1
|
Nair KA, Liu B. Navigating the landscape of the unfolded protein response in CD8 + T cells. Front Immunol 2024; 15:1427859. [PMID: 39026685 PMCID: PMC11254671 DOI: 10.3389/fimmu.2024.1427859] [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: 05/05/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024] Open
Abstract
Endoplasmic reticulum stress occurs due to large amounts of misfolded proteins, hypoxia, nutrient deprivation, and more. The unfolded protein is a complex intracellular signaling network designed to operate under this stress. Composed of three individual arms, inositol-requiring enzyme 1, protein kinase RNA-like ER kinase, and activating transcription factor-6, the unfolded protein response looks to resolve stress and return to proteostasis. The CD8+ T cell is a critical cell type for the adaptive immune system. The unfolded protein response has been shown to have a wide-ranging spectrum of effects on CD8+ T cells. CD8+ T cells undergo cellular stress during activation and due to environmental insults. However, the magnitude of the effects this response has on CD8+ T cells is still understudied. Thus, studying these pathways is important to unraveling the inner machinations of these powerful cells. In this review, we will highlight the recent literature in this field, summarize the three pathways of the unfolded protein response, and discuss their roles in CD8+ T cell biology and functionality.
Collapse
Affiliation(s)
- Keith Alan Nair
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Bei Liu
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| |
Collapse
|
2
|
Li P, Cui P, Yue Q, Xu Z, Liu Z. Exploring the potential biological significance of KDELR family genes in lung adenocarcinoma. Sci Rep 2024; 14:14820. [PMID: 38937522 PMCID: PMC11211404 DOI: 10.1038/s41598-024-65425-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024] Open
Abstract
The Lys-Asp-Glu-Leu receptor (KDELR) family genes play critical roles in a variety of biological processes in different tumors. Our study aimed to provide a comprehensive analysis of the potential roles of KDELRs in lung adenocarcinoma (LUAD). Utilizing data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database, as well as clinical samples, we conducted a series of analyses and validations using R software tools and various online resources. The results showed that KDELR family genes and proteins were highly expressed and associated with a poor prognosis of LUAD. Promoter hypomethylation and the competing endogenous RNA (ceRNA) network of PCAT6/hsa-miR-326/KDELR1 might be potential causes of aberrant KDELR1 overexpression in LUAD. Three key Transcription factors (TFs) (SPI1, EP300, and MAZ) and a TFs-miRNAs-KDELRs network (involving 11 TFs) might be involved in modulating KDELRs expression abnormalities. Gene Set Enrichment Analysis (GSEA) indicated enrichment of genes highly expressing KDELR1, KDELR2, and KDELR3 in MTORC1_SIGNALING, P53_PATHWAY, and ANGIOGENESIS. Negative correlations between KDELRs expression and CD8 + T cell infiltration, as well as CTLA-4 expression. Our multiple analyses suggested that the KDELRs are important signaling molecules in LUAD. These results provided novel insights for developing prognostic markers and novel therapies of LUAD.
Collapse
Affiliation(s)
- Peitong Li
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Pengfei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Qing Yue
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Zijun Xu
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Ziling Liu
- Cancer Center, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
3
|
Zhu M, Fang Z, Wu Y, Dong F, Wang Y, Zheng F, Ma X, Ma S, He J, Liu X, Yao X, Fu C. A KDELR-mediated ER-retrieval system modulates mitochondrial functions via the unfolded protein response in fission yeast. J Biol Chem 2024; 300:105754. [PMID: 38360270 PMCID: PMC10938167 DOI: 10.1016/j.jbc.2024.105754] [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: 11/17/2023] [Revised: 01/23/2024] [Accepted: 02/08/2024] [Indexed: 02/17/2024] Open
Abstract
KDELR (Erd2 [ER retention defective 2] in yeasts) is a receptor protein that retrieves endoplasmic reticulum (ER)-resident proteins from the Golgi apparatus. However, the role of the KDELR-mediated ER-retrieval system in regulating cellular homeostasis remains elusive. Here, we show that the absence of Erd2 triggers the unfolded protein response (UPR) and enhances mitochondrial respiration and reactive oxygen species in an UPR-dependent manner in the fission yeast Schizosaccharomyces pombe. Moreover, we perform transcriptomic analysis and find that the expression of genes related to mitochondrial respiration and the tricarboxylic acid cycle is upregulated in a UPR-dependent manner in cells lacking Erd2. The increased mitochondrial respiration and reactive oxygen species production is required for cell survival in the absence of Erd2. Therefore, our findings reveal a novel role of the KDELR-Erd2-mediated ER-retrieval system in modulating mitochondrial functions and highlight its importance for cellular homeostasis in the fission yeast.
Collapse
Affiliation(s)
- Mengdan Zhu
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Zheng Fang
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Yifan Wu
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Fenfen Dong
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Yuzhou Wang
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Fan Zheng
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Xiaopeng Ma
- Division of Life Sciences and Medicine, Department of General Surgery, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Shisong Ma
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Jiajia He
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Xing Liu
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Xuebiao Yao
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China.
| | - Chuanhai Fu
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
4
|
Zhang C, Xiao J, Fa L, Jiang F, Jiang H, Zhou L, Xu Z. Identification of co-expressed gene networks promoting CD8 + T cell infiltration and having prognostic value in uveal melanoma. BMC Ophthalmol 2023; 23:354. [PMID: 37563735 PMCID: PMC10416479 DOI: 10.1186/s12886-023-03098-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 08/01/2023] [Indexed: 08/12/2023] Open
Abstract
Current immunotherapies are unsatisfactory against uveal melanoma (UM); however, elevated CD8+ T cell infiltration level indicates poor prognosis in UM. Here, we aimed to identify co-expressed gene networks promoting CD8+ T cell infiltration in UM and created a prognostic hazard model based on the identified hub genes. Raw data and clinical information were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Stromal-immune comprehensive score (ESTIMATE) was used to evaluate the immune-infiltration landscape of the tumor microenvironment. Single-Sample Gene Set Enrichment Analysis (ssGSEA) and Weighted Correlation Network Analysis (WGCNA) were used to quantify CD8+ T cell infiltration level and identify hub genes. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed to analyze the biological processes. Least absolute shrinkage and selection operator (LASSO) Cox regression were used to establish a prognostic model, which was further validated. Finally, pan-cancer analysis evaluated these genes to be associated with CD8+ T cell infiltration in other tumors. In conclusion, the proposed four-gene (PTPN12, IDH2, P2RX4, and KDELR2) prognostic hazard model had satisfactory prognostic ability. These hub genes may promote CD8+ T cell infiltration in UM through antigen presentation, and CD8+ T cell possibly function as Treg, resulting in poor prognosis. These findings might facilitate the development of novel immunotherapies.
Collapse
Affiliation(s)
- Chun Zhang
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China
| | - Jing Xiao
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China
| | - Luzhong Fa
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China
| | - Fanwen Jiang
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China
| | - Hui Jiang
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China
| | - Lin Zhou
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China
| | - Zhuping Xu
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China.
| |
Collapse
|
5
|
Liu C, Xu S, Liu Q, Chai H, Luo Y, Li S. Identification of immune cells infiltrating in hippocampus and key genes associated with Alzheimer's disease. BMC Med Genomics 2023; 16:53. [PMID: 36915078 PMCID: PMC10009990 DOI: 10.1186/s12920-023-01458-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 02/13/2023] [Indexed: 03/16/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia and is primarily associated with memory impairment and cognitive decline, but the etiology of AD has not been elucidated. In recent years, evidence has shown that immune cells play critical roles in AD pathology. In the current study, we collected the transcriptomic data of the hippocampus from gene expression omnibus database, and investigated the effect of immune cell infiltration in the hippocampus on AD, and analyzed the key genes that influence the pathogenesis of AD patients. The results revealed that the relative abundance of immune cells in the hippocampus of AD patients was altered. Of all given 28 kinds of immune cells, monocytes were the important immune cell associated with AD. We identified 4 key genes associated with both AD and monocytes, including KDELR1, SPTAN1, CDC16 and RBBP6, and they differentially expressed in 5XFAD mice and WT mice. The logistic regression and random forest models based on the 4 key genes could effectively distinguish AD from healthy samples. Our research provided a new perspective on immunotherapy for AD patients.
Collapse
Affiliation(s)
- Chenming Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, Tongji University School of Medicine, Shanghai, 200092, China
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Sutong Xu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, Tongji University School of Medicine, Shanghai, 200092, China
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Qiulu Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, Tongji University School of Medicine, Shanghai, 200092, China
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Huazhen Chai
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, Tongji University School of Medicine, Shanghai, 200092, China
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Yuping Luo
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, Tongji University School of Medicine, Shanghai, 200092, China.
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Siguang Li
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, Tongji University School of Medicine, Shanghai, 200092, China.
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| |
Collapse
|
6
|
Eggleton P, De Alba J, Weinreich M, Calias P, Foulkes R, Corrigall VM. The therapeutic mavericks: Potent immunomodulating chaperones capable of treating human diseases. J Cell Mol Med 2023; 27:322-339. [PMID: 36651415 PMCID: PMC9889696 DOI: 10.1111/jcmm.17669] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/16/2022] [Accepted: 12/28/2022] [Indexed: 01/19/2023] Open
Abstract
Two major chaperones, calreticulin (CRT) and binding immunoglobulin protein (GRP78/BiP) dependent on their location, have immunoregulatory or anti-inflammatory functions respectively. CRT induces pro-inflammatory cytokines, dendritic cell (DC) maturation and activates cytotoxic T cells against tumours. By contrast, GRP78/BiP induces anti-inflammatory cytokines, inhibits DC maturation and heightens T-regulatory cell responses. These latter functions rebalance immune homeostasis in inflammatory diseases, such as rheumatoid arthritis. Both chaperones are therapeutically relevant agents acting primarily on monocytes/DCs. Endogenous exposure of CRT on cancer cell surfaces acts as an 'eat-me' signal and facilitates improved elimination of stressed and dying tumour cells by DCs. Therefore, therapeutics that promote endogenous CRT translocation to the cell surface can improve the removal of cancer cells. However, infused recombinant CRT dampens this cancer cell eradication by binding directly to the DCs. Low levels of endogenous BiP appear as a surface biomarker of endoplasmic reticulum (ER) stress in some types of tumour cells, a reflection of cells undergoing proliferation, in which resulting hypoxia and nutrient deprivation perturb ER homeostasis triggering the unfolded protein response, leading to increased expression of GRP78/BiP and altered cellular location. Conversely, infusion of an analogue of GRP78/BiP (IRL201805) can lead to long-term immune resetting and restoration of immune homeostasis. The therapeutic potential of both chaperones relies on them being relocated from their intracellular ER environment. Ongoing clinical trials are employing therapeutic interventions to either enhance endogenous cell surface CRT or infuse IRL201805, thereby triggering several disease-relevant immune responses leading to a beneficial clinical outcome.
Collapse
Affiliation(s)
- Paul Eggleton
- Revolo BiotherapeuticsNew OrleansLouisianaUSA,University of Exeter Medical SchoolExeterUK
| | | | | | | | | | - Valerie M. Corrigall
- Revolo BiotherapeuticsNew OrleansLouisianaUSA,Centre for Inflammation Biology and Cancer Immunology, King's College London, New Hunts HouseGuy' HospitalLondonUK
| |
Collapse
|
7
|
Graham PT, Nowak AK, Cornwall SMJ, Larma I, Nelson DJ. The STING agonist, DMXAA, reduces tumor vessels and enhances mesothelioma tumor antigen presentation yet blunts cytotoxic T cell function in a murine model. Front Immunol 2022; 13:969678. [PMID: 36466911 PMCID: PMC9716460 DOI: 10.3389/fimmu.2022.969678] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/20/2022] [Indexed: 08/22/2023] Open
Abstract
We assessed the murine Stimulator of Interferon Genes (STING) agonist, DMXAA, for anti-mesothelioma potential using the AE17-sOVA model that expresses ovalbumin (OVA) as a neo tumor antigen. Dose response experiments alongside testing different routes of administration identified a safe effective treatment regimen that induced 100% cures in mice with small or large tumors. Three doses of 25mg/kg DMXAA given intra-tumorally every 9 days induced tumor regression and long-term survival (>5 months). Re-challenge experiments showed that tumor-free mice developed protective memory. MTT and propidium-iodide assays showed that DMXAA exerted direct cytotoxic effects at doses >1mg/ml on the murine AE17 and AB1 mesothelioma cell lines. In-vivo studies using a CFSE-based in-vivo proliferation assay showed that DMXAA improved tumor-antigen presentation in tumor-draining lymph nodes, evidenced by OVA-specific OT-1 T cells undergoing more divisions. An in-vivo cytotoxic T lymphocyte (CTL) assay showed that DMXAA blunted the lytic quality of CTLs recognizing the dominant (SIINFEKL) and a subdominant (KVVRFDKL) OVA epitopes. DMXAA reduced tumor vessel size in-vivo and although the proportion of T cells infiltrating tumors reduced, the proportion of tumor-specific T cells increased. These data show careful dosing and treatment protocols reduce mesothelioma cell viability and modulate tumor vessels such that tumor-antigen specific CTLs access the tumor site. However, attempts to enhance DMXAA-induced anti-tumor responses by combination with an agonist anti-CD40 antibody or IL-2 reduced efficacy. These proof-of-concept data suggest that mesothelioma patients could benefit from treatment with a STING agonist, but combination with immunotherapy should be cautiously undertaken.
Collapse
Affiliation(s)
- Peter T. Graham
- School of Medicine, Curtin University, Bentley, WA, Australia
| | - Anna K. Nowak
- Medical School, University of Western Australia, Nedlands, WA, Australia
- National Centre for Asbestos Related Diseases, Nedlands, WA, Australia
- Institute of Respiratory Health, Nedlands, WA, Australia
| | | | - Irma Larma
- Becton Dickinson Pty Limited, Osborne Park, WA, Australia
| | - Delia J. Nelson
- School of Medicine, Curtin University, Bentley, WA, Australia
- Curtin Health Innovation Research Institute, Bentley, WA, Australia
| |
Collapse
|
8
|
Meng X, Li W, Yuan H, Dong W, Xiao W, Zhang X. KDELR2-KIF20A axis facilitates bladder cancer growth and metastasis by enhancing Golgi-mediated secretion. Biol Proced Online 2022; 24:12. [PMID: 36096734 PMCID: PMC9465899 DOI: 10.1186/s12575-022-00174-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Background Bladder cancer (BCa) is a fatal form of cancer worldwide associated with a poor prognosis. Identifying novel drivers of growth and metastasis hold therapeutic potential for the disease. Transport homeostasis between the endoplasmic reticulum and Golgi and the secretion of matrix metalloproteinases (MMPs) mediated by Golgi have been reported to be closely associated with tumor progression. However, to date, mechanistic studies remain limited. Results Here, we identified KDELR2 as a potential risk factor with prognostic value in patients with BCa, especially those harbouring the KDELR2 amplification. In addition, we found that KDELR2 is a regulator of BCa cell proliferation and tumorigenicity based on bioinformatic analysis with functional studies. Mechanistically, we revealed that KDELR2 could regulate the expression of KIF20A, thus stimulating the expression of MMP2, MMP9 and MKI67. Functionally, the overexpression of KDELR2 and KIF20A markedly promoted proliferation, migration, and invasion in vitro and enhanced tumor growth in vivo, while knockdown of KDELR2 and KIF20A exerted the opposite effects. And the overexpression of KDELR2 also enhanced lymph node metastasis in vivo. Conclusions Collectively, our findings clarified a hitherto unexplored mechanism of KDELR2-KIF20A axis in increasing Golgi-mediated secretion of MMPs to drive tumor progression in BCa. Supplementary Information The online version contains supplementary material available at 10.1186/s12575-022-00174-y.
Collapse
Affiliation(s)
- Xiangui Meng
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Weiquan Li
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hongwei Yuan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wei Dong
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China. .,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Wen Xiao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China. .,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China. .,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
9
|
Yuan Y, Yang B, Qi Z, Han Z, Cai J, Song J. KDELR1 Is an Independent Prognostic Predictor and Correlates With Immunity in Glioma. Front Oncol 2022; 12:783721. [PMID: 35814367 PMCID: PMC9263977 DOI: 10.3389/fonc.2022.783721] [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: 09/26/2021] [Accepted: 05/23/2022] [Indexed: 11/20/2022] Open
Abstract
Background Gliomas are the most malignant central nervous system tumors. With the development of sequencing technology, more potential biomarkers related to the treatment, prognosis, and molecular classification of glioma have been identified. Here, we intend to investigate the potential biological function and clinical value of a new biomarker in glioma. Methods KDELR1 expression data and the corresponding clinical information were downloaded from public databases and then preprocessed using R language. Correlation, Kaplan–Meier survival, and Cox regression analyses were performed to explore the clinical significance of KDELR1 in glioma patients. Furthermore, the immune infiltration and microenvironment parameters were evaluated via TIMER and CIBERSORT. Immunohistochemistry was conducted to confirm the KDELR1 expression and its correlation with immunity infiltration and prognosis. Results KDELR1 was upregulated in glioma samples compared with normal brain tissues, and its expression was significantly correlated with age, the World Health Organization (WHO) grade, recurrence, necrosis, microvascular proliferation, molecular classification, isocitrate dehydrogenase (IDH) mutation, and 1p/19q codeletion status. In addition, survival analysis showed that glioma patients with KDELR1 overexpression had shorter overall survival (OS) and disease-free survival times, and Cox regression analysis revealed that KDELR1 acted as an independent prognostic factor of OS in glioma patients. Gene set enrichment analysis indicated a significant enrichment of metabolism-associated pathways. KDELR1 expression was positively associated with immune infiltration (including infiltration by CD8+ T cells, CD4+ T cells, macrophages, and so on) and microenvironment parameters (including stromal, immune, and ESTIMATE scores) in gliomas. The expression of KDELR1 and its correlation with the tumor grade and prognosis were confirmed by immunohistochemistry in clinical samples (n = 119, P < 0.05). Conclusions Taken together, these findings suggest that KDELR1 is correlated with the tumor grade, molecular classifications, and immune infiltration; highlighting that KDELR1 is a novel and promising biomarker for molecular classification, treatment, and prognostic assessment may further indicate the treating effect of immune therapy.
Collapse
Affiliation(s)
- Yifan Yuan
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Biao Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Zengxin Qi
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Zhenyuan Han
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Jiajun Cai
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, China
- *Correspondence: Jianping Song, ; Jiajun Cai,
| | - Jianping Song
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- *Correspondence: Jianping Song, ; Jiajun Cai,
| |
Collapse
|
10
|
KDEL Receptors: Pathophysiological Functions, Therapeutic Options, and Biotechnological Opportunities. Biomedicines 2022; 10:biomedicines10061234. [PMID: 35740256 PMCID: PMC9220330 DOI: 10.3390/biomedicines10061234] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 02/07/2023] Open
Abstract
KDEL receptors (KDELRs) are ubiquitous seven-transmembrane domain proteins encoded by three mammalian genes. They bind to and retro-transport endoplasmic reticulum (ER)-resident proteins with a C-terminal Lys-Asp-Glu-Leu (KDEL) sequence or variants thereof. In doing this, KDELR participates in the ER quality control of newly synthesized proteins and the unfolded protein response. The binding of KDEL proteins to KDELR initiates signaling cascades involving three alpha subunits of heterotrimeric G proteins, Src family kinases, protein kinases A (PKAs), and mitogen-activated protein kinases (MAPKs). These signaling pathways coordinate membrane trafficking flows between secretory compartments and control the degradation of the extracellular matrix (ECM), an important step in cancer progression. Considering the basic cellular functions performed by KDELRs, their association with various diseases is not surprising. KDELR mutants unable to bind the collagen-specific chaperon heat-shock protein 47 (HSP47) cause the osteogenesis imperfecta. Moreover, the overexpression of KDELRs appears to be linked to neurodegenerative diseases that share pathological ER-stress and activation of the unfolded protein response (UPR). Even immune function requires a functional KDELR1, as its mutants reduce the number of T lymphocytes and impair antiviral immunity. Several studies have also brought to light the exploitation of the shuttle activity of KDELR during the intoxication and maturation/exit of viral particles. Based on the above, KDELRs can be considered potential targets for the development of novel therapeutic strategies for a variety of diseases involving proteostasis disruption, cancer progression, and infectious disease. However, no drugs targeting KDELR functions are available to date; rather, KDELR has been leveraged to deliver drugs efficiently into cells or improve antigen presentation.
Collapse
|
11
|
Del Giudice S, De Luca V, Parizadeh S, Russo D, Luini A, Di Martino R. Endogenous and Exogenous Regulatory Signaling in the Secretory Pathway: Role of Golgi Signaling Molecules in Cancer. Front Cell Dev Biol 2022; 10:833663. [PMID: 35399533 PMCID: PMC8984190 DOI: 10.3389/fcell.2022.833663] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 03/03/2022] [Indexed: 11/29/2022] Open
Abstract
The biosynthetic transport route that constitutes the secretory pathway plays a fundamental role in the cell, providing to the synthesis and transport of around one third of human proteins and most lipids. Signaling molecules within autoregulatory circuits on the intracellular membranes of the secretory pathway regulate these processes, especially at the level of the Golgi complex. Indeed, cancer cells can hijack several of these signaling molecules, and therefore also the underlying regulated processes, to bolster their growth or gain more aggressive phenotypes. Here, we review the most important autoregulatory circuits acting on the Golgi, emphasizing the role of specific signaling molecules in cancer. In fact, we propose to draw awareness to highlight the Golgi-localized regulatory systems as potential targets in cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | - Alberto Luini
- *Correspondence: Alberto Luini, ; Rosaria Di Martino,
| | | |
Collapse
|
12
|
Kye YC, Lee GW, Lee SW, Ju YJ, Kim HO, Yun CH, Cho JH. STAT1 maintains naïve CD8 + T cell quiescence by suppressing the type I IFN-STAT4-mTORC1 signaling axis. SCIENCE ADVANCES 2021; 7:eabg8764. [PMID: 34516905 PMCID: PMC8442933 DOI: 10.1126/sciadv.abg8764] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 07/13/2021] [Indexed: 06/13/2023]
Abstract
Naïve CD8+ T cell quiescence is maintained at a steady state. Although this state of quiescence involves various cell-intrinsic and cell-extrinsic regulators, the mechanisms underlying this regulation remain incompletely understood. Here, we found that signal transducer and activator of transcription 1 (STAT1), a key transcription factor downstream of interferon receptor (IFNR) signaling, plays a cell-intrinsic role in maintaining naïve CD8+ T cell quiescence. STAT1-deficient mice showed enhanced proliferation of peripheral naïve CD8+ T cells, which resulted in an abnormal increase in the number of CD44hi memory/activated phenotype cells and an enlargement of secondary lymphoid tissues. This phenomenon was not observed in IFNR-deficient mice but was paradoxically dependent on type I interferon and its alternative signaling pathway via the STAT4–RagD–lysosomal mTORC1 axis. Collectively, these findings underline the importance of STAT1 in regulating the homeostasis of peripheral naïve CD8+ T cells by suppressing their responsiveness to homeostatic cues at a steady state.
Collapse
Affiliation(s)
- Yoon-Chul Kye
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
- Institutes of Green-bio Science and Technology, Seoul National University, Pyeongchang 25354, Korea
| | - Gil-Woo Lee
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang 37673, Korea
- Department of Microbiology and Immunology and Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Hwasun 58128, Korea
- Immunotherapy Innovation Center, Hwasun Hospital, Chonnam National University Medical School, Hwasun 58128, Korea
| | - Sung-Woo Lee
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang 37673, Korea
- Department of Microbiology and Immunology and Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Hwasun 58128, Korea
- Immunotherapy Innovation Center, Hwasun Hospital, Chonnam National University Medical School, Hwasun 58128, Korea
| | - Young-Jun Ju
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
- Institutes of Green-bio Science and Technology, Seoul National University, Pyeongchang 25354, Korea
| | - Hee-Ok Kim
- Immunotherapy Innovation Center, Hwasun Hospital, Chonnam National University Medical School, Hwasun 58128, Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
- Institutes of Green-bio Science and Technology, Seoul National University, Pyeongchang 25354, Korea
| | - Jae-Ho Cho
- Department of Microbiology and Immunology and Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Hwasun 58128, Korea
- Immunotherapy Innovation Center, Hwasun Hospital, Chonnam National University Medical School, Hwasun 58128, Korea
- Biomedical Sciences Graduate Program, Chonnam National University, Hwasun 58128, Korea
| |
Collapse
|
13
|
Multiomics Profiling and Clustering of Low-Grade Gliomas Based on the Integrated Stress Status. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5554436. [PMID: 34368351 PMCID: PMC8343268 DOI: 10.1155/2021/5554436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 06/18/2021] [Accepted: 07/10/2021] [Indexed: 12/30/2022]
Abstract
Background Although the prognosis of low-grade glioma is better than that of glioblastoma, there are still some groups with poor prognosis. The integrated stress response contributes to the malignant progress of tumors. As there had limited research focused on the integrated stress status in LGG, it is urgent to profile and reclassify LGG based on the integrated stress response. Methods Information of glioma patients was obtained from the Chinese Glioma Genome Atlas, The Cancer Genome Atlas, and the GSE16011 cohorts. Statistical analyses were conducted using GraphPad Prism 8 and R language. Results We summarized and quantified four types of integrated stress responses. Relationships between these four types of stress states and the clinical characteristics were analyzed in low-grade glioma. We then reclassified the patients based on these four scores and found that cluster 2 had the worst prognosis, while cluster 1 had the best prognosis. We also established an accurate integrated stress response risk signature for predicting cluster 2. We found that immune response and suppressive immune cell components were more enriched in the high-risk group. We also profiled the genomic differences between the low- and high-risk groups, including the nonmissense mutation of driver genes and the copy number variations. Conclusion Low-grade glioma patients were divided into three clusters based on the integrated stress status, with cluster 2 exhibiting malignant transformation trends. The signature adequately reflected the traits of cluster 2, while a high risk score indicated a worse prognosis and an enriched inhibitory immune microenvironment.
Collapse
|
14
|
Wei H, Ma W, Lu X, Liu H, Lin K, Wang Y, Ye Z, Sun L, Huang Z, Pan T, Zhou Z, Cheng EY, Zhang H, Gao P, Zhong X. KDELR2 promotes breast cancer proliferation via HDAC3-mediated cell cycle progression. Cancer Commun (Lond) 2021; 41:904-920. [PMID: 34146461 PMCID: PMC8441056 DOI: 10.1002/cac2.12180] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/22/2021] [Accepted: 06/01/2021] [Indexed: 12/15/2022] Open
Abstract
Background Histone deacetylases (HDACs) engage in the regulation of various cellular processes by controlling global gene expression. The dysregulation of HDACs leads to carcinogenesis, making HDACs ideal targets for cancer therapy. However, the use of HDAC inhibitors (HDACi) as single agents has been shown to have limited success in treating solid tumors in clinical studies. This study aimed to identify a novel downstream effector of HDACs to provide a potential target for combination therapy. Methods Transcriptome sequencing and bioinformatics analysis were performed to screen for genes responsive to HDACi in breast cancer cells. The effects of HDACi on cell viability were detected using the MTT assay. The mRNA and protein levels of genes were determined by quantitative reverse transcription‐PCR (qRT‐PCR) and Western blotting. Cell cycle distribution and apoptosis were analyzed by flow cytometry. The binding of CREB1 (cAMP‐response element binding protein 1) to the promoter of the KDELR (The KDEL (Lys‐Asp‐Glu‐Leu) receptor) gene was validated by the ChIP (chromatin immunoprecipitation assay). The association between KDELR2 and protein of centriole 5 (POC5) was detected by immunoprecipitation. A breast cancer‐bearing mouse model was employed to analyze the effect of the HDAC3‐KDELR2 axis on tumor growth. Results KDELR2 was identified as a novel target of HDAC3, and its aberrant expression indicated the poor prognosis of breast cancer patients. We found a strong correlation between the protein expression patterns of HADC3 and KDELR2 in tumor tissues from breast cancer patients. The results of the ChIP assay and qRT‐PCR analysis validated that HDAC3 transactivated KDELR2 via CREB1. The HDAC3‐KDELR2 axis accelerated the cell cycle progression of cancer cells by protecting the centrosomal protein POC5 from proteasomal degradation. Moreover, the HDAC3‐KDELR2 axis promoted breast cancer cell proliferation and tumorigenesis in vitro and in vivo. Conclusion Our results uncovered a previously unappreciated function of KDELR2 in tumorigenesis, linking a critical Golgi‐the endoplasmic reticulum traffic transport protein to HDAC‐controlled cell cycle progression on the path of cancer development and thus revealing a potential therapeutical target for breast cancer.
Collapse
Affiliation(s)
- Haoran Wei
- Hefei National Laboratory for Physical Sciences at Microscale, the Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, P. R. China
| | - Wenhao Ma
- Hefei National Laboratory for Physical Sciences at Microscale, the Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, P. R. China
| | - Xiaofei Lu
- Hefei National Laboratory for Physical Sciences at Microscale, the Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, P. R. China
| | - Haiying Liu
- Hefei National Laboratory for Physical Sciences at Microscale, the Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, P. R. China
| | - Kashuai Lin
- School of Medicine and Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Yinghui Wang
- School of Medicine and Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Zijian Ye
- School of Medicine and Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Linchong Sun
- School of Medicine and Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Zhitong Huang
- School of Medicine and Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Tingting Pan
- Division of Life Sciences and Medicine, The First Affiliated Hospital of University of Science and Technology of China, University of Science and Technology of China, Hefei, Anhui, 230027, P. R. China
| | - Zilong Zhou
- Hefei National Laboratory for Physical Sciences at Microscale, the Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, P. R. China
| | - Eric Y Cheng
- College of Pharmacy, University of North Texas Health Science Center, Fort Worth, Texas, 76107, USA
| | - Huafeng Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, the Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, P. R. China
| | - Ping Gao
- Hefei National Laboratory for Physical Sciences at Microscale, the Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, P. R. China.,School of Medicine and Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Xiuying Zhong
- Hefei National Laboratory for Physical Sciences at Microscale, the Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, P. R. China.,School of Medicine and Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| |
Collapse
|
15
|
The Function of KDEL Receptors as UPR Genes in Disease. Int J Mol Sci 2021; 22:ijms22115436. [PMID: 34063979 PMCID: PMC8196686 DOI: 10.3390/ijms22115436] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 12/15/2022] Open
Abstract
The KDEL receptor retrieval pathway is essential for maintaining resident proteins in the endoplasmic reticulum (ER) lumen. ER resident proteins serve a variety of functions, including protein folding and maturation. Perturbations to the lumenal ER microenvironment, such as calcium depletion, can cause protein misfolding and activation of the unfolded protein response (UPR). Additionally, ER resident proteins are secreted from the cell by overwhelming the KDEL receptor retrieval pathway. Recent data show that KDEL receptors are also activated during the UPR through the IRE1/XBP1 signaling pathway as an adaptive response to cellular stress set forth to reduce the loss of ER resident proteins. This review will discuss the emerging connection between UPR activation and KDEL receptors as it pertains to ER proteostasis and disease states.
Collapse
|
16
|
Zhang J, Qin Q, Nan X, Guo Z, Liu Y, Jadoon S, Chen Y, Zhao L, Yan L, Hou S. Role of Protein Phosphatase1 Regulatory Subunit3 in Mediating the Abscisic Acid Response. PLANT PHYSIOLOGY 2020; 184:1317-1332. [PMID: 32948668 PMCID: PMC7608174 DOI: 10.1104/pp.20.01018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/09/2020] [Indexed: 05/06/2023]
Abstract
Protein phosphatase1 (PP1) plays important roles in eukaryotes, including in plant hormone responses, and functions as a holoenzyme that consists of catalytic and regulatory subunits. Animal genomes encode ∼200 PP1-interacting proteins; by contrast, only a few have been reported in plants. In this study, PP1 Regulatory Subunit3 (PP1R3), a protein that interacts with PP1 in Arabidopsis (Arabidopsis thaliana), was characterized by mass spectrometry. PP1R3 was widely expressed in various plant tissues and PP1R3 colocalized with Type One Protein Phosphatases (TOPPs) in the nucleus and cytoplasm. The pp1r3 mutants were hypersensitive to abscisic acid (ABA), similar to the dominant-negative mutant topp4-1 or the loss-of-function multiple mutants topp1 topp4-3, topp8 topp9, topp6/7/9, topp1/2/4-3/6/7/9, and topp1/4-3/5/6/7/8/9 (topp-7m). About two-thirds of differentially expressed genes in topp-7m showed the same gene expression changes as in pp1r3-2 In response to ABA, the phenotypes of pp1r3 topp1 topp4-3 and pp1r3 topp4-1 were consistent with those of pp1r3, while pp1r3 abi1-1 showed an additive effect of the pp1r3 and abi1-1 (mutation in Abscisic Acid Insensitive1 [ABI1]) single mutants. Moreover, pp1r3 could partially recover the ABA response-related phenotype, gene expression, and plant morphology of topp4-1 PP1R3 inhibited TOPP enzyme activity and facilitated the nuclear localization of TOPP4. By contrast, ABA treatment increased the amounts of TOPP1 and TOPP4 in the cytoplasm. Importantly, nuclear localization of TOPP4 partially restored the ABA-hypersensitive phenotype of topp4-1 Overall, our results suggest that the PP1R3:TOPP holoenzyme functions in parallel with ABI1 in the nucleus to regulate ABA signaling.
Collapse
Affiliation(s)
- Jing Zhang
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Qianqian Qin
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Xiaohui Nan
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Zilong Guo
- Root Biology Center, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yang Liu
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Sawaira Jadoon
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Yan Chen
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Lulu Zhao
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Longfeng Yan
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Suiwen Hou
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
17
|
Kokubun H, Jin H, Aoe T. Pathogenic Effects of Impaired Retrieval between the Endoplasmic Reticulum and Golgi Complex. Int J Mol Sci 2019; 20:ijms20225614. [PMID: 31717602 PMCID: PMC6888596 DOI: 10.3390/ijms20225614] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/31/2019] [Accepted: 11/07/2019] [Indexed: 12/15/2022] Open
Abstract
Cellular activities, such as growth and secretion, are dependent on correct protein folding and intracellular protein transport. Injury, like ischemia, malnutrition, and invasion of toxic substances, affect the folding environment in the endoplasmic reticulum (ER). The ER senses this information, following which cells adapt their response to varied situations through the unfolded protein response. Activation of the KDEL receptor, resulting from the secretion from the ER of chaperones containing the KDEL sequence, plays an important role in this adaptation. The KDEL receptor was initially shown to be necessary for the retention of KDEL sequence-containing proteins in the ER. However, it has become clear that the activated KDEL receptor also regulates bidirectional transport between the ER and the Golgi complex, as well as from the Golgi to the secretory pathway. In addition, it has been suggested that the signal for KDEL receptor activation may also affect several other cellular activities. In this review, we discuss KDEL receptor-mediated bidirectional transport and signaling and describe disease models and human diseases related to KDEL receptor dysfunction.
Collapse
Affiliation(s)
- Hiroshi Kokubun
- Department of Anesthesiology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Hisayo Jin
- Department of Anesthesiology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Tomohiko Aoe
- Department of Medicine, Pain Center, Chiba Medical Center, Teikyo University, Ichihara 299-0111, Japan
- Correspondence: ; Tel.: +81-436-62-1211
| |
Collapse
|
18
|
Liao Z, She C, Ma L, Sun Z, Li P, Zhang X, Wang P, Li W. KDELR2 Promotes Glioblastoma Tumorigenesis Targeted by HIF1a via mTOR Signaling Pathway. Cell Mol Neurobiol 2019; 39:1207-1215. [PMID: 31342232 DOI: 10.1007/s10571-019-00715-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/18/2019] [Indexed: 01/07/2023]
Abstract
The KDEL (Lys-Asp-Glu-Leu) receptors (KDELRs), proteins with seven transmembrane domains, are primarily responsible for endoplasmic reticulum (ER) homeostasis. Recent studies have found additional function of KDELRs in growth, cellular secretory traffic, immune response, and autophagy; however, its role in tumorigenesis is still poorly understood. Here, we showed that KDELR2 is highly expressed in glioblastoma (GBM) tissues. Reviewing the expression of KDELR2 in TCGA and REMBRANDT database, we found that higher expression of KDELR2 is associated with shorter survival of GBM patients. We explored the effect of KDELR2 on tumorigenesis in GBM cells and animal model (nude mice), and identified KDELR2 as oncogene promoting cell proliferation. Additionally, KDELR2 expression in GBM cells correlated positively with HIF1alpha (HIF1α) expression, and we demonstrated by ChIP-qPCR and luciferase reporter assay that the upstream region of the KDELR2 gene is directly targeted by HIF1alpha. Taken together, our data suggest that KDELR2 is a target gene downstream of HIF1-alpha driving the malignancy of GBM and could eventually serve as a therapeutic target for the treatment of GBM patients.
Collapse
Affiliation(s)
- Zhangyuan Liao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Chunhua She
- Department of Neurosurgery and Neuro-Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
| | - Li Ma
- Department of Neurosurgery and Neuro-Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Zengfeng Sun
- Department of Neurosurgery and Neuro-Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Peng Li
- Department of Neurosurgery and Neuro-Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Xiaohui Zhang
- Department of Neurosurgery and Neuro-Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Peng Wang
- Department of Neurosurgery and Neuro-Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Wenliang Li
- Department of Neurosurgery and Neuro-Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
| |
Collapse
|
19
|
Blum A, Khalifa S, Nordström K, Simon M, Schulz MH, Schmitt MJ. Transcriptomics of a KDELR1 knockout cell line reveals modulated cell adhesion properties. Sci Rep 2019; 9:10611. [PMID: 31337861 PMCID: PMC6650600 DOI: 10.1038/s41598-019-47027-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/05/2019] [Indexed: 11/10/2022] Open
Abstract
KDEL receptors (KDELRs) represent transmembrane proteins of the secretory pathway which regulate the retention of soluble ER-residents as well as retrograde and anterograde vesicle trafficking. In addition, KDELRs are involved in the regulation of cellular stress response and ECM degradation. For a deeper insight into KDELR1 specific functions, we characterised a KDELR1-KO cell line (HAP1) through whole transcriptome analysis by comparing KDELR1-KO cells with its respective HAP1 wild-type. Our data indicate more than 300 significantly and differentially expressed genes whose gene products are mainly involved in developmental processes such as cell adhesion and ECM composition, pointing out to severe cellular disorders due to a loss of KDELR1. Impaired adhesion capacity of KDELR1-KO cells was further demonstrated through in vitro adhesion assays, while collagen- and/or laminin-coating nearly doubled the adhesion property of KDELR1-KO cells compared to wild-type, confirming a transcriptional adaptation to improve or restore the cellular adhesion capability. Perturbations within the secretory pathway were verified by an increased secretion of ER-resident PDI and decreased cell viability under ER stress conditions, suggesting KDELR1-KO cells to be severely impaired in maintaining cellular homeostasis.
Collapse
Affiliation(s)
- Andrea Blum
- Molecular and Cell Biology, Department of Biosciences (FR 8.3) and Center of Human and Molecular Biology (ZHMB), Saarland University, D-66123, Saarbrücken, Germany.
| | - Saleem Khalifa
- Cluster of Excellence, Multimodal Computing and Interaction, Saarland University and Department for Computational Biology and Applied Algorithmics, Max Planck Institute for Informatics, Saarland Informatics Campus, Saarbrücken, Germany
| | - Karl Nordström
- Genetics/Epigenetics, Center for Human and Molecular Biology, Saarland University, Saarbrücken, Germany
| | - Martin Simon
- Molecular Cell Biology and Microbiology, Wuppertal University, D-42097, Wuppertal, Germany
| | - Marcel H Schulz
- Cluster of Excellence, Multimodal Computing and Interaction, Saarland University and Department for Computational Biology and Applied Algorithmics, Max Planck Institute for Informatics, Saarland Informatics Campus, Saarbrücken, Germany.,Institute for Cardiovascular Regeneration, Goethe University Frankfurt, 60590 Frankfurt am Main, Frankfurt, Germany
| | - Manfred J Schmitt
- Molecular and Cell Biology, Department of Biosciences (FR 8.3) and Center of Human and Molecular Biology (ZHMB), Saarland University, D-66123, Saarbrücken, Germany.
| |
Collapse
|
20
|
Stressed: The Unfolded Protein Response in T Cell Development, Activation, and Function. Int J Mol Sci 2019; 20:ijms20071792. [PMID: 30978945 PMCID: PMC6479341 DOI: 10.3390/ijms20071792] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 12/27/2022] Open
Abstract
The unfolded protein response (UPR) is a highly conserved pathway that allows cells to respond to stress in the endoplasmic reticulum caused by an accumulation of misfolded and unfolded protein. This is of great importance to secretory cells because, in order for proteins to traffic from the endoplasmic reticulum (ER), they need to be folded appropriately. While a wealth of literature has implicated UPR in immune responses, less attention has been given to the role of UPR in T cell development and function. This review discusses the importance of UPR in T cell development, homeostasis, activation, and effector functions. We also speculate about how UPR may be manipulated in T cells to ameliorate pathologies.
Collapse
|
21
|
Wu J, Chen YJ, Dobbs N, Sakai T, Liou J, Miner JJ, Yan N. STING-mediated disruption of calcium homeostasis chronically activates ER stress and primes T cell death. J Exp Med 2019; 216:867-883. [PMID: 30886058 PMCID: PMC6446864 DOI: 10.1084/jem.20182192] [Citation(s) in RCA: 201] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/29/2019] [Accepted: 02/25/2019] [Indexed: 12/31/2022] Open
Abstract
STING gain-of-function mutations cause lung disease and T cell cytopenia through unknown mechanisms. Here, we found that these mutants induce chronic activation of ER stress and unfolded protein response (UPR), leading to T cell death by apoptosis in the StingN153S/+ mouse and in human T cells. Mechanistically, STING-N154S disrupts calcium homeostasis in T cells, thus intrinsically primes T cells to become hyperresponsive to T cell receptor signaling-induced ER stress and the UPR, leading to cell death. This intrinsic priming effect is mediated through a novel region of STING that we name "the UPR motif," which is distinct from known domains required for type I IFN signaling. Pharmacological inhibition of ER stress prevented StingN153S/+ T cell death in vivo. By crossing StingN153S/+ to the OT-1 mouse, we fully restored CD8+ T cells and drastically ameliorated STING-associated lung disease. Together, our data uncover a critical IFN-independent function of STING that regulates calcium homeostasis, ER stress, and T cell survival.
Collapse
Affiliation(s)
- Jianjun Wu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX.,Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Yu-Ju Chen
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Nicole Dobbs
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX.,Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Tomomi Sakai
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX.,Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Jen Liou
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Jonathan J Miner
- Department of Medicine, Washington University School of Medicine, St. Louis, MO.,Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Nan Yan
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX .,Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
22
|
Si N, Song Z, Meng X, Li X, Xiao W, Zhang X. A novel MAF missense mutation leads to congenital nuclear cataract by impacting the transactivation of crystallin and noncrystallin genes. Gene 2019; 692:113-118. [PMID: 30659945 DOI: 10.1016/j.gene.2019.01.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/01/2019] [Accepted: 01/11/2019] [Indexed: 12/27/2022]
Abstract
The transcription factor v-maf avain musculoaponeurotic fibrosarcoma oncogene homolog (MAF) plays an important role in lens development. It contains a unique extended homology region (EHR) in the DNA binding domain. MAF mutations are associated with phenotypically distinct forms of congenital cataract and show different effects on the transactivation of target genes. Mutations in the MAF EHR region were rarely reported and their corresponding phenotype and impact on target genes' transactivation were not evaluated. A three- generation Chinese family with congenital cataract was recruited. The patients in the family present non-syndromic congenital nuclear and lamellar opacities. A novel MAF mutation (c.812 T > A, p.Val271Glu) was identified by targeted next-generation sequencing. The mutation is in highly conserved EHR region of MAF and co-segregates with the cataract in the family. It is predicted to be pathogenic by multiple algorithms and is absent in a control population. Dual luciferase activity assay shows the mutation significantly impair the transcriptional activity of four crystallin genes (CRYAA, CRYBA4, CRYBA1, and CRYGA) and two non-crystallin genes (HMOX1 and KDELR2). Herein, we report a novel missense mutation in the MAF EHR region of the DNA binding domain in a family with congenital cataract. The mutation is associated with non-syndromic bilateral nuclear cataract and impacts the transactivation of cataract associated genes involved in lens structure and stress response.
Collapse
Affiliation(s)
- Nuo Si
- McKusick-Zhang Center for Genetic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Zixun Song
- Department of Ophthalmology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Xiaolu Meng
- McKusick-Zhang Center for Genetic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Xinru Li
- Department of Ophthalmology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Wei Xiao
- Department of Ophthalmology, Shengjing Hospital, China Medical University, Shenyang, China.
| | - Xue Zhang
- McKusick-Zhang Center for Genetic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China.
| |
Collapse
|
23
|
KDELR2 Competes with Measles Virus Envelope Proteins for Cellular Chaperones Reducing Their Chaperone-Mediated Cell Surface Transport. Viruses 2019; 11:v11010027. [PMID: 30621148 PMCID: PMC6356275 DOI: 10.3390/v11010027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 12/21/2018] [Accepted: 01/03/2019] [Indexed: 12/19/2022] Open
Abstract
Recently, we found that the cytidine deaminase APOBEC3G (A3G) inhibits measles (MV) replication. Using a microarray, we identified differential regulation of several host genes upon ectopic expression of A3G. One of the up-regulated genes, the endoplasmic reticulum (ER) protein retention receptor KDELR2, reduced MV replication ~5 fold when it was over-expressed individually in Vero and CEM-SS T cells. Silencing of KDELR2 in A3G-expressing Vero cells abrogated the antiviral activity induced by A3G, confirming its role as an A3G-regulated antiviral host factor. Recognition of the KDEL (Lys-Asp-Glu-Leu) motif by KDEL receptors initiates the retrograde transport of soluble proteins that have escaped the ER and play an important role in ER quality control. Although KDELR2 over-expression reduced MV titers in cell cultures, we observed no interaction between KDELR2 and the MV hemagglutinin (H) protein. Instead, KDELR2 retained chaperones in the ER, which are required for the correct folding and transport of the MV envelope glycoproteins H and fusion protein (F) to the cell surface. Our data indicate that KDELR2 competes with MV envelope proteins for binding to calnexin and GRP78/Bip, and that this interaction limits the availability of the chaperones for MV proteins, causing the reduction of virus spread and titers.
Collapse
|
24
|
Mei M, Zhai C, Li X, Zhou Y, Peng W, Ma L, Wang Q, Iverson BL, Zhang G, Yi L. Characterization of aromatic residue-controlled protein retention in the endoplasmic reticulum of Saccharomyces cerevisiae. J Biol Chem 2017; 292:20707-20719. [PMID: 29038295 DOI: 10.1074/jbc.m117.812107] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/11/2017] [Indexed: 01/05/2023] Open
Abstract
An endoplasmic reticulum (ER) retention sequence (ERS) is a characteristic short sequence that mediates protein retention in the ER of eukaryotic cells. However, little is known about the detailed molecular mechanism involved in ERS-mediated protein ER retention. Using a new surface display-based fluorescence technique that effectively quantifies ERS-promoted protein ER retention within Saccharomyces cerevisiae cells, we performed comprehensive ERS analyses. We found that the length, type of amino acid residue, and additional residues at positions -5 and -6 of the C-terminal HDEL motif all determined the retention of ERS in the yeast ER. Moreover, the biochemical results guided by structure simulation revealed that aromatic residues (Phe-54, Trp-56, and other aromatic residues facing the ER lumen) in both the ERS (at positions -6 and -4) and its receptor, Erd2, jointly determined their interaction with each other. Our studies also revealed that this aromatic residue interaction might lead to the discriminative recognition of HDEL or KDEL as ERS in yeast or human cells, respectively. Our findings expand the understanding of ERS-mediated residence of proteins in the ER and may guide future research into protein folding, modification, and translocation affected by ER retention.
Collapse
Affiliation(s)
- Meng Mei
- From the Hubei Collaborative Innovation Center for Green Transformation of Bioresources, Hubei Key Laboratory of Industrial Biotechnology, Hubei University, Wuhan 430062, China
| | - Chao Zhai
- From the Hubei Collaborative Innovation Center for Green Transformation of Bioresources, Hubei Key Laboratory of Industrial Biotechnology, Hubei University, Wuhan 430062, China
| | - Xinzhi Li
- From the Hubei Collaborative Innovation Center for Green Transformation of Bioresources, Hubei Key Laboratory of Industrial Biotechnology, Hubei University, Wuhan 430062, China
| | - Yu Zhou
- From the Hubei Collaborative Innovation Center for Green Transformation of Bioresources, Hubei Key Laboratory of Industrial Biotechnology, Hubei University, Wuhan 430062, China
| | - Wenfang Peng
- From the Hubei Collaborative Innovation Center for Green Transformation of Bioresources, Hubei Key Laboratory of Industrial Biotechnology, Hubei University, Wuhan 430062, China
| | - Lixin Ma
- From the Hubei Collaborative Innovation Center for Green Transformation of Bioresources, Hubei Key Laboratory of Industrial Biotechnology, Hubei University, Wuhan 430062, China
| | - Qinhong Wang
- the Key Laboratory of Systems Microbial Biotechnology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China, and
| | - Brent L Iverson
- the Department of Chemistry, University of Texas, Austin, Texas 78712
| | - Guimin Zhang
- From the Hubei Collaborative Innovation Center for Green Transformation of Bioresources, Hubei Key Laboratory of Industrial Biotechnology, Hubei University, Wuhan 430062, China,
| | - Li Yi
- From the Hubei Collaborative Innovation Center for Green Transformation of Bioresources, Hubei Key Laboratory of Industrial Biotechnology, Hubei University, Wuhan 430062, China,
| |
Collapse
|
25
|
Seu L, Tidwell C, Timares L, Duverger A, Wagner FH, Goepfert PA, Westfall AO, Sabbaj S, Kutsch O. CD151 Expression Is Associated with a Hyperproliferative T Cell Phenotype. THE JOURNAL OF IMMUNOLOGY 2017; 199:3336-3347. [PMID: 28954890 DOI: 10.4049/jimmunol.1700648] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/28/2017] [Indexed: 02/06/2023]
Abstract
The tetraspanin CD151 is a marker of aggressive cell proliferation and invasiveness for a variety of cancer types. Given reports of CD151 expression on T cells, we explored whether CD151 would mark T cells in a hyperactivated state. Consistent with the idea that CD151 could mark a phenotypically distinct T cell subset, it was not uniformly expressed on T cells. CD151 expression frequency was a function of the T cell lineage (CD8 > CD4) and a function of the memory differentiation state (naive T cells < central memory T cells < effector memory T cells < T effector memory RA+ cells). CD151 and CD57, a senescence marker, defined the same CD28- T cell populations. However, CD151 also marked a substantial CD28+ T cell population that was not marked by CD57. Kinome array analysis demonstrated that CD28+CD151+ T cells form a subpopulation with a distinct molecular baseline and activation phenotype. Network analysis of these data revealed that cell cycle control and cell death were the most altered process motifs in CD28+CD151+ T cells. We demonstrate that CD151 in T cells is not a passive marker, but actively changed the cell cycle control and cell death process motifs of T cells. Consistent with these data, long-term T cell culture experiments in the presence of only IL-2 demonstrated that independent of their CD28 expression status, CD151+ T cells, but not CD151- T cells, would exhibit an Ag-independent, hyperresponsive proliferation phenotype. Not unlike its reported function as a tumor aggressiveness marker, CD151 in humans thus marks and enables hyperproliferative T cells.
Collapse
Affiliation(s)
- Lillian Seu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Christopher Tidwell
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Laura Timares
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Alexandra Duverger
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Frederic H Wagner
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Paul A Goepfert
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Andrew O Westfall
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Steffanie Sabbaj
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Olaf Kutsch
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
26
|
Jin H, Komita M, Aoe T. The Role of BiP Retrieval by the KDEL Receptor in the Early Secretory Pathway and its Effect on Protein Quality Control and Neurodegeneration. Front Mol Neurosci 2017; 10:222. [PMID: 28769758 PMCID: PMC5511815 DOI: 10.3389/fnmol.2017.00222] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 06/27/2017] [Indexed: 12/14/2022] Open
Abstract
Protein quality control in the early secretory pathway is a ubiquitous eukaryotic mechanism for adaptation to endoplasmic reticulum (ER) stress. An ER molecular chaperone, immunoglobulin heavy chain-binding protein (BiP), is one of the essential components in this process. BiP interacts with nascent proteins to facilitate their folding. BiP also plays an important role in preventing aggregation of misfolded proteins and regulating the ER stress response when cells suffer various injuries. BiP is a member of the 70-kDa heat shock protein (HSP70) family of molecular chaperones that resides in the ER. Interaction between BiP and unfolded proteins is mediated by a substrate-binding domain and a nucleotide-binding domain for ATPase activity, leading to protein folding and maturation. BiP also possesses a retrieval motif in its carboxyl terminal. When BiP is secreted from the ER, the Lys-Asp-Glu-Leu (KDEL) receptor in the post-ER compartments binds with the carboxyl terminal KDEL sequence of BiP and returns BiP to the ER via coat protein complex I (COPI) vesicular transport. Although yeast studies showed that BiP retrieval by the KDEL receptor is not essential in single cells, it is crucial for multicellular organisms, where some essential proteins require retrieval to facilitate folding and maturation. Experiments in knock-in mice expressing mutant BiP with the retrieval motif deleted revealed a unique role of BiP retrieval by the KDEL receptor in neuronal development and age-related neurodegeneration.
Collapse
Affiliation(s)
- Hisayo Jin
- Department of Anesthesiology, Graduate School of Medicine, Chiba UniversityChiba, Japan
| | - Mari Komita
- Department of Anesthesiology, Chiba Rosai HospitalIchihara, Japan
| | - Tomohiko Aoe
- Pain Center, Chiba Medical Center, Teikyo UniversityIchihara, Japan
| |
Collapse
|
27
|
Mohamed E, Cao Y, Rodriguez PC. Endoplasmic reticulum stress regulates tumor growth and anti-tumor immunity: a promising opportunity for cancer immunotherapy. Cancer Immunol Immunother 2017; 66:1069-1078. [PMID: 28577085 DOI: 10.1007/s00262-017-2019-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 05/14/2017] [Indexed: 11/30/2022]
Abstract
The endoplasmic reticulum (ER) stress is a cellular process that occurs as a consequence of several stress circumstances, such as the accumulation of unfolded proteins in the lumen of the ER or distinct insults that disturb the ER normal function. Different conditions in the tumor microenvironment (TME), including hypoxia, nutrient deprivation, and the elevated production of reactive oxygen and nitrogen species destabilize the loading and dispatching of the newly synthesized proteins, triggering ER stress in cancer cells and tumor-infiltrating leukocytes. In order to cope with TME-induced ER stress, tumor and stromal cells initiate an adaptive response process that aims to resolve ER stress and to restore cellular homeostasis, which is referred as the unfolded protein responses (UPR). Paradoxically, the UPR can also induce cell death under severe and/or permanent ER stress. The UPR is started through three mediators, the activation of the inositol-requiring enzyme-1α, the pancreatic ER kinase-like ER kinase, and the activating transcription factor 6. In this minireview, we will discuss the pro- and anti-tumorigenic role of the UPR in cancer cells. In addition, we will describe the effects of the TME-induced ER stress in the immunosuppressive activity of tumor-infiltrating myeloid cells. Also, we will review the results of emerging therapeutic interventions that target ER stress and the UPR mediators in cancer. We postulate that the inhibition of ER stress or the UPR-related elements could represent a significant approach to increase the efficacy of various forms of cancer immunotherapy.
Collapse
Affiliation(s)
- Eslam Mohamed
- Georgia Cancer Center, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd, Room CN-4125A, Augusta, GA, 30912, USA.
| | - Yu Cao
- Georgia Cancer Center, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd, Room CN-4125A, Augusta, GA, 30912, USA
| | - Paulo C Rodriguez
- Department of Medicine, Georgia Cancer Center, Augusta University, 1410 Laney Walker Blvd, Room CN-4114, Augusta, GA, 30912, USA.
| |
Collapse
|
28
|
H/KDEL receptors mediate host cell intoxication by a viral A/B toxin in yeast. Sci Rep 2016; 6:31105. [PMID: 27493088 PMCID: PMC4974620 DOI: 10.1038/srep31105] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 07/13/2016] [Indexed: 01/14/2023] Open
Abstract
A/B toxins such as cholera toxin, Pseudomonas exotoxin and killer toxin K28 contain a KDEL-like amino acid motif at one of their subunits which ensures retrograde toxin transport through the secretory pathway of a target cell. As key step in host cell invasion, each toxin binds to distinct plasma membrane receptors that are utilized for cell entry. Despite intensive efforts, some of these receptors are still unknown. Here we identify the yeast H/KDEL receptor Erd2p as membrane receptor of K28, a viral A/B toxin carrying an HDEL motif at its cell binding β-subunit. While initial toxin binding to the yeast cell wall is unaffected in cells lacking Erd2p, binding to spheroplasts and in vivo toxicity strongly depend on the presence of Erd2p. Consistently, Erd2p is not restricted to membranes of the early secretory pathway but extends to the plasma membrane where it binds and internalizes HDEL-cargo such as K28 toxin, GFP(HDEL) and Kar2p. Since human KDEL receptors are fully functional in yeast and restore toxin sensitivity in the absence of endogenous Erd2p, toxin uptake by H/KDEL receptors at the cell surface might likewise contribute to the intoxication efficiency of A/B toxins carrying a KDEL-motif at their cytotoxic A-subunit(s).
Collapse
|
29
|
Bustos-Morán E, Blas-Rus N, Martín-Cófreces NB, Sánchez-Madrid F. Orchestrating Lymphocyte Polarity in Cognate Immune Cell-Cell Interactions. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 327:195-261. [PMID: 27692176 DOI: 10.1016/bs.ircmb.2016.06.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The immune synapse (IS) is a specialized structure established between different immune cells that fulfills several functions, including a role as a communication bridge. This intimate contact between a T cell and an antigen-presenting cell promotes the proliferation and differentiation of lymphocytes involved in the contact. T-cell activation requires the specific triggering of the T-cell receptor (TCR), which promotes the activation of different signaling pathways inducing the polarization of the T cell. During this process, different adhesion and signaling receptors reorganize at specialized membrane domains, concomitantly to the polarization of the tubulin and actin cytoskeletons, forming stable polarization platforms. The centrosome also moves toward the IS, driving the movement of different organelles, such as the biosynthetic, secretory, degrading machinery, and mitochondria, to sustain T-cell activation. A proper orchestration of all these events is essential for T-cell effector functions and the accomplishment of a complete immune response.
Collapse
Affiliation(s)
- Eugenio Bustos-Morán
- Vascular Pathophysiology Area, Spanish National Center of Cardiovascular Research (CNIC), Madrid, Spain
| | - Noelia Blas-Rus
- Department of Immunology, La Princesa Hospital, Autonomus University of Madrid (UAM), Health Research Institute of Princesa Hospital (ISS-IP), Madrid, Spain
| | - Noa Beatriz Martín-Cófreces
- Vascular Pathophysiology Area, Spanish National Center of Cardiovascular Research (CNIC), Madrid, Spain.,Department of Immunology, La Princesa Hospital, Autonomus University of Madrid (UAM), Health Research Institute of Princesa Hospital (ISS-IP), Madrid, Spain
| | - Francisco Sánchez-Madrid
- Vascular Pathophysiology Area, Spanish National Center of Cardiovascular Research (CNIC), Madrid, Spain.,Department of Immunology, La Princesa Hospital, Autonomus University of Madrid (UAM), Health Research Institute of Princesa Hospital (ISS-IP), Madrid, Spain
| |
Collapse
|
30
|
Kamimura D, Atsumi T, Stofkova A, Nishikawa N, Ohki T, Suzuki H, Katsunuma K, Jiang JJ, Bando H, Meng J, Sabharwal L, Ogura H, Hirano T, Arima Y, Murakami M. Naïve T Cell Homeostasis Regulated by Stress Responses and TCR Signaling. Front Immunol 2016; 6:638. [PMID: 26734005 PMCID: PMC4681834 DOI: 10.3389/fimmu.2015.00638] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 12/04/2015] [Indexed: 11/13/2022] Open
Abstract
The survival of naïve T cells is believed to require signals from TCR–pMHC interactions and cytokines such as IL-7. In contrast, signals that negatively impact naïve T cell survival are less understood. We conducted a forward genetic screening of mice and found a mutant mouse line with reduced number of naïve T cells (T-Red mice). T-Red mice have a point mutation in the Kdelr1 gene, and their naïve T cells show enhanced integrated stress response (ISR), which eventually induces their apoptosis. Therefore, naïve T cells require a KDEL receptor-mediated mechanism that efficiently relieves cellular stress for their survival in vivo. Interestingly, naïve T cells expressing TCR with higher affinity/avidity to self-antigens survive in T-Red mice, suggesting the possible link between TCR-mediated survival and ISR-induced apoptosis. In this article, we discuss the regulation of naïve T cell homeostasis, keeping special attention on the ISR and TCR signal.
Collapse
Affiliation(s)
- Daisuke Kamimura
- Division of Molecular Neuroimmunology, Institute for Genomic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Toru Atsumi
- Division of Molecular Neuroimmunology, Institute for Genomic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Andrea Stofkova
- Division of Molecular Neuroimmunology, Institute for Genomic Medicine, Graduate School of Medicine, Hokkaido University , Sapporo , Japan
| | - Naoki Nishikawa
- Division of Molecular Neuroimmunology, Institute for Genomic Medicine, Graduate School of Medicine, Hokkaido University , Sapporo , Japan
| | - Takuto Ohki
- Division of Molecular Neuroimmunology, Institute for Genomic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hironao Suzuki
- Division of Molecular Neuroimmunology, Institute for Genomic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Kokichi Katsunuma
- Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Jing-Jing Jiang
- Division of Molecular Neuroimmunology, Institute for Genomic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hidenori Bando
- Division of Molecular Neuroimmunology, Institute for Genomic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Jie Meng
- Division of Molecular Neuroimmunology, Institute for Genomic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Lavannya Sabharwal
- Division of Molecular Neuroimmunology, Institute for Genomic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hideki Ogura
- Division of Molecular Neuroimmunology, Institute for Genomic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Medicine, Osaka University, Suita, Japan
| | | | - Yasunobu Arima
- Division of Molecular Neuroimmunology, Institute for Genomic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Masaaki Murakami
- Division of Molecular Neuroimmunology, Institute for Genomic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan; Laboratory of Developmental Immunology, WPI Immunology Frontier Research Center, Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
31
|
Kamimura D, Arima Y, Tsuruoka M, Jiang JJ, Bando H, Meng J, Sabharwal L, Stofkova A, Nishikawa N, Higuchi K, Ogura H, Atsumi T, Murakami M. Strong TCR-mediated signals suppress integrated stress responses induced by KDELR1 deficiency in naive T cells. Int Immunol 2015; 28:117-26. [PMID: 26489882 DOI: 10.1093/intimm/dxv059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 10/09/2015] [Indexed: 12/17/2022] Open
Abstract
KDEL receptor 1 (KDELR1) regulates integrated stress responses (ISR) to promote naive T-cell survival in vivo. In a mouse line having nonfunctional KDELR1, T-Red (naive T-cell reduced) mice, polyclonal naive T cells show excessive ISR and eventually undergo apoptosis. However, breeding T-Red mice with TCR-transgenic mice bearing relatively high TCR affinity rescued the T-Red phenotype, implying a link between ISR-induced apoptosis and TCR-mediated signaling. Here, we showed that strong TCR stimulation reduces ISR in naive T cells. In mice lacking functional KDELR1, surviving naive T cells expressed significantly higher levels of CD5, a surrogate marker of TCR self-reactivity. In addition, higher TCR affinity/avidity was confirmed using a tetramer dissociation assay on the surviving naive T cells, suggesting that among the naive T-cell repertoire, those that receive relatively stronger TCR-mediated signals via self-antigens survive enhanced ISR. Consistent with this observation, weak TCR stimulation with altered peptide ligands decreased the survival and proliferation of naive T cells, whereas stimulation with ligands having higher affinity had no such effect. These results suggest a novel role of TCR-mediated signals in the attenuation of ISR in vivo.
Collapse
Affiliation(s)
- Daisuke Kamimura
- Division of Molecular Neuroimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, Hokkaido 060-0815, Japan Laboratory of Developmental Immunology, Graduate School of Frontier Biosciences, Graduate School of Medicine and WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yasunobu Arima
- Division of Molecular Neuroimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, Hokkaido 060-0815, Japan Laboratory of Developmental Immunology, Graduate School of Frontier Biosciences, Graduate School of Medicine and WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Mineko Tsuruoka
- Laboratory of Developmental Immunology, Graduate School of Frontier Biosciences, Graduate School of Medicine and WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Jing-Jing Jiang
- Division of Molecular Neuroimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, Hokkaido 060-0815, Japan Laboratory of Developmental Immunology, Graduate School of Frontier Biosciences, Graduate School of Medicine and WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hidenori Bando
- Division of Molecular Neuroimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, Hokkaido 060-0815, Japan Laboratory of Developmental Immunology, Graduate School of Frontier Biosciences, Graduate School of Medicine and WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Jie Meng
- Division of Molecular Neuroimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, Hokkaido 060-0815, Japan Laboratory of Developmental Immunology, Graduate School of Frontier Biosciences, Graduate School of Medicine and WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Lavannya Sabharwal
- Division of Molecular Neuroimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, Hokkaido 060-0815, Japan Laboratory of Developmental Immunology, Graduate School of Frontier Biosciences, Graduate School of Medicine and WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Andrea Stofkova
- Division of Molecular Neuroimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, Hokkaido 060-0815, Japan
| | - Naoki Nishikawa
- Division of Molecular Neuroimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, Hokkaido 060-0815, Japan
| | - Kotaro Higuchi
- Division of Molecular Neuroimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, Hokkaido 060-0815, Japan
| | - Hideki Ogura
- Division of Molecular Neuroimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, Hokkaido 060-0815, Japan Laboratory of Developmental Immunology, Graduate School of Frontier Biosciences, Graduate School of Medicine and WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Toru Atsumi
- Division of Molecular Neuroimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, Hokkaido 060-0815, Japan Laboratory of Developmental Immunology, Graduate School of Frontier Biosciences, Graduate School of Medicine and WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masaaki Murakami
- Division of Molecular Neuroimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, Hokkaido 060-0815, Japan Laboratory of Developmental Immunology, Graduate School of Frontier Biosciences, Graduate School of Medicine and WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
32
|
Mutation of the ER retention receptor KDELR1 leads to cell-intrinsic lymphopenia and a failure to control chronic viral infection. Proc Natl Acad Sci U S A 2015; 112:E5706-14. [PMID: 26438836 DOI: 10.1073/pnas.1515619112] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Endoplasmic reticulum (ER)-resident proteins are continually retrieved from the Golgi and returned to the ER by Lys-Asp-Glu-Leu (KDEL) receptors, which bind to an eponymous tetrapeptide motif at their substrate's C terminus. Mice and humans possess three paralogous KDEL receptors, but little is known about their functional redundancy, or if their mutation can be physiologically tolerated. Here, we present a recessive mouse missense allele of the prototypical mammalian KDEL receptor, KDEL ER protein retention receptor 1 (KDELR1). Kdelr1 homozygous mutants were mildly lymphopenic, as were mice with a CRISPR/Cas9-engineered frameshift allele. Lymphopenia was cell intrinsic and, in the case of T cells, was associated with reduced expression of the T-cell receptor (TCR) and increased expression of CD44, and could be partially corrected by an MHC class I-restricted TCR transgene. Antiviral immunity was also compromised, with Kdelr1 mutant mice unable to clear an otherwise self-limiting viral infection. These data reveal a nonredundant cellular function for KDELR1, upon which lymphocytes distinctly depend.
Collapse
|