101
|
Guo Z, Zhang K, Wei X, Li Y, Ma X, Li Y, Han D, Du Q, Zhang T, Chen X, Wei H, Yan C, Zhang W, Pang Q, Wang P. Radiotherapy plus camrelizumab affects peripheral CD8 T-cell differentiation subsets expressing PD-1, TIGIT, and CTLA-4 in esophageal squamous cell carcinoma. J Leukoc Biol 2023; 113:11-17. [PMID: 36822161 DOI: 10.1093/jleuko/qiac018] [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: 03/22/2022] [Indexed: 01/11/2023] Open
Abstract
Our previous phase Ib trial (NCT03222440) showed that radiotherapy plus the anti-PD-1 antibody camrelizumab is a safe and feasible first-line therapy for locally advanced esophageal squamous cell carcinoma. In this study, we divided peripheral CD8 T-cell differentiation subsets into 4 subpopulations (naive T cells, central memory T cells, effector memory T cells, and CD45RA+ effector memory T cells). We then investigated the influence of radiotherapy plus camrelizumab therapy on the proportions of the 4 subsets and their PD-1, TIGIT, and CTLA-4 expression as well as their proliferative activity and compared the effects with those of concurrent chemoradiotherapy. Nineteen and 15 patients with esophageal squamous cell carcinoma who received radiotherapy plus camrelizumab therapy and concurrent chemoradiotherapy, respectively, were enrolled in this study. We isolated peripheral blood mononuclear cells from these patients before treatment and longitudinally after the delivery of 40 Gy radiotherapy. Flow cytometry was conducted to detect peripheral CD8 T-cell subsets and PD-1, TIGIT, CTLA-4, and Ki67 expression levels in patients with esophageal squamous cell carcinoma. We found that radiotherapy plus camrelizumab therapy did not change the proportions of the 4 subsets or the expression of CTLA-4, but this therapy decreased PD-1 expression by the 4 subsets and TIGIT expression by effector memory T cells, as well as significantly enhanced the proliferative activity of CD8 T cells, whereas concurrent chemoradiotherapy produced different effects. In addition, we further identified peripheral biomarkers that potentially predict the outcome of radiotherapy plus camrelizumab therapy.
Collapse
Affiliation(s)
- Zhoubo Guo
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Kunning Zhang
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Xiaoying Wei
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Yanqi Li
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Xiaoxue Ma
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Yang Li
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Dong Han
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Qingwu Du
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Tian Zhang
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Xi Chen
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Hui Wei
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Cihui Yan
- Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Wencheng Zhang
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Qingsong Pang
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| | - Ping Wang
- Departments of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhu West Road, Hexi District, Tianjin, China
| |
Collapse
|
102
|
Mass Cytometry Reveals the Imbalanced Immune State in the Peripheral Blood of Patients with Essential Hypertension. Cardiovasc Ther 2023; 2023:9915178. [PMID: 36891527 PMCID: PMC9988372 DOI: 10.1155/2023/9915178] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 03/02/2023] Open
Abstract
Mounting evidence has confirmed that essential hypertension (EH) is closely related to low-grade inflammation, but there is still a lack of in-depth understanding of the state of immune cells in the circulating blood of patients with EH. We analyzed whether hypertensive peripheral blood immune cell balance was destroyed. The peripheral blood mononuclear cells (PBMCs) of all subjects were analyzed using time-of-flight cytometry (CyTOF) based on 42 kinds of metal-binding antibodies. CD45+ cells were categorized into 32 kinds of subsets. Compared with the health control (HC) group, the percentage of total dendritic cells, two kinds of myeloid dendritic cell subsets, one intermediate/nonclassical monocyte subset and one CD4+ central memory T cell subset in the EH group, was significantly higher; the percentage of low-density neutrophils, four kinds of classical monocyte subsets, one CD14lowCD16- monocyte subset, one naive CD4+ and one naive CD8+ T cell subsets, one CD4+ effector and one CD4+ central memory T cell subsets, one CD8+ effector memory T cell subset, and one terminally differentiated γδ T cell subset, decreased significantly in EH. What is more, the expression of many important antigens was enhanced in CD45+ immune cells, granulocytes, and B cells in patients with EH. In conclusion, the altered number and antigen expression of immune cells reflect the imbalanced immune state of the peripheral blood in patients with EH.
Collapse
|
103
|
Li N, Shi T, Zhang M, He Y, Feng J, Mei Z, Su X, Jie Z. PLZF promotes the development of asthma tolerance via affecting memory phenotypes of immune cells. Int Immunopharmacol 2023; 114:109559. [PMID: 36525795 DOI: 10.1016/j.intimp.2022.109559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
Clarifying the pathogenesis of asthma and/or identifying the specific pathway underlying oral asthma tolerance (OT) would be of great significance. In our previous study, promyelocytic leukemia zinc finger (PLZF), which reportedly regulates memory phenotypes, was found to promote ovalbumin (OVA)-induced OT. Therefore, this study aimed to explore the regulatory effects of PLZF on memory phenotypes in asthma and OT mouse models. We found that Zbtb16 (encoding PLZF) and PLZF+ cells were highly increased in OT lungs compared with asthmatic lungs. PLZF was co-expressed with GATA3, and IL-4+PLZF+ cells were significantly lower in OT lungs than in asthmatic lungs. Notably, memory cells were decreased in OT mice, and these mice had PLZF+ cells that expressed lower levels of CD44 than those of asthmatic mice. When Zbtb16 was overexpressed in splenic lymphocytes, the number of CD44+ cells decreased. There were increased memory cells in splenic lymphocytes after treatment with the supernatant of OVA-treated airway epithelial cells; however, this was reversed by Zbtb16 overexpression. Early respiratory syncytial virus infection increased memory cells and reduced PLZF+ cells in the OT mice. Collectively, these results indicate that PLZF may reduce the proportion of memory cells, thereby, promoting the establishment of OT.
Collapse
Affiliation(s)
- Na Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China; Department of Medicine, Respiratory, Emergency and Intensive Care Medicine, The Affiliated Dushu Lake Hospital of Soochow University, Suzhou, China
| | - Tianyun Shi
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Meng Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Yanchao He
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Jingjing Feng
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Zhoufang Mei
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Xiao Su
- Unit of Respiratory Infection and Immunity, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.
| | - Zhijun Jie
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
104
|
Younger DS. Pediatric neuropsychiatric disorders with motor and nonmotor phenomena. HANDBOOK OF CLINICAL NEUROLOGY 2023; 196:367-387. [PMID: 37620079 DOI: 10.1016/b978-0-323-98817-9.00028-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
The concept of pediatric autoimmune neuropsychiatric disorders associated with group A beta-hemolytic streptococcus (PANDAS) has become seminal since first introduced more than two decades ago. At the time of this writing, most neurologists, pediatricians, psychiatrists, and general pediatricians will probably have heard of this association or treated an affected child with PANDAS. The concept of an acute-onset, and typically self-limited, postinfectious autoimmune neuropsychiatric disorder resembling PANDAS manifesting vocal and motor tics and obsessive-compulsive disorder has broadened to other putative microbes and related endogenous and exogenous disease triggers. These disorders with common features of hypometabolism in the medial temporal lobe and hippocampus in brain 18fluorodeoxyglucose positron emission tomography fused to magnetic resonance imaging (FDG PET-MRI), form a spectrum: with the neuropsychiatric disorder Tourette syndrome and PANDAS with its well-defined etiopathogenesis at one end, and pediatric abrupt-onset neuropsychiatric syndrome (PANS), alone or associated with specific bacterial and viral pathogens, at the other end. The designation of PANS in the absence of a specific trigger, as an exclusionary diagnosis, reflects the current problem in nosology.
Collapse
Affiliation(s)
- David S Younger
- Department of Clinical Medicine and Neuroscience, CUNY School of Medicine, New York, NY, United States; Department of Medicine, Section of Internal Medicine and Neurology, White Plains Hospital, White Plains, NY, United States.
| |
Collapse
|
105
|
Gao A, Zhao W, Wu R, Su R, Jin R, Luo J, Gao C, Li X, Wang C. Tissue-resident memory T cells: The key frontier in local synovitis memory of rheumatoid arthritis. J Autoimmun 2022; 133:102950. [PMID: 36356551 DOI: 10.1016/j.jaut.2022.102950] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/27/2022] [Accepted: 10/29/2022] [Indexed: 11/09/2022]
Abstract
Rheumatoid arthritis (RA) is a highly disabling, systemic autoimmune disease. It presents a remarkable tendency to recur, which renders it almost impossible for patients to live without drugs. Under such circumstances, many patients have to suffer the pain of recurrent attacks as well as the side effects of long-term medication. Current therapies for RA are primarily systemic treatments without targeting the problem that RA is more likely to recur locally. Emerging studies suggest the existence of a mechanism mediating local memory during RA, which is closely related to the persistent residence of tissue-resident memory T cells (TRM). TRM, one of the memory T cell subsets, reside in tissues providing immediate immune protection but driving recurrent local inflammation on the other hand. The heterogeneity among synovial TRM is unclear, with the dominated CD8+ TRM observed in inflamed synovium of RA patients coming into focus. Besides local arthritis relapse, TRM may also contribute to extra-articular organ involvement in RA due to their migration potential. Future integration of single-cell RNA sequencing (scRNA-seq) with spatial transcriptomics to explore the gene expression patterns of TRM in both temporal dimension and spatial dimension may help us identify specific therapeutic targets. Targeting synovial TRM to suppress local arthritis flares while using systemic therapies to prevent extra-articular organ involvement may provide a new perspective to address RA recurrence.
Collapse
Affiliation(s)
- Anqi Gao
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory for Immunomicroecology, Shanxi, China
| | - Wenpeng Zhao
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory for Immunomicroecology, Shanxi, China
| | - Ruihe Wu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory for Immunomicroecology, Shanxi, China
| | - Rui Su
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory for Immunomicroecology, Shanxi, China
| | - Ruqing Jin
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory for Immunomicroecology, Shanxi, China
| | - Jing Luo
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory for Immunomicroecology, Shanxi, China
| | - Chong Gao
- Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital/Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaofeng Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory for Immunomicroecology, Shanxi, China
| | - Caihong Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Key Laboratory for Immunomicroecology, Shanxi, China.
| |
Collapse
|
106
|
Meng J, Yu Z, Chen H, Yu X, Jiang M, Zeng XA, You J. Brucea javanica oil emulsion significantly improved the effect of anti-programmed cell death protein-1 immunotherapy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 107:154446. [PMID: 36182799 DOI: 10.1016/j.phymed.2022.154446] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 08/24/2022] [Accepted: 09/05/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Brucea javanica oil (BJO) is the active substance extracted from the dry and mature fruit of Brucea javanica. Its pharmaceutical preparation, BJO emulsion (BJOE), is one of the most widely studied traditional Chinese medicine preparations for the treatment of malignancy. However, the unrevealed anti-tumor mechanism immensely limits further development of BJOE. PURPOSE In this study, we delved into the anti-tumor mechanism of commercial BJOE, including its influence on the tumor microenvironment (TME) and the treatment effect when combined with anti-programmed cell death protein-1 (PD-1) therapy. METHODS The cytotoxicity of BJOE was tested in different cells in vitro, and a Förster resonance energy transfer system was also constructed to predict the release behavior of BJOE in vivo. Then, a B16 melanoma mouse model was used to explore the combination of BJOE and anti-mouse PD-1 antibody therapy. In addition, mass cytometry was used to test the impact of both drugs on the TME. RESULTS Out data revealed that BJOE did not directly kill tumor cells in vitro. However, BJOE was mainly released at the tumor site, converting an immunosuppressive TME into an immune-activated state, and its combination with anti-PD-1 therapy significantly inhibited the growth of melanoma and prolonged the survival time of the mice due to an increase in cytotoxic T lymph (CD8+ T) and helper/inducible T lymph (CD4+ T) cells in lymph nodes and tumors. CONCLUSIONS Our work explored the anti-tumor mechanism of commercial BJOE and the regulation of cytokines by BJOE when it was combined with anti-PD-1 therapy in vivo. The combination of these therapies could increase the numbers of CD4+ T-cells, CD8+ T-cells, and effective natural killer cells and the ratio of MI/M2 macrophages in tumor tissues, promoting inflammatory activity and enhancing the anti-tumor effect. This study provides a theoretical basis for advancing the modern development of traditional Chinese medicine preparations and stands as a reference for clinically improving the efficacy of PD-1 antibodies.
Collapse
Affiliation(s)
- Jun Meng
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510641, China
| | - Zhixin Yu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, United States
| | - Hongying Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Xin Yu
- College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, No. 26 Huatuo Dajie, Benxi, Liaoning 117004, China
| | - Mengshi Jiang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Xin-An Zeng
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510641, China.
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
107
|
Viano ME, Baez NS, Savid-Frontera C, Lidon NL, Hodge DL, Herbelin A, Gombert JM, Barbarin A, Rodriguez-Galan MC. Virtual Memory CD8 + T Cells: Origin and Beyond. J Interferon Cytokine Res 2022; 42:624-642. [PMID: 36083273 PMCID: PMC9835308 DOI: 10.1089/jir.2022.0053] [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: 03/09/2022] [Accepted: 04/19/2022] [Indexed: 01/21/2023] Open
Abstract
The presence of CD8+ T cells with a memory phenotype in nonimmunized mice has been noted for decades, but it was not until about 2 decades ago that they began to be studied in greater depth. Currently called virtual memory CD8+ T cells, they consist of a heterogeneous group of cells with memory characteristics, without any previous contact with their specific antigens. These cells were identified in mice, but a few years ago, a cell type with characteristics equivalent to the murine ones was described in healthy humans. In this review, we address the different aspects of its biology mainly developed in murine models and what is currently known about its cellular equivalent in humans.
Collapse
Affiliation(s)
- Maria Estefania Viano
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Natalia Soledad Baez
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Constanza Savid-Frontera
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Nicolás Leonel Lidon
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | | | - André Herbelin
- Inserm U1313, Poitiers, France
- Université de Poitiers, Poitiers, France
| | - Jean-Marc Gombert
- Inserm U1313, Poitiers, France
- Université de Poitiers, Poitiers, France
- Service d'Immunologie et Inflammation, CHU de Poitiers, Poitiers, France
| | - Alice Barbarin
- Inserm U1313, Poitiers, France
- CHU de Poitiers, Poitiers, France
| | | |
Collapse
|
108
|
Sjaastad FV, Huggins MA, Lucas ED, Skon-Hegg C, Swanson W, Martin MD, Salgado OC, Xu J, Pierson M, Dileepan T, Kucaba TA, Hamilton SE, Griffith TS. Reduced T Cell Priming in Microbially Experienced "Dirty" Mice Results from Limited IL-27 Production by XCR1+ Dendritic Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2149-2159. [PMID: 36426978 PMCID: PMC10065988 DOI: 10.4049/jimmunol.2200324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 09/28/2022] [Indexed: 01/04/2023]
Abstract
Successful vaccination strategies offer the potential for lifelong immunity against infectious diseases and cancer. There has been increased attention regarding the limited translation of some preclinical findings generated using specific pathogen-free (SPF) laboratory mice to humans. One potential reason for the difference between preclinical and clinical findings lies in maturation status of the immune system at the time of challenge. In this study, we used a "dirty" mouse model, where SPF laboratory mice were cohoused (CoH) with pet store mice to permit microbe transfer and immune system maturation, to investigate the priming of a naive T cell response after vaccination with a peptide subunit mixed with polyinosinic-polycytidylic acid and agonistic anti-CD40 mAb. Although this vaccination platform induced robust antitumor immunity in SPF mice, it failed to do so in microbially experienced CoH mice. Subsequent investigation revealed that despite similar numbers of Ag-specific naive CD4 and CD8 T cell precursors, the expansion, differentiation, and recall responses of these CD4 and CD8 T cell populations in CoH mice were significantly reduced compared with SPF mice after vaccination. Evaluation of the dendritic cell compartment revealed reduced IL-27p28 expression by XCR1+ dendritic cells from CoH mice after vaccination, correlating with reduced T cell expansion. Importantly, administration of recombinant IL-27:EBI3 complex to CoH mice shortly after vaccination significantly boosted Ag-specific CD8 and CD4 T cell expansion, further implicating the defect to be T cell extrinsic. Collectively, our data show the potential limitation of exclusive use of SPF mice when testing vaccine efficacy.
Collapse
Affiliation(s)
- Frances V Sjaastad
- Department of Urology, University of Minnesota, Minneapolis, MN
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN
| | - Matthew A Huggins
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Erin D Lucas
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Cara Skon-Hegg
- Department of Urology, University of Minnesota, Minneapolis, MN
| | - Whitney Swanson
- Department of Urology, University of Minnesota, Minneapolis, MN
| | | | - Oscar C Salgado
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Julie Xu
- Department of Urology, University of Minnesota, Minneapolis, MN
| | - Mark Pierson
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Thamotharampillai Dileepan
- Center for Immunology, University of Minnesota, Minneapolis, MN
- Department of Microbiology & Immunology, University of Minnesota, Minneapolis, MN
| | - Tamara A Kucaba
- Department of Urology, University of Minnesota, Minneapolis, MN
| | - Sara E Hamilton
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
- Center for Immunology, University of Minnesota, Minneapolis, MN
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN; and
| | - Thomas S Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN
- Center for Immunology, University of Minnesota, Minneapolis, MN
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN; and
- Minneapolis VA Health Care System, Minneapolis, MN
| |
Collapse
|
109
|
Doan Ngoc TM, Tilly G, Danger R, Bonizec O, Masset C, Guérif P, Bruneau S, Glemain A, Harb J, Cadoux M, Vivet A, Mai HL, Garcia A, Laplaud D, Liblau R, Giral M, Blandin S, Feyeux M, Dubreuil L, Pecqueur C, Cyr M, Ni W, Brouard S, Degauque N. Effector Memory-Expressing CD45RA (TEMRA) CD8 + T Cells from Kidney Transplant Recipients Exhibit Enhanced Purinergic P2X4 Receptor-Dependent Proinflammatory and Migratory Responses. J Am Soc Nephrol 2022; 33:2211-2231. [PMID: 36280286 PMCID: PMC9731633 DOI: 10.1681/asn.2022030286] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 08/22/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The mechanisms regulating CD8+ T cell migration to nonlymphoid tissue during inflammation have not been fully elucidated, and the migratory properties of effector memory CD8+ T cells that re-express CD45RA (TEMRA CD8+ T cells) remain unclear, despite their roles in autoimmune diseases and allotransplant rejection. METHODS We used single-cell proteomic profiling and functional testing of CD8+ T cell subsets to characterize their effector functions and migratory properties in healthy volunteers and kidney transplant recipients with stable or humoral rejection. RESULTS We showed that humoral rejection of a kidney allograft is associated with an accumulation of cytolytic TEMRA CD8+ T cells in blood and kidney graft biopsies. TEMRA CD8+ T cells from kidney transplant recipients exhibited enhanced migratory properties compared with effector memory (EM) CD8+ T cells, with enhanced adhesion to activated endothelium and transmigration in response to the chemokine CXCL12. CXCL12 directly triggers a purinergic P2×4 receptor-dependent proinflammatory response of TEMRA CD8+ T cells from transplant recipients. The stimulation with IL-15 promotes the CXCL12-induced migration of TEMRA and EM CD8+ T cells and promotes the generation of functional PSGL1, which interacts with the cell adhesion molecule P-selectin and adhesion of these cells to activated endothelium. Although disruption of the interaction between functional PSGL1 and P-selectin prevents the adhesion and transmigration of both TEMRA and EM CD8+ T cells, targeting VLA-4 or LFA-1 (integrins involved in T cell migration) specifically inhibited the migration of TEMRA CD8+ T cells from kidney transplant recipients. CONCLUSIONS Our findings highlight the active role of TEMRA CD8+ T cells in humoral transplant rejection and suggest that kidney transplant recipients may benefit from therapeutics targeting these cells.
Collapse
Affiliation(s)
- Tra-My Doan Ngoc
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Gaëlle Tilly
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Richard Danger
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, Nantes Université, Institut de Transplantation Urologie Néphrologie, Nantes, France
| | - Orianne Bonizec
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Christophe Masset
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, Nantes Université, Institut de Transplantation Urologie Néphrologie, Nantes, France
| | - Pierrick Guérif
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, Nantes Université, Institut de Transplantation Urologie Néphrologie, Nantes, France
| | - Sarah Bruneau
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Alexandre Glemain
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Jean Harb
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, Nantes Université, Institut de Transplantation Urologie Néphrologie, Nantes, France
| | - Marion Cadoux
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Anaïs Vivet
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Hoa Le Mai
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, Nantes Université, Institut de Transplantation Urologie Néphrologie, Nantes, France
| | - Alexandra Garcia
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - David Laplaud
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Roland Liblau
- CNRS, Institut National de la Santé et de la Recherche Médicale, UPS, Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, Toulouse, France
- Department of Immunology, Toulouse University Hospital, Toulouse, France
| | - Magali Giral
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, Nantes Université, Institut de Transplantation Urologie Néphrologie, Nantes, France
| | - Stéphanie Blandin
- CHU Nantes, CNRS, Institut National de la Santé et de la Recherche Médicale, BioCore, US16, SFR Bonamy, Nantes Université, Nantes, France
| | - Magalie Feyeux
- CHU Nantes, CNRS, Institut National de la Santé et de la Recherche Médicale, BioCore, US16, SFR Bonamy, Nantes Université, Nantes, France
| | | | - Claire Pecqueur
- Université d’Angers, Institut National de la Santé et de la Recherche Médicale, CNRS, CRCI2NA, Nantes Université, Nantes, France
| | - Matthew Cyr
- IsoPlexis Corporation, Branford, Connecticut
| | - Weiming Ni
- IsoPlexis Corporation, Branford, Connecticut
| | - Sophie Brouard
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, Nantes Université, Institut de Transplantation Urologie Néphrologie, Nantes, France
| | - Nicolas Degauque
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| |
Collapse
|
110
|
Intranasal Vaccination with rePcrV Protects against Pseudomonas aeruginosa and Generates Lung Tissue-Resident Memory T Cells. J Immunol Res 2022; 2022:1403788. [DOI: 10.1155/2022/1403788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/09/2022] [Accepted: 11/11/2022] [Indexed: 11/28/2022] Open
Abstract
Tissue-resident memory T (TRM) cells are immune sentinels that bear a key role in the local immune system and rapidly respond to infection. Our previous studies showed that mucosal immunization via intranasal pathways was more effective than intramuscular route. However, the mechanism of enhanced protective immunity remains unclear. Here, we formulated a Pseudomonas aeruginosa vaccine composed of type III secretion protein PcrV from P. aeruginosa and curdlan adjuvant and then administered by the intranasal route. Flow cytometry and immunofluorescence staining showed that the ratio of CD44+CD62L-CD69+CD4+ TRM cells induced by this vaccine was significantly increased, and IL-17A production was notably enhanced. Further analysis revealed that vaccinated mice can protect against the P. aeruginosa challenge even after administration with FTY720 treatment. What is more, our results showed that CD4+ TRM might be involved in the recruitment of neutrophils and provided partial protection against Pseudomonas aeruginosa. Taken together, these data demonstrated that CD4+ TRM cells were elicited in lung tissues after immunization with rePcrV and contributed to protective immunity. Furthermore, it provided novel strategies for the development of vaccines for P. aeruginosa and other respiratory-targeted vaccines.
Collapse
|
111
|
Shenoy AT, De Ana CL, Barker KA, Arafa EI, Martin IM, Mizgerd JP, Belkina AC. CPHEN-011: Comprehensive phenotyping of murine lung resident lymphocytes after recovery from pneumococcal pneumonia. Cytometry A 2022; 101:892-902. [PMID: 34854229 PMCID: PMC9160214 DOI: 10.1002/cyto.a.24522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 01/27/2023]
Abstract
Recovery from pneumococcal (Spn) pneumonia induces development of tissue resident memory CD4+ TRM cells, BRM cells, and antibody secreting plasma cells in experienced lungs. These tissue resident lymphocytes confer protection against subsequent lethal challenge by serotype mismatched Spn (termed as heterotypic immunity). While traditional flow cytometry and gating strategies support premeditated identification of cells using a limited set of markers, discovery of novel tissue resident lymphocytes necessitates stable platforms that can handle larger sets of phenotypic markers and lends itself to unbiased clustering approaches. In this report, we leverage the power of full spectrum flow cytometry (FSFC) to develop a comprehensive panel of phenotypic markers that allows identification of multiple subsets of tissue resident lymphocytes in Spn-experienced murine lungs. Using Phenograph algorithm on this multidimensional data, we identify unforeseen heterogeneity in lung resident adaptive immune landscape which includes unexpected subsets of TRM and BRM cells. Further, using conventional gating strategy informed by our unsupervised clustering data, we confirm their presence exquisitely in Spn-experienced lungs as potentially relevant to heterotypic immunity and define CD73 as a highly expressed marker on TRM cells. Thus, our study emphasizes the utility of FSFC for confirmatory and discovery studies relating to tissue resident adaptive immunity.
Collapse
Affiliation(s)
- Anukul T. Shenoy
- Pulmonary Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Carolina Lyon De Ana
- Pulmonary Center, Boston University School of Medicine, Boston, MA 02118, USA
- Dept. of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Kimberly A. Barker
- Pulmonary Center, Boston University School of Medicine, Boston, MA 02118, USA
- Dept. of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Emad I. Arafa
- Pulmonary Center, Boston University School of Medicine, Boston, MA 02118, USA
- Dept. of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Ian M.C. Martin
- Pulmonary Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Joseph P. Mizgerd
- Pulmonary Center, Boston University School of Medicine, Boston, MA 02118, USA
- Dept. of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
- Dept. of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- Dept. of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Anna C. Belkina
- Pulmonary Center, Boston University School of Medicine, Boston, MA 02118, USA
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, 02118, USA
- Dept. of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
112
|
Visvabharathy L, Hanson BA, Orban ZS, Lim PH, Palacio NM, Jimenez M, Clark JR, Graham EL, Liotta EM, Tachas G, Penaloza-MacMaster P, Koralnik IJ. T cell responses to SARS-CoV-2 in people with and without neurologic symptoms of long COVID. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2021.08.08.21261763. [PMID: 34401886 PMCID: PMC8366804 DOI: 10.1101/2021.08.08.21261763] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Many people experiencing long COVID syndrome, or post-acute sequelae of SARS-CoV-2 infection (PASC), suffer from debilitating neurologic symptoms (Neuro-PASC). However, whether virus-specific adaptive immunity is affected in Neuro-PASC patients remains poorly understood. We report that Neuro-PASC patients exhibit distinct immunological signatures composed of elevated humoral and cellular responses toward SARS-CoV-2 Nucleocapsid protein at an average of 6 months post-infection compared to healthy COVID convalescents. Neuro-PASC patients also had enhanced virus-specific production of IL-6 from and diminished activation of CD8+ T cells. Furthermore, the severity of cognitive deficits or quality of life disturbances in Neuro-PASC patients were associated with a reduced diversity of effector molecule expression in T cells but elevated IFN-γ production to the C-terminal domain of Nucleocapsid protein. Proteomics analysis showed enhanced plasma immunoregulatory proteins and reduced pro-inflammatory and antiviral response proteins in Neuro-PASC patients compared with healthy COVID convalescents, which were also correlated with worse neurocognitive dysfunction. These data provide new insight into the pathogenesis of long COVID syndrome and a framework for the rational design of predictive biomarkers and therapeutic interventions.
Collapse
Affiliation(s)
- Lavanya Visvabharathy
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago IL 60611 USA
| | - Barbara A. Hanson
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago IL 60611 USA
| | - Zachary S. Orban
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago IL 60611 USA
| | - Patrick H. Lim
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago IL 60611 USA
| | - Nicole M. Palacio
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago IL 60611 USA
| | - Millenia Jimenez
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago IL 60611 USA
| | - Jeffrey R. Clark
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago IL 60611 USA
| | - Edith L. Graham
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago IL 60611 USA
| | - Eric M. Liotta
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago IL 60611 USA
| | - George Tachas
- Director, Drug Discovery & Patents, Antisense Therapeutics Ltd., Melbourne, Australia
| | - Pablo Penaloza-MacMaster
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago IL 60611 USA
| | - Igor J. Koralnik
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago IL 60611 USA
| |
Collapse
|
113
|
Radziszewska A, Moulder Z, Jury EC, Ciurtin C. CD8 + T Cell Phenotype and Function in Childhood and Adult-Onset Connective Tissue Disease. Int J Mol Sci 2022; 23:11431. [PMID: 36232733 PMCID: PMC9569696 DOI: 10.3390/ijms231911431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/14/2022] [Accepted: 09/19/2022] [Indexed: 11/21/2022] Open
Abstract
CD8+ T cells are cytotoxic lymphocytes that destroy pathogen infected and malignant cells through release of cytolytic molecules and proinflammatory cytokines. Although the role of CD8+ T cells in connective tissue diseases (CTDs) has not been explored as thoroughly as that of other immune cells, research focusing on this key component of the immune system has recently gained momentum. Aberrations in cytotoxic cell function may have implications in triggering autoimmunity and may promote tissue damage leading to exacerbation of disease. In this comprehensive review of current literature, we examine the role of CD8+ T cells in systemic lupus erythematosus, Sjögren's syndrome, systemic sclerosis, polymyositis, and dermatomyositis with specific focus on comparing what is known about CD8+ T cell peripheral blood phenotypes, CD8+ T cell function, and CD8+ T cell organ-specific profiles in adult and juvenile forms of these disorders. Although, the precise role of CD8+ T cells in the initiation of autoimmunity and disease progression remains to be elucidated, increasing evidence indicates that CD8+ T cells are emerging as an attractive target for therapy in CTDs.
Collapse
Affiliation(s)
- Anna Radziszewska
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH), Great Ormond Street Hospital (GOSH), London WC1E 6JF, UK
- Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK
| | - Zachary Moulder
- University College London Medical School, University College London, London WC1E 6DE, UK
| | - Elizabeth C. Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH), Great Ormond Street Hospital (GOSH), London WC1E 6JF, UK
- Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK
| |
Collapse
|
114
|
Jiang ZF, Wu W, Hu HB, Li ZY, Zhong M, Zhang L. P2X7 receptor as the regulator of T-cell function in intestinal barrier disruption. World J Gastroenterol 2022; 28:5265-5279. [PMID: 36185635 PMCID: PMC9521516 DOI: 10.3748/wjg.v28.i36.5265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 06/20/2022] [Accepted: 09/01/2022] [Indexed: 02/06/2023] Open
Abstract
The intestinal mucosa is a highly compartmentalized structure that forms a direct barrier between the host intestine and the environment, and its dysfunction could result in a serious disease. As T cells, which are important components of the mucosal immune system, interact with gut microbiota and maintain intestinal homeostasis, they may be involved in the process of intestinal barrier dysfunction. P2X7 receptor (P2X7R), a member of the P2X receptors family, mediates the effects of extracellular adenosine triphosphate and is expressed by most innate or adaptive immune cells, including T cells. Current evidence has demonstrated that P2X7R is involved in inflammation and mediates the survival and differentiation of T lymphocytes, indicating its potential role in the regulation of T cell function. In this review, we summarize the available research about the regulatory role and mechanism of P2X7R on the intestinal mucosa-derived T cells in the setting of intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Zhi-Feng Jiang
- Center of Emergency & Intensive Care Unit, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Wei Wu
- Department of Critical Care Medicine, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Han-Bing Hu
- Center of Emergency & Intensive Care Unit, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Zheng-Yang Li
- Department of Gastroenterology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Ming Zhong
- Department of Critical Care Medicine, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Lin Zhang
- Center of Emergency & Intensive Care Unit, Jinshan Hospital of Fudan University, Shanghai 201508, China
| |
Collapse
|
115
|
Lobognon VD, Alard JE. Could AMPs and B-cells be the missing link in understanding periodontitis? Front Immunol 2022; 13:887147. [PMID: 36211356 PMCID: PMC9532695 DOI: 10.3389/fimmu.2022.887147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 08/01/2022] [Indexed: 12/29/2022] Open
Abstract
Periodontal diseases are common inflammatory conditions characterized by bone loss in response to simultaneous bacterial aggression and host defenses. The etiology of such diseases is still not completely understood, however. It has been shown that specific pathogens involved in the build-up of dysbiotic biofilms participate actively in the establishment of periodontitis. This multifactorial pathology also depends on environmental factors and host characteristics, especially defenses. The immune response to the pathogens seems to be critical in preventing the disease from starting but also contributes to tissue damage. It is known that small molecules known as antimicrobial peptides (AMPs) are key actors in the innate immune response. They not only target microbes, but also act as immuno-modulators. They can help to recruit or activate cells such as neutrophils, monocytes, dendritic cells, or lymphocytes. AMPs have already been described in the periodontium, and their expression seems to be connected to disease activity. Alpha and beta defensins and LL37 are the AMPs most frequently linked to periodontitis. Additionally, leukocyte infiltrates, especially B-cells, have also been linked to the severity of periodontitis. Indeed, the particular subpopulations of B-cells in these infiltrates have been linked to inflammation and bone resorption. A link between B-cells and AMP could be relevant to understanding B-cells' action. Some AMP receptors, such as chemokines receptors, toll-like receptors, or purinergic receptors, have been shown to be expressed by B-cells. Consequently, the action of AMPs on B-cell subpopulations could participate to B-cell recruitment, their differentiation, and their implication in both periodontal defense and destruction.
Collapse
Affiliation(s)
- Vanessa Dominique Lobognon
- B lymphocytes, Autoimmunity and Immunotherapies (LBAI), Mixed Research Unit (UMR)1227 INSERM, University of Brest, Brest, France
| | - Jean-Eric Alard
- B lymphocytes, Autoimmunity and Immunotherapies (LBAI), Mixed Research Unit (UMR)1227 INSERM, University of Brest, Brest, France,Service d’Odontologie, University Hospital (CHU) de Brest, Brest, France,*Correspondence: Jean-Eric Alard,
| |
Collapse
|
116
|
Papait A, Silini AR, Gazouli M, Malvicini R, Muraca M, O’Driscoll L, Pacienza N, Toh WS, Yannarelli G, Ponsaerts P, Parolini O, Eissner G, Pozzobon M, Lim SK, Giebel B. Perinatal derivatives: How to best validate their immunomodulatory functions. Front Bioeng Biotechnol 2022; 10:981061. [PMID: 36185431 PMCID: PMC9518643 DOI: 10.3389/fbioe.2022.981061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/12/2022] [Indexed: 11/27/2022] Open
Abstract
Perinatal tissues, mainly the placenta and umbilical cord, contain a variety of different somatic stem and progenitor cell types, including those of the hematopoietic system, multipotent mesenchymal stromal cells (MSCs), epithelial cells and amnion epithelial cells. Several of these perinatal derivatives (PnDs), as well as their secreted products, have been reported to exert immunomodulatory therapeutic and regenerative functions in a variety of pre-clinical disease models. Following experience with MSCs and their extracellular vesicle (EV) products, successful clinical translation of PnDs will require robust functional assays that are predictive for the relevant therapeutic potency. Using the examples of T cell and monocyte/macrophage assays, we here discuss several assay relevant parameters for assessing the immunomodulatory activities of PnDs. Furthermore, we highlight the need to correlate the in vitro assay results with preclinical or clinical outcomes in order to ensure valid predictions about the in vivo potency of therapeutic PnD cells/products in individual disease settings.
Collapse
Affiliation(s)
- Andrea Papait
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| | - Antonietta Rosa Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Maria Gazouli
- Department of Basic Medical Sciences, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ricardo Malvicini
- Department of Women and Children Health, University of Padova, Padova, Italy
- Laboratorio de Regulación Génica y Células Madre, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMeTTyB), Universidad Favaloro-CONICET, Buenos Aires, Argentina
| | - Maurizio Muraca
- Department of Women and Children Health, University of Padova, Padova, Italy
| | - Lorraine O’Driscoll
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
- Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity St. James’s Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - Natalia Pacienza
- Laboratorio de Regulación Génica y Células Madre, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMeTTyB), Universidad Favaloro-CONICET, Buenos Aires, Argentina
| | - Wei Seong Toh
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Gustavo Yannarelli
- Laboratorio de Regulación Génica y Células Madre, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMeTTyB), Universidad Favaloro-CONICET, Buenos Aires, Argentina
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| | - Günther Eissner
- Systems Biology Ireland, School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Michela Pozzobon
- Department of Women and Children Health, University of Padova, Padova, Italy
| | - Sai Kiang Lim
- Institute of Medical Biology and Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
117
|
Pani F, Caria P, Yasuda Y, Makoto M, Mariotti S, Leenhardt L, Roshanmehr S, Caturegli P, Buffet C. The Immune Landscape of Papillary Thyroid Cancer in the Context of Autoimmune Thyroiditis. Cancers (Basel) 2022; 14:cancers14174287. [PMID: 36077831 PMCID: PMC9454449 DOI: 10.3390/cancers14174287] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 12/26/2022] Open
Abstract
Simple Summary The association between papillary thyroid cancer and Hashimoto’s thyroiditis went through a long-standing human debate recently elucidated by the establishment of a novel mouse model. Papillary thyroid carcinoma is an excellent model for studying the tumor immune microenvironment because it is naturally accompanied by immune cells, making it a good candidate for the treatment with immune checkpoint inhibitors. Abstract Papillary thyroid cancer (PTC) often co-occurs with Hashimoto’s thyroiditis, an association that has long been reported in clinical studies, remaining controversial. Experimental evidence has recently shown that pre-existing thyroiditis has a beneficial effect on PTC growth and progression by a distinctive expansion of effector memory CD8 T cells. Although the link between inflammation and PTC might involve different components of the immune system, a deep characterization of them which includes T cells, B cells and tertiary lymphoid structures, Mye-loid cells, Neutrophils, NK cells and dendritic cells will be desirable. The present review article considers the role of the adaptive and innate immune response surrounding PTC in the context of Hashimoto’s thyroiditis. This review will focus on the current knowledge by in vivo and in vitro studies specifically performed on animals’ models; thyroid cancer cells and human samples including (i) the dual role of tumor-infiltrating lymphocytes; (ii) the emerging role of B cells and tertiary lymphoid structures; (iii) the role of myeloid cells, dendritic cells, and natural killer cells; (iv) the current knowledge of the molecular biomarkers implicated in the complex link between thyroiditis and PTC and the potential implication of cancer immunotherapy in PTC patients in the context of thyroiditis.
Collapse
Affiliation(s)
- Fabiana Pani
- Service des Pathologies Thyroïdiennes et Tumeurs Endocrines, AP-HP, Hôpital Pitié-Salpêtrière, Sorbonne Université, GRC n°16, GRC Tumeurs Thyroïdiennes, 75013 Paris, France
- Correspondence: or
| | - Paola Caria
- Department of Biomedical Sciences, Biochemistry, Biology and Genetics Unit, University of Cagliari, Cittadella Universitaria di Monserrato, SP 8, Km 0.700, Monserrato, 09042 Cagliari, Italy
| | - Yoshinori Yasuda
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Miyara Makoto
- Inserm, Centre d’Immunologie et des Maladies Infectieuses-Paris (CIMI-PARIS), AP-HP Hôpital Pitié-Salpêtrière, Sorbonne Université, 75013 Paris, France
| | - Stefano Mariotti
- Department of Medical Sciences and Public Health, Endocrinology Unit, University of Cagliari, Monserrato, 09042 Cagliari, Italy
| | - Laurence Leenhardt
- Service des Pathologies Thyroïdiennes et Tumeurs Endocrines, AP-HP, Hôpital Pitié-Salpêtrière, Sorbonne Université, GRC n°16, GRC Tumeurs Thyroïdiennes, 75013 Paris, France
| | - Solmaz Roshanmehr
- Division of Immunology, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Patrizio Caturegli
- Division of Immunology, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Camille Buffet
- Service des Pathologies Thyroïdiennes et Tumeurs Endocrines, AP-HP, Hôpital Pitié-Salpêtrière, Sorbonne Université, GRC n°16, GRC Tumeurs Thyroïdiennes, 75013 Paris, France
| |
Collapse
|
118
|
Zhang M, Li N, He Y, Shi T, Jie Z. Pulmonary resident memory T cells in respiratory virus infection and their inspiration on therapeutic strategies. Front Immunol 2022; 13:943331. [PMID: 36032142 PMCID: PMC9412965 DOI: 10.3389/fimmu.2022.943331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/26/2022] [Indexed: 11/25/2022] Open
Abstract
The immune system generates memory cells on infection with a virus for the first time. These memory cells play an essential role in protection against reinfection. Tissue-resident memory T (TRM) cells can be generated in situ once attacked by pathogens. TRM cells dominate the defense mechanism during early stages of reinfection and have gradually become one of the most popular focuses in recent years. Here, we mainly reviewed the development and regulation of various TRM cell signaling pathways in the respiratory tract. Moreover, we explored the protective roles of TRM cells in immune response against various respiratory viruses, such as Respiratory Syncytial Virus (RSV) and influenza. The complex roles of TRM cells against SARS-CoV-2 infection are also discussed. Current evidence supports the therapeutic strategies targeting TRM cells, providing more possibilities for treatment. Rational utilization of TRM cells for therapeutics is vital for defense against respiratory viruses.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Na Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Yanchao He
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Tianyun Shi
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Zhijun Jie
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
- Center of Community-Based Health Research, Fudan University, Shanghai, China
- *Correspondence: Zhijun Jie,
| |
Collapse
|
119
|
Chandiran K, Suarez-Ramirez JE, Hu Y, Jellison ER, Ugur Z, Low JS, McDonald B, Kaech SM, Cauley LS. SMAD4 and TGFβ are architects of inverse genetic programs during fate-determination of antiviral CTLs. eLife 2022; 11:76457. [PMID: 35942952 PMCID: PMC9402230 DOI: 10.7554/elife.76457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 08/05/2022] [Indexed: 11/25/2022] Open
Abstract
Transforming growth factor β (TGFβ) is an important differentiation factor for cytotoxic T lymphocytes (CTLs) and alters the expression levels of several of homing receptors during infection. SMAD4 is part of the canonical signaling network used by members of the transforming growth factor family. For this study, genetically modified mice were used to determine how SMAD4 and TGFβ receptor II (TGFβRII) participate in transcriptional programming of pathogen-specific CTLs. We show that these molecules are essential components of opposing signaling mechanisms, and cooperatively regulate a collection of genes that determine whether specialized populations of pathogen-specific CTLs circulate around the body, or settle in peripheral tissues. TGFβ uses a canonical SMAD-dependent signaling pathway to downregulate Eomesodermin (EOMES), KLRG1, and CD62L, while CD103 is induced. Conversely, in vivo and in vitro data show that EOMES, KLRG1, CX3CR1, and CD62L are positively regulated via SMAD4, while CD103 and Hobit are downregulated. Intravascular staining also shows that signaling via SMAD4 promotes formation of long-lived terminally differentiated CTLs that localize in the vasculature. Our data show that inflammatory molecules play a key role in lineage determination of pathogen-specific CTLs, and use SMAD-dependent signaling to alter the expression levels of multiple homing receptors and transcription factors with known functions during memory formation.
Collapse
Affiliation(s)
- Karthik Chandiran
- Department of Immunology, University of Connecticut Health Center, Farmington, United States
| | - Jenny E Suarez-Ramirez
- Department of Immunology, University of Connecticut Health Center, Farmington, United States
| | - Yinghong Hu
- Department of Microbiology and Immunology, Emory University, Atlanta, United States
| | - Evan R Jellison
- Department of Immunology, University of Connecticut Health Center, Farmington, United States
| | - Zenep Ugur
- Department of Immunology, University of Connecticut Health Center, Farmington, United States
| | - Jun-Siong Low
- Department of Immunobiology, Yale University, Bellinzona, Switzerland
| | - Bryan McDonald
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, San Diego, United States
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, San Diego, United States
| | - Linda S Cauley
- Department of Immunology, University of Connecticut Health Center, Farmington, United States
| |
Collapse
|
120
|
Álvarez B, Revilla C, Moreno S, Jiménez-Marín Á, Ramos E, Martínez de la Riva P, Poderoso T, Garrido JJ, Ezquerra Á, Domínguez J. CD9 expression in porcine blood CD4 + T cells delineates two subsets with phenotypic characteristics of central and effector memory cells. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 133:104431. [PMID: 35526640 DOI: 10.1016/j.dci.2022.104431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/20/2022] [Accepted: 05/02/2022] [Indexed: 06/14/2023]
Abstract
In this report, we describe the characterization of a new monoclonal antibody, named 4H5CR4, against porcine CD9. Its use in combination with antibodies to CD4, CD8α, and 2E3 allows to distinguish at least five main CD4+ T cell subsets. Analysis on these subsets of CD45RA, CD27, CD29, CD95, CCR7, and SLA-DR markers depicts a progressive model of CD4+ T cell development. CD4+ 2E3+ CD8α- CD9- cells are the least differentiated population of naïve cells, whereas the CD4+ 2E3- CD8α+CD9+ and CD4+ 2E3- CD8α+ CD9- cells display phenotypic features of central and effector memory T helper cells, respectively. The latter subsets were able to produce IFN-γ after polyclonal activation with PMA/Ionomycin; however, in vitro virus-specific IFN-γ production of PBMCs collected at 38-44 days after pseudorabies virus vaccination was dominated by cells with a CD9+ phenotype. Therefore, CD9 appears to be a useful marker to investigate CD4+ T cell heterogeneity in swine.
Collapse
Affiliation(s)
- Belén Álvarez
- Departamento de Biotecnología, Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, 28040, Spain
| | - Concepción Revilla
- Departamento de Biotecnología, Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, 28040, Spain
| | - Sara Moreno
- Departamento de Biotecnología, Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, 28040, Spain
| | - Ángeles Jiménez-Marín
- Grupo de Inmunogenómica y Patogénesis Molecular (IMIBIC), Departamento de Genética, Universidad de Córdoba, Campus Universitario de Rabanales, 14014, Córdoba, Spain
| | - Elena Ramos
- Departamento de Biotecnología, Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, 28040, Spain
| | - Paloma Martínez de la Riva
- Departamento de Biotecnología, Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, 28040, Spain
| | - Teresa Poderoso
- Departamento de Biotecnología, Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, 28040, Spain
| | - Juan J Garrido
- Grupo de Inmunogenómica y Patogénesis Molecular (IMIBIC), Departamento de Genética, Universidad de Córdoba, Campus Universitario de Rabanales, 14014, Córdoba, Spain
| | - Ángel Ezquerra
- Departamento de Biotecnología, Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, 28040, Spain.
| | - Javier Domínguez
- Departamento de Biotecnología, Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, 28040, Spain
| |
Collapse
|
121
|
Cai X, Li H, Wang M, Chu E, Wei N, Lin J, Hu Y, Dai J, Chen A, Zheng H, Zhang Q, Zhong Y, Chang R, Wu S, Xiao Y, Liu C. mTOR Participates in the Formation, Maintenance, and Function of Memory CD8 +T Cells Regulated by Glycometabolism. Biochem Pharmacol 2022; 204:115197. [PMID: 35926651 DOI: 10.1016/j.bcp.2022.115197] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/21/2022] [Accepted: 07/27/2022] [Indexed: 11/02/2022]
Abstract
Memory CD8+T cells participate in the fight against infection and tumorigenesis as well as in autoimmune disease progression because of their efficient and rapid immune response, long-term survival, and continuous differentiation. At each stage of their formation, maintenance, and function, the cell metabolism must be adjusted to match the functional requirements of the specific stage. Notably, enhanced glycolytic metabolism can generate sufficient levels of adenosine triphosphate (ATP) to form memory CD8+T cells, countering the view that glycolysis prevents the formation of memory CD8+T cells. This review focuses on how glycometabolism regulates memory CD8+T cells and highlights the key mechanisms through which the mammalian target of rapamycin (mTOR) signaling pathway affects memory CD8+T cell formation, maintenance, and function by regulating glycometabolism. In addition, different subpopulations of memory CD8+T cells exhibit different metabolic flexibility during their formation, survival, and functional stages, during which the energy metabolism may be critical. These findings which may explain why enhanced glycolytic metabolism can give rise to memory CD8+T cells. Modulating the metabolism of memory CD8+T cells to influence specific cell fates may be useful for disease treatment.
Collapse
Affiliation(s)
- Xuepei Cai
- Department of Orthodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Haokun Li
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Manyi Wang
- Department of Orthodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Edward Chu
- Department of Oncology and Cancer Therapeutics Program, Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ning Wei
- Department of Oncology and Cancer Therapeutics Program, Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jiayu Lin
- Department of Orthodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Yun Hu
- Department of Orthodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Jingtao Dai
- Department of Orthodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Aijie Chen
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Hua Zheng
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qianbing Zhang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yuxia Zhong
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ruoshui Chang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Sha Wu
- Department of Immunology, School of Basic Medical Sciences, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory of Functional Proteomics of Guangdong Province, Guangzhou, China; National Demonstration Center for Experimental Education of Basic Medical Sciences of China, Guangzhou, China.
| | - Yaomu Xiao
- Department of Orthodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China.
| | - Chufeng Liu
- Department of Orthodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
122
|
Sethi GS, Gracias D, Croft M. Contribution of circulatory cells to asthma exacerbations and lung tissue-resident CD4 T cell memory. Front Immunol 2022; 13:951361. [PMID: 35936001 PMCID: PMC9353789 DOI: 10.3389/fimmu.2022.951361] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Tissue-resident memory CD4 T cells (Trm) are thought to be a major contributor to asthma relapse, but the role of circulatory T cells in asthma exacerbations or to maintaining the population of lung Trm cells is not fully understood. Here, we used a house dust mite allergen-based murine model of asthma relapse, and monitored the development of lung effector/Trm phenotype CD44hiCD62LloCD69+ CD4 T cells. To determine the contribution of circulatory cells, mice were treated with FTY720, to block lymphocyte egress from lymph nodes. Inhibiting the primary migration of circulatory cells to the lungs mitigated the accumulation and expansion of allergen-driven Trm phenotype cells, but subsequent allergen challenges still resulted in strong lung inflammation and Trm cell accumulation. This was blocked if FTY720 was also given at the time of allergen re-exposure, showing that new circulatory cells contributed to this lung memory/effector T cell pool at times well after the initial sensitization. However, once lung-localized Trm cells developed at high frequency, circulatory cells were not required to maintain this population following allergen re-encounter, even though circulatory cells still were major contributors to the overall asthmatic lung inflammatory response. Our results suggest that strategies that target the response of circulatory memory T cells and Trm cells together might be required to strongly inhibit T cell reactivity to airborne allergens and to limit exacerbations of asthma and their reoccurrence, but the contribution of circulatory T cells might vary in long-term asthmatics possessing a large stable Trm cell population in the lungs.
Collapse
Affiliation(s)
- Gurupreet S. Sethi
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Donald Gracias
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Michael Croft
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
- *Correspondence: Michael Croft,
| |
Collapse
|
123
|
HSPG2 Mutation Association with Immune Checkpoint Inhibitor Outcome in Melanoma and Non-Small Cell Lung Cancer. Cancers (Basel) 2022; 14:cancers14143495. [PMID: 35884556 PMCID: PMC9315784 DOI: 10.3390/cancers14143495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/11/2022] [Accepted: 07/11/2022] [Indexed: 02/01/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) markedly promote the survival outcome of advanced melanoma and non-small cell lung cancer (NSCLC). Clinically, favorable ICI treatment efficacy is noticed only in a smaller proportion of patients. Heparan sulfate proteoglycan 2 (HSPG2) frequently mutates in both tumors. Herein, we aim to investigate the immunotherapeutic and immunological roles of HSPG2 mutations in melanoma and NSCLC. A total of 631 melanoma samples and 109 NSCLC samples with both somatic mutational profiles and clinical immunotherapy data were curated. In addition, by using The Cancer Genome Atlas data, genomic and immunological traits behind HSPG2 mutations were elucidated. Melanoma patients with HSPG2 mutations had a markedly extended ICI outcome than other patients. An association between HSPG2 mutations and the improved outcome was further confirmed in NSCLC. In addition, an elevated ICI response rate was presented in HSPG2-mutated NSCLC patients (81.8% vs. 29.7%, p = 0.002). Subsequent analyses revealed that HSPG2-mutated patients had a favorable abundance of response immunocytes, an inferior abundance of suppression immunocytes, enhanced mutational burden, and interferon response-relevant signaling pathways. We uncovered that HSPG2 mutations were predictive of a better ICI response and associated with preferable immunogenicity, which may be considered as a genomic determinant to customize biotherapy strategies.
Collapse
|
124
|
Zhang H, Liu J, Yang Z, Zeng L, Wei K, Zhu L, Tang L, Wang D, Zhou Y, Lv J, Zhou N, Tang K, Ma J, Huang B. TCR activation directly stimulates PYGB-dependent glycogenolysis to fuel the early recall response in CD8 + memory T cells. Mol Cell 2022; 82:3077-3088.e6. [PMID: 35738262 DOI: 10.1016/j.molcel.2022.06.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/30/2022] [Accepted: 05/31/2022] [Indexed: 12/12/2022]
Abstract
Glycolysis facilitates the rapid recall response of CD8+ memory T (Tm) cells. However, it remains unclear whether Tm cells uptake exogenous glucose or mobilize endogenous sugar to fuel glycolysis. Here, we show that intracellular glycogen rather than extracellular glucose acts as the major carbon source for the early recall response. Following antigenic stimulation, Tm cells exhibit high glycogen phosphorylase (brain form, PYGB) activity, leading to glycogenolysis and release of glucose-6-phosphate (G6P). Elevated G6P mainly flows to glycolysis but is also partially channeled to the pentose phosphate pathway, which maintains the antioxidant capacity necessary for later recall stages. Mechanistically, TCR signaling directly induces phosphorylation of PYGB by LCK-ZAP70. Functionally, the glycogenolysis-fueled early recall response of CD8+ Tm cells accelerates the clearance of OVA-Listeria monocytogenes in an infected mouse model. Thus, we uncover a specific dependency on glycogen for the initial activation of memory T cells, which may have therapeutic implications for adaptive immunity.
Collapse
Affiliation(s)
- Huafeng Zhang
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jincheng Liu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuoshun Yang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liping Zeng
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Keke Wei
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liyan Zhu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Tang
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dianheng Wang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yabo Zhou
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China
| | - Jiadi Lv
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China
| | - Nannan Zhou
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China
| | - Ke Tang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingwei Ma
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Huang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China; State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China.
| |
Collapse
|
125
|
Koyaman-Nasu R, Wang Y, Hasegawa I, Endo Y, Nakayama T, Kimura MY. The cellular and molecular basis of CD69 function in anti-tumor immunity. Int Immunol 2022; 34:555-561. [PMID: 35689672 DOI: 10.1093/intimm/dxac024] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/09/2022] [Indexed: 12/16/2022] Open
Abstract
Cancer immunotherapy utilizes our immune system to attack cancer cells and is an extremely promising strategy for cancer treatment. Although immune-checkpoint blockade, such as anti-PD-1 antibody (Ab), has demonstrated significant enhancement of anti-tumor immunity and has induced notable clinical outcomes, its response rates remain low, and adverse effects are always a matter of concern; therefore, new targets for cancer immunotherapy are always desired. In this situation, new concepts are needed to fuel the investigation of new target molecules for cancer immunotherapy. We propose that CD69 is one such target molecule. CD69 is known to be an activation marker of leukocytes and is also considered a crucial regulator of various immune responses through its interacting proteins. CD69 promotes T cell retention in lymphoid tissues via sphingosine-1-phosphate receptor 1 (S1P1) internalization and also plays roles in the pathogenesis of inflammatory disorders through interacting with its functional ligands Myl9/12 (myosin light chains 9, 12a and 12b). In anti-tumor immunity, CD69 is known to be expressed on T cells in the tumor microenvironment (TME) and tumor-draining lymph nodes (TDLNs). We revealed that CD69 negatively regulates the effector function of intratumoral T cells and importantly controls the 'exhaustion' of CD8 T cells. In addition, we and others showed that either CD69 deficiency or the administration of anti-CD69 monoclonal antibody enhances anti-tumor immunity. Thus, CD69 is an attractive target for cancer immunotherapy.
Collapse
Affiliation(s)
- Ryo Koyaman-Nasu
- Department of Experimental Immunology, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Yangsong Wang
- Department of Experimental Immunology, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Ichita Hasegawa
- Department of Experimental Immunology, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Yukihiro Endo
- Department of Experimental Immunology, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan.,AMED-CREST, AMED, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Motoko Y Kimura
- Department of Experimental Immunology, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| |
Collapse
|
126
|
He Y, Peng K, Li R, Zhang Z, Pan L, Zhang T, Lin A, Hong R, Nie Z, Guan Q, Jin L. Changes of T lymphocyte subpopulations and their roles in predicting the risk of Parkinson's disease. J Neurol 2022; 269:5368-5381. [PMID: 35608657 PMCID: PMC9467943 DOI: 10.1007/s00415-022-11190-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 12/29/2022]
Abstract
T lymphocytes are involved in the pathogenesis of Parkinson's disease (PD), while the heterogeneity of T-cell subpopulations remains elusive. In this study, we analyzed up to 22 subpopulations of T lymphocytes in 115 PD patients and 60 matched healthy controls (HC) using flow cytometry. We found that PD patients exhibited decreased naïve CD8+ T cells (CD3+ CD8+ CD45RA+ CD45RO-) and increased late-differentiated CD4+ T cells (CD3+ CD4+ CD28- CD27-), compared to HC, which were not affected by anti-parkinsonism medication administration. The proportion of naïve CD8+ T cells in PD patients was positively correlated with their severity of autonomic dysfunction and psychiatric complications, but negatively associated with the severity of rapid eye movement and sleep behavior disorder. The proportion of late-differentiated CD4+ T cells was negatively correlated with the onset age of the disease. We further developed individualized PD risk prediction models with high reliability and accuracy on the base of the T lymphocyte subpopulations. These data suggest that peripheral cellular immunity is disturbed in PD patients, and changes in CD8+ T cells and late-differentiated CD4+ T cells are representative and significant. Therefore, we recommend naïve CD8 + and late-differentiated CD4+ T cells as candidates for multicentric clinical study and pathomechanism study of PD.
Collapse
Affiliation(s)
- Yijing He
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China
| | - Kangwen Peng
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China
| | - Ruoyu Li
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China
| | - Zhuoyu Zhang
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China
| | - Lizhen Pan
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China
| | - Tianyu Zhang
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China
| | - Ao Lin
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China
| | - Ronghua Hong
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China
| | - Zhiyu Nie
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China
| | - Qiang Guan
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China.
| | - Lingjing Jin
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, 200065, Shanghai, People's Republic of China. .,Department of Neurology and Neurological Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, 200092, People's Republic of China. .,Shanghai Clinical Research Center for Aging and Medicine, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
127
|
Loi JK, Alexandre YO, Senthil K, Schienstock D, Sandford S, Devi S, Christo SN, Mackay LK, Chinnery HR, Osborne PB, Downie LE, Sloan EK, Mueller SN. Corneal tissue-resident memory T cells form a unique immune compartment at the ocular surface. Cell Rep 2022; 39:110852. [PMID: 35613584 DOI: 10.1016/j.celrep.2022.110852] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 03/27/2022] [Accepted: 04/29/2022] [Indexed: 11/03/2022] Open
Abstract
The eye is considered immune privileged such that immune responses are dampened to protect vision. As the most anterior compartment of the eye, the cornea is exposed to pathogens and can mount immune responses that recruit effector T cells. However, presence of immune memory in the cornea is not defined. Here, we use intravital 2-photon microscopy to examine T cell responses in the cornea in mice. We show that recruitment of CD8+ T cells in response to ocular virus infection results in the formation of tissue-resident memory T (TRM) cells. Motile corneal TRM cells patrol the cornea and rapidly respond in situ to antigen rechallenge. CD103+ TRM cell generation requires antigen and transforming growth factor β. In vivo imaging in humans also reveals highly motile cells that patrol the healthy cornea. Our study finds that TRM cells form in the cornea where they can provide local protective immunity.
Collapse
Affiliation(s)
- Joon Keit Loi
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Yannick O Alexandre
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Kirthana Senthil
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Dominik Schienstock
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Sarah Sandford
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Sapna Devi
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Susan N Christo
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Laura K Mackay
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Holly R Chinnery
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Peregrine B Osborne
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, VIC, Australia
| | - Laura E Downie
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Erica K Sloan
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia; Division of Surgery, Peter MacCallum Cancer Center, Melbourne, VIC, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
| |
Collapse
|
128
|
Kawasaki T, Ikegawa M, Kawai T. Antigen Presentation in the Lung. Front Immunol 2022; 13:860915. [PMID: 35615351 PMCID: PMC9124800 DOI: 10.3389/fimmu.2022.860915] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/15/2022] [Indexed: 12/28/2022] Open
Abstract
The lungs are constantly exposed to environmental and infectious agents such as dust, viruses, fungi, and bacteria that invade the lungs upon breathing. The lungs are equipped with an immune defense mechanism that involves a wide variety of immunological cells to eliminate these agents. Various types of dendritic cells (DCs) and macrophages (MACs) function as professional antigen-presenting cells (APCs) that engulf pathogens through endocytosis or phagocytosis and degrade proteins derived from them into peptide fragments. During this process, DCs and MACs present the peptides on their major histocompatibility complex class I (MHC-I) or MHC-II protein complex to naïve CD8+ or CD4+ T cells, respectively. In addition to these cells, recent evidence supports that antigen-specific effector and memory T cells are activated by other lung cells such as endothelial cells, epithelial cells, and monocytes through antigen presentation. In this review, we summarize the molecular mechanisms of antigen presentation by APCs in the lungs and their contribution to immune response.
Collapse
Affiliation(s)
| | | | - Taro Kawai
- *Correspondence: Takumi Kawasaki, ; Taro Kawai,
| |
Collapse
|
129
|
Li L, Tang W, Zhang Y, Jia M, Wang L, Li Q, Han Q, Peng X, Xie Y, Wu J, Wang Z, Zhen J, Wang X, Liu M, Sun Y, Zhang C, Yi F. Targeting tissue-resident memory CD8 + T cells in the kidney is a potential therapeutic strategy to ameliorate podocyte injury and glomerulosclerosis. Mol Ther 2022; 30:2746-2759. [PMID: 35514086 PMCID: PMC9372318 DOI: 10.1016/j.ymthe.2022.04.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/21/2022] [Accepted: 04/29/2022] [Indexed: 12/01/2022] Open
Abstract
Although tissue-resident memory T (TRM) cells, a recently identified non-circulating memory T cell population, play a crucial role in mediating local immune responses and protect against pathogens upon local reinfection, the composition, effector function, and specificity of TRM cells in the kidney and their relevance for chronic kidney disease remain unknown. In this study, we found that renal tissue displayed high abundance of tissue-resident lymphocytes and the proportion of CD8+ TRM cells was significantly increased in the kidney from patients and mice with focal segmental glomerulosclerosis (FSGS), diabetic kidney disease (DKD) and lupus nephritis (LN). Mechanistically, IL-15 significantly promoted CD8+ TRM cell formation and activation, thereby promoting podocyte injury and glomerulosclerosis. Interestingly, Sparsentan, the dual angiotensin II (Ang II) receptor and endothelin Type A receptor antagonist, can also reduce TRM cell responses by intervening IL-15 signaling, exploring its new pharmacological functions. Mechanistically, Sparsentan inhibited Ang II or endothelin-1 (ET-1)-mediated IL-15 signaling, thereby further regulating renal CD8+ TRM cell fates. Collectively, our studies provide direct evidence for the pivotal role of renal CD8+ TRM cells in podocyte injury, and further strengthen that targeting TRM cells represents a novel therapeutic strategy for patients with glomerular diseases.
Collapse
Affiliation(s)
- Liang Li
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Wei Tang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Yan Zhang
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Meng Jia
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Limei Wang
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China, 250012
| | - Quanxin Li
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Qingsheng Han
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Xiuping Peng
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Yusheng Xie
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Jichao Wu
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Ziying Wang
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Junhui Zhen
- Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Xiaojie Wang
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Min Liu
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Yu Sun
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China, 430022
| | - Fan Yi
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012.
| |
Collapse
|
130
|
TCF-1: a maverick in T cell development and function. Nat Immunol 2022; 23:671-678. [PMID: 35487986 PMCID: PMC9202512 DOI: 10.1038/s41590-022-01194-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/22/2022] [Indexed: 02/01/2023]
Abstract
The T cell-specific DNA-binding protein TCF-1 is a central regulator of T cell development and function along multiple stages and lineages. Because it interacts with β-catenin, TCF-1 has been classically viewed as a downstream effector of canonical Wnt signaling, although there is strong evidence for β-catenin-independent TCF-1 functions. TCF-1 co-binds accessible regulatory regions containing or lacking its conserved motif and cooperates with other nuclear factors to establish context-dependent epigenetic and transcription programs that are essential for T cell development and for regulating immune responses to infection, autoimmunity and cancer. Although it has mostly been associated with positive regulation of chromatin accessibility and gene expression, TCF-1 has the potential to reduce chromatin accessibility and thereby suppress gene expression. In addition, the binding of TCF-1 bends the DNA and affects the chromatin conformation genome wide. This Review discusses the current understanding of the multiple roles of TCF-1 in T cell development and function and their mechanistic underpinnings.
Collapse
|
131
|
Hitchcock J, Hughes K, Pensa S, Lloyd-Lewis B, Watson CJ. The immune environment of the mammary gland fluctuates during post-lactational regression and correlates with tumour growth rate. Development 2022; 149:275060. [PMID: 35420674 PMCID: PMC9124574 DOI: 10.1242/dev.200162] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 04/04/2022] [Indexed: 01/02/2023]
Abstract
Post-lactational mammary gland regression encompasses extensive programmed cell death and removal of milk-producing epithelial cells, breakdown of extracellular matrix components and redifferentiation of stromal adipocytes. This highly regulated involution process is associated with a transient increased risk of breast cancer in women. Using a syngeneic tumour model, we show that tumour growth is significantly altered depending on the stage of involution at which tumour cells are implanted. Tumour cells injected at day 3 involution grew faster than those in nulliparous mice, whereas tumours initiated at day 6 involution grew significantly slower. These differences in tumour progression correlate with distinct changes in innate immune cells, in particular among F4/80-expressing macrophages and among TCRδ+ unconventional T cells. Breast cancer post-pregnancy risk is exacerbated in older first-time mothers and, in our model, initial tumour growth is moderately faster in aged mice compared with young mice. Our results have implications for breast cancer risk and the use of anti-inflammatory therapeutics for postpartum breast cancers. Summary: Mammary gland involution is associated with dynamic changes in immune cell types and numbers at different stages that correlates with the initial rate of growth of implanted tumour cells.
Collapse
Affiliation(s)
- Jessica Hitchcock
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Katherine Hughes
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK
| | - Sara Pensa
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Bethan Lloyd-Lewis
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
- School of Cellular and Molecular Medicine, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - Christine J. Watson
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| |
Collapse
|
132
|
Chen Q, Li H, Liu Y, Zhao M. Epigenetic Regulation of Immune and Inflammatory Responses in Rheumatoid Arthritis. Front Immunol 2022; 13:881191. [PMID: 35479077 PMCID: PMC9035598 DOI: 10.3389/fimmu.2022.881191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/23/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose Rheumatoid arthritis (RA) is a disease associated with multiple factors. Epigenetics can affect gene expression without altering the DNA sequence. In this study, we aimed to comprehensively analyze epigenetic regulation in RA. Methods Using the Gene Expression Omnibus database, we identified a methylation chip, RNA-sequencing, and miRNA microarray for RA. First, we searched for DNA methylation, genes, and miRNAs associated with RA using differential analysis. Second, we determined the regulatory networks for RA-specific methylation, miRNA, and m6A using cross-analysis. Based on these three regulatory networks, we built a comprehensive epigenetic regulatory network and identified hub genes. Results Using a differential analysis, we identified 16,852 differentially methylated sites, 4877 differentially expressed genes, and 32 differentially expressed miRNAs. The methylation-expression regulatory network was mainly associated with the PI3K-Akt and T-cell receptor signaling pathways. The miRNA expression regulatory network was mainly related to the MAPK and chemokine signaling pathways. M6A regulatory network was mainly associated with the MAPK signaling pathway. Additionally, five hub genes were identified in the epigenetic regulatory network: CHD3, SETD1B, FBXL19, SMARCA4, and SETD1A. Functional analysis revealed that these five genes were associated with immune cells and inflammatory responses. Conclusion We constructed a comprehensive epigenetic network associated with RA and identified core regulatory genes. This study provides a new direction for future research on the epigenetic mechanisms of RA.
Collapse
Affiliation(s)
- Qi Chen
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hao Li
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yusi Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Min Zhao
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
133
|
Chiang EY, Mellman I. TIGIT-CD226-PVR axis: advancing immune checkpoint blockade for cancer immunotherapy. J Immunother Cancer 2022; 10:jitc-2022-004711. [PMID: 35379739 PMCID: PMC8981293 DOI: 10.1136/jitc-2022-004711] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2022] [Indexed: 12/22/2022] Open
Abstract
Recent advances in understanding the roles of immune checkpoints in allowing tumors to circumvent the immune system have led to successful therapeutic strategies that have fundamentally changed oncology practice. Thus far, immunotherapies against only two checkpoint targets have been approved, CTLA-4 and PD-L1/PD-1. Antibody blockade of these targets enhances the function of antitumor T cells at least in part by relieving inhibition of the T cell costimulatory receptor CD28. These successes have stimulated considerable interest in identifying other pathways that may bte targeted alone or together with existing immunotherapies. One such immune checkpoint axis is comprised of members of the PVR/nectin family that includes the inhibitory receptor T cell immunoreceptor with Ig and immunoreceptor tyrosine-based inhibitory domains (TIGIT). Interestingly, TIGIT acts to regulate the activity of a second costimulatory receptor CD226 that works in parallel to CD28. There are currently over two dozen TIGIT-directed blocking antibodies in various phases of clinical development, testament to the promise of modulating this pathway to enhance antitumor immune responses. In this review, we discuss the role of TIGIT as a checkpoint inhibitor, its interplay with the activating counter-receptor CD226, and its status as the next advance in cancer immunotherapy.
Collapse
Affiliation(s)
- Eugene Y Chiang
- Cancer Immunology, Genentech Inc, South San Francisco, California, USA
| | - Ira Mellman
- Cancer Immunology, Genentech Inc, South San Francisco, California, USA
| |
Collapse
|
134
|
Zhang H, Liu S, Li Y, Li J, Ni C, Yang M, Dong J, Wang Z, Qin Z. Dysfunction of S100A4 + effector memory CD8 + T cells aggravates asthma. Eur J Immunol 2022; 52:978-993. [PMID: 35340022 DOI: 10.1002/eji.202149572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 11/09/2022]
Abstract
Progressive loss of effector functions, especially IFN-γ secreting capability, in effector memory CD8+ T (CD8+ TEM ) cells plays a crucial role in asthma worsening. However, the mechanisms of CD8+ TEM cell dysfunction remain elusive. Here, we report that S100A4 drives CD8+ TEM cell dysfunction, impairing their protective memory response and promoting asthma worsening in an ovalbumin (OVA)-induced asthmatic murine model. We find that CD8+ TEM cells contain two subsets based on S100A4 expression. S100A4+ subsets exhibit dysfunctional effector phenotypes with increased proliferative capability, whereas S100A4- subsets retain effector function but are more inclined to apoptosis, giving rise a dysfunctional CD8+ TEM cell pool. Mechanistically, S100A4 upregulation of mitochondrial metabolism results in a decrease of acetyl-CoA levels, which impair the transcription of effector genes, especially ifn-γ, facilitating cell survival, tolerance and memory potential. Our findings thus reveal general insights into how S100A4 CD8+ TEM cells reprogram into dysfunctional and less protective phenotypes to aggravate asthma. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Huilei Zhang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China.,University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Shuangqing Liu
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China.,University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Yanan Li
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jianru Li
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Chen Ni
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ming Yang
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences & Pharmacy, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW 2300, Australia
| | - Jun Dong
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, 10117, Germany
| | - Zhaoqing Wang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhihai Qin
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China.,University of Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
135
|
Anang DC, Ramwadhdoebe TH, Hähnlein JS, van Kuijk B, Smits N, van Lienden KP, Maas M, Gerlag DM, Tak PP, de Vries N, van Baarsen LGM. Increased Frequency of CD4+ Follicular Helper T and CD8+ Follicular T Cells in Human Lymph Node Biopsies during the Earliest Stages of Rheumatoid Arthritis. Cells 2022; 11:cells11071104. [PMID: 35406668 PMCID: PMC8997933 DOI: 10.3390/cells11071104] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/16/2022] [Accepted: 03/22/2022] [Indexed: 01/27/2023] Open
Abstract
Follicular T helper cells (Tfh cells) provide key B-cell help and are essential in germinal center formation and (auto) antibody generation. To gain more insight into their role during the earliest phase of rheumatoid arthritis (RA), we analyzed their frequencies, phenotypes, and cytokine profiles in peripheral blood and lymph node biopsies of healthy controls (HCs), autoantibody-positive individuals at risk for developing RA (RA-risk individuals), and early RA patients. Subsequently, we confirmed their presence in lymph nodes and synovial tissue of RA patients using immunofluorescence microscopy. In the blood, the frequency of Tfh cells did not differ between study groups. In lymphoid and synovial tissues, Tfh cells were localized in B-cell areas, and their frequency correlated with the frequency of CD19+ B cells. Compared to lymphoid tissues of healthy controls, those of RA patients and RA-risk individuals showed more CD19+ B cells, CD4+CXCR5+ follicular helper T cells, and CD8+CXCR5+ follicular T cells. These Tfh cells produced less IL-21 upon ex vivo stimulation. These findings suggest that Tfh cells may present a novel rationale for therapeutic targeting during the preclinical stage of RA to prevent further disease progression.
Collapse
Affiliation(s)
- Dornatien Chuo Anang
- Amsterdam Rheumatology & Immunology Center (ARC), Department of Rheumatology & Clinical Immunology, 1007 MB Amsterdam, The Netherlands; (D.C.A.); (T.H.R.); (J.S.H.); (B.v.K.); (N.S.); (D.M.G.); (P.P.T.); (N.d.V.)
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, 1007 MB Amsterdam, The Netherlands
| | - Tamara H. Ramwadhdoebe
- Amsterdam Rheumatology & Immunology Center (ARC), Department of Rheumatology & Clinical Immunology, 1007 MB Amsterdam, The Netherlands; (D.C.A.); (T.H.R.); (J.S.H.); (B.v.K.); (N.S.); (D.M.G.); (P.P.T.); (N.d.V.)
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, 1007 MB Amsterdam, The Netherlands
| | - Janine S. Hähnlein
- Amsterdam Rheumatology & Immunology Center (ARC), Department of Rheumatology & Clinical Immunology, 1007 MB Amsterdam, The Netherlands; (D.C.A.); (T.H.R.); (J.S.H.); (B.v.K.); (N.S.); (D.M.G.); (P.P.T.); (N.d.V.)
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, 1007 MB Amsterdam, The Netherlands
| | - Bo van Kuijk
- Amsterdam Rheumatology & Immunology Center (ARC), Department of Rheumatology & Clinical Immunology, 1007 MB Amsterdam, The Netherlands; (D.C.A.); (T.H.R.); (J.S.H.); (B.v.K.); (N.S.); (D.M.G.); (P.P.T.); (N.d.V.)
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, 1007 MB Amsterdam, The Netherlands
| | - Noortje Smits
- Amsterdam Rheumatology & Immunology Center (ARC), Department of Rheumatology & Clinical Immunology, 1007 MB Amsterdam, The Netherlands; (D.C.A.); (T.H.R.); (J.S.H.); (B.v.K.); (N.S.); (D.M.G.); (P.P.T.); (N.d.V.)
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, 1007 MB Amsterdam, The Netherlands
| | - Krijn P. van Lienden
- Department of Radiology, Amsterdam UMC, University of Amsterdam, 1007 MB Amsterdam, The Netherlands; (K.P.v.L.); (M.M.)
| | - Mario Maas
- Department of Radiology, Amsterdam UMC, University of Amsterdam, 1007 MB Amsterdam, The Netherlands; (K.P.v.L.); (M.M.)
| | - Daniëlle M. Gerlag
- Amsterdam Rheumatology & Immunology Center (ARC), Department of Rheumatology & Clinical Immunology, 1007 MB Amsterdam, The Netherlands; (D.C.A.); (T.H.R.); (J.S.H.); (B.v.K.); (N.S.); (D.M.G.); (P.P.T.); (N.d.V.)
- UCB Pharma, Slough SL1 3XE, UK
| | - Paul P. Tak
- Amsterdam Rheumatology & Immunology Center (ARC), Department of Rheumatology & Clinical Immunology, 1007 MB Amsterdam, The Netherlands; (D.C.A.); (T.H.R.); (J.S.H.); (B.v.K.); (N.S.); (D.M.G.); (P.P.T.); (N.d.V.)
- Candel Therapeutics, Needham, MA 02494, USA
- Department of Internal Medicine, Cambridge University, Cambridge CB2 0QQ, UK
| | - Niek de Vries
- Amsterdam Rheumatology & Immunology Center (ARC), Department of Rheumatology & Clinical Immunology, 1007 MB Amsterdam, The Netherlands; (D.C.A.); (T.H.R.); (J.S.H.); (B.v.K.); (N.S.); (D.M.G.); (P.P.T.); (N.d.V.)
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, 1007 MB Amsterdam, The Netherlands
| | - Lisa G. M. van Baarsen
- Amsterdam Rheumatology & Immunology Center (ARC), Department of Rheumatology & Clinical Immunology, 1007 MB Amsterdam, The Netherlands; (D.C.A.); (T.H.R.); (J.S.H.); (B.v.K.); (N.S.); (D.M.G.); (P.P.T.); (N.d.V.)
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, 1007 MB Amsterdam, The Netherlands
- Correspondence: ; Tel.: +31-20-56-64969; Fax: +31-20-69-19658
| |
Collapse
|
136
|
Heikkilä N, Hetemäki I, Sormunen S, Isoniemi H, Kekäläinen E, Saramäki J, Arstila TP. Peripheral differentiation patterns of human T cells. Eur J Immunol 2022; 52:882-894. [PMID: 35307831 PMCID: PMC9313577 DOI: 10.1002/eji.202149465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 01/12/2022] [Accepted: 03/15/2022] [Indexed: 11/08/2022]
Abstract
Long-term T-cell memory is dependent on the maintenance of memory T cells in the lymphoid tissues, and at the surface interfaces that provide entry routes for pathogens. However, much of the current information on human T-cell memory is based on analyzing circulating T cells. Here, we have studied the distribution and age-related changes of memory T-cell subsets in samples from blood, mesenteric LNs, spleen, and ileum, obtained from donors ranging in age from 5 days to 67 years of age. Our data show that the main reservoir of polyclonal naive cells is found in the LNs, and the resting memory subsets capable of self-renewal are also prominent there. In contrast, nondividing but functionally active memory subsets dominate the spleen, and especially the ileum. In general, the replacement of naive cells with memory subsets continues throughout our period of observation, with no apparent plateau. In conclusion, the analysis of lymphoid and nonlymphoid tissues reveals a dynamic pattern of changes distinct to each tissue, and with substantial differences between CD4+ and CD8+ compartments.
Collapse
Affiliation(s)
- Nelli Heikkilä
- Translational Immunology Research Program (TRIMM), Research Programs Unit (RPU), Department of Bacteriology and Immunology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Iivo Hetemäki
- Translational Immunology Research Program (TRIMM), Research Programs Unit (RPU), Department of Bacteriology and Immunology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Silja Sormunen
- Department of Computer Science, Aalto University, Espoo, Finland
| | - Helena Isoniemi
- Division of Transplantation and Liver Surgery, Helsinki University Central Hospital, Helsinki, Finland
| | - Eliisa Kekäläinen
- Translational Immunology Research Program (TRIMM), Research Programs Unit (RPU), Department of Bacteriology and Immunology, Medicum, Faculty of Medicine, HUSLAB Clinical Microbiology, University of Helsinki, and Helsinki University Central Hospital, Helsinki, Finland
| | - Jari Saramäki
- Department of Computer Science, Aalto University, Espoo, Finland
| | - T Petteri Arstila
- Translational Immunology Research Program (TRIMM), Research Programs Unit (RPU), Department of Bacteriology and Immunology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
137
|
Deng B, Ma B, Ma Y, Cao P, Leng X, Huang P, Zhao Y, Ji T, Lu X, Liu L. Doxorubicin and CpG loaded liposomal spherical nucleic acid for enhanced Cancer treatment. J Nanobiotechnology 2022; 20:140. [PMID: 35303868 PMCID: PMC8932194 DOI: 10.1186/s12951-022-01353-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 03/04/2022] [Indexed: 01/01/2023] Open
Abstract
Chemotherapeutics that can trigger immunogenic cell death (ICD) and release tumor-specific antigens are effective on treating a variety of cancers. The codelivery of chemotherapeutics with adjuvants is a promising strategy to achieve synergistic therapeutic effect. However, low drug loading and complicated preparation of current delivery systems lead to carrier-associated toxicity and immunogenicity. Herein, we developed a facile approach to construct liposomal spherical nucleic acids (SNA) by the self-assembly of 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE)-doxorubicin conjugate and DOPE-matrix metalloproteinases-9 (MMP-9) responsive peptide-CpG conjugate (DOPE-MMP-CpG). Liposomal SNAs efficiently co-delivered DOX and CpG into tumors and released the two drugs upon biological stimuli of MMP-9 enzyme in tumor microenvironment (TME) and high concentration of endogenous glutathione in tumor cells. We demonstrated that liposomal SNA enhanced activation of dendritic cells (DCs), promoted expansion of CD8+ and CD4+ T cells in both tumors and spleen, inhibited tumor growth, and extended animal survival. This work provided a simple strategy of delivering chemotherapeutics and adjuvants to tumors with synergistic therapeutic effect and reduced side effect.
Collapse
Affiliation(s)
- Bo Deng
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Bing Ma
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Yingying Ma
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Pei Cao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Xigang Leng
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Pengyu Huang
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Yuanyuan Zhao
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Science, No. 2, 1st North Street, Zhongguancun, Beijing, 100190, People's Republic of China
| | - Tianjiao Ji
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Xueguang Lu
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Science, No. 2, 1st North Street, Zhongguancun, Beijing, 100190, People's Republic of China.
| | - Lanxia Liu
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China.
| |
Collapse
|
138
|
Ikumi NM, Matjila M. Preterm Birth in Women With HIV: The Role of the Placenta. Front Glob Womens Health 2022; 3:820759. [PMID: 35392117 PMCID: PMC8982913 DOI: 10.3389/fgwh.2022.820759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/08/2022] [Indexed: 01/12/2023] Open
Abstract
Maternal HIV infection is associated with an increased risk of preterm birth (PTB). However, the mechanisms underlying this increased risk in women with HIV remain poorly understood. In this regard, it is well-established that labor is an inflammatory process and premature activation of the pro-inflammatory signals (associated with labor) can result in preterm labor which can subsequently lead to PTB. HIV infection is known to cause severe immune dysregulation within its host characterized by altered immune profiles, chronic inflammation and eventually, the progressive failure of the immune system. The human placenta comprises different immune cell subsets, some of which play an important role during pregnancy including participating in the inflammatory processes that accompany labor. It is therefore plausible that HIV/antiretroviral therapy (ART)-associated immune dysregulation within the placental microenvironment may underlie the increased risk of PTB reported in women with HIV. Here, we review evidence from studies that point toward the placental origin of spontaneous PTB and discuss possible ways maternal HIV infection and/or ART could increase this risk. We focus on key cellular players in the maternal decidua including natural killer cells, CD4+ T cells including CD4+ regulatory T cells, CD8+ T cells as well as macrophages.
Collapse
|
139
|
Roquilly A, Mintern JD, Villadangos JA. Spatiotemporal Adaptations of Macrophage and Dendritic Cell Development and Function. Annu Rev Immunol 2022; 40:525-557. [PMID: 35130030 DOI: 10.1146/annurev-immunol-101320-031931] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Macrophages and conventional dendritic cells (cDCs) are distributed throughout the body, maintaining tissue homeostasis and tolerance to self and orchestrating innate and adaptive immunity against infection and cancer. As they complement each other, it is important to understand how they cooperate and the mechanisms that integrate their functions. Both are exposed to commensal microbes, pathogens, and other environmental challenges that differ widely among anatomical locations and over time. To adjust to these varying conditions, macrophages and cDCs acquire spatiotemporal adaptations (STAs) at different stages of their life cycle that determine how they respond to infection. The STAs acquired in response to previous infections can result in increased responsiveness to infection, termed training, or in reduced responses, termed paralysis, which in extreme cases can cause immunosuppression. Understanding the developmental stage and location where macrophages and cDCs acquire their STAs, and the molecular and cellular players involved in their induction, may afford opportunities to harness their beneficial outcomes and avoid or reverse their deleterious effects. Here we review our current understanding of macrophage and cDC development, life cycle, function, and STA acquisition before, during, and after infection. We propose a unified framework to explain how these two cell types adjust their activities to changing conditions over space and time to coordinate their immunosurveillance functions. Expected final online publication date for the Annual Review of Immunology, Volume 40 is April 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Antoine Roquilly
- Center for Research in Transplantation and Translational Immunology, INSERM, UMR 1064, CHU Nantes, University of Nantes, Nantes, France
| | - Justine D Mintern
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Jose A Villadangos
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia.,Department of Microbiology and Immunology, Doherty Institute of Infection and Immunity, The University of Melbourne, Parkville, Victoria, Australia;
| |
Collapse
|
140
|
Lai X, Keller C, Santos G, Schaft N, Dörrie J, Vera J. Multi-Level Computational Modeling of Anti-Cancer Dendritic Cell Vaccination Utilized to Select Molecular Targets for Therapy Optimization. Front Cell Dev Biol 2022; 9:746359. [PMID: 35186943 PMCID: PMC8847669 DOI: 10.3389/fcell.2021.746359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/23/2021] [Indexed: 01/18/2023] Open
Abstract
Dendritic cells (DCs) can be used for therapeutic vaccination against cancer. The success of this therapy depends on efficient tumor-antigen presentation to cytotoxic T lymphocytes (CTLs) and the induction of durable CTL responses by the DCs. Therefore, simulation of such a biological system by computational modeling is appealing because it can improve our understanding of the molecular mechanisms underlying CTL induction by DCs and help identify new strategies to improve therapeutic DC vaccination for cancer. Here, we developed a multi-level model accounting for the life cycle of DCs during anti-cancer immunotherapy. Specifically, the model is composed of three parts representing different stages of DC immunotherapy - the spreading and bio-distribution of intravenously injected DCs in human organs, the biochemical reactions regulating the DCs' maturation and activation, and DC-mediated activation of CTLs. We calibrated the model using quantitative experimental data that account for the activation of key molecular circuits within DCs, the bio-distribution of DCs in the body, and the interaction between DCs and T cells. We showed how such a data-driven model can be exploited in combination with sensitivity analysis and model simulations to identify targets for enhancing anti-cancer DC vaccination. Since other previous works show how modeling improves therapy schedules and DC dosage, we here focused on the molecular optimization of the therapy. In line with this, we simulated the effect in DC vaccination of the concerted modulation of combined intracellular regulatory processes and proposed several possibilities that can enhance DC-mediated immunogenicity. Taken together, we present a comprehensive time-resolved multi-level model for studying DC vaccination in melanoma. Although the model is not intended for personalized patient therapy, it could be used as a tool for identifying molecular targets for optimizing DC-based therapy for cancer, which ultimately should be tested in in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Xin Lai
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie and Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Christine Keller
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Guido Santos
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Departament of Biochemistry, Microbiology, Cell Biology and Genetics, Faculty of Sciences, University of La Laguna, San Cristóbal de La Laguna, Spain
| | - Niels Schaft
- Deutsches Zentrum Immuntherapie and Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- RNA Group, Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jan Dörrie
- Deutsches Zentrum Immuntherapie and Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- RNA Group, Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Julio Vera
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie and Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| |
Collapse
|
141
|
Tseng CY, Wang WX, Douglas TR, Chou LYT. Engineering DNA Nanostructures to Manipulate Immune Receptor Signaling and Immune Cell Fates. Adv Healthc Mater 2022; 11:e2101844. [PMID: 34716686 DOI: 10.1002/adhm.202101844] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/14/2021] [Indexed: 12/19/2022]
Abstract
Immune cells sense, communicate, and logically integrate a multitude of environmental signals to make important cell-fate decisions and fulfill their effector functions. These processes are initiated and regulated by a diverse array of immune receptors and via their dynamic spatiotemporal organization upon ligand binding. Given the widespread relevance of the immune system to health and disease, there have been significant efforts toward understanding the biophysical principles governing immune receptor signaling and activation, as well as the development of biomaterials which exploit these principles for therapeutic immune engineering. Here, how advances in the field of DNA nanotechnology constitute a growing toolbox for further pursuit of these endeavors is discussed. Key cellular players involved in the induction of immunity against pathogens or diseased cells are first summarized. How the ability to design DNA nanostructures with custom shapes, dynamics, and with site-specific incorporation of diverse guests can be leveraged to manipulate the signaling pathways that regulate these processes is then presented. It is followed by highlighting emerging applications of DNA nanotechnology at the crossroads of immune engineering, such as in vitro reconstitution platforms, vaccines, and adjuvant delivery systems. Finally, outstanding questions that remain for further advancing immune-modulatory DNA nanodevices are outlined.
Collapse
Affiliation(s)
- Chung Yi Tseng
- Institute of Biomedical Engineering University of Toronto Toronto Ontario M5S 3G9 Canada
| | - Wendy Xueyi Wang
- Institute of Biomedical Engineering University of Toronto Toronto Ontario M5S 3G9 Canada
| | - Travis Robert Douglas
- Institute of Biomedical Engineering University of Toronto Toronto Ontario M5S 3G9 Canada
| | - Leo Y. T. Chou
- Institute of Biomedical Engineering University of Toronto Toronto Ontario M5S 3G9 Canada
| |
Collapse
|
142
|
Tsuda S, Pipkin ME. Transcriptional Control of Cell Fate Determination in Antigen-Experienced CD8 T Cells. Cold Spring Harb Perspect Biol 2022; 14:a037945. [PMID: 34127445 PMCID: PMC8805646 DOI: 10.1101/cshperspect.a037945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Robust immunity to intracellular infections is mediated by antigen-specific naive CD8 T cells that become activated and differentiate into phenotypically and functionally diverse subsets of effector cells, some of which terminally differentiate and others that give rise to memory cells that provide long-lived protection. This developmental system is an outstanding model with which to elucidate how regulation of chromatin structure and transcriptional control establish gene expression programs that govern cell fate determination, insights from which are likely to be useful for informing the design of immunotherapeutic approaches to engineer durable immunity to infections and tumors. A unifying framework that describes how naive CD8 T cells develop into memory cells is still outstanding. We propose a model that incorporates a common early linear path followed by divergent paths that slowly lose capacity to interconvert and discuss classical and contemporary observations that support these notions, focusing on insights from transcriptional control and chromatin regulation.
Collapse
Affiliation(s)
- Shanel Tsuda
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Matthew E Pipkin
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida 33458, USA
| |
Collapse
|
143
|
Faraj S, Kemp EH, Gawkrodger DJ. Patho-immunological mechanisms of vitiligo: the role of the innate and adaptive immunities and environmental stress factors. Clin Exp Immunol 2022; 207:27-43. [PMID: 35020865 PMCID: PMC8802175 DOI: 10.1093/cei/uxab002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 10/04/2021] [Accepted: 10/15/2021] [Indexed: 12/17/2022] Open
Abstract
Epidermal melanocyte loss in vitiligo, triggered by stresses ranging from trauma to emotional stress, chemical exposure or metabolite imbalance, to the unknown, can stimulate oxidative stress in pigment cells, which secrete damage-associated molecular patterns that then initiate innate immune responses. Antigen presentation to melanocytes leads to stimulation of autoreactive T-cell responses, with further targeting of pigment cells. Studies show a pathogenic basis for cellular stress, innate immune responses and adaptive immunity in vitiligo. Improved understanding of the aetiological mechanisms in vitiligo has already resulted in successful use of the Jak inhibitors in vitiligo. In this review, we outline the current understanding of the pathological mechanisms in vitiligo and locate loci to which therapeutic attack might be directed.
Collapse
Affiliation(s)
- Safa Faraj
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | | | - David John Gawkrodger
- Department of Infection, Immunology and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| |
Collapse
|
144
|
Sun H, He T, Wu Y, Yuan H, Ning J, Zhang Z, Deng X, Li B, Wu C. Cytotoxin-Associated Gene A-Negative Helicobacter pylori Promotes Gastric Mucosal CX3CR1+CD4+ Effector Memory T Cell Recruitment in Mice. Front Microbiol 2022; 13:813774. [PMID: 35154057 PMCID: PMC8829513 DOI: 10.3389/fmicb.2022.813774] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/07/2022] [Indexed: 11/13/2022] Open
Abstract
Background Helicobacter pylori can cause many kinds of gastric disorders, ranging from gastritis to gastric cancer. Cytotoxin-associated gene A (CagA)+H. pylori is more likely to cause gastric histopathologic damage than CagA–H. pylori. However, the underlying mechanism needs to be further investigated. Materials and methods Mice were intragastrically administered equal amounts of CagA+ or CagA–H. pylori. Four weeks later, 24 chemokines in stomachs were measured using a mouse chemokine array, and the phenotypes of the recruited gastric CD4+ T cells were analyzed. The migration pathway was evaluated. Finally, the correlation between each pair among the recruited CD4+ T cell sub-population, H. pylori colonization level, and histopathologic damage score were determined by Pearson correlation analysis. Results The concentration of chemokines, CCL3 and CX3CL1, were significantly elevated in CagA–H. pylori-infected gastric mucosa than in CagA+H. pylori-infected gastric mucosa. Among them, CX3CL1 secreted by gastric epithelial cells, which was elicited more effectively by CagA–H. pylori than by the CagA+ strain, dramatically promoted mucosal CD4+ T cell migration. The expression of CX3CR1, the only known receptor of CX3CL1, was upregulated on the surface of gastric CD4+ T cells in CagA–H. pylori-infected stomach. In addition, most of the CX3CR1-positive gastric CD4+ T cells were CD44+CD69–CCR7– effector memory T cells (Tem). Pearson correlation analysis showed that the recruited CX3CR1+CD4+ Tem cell population was negatively correlated with H. pylori colonization level and histopathologic damage score. Conclusion CagA–H. pylori promotes gastric mucosal CX3CR1+CD4+ Tem recruitment in mice.
Collapse
Affiliation(s)
- Heqiang Sun
- Department of Laboratory Medicine, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Taojun He
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yanan Wu
- Peking University People’s Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Hanmei Yuan
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Jie Ning
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Zhenhua Zhang
- Department of Gastroenterology of the 305 Hospital of Chinese People’s Liberation Army, Beijing, China
| | - Xinli Deng
- Department of Laboratory Medicine, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Bin Li
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Bin Li,
| | - Chao Wu
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- *Correspondence: Chao Wu,
| |
Collapse
|
145
|
Mapalagamage M, Weiskopf D, Sette A, De Silva AD. Current Understanding of the Role of T Cells in Chikungunya, Dengue and Zika Infections. Viruses 2022; 14:v14020242. [PMID: 35215836 PMCID: PMC8878350 DOI: 10.3390/v14020242] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/14/2022] [Accepted: 01/15/2022] [Indexed: 02/06/2023] Open
Abstract
Arboviral infections such as Chikungunya (CHIKV), Dengue (DENV) and Zika (ZIKV) are a major disease burden in tropical and sub-tropical countries, and there are no effective vaccinations or therapeutic drugs available at this time. Understanding the role of the T cell response is very important when designing effective vaccines. Currently, comprehensive identification of T cell epitopes during a DENV infection shows that CD8 and CD4 T cells and their specific phenotypes play protective and pathogenic roles. The protective role of CD8 T cells in DENV is carried out through the killing of infected cells and the production of proinflammatory cytokines, as CD4 T cells enhance B cell and CD8 T cell activities. A limited number of studies attempted to identify the involvement of T cells in CHIKV and ZIKV infection. The identification of human immunodominant ZIKV viral epitopes responsive to specific T cells is scarce, and none have been identified for CHIKV. In CHIKV infection, CD8 T cells are activated during the acute phase in the lymph nodes/blood, and CD4 T cells are activated during the chronic phase in the joints/muscles. Studies on the role of T cells in ZIKV-neuropathogenesis are limited and need to be explored. Many studies have shown the modulating actions of T cells due to cross-reactivity between DENV-ZIKV co-infections and have repeated heterologous/homologous DENV infection, which is an important factor to consider when developing an effective vaccine.
Collapse
Affiliation(s)
- Maheshi Mapalagamage
- Department of Zoology and Environment Sciences, Faculty of Science, University of Colombo, Colombo 00700, Sri Lanka;
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; (D.W.); (A.S.)
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; (D.W.); (A.S.)
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; (D.W.); (A.S.)
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego (UCSD), La Jolla, CA 92037, USA
| | - Aruna Dharshan De Silva
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; (D.W.); (A.S.)
- Department of Paraclinical Sciences, Faculty of Medicine, General Sir John Kotelawala Defence University, Colombo 10390, Sri Lanka
- Correspondence:
| |
Collapse
|
146
|
T cell subtype profiling measures exhaustion and predicts anti-PD-1 response. Sci Rep 2022; 12:1342. [PMID: 35079117 PMCID: PMC8789795 DOI: 10.1038/s41598-022-05474-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/05/2022] [Indexed: 12/11/2022] Open
Abstract
Anti-PD-1 therapy can provide long, durable benefit to a fraction of patients. The on-label PD-L1 test, however, does not accurately predict response. To build a better biomarker, we created a method called T Cell Subtype Profiling (TCSP) that characterizes the abundance of T cell subtypes (TCSs) in FFPE specimens using five RNA models. These TCS RNA models are created using functional methods, and robustly discriminate between naïve, activated, exhausted, effector memory, and central memory TCSs, without the reliance on non-specific, classical markers. TCSP is analytically valid and corroborates associations between TCSs and clinical outcomes. Multianalyte biomarkers based on TCS estimates predicted response to anti-PD-1 therapy in three different cancers and outperformed the indicated PD-L1 test, as well as Tumor Mutational Burden. Given the utility of TCSP, we investigated the abundance of TCSs in TCGA cancers and created a portal to enable researchers to discover other TCSP-based biomarkers.
Collapse
|
147
|
Wu Y, Wang GJ, He HQ, Qin HH, Shen WT, Yu Y, Zhang X, Zhou ML, Fei JB. Low-dose intralesional injection of 5-fluorouracil and triamcinolone reduces tissue resident memory T cells in chronic eczema. World J Clin Cases 2022; 10:166-176. [PMID: 35071516 PMCID: PMC8727240 DOI: 10.12998/wjcc.v10.i1.166] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 10/29/2021] [Accepted: 11/28/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Tissue resident memory T (TRM) cells have been reported to play a significant role in the pathogenesis and relapse of chronic eczema.
AIM To compare the efficacy and safety of the intralesional injection of 5-fluorouracil (5-FU) and triamcinolone (TA) with those associated with TA alone for the treatment of chronic eczema.
METHODS A total of 168 patients were randomized to 5-FU+TA or TA groups and received a one-time intralesional injection of 5-FU+TA or TA only. Biopsies were collected before and 2 wk after treatment for evaluation of histopathological changes. All patients were followed up monthly for up to 1 year.
RESULTS No serious adverse event was observed in either group. Although the mean atopic dermatitis severity index scores and effective rates were comparable between the two groups after 2 wk of treatment, the relapse rate was significantly lower in the 5-FU+TA group than in the TA group. Histological examination showed significantly fewer CD8+ and CD103+ T cells but not CD4+ T cells in the 5-FU+TA group.
CONCLUSION One-time intralesional injection of 5-FU+TA is effective and safe for chronic eczema treatment and can further reduce the retention of TRM cells in the lesional skin and the relapse rate of chronic eczema.
Collapse
Affiliation(s)
- Yun Wu
- Department of Dermatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Guo-Jiang Wang
- Department of Dermatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Hui-Qiong He
- Department of Dermatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Hai-Hong Qin
- Department of Dermatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Wen-Tong Shen
- Department of Dermatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Yue Yu
- Department of Dermatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Xun Zhang
- Department of Dermatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Mao-Lin Zhou
- Department of Dermatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Jian-Biao Fei
- Department of Dermatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| |
Collapse
|
148
|
Roth GA, Picece VCTM, Ou BS, Luo W, Pulendran B, Appel EA. Designing spatial and temporal control of vaccine responses. NATURE REVIEWS. MATERIALS 2022; 7:174-195. [PMID: 34603749 PMCID: PMC8477997 DOI: 10.1038/s41578-021-00372-2] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/08/2021] [Indexed: 05/02/2023]
Abstract
Vaccines are the key technology to combat existing and emerging infectious diseases. However, increasing the potency, quality and durability of the vaccine response remains a challenge. As our knowledge of the immune system deepens, it becomes clear that vaccine components must be in the right place at the right time to orchestrate a potent and durable response. Material platforms, such as nanoparticles, hydrogels and microneedles, can be engineered to spatially and temporally control the interactions of vaccine components with immune cells. Materials-based vaccination strategies can augment the immune response by improving innate immune cell activation, creating local inflammatory niches, targeting lymph node delivery and controlling the time frame of vaccine delivery, with the goal of inducing enhanced memory immunity to protect against future infections. In this Review, we highlight the biological mechanisms underlying strong humoral and cell-mediated immune responses and explore materials design strategies to manipulate and control these mechanisms.
Collapse
Affiliation(s)
- Gillie A. Roth
- Department of Bioengineering, Stanford University, Stanford, CA USA
| | - Vittoria C. T. M. Picece
- Department of Materials Science & Engineering, Stanford University, Stanford, CA USA
- Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, Switzerland
| | - Ben S. Ou
- Department of Bioengineering, Stanford University, Stanford, CA USA
| | - Wei Luo
- Institute for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford, CA USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford, CA USA
- ChEM-H Institute, Stanford University, Stanford, CA USA
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA USA
- Program in Immunology, Stanford University School of Medicine, Stanford, CA USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA USA
| | - Eric A. Appel
- Department of Bioengineering, Stanford University, Stanford, CA USA
- Department of Materials Science & Engineering, Stanford University, Stanford, CA USA
- ChEM-H Institute, Stanford University, Stanford, CA USA
- Department of Paediatrics — Endocrinology, Stanford University School of Medicine, Stanford, CA USA
| |
Collapse
|
149
|
Understanding human immunity in idiopathic recurrent pregnancy loss. Eur J Obstet Gynecol Reprod Biol 2021; 270:17-29. [PMID: 35007974 DOI: 10.1016/j.ejogrb.2021.12.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 12/12/2021] [Accepted: 12/21/2021] [Indexed: 01/03/2023]
Abstract
Miscarriage, defined as the loss of a pregnancy before a viable gestation, affects 1 in 6 couples. Recurrent pregnancy loss (RPL), defined as two or more miscarriages, affects up to 1.9% of couples. The physical, psychological, and financial impact of miscarriage can be substantial. However, despite its multifactorial etiology, for up to 50% of couples a reason behind this condition cannot be identified, termed 'idiopathic RPL'. Much recent research has strived to understand this, with immune dysregulation being a source of particular interest. In this short review we summarize the current evidence on the complex role of the immune system both pre- and early post-conception in RPL. A key question is whether systemic peripheral blood markers, in particular natural killer cell and T cells, may be utilized to accurately predict and/ or diagnose those pregnancies at high risk of loss. Given the invasive nature of endometrial testing, identification of reliable peripheral immune biomarkers is particularly appealing. Clinical trials using potent immunomodulatory agents, including intravenous immunoglobulin, donor leukocyte immunization, and tumor necrosis factor (TNF)-α inhibitors, have been undertaken with the primary objective of preventing miscarriage in women with RPL. Standardisation of both diagnostic and prognostic immune cell testing assays is required to permit accurate identification of those women who may benefit from immunomodulation. Prompt clarification is required to meet the increasing expectation from couples and clinicians, as without these advancements women are at risk of exposure to potent immune-therapies and subsequent studies are at risk of failure, generating further controversy regarding the role of immune dysregulation in women with RPL. Through this review we highlight clear gaps in our current knowledge on immune activity in RPL.
Collapse
|
150
|
Cai J, Zhao J, Liu D, Xie H, Qi H, Ma J, Sun Z, Zhao H. Efficacy and Safety of Central Memory T Cells Combined With Adjuvant Therapy to Prevent Recurrence of Hepatocellular Carcinoma With Microvascular Invasion: A Pilot Study. Front Oncol 2021; 11:781029. [PMID: 34926296 PMCID: PMC8679661 DOI: 10.3389/fonc.2021.781029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/08/2021] [Indexed: 01/27/2023] Open
Abstract
Background Postoperative adjuvant transcatheter arterial chemoembolization (TACE) following curative hepatectomy has been reported to improve the clinical outcomes of hepatocellular carcinoma (HCC) patients with microvascular invasion (MVI), but more endeavors are required to achieve greater clinical benefit. Central memory T-cell (Tcm) self-transfusion has shown superior antitumor activity in several preclinical studies; however, clinical studies are rare. The aim of this study was to evaluate the clinical benefit and safety of combination treatment with Tcm self-transfusion and TACE as adjuvant treatment in HCC patients with MVI after curative hepatectomy. Methods From October 2016 to September 2018, primary HCC patients with histologically confirmed MVI who underwent curative hepatectomy at the Cancer Hospital of the Chinese Academy of Medical Sciences were recruited for this study. The patients were divided into a Tcm group (combined Tcm self-transfusion with TACE treatment) or a control group (TACE treatment alone) according to their willingness. The recurrence-free survival (RFS), quality-of-life (QOL) score, and adverse events of each patient were recorded within 2 years. Results A total of 52 patients were enrolled, and 48 were eligible for the final data analysis. The median follow-up time was 20.5 months (95% CI: 17.05–22.55 months). The median RFS time was 9.5 months in the control group; the cutoff date was not reached in the Tcm group (when the follow-up duration was 12 months, p = 0.049, HR = 0.40; 95% CI: 0.16–0.99). Compared with the control group, 1- and 2-year RFS rates were higher in the Tcm group (72.0% vs. 46.4% and 58.18% vs. 39.14%, respectively). Multivariate analysis did not indicate that Tcm treatment was an independent prognostic factor associated with HCC recurrence (p = 0.107, HR = 2.312; 95% CI: 0.835–6.400), which might be due to the small sample size of this study. Nevertheless, Tcm treatment effectively improved a reduced QOL due to HCC and liver function injury. Finally, the safety profile of Tcm treatment in this study was good, without any serious adverse events. Conclusions This pilot study showed that Tcm self-transfusion combined with TACE treatment might be a beneficial adjuvant therapy with good safety for primary HCC patients with MVI after curative hepatectomy. Trial registration number NCT03575806
Collapse
Affiliation(s)
- Jianqiang Cai
- Department of Hepatobiliary Surgery, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianjun Zhao
- Department of Hepatobiliary Surgery, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Defang Liu
- Department of New Drug Registration, Hebei Immune Cell Application Engineering Research Center/Baoding Newish Technology Co., LTD/Newish Technology (Beijing) Co., LTD, Beijing, China
| | - Huangfan Xie
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Hailong Qi
- Department of New Drug Registration, Hebei Immune Cell Application Engineering Research Center/Baoding Newish Technology Co., LTD/Newish Technology (Beijing) Co., LTD, Beijing, China.,School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Junfan Ma
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Zhongjie Sun
- State Key Laboratory of Elemento-Organic Chemistry, College of Chemistry, Nankai University, Tianjin, China
| | - Hong Zhao
- Department of Hepatobiliary Surgery, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|