For decades, GC B cells have been defined by their reactivity to the plant lectin peanut agglutinin (PNA), which binds serine/threonine (O-linked) glycans containing the asialylated disaccharide Gal-β1,3-GalNAc-Ser/Thr (also called T-antigen). In T cells, acquisition of PNA binding by activated T cells and thymocytes has been linked with altered tissue homing patterns, cell signaling, and survival. Yet, in GC B cells, the glycobiological basis and significance of PNA binding remains surprisingly unresolved. Here, we investigated the basis for PNA reactivity of GC B cells. We found that GC B cell binding to PNA is associated with downregulation of the α2,3 sialyltransferase, ST3GAL1 (ST3Gal1), and overexpression of ST3Gal1 was sufficient to reverse PNA binding in B cell lines. Moreover, we found that the primary scaffold for PNA-reactive O-glycans in B cells is the B cell receptor-associated receptor-type tyrosine phosphatase CD45, suggesting a role for altered O-glycosylation in antigen receptor signaling. Consistent with similar reports in T cells, ST3Gal1 overexpression in B cells in vitro induced drastic shortening in O-glycans, which we confirmed by both antibody staining and mass spectrometric O-glycomic analysis. Unexpectedly, ST3Gal1-induced changes in O-glycan length also correlated with altered binding of two glycosylation-sensitive CD45 antibodies, RA3-6B2 (more commonly called B220) and MEM55, which (in humans) have previously been reported to favor binding to naïve/GC subsets and memory/plasmablast subsets, respectively. Analysis of primary B cell binding to B220, MEM55, and several plant lectins suggested that B cell differentiation is accompanied by significant loss of O-glycan complexity, including loss of extended Core 2 O-glycans. To our surprise, decreased O-glycan length from naïve to post-GC fates best correlated not with ST3Gal1, but rather downregulation of the Core 2 branching enzyme GCNT1. Thus, our data suggest that O-glycan remodeling is a feature of B cell differentiation, dually regulated by ST3Gal1 and GCNT1, that ultimately results in expression of distinct O-glycosylation states/CD45 glycoforms at each stage of B cell differentiation.
Collapse
Affiliation(s)
- Nicholas Giovannone
- Department of Dermatology, Brigham and Women's Hospital, Boston MA, United States.,Harvard Medical School, Boston MA, United States
| | | | - Jennifer Liang
- Department of Dermatology, Brigham and Women's Hospital, Boston MA, United States
| | - Jenna Geddes Sweeney
- Department of Dermatology, Brigham and Women's Hospital, Boston MA, United States.,Harvard Medical School, Boston MA, United States
| | - Matthew R Kudelka
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, United States.,Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, United States
| | - Sandra L King
- Department of Dermatology, Brigham and Women's Hospital, Boston MA, United States
| | - Gi Soo Lee
- Department of Otology and Laryngology, Harvard Medical School, Boston, MA, United States
| | - Richard D Cummings
- Harvard Medical School, Boston MA, United States.,Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Anne Dell
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Steven R Barthel
- Department of Dermatology, Brigham and Women's Hospital, Boston MA, United States
| | - Hans R Widlund
- Department of Dermatology, Brigham and Women's Hospital, Boston MA, United States.,Harvard Medical School, Boston MA, United States
| | - Stuart M Haslam
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Charles J Dimitroff
- Department of Dermatology, Brigham and Women's Hospital, Boston MA, United States.,Harvard Medical School, Boston MA, United States
| |
Collapse
|
28
|
Borsig L. Selectins in cancer immunity. Glycobiology 2018; 28:648-655. [PMID: 29272415 DOI: 10.1093/glycob/cwx105] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 12/18/2017] [Indexed: 12/12/2022] Open
Abstract
Selectins are vascular adhesion molecules that mediate physiological responses such as inflammation, immunity and hemostasis. During cancer progression, selectins promote various steps enabling the interactions between tumor cells and the blood constituents, including platelets, endothelial cells and leukocytes. Selectins are carbohydrate-binding molecules that bind to sialylated, fucosylated glycan structures. The increased selectin ligand expression on tumor cells correlates with enhanced metastasis and poor prognosis for cancer patients. While, many studies focused on the role of selectin as a mediator of tumor cell adhesion and extravasation during metastasis, there is evidence for selectins to activate signaling cascade that regulates immune responses within a tumor microenvironment. L-Selectin binding induces activation of leukocytes, which can be further modulated by selectin-mediated interactions with platelets and endothelial cells. Selectin ligand on leukocytes, PSGL-1, triggers intracellular signaling in leukocytes that are induced through platelet's P-selectin or endothelial E-selectin binding. In this review, I summarize the evidence for selectin-induced immune modulation in cancer progression that represents a possible target for controlling tumor immunity.
Collapse
Affiliation(s)
- Lubor Borsig
- Institute of Physiology, University of Zurich and Zurich Center for Integrative Human Physiology, Winterthurerstrasse 190, Zurich, Switzerland
| |
Collapse
|
29
|
Dias AM, Pereira MS, Padrão NA, Alves I, Marcos-Pinto R, Lago P, Pinho SS. Glycans as critical regulators of gut immunity in homeostasis and disease. Cell Immunol 2018; 333:9-18. [DOI: 10.1016/j.cellimm.2018.07.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 07/04/2018] [Accepted: 07/17/2018] [Indexed: 12/19/2022]
|
30
|
Osborn JF, Mooster JL, Hobbs SJ, Munks MW, Barry C, Harty JT, Hill AB, Nolz JC. Enzymatic synthesis of core 2 O-glycans governs the tissue-trafficking potential of memory CD8 + T cells. Sci Immunol 2018; 2:2/16/eaan6049. [PMID: 29030501 DOI: 10.1126/sciimmunol.aan6049] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 08/08/2017] [Indexed: 12/14/2022]
Abstract
Trafficking of memory CD8+ T cells out of the circulation is essential to provide protective immunity against intracellular pathogens in nonlymphoid tissues. However, the molecular mechanisms that dictate the trafficking potential of diverse memory CD8+ T cell populations are not completely defined. We show that after infection or inflammatory challenge, central memory (TCM) CD8+ T cells rapidly traffic into nonlymphoid tissues, whereas most effector memory cells remain in the circulation. Furthermore, we demonstrate that cellular migration of memory CD8+ T cells into nonlymphoid tissues is driven by interleukin-15 (IL-15)-stimulated enzymatic synthesis of core 2 O-glycans, which generates functional ligands for E- and P-selectins. Given that IL-15-stimulated expression of glycosyltransferase enzymes is largely a feature of TCM CD8+ T cells, this allows TCM to selectively migrate out of the circulation and into nonlymphoid tissues. Collectively, our data indicate that entry of memory CD8+ T cells into inflamed, nonlymphoid tissues is primarily restricted to TCM cells that have the capacity to synthesize core 2 O-glycans.
Collapse
Affiliation(s)
- Jossef F Osborn
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Jana L Mooster
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Samuel J Hobbs
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Michael W Munks
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Conrad Barry
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239, USA
| | - John T Harty
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA
| | - Ann B Hill
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Jeffrey C Nolz
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239, USA. .,Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR 97239, USA.,Department of Radiation Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| |
Collapse
|
31
|
Hobbs SJ, Osborn JF, Nolz JC. Activation and trafficking of CD8 + T cells during viral skin infection: immunological lessons learned from vaccinia virus. Curr Opin Virol 2018; 28:12-19. [PMID: 29080420 PMCID: PMC5835170 DOI: 10.1016/j.coviro.2017.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 10/07/2017] [Indexed: 01/13/2023]
Abstract
Epicutaneous delivery of vaccinia virus (VacV) by scarification of the skin generates robust and durable protective immunity, which was ultimately responsible for eradicating smallpox from the human race. Therefore, infection of the skin with VacV is often used in experimental model systems to study the activation of adaptive immunity, as well as the development and functional features of immunological memory. Here, we describe recent advances using this viral infection to identify and characterize the mechanisms regulating the activation and trafficking of cytotoxic CD8+ T cells into the inflamed skin, the migratory features of CD8+ T cells within the skin microenvironment, and finally, their subsequent differentiation into tissue-resident memory cells.
Collapse
Affiliation(s)
- Samuel J Hobbs
- Departments of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, United States
| | - Jossef F Osborn
- Departments of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, United States
| | - Jeffrey C Nolz
- Departments of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, United States; Departments of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, United States; Departments of Radiation Medicine, Oregon Health & Science University, Portland, OR 97239, United States.
| |
Collapse
|