1
|
Hu B, Liu G, Zhao K, Zhang G. Diversity of extracellular HSP70 in cancer: advancing from a molecular biomarker to a novel therapeutic target. Front Oncol 2024; 14:1388999. [PMID: 38646439 PMCID: PMC11026673 DOI: 10.3389/fonc.2024.1388999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 03/25/2024] [Indexed: 04/23/2024] Open
Abstract
Heat shock protein 70 (HSP70) is a highly conserved protein functioning as a "molecular chaperone", which is integral to protein folding and maturation. In addition to its high expression within cells upon stressful challenges, HSP70 can be translocated to the cell membrane or released from cells in free form or within extracellular vesicles (EVs). Such trafficking of HSP70 is also present in cancer cells, as HSP70 is overexpressed in various types of patient samples across a range of common malignancies, signifying that extracellular HSP70 (eHSP70) can serve as a tumor biomarker. eHSP70 is involved in a broad range of cancer-related events, including cell proliferation and apoptosis, extracellular matrix (ECM) remodeling, epithelial-mesenchymal transition (EMT), angiogenesis, and immune response. eHSP70 can also induce cancer cell resistance to various treatments, such as chemotherapy, radiotherapy, and anti-programmed death-1 (PD-1) immunotherapy. Though the role of eHSP70 in tumors is contradictory, characterized by both pro-tumor and anti-tumor effects, eHSP70 serves as a promising target in cancer treatment. In this review, we comprehensively summarized the current knowledge about the role of eHSP70 in cancer progression and treatment resistance and discussed the feasibility of eHSP70 as a cancer biomarker and therapeutic target.
Collapse
Affiliation(s)
- Binbin Hu
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guihong Liu
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kejia Zhao
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Chengdu, Sichuan, China
| | - Gao Zhang
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
2
|
Chen J, Lynn EG, Yousof TR, Sharma H, MacDonald ME, Byun JH, Shayegan B, Austin RC. Scratching the Surface—An Overview of the Roles of Cell Surface GRP78 in Cancer. Biomedicines 2022; 10:biomedicines10051098. [PMID: 35625836 PMCID: PMC9138746 DOI: 10.3390/biomedicines10051098] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/01/2022] [Accepted: 05/05/2022] [Indexed: 02/04/2023] Open
Abstract
The 78 kDa glucose-regulated protein (GRP78) is considered an endoplasmic reticulum (ER)-resident molecular chaperone that plays a crucial role in protein folding homeostasis by regulating the unfolded protein response (UPR) and inducing numerous proapoptotic and autophagic pathways within the eukaryotic cell. However, in cancer cells, GRP78 has also been shown to migrate from the ER lumen to the cell surface, playing a role in several cellular pathways that promote tumor growth and cancer cell progression. There is another insidious consequence elicited by cell surface GRP78 (csGRP78) on cancer cells: the accumulation of csGRP78 represents a novel neoantigen leading to the production of anti-GRP78 autoantibodies that can bind csGRP78 and further amplify these cellular pathways to enhance cell growth and mitigate apoptotic cell death. This review examines the current body of literature that delineates the mechanisms by which ER-resident GRP78 localizes to the cell surface and its consequences, as well as potential therapeutics that target csGRP78 and block its interaction with anti-GRP78 autoantibodies, thereby inhibiting further amplification of cancer cell progression.
Collapse
Affiliation(s)
- Jack Chen
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
| | - Edward G. Lynn
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
| | - Tamana R. Yousof
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
| | - Hitesh Sharma
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
| | - Melissa E. MacDonald
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
| | - Jae Hyun Byun
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
| | - Bobby Shayegan
- Department of Surgery, Division of Urology, The Research Institute of St. Joe′s Hamilton, McMaster University, ON L8N 4A6, Canada;
| | - Richard C. Austin
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
- Correspondence: ; Tel.: +1-905-522-1155 (ext. 35175)
| |
Collapse
|
3
|
Hernandez I, Cohen M. Linking cell-surface GRP78 to cancer: From basic research to clinical value of GRP78 antibodies. Cancer Lett 2022; 524:1-14. [PMID: 34637844 DOI: 10.1016/j.canlet.2021.10.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/10/2021] [Accepted: 10/05/2021] [Indexed: 01/01/2023]
Abstract
Glucose-related protein 78 (GRP78) is a chaperone protein localized primarily in the endoplasmic reticulum (ER) lumen, where it helps in proper protein folding by targeting misfolded proteins and facilitating protein assembly. In stressed cells, GRP78 is translocated to the cell surface (csGRP78) where it binds to various ligands and triggers different intracellular pathways. Thus, csGRP78 expression is associated with cancer, involved in the maintenance and progression of the disease. Extracellular exposition of csGRP78 leads to the production of autoantibodies as observed in patients with prostate or ovarian cancer, in which the ability to target csGRP78 affects the tumor development. Present on the surface of cancer cells and not normal cells in vivo, csGRP78 represents an interesting target for therapeutic antibody strategies. Here we give an overview of the csGRP78 function in the cell and its role in oncogenesis, thereby providing insight into the clinical value of GRP78 monoclonal antibodies for cancer prognosis and treatment.
Collapse
Affiliation(s)
- Isabelle Hernandez
- Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Translational Research Centre in Oncohaematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Marie Cohen
- Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Translational Research Centre in Oncohaematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
4
|
Lin J, Jiang X, Dong M, Liu X, Shen Q, Huang Y, Zhang H, Ye R, Zhou H, Yan C, Yuan S, Wu X, Chen L, Wang Y, He M, Tao Y, Zhang Z, Jin W. Hepatokine Pregnancy Zone Protein Governs the Diet-Induced Thermogenesis Through Activating Brown Adipose Tissue. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101991. [PMID: 34514733 PMCID: PMC8564441 DOI: 10.1002/advs.202101991] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/23/2021] [Indexed: 05/06/2023]
Abstract
Intermittent fasting (IF), as a dietary intervention for weight loss, takes effects primarily through increasing energy expenditure. However, whether inter-organ systems play a key role in IF remains unclear. Here, a novel hepatokine, pregnancy zone protein (PZP) is identified, which has significant induction during the refeeding stage of IF. Further, loss of function studies and protein therapeutic experiment in mice revealed that PZP promotes diet-induced thermogenesis through activating brown adipose tissue (BAT). Mechanistically, circulating PZP can bind to cell surface glucose-regulated protein of 78 kDa (GRP78) to promote uncoupling protein 1 (UCP1) expression via a p38 MAPK-ATF2 signaling pathway in BAT. These studies illuminate a systemic regulation in which the IF promotes BAT thermogenesis through the endocrinal system and provide a novel potential target for treating obesity and related disorders.
Collapse
Affiliation(s)
- Jun Lin
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
| | - Xiaoxiao Jiang
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Meng Dong
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
| | - Xiaomeng Liu
- Institute of Neuroscience and Translational MedicineCollege of Life Science and AgronomyZhoukou Normal UniversityZhoukou466000China
| | - Qiwei Shen
- Department of General SurgeryHuashan HospitalFudan UniversityShanghaiChina
| | - Yuanyuan Huang
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Hanlin Zhang
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Rongcai Ye
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Huiqiao Zhou
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Chunlong Yan
- College of AgricultureYanbian UniversityYanji133000China
| | - Shouli Yuan
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Xiangnan Wu
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Li Chen
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yanfang Wang
- State Key Laboratory of Animal NutritionInstitute of Animal ScienceChinese Academy of Agricultural SciencesBeijing100193China
| | - Min He
- Division of Endocrinology and MetabolismHuashan HospitalFudan UniversityShanghaiChina
| | - Yi Tao
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
| | - Zhaoyun Zhang
- Division of Endocrinology and MetabolismHuashan HospitalFudan UniversityShanghaiChina
| | - Wanzhu Jin
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
5
|
Indomethacin Disrupts the Formation of β-Amyloid Plaques via an α2-Macroglobulin-Activating lrp1-Dependent Mechanism. Int J Mol Sci 2021; 22:ijms22158185. [PMID: 34360951 PMCID: PMC8348656 DOI: 10.3390/ijms22158185] [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: 06/15/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 12/26/2022] Open
Abstract
Epidemiological studies have implied that the nonsteroidal anti-inflammatory drug (NSAID) indomethacin slows the development and progression of Alzheimer’s disease (AD). However, the underlying mechanisms are notably understudied. Using a chimeric mouse/human amyloid precursor protein (Mo/HuAPP695swe) and a mutant human presenilin 1 (PS1-dE9) (APP/PS1) expressing transgenic (Tg) mice and neuroblastoma (N) 2a cells as in vivo and in vitro models, we revealed the mechanisms of indomethacin in ameliorating the cognitive decline of AD. By screening AD-associated genes, we observed that a marked increase in the expression of α2-macroglobulin (A2M) was markedly induced after treatment with indomethacin. Mechanistically, upregulation of A2M was caused by the inhibition of cyclooxygenase-2 (COX-2) and lipocalin-type prostaglandin D synthase (L-PGDS), which are responsible for the synthesis of prostaglandin (PG)H2 and PGD2, respectively. The reduction in PGD2 levels induced by indomethacin alleviated the suppression of A2M expression through a PGD2 receptor 2 (CRTH2)-dependent mechanism. Highly activated A2M not only disrupted the production and aggregation of β-amyloid protein (Aβ) but also induced Aβ efflux from the brain. More interestingly, indomethacin decreased the degradation of the A2M receptor, low-density lipoprotein receptor-related protein 1 (LRP1), which facilitated the brain efflux of Aβ. Through the aforementioned mechanisms, indomethacin ameliorated cognitive decline in APP/PS1 Tg mice by decreasing Aβ production and clearing Aβ from the brains of AD mice.
Collapse
|
6
|
Yan JB, Lai CC, Jhu JW, Gongol B, Marin TL, Lin SC, Chiu HY, Yen CJ, Wang LY, Peng IC. Insulin and Metformin Control Cell Proliferation by Regulating TDG-Mediated DNA Demethylation in Liver and Breast Cancer Cells. MOLECULAR THERAPY-ONCOLYTICS 2020; 18:282-294. [PMID: 32728616 PMCID: PMC7378318 DOI: 10.1016/j.omto.2020.06.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 06/19/2020] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is a frequent comorbidity of cancer. Hyperinsulinemia secondary to T2DM promotes cancer progression, whereas antidiabetic agents, such as metformin, have anticancer effects. However, the detailed mechanism for insulin and metformin-regulated cancer cell proliferation remains unclear. This study identified a mechanism by which insulin upregulated the expression of c-Myc, sterol regulatory element-binding protein 1 (SREBP1), and acetyl-coenzyme A (CoA) carboxylase 1 (ACC1), which are important regulators of lipogenesis and cell proliferation. Thymine DNA glycosylase (TDG), a DNA demethylase, was transactivated by c-Myc upon insulin treatment, thereby decreasing 5-carboxylcytosine (5caC) abundance in the SREBP1 promoter. On the other hand, metformin-activated AMP-activated protein kinase (AMPK) increased DNA methyltransferase 3A (DNMT3A) activity to increase 5-methylcytosine (5mC) abundance in the TDG promoter. This resulted in decreased TDG expression and enhanced 5caC abundance in the SREBP1 promoter. These findings demonstrate that c-Myc activates, whereas AMPK inhibits, TDG-mediated DNA demethylation of the SREBP1 promoter in insulin-promoted and metformin-suppressed cancer progression, respectively. This study indicates that TDG is an epigenetic-based therapeutic target for cancers associated with T2DM.
Collapse
Affiliation(s)
- Jia-Bao Yan
- Department of Life Sciences, National Cheng Kung University, Tainan City 701, Taiwan
| | - Chien-Cheng Lai
- Department of Life Sciences, National Cheng Kung University, Tainan City 701, Taiwan
| | - Jin-Wei Jhu
- Department of Life Sciences, National Cheng Kung University, Tainan City 701, Taiwan
| | - Brendan Gongol
- Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA
| | - Traci L Marin
- Department of Health Sciences, Victor Valley College, Victorville, CA 92395, USA
| | - Shih-Chieh Lin
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City 701, Taiwan
| | - Hsiang-Yi Chiu
- Department of Life Sciences, National Cheng Kung University, Tainan City 701, Taiwan
| | - Chia-Jui Yen
- Division of Hematology and Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan City 701, Taiwan
| | - Liang-Yi Wang
- Department of Public Health, National Cheng Kung University, Tainan City 701, Taiwan
| | - I-Chen Peng
- Department of Life Sciences, National Cheng Kung University, Tainan City 701, Taiwan
| |
Collapse
|
7
|
Liu C, Li Z, Xu L, Shi Y, Zhang X, Shi S, Hou K, Fan Y, Li C, Wang X, Zhou L, Liu Y, Qu X, Che X. GALNT6 promotes breast cancer metastasis by increasing mucin-type O-glycosylation of α2M. Aging (Albany NY) 2020; 12:11794-11811. [PMID: 32559179 PMCID: PMC7343513 DOI: 10.18632/aging.103349] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 05/14/2020] [Indexed: 01/22/2023]
Abstract
Breast cancer is the most lethal malignancy in women. N-acetylgalactosaminyltransferase 6 (GALNT6) is an enzyme which mediates the initial step of mucin-type O-glycosylation, and has been reported to be involved in mammary carcinogenesis. However, the molecular mechanism of GALNT6 in breast cancer metastasis has not been fully explored. In this study, based on online database analyses and tissue microarrays, the overall survival (OS) of breast cancer patients with high expression of GALNT6 was found to be shorter than those with low expression of GALNT6. Also, high GALNT6 expression was positively correlated with advanced pN stage and pTNM stage. GALNT6 was shown to be able to promote the migration and invasion of breast cancer cells, and enhance the level of mucin-type O-glycosylation of substrates in the supernatants of breast cancer cells. Qualitative mucin-type glycosylomics analysis identified α2M as a novel substrate of GALNT6. Further investigation showed that GALNT6 increased O-glycosylation of α2M, and the following activation of the downstream PI3K/Akt signaling pathway was involved in the promotion of migration and invasion of breast cancer cells. This study identified a new substrate of GALNT6 and provides novel understanding of the role of GALNT6 in promoting metastasis and poor prognosis in breast cancer.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Breast/pathology
- Breast/surgery
- Breast Neoplasms/diagnosis
- Breast Neoplasms/mortality
- Breast Neoplasms/pathology
- Breast Neoplasms/surgery
- Breast Neoplasms, Male/diagnosis
- Breast Neoplasms, Male/mortality
- Breast Neoplasms, Male/pathology
- Breast Neoplasms, Male/surgery
- Carcinoma, Ductal, Breast/diagnosis
- Carcinoma, Ductal, Breast/mortality
- Carcinoma, Ductal, Breast/secondary
- Carcinoma, Ductal, Breast/surgery
- Cell Line, Tumor
- Datasets as Topic
- Female
- Follow-Up Studies
- Glycosylation
- Humans
- Kaplan-Meier Estimate
- Male
- Mastectomy
- Middle Aged
- N-Acetylgalactosaminyltransferases/metabolism
- Neoplasm Metastasis/pathology
- Neoplasm Staging
- Phosphatidylinositol 3-Kinases/metabolism
- Prognosis
- Proto-Oncogene Proteins c-akt/metabolism
- Signal Transduction
- Tissue Array Analysis
- alpha-Macroglobulins/metabolism
- Polypeptide N-acetylgalactosaminyltransferase
Collapse
Affiliation(s)
- Chang Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
- Department of Internal Medicine, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China
| | - Zhi Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Lu Xu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yu Shi
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiaojie Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Sha Shi
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Kezuo Hou
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yibo Fan
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Ce Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiaoxun Wang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Lu Zhou
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiaofang Che
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
8
|
Thiebaut AM, Hedou E, Marciniak SJ, Vivien D, Roussel BD. Proteostasis During Cerebral Ischemia. Front Neurosci 2019; 13:637. [PMID: 31275110 PMCID: PMC6594416 DOI: 10.3389/fnins.2019.00637] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/03/2019] [Indexed: 12/21/2022] Open
Abstract
Cerebral ischemia is a complex pathology involving a cascade of cellular mechanisms, which deregulate proteostasis and lead to neuronal death. Proteostasis refers to the equilibrium between protein synthesis, folding, transport, and protein degradation. Within the brain proteostasis plays key roles in learning and memory by controlling protein synthesis and degradation. Two important pathways are implicated in the regulation of proteostasis: the unfolded protein response (UPR) and macroautophagy (called hereafter autophagy). Both are necessary for cell survival, however, their over-activation in duration or intensity can lead to cell death. Moreover, UPR and autophagy can activate and potentiate each other to worsen the issue of cerebral ischemia. A better understanding of autophagy and ER stress will allow the development of therapeutic strategies for stroke, both at the acute phase and during recovery. This review summarizes the latest therapeutic advances implicating ER stress or autophagy in cerebral ischemia. We argue that the processes governing proteostasis should be considered together in stroke, rather than focusing either on ER stress or autophagy in isolation.
Collapse
Affiliation(s)
- Audrey M Thiebaut
- INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, University of Caen Normandy, Caen, France
| | - Elodie Hedou
- INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, University of Caen Normandy, Caen, France
| | - Stefan J Marciniak
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom.,Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Denis Vivien
- INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, University of Caen Normandy, Caen, France.,Department of Clinical Research, University of Caen Normandy, Caen, France
| | - Benoit D Roussel
- INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, University of Caen Normandy, Caen, France
| |
Collapse
|
9
|
Huang YX, Song H, Tao Y, Shao XB, Zeng XS, Xu XL, Qi JL, Sun JF. Ovostatin 2 knockdown significantly inhibits the growth, migration, and tumorigenicity of cutaneous malignant melanoma cells. PLoS One 2018; 13:e0195610. [PMID: 29684087 PMCID: PMC5912766 DOI: 10.1371/journal.pone.0195610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 03/26/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND We previously identified ovostatin 2 (OVOS2) as a new candidate gene for cutaneous malignant melanoma (CMM) in a Chinese population. In this study, we aimed to investigate the exact role of OVOS2 in cell proliferation, invasion, and tumorigenesis of melanoma A375 cells. METHODS The downregulation of OVOS2 expression was performed using lentiviral vectors with specific shRNA. The effects of OVOS2 expression on cell proliferation, cell cycle, cell migration, cell invasion, and potential of tumorigenesis were further investigated. RESULTS The downregulation of OVOS2 significantly suppressed the proliferation of A375 cells and led to a G2/M phase block. The transwell cell migration assay showed that the reduced expression of OVOS2 also significantly inhibited the transmigration of A375 cells. The western blot results showed downregulated expression of p-FAK, p-AKT, and p-ERK. This was accompanied by the upregulated epithelial phenotypes E-cadherin and β-catenin, and downregulated expression of mesenchymal phenotype N-cadherin after OVOS2 knockdown. The transplantation tumor experiment in BALB/C nude mouse showed that after an observation period of 32 days, the growth speed and weight of the transplanted tumors were significantly suppressed in the BALB/c nude mice subcutaneously injected with OVOS2 knocked-down A375 cells. CONCLUSION The inhibition of OVOS2 had significant suppressive effects on the proliferation, motility, and migration capabilities of A375 cells, suggesting a crucial promotive role of OVOS2 in the pathogenesis and progression of CMM. The involved mechanisms are at least partly associated with the overactivation of FAK/MAPK/ERK and FAK/PI3K/AKT signals.
Collapse
Affiliation(s)
- Ying-Xue Huang
- Institute of Dermatology, Chinese Academy of Medical Sciences, Nanjing, P. R. China
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, P. R. China
| | - Hao Song
- Institute of Dermatology, Chinese Academy of Medical Sciences, Nanjing, P. R. China
| | - Yue Tao
- Drum Tower Hospital, Medical School of Nanjing University, Nanjing, P. R. China
| | - Xue-Bao Shao
- Institute of Dermatology, Chinese Academy of Medical Sciences, Nanjing, P. R. China
| | - Xue-Si Zeng
- Institute of Dermatology, Chinese Academy of Medical Sciences, Nanjing, P. R. China
| | - Xiu-Lian Xu
- Institute of Dermatology, Chinese Academy of Medical Sciences, Nanjing, P. R. China
| | - Jin-Liang Qi
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, P. R. China
| | - Jian-Fang Sun
- Institute of Dermatology, Chinese Academy of Medical Sciences, Nanjing, P. R. China
| |
Collapse
|
10
|
mTOR inhibitors activate PERK signaling and favor viability of gastrointestinal neuroendocrine cell lines. Oncotarget 2017; 8:20974-20987. [PMID: 28423496 PMCID: PMC5400559 DOI: 10.18632/oncotarget.15469] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 02/06/2017] [Indexed: 02/06/2023] Open
Abstract
mTOR and Unfolded Protein Response (UPR) are two signaling pathways frequently activated in cancer cells. The mTOR pathway has been shown to be up-regulated in most gastroenteropancreatic neuroendocrine tumors. In contrast, little is known about the UPR status in neoplastic neuroendocrine cells. However, these hormone-producing cells are likely to present distinctive adaptations of this pathway, as other secretory cells. We therefore analyzed the status of the three axes of UPR and their relation to mTOR pathway in two gastrointestinal neuroendocrine tumors (GI-NET) cell lines STC-1 and GluTag. At baseline, pharmacological inducers activate the three arms of UPR: PERK, ATF6 and IRE1. Although hypoxia stimulates the PERK, ATF6 and IRE-1 pathways in both cell lines, glucose depletion activates UPR only in STC-1 cell line. Strikingly, P-p70S6K1 increases concomitantly to P-PERK and BiP in response to thapsigargin treatment, glucose depletion or hypoxia. We found that different mTOR inhibitors activate the PERK signaling pathway. To confirm that mTOR inhibition modulates PERK activation, we inhibited PERK and showed that it decreased cell viability when associated to mTOR inhibition, indicating that mTOR drives a PERK-dependent survival pathway. In conclusion, in GI-NET cell lines, UPR signaling is functional and PERK arm is induced by mTOR inhibition. These observations open up new perspectives for therapeutic strategies: the crosstalk between mTOR and UPR might contribute to the resistance to mTOR inhibitors and could be targeted by mTOR and PERK inhibitors in combination therapy.
Collapse
|
11
|
Bellezza I, Scarpelli P, Pizzo SV, Grottelli S, Costanzi E, Minelli A. ROS-independent Nrf2 activation in prostate cancer. Oncotarget 2017; 8:67506-67518. [PMID: 28978049 PMCID: PMC5620189 DOI: 10.18632/oncotarget.18724] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 05/23/2017] [Indexed: 12/16/2022] Open
Abstract
In prostate cancer, oxidative stress and the subsequent Nrf2 activation promote the survival of cancer cells and acquired chemoresistance. Nrf2 links prostate cancer to endoplasmic reticulum stress, an event that triggers the unfolded protein response, aiming to restore cellular homeostasis as well as an adaptive survival mechanism. Glucose-regulated protein of 78 kD /immunoglobulin heavy chain binding protein (GRP78/BiP) is a key molecular chaperone in the endoplasmic reticulum that, when expressed at the cell surface, acts as a receptor for several signaling pathways enhancing antiapoptotic and proliferative signals. We showed GRP78/BiP translocation to PC3 cell surface in the presence of tunicamycin, an ER stress inductor, and demonstrated the existence of a GRP78/BiP-dependent non-canonical Nrf2 activation, responsible for increased resistance to ER-stress induced apoptosis. We found that, even in the absence of ROS production, tunicamycin causes Nrf2 activation, and activates Akt signaling, events bulnted by anti-GRP78/BiP antibody treatment. The presence of GRP78/BiP at the cell surface might be exploited for the immunotherapeutic strategy of prostate cancer since its blockage by anti-GRP78/BiP antibodies might promote cancer death by suppressing some of the several molecular protective mechanisms found in aggressive cancer cells.
Collapse
Affiliation(s)
- Ilaria Bellezza
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Paolo Scarpelli
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Silvia Grottelli
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Egidia Costanzi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Alba Minelli
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
12
|
Yip CW, Lam CY, Poon TCW, Cheung TT, Cheung PFY, Fung SW, Wang XQ, Leung ICY, Ng LWC, Lo CM, Tsao GSW, Cheung ST. Granulin-epithelin precursor interacts with 78-kDa glucose-regulated protein in hepatocellular carcinoma. BMC Cancer 2017; 17:409. [PMID: 28601093 PMCID: PMC5466756 DOI: 10.1186/s12885-017-3399-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/01/2017] [Indexed: 08/21/2023] Open
Abstract
Background Granulin-epithelin precursor (GEP) is a secretory growth factor, which has been demonstrated to control cancer growth, invasion, drug resistance and immune escape. Our previous studies and others also demonstrated its potential in targeted therapy. Comprehensive characterization of GEP partner on cancer cells are warranted. We have previously shown that GEP interacted with heparan sulfate on the surface of liver cancer cells and the interaction is crucial for GEP-mediated signaling transduction. This study aims to characterize GEP protein partner at the cell membrane with the co-immunoprecipitation and mass spectrometry approach. Methods The membrane fraction from liver cancer model Hep3B was used for capturing binding partner with the specific monoclonal antibody against GEP. The precipitated proteins were analyzed by mass spectrometry. After identifying the GEP binding partner, this specific interaction was validated in additional liver cancer cell line HepG2 by co-immunoprecipitation using GRP78 and GEP antibodies, respectively, as the bait. GRP78 transcript levels in hepatocellular carcinoma (HCC) clinical samples (n = 77 pairs) were examined by real-time quantitative RT-PCR. GEP and GRP78 protein expressions were investigated by immunohistochemistry on paraffin sections. Results We identified the GEP-binding protein as 78-kDa glucose-regulated protein (GRP78, also named heat shock 70-kDa protein 5, HSPA5). This interaction was validated in independent HCC cell lines. Increased GRP78 mRNA levels were demonstrated in liver cancer tissues compared with the paralleled liver tissues (t-test, P = 0.002). GRP78 and GEP transcript levels were significantly correlated (Spearman’s correlation, P = 0.001), and the proteins were also detectable in the cytoplasm of liver cancer cells by immunohistochemical staining. Conclusions GRP78 and GEP are interacting protein partners in liver cancer cells and may play a role in GEP-mediated cancer progression in HCC.
Collapse
Affiliation(s)
- Chi Wai Yip
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China.,Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - Ching Yan Lam
- Department of Surgery, The University of Hong Kong, Hong Kong, China.,Department of Health, The Government of the Hong Kong Special Administrative Region, Hong Kong, China
| | | | - Tan To Cheung
- Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - Phyllis F Y Cheung
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China.,Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong, China
| | - Sze Wai Fung
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China.,Department of Surgery, The University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Xiao Qi Wang
- Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - Idy C Y Leung
- Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - Linda W C Ng
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Chung Mau Lo
- Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - George S W Tsao
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Siu Tim Cheung
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China. .,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China. .,Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.
| |
Collapse
|
13
|
Carli AV, Harvey EJ, Azeddine B, Gao C, Li Y, Li A, Sayegh M, Wang H, Nahal A, Michel RP, Henderson JE, Séguin C. Substrain-specific differences in bone parameters, alpha-2-macroglobulin circulating levels, and osteonecrosis incidence in a rat model. J Orthop Res 2017; 35:1183-1194. [PMID: 26895739 DOI: 10.1002/jor.23199] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 02/08/2016] [Indexed: 02/04/2023]
Abstract
Osteonecrosis of the femoral head (ONFH) is a potentially devastating complication that occurs in up to 40% of young adults receiving chronic glucocorticoid (GC) therapy. Through a validated GC therapy rat model, we have previously shown that Wistar Kyoto (WK) rats exhibit a genetic susceptibility to GC-induced ONFH compared to Sasco Fischer (F344) rats. We have undertaken this study in order to investigate differences between these two strains for their bone parameters, alpha-2-macroglobulin (A2M) circulating levels and incidence of GC-induced osteonecrosis of the femoral head. WK and F344 rats were treated either with 1.5 mg/kg/day of prednisone or placebo for 6 months. Blood was taken every month. The femoral heads were harvested for histological examination to detect ONFH and analyzed with micro-computed tomography. After 3 months of GC-therapy, plasma A2M was elevated in treated rats only. GC-treated WK rats exhibited histological evidence of early ONFH through higher rates of cellular apoptosis and empty osteocyte lacunae in the subchondral bone compared to placebos and to F344 rats. Furthermore, micro-CT analysis exhibited femoral head collapse only in GC-treated WK rats. Interestingly, GC-treated F344 rats exhibited significant micro-CT changes, but such changes were less concentrated in the articular region and were accompanied histologically with increased marrow fat. These µCT and histological findings suggest that elevated A2M serum level is not predictive and suitable as an indicative biomarker for early GC-induced ONFH in rodents. Elevated A2M levels observed during GC treatment suggests that it plays role in the host reparative response to GC-associated effects. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:1183-1194, 2017.
Collapse
Affiliation(s)
- Alberto V Carli
- Vascular, Biology Research Lab, Research Institute, McGill University Health Centre, C9 Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada.,Bone Engineering Labs, Surgical Research, Research Institute, McGill University Health Centre, Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada.,Department of Surgery, Division of Orthopaedic Surgery, McGill University Health Centre, B5 Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada
| | - Edward J Harvey
- Vascular, Biology Research Lab, Research Institute, McGill University Health Centre, C9 Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada.,Bone Engineering Labs, Surgical Research, Research Institute, McGill University Health Centre, Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada.,Department of Surgery, Division of Orthopaedic Surgery, McGill University Health Centre, B5 Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada
| | - Bouziane Azeddine
- Vascular, Biology Research Lab, Research Institute, McGill University Health Centre, C9 Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada
| | - Chan Gao
- Bone Engineering Labs, Surgical Research, Research Institute, McGill University Health Centre, Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada
| | - Yongbiao Li
- Vascular, Biology Research Lab, Research Institute, McGill University Health Centre, C9 Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada
| | - Ailian Li
- Bone Engineering Labs, Surgical Research, Research Institute, McGill University Health Centre, Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada
| | - Mireille Sayegh
- Vascular, Biology Research Lab, Research Institute, McGill University Health Centre, C9 Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada
| | - Huifen Wang
- Bone Engineering Labs, Surgical Research, Research Institute, McGill University Health Centre, Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada
| | - Ayoub Nahal
- Department of Pathology, McGill University Health Centre (MUHC), C3 Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada and McGill University Health Centre (MUHC), Glen site, 1001 Décarie Blvd, Montreal, QC H4A 3J1, Canada
| | - René P Michel
- Department of Pathology, McGill University Health Centre (MUHC), C3 Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada and McGill University Health Centre (MUHC), Glen site, 1001 Décarie Blvd, Montreal, QC H4A 3J1, Canada
| | - Janet E Henderson
- Bone Engineering Labs, Surgical Research, Research Institute, McGill University Health Centre, Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada
| | - Chantal Séguin
- Vascular, Biology Research Lab, Research Institute, McGill University Health Centre, C9 Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada.,Department of Medicine, Division of Hematology and Oncology, McGill University Health Centre, Glen site, 1001 Décarie Blvd, room D02-7519, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
14
|
Zhou W, Liu L, Xue Y, Zheng J, Liu X, Ma J, Li Z, Liu Y. Combination of Endothelial-Monocyte-Activating Polypeptide-II with Temozolomide Suppress Malignant Biological Behaviors of Human Glioblastoma Stem Cells via miR-590-3p/MACC1 Inhibiting PI3K/AKT/mTOR Signal Pathway. Front Mol Neurosci 2017; 10:68. [PMID: 28348518 PMCID: PMC5346543 DOI: 10.3389/fnmol.2017.00068] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 02/28/2017] [Indexed: 12/17/2022] Open
Abstract
This study aims to investigate the effect of Endothelial-Monocyte-Activating Polypeptide-II (EMAP-II) combined with temozolomide (TMZ) upon glioblastoma stem cells (GSCs) and its possible molecular mechanisms. In this study, combination of EMAP-II with TMZ inhibited cell viability, migration and invasion in GSCs, and autophagy inhibitor 3-methyl adenine (3-MA) and chloroquine (CQ) partly reverse the anti-proliferative effect of the combination treatment. Autophagic vacuoles were formed in GSCs after the combination therapy, accompanied with the up-regulation of LC3-II and Beclin-1 as well as the down-regulation of p62/SQSTM1. Further, miR-590-3p was up-regulated and Metastasis-associated in colon cancer 1 (MACC1) was down-regulated by the combination treatment in GSCs; MiR-590-3p overexpression and MACC1 knockdown up-regulated LC3-II and Beclin-1 as well as down-regulated p62/SQSTM1 in GSCs; MACC1 was identified as a direct target of miR-590-3p, mediating the effects of miR-590-3p in the combination treatment. Furthermore, the combination treatment and MACC1 knockdown decreased p-PI3K, p-Akt, p-mTOR, p-S6 and p-4EBP in GSCs; PI3K/Akt agonist insulin-like growth factor-1(IGF-1) partly blocked the effect of the combination treatment. Moreover, in vivo xenograft models, the mice given stable overexpressed miR-590-3p cells and treated with EMAP-II and TMZ had the smallest tumor sizes, besides, miR-590-3p + EMAP-II + TMZ up-regulated the expression level of miR-590-3p, LC3-II and Beclin-1 as well as down-regulated p62/SQSTM1. In conclusion, these results elucidated anovel molecular mechanism of EMAP-II in combination with TMZ suppressed malignant biological behaviors of GSCs via miR-590-3p/MACC1 inhibiting PI3K/AKT/mTOR signaling pathway, and might provide potential therapeutic approaches for human GSCs.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Neurosurgery, Shengjing Hospital of China Medical UniversityShenyang, China; Liaoning Research Center for Translational Medicine in Nervous System DiseaseShenyang, China
| | - Libo Liu
- Department of Neurobiology, College of Basic Medicine, China Medical UniversityShenyang, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical UniversityShenyang, China
| | - Yixue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical UniversityShenyang, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical UniversityShenyang, China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical UniversityShenyang, China; Liaoning Research Center for Translational Medicine in Nervous System DiseaseShenyang, China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical UniversityShenyang, China; Liaoning Research Center for Translational Medicine in Nervous System DiseaseShenyang, China
| | - Jun Ma
- Department of Neurobiology, College of Basic Medicine, China Medical UniversityShenyang, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical UniversityShenyang, China
| | - Zhen Li
- Department of Neurosurgery, Shengjing Hospital of China Medical UniversityShenyang, China; Liaoning Research Center for Translational Medicine in Nervous System DiseaseShenyang, China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical UniversityShenyang, China; Liaoning Research Center for Translational Medicine in Nervous System DiseaseShenyang, China
| |
Collapse
|
15
|
Gopal U, Gonzalez-Gronow M, Pizzo SV. Activated α2-Macroglobulin Regulates Transcriptional Activation of c-MYC Target Genes through Cell Surface GRP78 Protein. J Biol Chem 2016; 291:10904-15. [PMID: 27002159 DOI: 10.1074/jbc.m115.708131] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Indexed: 12/25/2022] Open
Abstract
Activated α2-macroglobulin (α2M*) signals predominantly through cell surface GRP78 (CS-GRP78) to promote proliferation and survival of cancer cells; however, the molecular mechanism remains obscure. c-MYC is an essential transcriptional regulator that controls cell proliferation. We hypothesize that α2M*/CS-GRP78-evoked key signaling events are required for transcriptional activation of c-MYC target genes. Activation of CS-GRP78 by α2M* requires ligation of the GRP78 primary amino acid sequence (Leu(98)-Leu(115)). After stimulation with α2M*, CS-GRP78 signaling activates 3-phosphoinositide-dependent protein kinase-1 (PDK1) to induce phosphorylation of PLK1, which in turn induces c-MYC transcription. We demonstrate that PLK1 binds directly to c-MYC and promotes its transcriptional activity by phosphorylating Ser(62) Moreover, activated c-MYC is recruited to the E-boxes of target genes FOSL1 and ID2 by phosphorylating histone H3 at Ser(10) In addition, targeting the carboxyl-terminal domain of CS-GRP78 with a mAb suppresses transcriptional activation of c-MYC target genes and impairs cell proliferation. This work demonstrates that α2M*/CS-GRP78 acts as an upstream regulator of the PDK1/PLK1 signaling axis to modulate c-MYC transcription and its target genes, suggesting a therapeutic strategy for targeting c-MYC-associated malignant progression.
Collapse
Affiliation(s)
- Udhayakumar Gopal
- From the Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710
| | - Mario Gonzalez-Gronow
- From the Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710
| | - Salvatore Vincent Pizzo
- From the Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710
| |
Collapse
|
16
|
Vlachostergios PJ, Balmiki RL, Daya R. GRP78 and α2-macroglobulin are new promising targets for metastatic castrate-resistant prostate cancer treatment. Clin Transl Oncol 2015; 17:932-4. [DOI: 10.1007/s12094-015-1324-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 06/05/2015] [Indexed: 10/23/2022]
|
17
|
Abstract
Over 230,000 new cases of invasive breast cancer are diagnosed annually within the USA. Recurrent breast cancer remains a mostly incurable disease with drug resistance, tumor latency and distant metastases driving breast tumor recurrence and morbidity. Understanding drug resistance is a critical component of combating breast cancer. Recently, the protein chaperone GRP78 and the unfolded protein response were implicated as drivers of drug resistance. Preclinical studies show inhibiting GRP78 can reverse drug resistance. Furthermore, drugs developed to target GRP78 show clinical promise in several ongoing clinical trials.
Collapse
|
18
|
Yao X, Liu H, Zhang X, Zhang L, Li X, Wang C, Sun S. Cell Surface GRP78 Accelerated Breast Cancer Cell Proliferation and Migration by Activating STAT3. PLoS One 2015; 10:e0125634. [PMID: 25973748 PMCID: PMC4431800 DOI: 10.1371/journal.pone.0125634] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 03/16/2015] [Indexed: 11/18/2022] Open
Abstract
High levels of cell surface glucose regulated protein 78 (sGRP78) have been implicated in cancer growth, survival, metastasis, and chemotherapy resistance. However, the underlying mechanism remains largely unknown. Here we report that the level of sGRP78 expression in human breast tumors gradually increases during cancer progression. Overexpression of GRP78 significantly enhanced its membrane distribution in human MCF-7 breast cancer cells, but had no effect on endoplasmic reticulum (ER) stress. High levels of sGRP78 facilitated cell proliferation and migration, as well as suppressed cell apoptosis. Neutralization of sGRP78 by a specific antibody against GRP78 alleviated sGRP78-induced cell growth and migration. Importantly, high phosphorylation levels of the signal transducer and activator of transcription 3 (STAT3) were found in human breast tumors that express sGRP78 and MCF-7 cells infected with adenovirus encoding human GRP78. Pretreatment with a GRP78 antibody suppressed STAT3 phosphorylation. Furthermore, genetic and pharmacological inhibition of STAT3 reversed the impacts of GRP78 on cell proliferation, apoptosis, and migration. These findings indicate that STAT3 mediates sGRP78-promoted breast cancer cell growth and migration.
Collapse
Affiliation(s)
- Xiaoli Yao
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Hua Liu
- Department of Clinical Pathology, The First People’s Hospital of Lianyungang, Lianyungang, 222000, Jiangsu Province, China
- Department of Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, Hubei Province, China
| | - Xinghua Zhang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Liang Zhang
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Xiang Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Changhua Wang
- Department of Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, Hubei Province, China
| | - Shengrong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
- * E-mail:
| |
Collapse
|
19
|
Selection and identification of ligand peptides targeting a model of castrate-resistant osteogenic prostate cancer and their receptors. Proc Natl Acad Sci U S A 2015; 112:3776-81. [PMID: 25762070 DOI: 10.1073/pnas.1500128112] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
We performed combinatorial peptide library screening in vivo on a novel human prostate cancer xenograft that is androgen-independent and induces a robust osteoblastic reaction in bonelike matrix and soft tissue. We found two peptides, PKRGFQD and SNTRVAP, which were enriched in the tumors, targeted the cell surface of androgen-independent prostate cancer cells in vitro, and homed to androgen receptor-null prostate cancer in vivo. Purification of tumor homogenates by affinity chromatography on these peptides and subsequent mass spectrometry revealed a receptor for the peptide PKRGFQD, α-2-macroglobulin, and for SNTRVAP, 78-kDa glucose-regulated protein (GRP78). These results indicate that GRP78 and α-2-macroglobulin are highly active in osteoblastic, androgen-independent prostate cancer in vivo. These previously unidentified ligand-receptor systems should be considered for targeted drug development against human metastatic androgen-independent prostate cancer.
Collapse
|
20
|
Misra UK, Pizzo SV. Activated α2-macroglobulin binding to human prostate cancer cells triggers insulin-like responses. J Biol Chem 2015; 290:9571-87. [PMID: 25720493 DOI: 10.1074/jbc.m114.617837] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Indexed: 12/21/2022] Open
Abstract
Ligation of cell surface GRP78 by activated α2-macroglobulin (α2M*) promotes cell proliferation and suppresses apoptosis. α2M*-treated human prostate cancer cells exhibit a 2-3-fold increase in glucose uptake and lactate secretion, an effect similar to insulin treatment. In both α2M* and insulin-treated cells, the mRNA levels of SREBP1-c, SREBP2, fatty-acid synthase, acetyl-CoA carboxylase, ATP citrate lyase, and Glut-1 were significantly increased together with their protein levels, except for SREBP2. Pretreatment of cells with α2M* antagonist antibody directed against the carboxyl-terminal domain of GRP78 blocks these α2M*-mediated effects, and silencing GRP78 expression by RNAi inhibits up-regulation of ATP citrate lyase and fatty-acid synthase. α2M* induces a 2-3-fold increase in lipogenesis as determined by 6-[(14)C]glucose or 1-[(14)C]acetate incorporation into free cholesterol, cholesterol esters, triglycerides, free fatty acids, and phosphatidylcholine, which is blocked by inhibitors of fatty-acid synthase, PI 3-kinase, mTORC, or an antibody against the carboxyl-terminal domain of GRP78. We also assessed the incorporation of [(14)CH3]choline into phosphatidylcholine and observed similar effects. Lipogenesis is significantly affected by pretreatment of prostate cancer cells with fatostatin A, which blocks sterol regulatory element-binding protein proteolytic cleavage and activation. This study demonstrates that α2M* functions as a growth factor, leading to proliferation of prostate cancer cells by promoting insulin-like responses. An antibody against the carboxyl-terminal domain of GRP78 may have important applications in prostate cancer therapy.
Collapse
Affiliation(s)
- Uma Kant Misra
- From the Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710
| | - Salvatore Vincent Pizzo
- From the Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710
| |
Collapse
|
21
|
Wang H, Pezeshki AM, Yu X, Guo C, Subjeck JR, Wang XY. The Endoplasmic Reticulum Chaperone GRP170: From Immunobiology to Cancer Therapeutics. Front Oncol 2015; 4:377. [PMID: 25629003 PMCID: PMC4290550 DOI: 10.3389/fonc.2014.00377] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 12/16/2014] [Indexed: 01/09/2023] Open
Abstract
Glucose-regulated protein 170 (GRP170) is the largest member of glucose-regulated protein family that resides in the endoplasmic reticulum (ER). As a component of the ER chaperone network, GRP170 assists in protein folding, assembly, and transportation of secretory or transmembrane proteins. The well documented cytoprotective activity of intracellular GRP170 due to its intrinsic chaperoning property has been shown to provide a survival benefit in cancer cells during tumor progression or metastasis. Accumulating evidence shows that extracellular GRP170 displays a superior capacity in delivering tumor antigens to specialized antigen-presenting cells for cross-presentation, resulting in generation of an anti-tumor immune response dependent on cytotoxic CD8+ T cells. This unique feature of GRP170 provides a molecular basis for using GRP170 as an immunostimulatory adjuvant to develop a recombinant vaccine for therapeutic immunization against cancers. This review summarizes the latest findings in understanding the biological effects of GRP170 on cell functions and tumor progression. The immunomodulating activities of GRP170 during interactions with the innate and adaptive arms of the immune system as well as its therapeutic applications in cancer immunotherapy will be discussed.
Collapse
Affiliation(s)
- Hongxia Wang
- Department of Human Molecular Genetics, Virginia Commonwealth University , Richmond, VA , USA
| | - Abdul Mohammad Pezeshki
- Department of Human Molecular Genetics, Virginia Commonwealth University , Richmond, VA , USA
| | - Xiaofei Yu
- Department of Human Molecular Genetics, Virginia Commonwealth University , Richmond, VA , USA
| | - Chunqing Guo
- Department of Human Molecular Genetics, Virginia Commonwealth University , Richmond, VA , USA
| | - John R Subjeck
- Department of Cell Stress Biology, Roswell Park Cancer Institute , Buffalo, NY , USA
| | - Xiang-Yang Wang
- Department of Human Molecular Genetics, Virginia Commonwealth University , Richmond, VA , USA ; Massey Cancer Center, Virginia Commonwealth University , Richmond, VA , USA ; Institute of Molecular Medicine, Virginia Commonwealth University , Richmond, VA , USA
| |
Collapse
|
22
|
Zhao L, Li H, Shi Y, Wang G, Liu L, Su C, Su R. Nanoparticles inhibit cancer cell invasion and enhance antitumor efficiency by targeted drug delivery via cell surface-related GRP78. Int J Nanomedicine 2014; 10:245-56. [PMID: 25565817 PMCID: PMC4283987 DOI: 10.2147/ijn.s74868] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Nanoparticles (NPs) which target specific agents could effectively recognize the target cells and increase the stability of chemical agents by encapsulation. As such, NPs have been widely used in cancer treatment research. Recently, over 90% of treatment failure cases in patients with metastatic cancer were attributed to resistance to chemotherapy. Surface-exposed glucose-regulated protein of 78 kDa (GRP78) is expressed highly on many tumor cell surfaces in many human cancers and is related to the regulation of invasion and metastasis. Herein, we report that NPs conjugated with antibody against GRP78 (mAb GRP78-NPs) inhibit the adhesion, invasion, and metastasis of hepatocellular carcinoma (HCC) and promote drug delivery of 5-fluorouracil into GRP78 high-expressed human hepatocellular carcinoma cells. Our new findings suggest that mAb GRP78-NPs could enhance drug accumulation by effectively transporting NPs into cell surface GRP78-overexpressed human hepatocellular carcinoma cells and then inhibit cell proliferation and viability and induce cell apoptosis by regulating caspase-3. In brief, mAb GRP78-NPs effectively inhibit cancer cell invasion and enhance antitumor efficiency by targeted drug delivery.
Collapse
Affiliation(s)
- Liang Zhao
- School of Pharmacy, Liaoning Medical University, Jinzhou, People's Republic of China
| | - Hongdan Li
- Central Laboratory of Liaoning Medical University, Jinzhou, People's Republic of China
| | - Yijie Shi
- School of Pharmacy, Liaoning Medical University, Jinzhou, People's Republic of China
| | - Guan Wang
- Central Laboratory of Liaoning Medical University, Jinzhou, People's Republic of China
| | - Liwei Liu
- School of Pharmacy, Liaoning Medical University, Jinzhou, People's Republic of China
| | - Chang Su
- School of Veterinary Medicine, Liaoning Medical University, Jinzhou, People's Republic of China
| | - Rongjian Su
- Central Laboratory of Liaoning Medical University, Jinzhou, People's Republic of China
| |
Collapse
|
23
|
Abstract
The glucose-regulated proteins (GRPs) are stress-inducible chaperones that mostly reside in the endoplasmic reticulum or the mitochondria. Recent advances show that the GRPs have functions that are distinct from those of the related heat shock proteins, and they can be actively translocated to other cellular locations and assume novel functions that control signalling, proliferation, invasion, apoptosis, inflammation and immunity. Mouse models further identified their specific roles in development, tumorigenesis, metastasis and angiogenesis. This Review describes their discovery and regulation, as well as their biological functions in cancer. Promising agents that use or target the GRPs are being developed, and their efficacy as anticancer therapeutics is also discussed.
Collapse
Affiliation(s)
- Amy S Lee
- Department of Biochemistry and Molecular Biology, University of Southern California Norris Comprehensive Cancer Center, 1441 Eastlake Avenue, Room 5308, Los Angeles, California 900899176, USA
| |
Collapse
|
24
|
Feng C, He K, Zhang C, Su S, Li B, Li Y, Duan CY, Chen S, Chen R, Liu Y, Li H, Wei M, Xia X, Dai R. JNK contributes to the tumorigenic potential of human cholangiocarcinoma cells through the mTOR pathway regulated GRP78 induction. PLoS One 2014; 9:e90388. [PMID: 24587347 PMCID: PMC3938720 DOI: 10.1371/journal.pone.0090388] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 01/28/2014] [Indexed: 12/14/2022] Open
Abstract
Less is known about the roles of c-Jun N-terminal kinase (JNK) in cholangiocarcinoma (CCA). Here, we report that JNK exerts its oncogenic action in human CCA cells, partially due to the mammalian target of rapamycin (mTOR) pathway regulated glucose-regulated protein 78 (GRP78) induction. In human CCA cells, the phosphorylation of eukaryotic initiation factor alpha (eIF2α) results in the accumulation of activating transcription factor 4 (ATF4) and GRP78 independent of unfolded protein response (UPR). Suppression of GRP78 expression decreases the proliferation and invasion of human CCA cells. It's notable that mTOR is required for eIF2α phosphorylation-induced ATF4 and GRP78 expression. Importantly, JNK promotes eIF2α/ATF4-mediated GRP78 induction through regulating the activity of mTOR. Thus, our study implicates JNK/mTOR signaling plays an important role in cholangiocarcinogenesis, partially through promoting the eIF2α/ATF4/GRP78 pathway.
Collapse
Affiliation(s)
- Chunhong Feng
- Department of Hepatobiliary Surgery of the Affiliated Hospital, Luzhou Medical College, Luzhou, Sichuan, China
| | - Kai He
- Department of Hepatobiliary Surgery of the Affiliated Hospital, Luzhou Medical College, Luzhou, Sichuan, China
| | - Chunyan Zhang
- Department of Biochemistry and Molecular Biology, Luzhou Medical College, Luzhou, Sichuan, China
| | - Song Su
- Department of Hepatobiliary Surgery of the Affiliated Hospital, Luzhou Medical College, Luzhou, Sichuan, China
| | - Bo Li
- Department of Hepatobiliary Surgery of the Affiliated Hospital, Luzhou Medical College, Luzhou, Sichuan, China
| | - Yuxiao Li
- Department of Biochemistry and Molecular Biology, Luzhou Medical College, Luzhou, Sichuan, China
| | - Chun-Yan Duan
- Department of Biochemistry and Molecular Biology, Luzhou Medical College, Luzhou, Sichuan, China
| | - Shaokun Chen
- Department of Biochemistry and Molecular Biology, Luzhou Medical College, Luzhou, Sichuan, China
| | - Run Chen
- Department of Public Health, Luzhou Medical College, Luzhou, Sichuan, China
| | - Youping Liu
- Department of Biochemistry and Molecular Biology, Luzhou Medical College, Luzhou, Sichuan, China
| | - Hong Li
- Department of Biochemistry and Molecular Biology, Luzhou Medical College, Luzhou, Sichuan, China
| | - Mei Wei
- Affiliated Hospital of Chinese Traditional Medicine, Luzhou Medical College, Luzhou, Sichuan, China
| | - Xianming Xia
- Department of Hepatobiliary Surgery of the Affiliated Hospital, Luzhou Medical College, Luzhou, Sichuan, China
| | - Rongyang Dai
- Department of Hepatobiliary Surgery of the Affiliated Hospital, Luzhou Medical College, Luzhou, Sichuan, China
- Department of Biochemistry and Molecular Biology, Luzhou Medical College, Luzhou, Sichuan, China
| |
Collapse
|
25
|
Misra UK, Pizzo SV. Activated α2-macroglobulin binding to cell surface GRP78 induces T-loop phosphorylation of Akt1 by PDK1 in association with Raptor. PLoS One 2014; 9:e88373. [PMID: 24516643 PMCID: PMC3916429 DOI: 10.1371/journal.pone.0088373] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 01/13/2014] [Indexed: 12/14/2022] Open
Abstract
PDK1 phosphorylates multiple substrates including Akt by PIP3-dependent mechanisms. In this report we provide evidence that in prostate cancer cells stimulated with activated α2-macroglobulin (α2M*) PDK1 phosphorylates Akt in the T-loop at Thr(308) by using Raptor in the mTORC1 complex as a scaffold protein. First we demonstrate that PDK1, Raptor, and mTOR co-immunoprecipitate. Silencing the expression, not only of PDK1, but also Raptor by RNAi nearly abolished Akt phosphorylation at Akt(Thr308) in Raptor-immunoprecipitates of α2M*-stimulated prostate cancer cells. Immunodepleting Raptor or PDK from cell lysates of cells treated with α2M* drastically reduced Akt phosphorylation at Thr(308), which was recovered by adding the supernatant of Raptor- or PDK1-depleted cell lysates, respectively. Studies of insulin binding to its receptor on prostate cancer cells yielded similar results. We thus demonstrate that phosphorylating the T-loop Akt residue Thr(308) by PDK1 requires Raptor of the mTORC1 complex as a platform or scaffold protein.
Collapse
Affiliation(s)
- Uma Kant Misra
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Salvatore Vincent Pizzo
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|
26
|
Liu R, Li X, Gao W, Zhou Y, Wey S, Mitra SK, Krasnoperov V, Dong D, Liu S, Li D, Zhu G, Louie S, Conti PS, Li Z, Lee AS, Gill PS. Monoclonal antibody against cell surface GRP78 as a novel agent in suppressing PI3K/AKT signaling, tumor growth, and metastasis. Clin Cancer Res 2013; 19:6802-11. [PMID: 24048331 DOI: 10.1158/1078-0432.ccr-13-1106] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
PURPOSE The ER chaperone GRP78 translocates to the surface of tumor cells and promotes survival, metastasis, and resistance to therapy. An oncogenic function of cell surface GRP78 has been attributed to the activation of the phosphoinositide 3-kinase (PI3K) pathway. We intend to use a novel anti-GRP78 monoclonal antibody (MAb159) to attenuate PI3K signaling and inhibit tumor growth and metastasis. EXPERIMENTAL DESIGN MAb159 was characterized biochemically. Antitumor activity was tested in cancer cell culture, tumor xenograft models, tumor metastasis models, and spontaneous tumor models. Cancer cells and tumor tissues were analyzed for PI3K activity. MAb159 was humanized and validated for diagnostic and therapeutic application. RESULTS MAb159 specifically recognized surface GRP78, triggered GRP78 endocytosis, and localized to tumors but not to normal organs in vivo. MAb159 inhibited tumor cell proliferation and enhanced tumor cell death both in vitro and in vivo. In MAb159-treated tumors, PI3K signaling was inhibited without compensatory MAPK pathway activation. Furthermore, MAb159 halted or reversed tumor progression in the spontaneous PTEN-loss-driven prostate and leukemia tumor models, and inhibited tumor growth and metastasis in xenograft models. Humanized MAb159, which retains high affinity, tumor specific localization, and the antitumor activity, was nontoxic in mice, and had desirable pharmacokinetics. CONCLUSIONS GRP78-specific antibody MAb159 modulates the PI3K pathway and inhibits tumor growth and metastasis. Humanized MAb159 will enter human trials shortly.
Collapse
Affiliation(s)
- Ren Liu
- Authors' Affiliations: Departments of Pathology, Biochemistry and Molecular Biology, and Radiology; School of Pharmacy, University of Southern California Keck School of Medicine, USC Norris Comprehensive Cancer Center; and VasGene Therapeutics Inc., Los Angeles, California
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Misra UK, Payne S, Pizzo SV. The Monomeric Receptor Binding Domain of Tetrameric α2-Macroglobulin Binds to Cell Surface GRP78 Triggering Equivalent Activation of Signaling Cascades. Biochemistry 2013; 52:4014-25. [DOI: 10.1021/bi400376s] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Uma Kant Misra
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710, United
States
| | - Sturgis Payne
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710, United
States
| | - Salvatore Vincent Pizzo
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710, United
States
| |
Collapse
|
28
|
Evidence for a pro-proliferative feedback loop in prostate cancer: the role of Epac1 and COX-2-dependent pathways. PLoS One 2013; 8:e63150. [PMID: 23646189 PMCID: PMC3640024 DOI: 10.1371/journal.pone.0063150] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 03/29/2013] [Indexed: 12/15/2022] Open
Abstract
Objective In human prostate cancer cells, a selective Epac agonist, 8-CPT-2Me-cAMP, upregulates cell proliferation and survival via activation of Ras-MAPK and PI- 3-kinase-Akt-mTOR signaling cascades. Here we examine the role of inflammatory mediators in Epac1-induced cellular proliferation by determining the expression of the pro-inflammatory markers p-cPLA2, COX-2, and PGE2 in prostate cancer cells treated with 8-CPT-2Me-cAMP. Methods We employed inhibitors of COX-2, mTORC1, and mTORC2 to probe cyclic AMP-dependent pathways in human prostate cancer cells. RNAi targeting Epac1, Raptor, and Rictor was also employed in these studies. Results 8-CPT-2Me-cAMP treatment caused a 2–2.5-fold increase of p-cPLA2S505, COX-2, and PGE2 levels in human prostate cancer cell lines. Pretreatment of cells with the COX-2 inhibitor SC-58125 or the EP4 antagonist AH-23848, or with an inhibitor of mTORC1 and mTORC2, Torin1, significantly reduced the Epac1-dependent increase of p-cPLA2 and COX-2, p-S6-kinaseT389, and p-AKTS473. In addition, Epac1-induced protein and DNA synthesis were greatly reduced upon pretreatment of cells with either COX-2, EP4, or mTOR inhibitors. Transfection of prostate cancer cells with Epac1 dsRNA, Raptor dsRNA, or Rictor dsRNA profoundly reduced Epac1-dependent increases in p-cPLA2 and COX-2. Conclusion We show that Epac1, a downstream effector of cAMP, functions as a pro-inflammatory modulator in prostate cancer cells and promotes cell proliferation and survival by upregulating Ras-MAPK, and PI 3-kinase-Akt-mTOR signaling.
Collapse
|