1
|
Nath PR, Isakov N. Mechanisms of Cbl-Mediated Ubiquitination of Proteins in T and Natural Killer Cells and Effects on Immune Cell Functions. Life (Basel) 2024; 14:1592. [PMID: 39768300 PMCID: PMC11677474 DOI: 10.3390/life14121592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/17/2024] [Accepted: 11/30/2024] [Indexed: 01/11/2025] Open
Abstract
Post-translational ubiquitination is an essential mechanism for the regulation of protein stability and function, which contributes to the regulation of the immune system. Cbl, an E3 ubiquitin ligase, is particularly well-characterized in the context of T and NK cell signaling, where it serves as a key regulator of receptor downstream signaling events and as a modulator of cell activation. Cbl promotes the proteasomal degradation of TCR/CD3 subunits as well as the protein kinases Fyn and Lck in T cells. Additionally, the scaffold protein linker for activation of T cells (LAT) is a universal target for Cbl-mediated ubiquitination and degradation in both T and NK cells. Recent findings suggest that CrkII-mediated ubiquitination and degradation of C3G by Cbl during early T cell activation may also be relevant to NK cell signaling. Given its role in modulating immune responses and its manageable impact on autoimmunity, Cbl is being investigated as a target for cancer immunotherapy. This review explores the ubiquitin ligase activity of Cbl and its implications for CAR T and NK cell immunotherapies.
Collapse
Affiliation(s)
- Pulak Ranjan Nath
- Lentigen Technology Inc., A Miltenyi Biotec Company, 910 Clopper Road, Gaithersburg, MD 20878, USA;
| | - Noah Isakov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of the Negev, P.O. Box 653, Beer Sheva 84105, Israel
| |
Collapse
|
2
|
Seo S, Hattori M, Yoshida T. Establishing an evaluation system for T cell activation and anergy based on CD25 expression levels as an indicator. Cytotechnology 2024; 76:749-759. [PMID: 39435413 PMCID: PMC11490625 DOI: 10.1007/s10616-024-00651-0] [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: 04/12/2024] [Accepted: 07/31/2024] [Indexed: 10/23/2024] Open
Abstract
T cell anergy refers to a state where T cells become unresponsive, playing an important role in several types of immune tolerance, such as oral tolerance. This tolerance is vital for preventing some diseases, including food allergies. Understanding the mechanism underlying T cell anergy is essential to addressing food allergies. Previous studies often identified anergic T cells by their decreased ability to produce cytokine compared to the control cells. In the studies, unstimulated or naïve T cells were commonly used as the control cells. These systems could evaluate the hyporesponsiveness of anergic T cells; however, it was challenging to distinguish whether the decrease in cytokine production by anergic T cells was owing to anergy induction or merely a temporarily response to a certain stimulation. This complexity arises because some T cell responses are temporarily suppressed, even by activating stimuli. Therefore, this study aims to explore a new evaluation index that can differentiate the responsiveness of activated T cells from that of anergic T cells compared to the control cells. It was demonstrated that CD25 expression levels serve as an appropriate indicator for distinguishing between T-cell activation and anergy. Conversely, cytokine-producing ability proved inadequate for this purpose. It was found that CD25 expression increased in activated T cells than in naïve T cells, whereas it decreased in anergic T cells after restimulation. This occurred despite decreased cytokine production in the activated and anergic T cells than in the naïve T cells. This new evaluation system, centered on CD25 expression, may help in identifying the mechanism for determining T cell activation and anergy.
Collapse
Affiliation(s)
- Sangwon Seo
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu City, Tokyo, 183-8509 Japan
| | - Makoto Hattori
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu City, Tokyo, 183-8509 Japan
| | - Tadashi Yoshida
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu City, Tokyo, 183-8509 Japan
| |
Collapse
|
3
|
Chudnovskiy A, Castro TBR, Nakandakari-Higa S, Cui A, Lin CH, Sade-Feldman M, Phillips BK, Pae J, Mesin L, Bortolatto J, Schweitzer LD, Pasqual G, Lu LF, Hacohen N, Victora GD. Proximity-dependent labeling identifies dendritic cells that drive the tumor-specific CD4 + T cell response. Sci Immunol 2024; 9:eadq8843. [PMID: 39365874 DOI: 10.1126/sciimmunol.adq8843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/26/2024] [Indexed: 10/06/2024]
Abstract
Dendritic cells (DCs) are uniquely capable of transporting tumor antigens to tumor-draining lymph nodes (tdLNs) and interact with effector T cells in the tumor microenvironment (TME) itself, mediating both natural antitumor immunity and the response to checkpoint blockade immunotherapy. Using LIPSTIC (Labeling Immune Partnerships by SorTagging Intercellular Contacts)-based single-cell transcriptomics, we identified individual DCs capable of presenting antigen to CD4+ T cells in both the tdLN and TME. Our findings revealed that DCs with similar hyperactivated transcriptional phenotypes interact with helper T cells both in tumors and in the tdLN and that checkpoint blockade drugs enhance these interactions. These findings show that a relatively small fraction of DCs is responsible for most of the antigen presentation in the tdLN and TME to both CD4+ and CD8+ tumor-specific T cells and that classical checkpoint blockade enhances CD40-driven DC activation at both sites.
Collapse
Affiliation(s)
- Aleksey Chudnovskiy
- Laboratory of Lymphocyte Dynamics, Rockefeller University, New York, NY, USA
| | - Tiago B R Castro
- Laboratory of Lymphocyte Dynamics, Rockefeller University, New York, NY, USA
| | | | - Ang Cui
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard School of Dental Medicine, Harvard University, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Chia-Hao Lin
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | | | - Brooke K Phillips
- Laboratory of Lymphocyte Dynamics, Rockefeller University, New York, NY, USA
| | - Juhee Pae
- Laboratory of Lymphocyte Dynamics, Rockefeller University, New York, NY, USA
| | - Luka Mesin
- Laboratory of Lymphocyte Dynamics, Rockefeller University, New York, NY, USA
| | - Juliana Bortolatto
- Laboratory of Lymphocyte Dynamics, Rockefeller University, New York, NY, USA
| | | | - Giulia Pasqual
- Laboratory of Synthetic Immunology, Oncology and Immunology Section, Department of Surgery Oncology and Gastroenterology, University of Padua, Padua, Italy
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Li-Fan Lu
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Gabriel D Victora
- Laboratory of Lymphocyte Dynamics, Rockefeller University, New York, NY, USA
| |
Collapse
|
4
|
Wang J, Han X, Hao Y, Chen S, Pang B, Zou L, Han X, Wang W, Liu L, Shen M, Jin A. Cbl-b inhibition promotes less differentiated phenotypes of T cells with enhanced cytokine production. Cell Immunol 2024; 403-404:104863. [PMID: 39186873 DOI: 10.1016/j.cellimm.2024.104863] [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: 07/05/2024] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 08/28/2024]
Abstract
For adoptive therapy with T cell receptor engineered T (TCR-T) cells, the quantity and quality of the final cell product directly affect their anti-tumor efficacy. The post-transfer efficacy window of TCR-T cells is keen to optimizing attempts during the manufacturing process. Cbl-b is a E3 ubiquitin ligase previously shown with critical negative impact in T cell functions. This study investigated whether strategic inclusion of a commercially available small inhibitor targeting Cbl-b (Cbl-b-IN-1) prior to T cell activation could enhance the quality of the final TCR-T cell product. Examination with both PBMCs and TCR-T cells revealed that Cbl-b-IN-1 treatment promoted TCR expression efficiency, T cell proliferation potential and, specifically, cell survival capability post antigenic stimulation. Cbl-b-IN-1 exposure facilitated T cells in maintaining less differentiated states with enhanced cytokine production. Further, we found that Cbl-b-IN-1 effectively augmented the activation of TCR signaling, shown by increased phosphorylation levels of Zeta-chain-associated protein kinase 70 (ZAP70) and phospholipase c-γ1 (PLCγ1). In conclusion, our results evidence that the inclusion of Cbl-b inhibitor immediately prior to TCR-T cell activation may enhance their proliferation, survival, and function potentials, presenting an applicable optimization strategy for immunotherapy with adoptive cell transfer.
Collapse
Affiliation(s)
- Junfan Wang
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400010, China; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing 400010, China
| | - XiaoJian Han
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400010, China; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing 400010, China
| | - Yanan Hao
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400010, China; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing 400010, China
| | - Siyin Chen
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400010, China; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing 400010, China
| | - Bo Pang
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400010, China; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing 400010, China
| | - Lin Zou
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400010, China; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing 400010, China
| | - Xiaxia Han
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400010, China; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing 400010, China
| | - Wang Wang
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400010, China; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing 400010, China
| | - Li Liu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Meiying Shen
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing 400010, China; Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Aishun Jin
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400010, China; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing 400010, China.
| |
Collapse
|
5
|
De Sanctis JB, Garmendia JV, Duchová H, Valentini V, Puskasu A, Kubíčková A, Hajdúch M. Lck Function and Modulation: Immune Cytotoxic Response and Tumor Treatment More Than a Simple Event. Cancers (Basel) 2024; 16:2630. [PMID: 39123358 PMCID: PMC11311849 DOI: 10.3390/cancers16152630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Lck, a member of the Src kinase family, is a non-receptor tyrosine kinase involved in immune cell activation, antigen recognition, tumor growth, and cytotoxic response. The enzyme has usually been linked to T lymphocyte activation upon antigen recognition. Lck activation is central to CD4, CD8, and NK activation. However, recently, it has become clearer that activating the enzyme in CD8 cells can be independent of antigen presentation and enhance the cytotoxic response. The role of Lck in NK cytotoxic function has been controversial in a similar fashion as the role of the enzyme in CAR T cells. Inhibiting tyrosine kinases has been a highly successful approach to treating hematologic malignancies. The inhibitors may be useful in treating other tumor types, and they may be useful to prevent cell exhaustion. New, more selective inhibitors have been documented, and they have shown interesting activities not only in tumor growth but in the treatment of autoimmune diseases, asthma, and graft vs. host disease. Drug repurposing and bioinformatics can aid in solving several unsolved issues about the role of Lck in cancer. In summary, the role of Lck in immune response and tumor growth is not a simple event and requires more research.
Collapse
Affiliation(s)
- Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (J.V.G.); (V.V.); (A.K.); (M.H.)
- Czech Advanced Technologies and Research Institute (CATRIN), 77900 Olomouc, Czech Republic
| | - Jenny Valentina Garmendia
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (J.V.G.); (V.V.); (A.K.); (M.H.)
| | - Hana Duchová
- Faculty of Science, Palacky University, 77900 Olomouc, Czech Republic; (H.D.); (A.P.)
| | - Viktor Valentini
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (J.V.G.); (V.V.); (A.K.); (M.H.)
| | - Alex Puskasu
- Faculty of Science, Palacky University, 77900 Olomouc, Czech Republic; (H.D.); (A.P.)
| | - Agáta Kubíčková
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (J.V.G.); (V.V.); (A.K.); (M.H.)
- Czech Advanced Technologies and Research Institute (CATRIN), 77900 Olomouc, Czech Republic
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (J.V.G.); (V.V.); (A.K.); (M.H.)
- Czech Advanced Technologies and Research Institute (CATRIN), 77900 Olomouc, Czech Republic
- Laboratory of Experimental Medicine, University Hospital Olomouc, 77900 Olomouc, Czech Republic
| |
Collapse
|
6
|
Wang B, Zhou J, An N. Investigating molecular markers linked to acute myocardial infarction and cuproptosis: bioinformatics analysis and validation in the AMI mice model. PeerJ 2024; 12:e17280. [PMID: 38827298 PMCID: PMC11143973 DOI: 10.7717/peerj.17280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 04/01/2024] [Indexed: 06/04/2024] Open
Abstract
Cuproptosis-related key genes play a significant role in the pathological processes of acute myocardial infarction (AMI). However, a complete understanding of the molecular mechanisms behind this participation remains elusive. This study was designed to identify genes and immune cells critical to AMI pathogenesis. Based on the GSE48060 dataset (31 AMI patients and 21 healthy persons, GPL570-55999), we identified genes associated with dysregulated cuproptosis and the activation of immune responses between normal subjects and patients with a first myocardial attack. Two molecular clusters associated with cuproptosis were defined in patients with AMI. Immune infiltration analysis showed that there was significant immunity heterogeneity among different clusters. Multiple immune responses were closely associated with Cluster2-specific differentially expressed genes (DEGs). The generalized linear model machine model presented the best discriminative performance with relatively lower residual and root mean square error, and a higher area under the curve (AUC = 0.870). A final two-gene-based generalized linear model was constructed, exhibiting satisfactory performance in two external validation datasets (AUC = 0.719, GSE66360 and AUC = 0.856, GSE123342). Column graph, calibration curve, and decision curve analyses also proved the accuracy of AMI prediction. We also constructed a mouse C57BL/6 model of AMI (3 h, 48 h, and 1 week) and used qRT-PCR and immunofluorescence to detect the expression changes of CBLB and ZNF302. In this study, we present a systematic analysis of the complex relationship between cuproptosis and a first AMI attack, and provide new insights into the diagnosis and treatment of AMI.
Collapse
Affiliation(s)
- Bingyu Wang
- Ningbo Medical Center Lihuili Hospital, Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Jianqing Zhou
- Ningbo Medical Center Lihuili Hospital, Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Ning An
- Ningbo Medical Center Lihuili Hospital, Medical School of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
7
|
Liu L, Xiang Y, Shao L, Yuan C, Song X, Sun M, Liu Y, Zhang X, Du S, Hou M, Peng J, Shi Y. E3 ubiquitin ligase casitas B-lineage lymphoma-b modulates T-cell anergic resistance via phosphoinositide 3-kinase signaling in patients with immune thrombocytopenia. J Thromb Haemost 2024; 22:1202-1214. [PMID: 38184203 DOI: 10.1016/j.jtha.2023.12.032] [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: 08/13/2023] [Revised: 12/01/2023] [Accepted: 12/24/2023] [Indexed: 01/08/2024]
Abstract
BACKGROUND The E3 ubiquitin ligase casitas B-lineage lymphoma-b (CBLB) is a newly identified component of the ubiquitin-dependent protein degradation system and is considered an important negative regulator of immune cells. CBLB is essential for establishing a threshold of T-cell activation and regulating peripheral T-cell tolerance through various mechanisms. However, the involvement of CBLB in the pathogenesis of immune thrombocytopenia (ITP) is unknown. OBJECTIVES We aimed to investigate the expression and role of CBLB in CD4+ T cells obtained from patients with ITP through quantitative proteomics analyses. METHODS CD4+ T cells were transfected with adenoviral vectors overexpressing CBLB to clarify the effect of CBLB on anergic induction of T cells in patients with ITP. DNA methylation levels of the CBLB promoter and 5' untranslated region (UTR) in patient-derived CD4+ T cells were detected via MassARRAY EpiTYPER assay (Agena Bioscience). RESULTS CD4+ T cells from patients with ITP showed resistance to anergic induction, highly activated phosphoinositide 3-kinase-protein kinase B (AKT) signaling, decreased CBLB expression, and 5' UTR hypermethylation of CBLB. CBLB overexpression in T cells effectively attenuated the elevated phosphorylated protein kinase B level and resistance to anergy. Low-dose decitabine treatment led to significantly elevated levels of CBLB expression in CD4+ T cells from 7 patients showing a partial or complete response. CONCLUSION These results indicate that the 5' UTR hypermethylation of CBLB in CD4+ T cells induces resistance to T-cell anergy in ITP. Thus, the upregulation of CBLB expression by low-dose decitabine treatment may represent a potential therapeutic approach to ITP.
Collapse
Affiliation(s)
- Lu Liu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Department of Hematology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, Shandong, China
| | - Yujiao Xiang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Experimental Asthma and Allergy Research Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Linlin Shao
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Chenglu Yuan
- Department of Hematology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, Shandong, China
| | - Xiaofeng Song
- Department of Hand and Foot Surgery, Qilu Hospital (Qingdao) of Shandong University, Qingdao, Shandong, China
| | - Meng Sun
- Jinan Vocational College of Nursing, Jinan, Shandong, China
| | - Yanfeng Liu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xianlei Zhang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Shenghong Du
- Department of Hematology, Taian Central Hospital, Taian, Shandong, China
| | - Ming Hou
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Shandong Provincial Clinical Research Center in Hematological Diseases, Jinan, Shandong, China; Leading Research Group of Scientific Innovation, Department of Science and Technology of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jun Peng
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yan Shi
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
8
|
Vos WG, van Os BW, den Toom M, Beckers L, van Roomen CP, van Tiel CM, Mohapatra BC, Band H, Nitz K, Weber C, Atzler D, de Winther MP, Bosmans LA, Lutgens E, Seijkens TT. T cell specific deletion of Casitas B lineage lymphoma-b reduces atherosclerosis, but increases plaque T cell infiltration and systemic T cell activation. Front Immunol 2024; 15:1297893. [PMID: 38504977 PMCID: PMC10949527 DOI: 10.3389/fimmu.2024.1297893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 02/19/2024] [Indexed: 03/21/2024] Open
Abstract
Introduction Atherosclerosis is a lipid-driven inflammatory disease of the arterial wall, and the underlying cause of the majority of cardiovascular diseases. Recent advances in high-parametric immunophenotyping of immune cells indicate that T cells constitute the major leukocyte population in the atherosclerotic plaque. The E3 ubiquitin ligase Casitas B-lymphoma proto-oncogene-B (CBL-B) is a critical intracellular regulator that sets the threshold for T cell activation, making CBL-B a potential therapeutic target to modulate inflammation in atherosclerosis. We previously demonstrated that complete knock-out of CBL-B aggravated atherosclerosis in Apoe-/- mice, which was attributed to increased macrophage recruitment and increased CD8+ T cell activation in the plaque. Methods To further study the T cell specific role of CBL-B in atherosclerosis, Apoe-/- CD4cre Cblb fl/fl (Cbl-bcKO) mice and Apoe-/-CD4WTCblbfl/fl littermates (Cbl-bfl/fl) were fed a high cholesterol diet for ten weeks. Results Cbl-bcKO mice had smaller atherosclerotic lesions in the aortic arch and root compared to Cbl-bfl/fl, and a substantial increase in CD3+ T cells in the plaque. Collagen content in the plaque was decreased, while other plaque characteristics including plaque necrotic core, macrophage content, and smooth muscle cell content, remained unchanged. Mice lacking T cell CBL-B had a 1.4-fold increase in CD8+ T cells and a 1.8-fold increase in regulatory T cells in the spleen. Splenic CD4+ and CD8+ T cells had increased expression of C-X-C Motif Chemokine Receptor 3 (CXCR3) and interferon-γ (IFN-γ), indicating a T helper 1 (Th1)-like/effector CD8+ T cell-like phenotype. Conclusion In conclusion, Cbl-bcKO mice have reduced atherosclerosis but show increased T cell accumulation in the plaque accompanied by systemic T cell activation.
Collapse
Affiliation(s)
- Winnie G. Vos
- Department of Medical Biochemistry, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands
- Amsterdam Immunity and Infection, Inflammatory Diseases, Amsterdam, Netherlands
| | - Bram W. van Os
- Department of Medical Biochemistry, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands
- Amsterdam Immunity and Infection, Inflammatory Diseases, Amsterdam, Netherlands
| | - Myrthe den Toom
- Department of Medical Biochemistry, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
| | - Linda Beckers
- Department of Medical Biochemistry, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
| | - Cindy P.A.A. van Roomen
- Department of Medical Biochemistry, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
| | - Claudia M. van Tiel
- Department of Medical Biochemistry, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
| | - Bhopal C. Mohapatra
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Hamid Band
- Eppley Institute for Research in Cancer and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| | - Katrin Nitz
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Department of Cardiovascular Medicine and Immunology, Mayo Clinic, Rochester, MN, United States
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
| | - Dorothee Atzler
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Walther Straub Institute of Parmacology and Toxicology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Menno P.J. de Winther
- Department of Medical Biochemistry, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands
- Amsterdam Immunity and Infection, Inflammatory Diseases, Amsterdam, Netherlands
| | - Laura A. Bosmans
- Department of Medical Biochemistry, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands
- Amsterdam Immunity and Infection, Inflammatory Diseases, Amsterdam, Netherlands
| | - Esther Lutgens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Department of Cardiovascular Medicine and Immunology, Mayo Clinic, Rochester, MN, United States
| | - Tom T.P. Seijkens
- Department of Medical Biochemistry, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
9
|
Zhou L, Yang J, Zhang K, Wang T, Jiang S, Zhang X. Rising Star in Immunotherapy: Development and Therapeutic Potential of Small-Molecule Inhibitors Targeting Casitas B Cell Lymphoma-b (Cbl-b). J Med Chem 2024; 67:816-837. [PMID: 38181380 DOI: 10.1021/acs.jmedchem.3c01361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2024]
Abstract
Casitas B cell lymphoma-b (Cbl-b) is a vital negative regulator of TCR and BCR signaling pathways, playing a significant role in setting an appropriate threshold for the activation of T cells and controlling the tolerance of peripheral T cells via a variety of mechanisms. Overexpression of Cbl-b leads to immune hyporesponsiveness of T cells. Conversely, the deficiency of Cbl-b in T cells results in markedly increased production of IL-2, even in the lack of CD28 costimulation in vitro. And Cbl-b-/- mice spontaneously reject multifarious cancers. Therefore, Cbl-b may be associated with immune-mediated diseases, and blocking Cbl-b could be considered as a new antitumor immunotherapy strategy. In this review, the possible regulatory mechanisms and biological potential of Cbl-b for antitumor immunotherapy are summarized. Besides, the potential roles of Cbl-b in immune-mediated diseases are comprehensively discussed, with emphasis on Cbl-b immune-oncology agents in the preclinical stage and clinical trials.
Collapse
Affiliation(s)
- Lixin Zhou
- Department of Medicinal Chemistry, School of Pharmacy and School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Jiamei Yang
- Department of Medicinal Chemistry, School of Pharmacy and School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Kuojun Zhang
- Department of Medicinal Chemistry, School of Pharmacy and School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Tianyu Wang
- Department of Medicinal Chemistry, School of Pharmacy and School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Sheng Jiang
- Department of Medicinal Chemistry, School of Pharmacy and School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Xiangyu Zhang
- Department of Medicinal Chemistry, School of Pharmacy and School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
10
|
Mohammadian Gol T, Kim M, Sinn R, Ureña-Bailén G, Stegmeyer S, Gratz PG, Zahedipour F, Roig-Merino A, Antony JS, Mezger M. CRISPR-Cas9-Based Gene Knockout of Immune Checkpoints in Expanded NK Cells. Int J Mol Sci 2023; 24:16065. [PMID: 38003255 PMCID: PMC10671270 DOI: 10.3390/ijms242216065] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Natural killer (NK) cell immunotherapy has emerged as a novel treatment modality for various cancer types, including leukemia. The modulation of inhibitory signaling pathways in T cells and NK cells has been the subject of extensive investigation in both preclinical and clinical settings in recent years. Nonetheless, further research is imperative to optimize antileukemic activities, especially regarding NK-cell-based immunotherapies. The central scientific question of this study pertains to the potential for boosting cytotoxicity in expanded and activated NK cells through the inhibition of inhibitory receptors. To address this question, we employed the CRISPR-Cas9 system to target three distinct inhibitory signaling pathways in NK cells. Specifically, we examined the roles of A2AR within the metabolic purinergic signaling pathway, CBLB as an intracellular regulator in NK cells, and the surface receptors NKG2A and CD96 in enhancing the antileukemic efficacy of NK cells. Following the successful expansion of NK cells, they were transfected with Cas9+sgRNA RNP to knockout A2AR, CBLB, NKG2A, and CD96. The analysis of indel frequencies for all four targets revealed good knockout efficiencies in expanded NK cells, resulting in diminished protein expression as confirmed by flow cytometry and Western blot analysis. Our in vitro killing assays demonstrated that NKG2A and CBLB knockout led to only a marginal improvement in the cytotoxicity of NK cells against AML and B-ALL cells. Furthermore, the antileukemic activity of CD96 knockout NK cells did not yield significant enhancements, and the blockade of A2AR did not result in significant improvement in killing efficiency. In conclusion, our findings suggest that CRISPR-Cas9-based knockout strategies for immune checkpoints might not be sufficient to efficiently boost the antileukemic functions of expanded (and activated) NK cells and, at the same time, point to the need for strong cellular activating signals, as this can be achieved, for example, via transgenic chimeric antigen receptor expression.
Collapse
Affiliation(s)
- Tahereh Mohammadian Gol
- Department of General Pediatrics, Hematology and Oncology, University Children’s Hospital Tübingen, 72076 Tübingen, Germany; (T.M.G.); (M.K.); (P.G.G.); (F.Z.)
| | - Miso Kim
- Department of General Pediatrics, Hematology and Oncology, University Children’s Hospital Tübingen, 72076 Tübingen, Germany; (T.M.G.); (M.K.); (P.G.G.); (F.Z.)
| | - Ralph Sinn
- Department of General Pediatrics, Hematology and Oncology, University Children’s Hospital Tübingen, 72076 Tübingen, Germany; (T.M.G.); (M.K.); (P.G.G.); (F.Z.)
| | - Guillermo Ureña-Bailén
- Department of General Pediatrics, Hematology and Oncology, University Children’s Hospital Tübingen, 72076 Tübingen, Germany; (T.M.G.); (M.K.); (P.G.G.); (F.Z.)
| | - Sarah Stegmeyer
- Department of General Pediatrics, Hematology and Oncology, University Children’s Hospital Tübingen, 72076 Tübingen, Germany; (T.M.G.); (M.K.); (P.G.G.); (F.Z.)
| | - Paul Gerhard Gratz
- Department of General Pediatrics, Hematology and Oncology, University Children’s Hospital Tübingen, 72076 Tübingen, Germany; (T.M.G.); (M.K.); (P.G.G.); (F.Z.)
| | - Fatemeh Zahedipour
- Department of General Pediatrics, Hematology and Oncology, University Children’s Hospital Tübingen, 72076 Tübingen, Germany; (T.M.G.); (M.K.); (P.G.G.); (F.Z.)
| | | | - Justin S. Antony
- Department of General Pediatrics, Hematology and Oncology, University Children’s Hospital Tübingen, 72076 Tübingen, Germany; (T.M.G.); (M.K.); (P.G.G.); (F.Z.)
| | - Markus Mezger
- Department of General Pediatrics, Hematology and Oncology, University Children’s Hospital Tübingen, 72076 Tübingen, Germany; (T.M.G.); (M.K.); (P.G.G.); (F.Z.)
| |
Collapse
|
11
|
Dong Y, Chen Y, Ma G, Cao H. The role of E3 ubiquitin ligases in bone homeostasis and related diseases. Acta Pharm Sin B 2023; 13:3963-3987. [PMID: 37799379 PMCID: PMC10547920 DOI: 10.1016/j.apsb.2023.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 10/07/2023] Open
Abstract
The ubiquitin-proteasome system (UPS) dedicates to degrade intracellular proteins to modulate demic homeostasis and functions of organisms. These enzymatic cascades mark and modifies target proteins diversly through covalently binding ubiquitin molecules. In the UPS, E3 ubiquitin ligases are the crucial constituents by the advantage of recognizing and presenting proteins to proteasomes for proteolysis. As the major regulators of protein homeostasis, E3 ligases are indispensable to proper cell manners in diverse systems, and they are well described in physiological bone growth and bone metabolism. Pathologically, classic bone-related diseases such as metabolic bone diseases, arthritis, bone neoplasms and bone metastasis of the tumor, etc., were also depicted in a UPS-dependent manner. Therefore, skeletal system is versatilely regulated by UPS and it is worthy to summarize the underlying mechanism. Furthermore, based on the current status of treatment, normal or pathological osteogenesis and tumorigenesis elaborated in this review highlight the clinical significance of UPS research. As a strategy possibly remedies the limitations of UPS treatment, emerging PROTAC was described comprehensively to illustrate its potential in clinical application. Altogether, the purpose of this review aims to provide more evidence for exploiting novel therapeutic strategies based on UPS for bone associated diseases.
Collapse
Affiliation(s)
| | | | - Guixing Ma
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Shenzhen 518055, China
| | - Huiling Cao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Shenzhen 518055, China
| |
Collapse
|
12
|
Zhou L, Ye X, Wang K, Shen H, Wang T, Zhang X, Jiang S, Xiao Y, Zhang K. Discovery of diaminotriazine carboxamides as potent inhibitors of hematopoetic progenitor kinase 1. Bioorg Chem 2023; 138:106682. [PMID: 37339563 DOI: 10.1016/j.bioorg.2023.106682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 06/01/2023] [Accepted: 06/12/2023] [Indexed: 06/22/2023]
Abstract
Hematopoietic progenitor kinase 1 (HPK1), a member of mitogen-activated protein kinase kinase kinase kinase (MAP4K) family of Ste20 serine/threonine kinases, is a negative regulator of T-cell receptor (TCR) signaling. Inactivating HPK1 kinase has been reported to be sufficient to elicit antitumor immune response. Therefore, HPK1 has attracted much attention as a promising target for tumor immunotherapy. A few of HPK1 inhibitors have been reported, and none of them have been approved for clinical applications. Hence, more effective HPK1 inhibitors are needed. Herein, a series of structurally novel diaminotriazine carboxamides were rationally designed, synthesized and evaluated for their inhibitory activity against HPK1 kinase. Most of them exhibited potent inhibitory potency against HPK1 kinase. In particular, compound 15b showed more robust HPK1 inhibitory activity than that of 11d developed by Merck in kinase activity assay (IC50 = 3.1 and 8.2 nM, respectively). The significant inhibitory potency against SLP76 phosphorylation in Jurkat T cells further confirmed the efficacy of compound 15b. In human peripheral blood mononuclear cell (PBMC) functional assays, compound 15b more significantly induced the production of interleukin 2 (IL-2) and interferon γ (IFN-γ) relative to 11d. Furthermore, 15b alone or in combination with anti-PD-1 antibodies showed potent in vivo antitumor efficacy in MC38 tumor-bearing mice. Compound 15b represents a promising lead for the development of effective HPK1 small-molecule inhibitors.
Collapse
Affiliation(s)
- Lixin Zhou
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiuquan Ye
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Kaizhen Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Hongtao Shen
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Tianyu Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiangyu Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Sheng Jiang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Yibei Xiao
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Kuojun Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
13
|
Zheng Y, Yu M, Chen Y, Xue L, Zhu W, Fu G, Morris SW, Wen R, Wang D. CARD19, a Novel Regulator of the TAK1/NF-κB Pathway in Self-Reactive B Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1222-1235. [PMID: 36961449 PMCID: PMC10156913 DOI: 10.4049/jimmunol.2200639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 02/22/2023] [Indexed: 03/25/2023]
Abstract
The caspase recruitment domain family member (CARD)11-Bcl10-Malt1 signalosome controls TGF-β-activated kinase 1 (TAK1) activation and regulates BCR-induced NF-κB activation. In this study, we discovered that CARD19 interacted with TAK1 and inhibited TAB2-mediated TAK1 ubiquitination and activation. Although CARD19 deficiency in mice did not affect B cell development, it enhanced clonal deletion, receptor editing, and anergy of self-reactive B cells, and it reduced autoantibody production. Mechanistically, CARD19 deficiency increased BCR/TAK1-mediated NF-κB activation, leading to increased expression of transcription factors Egr2/3, as well as the E3 ubiquitin ligases c-Cbl/Cbl-b, which are known inducers of B cell tolerance in self-reactive B cells. RNA sequencing analysis revealed that although CARD19 deficiency did not affect the overall Ag-induced gene expression in naive B cells, it suppressed BCR signaling and increased hyporesponsiveness of self-reactive B cells. As a result, CARD19 deficiency prevented Bm12-induced experimental systemic lupus erythematosus. In summary, CARD19 negatively regulates BCR/TAK1-induced NF-κB activation and its deficiency increases Egr2/3 and c-Cbl/Cbl-b expression in self-reactive B cells, thereby enhancing B cell tolerance.
Collapse
Affiliation(s)
| | - Mei Yu
- Versiti Blood Research Institute, Milwaukee, WI
| | - Yuhong Chen
- Versiti Blood Research Institute, Milwaukee, WI
| | | | - Wen Zhu
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Guoping Fu
- Versiti Blood Research Institute, Milwaukee, WI
| | | | - Renren Wen
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Demin Wang
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
14
|
Yoon JS, Lee CW. Protein phosphatases regulate the liver microenvironment in the development of hepatocellular carcinoma. Exp Mol Med 2022; 54:1799-1813. [PMID: 36380016 PMCID: PMC9722691 DOI: 10.1038/s12276-022-00883-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
The liver is a complicated heterogeneous organ composed of different cells. Parenchymal cells called hepatocytes and various nonparenchymal cells, including immune cells and stromal cells, are distributed in liver lobules with hepatic architecture. They interact with each other to compose the liver microenvironment and determine its characteristics. Although the liver microenvironment maintains liver homeostasis and function under healthy conditions, it also shows proinflammatory and profibrogenic characteristics that can induce the progression of hepatitis and hepatic fibrosis, eventually changing to a protumoral microenvironment that contributes to the development of hepatocellular carcinoma (HCC). According to recent studies, phosphatases are involved in liver diseases and HCC development by regulating protein phosphorylation in intracellular signaling pathways and changing the activities and characteristics of liver cells. Therefore, this review aims to highlight the importance of protein phosphatases in HCC development and in the regulation of the cellular components in the liver microenvironment and to show their significance as therapeutic targets.
Collapse
Affiliation(s)
- Joon-Sup Yoon
- grid.264381.a0000 0001 2181 989XDepartment of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, 16419 Republic of Korea
| | - Chang-Woo Lee
- grid.264381.a0000 0001 2181 989XDepartment of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, 16419 Republic of Korea ,grid.264381.a0000 0001 2181 989XDepartment of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351 Republic of Korea
| |
Collapse
|
15
|
Hodgson R, Xu X, Anzilotti C, Deobagkar-Lele M, Crockford TL, Kepple JD, Cawthorne E, Bhandari A, Cebrian-Serrano A, Wilcock MJ, Davies B, Cornall RJ, Bull KR. NDRG1 is induced by antigen-receptor signaling but dispensable for B and T cell self-tolerance. Commun Biol 2022; 5:1216. [PMID: 36357486 PMCID: PMC9649591 DOI: 10.1038/s42003-022-04118-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 10/17/2022] [Indexed: 11/12/2022] Open
Abstract
Peripheral tolerance prevents the initiation of damaging immune responses by autoreactive lymphocytes. While tolerogenic mechanisms are tightly regulated by antigen-dependent and independent signals, downstream pathways are incompletely understood. N-myc downstream-regulated gene 1 (NDRG1), an anti-cancer therapeutic target, has previously been implicated as a CD4+ T cell clonal anergy factor. By RNA-sequencing, we identified Ndrg1 as the third most upregulated gene in anergic, compared to naïve follicular, B cells. Ndrg1 is upregulated by B cell receptor activation (signal one) and suppressed by co-stimulation (signal two), suggesting that NDRG1 may be important in B cell tolerance. However, though Ndrg1-/- mice have a neurological defect mimicking NDRG1-associated Charcot-Marie-Tooth (CMT4d) disease, primary and secondary immune responses were normal. We find that B cell tolerance is maintained, and NDRG1 does not play a role in downstream responses during re-stimulation of in vivo antigen-experienced CD4+ T cells, demonstrating that NDGR1 is functionally redundant for lymphocyte anergy.
Collapse
Affiliation(s)
- Rose Hodgson
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Xijin Xu
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Consuelo Anzilotti
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mukta Deobagkar-Lele
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Tanya L Crockford
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Jessica D Kepple
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Eleanor Cawthorne
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Aneesha Bhandari
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Alberto Cebrian-Serrano
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Martin J Wilcock
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Benjamin Davies
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Richard J Cornall
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Katherine R Bull
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
16
|
Kasmani MY, Ciecko AE, Brown AK, Petrova G, Gorski J, Chen YG, Cui W. Autoreactive CD8 T cells in NOD mice exhibit phenotypic heterogeneity but restricted TCR gene usage. Life Sci Alliance 2022; 5:5/10/e202201503. [PMID: 35667687 PMCID: PMC9170949 DOI: 10.26508/lsa.202201503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 11/27/2022] Open
Abstract
Paired scRNA-seq and scTCR-seq reveals that diabetogenic CD8 T cells in the islets and spleens of NOD mice exhibit phenotypic and clonal heterogeneity despite restricted TCR gene usage. Expression of certain TCR genes correlates with clonal proliferation and effector phenotype. Type 1 diabetes (T1D) is an autoimmune disorder defined by CD8 T cell–mediated destruction of pancreatic β cells. We have previously shown that diabetogenic CD8 T cells in the islets of non-obese diabetic mice are phenotypically heterogeneous, but clonal heterogeneity remains relatively unexplored. Here, we use paired single-cell RNA and T-cell receptor sequencing (scRNA-seq and scTCR-seq) to characterize autoreactive CD8 T cells from the islets and spleens of non-obese diabetic mice. scTCR-seq demonstrates that CD8 T cells targeting the immunodominant β-cell epitope IGRP206-214 exhibit restricted TCR gene usage. scRNA-seq identifies six clusters of autoreactive CD8 T cells in the islets and six in the spleen, including memory and exhausted cells. Clonal overlap between IGRP206-214–reactive CD8 T cells in the islets and spleen suggests these cells may circulate between the islets and periphery. Finally, we identify correlations between TCR genes and T-cell clonal expansion and effector fate. Collectively, our work demonstrates that IGRP206-214–specific CD8 T cells are phenotypically heterogeneous but clonally restricted, raising the possibility of selectively targeting these TCR structures for therapeutic benefit.
Collapse
Affiliation(s)
- Moujtaba Y Kasmani
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA.,Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, USA
| | - Ashley E Ciecko
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA.,Max McGee National Research Center for Juvenile Diabetes, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ashley K Brown
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA.,Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, USA
| | - Galina Petrova
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jack Gorski
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA.,Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, USA
| | - Yi-Guang Chen
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA .,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA.,Max McGee National Research Center for Juvenile Diabetes, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Weiguo Cui
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA .,Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
17
|
Cbl-b restrains priming of pathogenic Th17 cells via the inhibition of IL-6 production by macrophages. iScience 2022; 25:105151. [PMID: 36185364 PMCID: PMC9523381 DOI: 10.1016/j.isci.2022.105151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 08/06/2022] [Accepted: 09/12/2022] [Indexed: 11/23/2022] Open
Abstract
E3 ubiquitin ligase Cbl-b is involved in the maintenance of a balance between immunity and tolerance. Mice lacking Cbl-b are highly susceptible to experimental autoimmune encephalomyelitis (EAE), a Th17-mediated autoimmune disease. However, how Cbl-b regulates Th17 cell responses remains unclear. In this study, utilizing adoptive transfer and cell type-specific Cblb knockout strains, we show that Cbl-b expression in macrophages, but not T cells or dendritic cells (DCs), restrains the generation of pathogenic Th17 cells and the development of EAE. Cbl-b inhibits IL-6 production by macrophages that is induced by signaling from CARD9-dependent C-type lectin receptor (CLR) pathways, which directs T cells to generate pathogenic Th17 cells. Therefore, our data unveil a previously unappreciated function for Cbl-b in the regulation of pathogenic Th17 responses. E3 ubiquitin ligase Cbl-b inhibits EAE disease progression Cbl-b dampens pathogenic Th17 response via inhibiting macrophage-derived IL-6 Cbl-b controls macrophage-derived IL-6 via a CARD9-dependent manner
Collapse
|
18
|
Zhang S, Yu F, Che A, Tan B, Huang C, Chen Y, Liu X, Huang Q, Zhang W, Ma C, Qian M, Liu M, Qin J, Du B. Neuroendocrine Regulation of Stress-Induced T Cell Dysfunction during Lung Cancer Immunosurveillance via the Kisspeptin/GPR54 Signaling Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104132. [PMID: 35224894 PMCID: PMC9069377 DOI: 10.1002/advs.202104132] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 02/10/2022] [Indexed: 06/01/2023]
Abstract
Emerging evidence suggests that physiological distress is highly correlated with cancer incidence and mortality. However, the mechanisms underlying psychological challenges-mediated tumor immune evasion are not systematically explored. Here, it is demonstrated that acute restraint (AR) increases the level of the plasma neuropeptide hormones, kisspeptin, and the expression levels of its receptor, Gpr54, in the hypothalamus, splenic and tumor-infiltrating T cells, suggesting a correlation between the neuroendocrine system and tumor microenvironment. Accordingly, administration of kisspeptin-10 significantly impairs T cell function, whereas knockout of Gpr54 in T cells inhibits lung tumor progression by suppressing T cell dysfunction and exhaustion with or without AR. In addition, Gpr54 defective OT-1 T cells show superior antitumor activity against OVA peptide-positive tumors. Mechanistically, ERK5-mediated NR4A1 activation is found to be essential for kisspeptin/GPR54-facilitated T cell dysfunction. Meanwhile, pharmacological inhibition of ERK5 signaling by XMD8-92 significantly reduces the tumor growth by enhancing CD8+ T cell antitumor function. Furthermore, depletion of GPR54 or ERK5 by CRISPR/Cas9 in CAR T cells intensifies the antitumor responses to both PSMA+ and CD19+ tumor cells, while eliminating T cell exhaustion. Taken together, these results indicate that kisspeptin/GPR54 signaling plays a nonredundant role in the stress-induced tumor immune evasion.
Collapse
Affiliation(s)
- Su Zhang
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| | - Fangfei Yu
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| | - Anran Che
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| | | | - Chenshen Huang
- Department of General SurgeryTongji HospitalSchool of MedicineTongji UniversityShanghai200065China
| | - Yuxue Chen
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| | - Xiaohong Liu
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| | - Qi Huang
- Department of General SurgeryTongji HospitalSchool of MedicineTongji UniversityShanghai200065China
| | - Wenying Zhang
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| | - Chengbin Ma
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| | - Min Qian
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| | - Mingyao Liu
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| | - Juliang Qin
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| | - Bing Du
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesChangning Maternity and Infant Health HospitalEast China Normal UniversityShanghai200241China
| |
Collapse
|
19
|
Xu F, Liu C, Dong Y, Wu W, Xu J, Yan Y, Shao Y, Hao C, Yang Y, Zhang J. Ablation of Cbl-b and c-Cbl in dendritic cells causes spontaneous liver cirrhosis via altering multiple properties of CD103 + cDC1s. Cell Death Dis 2022; 8:142. [PMID: 35354799 PMCID: PMC8967913 DOI: 10.1038/s41420-022-00953-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 03/08/2022] [Accepted: 03/17/2022] [Indexed: 12/02/2022]
Abstract
The Casitas B-lineage lymphoma (Cbl) family proteins are E3 ubiquitin ligases implicated in the regulation of various immune cells. However, their function in dendritic cells (DCs) remains unclear. To investigate the role of Cbl family members in DCs, we created dendritic cell double-deficient Casitas B lymphoma-b (Cbl-b) and Casitas B lineage lymphoma (c-Cbl) mice by crossing Cbl-b−/− mice with c-Cblflox/flox CD11c-Cre+ mice. We found that specific deletion of Cbl-b and c-Cbl in CD11c+ cells, predominantly in DCs, led to liver fibrosis, cirrhosis, and accumulation of systemic conventional Type I DCs (cDC1s) due to enhanced cell proliferation and decreased cell apoptosis. In addition to a change in DC number, double knockout (dKO) cDC1s exhibited a partially activated status as indicated by high basal expression levels of certain cytokines and possessed an enhanced capacity to prime T cells. After adoptive transfer, dKO cDC1s could drive liver fibrosis too. In further experiments, we demonstrated that Cbl-b and c-Cbl could target signal transducer and activator of transcription 5 (STAT5), a transcriptional repressor for the pro-apoptotic protein Bim, to promote ubiquitination-mediated degradation and cell apoptosis in cDC1s. Further extensive experiments revealed that Cbl-b mediated K27-linked ubiquitination of lysine 164 of STAT5a while c-Cbl induced K29-linked ubiquitination of lysine 696 of STAT5a and K27-linked ubiquitination of lysine 140 and 694 of STAT5b. Thus, our findings indicate a functional redundancy of Cbl-b and c-Cbl in cDC homeostasis and maturation.
Collapse
Affiliation(s)
- Fei Xu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Chen Liu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Yongli Dong
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Wenyan Wu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Jie Xu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Yunqiu Yan
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Yu Shao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Chuangli Hao
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, People's Republic of China.
| | - Yi Yang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China.
| | - Jinping Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China.
| |
Collapse
|
20
|
Gumber D, Wang LD. Improving CAR-T immunotherapy: Overcoming the challenges of T cell exhaustion. EBioMedicine 2022; 77:103941. [PMID: 35301179 PMCID: PMC8927848 DOI: 10.1016/j.ebiom.2022.103941] [Citation(s) in RCA: 159] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/27/2022] [Accepted: 03/01/2022] [Indexed: 12/15/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has emerged as a cancer treatment with enormous potential, demonstrating impressive antitumor activity in the treatment of hematological malignancies. However, CAR T cell exhaustion is a major limitation to their efficacy, particularly in the application of CAR T cells to solid tumors. CAR T cell exhaustion is thought to be due to persistent antigen stimulation, as well as an immunosuppressive tumor microenvironment, and mitigating exhaustion to maintain CAR T cell effector function and persistence and achieve clinical potency remains a central challenge. Here, we review the underlying mechanisms of exhaustion and discuss emerging strategies to prevent or reverse exhaustion through modifications of the CAR receptor or CAR independent pathways. Additionally, we discuss the potential of these strategies for improving clinical outcomes of CAR T cell therapy.
Collapse
Affiliation(s)
- Diana Gumber
- Irell and Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Beckman Research Institute, Duarte CA, United States; Department of Immunooncology, City of Hope National Medical Center, Beckman Research Institute, Duarte, CA, United States
| | - Leo D Wang
- Irell and Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Beckman Research Institute, Duarte CA, United States; Department of Immunooncology, City of Hope National Medical Center, Beckman Research Institute, Duarte, CA, United States; Department of Pediatrics, City of Hope National Medical Center, Duarte, CA, United States.
| |
Collapse
|
21
|
Lewis EL, Xu R, Beltra JC, Ngiow SF, Cohen J, Telange R, Crane A, Sawinski D, Wherry EJ, Porrett PM. NFAT-dependent and -independent exhaustion circuits program maternal CD8 T cell hypofunction in pregnancy. J Exp Med 2022; 219:e20201599. [PMID: 34882194 PMCID: PMC8666877 DOI: 10.1084/jem.20201599] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/09/2021] [Accepted: 11/18/2021] [Indexed: 11/21/2022] Open
Abstract
Pregnancy is a common immunization event, but the molecular mechanisms and immunological consequences provoked by pregnancy remain largely unknown. We used mouse models and human transplant registry data to reveal that pregnancy induced exhausted CD8 T cells (Preg-TEX), which associated with prolonged allograft survival. Maternal CD8 T cells shared features of exhaustion with CD8 T cells from cancer and chronic infection, including transcriptional down-regulation of ribosomal proteins and up-regulation of TOX and inhibitory receptors. Similar to other models of T cell exhaustion, NFAT-dependent elements of the exhaustion program were induced by fetal antigen in pregnancy, whereas NFAT-independent elements did not require fetal antigen. Despite using conserved molecular circuitry, Preg-TEX cells differed from TEX cells in chronic viral infection with respect to magnitude and dependency of T cell hypofunction on NFAT-independent signals. Altogether, these data reveal the molecular mechanisms and clinical consequences of maternal CD8 T cell hypofunction and identify pregnancy as a previously unappreciated context in which T cell exhaustion may occur.
Collapse
Affiliation(s)
- Emma L. Lewis
- Department of Obstetrics and Gynecology, The University of Pennsylvania, Philadelphia, PA
| | - Rong Xu
- Department of Surgery, The University of Pennsylvania, Philadelphia, PA
| | - Jean-Christophe Beltra
- Department of Systems Pharmacology and Translational Therapeutics, The University of Pennsylvania, Philadelphia, PA
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA
| | - Shin Foong Ngiow
- Department of Systems Pharmacology and Translational Therapeutics, The University of Pennsylvania, Philadelphia, PA
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA
| | - Jordana Cohen
- Department of Medicine, The University of Pennsylvania, Philadelphia, PA
| | - Rahul Telange
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL
| | - Alexander Crane
- Department of Surgery, The University of Pennsylvania, Philadelphia, PA
| | - Deirdre Sawinski
- Department of Medicine, The University of Pennsylvania, Philadelphia, PA
| | - E. John Wherry
- Department of Systems Pharmacology and Translational Therapeutics, The University of Pennsylvania, Philadelphia, PA
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA
| | - Paige M. Porrett
- Department of Surgery, The University of Pennsylvania, Philadelphia, PA
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL
- Comprehensive Transplant Institute, The University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
22
|
Ashouri JF, Lo W, Nguyen TTT, Shen L, Weiss A. ZAP70, too little, too much can lead to autoimmunity*. Immunol Rev 2021; 307:145-160. [PMID: 34923645 PMCID: PMC8986586 DOI: 10.1111/imr.13058] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/05/2021] [Indexed: 12/21/2022]
Abstract
Establishing both central and peripheral tolerance requires the appropriate TCR signaling strength to discriminate self‐ from agonist‐peptide bound to self MHC molecules. ZAP70, a cytoplasmic tyrosine kinase, directly interacts with the TCR complex and plays a central and requisite role in TCR signaling in both thymocytes and peripheral T cells. By studying ZAP70 hypomorphic mutations in mice and humans with a spectrum of hypoactive or hyperactive activities, we have gained insights into mechanisms of central and peripheral tolerance. Interestingly, both hypoactive and hyperactive ZAP70 can lead to the development of autoimmune diseases, albeit through distinct mechanisms. Immature thymocytes and mature T cells rely on normal ZAP70 function to complete their development in the thymus and to modulate T cell responses in the periphery. Hypoactive ZAP70 function compromises key developmental checkpoints required to establish central tolerance, allowing thymocytes with potentially self‐reactive TCRs a greater chance to escape negative selection. Such ‘forbidden clones’ may escape into the periphery and may pose a greater risk for autoimmune disease development since they may not engage negative regulatory mechanisms as effectively. Hyperactive ZAP70 enhances thymic negative selection but some thymocytes will, nonetheless, escape negative selection and have greater sensitivity to weak and self‐ligands. Such cells must be controlled by mechanisms involved in anergy, expansion of Tregs, and upregulation of inhibitory receptors or signaling molecules. However, such potentially autoreactive cells may still be able to escape control by peripheral negative regulatory constraints. Consistent with findings in Zap70 mutants, the signaling defects in at least one ZAP70 substrate, LAT, can also lead to autoimmune disease. By dissecting the similarities and differences among mouse models of patient disease or mutations in ZAP70 that affect TCR signaling strength, we have gained insights into how perturbed ZAP70 function can lead to autoimmunity. Because of our work and that of others on ZAP70, it is likely that perturbations in other molecules affecting TCR signaling strength will be identified that also overcome tolerance mechanisms and cause autoimmunity. Delineating these molecular pathways could lead to the development of much needed new therapeutic targets in these complex diseases.
Collapse
Affiliation(s)
- Judith F. Ashouri
- Department of Medicine Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center University of California, San Francisco San Francisco California USA
| | - Wan‐Lin Lo
- Division of Microbiology and Immunology Department of Pathology University of Utah Salt Lake City Utah USA
| | - Trang T. T. Nguyen
- Department of Medicine Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center University of California, San Francisco San Francisco California USA
| | - Lin Shen
- Department of Medicine Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center University of California, San Francisco San Francisco California USA
| | - Arthur Weiss
- Department of Medicine Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center University of California, San Francisco San Francisco California USA
- Howard Hughes Medical Institute University of California, San Francisco San Francisco California USA
| |
Collapse
|
23
|
Gavali S, Liu J, Li X, Paolino M. Ubiquitination in T-Cell Activation and Checkpoint Inhibition: New Avenues for Targeted Cancer Immunotherapy. Int J Mol Sci 2021; 22:10800. [PMID: 34639141 PMCID: PMC8509743 DOI: 10.3390/ijms221910800] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/15/2022] Open
Abstract
The advent of T-cell-based immunotherapy has remarkably transformed cancer patient treatment. Despite their success, the currently approved immunotherapeutic protocols still encounter limitations, cause toxicity, and give disparate patient outcomes. Thus, a deeper understanding of the molecular mechanisms of T-cell activation and inhibition is much needed to rationally expand targets and possibilities to improve immunotherapies. Protein ubiquitination downstream of immune signaling pathways is essential to fine-tune virtually all immune responses, in particular, the positive and negative regulation of T-cell activation. Numerous studies have demonstrated that deregulation of ubiquitin-dependent pathways can significantly alter T-cell activation and enhance antitumor responses. Consequently, researchers in academia and industry are actively developing technologies to selectively exploit ubiquitin-related enzymes for cancer therapeutics. In this review, we discuss the molecular and functional roles of ubiquitination in key T-cell activation and checkpoint inhibitory pathways to highlight the vast possibilities that targeting ubiquitination offers for advancing T-cell-based immunotherapies.
Collapse
Affiliation(s)
| | | | | | - Magdalena Paolino
- Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital Solna, 17176 Solna, Sweden; (S.G.); (J.L.); (X.L.)
| |
Collapse
|
24
|
Rodriguez RM, Hernández-Fuentes MP, Corte-Iglesias V, Saiz ML, Lozano JJ, Cortazar AR, Mendizabal I, Suarez-Fernandez ML, Coto E, López-Vázquez A, Díaz-Corte C, Aransay AM, López-Larrea C, Suarez-Álvarez B. Defining a Methylation Signature Associated With Operational Tolerance in Kidney Transplant Recipients. Front Immunol 2021; 12:709164. [PMID: 34489960 PMCID: PMC8417883 DOI: 10.3389/fimmu.2021.709164] [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: 05/13/2021] [Accepted: 08/03/2021] [Indexed: 12/27/2022] Open
Abstract
Operational tolerance after kidney transplantation is defined as stable graft acceptance without the need for immunosuppression therapy. However, it is not clear which cellular and molecular pathways are driving tolerance in these patients. We performed genome-wide analysis of DNA methylation in peripheral blood mononuclear cells from kidney transplant recipients with chronic rejection and operational tolerance from the Genetic Analysis of Molecular Biomarkers of Immunological Tolerance (GAMBIT) study. Our results showed that both clinical stages diverge in 2737 genes, indicating that each one has a specific methylation signature associated with transplant outcome. We also observed that tolerance is associated with demethylation in genes involved in immune function, including B and T cell activation and Th17 differentiation, while in chronic rejection it is associated with intracellular signaling and ubiquitination pathways. Using co-expression network analysis, we selected 12 genomic regions that are specifically hypomethylated or hypermethylated in tolerant patients. Analysis of these genes in transplanted patients with low dose of steroids showed that these have a similar methylation signature to that of tolerant recipients. Overall, these results demonstrate that methylation analysis can mirror the immune status associated with transplant outcome and provides a starting point for understanding the epigenetic mechanisms associated with tolerance.
Collapse
Affiliation(s)
- Ramon M Rodriguez
- Translation Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias-ISPA, Oviedo, Spain.,Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Lipids in Human Pathology, Institut d'Investigació Sanitària Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), Palma, Spain
| | - María P Hernández-Fuentes
- MRC Centre for Transplantation, King's Health Partners, Guy's Hospital, King's College London, London, United Kingdom
| | - Viviana Corte-Iglesias
- Translation Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias-ISPA, Oviedo, Spain
| | - María Laura Saiz
- Translation Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias-ISPA, Oviedo, Spain
| | - Juan José Lozano
- Bioinformatics Platform, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Ana R Cortazar
- Genome Analysis Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Derio, Spain
| | - Isabel Mendizabal
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | | | - Eliecer Coto
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Genética Molecular, Hospital Universitario Central Asturias, Oviedo, Spain
| | - Antonio López-Vázquez
- Translation Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias-ISPA, Oviedo, Spain.,Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Immunology Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Carmen Díaz-Corte
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Nephrology Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Ana M Aransay
- Genome Analysis Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Derio, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Carlos López-Larrea
- Translation Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias-ISPA, Oviedo, Spain.,Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Immunology Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Beatriz Suarez-Álvarez
- Translation Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias-ISPA, Oviedo, Spain.,Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
25
|
Patel T, Henrickson SE, Moser EK, Field NS, Maurer K, Dawany N, Conrad M, Bunin N, Freedman JL, Heimall J, Arnold DE, Wang J, Markowitz JE, Payne-Poff SB, Williams KW, Russo PA, Wherry EJ, Devoto M, Oliver P, Sullivan KE, Kelsen JR. Immune Dysregulation in Human ITCH Deficiency Successfully Treated with Hematopoietic Cell Transplantation. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2021; 9:2885-2893.e3. [PMID: 33894394 PMCID: PMC9053103 DOI: 10.1016/j.jaip.2021.04.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 02/24/2021] [Accepted: 04/03/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Mutations in ITCH, which encodes an E3 ubiquitin-protein ligase, can result in systemic autoimmunity and immunodeficiency. The clinical phenotype and mechanism of disease have not been fully characterized, resulting in a paucity of therapeutic options for this potentially fatal disease. OBJECTIVE We aimed to (1) expand the understanding about the phenotype of human ITCH deficiency (2) further characterize the associated immune dysregulation, and (3) report the first successful hematopoietic cell transplant (HCT) in a patient with ITCH deficiency. METHODS Disease profiling was performed in a patient with multisystem immune dysregulation. Whole exome sequencing with trio analysis and functional validation of candidate disease variants were performed, including mRNA and protein expression. Analyses to further delineate the immunophenotype included quantitative evaluation of lymphoid and myeloid subsets with flow cytometry and mass cytometry. RESULTS A patient with multisystem immune dysregulation presenting with growth failure, very-early-onset inflammatory bowel disease, arthritis, uveitis, psoriasis, and type 1 diabetes mellitus underwent whole exome sequencing, which identified novel compound heterozygous mutations in ITCH. Reduced expression of ITCH mRNA and absent ITCH protein were found. Abnormalities in both lymphoid and myeloid lineages were identified. The patient underwent HCT. He demonstrated excellent immune reconstitution and resolution of many manifestations of his systemic disease. CONCLUSIONS Here we report ITCH deficiency with unique clinical features of colonic very-early-onset inflammatory bowel disease, arthritis, and uveitis in the setting of immune dysregulation and further characterize the underlying immune dysregulation. We demonstrate that HCT can be an effective, and potentially curative, therapy for ITCH deficiency.
Collapse
Affiliation(s)
- Trusha Patel
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pa; Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pa.
| | - Sarah E Henrickson
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pa; Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Emily K Moser
- Division of Pulmonary Critical Care and Sleep Medicine, University of Florida, Gainesville, Fla; Division of Protective Immunity, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Natania S Field
- Division of Protective Immunity, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Kelly Maurer
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Noor Dawany
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Maire Conrad
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pa; Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pa
| | - Nancy Bunin
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pa; Blood and Marrow Transplant Section, Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Jason L Freedman
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pa; Blood and Marrow Transplant Section, Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Jennifer Heimall
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pa; Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Danielle E Arnold
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Jing Wang
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, Pa; Division of Anatomic Pathology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Jonathan E Markowitz
- Pediatric Gastroenterology, Prisma Health Children's Hospital Upstate, Greenville, SC; University of South Carolina School of Medicine-Greenville, Greenville, SC
| | - Sarah Beth Payne-Poff
- University of South Carolina School of Medicine-Greenville, Greenville, SC; Pediatric Rheumatology, Prisma Health Children's Hospital Upstate, Greenville, SC
| | - Kelli W Williams
- Division of Pediatric Pulmonology, Allergy and Immunology, Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Pierre A Russo
- Division of Anatomic Pathology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pa; Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pa
| | - E John Wherry
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Pa; Institute for Immunology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pa
| | - Marcella Devoto
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pa; Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pa; Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Paula Oliver
- Division of Protective Immunity, Children's Hospital of Philadelphia, Philadelphia, Pa; Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pa
| | - Kathleen E Sullivan
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pa; Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Judith R Kelsen
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pa; Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pa
| |
Collapse
|
26
|
Bevington SL, Ng STH, Britton GJ, Keane P, Wraith DC, Cockerill PN. Chromatin Priming Renders T Cell Tolerance-Associated Genes Sensitive to Activation below the Signaling Threshold for Immune Response Genes. Cell Rep 2021; 31:107748. [PMID: 32521273 PMCID: PMC7296351 DOI: 10.1016/j.celrep.2020.107748] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/20/2020] [Accepted: 05/18/2020] [Indexed: 12/13/2022] Open
Abstract
Immunological homeostasis in T cells is maintained by a tightly regulated signaling and transcriptional network. Full engagement of effector T cells occurs only when signaling exceeds a critical threshold that enables induction of immune response genes carrying an epigenetic memory of prior activation. Here we investigate the underlying mechanisms causing the suppression of normal immune responses when T cells are rendered anergic by tolerance induction. By performing an integrated analysis of signaling, epigenetic modifications, and gene expression, we demonstrate that immunological tolerance is established when both signaling to and chromatin priming of immune response genes are weakened. In parallel, chromatin priming of immune-repressive genes becomes boosted, rendering them sensitive to low levels of signaling below the threshold needed to activate immune response genes. Our study reveals how repeated exposure to antigens causes an altered epigenetic state leading to T cell anergy and tolerance, representing a basis for treating auto-immune diseases. Activation of immune response genes is suppressed in tolerant T cells Epigenetic priming of repressive genes is boosted when tolerance is established Inhibitory receptor genes have a lower threshold of activation in tolerant cells Induction of tolerance by peptides points toward a therapy for multiple sclerosis
Collapse
Affiliation(s)
- Sarah L Bevington
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Sky T H Ng
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Graham J Britton
- Precision Immunology Institute and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Peter Keane
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - David C Wraith
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | - Peter N Cockerill
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
27
|
Hu LB, Hu XQ, Zhang Q, You QD, Jiang ZY. An affinity prediction approach for the ligand of E3 ligase Cbl-b and an insight into substrate binding pattern. Bioorg Med Chem 2021; 38:116130. [PMID: 33848699 DOI: 10.1016/j.bmc.2021.116130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 10/21/2022]
Abstract
Protein-protein interactions (PPIs) are essentially fundamental to all cellular processes, so that developing small molecule inhibitors of PPIs have great significance despite representing a huge challenge. Studying PPIs with the help of peptide motifs could obtain the structural information and reference significance to reduce the difficulty in the development of small molecules. Computational methods are powerful tools to characterize peptide-protein interactions, especially molecular dynamics simulation and binding free energy calculation. Here, we established an affinity prediction model suitable for Casitas B lymphoma-b (Cbl-b) and phosphorylated motif system. According to the affinity data set of multiple truncated peptides, the force field, solvent model, and internal dielectric constant of molecular mechanics/generalized Born surface area (MM/GBSA) method were optimized. Further, we predicted the affinity of the rationally designed new sequences through this model and obtained a new 6-mer motif with a 7-fold increase in affinity and the comprehensive structure-activity relationship. Moreover, we proposed an insight of unexpected activity of the truncated 5-mer peptide and revealed the possible binding mode of the new highly active 6-mer motif by extended simulation. Our results showed that the activity enhancement of the truncated peptide was caused by the acetyl-mediated conformation change. The side chain of Arg and pTyr in the 6-mer motif co-occupied the site p1 to form numerous hydrogen bond interactions and increased hydrophobic interaction formed with Tyr266, leading to the higher affinity. The present work provided a reference to investigate the PPI of Cbl-b and phosphorylated substrates and guided the development of Cbl-b inhibitors.
Collapse
Affiliation(s)
- Lv-Bin Hu
- State Key Laboratory of Natural Medicines, and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiu-Qi Hu
- State Key Laboratory of Natural Medicines, and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qiong Zhang
- State Key Laboratory of Natural Medicines, and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qi-Dong You
- State Key Laboratory of Natural Medicines, and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Zheng-Yu Jiang
- State Key Laboratory of Natural Medicines, and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
28
|
Nguyen TTT, Wang ZE, Shen L, Schroeder A, Eckalbar W, Weiss A. Cbl-b deficiency prevents functional but not phenotypic T cell anergy. J Exp Med 2021; 218:212089. [PMID: 33974042 PMCID: PMC8117209 DOI: 10.1084/jem.20202477] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/05/2021] [Accepted: 04/12/2021] [Indexed: 11/29/2022] Open
Abstract
T cell anergy is an important peripheral tolerance mechanism. We studied how T cell anergy is established using an anergy model in which the Zap70 hypermorphic mutant W131A is coexpressed with the OTII TCR transgene (W131AOTII). Anergy was established in the periphery, not in the thymus. Contrary to enriched tolerance gene signatures and impaired TCR signaling in mature peripheral CD4 T cells, CD4SP thymocytes exhibited normal TCR signaling in W131AOTII mice. Importantly, the maintenance of T cell anergy in W131AOTII mice required antigen presentation via MHC-II. We investigated the functional importance of the inhibitory receptor PD-1 and the E3 ubiquitin ligases Cbl-b and Grail in this model. Deletion of each did not affect expression of phenotypic markers of anergic T cells or T reg numbers. However, deletion of Cbl-b, but not Grail or PD-1, in W131AOTII mice restored T cell responsiveness and signaling. Thus, Cbl-b plays an essential role in the establishment and/or maintenance of unresponsiveness in T cell anergy.
Collapse
Affiliation(s)
- Trang T T Nguyen
- Russell/Engleman Rheumatology Research Center, Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Zhi-En Wang
- Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Lin Shen
- Russell/Engleman Rheumatology Research Center, Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Andrew Schroeder
- Division of Transplant Surgery, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Walter Eckalbar
- Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Arthur Weiss
- Russell/Engleman Rheumatology Research Center, Department of Medicine, University of California, San Francisco, San Francisco, CA.,Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
29
|
Guo X, Mahlakõiv T, Ye Q, Somanchi S, He S, Rana H, DiFiglia A, Gleason J, van der Touw W, Hariri R, Zhang X. CBLB ablation with CRISPR/Cas9 enhances cytotoxicity of human placental stem cell-derived NK cells for cancer immunotherapy. J Immunother Cancer 2021; 9:e001975. [PMID: 33741730 PMCID: PMC7986888 DOI: 10.1136/jitc-2020-001975] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Tumors often develop resistance to surveillance by endogenous immune cells, which include natural killer (NK) cells. Ex vivo activated and/or expanded NK cells demonstrate cytotoxicity against various tumor cells and are promising therapeutics for adoptive cancer immunotherapy. Genetic modification can further enhance NK effector cell activity or activation sensitization. Here, we evaluated the effect of the genetic deletion of ubiquitin ligase Casitas B-lineage lymphoma pro-oncogene-b (CBLB), a negative regulator of lymphocyte activity, on placental CD34+ cell-derived NK (PNK) cell cytotoxicity against tumor cells. METHODS Using CRISPR/Cas9 technology, CBLB was knocked out in placenta-derived CD34+ hematopoietic stem cells, followed by differentiation into PNK cells. Cell expansion, phenotype and cytotoxicity against tumor cells were characterized in vitro. The antitumor efficacy of CBLB knockout (KO) PNK cells was tested in an acute myeloid leukemia (HL-60) tumor model in NOD-scid IL2R gammanull (NSG) mice. PNK cell persistence, biodistribution, proliferation, phenotype and antitumor activity were evaluated. RESULTS 94% of CBLB KO efficacy was achieved using CRISPR/Cas9 gene editing technology. CBLB KO placental CD34+ cells differentiated into PNK cells with high cell yield and >90% purity determined by CD56+ CD3- cell identity. Ablation of CBLB did not impact cell proliferation, NK cell differentiation or phenotypical characteristics of PNK cells. When compared with the unmodified PNK control, CBLB KO PNK cells exhibited higher cytotoxicity against a range of liquid and solid tumor cell lines in vitro. On infusion into busulfan-conditioned NSG mice, CBLB KO PNK cells showed in vivo proliferation and maturation as evidenced by increased expression of CD16, killer Ig-like receptors and NKG2A over 3 weeks. Additionally, CBLB KO PNK cells showed greater antitumor activity in a disseminated HL60-luciferase mouse model compared with unmodified PNK cells. CONCLUSION CBLB ablation increased PNK cell effector function and proliferative capacity compared with non-modified PNK cells. These data suggest that targeting CBLB may offer therapeutic advantages via enhancing antitumor activities of NK cell therapies.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/deficiency
- Adaptor Proteins, Signal Transducing/genetics
- Animals
- Antigens, CD34/metabolism
- CRISPR-Associated Protein 9/genetics
- CRISPR-Associated Protein 9/metabolism
- CRISPR-Cas Systems
- Clustered Regularly Interspaced Short Palindromic Repeats
- Coculture Techniques
- Cytotoxicity, Immunologic
- Female
- GPI-Linked Proteins/metabolism
- Gene Knockout Techniques
- HL-60 Cells
- Humans
- Immunotherapy, Adoptive
- K562 Cells
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Killer Cells, Natural/transplantation
- Mice, Inbred NOD
- Mice, SCID
- NK Cell Lectin-Like Receptor Subfamily C/metabolism
- Neoplasms/immunology
- Neoplasms/metabolism
- Neoplasms/therapy
- Phenotype
- Placenta/cytology
- Pregnancy
- Proto-Oncogene Proteins c-cbl/deficiency
- Proto-Oncogene Proteins c-cbl/genetics
- Receptors, IgG/metabolism
- Stem Cells/immunology
- Stem Cells/metabolism
- Xenograft Model Antitumor Assays
- Mice
Collapse
Affiliation(s)
- Xuan Guo
- Celularity Inc, Florham Park, New Jersey, USA
| | | | - Qian Ye
- Celularity Inc, Florham Park, New Jersey, USA
| | | | - Shuyang He
- Celularity Inc, Florham Park, New Jersey, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Lu T, Chen L, Mansour AG, Yu MJ, Brooks N, Teng KY, Li Z, Zhang J, Barr T, Yu J, Caligiuri MA. Cbl-b Is Upregulated and Plays a Negative Role in Activated Human NK Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:677-685. [PMID: 33419766 PMCID: PMC8184061 DOI: 10.4049/jimmunol.2000177] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022]
Abstract
The E3 ubiquitin ligase Cbl-b has been characterized as an intracellular checkpoint in T cells; however, the function of Cbl-b in primary human NK cells, an innate immune anti-tumor effector cell, is not well defined. In this study, we show that the expression of Cbl-b is significantly upregulated in primary human NK cells activated by IL-15, IL-2, and the human NK cell-sensitive tumor cell line K562 that lacks MHC class I expression. Pretreatment with JAK or AKT inhibitors prior to IL-15 stimulation reversed Cbl-b upregulation. Downregulation of Cbl-b resulted in significant increases in granzyme B and perforin expression, IFN-γ production, and cytotoxic activity against tumor cells. Collectively, we demonstrate upregulation of Cbl-b and its inhibitory effects in IL-15/IL-2/K562-activated human NK cells, suggesting that Cbl-b plays a negative feedback role in human NK cells.
Collapse
Affiliation(s)
- Ting Lu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010
| | - Li Chen
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010
| | - Anthony G Mansour
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010
| | - Melissa J Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010
| | - Noah Brooks
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010
| | - Kun-Yu Teng
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010
| | - Zhenlong Li
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010
| | - Jianying Zhang
- Department of Computational and Quantitative Medicine, City of Hope National Medical Center, Duarte, CA 91010
| | - Tasha Barr
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA 91010
- Department of Immuno-Oncology, Duarte, CA 91010; and
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010
| | - Michael A Caligiuri
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010;
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA 91010
- Department of Immuno-Oncology, Duarte, CA 91010; and
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010
| |
Collapse
|
31
|
CNOT4 suppresses non-small cell lung cancer progression and is required for effector cytolytic T lymphocytes cell responses to lung cancer cells. Mol Immunol 2021; 132:165-171. [PMID: 33592572 DOI: 10.1016/j.molimm.2020.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/03/2020] [Accepted: 12/11/2020] [Indexed: 12/09/2022]
Abstract
The therapeutic options of non-small cell lung cancer (NSCLC) are limited, although a combination of targeted therapy and immunotherapy is promising. To explore novel targets for immunotherapy, we explored the role of Ccr4-Not transcription complex subunit 4 (CNOT4) in NSCLC. The expression of CNOT4 in tumor tissues was determined by immunohistochemistry staining and western blotting. The cell lines that stably express CNOT4 were established in H1299 and A549 cells. Direct cell counting, MTT assay, and colony formation were used to determine the ability of cell proliferation. Cell apoptosis and cell cycle were next analyzed by PI/Annexin V staining. Cell invasion and migration were examined by transwell assays. To further explore the function of CNOT4 in cytotoxic T lymphocytes (CTLs) mediated cytotoxicity, an in vitro co-culture system of CNOT4 overexpressing and control H1299 cells with CTLs was developed. CNOT4 was down-regulated in tumor tissues compared with paired normal tissues from patients with lung cancers. CNOT4 overexpression significantly inhibited tumor cell proliferation, colony formation, cell migration, and invasion, but promoted cell apoptosis. Furthermore, overexpression of CNOT4 enhanced cytotoxicity of CTLs to H1299. CNOT4 functions as a potential tumor suppressor of NSCLC via inhibiting tumor cell function and increasing the sensitivity to CTLs.
Collapse
|
32
|
Trendel N, Kruger P, Gaglione S, Nguyen J, Pettmann J, Sontag ED, Dushek O. Perfect adaptation of CD8 + T cell responses to constant antigen input over a wide range of affinities is overcome by costimulation. Sci Signal 2021; 14:eaay9363. [PMID: 34855472 PMCID: PMC7615691 DOI: 10.1126/scisignal.aay9363] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Reduced T cell responses by contrast antigen stimulation can be rescued by signals from costimulatory receptors.
Collapse
Affiliation(s)
- Nicola Trendel
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, Oxford, UK
| | - Philipp Kruger
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, Oxford, UK
| | - Stephanie Gaglione
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, Oxford, UK
| | - John Nguyen
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, Oxford, UK
| | - Johannes Pettmann
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, Oxford, UK
| | - Eduardo D Sontag
- Electrical and Computer Engineering & Bioengineering, Northeastern University, USA
| | - Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, Oxford, UK
| |
Collapse
|
33
|
Jafari D, Mousavi MJ, Keshavarz Shahbaz S, Jafarzadeh L, Tahmasebi S, Spoor J, Esmaeilzadeh A. E3 ubiquitin ligase Casitas B lineage lymphoma-b and its potential therapeutic implications for immunotherapy. Clin Exp Immunol 2021; 204:14-31. [PMID: 33306199 DOI: 10.1111/cei.13560] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/17/2020] [Accepted: 12/02/2020] [Indexed: 12/25/2022] Open
Abstract
The distinction of self from non-self is crucial to prevent autoreactivity and ensure protection from infectious agents and tumors. Maintaining the balance between immunity and tolerance of immune cells is strongly controlled by several sophisticated regulatory mechanisms of the immune system. Among these, the E3 ligase ubiquitin Casitas B cell lymphoma-b (Cbl-b) is a newly identified component in the ubiquitin-dependent protein degradation system, which is thought to be an important negative regulator of immune cells. An update on the current knowledge and new concepts of the relevant immune homeostasis program co-ordinated by Cbl-b in different cell populations could pave the way for future immunomodulatory therapies of various diseases, such as autoimmune and allergic diseases, infections, cancers and other immunopathological conditions. In the present review, the latest findings are comprehensively summarized on the molecular structural basis of Cbl-b and the suppressive signaling mechanisms of Cbl-b in physiological and pathological immune responses, as well as its emerging potential therapeutic implications for immunotherapy in animal models and human diseases.
Collapse
Affiliation(s)
- D Jafari
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.,Immunotherapy Research and Technology Group, Zanjan University of Medical Sciences, Zanjan, Iran
| | - M J Mousavi
- Department of Hematology, Faculty of Allied medicine, Bushehr University of Medical Sciences, Bushehr, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - S Keshavarz Shahbaz
- Department of Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - L Jafarzadeh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - S Tahmasebi
- Department of Immunology, School of public health, Tehran University of Medical Sciences, Tehran, Iran
| | - J Spoor
- Erasmus University Medical Centre, Erasmus University Rotterdam, Rotterdam, the Netherlands
| | - A Esmaeilzadeh
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.,Immunotherapy Research and Technology Group, Zanjan University of Medical Sciences, Zanjan, Iran.,Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
34
|
You D, Hillerman S, Locke G, Chaudhry C, Stromko C, Murtaza A, Fan Y, Koenitzer J, Chen Y, Briceno S, Bhadra R, Duperret E, Gullo-Brown J, Gao C, Zhao D, Feder J, Curtin J, Degnan AP, Kumi G, Wittman M, Johnson BM, Parrish KE, Gokulrangan G, Morrison J, Quigley M, Hunt JT, Salter-Cid L, Lees E, Sanjuan MA, Liu J. Enhanced antitumor immunity by a novel small molecule HPK1 inhibitor. J Immunother Cancer 2021; 9:jitc-2020-001402. [PMID: 33408094 PMCID: PMC7789447 DOI: 10.1136/jitc-2020-001402] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2020] [Indexed: 12/13/2022] Open
Abstract
Background Hematopoietic progenitor kinase 1 (HPK1 or MAP4K1) has been demonstrated as a negative intracellular immune checkpoint in mediating antitumor immunity in studies with HPK1 knockout and kinase dead mice. Pharmacological inhibition of HPK1 is desirable to investigate the role of HPK1 in human immune cells with therapeutic implications. However, a significant challenge remains to identify a small molecule inhibitor of HPK1 with sufficient potency, selectivity, and other drug-like properties suitable for proof-of-concept studies. In this report, we identified a novel, potent, and selective HPK1 small molecule kinase inhibitor, compound K (CompK). A series of studies were conducted to investigate the mechanism of action of CompK, aiming to understand its potential application in cancer immunotherapy. Methods Human primary T cells and dendritic cells (DCs) were investigated with CompK treatment under conditions relevant to tumor microenvironment (TME). Syngeneic tumor models were used to assess the in vivo pharmacology of CompK followed by human tumor interrogation ex vivo. Results CompK treatment demonstrated markedly enhanced human T-cell immune responses under immunosuppressive conditions relevant to the TME and an increased avidity of the T-cell receptor (TCR) to recognize viral and tumor-associated antigens (TAAs) in significant synergy with anti-PD1. Animal model studies, including 1956 sarcoma and MC38 syngeneic models, revealed improved immune responses and superb antitumor efficacy in combination of CompK with anti-PD-1. An elevated immune response induced by CompK was observed with fresh tumor samples from multiple patients with colorectal carcinoma, suggesting a mechanistic translation from mouse model to human disease. Conclusion CompK treatment significantly improved human T-cell functions, with enhanced TCR avidity to recognize TAAs and tumor cytolytic activity by CD8+ T cells. Additional benefits include DC maturation and priming facilitation in tumor draining lymph node. CompK represents a novel pharmacological agent to address cancer treatment resistance.
Collapse
Affiliation(s)
- Dan You
- Oncology Discovery, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | - Stephen Hillerman
- Oncology Discovery, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | - Gregory Locke
- Oncology Discovery, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | - Charu Chaudhry
- Oncology Discovery, Johnson and Johnson Limited, Spring House, Pennsylvania, USA
| | - Caitlyn Stromko
- Oncology Discovery, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | - Anwar Murtaza
- Oncology Discovery, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | - Yi Fan
- Oncology Discovery, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | | | - Yali Chen
- Oncology Discovery, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | - Stephanie Briceno
- Oncology Discovery, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | | | | | | | - Chan Gao
- Oncology Discovery, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | - Dandan Zhao
- Oncology Discovery, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | - John Feder
- Oncology Discovery, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | - Joshua Curtin
- Oncology Discovery, Johnson and Johnson Limited, Spring House, Pennsylvania, USA
| | - Andrew P Degnan
- Oncology Discovery, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | - Godwin Kumi
- Oncology Discovery, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | - Mark Wittman
- Oncology Discovery, Bristol-Myers Squibb Co, Cambridge, Massachusetts, USA
| | - Benjamin M Johnson
- Oncology Discovery, Bristol-Myers Squibb Co, Cambridge, Massachusetts, USA
| | - Karen E Parrish
- Oncology Discovery, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | | | - John Morrison
- Oncology Discovery, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | - Michael Quigley
- Oncology Discovery, Gilead Sciences Inc, Foster City, California, USA
| | - John T Hunt
- Oncology Discovery, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | | | - Emma Lees
- Oncology Discovery, Bristol-Myers Squibb Co, Cambridge, Massachusetts, USA
| | - Miguel A Sanjuan
- Oncology Discovery, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | - Jinqi Liu
- Oncology Discovery, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| |
Collapse
|
35
|
Khan M, Zhao Z, Arooj S, Fu Y, Liao G. Soluble PD-1: Predictive, Prognostic, and Therapeutic Value for Cancer Immunotherapy. Front Immunol 2020; 11:587460. [PMID: 33329567 PMCID: PMC7710690 DOI: 10.3389/fimmu.2020.587460] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 10/16/2020] [Indexed: 12/13/2022] Open
Abstract
Programmed death protein 1 (PD-1) interaction with PD-L1 deliver immunosuppressive environment for tumor growth, and its blockade with directed monoclonal antibodies (anti-PD-1/anti-PD-L1) has shown remarkable clinical outcome. Lately, their soluble counterparts, sPD-1 and sPD-L1, have been detected in plasma, and elevated levels have been associated with advanced disease, clinical stages, and worst prognosis for cancer patients. Elevated plasma levels of sPD-L1 have been correlated with worst prognosis in several studies and has displayed a persistent outlook. On the other hand, sPD-1 levels have been inconsistent in their predictive and prognostic ability. Pretherapeutic higher sPD-1 plasma levels have shown to predict advanced disease state and to a lesser extent worst prognosis. Any increase in sPD-1 plasma level post therapeutically have been correlated with improved survival for various cancers. In vitro and in vivo studies have shown sPD-1 ability to bind PD-L1 and PD-L2 and block PD-1/PD-L1 interaction. Local delivery of sPD-1 in cancer tumor microenvironment through local gene therapy have demonstrated an increase in tumor specific CD8+ T cell immunity and tumor growth reduction. It had also exhibited enhancement of T cell immunity induced by vaccination and other gene therapeutic agents. Furthermore, it may also lessen the inhibitory effect of circulating sPD-L1 and enhance the effects of mAb-based immunotherapy. In this review, we highlight various aspects of sPD-1 role in cancer prediction, prognosis, and anti-cancer immunity, as well as, its therapeutic value for local gene therapy or systemic immunotherapy in blocking the PD-1 and PD-L1 checkpoint interactions.
Collapse
Affiliation(s)
- Muhammad Khan
- Department of Radiation Oncology, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China.,Department of Oncology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhihong Zhao
- Department of Nephrology, Shenzhen People's Hospital, Second Clinical Medicine Centre, Jinan University, Shenzhen, China
| | - Sumbal Arooj
- Department of Radiation Oncology, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China.,Department of Biochemistry, University of Sialkot, Sialkot, Pakistan
| | - Yuxiang Fu
- Department of Radiation Oncology, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Guixiang Liao
- Department of Radiation Oncology, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
36
|
Tang J, Tu S, Lin G, Guo H, Yan C, Liu Q, Huang L, Tang N, Xiao Y, Pope RM, Rajaram MVS, Amer AO, Ahmer BM, Gunn JS, Wozniak DJ, Tao L, Coppola V, Zhang L, Langdon WY, Torrelles JB, Lipkowitz S, Zhang J. Sequential ubiquitination of NLRP3 by RNF125 and Cbl-b limits inflammasome activation and endotoxemia. J Exp Med 2020; 217:133674. [PMID: 31999304 PMCID: PMC7144527 DOI: 10.1084/jem.20182091] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 04/26/2019] [Accepted: 12/04/2019] [Indexed: 12/11/2022] Open
Abstract
Aberrant NLRP3 inflammasome activation contributes to the development of endotoxemia. The importance of negative regulation of NLRP3 inflammasomes remains poorly understood. Here, we show that the E3 ubiquitin ligase Cbl-b is essential for preventing endotoxemia induced by a sub-lethal dose of LPS via a caspase-11/NLRP3-dependent manner. Further studies show that NLRP3 undergoes both K63- and K48-linked polyubiquitination. Cbl-b binds to the K63-ubiquitin chains attached to the NLRP3 leucine-rich repeat domain (LRR) via its ubiquitin-associated region (UBA) and then targets NLRP3 at K496 for K48-linked ubiquitination and proteasome-mediated degradation. We also identify RNF125 as an additional E3 ubiquitin ligase that initiates K63-linked ubiquitination of the NLRP3 LRR domain. Therefore, NLRP3 is sequentially ubiquitinated by K63- and K48-linked ubiquitination, thus keeping the NLRP3 inflammasomes in check and restraining endotoxemia.
Collapse
Affiliation(s)
- Juan Tang
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH.,Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Sha Tu
- Department of Pathology, University of Iowa, Iowa City, IA.,Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Guoxin Lin
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH.,Department of Pathology, University of Iowa, Iowa City, IA.,Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Hui Guo
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH.,Department of Pathology, University of Iowa, Iowa City, IA
| | - Chengkai Yan
- Department of Pathology, University of Iowa, Iowa City, IA
| | - Qingjun Liu
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - Ling Huang
- Department of Pathology, University of Iowa, Iowa City, IA
| | - Na Tang
- Department of Pathology, University of Iowa, Iowa City, IA
| | - Yizhi Xiao
- Department of Pathology, University of Iowa, Iowa City, IA
| | - R Marshall Pope
- Proteomics Facility, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Murugesan V S Rajaram
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - Amal O Amer
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - Brian M Ahmer
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - John S Gunn
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - Daniel J Wozniak
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - Lijian Tao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Vincenzo Coppola
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH
| | - Liwen Zhang
- Mass Spectrometry and Proteomics Facility, The Ohio State University, Columbus, OH
| | - Wallace Y Langdon
- School of Biomedical Science, University of Western Australia, Perth, Australia
| | - Jordi B Torrelles
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - Stanley Lipkowitz
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jian Zhang
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH.,Department of Pathology, University of Iowa, Iowa City, IA
| |
Collapse
|
37
|
Seijkens TTP, Poels K, Meiler S, van Tiel CM, Kusters PJH, Reiche M, Atzler D, Winkels H, Tjwa M, Poelman H, Slütter B, Kuiper J, Gijbels M, Kuivenhoven JA, Matic LP, Paulsson-Berne G, Hedin U, Hansson GK, Nicolaes GAF, Daemen MJAP, Weber C, Gerdes N, de Winther MPJ, Lutgens E. Deficiency of the T cell regulator Casitas B-cell lymphoma-B aggravates atherosclerosis by inducing CD8+ T cell-mediated macrophage death. Eur Heart J 2020; 40:372-382. [PMID: 30452556 PMCID: PMC6340101 DOI: 10.1093/eurheartj/ehy714] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 10/15/2018] [Indexed: 12/13/2022] Open
Abstract
Aims The E3-ligase CBL-B (Casitas B-cell lymphoma-B) is an important negative regulator of T cell activation that is also expressed in macrophages. T cells and macrophages mediate atherosclerosis, but their regulation in this disease remains largely unknown; thus, we studied the function of CBL-B in atherogenesis. Methods and results The expression of CBL-B in human atherosclerotic plaques was lower in advanced lesions compared with initial lesions and correlated inversely with necrotic core area. Twenty weeks old Cblb−/−Apoe−/− mice showed a significant increase in plaque area in the aortic arch, where initial plaques were present. In the aortic root, a site containing advanced plaques, lesion area rose by 40%, accompanied by a dramatic change in plaque phenotype. Plaques contained fewer macrophages due to increased apoptosis, larger necrotic cores, and more CD8+ T cells. Cblb−/−Apoe−/− macrophages exhibited enhanced migration and increased cytokine production and lipid uptake. Casitas B-cell lymphoma-B deficiency increased CD8+ T cell numbers, which were protected against apoptosis and regulatory T cell-mediated suppression. IFNγ and granzyme B production was enhanced in Cblb−/−Apoe−/− CD8+ T cells, which provoked macrophage killing. Depletion of CD8+ T cells in Cblb−/−Apoe−/− bone marrow chimeras rescued the phenotype, indicating that CBL-B controls atherosclerosis mainly through its function in CD8+ T cells. Conclusion Casitas B-cell lymphoma-B expression in human plaques decreases during the progression of atherosclerosis. As an important regulator of immune responses in experimental atherosclerosis, CBL-B hampers macrophage recruitment and activation during initial atherosclerosis and limits CD8+ T cell activation and CD8+ T cell-mediated macrophage death in advanced atherosclerosis, thereby preventing the progression towards high-risk plaques. ![]()
Collapse
Affiliation(s)
- Tom T P Seijkens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Pettenkoferstraße 8a & 9, Munich, Germany
| | - Kikkie Poels
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Svenja Meiler
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Claudia M van Tiel
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Pascal J H Kusters
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Myrthe Reiche
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Dorothee Atzler
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Pettenkoferstraße 8a & 9, Munich, Germany.,Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Goethestraße 33D, Munich, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Pettenkoferstraße 8a & 9, Munich, Germany
| | - Holger Winkels
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Pettenkoferstraße 8a & 9, Munich, Germany
| | - Marc Tjwa
- Laboratory of Vascular Hematology/Angiogenesis, Institute for Transfusion Medicine, Goethe University Frankfurt, Sandhofstraße 1D, Germany
| | - Hessel Poelman
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER, Maastricht University, Maastricht, the Netherlands
| | - Bram Slütter
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einstein weg 55, 2333 CC, Leiden, the Netherlands
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einstein weg 55, 2333 CC, Leiden, the Netherlands
| | - Marion Gijbels
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Jan Albert Kuivenhoven
- Department of Pediatrics, Section Molecular Genetics, University of Groningen, University Medical Center Groningen, Postbus 72, AB Groningen, The Netherlands
| | - Ljubica Perisic Matic
- Department of Molecular Medicine and Surgery, Karolinska University Hospital, Karolinska Institutet, Solna, SE-171 76, Stockholm, Sweden
| | - Gabrielle Paulsson-Berne
- Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Solna SE-171 76 Stockholm, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska University Hospital, Karolinska Institutet, Solna, SE-171 76, Stockholm, Sweden
| | - Göran K Hansson
- Department of Molecular Medicine and Surgery, Karolinska University Hospital, Karolinska Institutet, Solna, SE-171 76, Stockholm, Sweden
| | - Gerry A F Nicolaes
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER, Maastricht University, Maastricht, the Netherlands
| | - Mat J A P Daemen
- Department of Pathology, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Pettenkoferstraße 8a & 9, Munich, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Pettenkoferstraße 8a & 9, Munich, Germany
| | - Norbert Gerdes
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Pettenkoferstraße 8a & 9, Munich, Germany.,Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Moorenstraße 5m 0225 Düsseldorf, Germany
| | - Menno P J de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Pettenkoferstraße 8a & 9, Munich, Germany
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Pettenkoferstraße 8a & 9, Munich, Germany
| |
Collapse
|
38
|
Poels K, Vos WG, Lutgens E, Seijkens TTP. E3 Ubiquitin Ligases as Immunotherapeutic Target in Atherosclerotic Cardiovascular Disease. Front Cardiovasc Med 2020; 7:106. [PMID: 32582770 PMCID: PMC7292335 DOI: 10.3389/fcvm.2020.00106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/20/2020] [Indexed: 12/14/2022] Open
Abstract
Chronic low-grade inflammation drives atherosclerosis and despite optimal pharmacological treatment of classical cardiovascular risk factors, one third of the patients with atherosclerotic cardiovascular disease has elevated inflammatory biomarkers. Additional anti-inflammatory strategies to target this residual inflammatory cardiovascular risk are therefore required. T-cells are a dominant cell type in human atherosclerotic lesions. Modulation of T-cell activation is therefore a potential strategy to target inflammation in atherosclerosis. Ubiquitination is an important regulatory mechanism of T-cell activation and several E3 ubiquitin ligases, including casitas B-lineage lymphoma proto-oncogene B (Cbl-B), itchy homolog (Itch), and gene related to anergy in lymphocytes (GRAIL), function as a natural brake on T-cell activation. In this review we discuss recent insights on the role of Cbl-B, Itch, and GRAIL in atherosclerosis and explore the therapeutic potential of these E3 ubiquitin ligases in cardiovascular medicine.
Collapse
Affiliation(s)
- Kikkie Poels
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Winnie G Vos
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian's University, Munich, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Tom T P Seijkens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Internal Medicine, Amsterdam UMC, Location VUmc, VU University, Amsterdam, Netherlands.,Department of Hematology, Amsterdam UMC, Location VUmc, VU University, Amsterdam, Netherlands
| |
Collapse
|
39
|
CD5 signalosome coordinates antagonist TCR signals to control the generation of Treg cells induced by foreign antigens. Proc Natl Acad Sci U S A 2020; 117:12969-12979. [PMID: 32434911 DOI: 10.1073/pnas.1917182117] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
CD5 is characterized as an inhibitory coreceptor with an important regulatory role during T cell development. The molecular mechanism by which CD5 operates has been puzzling and its function in mature T cells suggests promoting rather than repressing effects on immune responses. Here, we combined quantitative mass spectrometry and genetic studies to analyze the components and the activity of the CD5 signaling machinery in primary T cells. We found that T cell receptor (TCR) engagement induces the selective phosphorylation of CD5 tyrosine 429, which serves as a docking site for proteins with adaptor functions (c-Cbl, CIN85, CRKL), connecting CD5 to positive (PI3K) and negative (UBASH3A, SHIP1) regulators of TCR signaling. c-CBL acts as a coordinator in this complex enabling CD5 to synchronize positive and negative feedbacks on TCR signaling through the other components. Disruption of CD5 signalosome in mutant mice reveals that it modulates TCR signal outputs to selectively repress the transactivation of Foxp3 and limit the inopportune induction of peripherally induced regulatory T cells during immune responses against foreign antigen. Our findings bring insights into the paradigm of coreceptor signaling, suggesting that, in addition to providing dualistic enhancing or dampening inputs, coreceptors can engage concomitant stimulatory and inhibitory signaling events, which act together to promote specific functional outcomes.
Collapse
|
40
|
Matson CA, Singh NJ. Manipulating the TCR signaling network for cellular immunotherapy: Challenges & opportunities. Mol Immunol 2020; 123:64-73. [PMID: 32422416 DOI: 10.1016/j.molimm.2020.04.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 02/24/2020] [Accepted: 04/11/2020] [Indexed: 02/06/2023]
Abstract
T cells can help confer protective immunity by eliminating infections and tumors or drive immunopathology by damaging host cells. Both outcomes require a series of steps from the activation of naïve T cells to their clonal expansion, differentiation and migration to tissue sites. In addition to specific recognition of the antigen via the T cell receptor (TCR), multiple accessory signals from costimulatory molecules, cytokines and metabolites also influence each step along the progression of the T cell response. Current efforts to modify effector T cell function in many clinical contexts focus on the latter - which encompass antigen-independent and broad, contextual regulators. Not surprisingly, such approaches are often accompanied by adverse events, as they also affect T cells not relevant to the specific treatment. In contrast, fine tuning T cell responses by precisely targeting antigen-specific TCR signals has the potential to radically alter therapeutic strategies in a focused manner. Development of such approaches, however, requires a better understanding of functioning of the TCR and the biochemical signaling network coupled to it. In this article, we review some of the recent advances which highlight important roles of TCR signals throughout the activation and differentiation of T cells during an immune response. We discuss how, an appreciation of specific signaling modalities and variant ligands that influence the function of the TCR has the potential to influence design principles for the next generation of pharmacologic and cellular therapies, especially in the context of tumor immunotherapies involving adoptive cell transfers.
Collapse
Affiliation(s)
- Courtney A Matson
- Department of Microbiology & Immunology, University of Maryland School of Medicine, 685 W Baltimore St, HSF1, Room 380, Baltimore, MD 21201, United States
| | - Nevil J Singh
- Department of Microbiology & Immunology, University of Maryland School of Medicine, 685 W Baltimore St, HSF1, Room 380, Baltimore, MD 21201, United States.
| |
Collapse
|
41
|
Han S, Chung DC, St Paul M, Liu ZQ, Garcia-Batres C, Elford AR, Tran CW, Chapatte L, Ohashi PS. Overproduction of IL-2 by Cbl-b deficient CD4 + T cells provides resistance against regulatory T cells. Oncoimmunology 2020; 9:1737368. [PMID: 32313719 PMCID: PMC7153846 DOI: 10.1080/2162402x.2020.1737368] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 12/17/2019] [Accepted: 12/26/2019] [Indexed: 12/22/2022] Open
Abstract
Regulatory T cells are integral to the regulation of autoimmune and anti-tumor immune responses. However, several studies have suggested that changes in T cell signaling networks can result in T cells that are resistant to the suppressive effects of regulatory T cells. Here, we investigated the role of Cbl-b, an E3 ubiquitin ligase, in establishing resistance to Treg-mediated suppression. We found that the absence of Cbl-b, a negative regulator of multiple TCR signaling pathways, rendered T cells impartial to Treg suppression by regulating cytokine networks leading to improved anti-tumor immunity despite the presence of Treg cells in the tumor. Specifically, Cbl-b KO CD4+FoxP3− T cells hyper-produced IL-2 and together with IL-2 Rα upregulation served as an essential mechanism to escape suppression by Treg cells. Furthermore, we report that IL-2 serves as the central molecule required for cytokine-induced Treg resistance. Collectively our data emphasize the role of IL-2 as a key mechanism that renders CD4+ T cells resistant to the inhibitory effects of Treg cells.
Collapse
Affiliation(s)
- SeongJun Han
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Douglas C Chung
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Michael St Paul
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Zhe Qi Liu
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Carlos Garcia-Batres
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Alisha R Elford
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Charles W Tran
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Laurence Chapatte
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Pamela S Ohashi
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
42
|
Beck JD, Birtel M, Haefner E, Keil IS, Reidenbach D, Salomon N, Yildiz IG, Diken M. CIMT 2019: report on the 17th Annual Meeting of the Association for Cancer Immunotherapy. Hum Vaccin Immunother 2020; 16:808-815. [PMID: 31584850 PMCID: PMC7227697 DOI: 10.1080/21645515.2019.1675459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Jan David Beck
- TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Matthias Birtel
- TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Erik Haefner
- TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Isabell Sofia Keil
- TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Daniel Reidenbach
- TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Nadja Salomon
- TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Ikra Gizem Yildiz
- TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Mustafa Diken
- TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| |
Collapse
|
43
|
Abstract
There are now 354 inborn errors of immunity (primary immunodeficiency diseases (PIDDs)) with 344 distinct molecular etiologies reported according to the International Union of Immunological Sciences (IUIS) (Clin Gastroenterol Hepatol 11: p. 1050-63, 2013, Semin Gastrointest Dis 8: p. 22-32, 1997, J Clin Immunol 38: p. 96-128, 2018). Using the IUIS document as a reference and cross-checking PubMed ( www.ncbi.nlm.nih.pubmed.gov ), we found that approximately one third of the 354 diseases of impaired immunity have a gastrointestinal component [J Clin Immunol 38: p. 96-128, 2018]. Often, the gastrointestinal symptomatology and pathology is the heralding sign of a PIDD; therefore, it is important to recognize patterns of disease which may manifest along the gastrointestinal tract as a more global derangement of immune function. As such, holistic consideration of immunity is warranted in patients with clinically significant gastrointestinal disease. Here, we discuss the manifold presentations and GI-specific complications of PIDDs which could lead patients to seek advice from a variety of clinician specialists. Often, patients with these medical problems will engage general pediatricians, surgeons, gastroenterologists, rheumatologists, and clinical immunologists among others. Following delineation of the presenting concern, accurate and often molecular diagnosis is imperative and a multi-disciplinary approach warranted for optimal management. In this review, we will summarize the current state of understanding of PIDD gastrointestinal disease involvement. We will do so by focusing upon gastrointestinal disease categories (i.e., inflammatory, diarrhea, nodular lymphoid hyperplasia, liver/biliary tract, structural disease, and oncologic disease) with an intent to aid the healthcare provider who may encounter a patient with an as-yet undiagnosed PIDD who presents initially with a gastrointestinal symptom, sign, or problem.
Collapse
|
44
|
Martinov T, Fife BT. Type 1 diabetes pathogenesis and the role of inhibitory receptors in islet tolerance. Ann N Y Acad Sci 2020; 1461:73-103. [PMID: 31025378 PMCID: PMC6994200 DOI: 10.1111/nyas.14106] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/25/2019] [Accepted: 04/03/2019] [Indexed: 12/15/2022]
Abstract
Type 1 diabetes (T1D) affects over a million Americans, and disease incidence is on the rise. Despite decades of research, there is still no cure for this disease. Exciting beta cell replacement strategies are being developed, but in order for such approaches to work, targeted immunotherapies must be designed. To selectively halt the autoimmune response, researchers must first understand how this response is regulated and which tolerance checkpoints fail during T1D development. Herein, we discuss the current understanding of T1D pathogenesis in humans, genetic and environmental risk factors, presumed roles of CD4+ and CD8+ T cells as well as B cells, and implicated autoantigens. We also highlight studies in non-obese diabetic mice that have demonstrated the requirement for CD4+ and CD8+ T cells and B cells in driving T1D pathology. We present an overview of central and peripheral tolerance mechanisms and comment on existing controversies in the field regarding central tolerance. Finally, we discuss T cell- and B cell-intrinsic tolerance mechanisms, with an emphasis on the roles of inhibitory receptors in maintaining islet tolerance in humans and in diabetes-prone mice, and strategies employed to date to harness inhibitory receptor signaling to prevent or reverse T1D.
Collapse
Affiliation(s)
- Tijana Martinov
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Brian T Fife
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
45
|
Sioud M. Unleashing the Therapeutic Potential of Dendritic and T Cell Therapies Using RNA Interference. Methods Mol Biol 2020; 2115:259-280. [PMID: 32006406 DOI: 10.1007/978-1-0716-0290-4_15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Therapeutic dendritic cell (DC) cancer vaccines work to boost the body's immune system to fight a cancer. Although this type of immunotherapy often leads to the activation of tumor-specfic T cells, clinical responses are fairly low, arguing for the need to improve the design of DC-based vaccines. Recent studies revealed a promising strategy of combining DC vaccines with small interfering RNAs (siRNAs) targeting immunosuppressive signals such as checkpoint receptors. Similarly, incorporating checkpoint siRNA blockers in adoptive T-cell therapy to amplify cytotoxic T lymphocyte responses is now being tested in the clinic. The development of the next generation of cancer immunotherapies using siRNA technology will hopefuly benefit patients with various cancer types including those who did not respond to current therapies. This review highlights the latest advances in RNA interference technology to improve the therapeutic efficacy of DC cancer vaccines and T cell therapy.
Collapse
Affiliation(s)
- Mouldy Sioud
- Department of Immunology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Ullernchausseen 70, Oslo, Norway.
| |
Collapse
|
46
|
Wagle MV, Marchingo JM, Howitt J, Tan SS, Goodnow CC, Parish IA. The Ubiquitin Ligase Adaptor NDFIP1 Selectively Enforces a CD8 + T Cell Tolerance Checkpoint to High-Dose Antigen. Cell Rep 2019; 24:577-584. [PMID: 30021156 DOI: 10.1016/j.celrep.2018.06.060] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 05/17/2018] [Accepted: 06/12/2018] [Indexed: 01/11/2023] Open
Abstract
Escape from peripheral tolerance checkpoints that control cytotoxic CD8+ T cells is important for cancer immunotherapy and autoimmunity, but pathways enforcing these checkpoints are mostly uncharted. We reveal that the HECT-type ubiquitin ligase activator, NDFIP1, enforces a cell-intrinsic CD8+ T cell checkpoint that desensitizes TCR signaling during in vivo exposure to high antigen levels. Ndfip1-deficient OT-I CD8+ T cells responding to high exogenous tolerogenic antigen doses that normally induce anergy aberrantly expanded and differentiated into effector cells that could precipitate autoimmune diabetes in RIP-OVAhi mice. In contrast, NDFIP1 was dispensable for peripheral deletion to low-dose exogenous or pancreatic islet-derived antigen and had little impact upon effector responses to Listeria or acute LCMV infection. These data provide evidence that NDFIP1 mediates a CD8+ T cell tolerance checkpoint, with a different mechanism to CD4+ T cells, and indicates that CD8+ T cell deletion and anergy are molecularly separable checkpoints.
Collapse
Affiliation(s)
- Mayura V Wagle
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia; Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Julia M Marchingo
- Division of Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Jason Howitt
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia; Department of Health and Medical Sciences, Swinburne University, Melbourne, VIC, Australia
| | - Seong-Seng Tan
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Christopher C Goodnow
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia; Garvan Institute of Medical Research, Sydney, NSW, Australia; St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia.
| | - Ian A Parish
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia; Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
| |
Collapse
|
47
|
Beyond the Cell Surface: Targeting Intracellular Negative Regulators to Enhance T cell Anti-Tumor Activity. Int J Mol Sci 2019; 20:ijms20235821. [PMID: 31756921 PMCID: PMC6929154 DOI: 10.3390/ijms20235821] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 02/07/2023] Open
Abstract
It is well established that extracellular proteins that negatively regulate T cell function, such as Cytotoxic T-Lymphocyte-Associated protein 4 (CTLA-4) and Programmed Cell Death protein 1 (PD-1), can be effectively targeted to enhance cancer immunotherapies and Chimeric Antigen Receptor T cells (CAR-T cells). Intracellular proteins that inhibit T cell receptor (TCR) signal transduction, though less well studied, are also potentially useful therapeutic targets to enhance T cell activity against tumor. Four major classes of enzymes that attenuate TCR signaling include E3 ubiquitin kinases such as the Casitas B-lineage lymphoma proteins (Cbl-b and c-Cbl), and Itchy (Itch), inhibitory tyrosine phosphatases, such as Src homology region 2 domain-containing phosphatases (SHP-1 and SHP-2), inhibitory protein kinases, such as C-terminal Src kinase (Csk), and inhibitory lipid kinases such as Src homology 2 (SH2) domain-containing inositol polyphosphate 5-phosphatase (SHIP) and Diacylglycerol kinases (DGKs). This review describes the mechanism of action of eighteen intracellular inhibitory regulatory proteins in T cells within these four classes, and assesses their potential value as clinical targets to enhance the anti-tumor activity of endogenous T cells and CAR-T cells.
Collapse
|
48
|
Suboptimal stimulation by weak agonist epitope variants does not drive dysfunction of HIV-1-specific cytotoxic T lymphocyte clones. AIDS 2019; 33:1565-1574. [PMID: 31306165 DOI: 10.1097/qad.0000000000002259] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To assess whether weakly recognized epitope variants induce anergy in HIV-1-specific CD8 T lymphocyte (CTL) clones as a mechanism of dysfunction. DESIGN HIV-1-specific CTL clones were exposed to suboptimally recognized epitope variants, and screened for anergy and other T-cell dysfunction markers, and subsequent capability to kill target cells bearing index epitope. METHODS In addition to the optimally recognized index epitope, two suboptimally recognized epitope variants were selected based on titration curves for killing of peptide-labeled target cells by three HIV-1-specific CTL clones targeting the epitopes SLYNTVATL (Gag 77-85, A02-restricted), RPAEPVPLQL (Rev 66-75, B07-restricted), and KRWIIMGLNK (Gag 263-272, B27-restricted). Consequences of suboptimal stimulation were assessed by cytokine secretion, gene expression, and capacity to kill index epitope-labeled target cells upon rechallenge. RESULTS Suboptimal recognition of epitope variants reduced cytokine production by CTL similarly to reduction in killing of target cells. Gene expression profiles after suboptimal stimulation demonstrated no patterns consistent with T-cell dysfunction due to anergy, exhaustion, or apoptosis. Preexposure of CTL to epitope variants had no discernable impact on their subsequent capacity to kill index epitope-bearing target cells. CONCLUSION Our data explore the hypothesis that poorly recognized epitope variants not only facilitate HIV-1 evasion of CTL recognition, but also induce CTL dysfunction through suboptimal signaling causing anergy. However, the results do not suggest that suboptimal signaling induces anergy (or exhaustion or apoptosis), indicating that the major role of CTL epitope variation is likely viral escape.
Collapse
|
49
|
Policheni A, Horikawa K, Milla L, Kofler J, Bouillet P, Belz GT, O'Reilly LA, Goodnow CC, Strasser A, Gray DHD. CARD11 is dispensable for homeostatic responses and suppressive activity of peripherally induced FOXP3
+
regulatory T cells. Immunol Cell Biol 2019; 97:740-752. [DOI: 10.1111/imcb.12268] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 05/12/2019] [Accepted: 05/13/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Antonia Policheni
- The Walter and Eliza Hall Institute of Medical Research Melbourne VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| | - Keisuke Horikawa
- Australian Cancer Research Foundation Department of Cancer Biology and Therapeutics The John Curtin School of Medical Research The Australian National University Canberra ACT Australia
| | - Liz Milla
- The Walter and Eliza Hall Institute of Medical Research Melbourne VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| | - Jennifer Kofler
- Australian Cancer Research Foundation Department of Cancer Biology and Therapeutics The John Curtin School of Medical Research The Australian National University Canberra ACT Australia
| | - Philippe Bouillet
- The Walter and Eliza Hall Institute of Medical Research Melbourne VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| | - Gabrielle T Belz
- The Walter and Eliza Hall Institute of Medical Research Melbourne VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| | - Lorraine A O'Reilly
- The Walter and Eliza Hall Institute of Medical Research Melbourne VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| | | | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research Melbourne VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| | - Daniel HD Gray
- The Walter and Eliza Hall Institute of Medical Research Melbourne VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| |
Collapse
|
50
|
Kugyelka R, Prenek L, Olasz K, Kohl Z, Botz B, Glant TT, Berki T, Boldizsár F. ZAP-70 Regulates Autoimmune Arthritis via Alterations in T Cell Activation and Apoptosis. Cells 2019; 8:cells8050504. [PMID: 31137740 PMCID: PMC6562615 DOI: 10.3390/cells8050504] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/21/2019] [Accepted: 05/24/2019] [Indexed: 01/08/2023] Open
Abstract
T cells play an essential role in the pathogenesis of both human rheumatoid arthritis (RA) and its murine models. A key molecule in T cell activation is ZAP-70, therefore we aimed to investigate the effects of partial ZAP-70 deficiency on the pathogenesis of recombinant human G1(rhG1)-induced arthritis (GIA), a well-established mouse model of RA. Arthritis was induced in BALB/c and ZAP-70+/- heterozygous mice. Disease progression was monitored using a scoring system and in vivo imaging, antigen-specific proliferation, cytokine and autoantibody production was measured and T cell apoptotic pathways were analyzed. ZAP-70+/- mice developed a less severe arthritis, as shown by both clinical picture and in vitro parameters (decreased T cell proliferation, cytokine and autoantibody production). The amount of cleaved Caspase-3 increased in arthritic ZAP-70+/- T cells, with no significant changes in cleaved Caspase-8 and -9 levels; although expression of Bim, Bcl-2 and Cytochrome C showed alterations. Tyrosine phosphorylation was less pronounced in arthritic ZAP-70+/- T cells and the amount of Cbl-b-a negative regulator of T cell activation-decreased as well. We hypothesize that the less severe disease seen in the partial absence of ZAP-70 might be caused by the decreased T cell activation accompanied by increased apoptosis.
Collapse
Affiliation(s)
- Réka Kugyelka
- Department of Immunology and Biotechnology, Medical School, University of Pécs, H-7624 Pécs, Hungary.
| | - Lilla Prenek
- Department of Immunology and Biotechnology, Medical School, University of Pécs, H-7624 Pécs, Hungary.
| | - Katalin Olasz
- Department of Immunology and Biotechnology, Medical School, University of Pécs, H-7624 Pécs, Hungary.
| | - Zoltán Kohl
- Department of Immunology and Biotechnology, Medical School, University of Pécs, H-7624 Pécs, Hungary.
| | - Bálint Botz
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, H-7624 Pécs, Hungary.
- Department of Radiology, Medical School, University of Pécs, H-7624 Pécs, Hungary.
| | - Tibor T Glant
- Department of Molecular Medicine, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Timea Berki
- Department of Immunology and Biotechnology, Medical School, University of Pécs, H-7624 Pécs, Hungary.
| | - Ferenc Boldizsár
- Department of Immunology and Biotechnology, Medical School, University of Pécs, H-7624 Pécs, Hungary.
| |
Collapse
|