1
|
Rashwan ME, Ahmed MR, Elfiky AA. In silico prediction of GRP78-CRIPTO binding sites to improve therapeutic targeting in glioblastoma. Sci Rep 2025; 15:16660. [PMID: 40360533 PMCID: PMC12075867 DOI: 10.1038/s41598-025-00125-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most malignant tumors in central nervous system (CNS) tumors. The glucose-regulated protein 78 (GRP78) and CRIPTO (Cripto-1), a protein that belongs to the EGF-CFC (epidermal growth factor cripto-1 FRL-1 cryptic) family, are overexpressed in GBM. A complex between GRP78 SBDβ (substrate binding domain beta) and CRIPTO CFC domain was reported in previous studies. This complex activates MAPK/AKT signaling, Src/PI3K/AKT, and Smad2/3 pathways which is a reason for tumor proliferation. In this work, we study how the two proteins form the complex figuring out binding sites between GRP78 and CRIPTO utilizing computational biophysics and bioinformatics tools, such as protein-protein docking, molecular dynamics simulation and MMGBSA calculations. Haddock web server results of 4 regions from the CFC domain (region1 (- 70.4), region2 (- 78.7), region3 (- 74.2), region4 (- 86.8)) with selected residues of the SBDβ are then simulated for 100 ns MDS then MMGBSA were calculated for the four complexes. The results reveal the stability of the complexes with binding free energy (complex1 (- 15.07 kcal/mol), complex2 (- 59.78 kcal/mol), complex3 (- 81.92 kcal/mol), complex4 (- 126.26 kcal/mol). All these findings ensure that GRP78 SBDβ associates with the CRIPTO CFC domain, and the binding sites suggested make stable interactions between the proteins.
Collapse
Affiliation(s)
- Mahmoud E Rashwan
- Physics Department, Faculty of Science, Sohag University, Sohag, 82524, Egypt.
| | - Mahrous R Ahmed
- Physics Department, Faculty of Science, Sohag University, Sohag, 82524, Egypt
| | - Abdo A Elfiky
- Biophysics Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| |
Collapse
|
2
|
Yang Y, Li W, Zhao Y, Sun M, Xing F, Yang J, Zhou Y. GRP78 in Glioma Progression and Therapy: Implications for Targeted Approaches. Biomedicines 2025; 13:382. [PMID: 40002794 PMCID: PMC11852679 DOI: 10.3390/biomedicines13020382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/24/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Glioma is the most common primary malignant brain tumor, accounting for the majority of brain cancer-related deaths. Considering the limited efficacy of conventional therapies, novel molecular targeted therapies have been developed to improve outcomes and minimize toxicity. Glucose-regulated protein 78 (GRP78), a molecular chaperone primarily localized in the endoplasmic reticulum (ER), has received increasing attention for its role in glioma progression and resistance to conventional therapies. Overexpressed in gliomas, GRP78 supports tumor growth, survival, and therapeutic resistance by maintaining cellular homeostasis and regulating multiple signaling pathways. Its aberrant expression correlates with higher tumor grades and poorer patient prognosis. Beyond its intracellular functions, GRP78's presence on the cell surface and its role in the tumor microenvironment underscore its potential as a therapeutic target. Recent studies have explored innovative strategies to target GRP78, including small molecule inhibitors, monoclonal antibodies, and chimeric antigen receptor (CAR) T cell therapy, showing significant potential in glioma treatment. This review explores the biological characteristics of GRP78, its role in glioma pathophysiology, and the potential of GRP78-targeted therapy as a novel strategy to overcome treatment resistance and improve clinical outcomes. GRP78-targeted therapy, either alone or in combination with conventional treatments, could be a novel and attractive strategy for future glioma treatment.
Collapse
Affiliation(s)
- Yue Yang
- Department of Chemistry, College of Sciences, Shanghai University, Shanghai 200444, China
| | - Wen Li
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (W.L.); (Y.Z.)
- Department of Biomaterials and Stem Cells, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Yu Zhao
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (W.L.); (Y.Z.)
- Department of Biomaterials and Stem Cells, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Minxuan Sun
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (W.L.); (Y.Z.)
- Department of Biomaterials and Stem Cells, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Feifei Xing
- Department of Chemistry, College of Sciences, Shanghai University, Shanghai 200444, China
| | - Jiao Yang
- Suzhou Research Center of Medical School, Institute of Clinical Medicine Research, Suzhou Hospital, The Affiliated Hospital of Medical School, Nanjing University, Lijiang Road No. 1, Suzhou 215153, China
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, Suzhou 215009, China
| | - Yuanshuai Zhou
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (W.L.); (Y.Z.)
- Department of Biomaterials and Stem Cells, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| |
Collapse
|
3
|
Karamali N, Daraei A, Rostamlou A, Mahdavi R, Akbari Jonoush Z, Ghadiri N, Mahmoudi Z, Mardi A, Javidan M, Sohrabi S, Baradaran B. Decoding contextual crosstalk: revealing distinct interactions between non-coding RNAs and unfolded protein response in breast cancer. Cancer Cell Int 2024; 24:104. [PMID: 38468244 PMCID: PMC10926595 DOI: 10.1186/s12935-024-03296-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 03/06/2024] [Indexed: 03/13/2024] Open
Abstract
Breast cancer is significantly influenced by endoplasmic reticulum (ER) stress, impacting both its initiation and progression. When cells experience an accumulation of misfolded or unfolded proteins, they activate the unfolded protein response (UPR) to restore cellular balance. In breast cancer, the UPR is frequently triggered due to challenging conditions within tumors. The UPR has a dual impact on breast cancer. On one hand, it can contribute to tumor growth by enhancing cell survival and resistance to programmed cell death in unfavorable environments. On the other hand, prolonged and severe ER stress can trigger cell death mechanisms, limiting tumor progression. Furthermore, ER stress has been linked to the regulation of non-coding RNAs (ncRNAs) in breast cancer cells. These ncRNAs, including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), play essential roles in cancer development by influencing gene expression and cellular processes. An improved understanding of how ER stress and ncRNAs interact in breast cancer can potentially lead to new treatment approaches. Modifying specific ncRNAs involved in the ER stress response might interfere with cancer cell survival and induce cell death. Additionally, focusing on UPR-associated proteins that interact with ncRNAs could offer novel therapeutic possibilities. Therefore, this review provides a concise overview of the interconnection between ER stress and ncRNAs in breast cancer, elucidating the nuanced effects of the UPR on cell fate and emphasizing the regulatory roles of ncRNAs in breast cancer progression.
Collapse
Affiliation(s)
- Negin Karamali
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arshia Daraei
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Arman Rostamlou
- Department of Medical Biology, School of Medicine, University of EGE, Bornova, Izmir, Turkey
| | - Roya Mahdavi
- Student Research Committee, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Akbari Jonoush
- Student Research Committee, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nooshin Ghadiri
- Student Research Committee, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Mahmoudi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amirhossein Mardi
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Moslem Javidan
- Student Research Committee, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sepideh Sohrabi
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Kusaczuk M, Ambel ET, Naumowicz M, Velasco G. Cellular stress responses as modulators of drug cytotoxicity in pharmacotherapy of glioblastoma. Biochim Biophys Acta Rev Cancer 2024; 1879:189054. [PMID: 38103622 DOI: 10.1016/j.bbcan.2023.189054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 11/21/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
Despite the extensive efforts to find effective therapeutic strategies, glioblastoma (GBM) remains a therapeutic challenge with dismal prognosis of survival. Over the last decade the role of stress responses in GBM therapy has gained a great deal of attention, since depending on the duration and intensity of these cellular programs they can be cytoprotective or promote cancer cell death. As such, initiation of the UPR, autophagy or oxidative stress may either impede or facilitate drug-mediated cell killing. In this review, we summarize the mechanisms that regulate ER stress, autophagy, and oxidative stress during GBM development and progression to later discuss the involvement of these stress pathways in the response to different treatments. We also discuss how a precise understanding of the molecular mechanisms regulating stress responses evoked by different pharmacological agents could decisively contribute to the design of novel and more effective combinational treatments against brain malignancies.
Collapse
Affiliation(s)
- Magdalena Kusaczuk
- Department of Pharmaceutical Biochemistry, Medical University of Bialystok, Mickiewicza 2A, 15-222 Bialystok, Poland.
| | - Elena Tovar Ambel
- Department of Biochemistry and Molecular Biology, School of Biology, Complutense University, Instituto de Investigación Sanitaria San Carlos IdISSC, 28040 Madrid, Spain
| | - Monika Naumowicz
- Department of Physical Chemistry, Faculty of Chemistry, University of Bialystok, K. Ciolkowskiego 1K, 15-245 Bialystok, Poland
| | - Guillermo Velasco
- Department of Biochemistry and Molecular Biology, School of Biology, Complutense University, Instituto de Investigación Sanitaria San Carlos IdISSC, 28040 Madrid, Spain.
| |
Collapse
|
5
|
Akinyemi AO, Simpson KE, Oyelere SF, Nur M, Ngule CM, Owoyemi BCD, Ayarick VA, Oyelami FF, Obaleye O, Esoe DP, Liu X, Li Z. Unveiling the dark side of glucose-regulated protein 78 (GRP78) in cancers and other human pathology: a systematic review. Mol Med 2023; 29:112. [PMID: 37605113 PMCID: PMC10464436 DOI: 10.1186/s10020-023-00706-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/26/2023] [Indexed: 08/23/2023] Open
Abstract
Glucose-Regulated Protein 78 (GRP78) is a chaperone protein that is predominantly expressed in the lumen of the endoplasmic reticulum. GRP78 plays a crucial role in protein folding by assisting in the assembly of misfolded proteins. Under cellular stress conditions, GRP78 can translocate to the cell surface (csGRP78) were it interacts with different ligands to initiate various intracellular pathways. The expression of csGRP78 has been associated with tumor initiation and progression of multiple cancer types. This review provides a comprehensive analysis of the existing evidence on the roles of GRP78 in various types of cancer and other human pathology. Additionally, the review discusses the current understanding of the mechanisms underlying GRP78's involvement in tumorigenesis and cancer advancement. Furthermore, we highlight recent innovative approaches employed in downregulating GRP78 expression in cancers as a potential therapeutic target.
Collapse
Affiliation(s)
| | | | | | - Maria Nur
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, USA
| | | | | | | | - Felix Femi Oyelami
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, USA
| | | | - Dave-Preston Esoe
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, USA
| | - Xiaoqi Liu
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, USA
- Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, USA
| | - Zhiguo Li
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, USA.
| |
Collapse
|
6
|
Wang S, Wei W, Yuan Y, Sun B, Yang D, Liu N, Zhao X. Chimeric antigen receptor T cells targeting cell surface GRP78 efficiently kill glioblastoma and cancer stem cells. J Transl Med 2023; 21:493. [PMID: 37481592 PMCID: PMC10362566 DOI: 10.1186/s12967-023-04330-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/07/2023] [Indexed: 07/24/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) is recognized as among the most aggressive forms of brain tumor. Patients typically present with a five-year survival rate of less than 6% with traditional surgery and chemoradiotherapy, which calls for novel immunotherapies like chimeric antigen receptor T (CAR-T) cells therapy. In response to endoplasmic reticulum (ER) stress in multiple tumor cells including GBM, the glucose-regulated protein 78 (GRP78) expression increases and the protein is partially translocated to the cell surface, while it is restricted to the cytoplasm and the nucleus in normal cells. METHODS In this study, to target the cell surface GRP78 (csGRP78), CAR-T cells based on its binding peptide were generated. In vitro two GBM cell lines and glioma stem cells (GSCs) were used to confirm the localization of csGRP78 and the cytotoxicity of the CAR-T cells. In vivo a GBM xenograft model was used to assess the killing activity and the safety of the CAR-T cells. RESULTS We confirmed the localization of csGRP78 at the cell surface of two GBM cell lines (U-251MG and U-87MG) and in GSCs. Co-culture experiments revealed that the CAR-T cells could specifically kill the GBM tumor cells and GSCs with specific IFN-γ release. Furthermore, in the tumor xenograft model, the CAR-T cells could decrease the number of GSCs and significantly suppress tumor cell growth. Importantly, we found no obvious off-target effects or T cell infiltration in major organs following systemic administration of these cells. CONCLUSIONS The csGRP78 targeted CAR-T cells efficiently kill GBM tumor cells and GSCs both in vitro and in vivo, and ultimately suppress the xenograft tumors growth without obvious tissue injuries. Therefore, our study demonstrates that csGRP78 represents a valuable target and the csGRP78-targeted CAR-T cells strategy is an effective immunotherapy against GBM.
Collapse
Affiliation(s)
- Shijie Wang
- Department of Targeting Therapy and Immunology and Laboratory of Animal Tumor Models, Cancer Center and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenwen Wei
- Department of Targeting Therapy and Immunology and Laboratory of Animal Tumor Models, Cancer Center and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuncang Yuan
- Department of Targeting Therapy and Immunology and Laboratory of Animal Tumor Models, Cancer Center and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bin Sun
- Department of Targeting Therapy and Immunology and Laboratory of Animal Tumor Models, Cancer Center and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dong Yang
- Department of Targeting Therapy and Immunology and Laboratory of Animal Tumor Models, Cancer Center and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Nan Liu
- Department of Targeting Therapy and Immunology and Laboratory of Animal Tumor Models, Cancer Center and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xudong Zhao
- Department of Targeting Therapy and Immunology and Laboratory of Animal Tumor Models, Cancer Center and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
7
|
Gu Z, Wang L, Dong Q, Xu K, Ye J, Shao X, Yang S, Lu C, Chang C, Hou Y, Zhai Y, Wang X, He F, Sun A. Aberrant LYZ expression in tumor cells serves as the potential biomarker and target for HCC and promotes tumor progression via csGRP78. Proc Natl Acad Sci U S A 2023; 120:e2215744120. [PMID: 37428911 PMCID: PMC10629575 DOI: 10.1073/pnas.2215744120] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 05/02/2023] [Indexed: 07/12/2023] Open
Abstract
Hepatocellular carcinoma (HCC) takes the predominant malignancy of hepatocytes with bleak outcomes owing to high heterogeneity among patients. Personalized treatments based on molecular profiles will better improve patients' prognosis. Lysozyme (LYZ), a secretory protein with antibacterial function generally expressed in monocytes/macrophages, has been observed for the prognostic implications in different types of tumors. However, studies about the explicit applicative scenarios and mechanisms for tumor progression are still quite limited, especially for HCC. Here, based on the proteomic molecular classification data of early-stage HCC, we revealed that the LYZ level was elevated significantly in the most malignant HCC subtype and could serve as an independent prognostic predictor for HCC patients. Molecular profiles of LYZ-high HCCs were typical of those for the most malignant HCC subtype, with impaired metabolism, along with promoted proliferation and metastasis characteristics. Further studies demonstrated that LYZ tended to be aberrantly expressed in poorly differentiated HCC cells, which was regulated by STAT3 activation. LYZ promoted HCC proliferation and migration in both autocrine and paracrine manners independent of the muramidase activity through the activation of downstream protumoral signaling pathways via cell surface GRP78. Subcutaneous and orthotopic xenograft tumor models indicated that targeting LYZ inhibited HCC growth markedly in NOD/SCID mice. These results propose LYZ as a prognostic biomarker and therapeutic target for the subclass of HCC with an aggressive phenotype.
Collapse
Affiliation(s)
- Zhiwen Gu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing102206, China
- Research Unit of Proteomics-driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing102206, China
| | - Lei Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing102206, China
- Faculty of Environment and Life, Beijing University of Technology, Beijing100124, China
| | - Qian Dong
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing102206, China
| | - Kaikun Xu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing102206, China
| | - Jingnan Ye
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing102206, China
| | - Xianfeng Shao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing102206, China
| | - Songpeng Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing102206, China
| | - Cuixiu Lu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing102206, China
| | - Cheng Chang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing102206, China
- Research Unit of Proteomics-driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing102206, China
| | - Yushan Hou
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing102206, China
| | - Yuanjun Zhai
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing102206, China
- Research Unit of Proteomics-driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing102206, China
| | - Xinxin Wang
- Department of Pathology, Beijing You’an Hospital, Capital Medical University, Beijing100069, China
| | - Fuchu He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing102206, China
- Research Unit of Proteomics-driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing102206, China
| | - Aihua Sun
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing102206, China
- Research Unit of Proteomics-driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing102206, China
| |
Collapse
|
8
|
Dos Santos NS, Gonçalves DR, Balbinot B, Visioli F. Is GRP78 (Glucose-regulated protein 78) a prognostic biomarker in differents types of cancer? A systematic review and meta-analysis. Pathol Res Pract 2023; 242:154301. [PMID: 36610326 DOI: 10.1016/j.prp.2023.154301] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/31/2022] [Accepted: 01/02/2023] [Indexed: 01/05/2023]
Abstract
GRP78 is a chaperone with anti-apoptotic function associated with aggressive tumors. This systematic review aimed to evaluate GRP78 expression in cancer and its relation to prognosis outcomes. This review was conducted in different databases searching for human cancer studies assessing GRP78 immunohistochemical levels on tissue samples. A total of 98 manuscripts were included. In 62% of the studies, GRP78 was associated with a worse prognosis. A meta-analysis included 29 studies that detected a significantly higher expression of GRP78 in cancer tissues (RR= 2.35, 95% CI 1.75-3.15) compared to control. A meta-analysis of 3 and 5-years Overall Survival revealed an increased risk of death for tumors with high expression of GRP78 (RR=1.36, 95%CI 1.16-1,59, I2 = 57%) and (RR=1.65, 95%CI 1.22-2.21, I2 =64%), respectively. GRP78 is an important prognostic biomarker for different types of cancer and a promising therapeutic target.
Collapse
Affiliation(s)
- Natália Souza Dos Santos
- Oral Pathology Department, School of Dentistry, Universidade Federal do Rio Grande do Sul, Brazil
| | - Douglas Rodrigues Gonçalves
- Oral Pathology Department, School of Dentistry, Universidade Federal do Rio Grande do Sul, Brazil; Oral Medicine Unit, Otorhinolaryngology Service, Hospital de Clínicas de Porto Alegre, Brazil
| | - Bianca Balbinot
- Oral Pathology Department, School of Dentistry, Universidade Federal do Rio Grande do Sul, Brazil
| | - Fernanda Visioli
- Oral Pathology Department, School of Dentistry, Universidade Federal do Rio Grande do Sul, Brazil; Experimental Center Research, Hospital de Clínicas de Porto Alegre, Brazil.
| |
Collapse
|
9
|
Chern YJ, Tai IT. Adaptive response of resistant cancer cells to chemotherapy. Cancer Biol Med 2020; 17:842-863. [PMID: 33299639 PMCID: PMC7721100 DOI: 10.20892/j.issn.2095-3941.2020.0005] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
Despite advances in cancer therapeutics and the integration of personalized medicine, the development of chemoresistance in many patients remains a significant contributing factor to cancer mortality. Upon treatment with chemotherapeutics, the disruption of homeostasis in cancer cells triggers the adaptive response which has emerged as a key resistance mechanism. In this review, we summarize the mechanistic studies investigating the three major components of the adaptive response, autophagy, endoplasmic reticulum (ER) stress signaling, and senescence, in response to cancer chemotherapy. We will discuss the development of potential cancer therapeutic strategies in the context of these adaptive resistance mechanisms, with the goal of stimulating research that may facilitate the development of effective cancer therapy.
Collapse
Affiliation(s)
- Yi-Jye Chern
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, British Columbia V5Z1L3, Canada.,Michael Smith Genome Sciences Center, British Columbia Cancer Agency, Vancouver, British Columbia V5Z1L3, Canada
| | - Isabella T Tai
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, British Columbia V5Z1L3, Canada.,Michael Smith Genome Sciences Center, British Columbia Cancer Agency, Vancouver, British Columbia V5Z1L3, Canada
| |
Collapse
|
10
|
Liu K, Tsung K, Attenello FJ. Characterizing Cell Stress and GRP78 in Glioma to Enhance Tumor Treatment. Front Oncol 2020; 10:608911. [PMID: 33363039 PMCID: PMC7759649 DOI: 10.3389/fonc.2020.608911] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is the most common primary brain tumor, carrying a very poor prognosis, with median overall survival at about 12 to 15 months despite surgical resection, chemotherapy with temozolomide (TMZ), and radiation therapy. GBM recurs in the vast majority of patients, with recurrent tumors commonly displaying increase in resistance to standard of care chemotherapy, TMZ, as well as radiotherapy. One of the most commonly cited mechanisms of chemotherapeutic and radio-resistance occurs via the glucose-regulated protein 78 (GRP78), a well-studied mediator of the unfolded protein response (UPR), that has also demonstrated potential as a biomarker in GBM. Overexpression of GRP78 has been directly correlated with malignant tumor characteristics, including higher tumor grade, cellular proliferation, migration, invasion, poorer responses to TMZ and radiation therapy, and poorer patient outcomes. GRP78 expression is also higher in GBM tumor cells upon recurrence. Meanwhile, knockdown or suppression of GRP78 has been shown to sensitize cells to TMZ and radiation therapy. In light of these findings, various novel developing therapies are targeting GRP78 as monotherapies, combination therapies that enhance the effects of TMZ and radiation therapy, and as treatment delivery modalities. In this review, we delineate the mechanisms by which GRP78 has been noted to specifically modulate glioblastoma behavior and discuss current developing therapies involving GRP78 in GBM. While further research is necessary to translate these developing therapies into clinical settings, GRP78-based therapies hold promise in improving current standard-of-care GBM therapy and may ultimately lead to improved patient outcomes.
Collapse
Affiliation(s)
- Kristie Liu
- Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| | - Kathleen Tsung
- Department of Neurosurgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| | - Frank J Attenello
- Department of Neurosurgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
11
|
Cell surface GRP78 promotes stemness in normal and neoplastic cells. Sci Rep 2020; 10:3474. [PMID: 32103065 PMCID: PMC7044190 DOI: 10.1038/s41598-020-60269-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 01/31/2020] [Indexed: 01/07/2023] Open
Abstract
Reliable approaches to identify stem cell mechanisms that mediate aggressive cancer could have great therapeutic value, based on the growing evidence of embryonic signatures in metastatic cancers. However, how to best identify and target stem-like mechanisms aberrantly acquired by cancer cells has been challenging. We harnessed the power of reprogramming to examine GRP78, a chaperone protein generally restricted to the endoplasmic reticulum in normal tissues, but which is expressed on the cell surface of human embryonic stem cells and many cancer types. We have discovered that (1) cell surface GRP78 (sGRP78) is expressed on iPSCs and is important in reprogramming, (2) sGRP78 promotes cellular functions in both pluripotent and breast cancer cells (3) overexpression of GRP78 in breast cancer cells leads to an induction of a CD24−/CD44+ tumor initiating cell (TIC) population (4) sGRP78+ breast cancer cells are enriched for stemness genes and appear to be a subset of TICs (5) sGRP78+ breast cancer cells show an enhanced ability to seed metastatic organ sites in vivo. These collective findings show that GRP78 has important functions in regulating both pluripotency and oncogenesis, and suggest that sGRP78 marks a stem-like population in breast cancer cells that has increased metastatic potential in vivo.
Collapse
|
12
|
Dadey DYA, Kapoor V, Khudanyan A, Thotala D, Hallahan DE. PERK Regulates Glioblastoma Sensitivity to ER Stress Although Promoting Radiation Resistance. Mol Cancer Res 2018; 16:1447-1453. [PMID: 29991528 DOI: 10.1158/1541-7786.mcr-18-0224] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/16/2018] [Accepted: 06/20/2018] [Indexed: 11/16/2022]
Abstract
The aggressive nature and inherent therapeutic resistance of glioblastoma multiforme (GBM) has rendered the median survival of afflicted patients to 14 months. Therefore, it is imperative to understand the molecular biology of GBM to provide new treatment options to overcome this disease. It has been demonstrated that the protein kinase R-like endoplasmic reticulum kinase (PERK) pathway is an important regulator of the endoplasmic reticulum (ER) stress response. PERK signaling has been observed in other model systems after radiation; however, less is known in the context of GBM, which is frequently treated with radiation-based therapies. To investigate the significance of PERK, we studied activation of the PERK-eIF2α-ATF4 pathway in GBM after ionizing radiation (IR). By inhibiting PERK, it was determined that ionizing radiation (IR)-induced PERK activity led to eIF2α phosphorylation. IR enhanced the prodeath component of PERK signaling in cells treated with Sal003, an inhibitor of phospho-eIF2α phosphatase. Mechanistically, ATF4 mediated the prosurvival activity during the radiation response. The data support the notion that induction of ER stress signaling by radiation contributes to adaptive survival mechanisms during radiotherapy. The data also support a potential role for the PERK/eIF2α/ATF4 axis in modulating cell viability in irradiated GBM.Implications: The dual function of PERK as a mediator of survival and death may be exploited to enhance the efficacy of radiation therapy.Visual Overview: http://mcr.aacrjournals.org/content/16/10/1447/F1.large.jpg Mol Cancer Res; 16(10); 1447-53. ©2018 AACR.
Collapse
Affiliation(s)
- David Y A Dadey
- Department of Radiation Oncology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri.,Medical Scientist Training Program, School of Medicine, Washington University in St. Louis, St. Louis, Missouri.,Department of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Vaishali Kapoor
- Department of Radiation Oncology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Arpine Khudanyan
- Department of Radiation Oncology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri.,Oregon Health and Science University, School of Medicine, Portland, Oregon
| | - Dinesh Thotala
- Department of Radiation Oncology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri.,Siteman Cancer Center, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Dennis E Hallahan
- Department of Radiation Oncology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri. .,Siteman Cancer Center, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| |
Collapse
|
13
|
Jin X, Kim DK, Riew TR, Kim HL, Lee MY. Cellular and Subcellular Localization of Endoplasmic Reticulum Chaperone GRP78 Following Transient Focal Cerebral Ischemia in Rats. Neurochem Res 2018; 43:1348-1362. [PMID: 29774449 DOI: 10.1007/s11064-018-2550-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/13/2018] [Accepted: 05/13/2018] [Indexed: 12/20/2022]
Abstract
The 78-kDa glucose-regulated protein (GRP78), a chaperone protein located in the endoplasmic reticulum (ER), has been reported to have neuroprotective effects in the injured central nervous system. Our aim was to examine the expression profiles and subcellular distributions of GRP78 and its association with the neuroglial reaction in the rat striatum after transient, focal cerebral ischemia. In sham-operated rats, constitutive, specific immunoreactivity for GRP78 was almost exclusively localized to the rough ER of striatal neurons, with none in the resting, ramified microglia or astrocytes. At 1 day post reperfusion, increased expression was observed in ischemia-resistant cholinergic interneurons, when most striatal neurons had lost GRP78 expression (this occurred earlier than the loss of other neuronal markers). By 3 days post reperfusion, GRP78 expression had re-emerged in association with the activation of glial cells in both infarct and peri-infarct areas but showed different patterns in the two regions. Most of the expression induced in the infarct area could be attributed to brain macrophages, while expression in the peri-infarct area predominantly occurred in neurons and reactive astrocytes. A gradual, sustained induction of GRP78 immunoreactivity occurred in reactive astrocytes localized to the astroglial scar, lasting for at least 28 days post reperfusion. Using correlative light- and electron-microscopy, we found conspicuous GRP78 protein localized to abnormally prominent, dilated rough ER in both glial cell types. Thus, our data indicate a link between GRP78 expression and the activated functional status of neuroglial cells, predominantly microglia/macrophages and astrocytes, occurring in response to ischemia-induced ER stress.
Collapse
Affiliation(s)
- Xuyan Jin
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06501, Republic of Korea
| | - Dong Kyu Kim
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06501, Republic of Korea
| | - Tae-Ryong Riew
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06501, Republic of Korea
| | - Hong Lim Kim
- Integrative Research Support Center, Laboratory of Electron Microscopy, College of Medicine, The Catholic University of Korea, Seoul, 06501, Republic of Korea
| | - Mun-Yong Lee
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06501, Republic of Korea.
| |
Collapse
|
14
|
Cornec-Le Gall E, Olson RJ, Besse W, Heyer CM, Gainullin VG, Smith JM, Audrézet MP, Hopp K, Porath B, Shi B, Baheti S, Senum SR, Arroyo J, Madsen CD, Férec C, Joly D, Jouret F, Fikri-Benbrahim O, Charasse C, Coulibaly JM, Yu AS, Khalili K, Pei Y, Somlo S, Le Meur Y, Torres VE, Harris PC. Monoallelic Mutations to DNAJB11 Cause Atypical Autosomal-Dominant Polycystic Kidney Disease. Am J Hum Genet 2018; 102:832-844. [PMID: 29706351 PMCID: PMC5986722 DOI: 10.1016/j.ajhg.2018.03.013] [Citation(s) in RCA: 212] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/08/2018] [Indexed: 01/05/2023] Open
Abstract
Autosomal-dominant polycystic kidney disease (ADPKD) is characterized by the progressive development of kidney cysts, often resulting in end-stage renal disease (ESRD). This disorder is genetically heterogeneous with ∼7% of families genetically unresolved. We performed whole-exome sequencing (WES) in two multiplex ADPKD-like pedigrees, and we analyzed a further 591 genetically unresolved, phenotypically similar families by targeted next-generation sequencing of 65 candidate genes. WES identified a DNAJB11 missense variant (p.Pro54Arg) in two family members presenting with non-enlarged polycystic kidneys and a frameshifting change (c.166_167insTT) in a second family with small renal and liver cysts. DNAJB11 is a co-factor of BiP, a key chaperone in the endoplasmic reticulum controlling folding, trafficking, and degradation of secreted and membrane proteins. Five additional multigenerational families carrying DNAJB11 mutations were identified by the targeted analysis. The clinical phenotype was consistent in the 23 affected members, with non-enlarged cystic kidneys that often evolved to kidney atrophy; 7 subjects reached ESRD from 59 to 89 years. The lack of kidney enlargement, histologically evident interstitial fibrosis in non-cystic parenchyma, and recurring episodes of gout (one family) suggested partial phenotypic overlap with autosomal-dominant tubulointerstitial diseases (ADTKD). Characterization of DNAJB11-null cells and kidney samples from affected individuals revealed a pathogenesis associated with maturation and trafficking defects involving the ADPKD protein, PC1, and ADTKD proteins, such as UMOD. DNAJB11-associated disease is a phenotypic hybrid of ADPKD and ADTKD, characterized by normal-sized cystic kidneys and progressive interstitial fibrosis resulting in late-onset ESRD.
Collapse
Affiliation(s)
- Emilie Cornec-Le Gall
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA; Department of Nephrology, University Hospital, European University of Brittany, Brest, Brittany 29200, France; Department of Molecular Genetics, National Institute of Health and Medical Sciences, INSERM U1078, Brest 29200, France
| | - Rory J Olson
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Whitney Besse
- Section of Nephrology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Christina M Heyer
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Jessica M Smith
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Marie-Pierre Audrézet
- Department of Molecular Genetics, National Institute of Health and Medical Sciences, INSERM U1078, Brest 29200, France
| | - Katharina Hopp
- Division of Renal Diseases and Hypertension, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80202, USA
| | - Binu Porath
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Beili Shi
- Division of Nephrology, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Saurabh Baheti
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, USA
| | - Sarah R Senum
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Jennifer Arroyo
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Charles D Madsen
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Claude Férec
- Department of Molecular Genetics, National Institute of Health and Medical Sciences, INSERM U1078, Brest 29200, France
| | - Dominique Joly
- Service of Nephrology, Necker Hospital, Paris 75231, France
| | - François Jouret
- Division of Nephrology, University of Liège, Liège 4000, Belgium
| | | | | | | | - Alan S Yu
- Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Korosh Khalili
- Department of Medical Imaging, University Health Network, Toronto, ON M5G 2C4, Canada
| | - York Pei
- Division of Nephrology, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Stefan Somlo
- Section of Nephrology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yannick Le Meur
- Department of Nephrology, University Hospital, European University of Brittany, Brest, Brittany 29200, France
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
15
|
Zheng HC, Gong BC, Zhao S. The meta and bioinformatics analysis of GRP78 expression in gastric cancer. Oncotarget 2017; 8:73017-73028. [PMID: 29069845 PMCID: PMC5641188 DOI: 10.18632/oncotarget.20318] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 08/04/2017] [Indexed: 01/16/2023] Open
Abstract
GRP78 is a molecular chaperone located in endoplasmic reticulum, and induces folding and assembly of newly-synthesized proteins, proteasome degradation of aberrant proteins, and translocation of secretory proteins, autophagy, and epithelial-mesenchymal transition. We performed a systematic meta- and bioinformatics analysis through multiple online databases up to March 14, 2017. It was found that up-regulated GRP78 expression in gastric cancer, compared with normal mucosa at both protein and mRNA levels (p < 0.05). GRP78 expression was positively correlated with depth of invasion, TNM staging and dedifferentiation of gastric cancer (p < 0.05), while its mRNA expression was negatively correlated with depth of invasion, histological grading and dedifferentiation (p < 0.05). A positive association between GRP78 expression and unfavorable overall survival was found in patients with gastric cancer (p < 0.005). A higher GRP78 mRNA expression was positively correlated with overall and progression-free survival rates of all cancer patients, even stratified by aggressive parameters, or as an independent factor (p < 0.05). These findings indicated that GRP78 expression might be employed as a potential marker to indicate gastric carcinogenesis and subsequent progression, even prognosis.
Collapse
Affiliation(s)
- Hua-Chuan Zheng
- Department of Experimental Oncology and Animal Center, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Bao-Cheng Gong
- Department of Experimental Oncology and Animal Center, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Shuang Zhao
- Department of Experimental Oncology and Animal Center, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
16
|
GRP78 haploinsufficiency suppresses acinar-to-ductal metaplasia, signaling, and mutant Kras-driven pancreatic tumorigenesis in mice. Proc Natl Acad Sci U S A 2017; 114:E4020-E4029. [PMID: 28461470 DOI: 10.1073/pnas.1616060114] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a highly lethal disease in critical need of new therapeutic strategies. Here, we report that the stress-inducible 78-kDa glucose-regulated protein (GRP78/HSPA5), a key regulator of endoplasmic reticulum homeostasis and PI3K/AKT signaling, is overexpressed in the acini and PDAC of Pdx1-Cre;KrasG12D/+;p53f/+ (PKC) mice as early as 2 mo, suggesting that GRP78 could exert a protective effect on acinar cells under stress, as during PDAC development. The PKC pancreata bearing wild-type Grp78 showed detectable PDAC by 3 mo and rapid subsequent tumor growth. In contrast, the PKC pancreata bearing a Grp78f/+ allele (PKC78f/+ mice) expressing about 50% of GRP78 maintained normal sizes during the early months, with reduced proliferation and suppression of AKT, S6, ERK, and STAT3 activation. Acinar-to-ductal metaplasia (ADM) has been identified as a key tumor initiation mechanism of PDAC. Compared with PKC, the PKC78f/+ pancreata showed substantial reduction of ADM as well as pancreatic intraepithelial neoplasia-1 (PanIN-1), PanIN-2, and PanIN-3 and delayed onset of PDAC. ADM in response to transforming growth factor α was also suppressed in ex vivo cultures of acinar cell clusters isolated from mouse pancreas bearing targeted heterozygous knockout of Grp78 (c78f/+ ) and subjected to 3D culture in collagen. We further discovered that GRP78 haploinsufficiency in both the PKC78f/+ and c78f/+ pancreata leads to reduction of epidermal growth factor receptor, which is critical for ADM initiation. Collectively, our studies establish a role for GRP78 in ADM and PDAC development.
Collapse
|
17
|
Dadey DYA, Kapoor V, Khudanyan A, Urano F, Kim AH, Thotala D, Hallahan DE. The ATF6 pathway of the ER stress response contributes to enhanced viability in glioblastoma. Oncotarget 2016; 7:2080-92. [PMID: 26716508 PMCID: PMC4811517 DOI: 10.18632/oncotarget.6712] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 11/21/2015] [Indexed: 11/25/2022] Open
Abstract
Therapeutic resistance is a major barrier to improvement of outcomes for patients with glioblastoma. The endoplasmic reticulum stress response (ERSR) has been identified as a contributor to chemoresistance in glioblastoma; however the contributions of the ERSR to radioresistance have not been characterized. In this study we found that radiation can induce ER stress and downstream signaling associated with the ERSR. Induction of ER stress appears to be linked to changes in ROS balance secondary to irradiation. Furthermore, we observed global induction of genes downstream of the ERSR in irradiated glioblastoma. Knockdown of ATF6, a regulator of the ERSR, was sufficient to enhance radiation induced cell death. Also, we found that activation of ATF6 contributes to the radiation-induced upregulation of glucose regulated protein 78 (GRP78) and NOTCH1. Our results reveal ATF6 as a potential therapeutic target to enhance the efficacy of radiation therapy.
Collapse
Affiliation(s)
- David Y A Dadey
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA.,Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO, USA
| | - Vaishali Kapoor
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Arpine Khudanyan
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Fumihiko Urano
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.,Department of Pathology, Washington University School of Medicine, St. Louis, MO, USA
| | - Albert H Kim
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Dinesh Thotala
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Dennis E Hallahan
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA.,Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
18
|
Dadey DYA, Kapoor V, Hoye K, Khudanyan A, Collins A, Thotala D, Hallahan DE. Antibody Targeting GRP78 Enhances the Efficacy of Radiation Therapy in Human Glioblastoma and Non-Small Cell Lung Cancer Cell Lines and Tumor Models. Clin Cancer Res 2016; 23:2556-2564. [PMID: 27815359 DOI: 10.1158/1078-0432.ccr-16-1935] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/07/2016] [Accepted: 10/12/2016] [Indexed: 11/16/2022]
Abstract
Purpose: Non-small cell lung cancer (NSCLC) and glioblastoma multiforme (GBM) have poor median survival. NSCLC and GBM overexpress glucose regulated protein 78 (GRP78), which has a role in radioresistance and recurrence. In this study, we determined the effect of anti-GRP78 antibody and the combined effect of the anti-GRP78 antibody with ionizing radiation (XRT) on NSCLC and GBM cell lines both in vitro and in vivoExperimental Design: NSCLC and GBM cancer cell lines were treated with anti-GRP78 antibodies and evaluated for proliferation, colony formation, cell death, and PI3K/Akt/mTOR signaling. The efficacy of anti-GRP78 antibodies on tumor growth in combination with XRT was determined in vivo in mouse xenograft models.Results: GBM and NSCLC cells treated with anti-GRP78 antibodies showed attenuated cell proliferation, colony formation, and enhanced apoptosis. GBM and NSCLC cells treated with anti-GRP78 antibodies also showed global suppression of PI3K/Akt/mTOR signaling. Combining antibody with XRT resulted in significant tumor growth delay in both NSCLC and GBM heterotopic tumor models.Conclusions: Antibodies targeting GRP78 exhibited antitumor activity and enhanced the efficacy of radiation in NSCLC and GBM both in vitro and in vivo GRP78 is a promising novel target, and anti-GRP78 antibodies could be used as an effective cancer therapy alone or in combination with XRT. Clin Cancer Res; 23(10); 2556-64. ©2016 AACR.
Collapse
Affiliation(s)
- David Y A Dadey
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri.,Medical Scientist Training Program, Washington University in St. Louis, St. Louis, Missouri
| | - Vaishali Kapoor
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri
| | - Kelly Hoye
- Medical Guidance Systems, St. Louis, Missouri
| | - Arpine Khudanyan
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri
| | - Andrea Collins
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri
| | - Dinesh Thotala
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri. .,Siteman Cancer Center, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Dennis E Hallahan
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri. .,Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri.,Siteman Cancer Center, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| |
Collapse
|
19
|
Zhou X, Xing X, Zhang S, Liu L, Wang C, Li L, Ji Q, Liu H. Glucose-regulated protein 78 contributes to the proliferation and tumorigenesis of human colorectal carcinoma via AKT and ERK pathways. Oncol Rep 2016; 36:2723-2730. [PMID: 27634156 DOI: 10.3892/or.2016.5097] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 07/28/2016] [Indexed: 11/06/2022] Open
Abstract
Glucose-regulated protein 78 (GRP78), a molecular chaperon in the endoplasmic reticulum (ER), is overexpressed in a variety of tumor types and plays a critical role in cancer cell proliferation, migration, invasion and drug resistance. However, the mechanisms underlying the role of GRP78 in tumor carcinogenesis remain largely unknown. In the present study, we found that GRP78 knockdown in colorectal carcinoma (CRC) cells significantly inhibited cell proliferation, colony formation and tumorigenesis in vitro and in vivo. The proliferation inhibition of CRC cells by GRP78 knockdown was associated with an S phase arrest, a reduced G1/S transition, and a downregulation of phosphorylation of AKT and ERK1/2, key cell cycle regulatory proteins. In addition, GRP78 knockdown enhanced the apoptosis induced by 5-fluorouracil (5-FU) in CRC cells. Taken together, our results indicate that GRP78 plays an important role in the development and progression of CRC and may have therapeutic potential for CRC patients.
Collapse
Affiliation(s)
- Xuan Zhou
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Xiaoming Xing
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Shuping Zhang
- Department of Obstetrics, Qingdao Municipal Hospital, Qingdao, Shandong 266000, P.R. China
| | - Lili Liu
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Chengqin Wang
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Lin Li
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Qiuxia Ji
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Huamin Liu
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| |
Collapse
|
20
|
Xipell E, Aragón T, Martínez-Velez N, Vera B, Idoate MA, Martínez-Irujo JJ, Garzón AG, Gonzalez-Huarriz M, Acanda AM, Jones C, Lang FF, Fueyo J, Gomez-Manzano C, Alonso MM. Endoplasmic reticulum stress-inducing drugs sensitize glioma cells to temozolomide through downregulation of MGMT, MPG, and Rad51. Neuro Oncol 2016; 18:1109-19. [PMID: 26951384 DOI: 10.1093/neuonc/now022] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 01/29/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Endoplasmic reticulum (ER) stress results from protein misfolding imbalance and has been postulated as a therapeutic strategy. ER stress activates the unfolded protein response which leads to a complex cellular response, including the upregulation of aberrant protein degradation in the ER, with the goal of resolving that stress. O(6)-methylguanine DNA methyltransferase (MGMT), N-methylpurine DNA glycosylase (MPG), and Rad51 are DNA damage repair proteins that mediate resistance to temozolomide in glioblastoma. In this work we sought to evaluate whether ER stress-inducing drugs were able to downmodulate DNA damage repair proteins and become candidates to combine with temozolomide. METHODS MTT assays were performed to evaluate the cytotoxicity of the treatments. The expression of proteins was evaluated using western blot and immunofluorescence. In vivo studies were performed using 2 orthotopic glioblastoma models in nude mice to evaluate the efficacy of the treatments. All statistical tests were 2-sided. RESULTS Treatment of glioblastoma cells with ER stress-inducing drugs leads to downregulation of MGMT, MPG, and Rad51. Inhibition of ER stress through pharmacological treatment resulted in rescue of MGMT, MPG, and Rad51 protein levels. Moreover, treatment of glioblastoma cells with salinomycin, an ER stress-inducing drug, and temozolomide resulted in enhanced DNA damage and a synergistic antitumor effect in vitro. Of importance, treatment with salinomycin/temozolomide resulted in a significant antiglioma effect in 2 aggressive orthotopic intracranial brain tumor models. CONCLUSIONS These findings provide a strong rationale for combining temozolomide with ER stress-inducing drugs as an alternative therapeutic strategy for glioblastoma.
Collapse
Affiliation(s)
- Enric Xipell
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Tomás Aragón
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Naiara Martínez-Velez
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Beatriz Vera
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Miguel Angel Idoate
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Juan José Martínez-Irujo
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Antonia García Garzón
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Marisol Gonzalez-Huarriz
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Arlet M Acanda
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Chris Jones
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Frederick F Lang
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Juan Fueyo
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Candelaria Gomez-Manzano
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Marta M Alonso
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| |
Collapse
|
21
|
YERUSHALMI RINAT, RAITER ANNAT, NALBANDYAN KAREN, HARDY BRITTA. Cell surface GRP78: A potential marker of good prognosis and response to chemotherapy in breast cancer. Oncol Lett 2015; 10:2149-2155. [PMID: 26622810 PMCID: PMC4579811 DOI: 10.3892/ol.2015.3579] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 05/29/2015] [Indexed: 02/07/2023] Open
Abstract
The 78-kDa glucose-regulated protein (GRP78) is a stress induced heat shock protein which, under limiting conditions, functions as a cell surface signaling receptor. Tumor cells are considered to be subjected to a physiologically stressful microenvironment due to their excessive growth. The role of GRP78 in tumor survival has been of notable interest. The present study aimed to assess the potential prognostic and predictive value of cell surface GRP78 expression in breast cancer tumor cells. Cell surface and cytoplasmic expression of GRP78 was examined by immunohistochemical staining of GRP78 in breast cancer archival paraffin-embedded tumor specimens. The cohort studied included breast cancer patients with operable T1,2, estrogen receptor-positive, node-negative cancer who were assessed using the Oncotype DX gene profile, as well as patients with locally advanced disease prior to and following neoadjuvant systemic treatment. GRP78 values were compared between the 2 groups, and prior to and following systemic treatment. Association analyses between GRP78 expression and prognostic markers were also performed. Cox regression analysis was used to examine the impact of these variables on disease-free survival (DFS). No differences in cytoplasmic GRP78 expression were observed. By contrast, the rates of cell surface GRP78 expression were 74.1% in the early stage operable patients, 36% in neoadjuvant systemic treatment patients prior to treatment and 62.5% in patients following systemic treatment (P<0.039). Positive cell surface GRP78 expression was associated with increased expression of the progesterone receptor (P=0.024), p53 expression (P=0.022) and improved DFS (P=0.047). In the case of GRP78 positivity, a trend for a superior response to chemotherapy was observed (P=0.19). The results of the present study indicated that cell surface GRP78 may be used as a marker for good prognosis in breast cancer and a potential marker for response to chemotherapy.
Collapse
Affiliation(s)
- RINAT YERUSHALMI
- Davidoff Cancer Center, Rabin Medical Center, Beilinson Campus, Petah Tikva 49100, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - ANNAT RAITER
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Felsenstein Medical Research Center, Rabin Medical Center, Beilinson Campus, Petah Tikva 49100, Israel
| | - KAREN NALBANDYAN
- Department of Pathology, Rabin Medical Center, Beilinson Campus, Petah Tikva 49100, Israel
| | - BRITTA HARDY
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Felsenstein Medical Research Center, Rabin Medical Center, Beilinson Campus, Petah Tikva 49100, Israel
| |
Collapse
|
22
|
Zhang Y, Li N, Wang D, Chen Y, Li G. Expression and significance of glucose-regulated protein 78 in human osteosarcoma. Oncol Lett 2015; 9:2268-2274. [PMID: 26137054 DOI: 10.3892/ol.2015.3030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 02/20/2015] [Indexed: 12/29/2022] Open
Abstract
The present study aimed to investigate the expression of glucose-regulated protein 78 (GRP78) in osteosarcoma cells, and analyze the differences in expression between tumor and normal tissues, pre- and post-chemotherapy patients and metastatic and non-metastatic tumors. According to these results, the associations between the expression of GRP78 and tumor growth, metastasis and chemotherapeutics could be determined. Between 2007 and 2012, 60 patients who had been diagnosed with osteosarcoma were selected for the present study. Of these patients, 20 presented with non-metastatic tumors and 40 with metastatic tumors, and 20 had been treated without chemotherapy and 40 with chemotherapy. In addition, 60 specimens obtained from adjacent normal tissues were collected for the control groups. Immunofluorescence staining was used to examine the expression of GRP78 in the different tissues. The total RNA and protein were extracted from crushed tissues and used in the reverse transcription polymerase chain reaction and western blot analysis. GRP78 was primarily located in the intracavity of the endoplasmic reticulum. The expression level of GRP78 in the tumor tissue was higher than that in the normal tissue surrounding the tumor (P<0.01). In addition, the level was higher in the metastatic tumors compared with the non-metastatic tumors (P<0.05), and in the non-chemotherapy-treated patients compared with the chemotherapy-treated patients (P<0.01). The expression level of GRP78 mRNA in the tumor tissue was higher than that in the normal tissue (P<0.01). Furthermore, the level was higher in the metastasis group than in the non-metastasis group (P<0.05), and in the non-chemotherapy group than in the chemotherapy group (P<0.01). The expression level of GRP78 protein was higher in the tumor tissue compared with the normal tissue (P<0.01), in the metastasis group compared with the non-metastasis group (P<0.05), and in the non-chemotherapy group compared with the chemotherapy group (P<0.01). In conclusion, the present study detected the expression of GRP78 in patients with osteosarcoma and revealed a higher expression level in the tumor tissues compared with the normal tissues around the tumor, in the metastasis group compared with the non-metastasis group and in the non-chemotherapy-treated group compared with the chemotherapy-treated group.
Collapse
Affiliation(s)
- Yongkui Zhang
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, P.R. China
| | - Nianhu Li
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, P.R. China
| | - Dongli Wang
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, P.R. China
| | - Yiqiang Chen
- Department of Orthopedics, The First People's Hospital of Tai'an City, Tai'an, Shandong, P.R. China
| | - Gang Li
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, P.R. China
| |
Collapse
|
23
|
Targeting the glucose-regulated protein-78 abrogates Pten-null driven AKT activation and endometrioid tumorigenesis. Oncogene 2015; 34:5418-26. [PMID: 25684138 PMCID: PMC4537850 DOI: 10.1038/onc.2015.4] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 12/23/2014] [Accepted: 01/02/2015] [Indexed: 12/21/2022]
Abstract
Rates of the most common gynecologic cancer, endometrioid adenocarcinoma (EAC), continue to rise, mirroring the global epidemic of obesity, a well-known EAC risk factor. Thus, identifying novel molecular targets to prevent and/or mitigate EAC is imperative. The prevalent Type 1 EAC commonly harbors loss of the tumor suppressor, Pten, leading to AKT activation. The major endoplasmic reticulum (ER) chaperone, GRP78, is a potent pro-survival protein to maintain ER homeostasis, and as a cell surface protein, is known to regulate the PI3K/AKT pathway. To determine whether targeting GRP78 could suppress EAC development, we created a conditional knockout mouse model utilizing progesterone receptor (PR)-Cre-recombinase to achieve Pten and Grp78 (cPtenf/fGrp78f/f) deletion in the endometrial epithelium. Mice with a single Pten (cPtenf/f) deletion developed well-differentiated EAC by 4 weeks. In contrast, no cPtenf/fGrp78f/f mice developed EAC, even after more than 8 months of observation. Histologic examination of uteri from cPtenf/fGrp78f/f mice also revealed no complex atypical hyperplasia (CAH), a well-established EAC precursor. These histologic observations among the cPtenf/fGrp78f/f murine uteri also corresponded to abrogation of AKT activation within the endometrium. We further observed that GRP78 co-localized with activated AKT on the surface of EAC thus providing an opportunity for therapeutic targeting. Consistent with previous findings that cell surface GRP78 is an upstream regulator of PI3K/AKT signaling, we show here that in vivo short-term systemic treatment with a highly specific monoclonal antibody against GRP78 suppressed AKT activation and increased apoptosis in the cPtenf/f tumors. Collectively, these findings present GRP78-targeting therapy as an efficacious therapeutic option for EAC.
Collapse
|
24
|
Yamamoto K, Shichiri H, Uda A, Yamashita K, Nishioka T, Kume M, Makimoto H, Nakagawa T, Hirano T, Hirai M. Apoptotic Effects of the Extracts of Cordyceps militaris
via Erk Phosphorylation in a Renal Cell Carcinoma Cell Line. Phytother Res 2015; 29:707-13. [DOI: 10.1002/ptr.5305] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 12/17/2014] [Accepted: 01/07/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Kazuhiro Yamamoto
- Department of Pharmacy; Kobe University Hospital; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
| | - Hiroaki Shichiri
- Division of Pharmacokinetics, Department of Internal Related; Kobe University Graduate School of Medicine; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
| | - Atsushi Uda
- Department of Pharmacy; Kobe University Hospital; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
| | - Kazuhiko Yamashita
- Department of Pharmacy; Kobe University Hospital; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
| | - Tatsuya Nishioka
- Department of Pharmacy; Kobe University Hospital; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
| | - Manabu Kume
- Department of Pharmacy; Kobe University Hospital; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
| | - Hiroo Makimoto
- Department of Pharmacy; Kobe University Hospital; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
| | - Tsutomu Nakagawa
- Department of Pharmacy; Kobe University Hospital; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
- Division of Pharmacokinetics, Department of Internal Related; Kobe University Graduate School of Medicine; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
| | - Takeshi Hirano
- Department of Pharmacy; Kobe University Hospital; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
- Division of Pharmacokinetics, Department of Internal Related; Kobe University Graduate School of Medicine; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
| | - Midori Hirai
- Department of Pharmacy; Kobe University Hospital; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
- Division of Pharmacokinetics, Department of Internal Related; Kobe University Graduate School of Medicine; 7-5-2 Kusunoki-cho Chuo-ku Kobe 650-0017 Japan
| |
Collapse
|
25
|
WU XINYU, FAN RUITAI, YAN XINHUI, CUI JING, XU JUNLING, GU HAO, GAO YONGJU. Endoplasmic reticulum stress protects human thyroid carcinoma cell lines against ionizing radiation-induced apoptosis. Mol Med Rep 2014; 11:2341-7. [DOI: 10.3892/mmr.2014.2956] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 10/31/2014] [Indexed: 11/06/2022] Open
|
26
|
Lu T, Yang W, Wang Z, Hu Z, Zeng X, Yang C, Wang Y, Zhang Y, Li F, Liu Z, Wang D, Ye Z. Knockdown of glucose-regulated protein 78/binding immunoglobulin heavy chain protein expression by asymmetric small interfering RNA induces apoptosis in prostate cancer cells and attenuates migratory capability. Mol Med Rep 2014; 11:249-56. [PMID: 25338653 DOI: 10.3892/mmr.2014.2737] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Accepted: 04/07/2014] [Indexed: 11/05/2022] Open
Abstract
Glucose-regulated protein 78 [GRP78, also termed binding immunoglobulin heavy chain protein (Bip)] may be involved in cancer progression and metastasis. However, to date there has been minimal investigation into its potential role in human prostate cancer cells. Recent studies have demonstrated that asymmetric small interfering RNA (asiRNA)-mediated gene silencing is more effective and longer-lasting than conventional symmetric siRNA. Thus, the current study aimed to investigate the effects of GRP78-specific asiRNA on human prostate cancer cells. A series of asiRNAs was synthesized and their efficiency in silencing GRP78 expression in PC-3 human prostate cancer cells was evaluated. The effects of knockdown using asiRNAs were compared to those of knockdown using symmetric siRNAs. The effect of GRP78 silencing on PC-3 cell apoptosis and migration, and the possible mechanisms governing these biological processes were examined. Compared with the symmetric siRNA, transfection with the 15 base pair asiRNA (asiGRP78-3) resulted in greater downregulation of GRP78 expression. GRP78 depletion in PC-3 cells resulted in increased apoptosis and decreased migration of these cells. Experiments investigating the underlying mechanisms of these effects revealed that knockdown of GRP78 attenuated protein kinase B activation and decreased the expression of pro-caspase 9, pro-caspase 3 and vimentin. In conclusion, knockdown of GRP78/Bip expression with asymmetric siRNA led to increased prostate cancer cell apoptosis and reduced cellular migration.
Collapse
Affiliation(s)
- Tong Lu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Weimin Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhihua Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhiquan Hu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xing Zeng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Chunguang Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Ye Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yong Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Fan Li
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhuo Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Dongbiao Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhangqun Ye
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
27
|
Gonzalez-Gronow M, Gomez CF, de Ridder GG, Ray R, Pizzo SV. Binding of tissue-type plasminogen activator to the glucose-regulated protein 78 (GRP78) modulates plasminogen activation and promotes human neuroblastoma cell proliferation in vitro. J Biol Chem 2014; 289:25166-76. [PMID: 25059665 DOI: 10.1074/jbc.m114.589341] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
The glucose-regulated protein 78 (GRP78) is a plasminogen (Pg) receptor on the cell surface. In this study, we demonstrate that GRP78 also binds the tissue-type plasminogen activator (t-PA), which results in a decrease in K(m) and an increase in the V(max) for both its amidolytic activity and activation of its substrate, Pg. This results in accelerated Pg activation when GRP78, t-PA, and Pg are bound together. The increase in t-PA activity is the result of a mechanism involving a t-PA lysine-dependent binding site in the GRP78 amino acid sequence (98)LIGRTWNDPSVQQDIKFL(115). We found that GRP78 is expressed on the surface of neuroblastoma SK-N-SH cells where it is co-localized with the voltage-dependent anion channel (VDAC), which is also a t-PA-binding protein in these cells. We demonstrate that both Pg and t-PA serve as a bridge between GRP78 and VDAC bringing them together to facilitate Pg activation. t-PA induces SK-N-SH cell proliferation via binding to GRP78 on the cell surface. Furthermore, Pg binding to the COOH-terminal region of GRP78 stimulates cell proliferation via its microplasminogen domain. This study confirms previous findings from our laboratory showing that GRP78 acts as a growth factor-like receptor and that its association with t-PA, Pg, and VDAC on the cell surface may be part of a system controlling cell growth.
Collapse
Affiliation(s)
- Mario Gonzalez-Gronow
- From the Department of Biological Sciences, Laboratory of Environmental Neurotoxicology Faculty of Medicine, Universidad Católica del Norte, Coquimbo 1781421, Chile and the Department of Pathology, Duke University, Medical Center, Durham, North Carolina 27710
| | - Cristian Farias Gomez
- From the Department of Biological Sciences, Laboratory of Environmental Neurotoxicology Faculty of Medicine, Universidad Católica del Norte, Coquimbo 1781421, Chile and
| | - Gustaaf G de Ridder
- the Department of Pathology, Duke University, Medical Center, Durham, North Carolina 27710
| | - Rupa Ray
- the Department of Pathology, Duke University, Medical Center, Durham, North Carolina 27710
| | - Salvatore V Pizzo
- the Department of Pathology, Duke University, Medical Center, Durham, North Carolina 27710
| |
Collapse
|
28
|
Parmar JH, Cook KL, Shajahan-Haq AN, Clarke PAG, Tavassoly I, Clarke R, Tyson JJ, Baumann WT. Modelling the effect of GRP78 on anti-oestrogen sensitivity and resistance in breast cancer. Interface Focus 2014; 3:20130012. [PMID: 24511377 DOI: 10.1098/rsfs.2013.0012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Understanding the origins of resistance to anti-oestrogen drugs is of critical importance to many breast cancer patients. Recent experiments show that knockdown of GRP78, a key gene in the unfolded protein response (UPR), can re-sensitize resistant cells to anti-oestrogens, and overexpression of GRP78 in sensitive cells can cause them to become resistant. These results appear to arise from the operation and interaction of three cellular systems: the UPR, autophagy and apoptosis. To determine whether our current mechanistic understanding of these systems is sufficient to explain the experimental results, we built a mathematical model of the three systems and their interactions. We show that the model is capable of reproducing previously published experimental results and some new data gathered specifically for this paper. The model provides us with a tool to better understand the interactions that bring about anti-oestrogen resistance and the effects of GRP78 on both sensitive and resistant breast cancer cells.
Collapse
Affiliation(s)
- Jignesh H Parmar
- Department of Biological Sciences , Virginia Polytechnic Institute and State University , Blacksburg, VA 24061 , USA
| | - Katherine L Cook
- Department of Oncology, Lombardi Comprehensive Cancer Center , Georgetown University Medical Center , Washington, DC 20057 , USA
| | - Ayesha N Shajahan-Haq
- Department of Oncology, Lombardi Comprehensive Cancer Center , Georgetown University Medical Center , Washington, DC 20057 , USA
| | - Pamela A G Clarke
- Department of Oncology, Lombardi Comprehensive Cancer Center , Georgetown University Medical Center , Washington, DC 20057 , USA
| | - Iman Tavassoly
- Department of Biological Sciences , Virginia Polytechnic Institute and State University , Blacksburg, VA 24061 , USA
| | - Robert Clarke
- Department of Oncology, Lombardi Comprehensive Cancer Center , Georgetown University Medical Center , Washington, DC 20057 , USA
| | - John J Tyson
- Department of Biological Sciences , Virginia Polytechnic Institute and State University , Blacksburg, VA 24061 , USA
| | - William T Baumann
- Bradley Department of Electrical and Computer Engineering , Virginia Polytechnic Institute and State University , Blacksburg, VA 24061 , USA
| |
Collapse
|
29
|
Ouyang YB, Giffard RG. MicroRNAs regulate the chaperone network in cerebral ischemia. Transl Stroke Res 2013; 4:693-703. [PMID: 24323423 PMCID: PMC3864745 DOI: 10.1007/s12975-013-0280-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 07/24/2013] [Accepted: 07/30/2013] [Indexed: 01/17/2023]
Abstract
The highly evolutionarily conserved 70 kDa heat shock protein (HSP70) family was first understood for its role in protein folding and response to stress. Subsequently, additional functions have been identified for it in regulation of organelle interaction, of the inflammatory response, and of cell death and survival. Overexpression of HSP70 family members is associated with increased resistance to and improved recovery from cerebral ischemia. MicroRNAs (miRNAs) are important posttranscriptional regulators that interact with multiple target messenger RNAs (mRNA) coordinately regulating target genes, including chaperones. The members of the HSP70 family are now appreciated to work together as networks to facilitate organelle communication and regulate inflammatory signaling and cell survival after cerebral ischemia. This review will focus on the new concept of the role of the chaperone network in the organelle network and its novel regulation by miRNA.
Collapse
Affiliation(s)
- Yi-Bing Ouyang
- Department of Anesthesia, Stanford University School of Medicine, 300 Pasteur Drive, S272A and S290, Stanford, CA, 94305-5117, USA,
| | | |
Collapse
|
30
|
Zhang D, Zhou B, Li Y, Wang M, Wang C, Zhou Z, Sun X. Polymorphisms of glucose-regulated protein 78 and risk of colorectal cancer: a case-control study in southwest China. PLoS One 2013; 8:e66791. [PMID: 23818965 PMCID: PMC3688565 DOI: 10.1371/journal.pone.0066791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 05/12/2013] [Indexed: 02/05/2023] Open
Abstract
Glucose-regulated protein 78 (GRP78), an endoplasmic reticulum chaperone, up-regulation serves as an efficient mechanism to promote malignant transformation of colorectal cancer (CRC) and protect CRC cells against apoptosis. Recently, the analysis of GRP78 polymorphisms has already determined that GRP78 rs391957 polymorphism could predict clinical outcome in CRC patients. Thus, we tested whether GRP78 polymorphisms are related to the risk of CRC. In this study, we detected two GRP78 polymorphisms (rs391957 (C>T) and rs430397 (G>A)) in 414 CRC cases and 502 hospital-based cancer-free healthy controls in Southwest China using a polymerase chain reaction-restriction fragment length polymorphism technique. Compared with the CC genotype, carriers of CT and TT genotypes of rs391957 polymorphism had higher risks of CRC (odds ratio (OR) = 1.39, 95% confidence interval (CI) = 1.06-1.83 for CT genotype and OR = 2.10, 95% CI = 1.06-4.14 for TT genotype, respectively). In CRC cases, the variant T allele was significantly associated with tumor invasion stage (P = 0.030), but not with status of lymph nodes metastasis (P = 0.052). Compared with the GG genotype, carriers of GA and AA genotypes of rs430397 polymorphism had higher risks of CRC (OR = 1.63, 95% CI = 1.23-2.15 for GA genotype and OR = 2.92, 95% CI = 1.23-6.94 for AA genotype, respectively). The rs430397 polymorphism was not associated with the clinicopathological characteristics of CRC. These data provide the first evidence that GRP78 rs391957 and rs430397 polymorphisms could serve as markers to predict the risk of CRC.
Collapse
Affiliation(s)
- Dan Zhang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Digestive Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Bin Zhou
- Laboratory of Digestive Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuan Li
- Laboratory of Digestive Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Mojin Wang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Digestive Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Cun Wang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Digestive Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zongguang Zhou
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Digestive Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- * E-mail:
| | - Xiaofeng Sun
- Division of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, County Council of Östergötland, University of Linköping, Linköping, Sweden
| |
Collapse
|
31
|
Avila MF, Cabezas R, Torrente D, Gonzalez J, Morales L, Alvarez L, Capani F, Barreto GE. Novel interactions of GRP78: UPR and estrogen responses in the brain. Cell Biol Int 2013; 37:521-32. [DOI: 10.1002/cbin.10058] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 01/22/2013] [Indexed: 12/21/2022]
Affiliation(s)
- Marco Fidel Avila
- Departamento de Nutrición y Bioquímica; Facultad de Ciencias, Pontificia Universidad Javeriana; Bogotá D.C., Colombia
| | - Ricardo Cabezas
- Departamento de Nutrición y Bioquímica; Facultad de Ciencias, Pontificia Universidad Javeriana; Bogotá D.C., Colombia
| | - Daniel Torrente
- Departamento de Nutrición y Bioquímica; Facultad de Ciencias, Pontificia Universidad Javeriana; Bogotá D.C., Colombia
| | - Janneth Gonzalez
- Departamento de Nutrición y Bioquímica; Facultad de Ciencias, Pontificia Universidad Javeriana; Bogotá D.C., Colombia
| | - Ludis Morales
- Departamento de Nutrición y Bioquímica; Facultad de Ciencias, Pontificia Universidad Javeriana; Bogotá D.C., Colombia
| | - Lisandro Alvarez
- Laboratorio de Citoarquitectura y Plasticidad Neuronal, Instituto de Investigaciones Cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), Facultad de Medicina, UBA-CONICET; Marcelo T. de Alvear 2270, C1122AAJ Buenos Aires; Argentina
| | - Francisco Capani
- Laboratorio de Citoarquitectura y Plasticidad Neuronal, Instituto de Investigaciones Cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), Facultad de Medicina, UBA-CONICET; Marcelo T. de Alvear 2270, C1122AAJ Buenos Aires; Argentina
| | - George E. Barreto
- Departamento de Nutrición y Bioquímica; Facultad de Ciencias, Pontificia Universidad Javeriana; Bogotá D.C., Colombia
| |
Collapse
|
32
|
Young CN, Cao X, Guruju MR, Pierce JP, Morgan DA, Wang G, Iadecola C, Mark AL, Davisson RL. ER stress in the brain subfornical organ mediates angiotensin-dependent hypertension. J Clin Invest 2012; 122:3960-4. [PMID: 23064361 DOI: 10.1172/jci64583] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 08/23/2012] [Indexed: 12/28/2022] Open
Abstract
Although endoplasmic reticulum (ER) stress is a pathologic mechanism in a variety of chronic diseases, it is unclear what role it plays in chronic hypertension (HTN). Dysregulation of brain mechanisms controlling arterial pressure is strongly implicated in HTN, particularly in models involving angiotensin II (Ang II). We tested the hypothesis that ER stress in the brain is causally linked to Ang II-dependent HTN. Chronic systemic infusion of low-dose Ang II in C57BL/6 mice induced slowly developing HTN, which was abolished by co-infusion of the ER stress inhibitor tauroursodeoxycholic acid (TUDCA) into the lateral cerebroventricle. Investigations of the brain regions involved revealed robust increases in ER stress biomarkers and profound ER morphological abnormalities in the circumventricular subfornical organ (SFO), a region outside the blood-brain barrier and replete with Ang II receptors. Ang II-induced HTN could be prevented in this model by selective genetic supplementation of the ER chaperone 78-kDa glucose-regulated protein (GRP78) in the SFO. These data demonstrate that Ang II-dependent HTN is mediated by ER stress in the brain, particularly the SFO. To our knowledge, this is the first report that ER stress, notably brain ER stress, plays a key role in chronic HTN. Taken together, these findings may have broad implications for the pathophysiology of this disease.
Collapse
Affiliation(s)
- Colin N Young
- Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853-6401, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Hill R, Li Y, Tran LM, Dry S, Calvopina JH, Garcia A, Kim C, Wang Y, Donahue TR, Herschman HR, Wu H. Cell intrinsic role of COX-2 in pancreatic cancer development. Mol Cancer Ther 2012; 11:2127-37. [PMID: 22784710 DOI: 10.1158/1535-7163.mct-12-0342] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
COX-2 is upregulated in pancreatic ductal adenocarcinomas (PDAC). However, how COX-2 promotes PDAC development is unclear. While previous studies have evaluated the efficacy of COX-2 inhibition via the use of nonsteroidal anti-inflammatory drugs (NSAID) or the COX-2 inhibitor celecoxib in PDAC models, none have addressed the cell intrinsic versus microenvironment roles of COX-2 in modulating PDAC initiation and progression. We tested the cell intrinsic role of COX-2 in PDAC progression using both loss-of-function and gain-of-function approaches. Cox-2 deletion in Pdx1+ pancreatic progenitor cells significantly delays the development of PDAC in mice with K-ras activation and Pten haploinsufficiency. Conversely, COX-2 overexpression promotes early onset and progression of PDAC in the K-ras mouse model. Loss of PTEN function is a critical factor in determining lethal PDAC onset and overall survival. Mechanistically, COX-2 overexpression increases p-AKT levels in the precursor lesions of Pdx1(+); K-ras(G12D)(/+); Pten(lox)(/+) mice in the absence of Pten LOH. In contrast, Cox-2 deletion in the same setting diminishes p-AKT levels and delays cancer progression. These data suggest an important cell intrinsic role for COX-2 in tumor initiation and progression through activation of the PI3K/AKT pathway. PDAC that is independent of intrinsic COX-2 expression eventually develops with decreased FKBP5 and increased GRP78 expression, two alternate pathways leading to AKT activation. Together, these results support a cell intrinsic role for COX-2 in PDAC development and suggest that while anti-COX-2 therapy may delay the development and progression of PDAC, mechanisms known to increase chemoresistance through AKT activation must also be overcome.
Collapse
Affiliation(s)
- Reginald Hill
- Corresponding Author: Hong Wu, Department of Molecular and Medical Pharmacology, CHS 33-131, 650 CE Young Drive South, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Sokolowska I, Woods AG, Gawinowicz MA, Roy U, Darie CC. Identification of a potential tumor differentiation factor receptor candidate in prostate cancer cells. FEBS J 2012; 279:2579-94. [PMID: 22613557 DOI: 10.1111/j.1742-4658.2012.08641.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Tumor differentiation factor (TDF) is a pituitary protein that is secreted into the bloodstream and has an endocrine function. TDF and TDF-P1, a 20-residue peptide selected from the ORF of TDF, induce differentiation in human breast and prostate cancer cells, but not in other cells. TDF has no known mechanism of action. In our recent study, we identified heat shock 70 kDa proteins (HSP70s) as TDF receptors (TDF-Rs) in breast cancer cells. Therefore, we sought to investigate whether TDF-R candidates from prostate cancer cells are the same as those identified in breast cancer cells. Here, we used TDF-P1 to purify the potential TDF-R candidates by affinity purification chromatography from DU145 and PC3 steroid-resistant prostate cancer cells, LNCaP steroid-responsive prostate cancer cells, and nonprostate NG108 neuroblastoma and BLK CL.4 fibroblast-like cells. We identified the purified proteins by MS, and validated them by western blotting, immunofluorescence microscopy, immunoaffinity purification chromatography, and structural biology. We identified seven candidate proteins, of which three were from the HSP70 family. These three proteins were validated as potential TDF-R candidates in LNCaP steroid-responsive and in DU145 and PC3 steroid-resistant prostate cancer cells, but not in NG108 neuroblastoma and BLK CL.4 fibroblast-like cells. Our previous study and the current study suggest that GRP78, and perhaps HSP70s, are strong TDF-R candidates, and further suggest that TDF interacts with its receptors exclusively in breast and prostate cells, inducing cell differentiation through a novel, steroid-independent pathway.
Collapse
Affiliation(s)
- Izabela Sokolowska
- Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | | | | | | | | |
Collapse
|
35
|
Chang YJ, Huang YP, Li ZL, Chen CH. GRP78 knockdown enhances apoptosis via the down-regulation of oxidative stress and Akt pathway after epirubicin treatment in colon cancer DLD-1 cells. PLoS One 2012; 7:e35123. [PMID: 22529978 PMCID: PMC3329422 DOI: 10.1371/journal.pone.0035123] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 03/13/2012] [Indexed: 01/22/2023] Open
Abstract
INTRODUCTION The 78-kDa glucose-regulated protein (GRP78) is induced in the cancer microenvironment and can be considered as a novel predictor of responsiveness to chemotherapy in many cancers. In this study, we found that intracellular reactive oxygen species (ROS) and nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation were higher in GRP78 knockdown DLD-1 colon cancer cells compared with scrambled control cells. METHODOLOGY/PRINCIPAL FINDINGS Treatment with epirubicin in GRP78 knockdown DLD-1 cells enhanced apoptosis and was associated with decreased production of intracellular ROS. In addition, apoptosis was increased by the antioxidants propyl gallate (PG) and dithiothreitol (DTT) in epirubicin-treated scrambled control cells. Epirubicin-treated GRP78 knockdown cells resulted in more inactivated Akt pathway members, such as phosphorylated Akt and GSK-3β, as well as downstream targets of β-catenin expression. Knockdown of Nrf2 with small interfering RNA (siRNA) increased apoptosis in epirubicin-treated GRP78 knockdown cells, which suggested that Nrf2 may be a primary defense mechanism in GRP78 knockdown cells. We also demonstrated that epirubicin-treated GRP78 knockdown cells could decrease survival pathway signaling through the redox activation of protein phosphatase 2A (PP2A), which is a serine/threonine phosphatase that negatively regulates the Akt pathway. CONCLUSIONS Our results indicate that epirubicin decreased the intracellular ROS in GRP78 knockdown cells, which decreased survival signaling through both the Akt pathway and the activation of PP2A. Together, these mechanisms contributed to the enhanced level of epirubicin-induced apoptosis that was observed in the GRP78 knockdown cells.
Collapse
Affiliation(s)
- Yu-Jia Chang
- Graduate Institute of Clinical Medicine College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan
- Cancer Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yi-Ping Huang
- Department of Microbiology, Immunology and Biopharmaceuticals, College of Life Sciences, National Chiayi University, Chiayi City, Taiwan
| | - Zih-Ling Li
- Department of Microbiology, Immunology and Biopharmaceuticals, College of Life Sciences, National Chiayi University, Chiayi City, Taiwan
| | - Ching-Hsein Chen
- Department of Microbiology, Immunology and Biopharmaceuticals, College of Life Sciences, National Chiayi University, Chiayi City, Taiwan
- * E-mail:
| |
Collapse
|
36
|
ER-Mitochondria Crosstalk during Cerebral Ischemia: Molecular Chaperones and ER-Mitochondrial Calcium Transfer. Int J Cell Biol 2012; 2012:493934. [PMID: 22577383 PMCID: PMC3335182 DOI: 10.1155/2012/493934] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 01/12/2012] [Indexed: 11/24/2022] Open
Abstract
It is commonly believed that sustained elevations in the mitochondrial matrix Ca2+ concentration are a major feature of the intracellular cascade of lethal events during cerebral ischemia. The physical association between the endoplasmic reticulum (ER) and mitochondria, known as the mitochondria-associated ER membrane (MAM), enables highly efficient transmission of Ca2+ from the ER to mitochondria under both physiological and pathological conditions. Molecular chaperones are well known for their protective effects during cerebral ischemia. It has been demonstrated recently that many molecular chaperones coexist with MAM and regulate the MAM and thus Ca2+ concentration inside mitochondria. Here, we review recent research on cerebral ischemia and MAM, with a focus on molecular chaperones and ER-mitochondrial calcium transfer.
Collapse
|
37
|
Li Z, Li Z. Glucose regulated protein 78: a critical link between tumor microenvironment and cancer hallmarks. Biochim Biophys Acta Rev Cancer 2012; 1826:13-22. [PMID: 22426159 DOI: 10.1016/j.bbcan.2012.02.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 02/26/2012] [Accepted: 02/27/2012] [Indexed: 12/27/2022]
Abstract
Glucose regulated protein 78 (GRP78) has long been recognized as a molecular chaperone in the endoplasmic reticulum (ER) and can be induced by the ER stress response. Besides its location in the ER, GRP78 has been found to be present in cell plasma membrane, cytoplasm, mitochondria, nucleus as well as cellular secretions. GRP78 is implicated in tumor cell proliferation, apoptosis resistance, immune escape, metastasis and angiogenesis, and its elevated expression usually correlates with a variety of tumor microenvironmental stresses, including hypoxia, glucose deprivation, lactic acidosis and inflammatory response. GRP78 protein acts as a centrally located sensor of stress, which feels and adapts to the alteration in the tumor microenvironment. This article reviews the potential contributions of GRP78 to the acquisition of cancer hallmarks based on intervening in stress responses caused by tumor niche alterations. The paper also introduces several potential GRP78 relevant targeted therapies.
Collapse
Affiliation(s)
- Zongwei Li
- Institute of Biotechnology, The Key Laboratory of Clinical Biology and Molecular Engineering of Education Ministry, Shanxi University, 030006 Taiyuan, PR China
| | | |
Collapse
|
38
|
Goldenberg-Cohen N, Raiter A, Gaydar V, Dratviman-Storobinsky O, Goldstein T, Weizman A, Hardy B. Peptide-binding GRP78 protects neurons from hypoxia-induced apoptosis. Apoptosis 2012; 17:278-288. [PMID: 22120956 DOI: 10.1007/s10495-011-0678-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Brain ischemia has major consequences leading to the apoptosis of astrocytes and neurons. Glucose-regulated protein 78 (GRP78) known for its role in endoplasmic reticulum stress alleviation was discovered on several cell surfaces acting as a receptor for signaling pathways. We have previously described peptides that bind cell surface GRP78 on endothelial cells to induce angiogenesis. We have also reported that ADoPep1 binds cardiomyocytes to prevent apoptosis of ischemic heart cells. In this study we describe the effect of hypoxia on astrocytes and neurons cell surface GRP78. Under hypoxic conditions, there was an increase of more than fivefold in GRP78 on cell surface of neurons while astrocytes were not affected. The addition of the GRP78 binding peptide, ADoPep1, to neurons decreased the percentage of GRP78 positive cells and did not change the percent of astrocytes. However, a significant increase in early and late apoptosis of both astrocytes and neurons under hypoxia was attenuated in the presence of ADoPep1. Intravitreal administration of ADoPep1 to mice in a model of optic nerve crush significantly reduced retinal cell loss after 21 days compared to the crush-damaged eyes without treatment or by control saline vehicle injection. Histological staining demonstrated reduced GRP78 after ADoPep1 treatment. The mechanism of peptide neuroprotection was demonstrated by the inhibition of hypoxia induced caspase 3/7 activity, cytochrome c release and p38 phosphorylation. This study is the first report on hypoxic neuronal and astrocyte cell surface GRP78 and suggests a potential therapeutic target for neuroprotection.
Collapse
Affiliation(s)
- Nitza Goldenberg-Cohen
- Eye Research Laboratory, Felsenstein Medical Research Center, Tel-Aviv University, School of Medicine, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel
| | | | | | | | | | | | | |
Collapse
|
39
|
Aryal BP, Paunesku T, Woloschak GE, He C, Jensen MP. A proteomic approach to identification of plutonium-binding proteins in mammalian cells. J Proteomics 2011; 75:1505-14. [PMID: 22146473 DOI: 10.1016/j.jprot.2011.11.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 10/29/2011] [Accepted: 11/19/2011] [Indexed: 11/18/2022]
Abstract
Plutonium can enter the body through different routes and remains there for decades; however its specific biochemical interactions are poorly defined. We, for the first time, have studied plutonium-binding proteins using a metalloproteomic approach with rat PC12 cells. A combination of immobilized metal ion chromatography, 2D gel electrophoresis, and mass spectrometry was employed to analyze potential plutonium-binding proteins. Our results show that several proteins from PC12 cells show affinity towards Pu(4+)-NTA (plutonium bound to nitrilotriacetic acid). Proteins from seven different spots in the 2D gel were identified. In contrast to the previously known plutonium-binding proteins transferrin and ferritin, which bind ferric ions, most identified proteins in our experiment are known to bind calcium, magnesium, or divalent transition metal ions. The identified plutonium interacting proteins also have functional roles in downregulation of apoptosis and other pro-proliferative processes. MetaCore™ analysis based on this group of proteins produced a pathway with a statistically significant association with development of neoplastic diseases.
Collapse
Affiliation(s)
- Baikuntha P Aryal
- Chemical Science and Engineering Division, Argonne National Lab, Argonne, IL, USA
| | | | | | | | | |
Collapse
|
40
|
Sokolowska I, Woods AG, Gawinowicz MA, Roy U, Darie CC. Identification of potential tumor differentiation factor (TDF) receptor from steroid-responsive and steroid-resistant breast cancer cells. J Biol Chem 2011; 287:1719-33. [PMID: 22130669 DOI: 10.1074/jbc.m111.284091] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tumor differentiation factor (TDF) is a recently discovered protein, produced by the pituitary gland and secreted into the bloodstream. TDF and TDF-P1, a 20-amino acid peptide selected from the open reading frame of TDF, induce differentiation in human breast and prostate cancer cells but not in other cells. TDF protein has no identified site of action or receptor, and its mechanism of action is unknown. Here, we used TDF-P1 to purify and identify potential TDF receptor (TDF-R) candidates from MCF7 steroid-responsive breast cancer cells and non-breast HeLa cancerous cells using affinity purification chromatography (AP), and mass spectrometry (MS). We identified four candidate proteins from the 70-kDa heat shock protein (HSP70) family in MCF7 cells. Experiments in non-breast HeLa cancerous cells did not identify any TDF-R candidates. AP and MS experiments were validated by AP and Western blotting (WB). We additionally looked for TDF-R in steroid-resistant BT-549 cells and human dermal fibroblasts (HDF-a) using AP and WB. TDF-P1 interacts with potential TDF-R candidates from MCF7 and BT-549 breast cells but not from HeLa or HDF-a cells. Immunofluorescence (IF) experiments identified GRP78, a TDF-R candidate, at the cell surface of MCF7, BT-549 breast cells, and HeLa cells but not HDF-a cells. IF of other HSP70 proteins demonstrated labeling on all four cell types. These results point toward GRP78 and HSP70 proteins as strong TDF-R candidates and suggest that TDF interacts with its receptor, exclusively on breast cells, through a steroid-independent pathway.
Collapse
Affiliation(s)
- Izabela Sokolowska
- Department of Chemistry and Biomolecular Science, Biochemistry and Proteomics Group, Clarkson University, Potsdam, New York 13699-5810, USA
| | | | | | | | | |
Collapse
|
41
|
Uckun FM, Qazi S, Ozer Z, Garner AL, Pitt J, Ma H, Janda KD. Inducing apoptosis in chemotherapy-resistant B-lineage acute lymphoblastic leukaemia cells by targeting HSPA5, a master regulator of the anti-apoptotic unfolded protein response signalling network. Br J Haematol 2011; 153:741-52. [PMID: 21517817 DOI: 10.1111/j.1365-2141.2011.08671.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We present previously unknown evidence that the immunoglobulin heavy chain binding protein BIP/HSPA5, also known as glucose regulated protein (GRP)78, serving as a pivotal component of the pro-survival axis of the unfolded protein response (UPR) signalling network, is abundantly expressed in relapsed B-lineage acute lymphoblastic leukaemia (ALL) and contributes to chemotherapy resistance of leukaemic B-cell precursors. The resistance of B-lineage ALL cells to the standard anti-leukaemic drug vincristine was overcome by the HSPA5 inhibitor epigallocatechin gallate, which inhibits the anti-apoptotic function of HSPA5 by targeting its ATP-binding domain. Notably, chemotherapy-resistant B-lineage ALL cells underwent apoptosis within 48 h of exposure to a doxorubicin-conjugated cell-penetrating cyclic anti-HSPA5 peptide targeting surface-expressed HSPA5 molecules on leukaemia cells. The identification of the HSPA5 as a chemoresistance biomarker and molecular target for B-lineage ALL may lead to new anti-leukaemic treatment strategies that are much needed.
Collapse
Affiliation(s)
- Fatih M Uckun
- Department of Pediatrics, Division of Hematology-Oncology, University of Southern California Keck School of Medicine, Los Angeles, CA, USA.
| | | | | | | | | | | | | |
Collapse
|