1
|
Zare H, Bakherad H, Esfahani AN, Aghamollaei H, Gargari SLM, Aliomrani M, Ebrahimizadeh W. Investigating the effect of cGRP78 vaccine against different cancer cells and its role in reducing melanoma metastasis. Res Pharm Sci 2024; 19:73-82. [PMID: 39006979 PMCID: PMC11244710 DOI: 10.4103/1735-5362.394822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 04/30/2023] [Accepted: 11/08/2023] [Indexed: 07/16/2024] Open
Abstract
Background and purpose Treatment of malignancies with chemotherapy and surgery is often associated with disease recurrence and metastasis. Immunotherapy improves cancer treatment by creating an active response against tumor antigens. Various cancer cells express a large amount of glucose-regulated protein 78 (GRP78) protein on their surface. Stimulating the immune system against this antigen can expose cancer cells to the immune system. Herein, we investigated the effectiveness of a cGRP78-based vaccine against different cancer cells. Experimental approach BALB/c mice were immunized with the cGRP78. The humoral immune response against different cancer cells was assessed by Cell-ELISA. The cellular immunity response was determined by splenocyte proliferation assay with different cancer antigens. The effect of vaccination on metastasis was investigated in vaccinated mice by injecting melanoma cancer cells into the tail of mice. Findings/Results These results indicated that the cGRP78 has acceptable antigenicity and stimulates the immune system to produce antibodies. After three injections, the amount of produced antibody was significantly different from the control group. Compared to the other three cell types, Hela and HepG2 showed the highest reaction to the serum of vaccinated mice. Cellular immunity against the B16F10 cell line had the best results compared to other cells. The metastasis results showed that after 30 days, the growth of B16F10 melanoma cancer cells was not noticeable in the lung tissue of vaccinated mice. Conclusion and implications Considering the resistance of vaccinated mice to metastasis, this vaccine offers a promising prospect for cancer treatment by inhibiting the spread of cancer cells.
Collapse
Affiliation(s)
- Hamed Zare
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Hamid Bakherad
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
- Pharmaceutical Sciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arman Nasr Esfahani
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Aghamollaei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Mahdi Aliomrani
- Department of Pharmacology and Toxicology, Isfahan Pharmaceutical Science Research Center, Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Walead Ebrahimizadeh
- Department of Surgery, Division of Urology, McGill University and the Research Institute of the McGill University Health Centre (RI MUHC), Montreal, Quebec, Canada
| |
Collapse
|
2
|
Yu W, Zhang H, Yuan Y, Tang J, Chen X, Liu T, Zhao X. Chimeric Antigen Receptor T Cells Targeting Cell Surface GRP78 to Eradicate Acute Myeloid Leukemia. Front Cell Dev Biol 2022; 10:928140. [PMID: 35990606 PMCID: PMC9387679 DOI: 10.3389/fcell.2022.928140] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/31/2022] [Indexed: 11/19/2022] Open
Abstract
Acute myeloid leukemia (AML) is a serious, life-threatening hematological malignancy. The treatment outcome of relapsed or refractory AML patients remains dismal, and new treatment options are needed. Chimeric antigen receptor (CAR) T cells have been successful in improving the prognosis for B-lineage acute lymphoblastic leukemia and lymphoma by targeting CD19. However, CAR T-cell therapy for AML is still elusive, owing to the lack of a tumor-specific cell surface antigen and spare hematopoietic stem cells (HSCs). This study generated a novel CAR construction that targets the cell surface protein glucose-regulated protein 78 (GRP78) (csGRP78). We confirmed that GRP78-CAR T cells demonstrate an anti-tumor effect against human AML cells in vitro. In xenograft models, GRP78-CAR T cells effectively eliminate AML cells and protect mice against systemic leukemia, in the meanwhile, prolonging survival. In addition, GRP78-CAR T cells also specifically eradicate the primary AML patient-derived blast. In particular, GRP78-CAR T cells spare normal HSCs, highlighting that GRP78-CAR is a promising approach for the therapy of AML.
Collapse
Affiliation(s)
- Wei Yu
- Laboratory of Animal Tumor Models, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Hang Zhang
- Department of Hematology, Institute of Hematology, West China Hospital of Sichuan University, Chengdu, China
| | - Yuncang Yuan
- Laboratory of Animal Tumor Models, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Tang
- Laboratory of Animal Tumor Models, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xinchuan Chen
- Department of Hematology, Institute of Hematology, West China Hospital of Sichuan University, Chengdu, China
| | - Ting Liu
- Department of Hematology, Institute of Hematology, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Ting Liu, ; Xudong Zhao,
| | - Xudong Zhao
- Laboratory of Animal Tumor Models, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Ting Liu, ; Xudong Zhao,
| |
Collapse
|
3
|
Chen J, Lynn EG, Yousof TR, Sharma H, MacDonald ME, Byun JH, Shayegan B, Austin RC. Scratching the Surface—An Overview of the Roles of Cell Surface GRP78 in Cancer. Biomedicines 2022; 10:biomedicines10051098. [PMID: 35625836 PMCID: PMC9138746 DOI: 10.3390/biomedicines10051098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/01/2022] [Accepted: 05/05/2022] [Indexed: 02/04/2023] Open
Abstract
The 78 kDa glucose-regulated protein (GRP78) is considered an endoplasmic reticulum (ER)-resident molecular chaperone that plays a crucial role in protein folding homeostasis by regulating the unfolded protein response (UPR) and inducing numerous proapoptotic and autophagic pathways within the eukaryotic cell. However, in cancer cells, GRP78 has also been shown to migrate from the ER lumen to the cell surface, playing a role in several cellular pathways that promote tumor growth and cancer cell progression. There is another insidious consequence elicited by cell surface GRP78 (csGRP78) on cancer cells: the accumulation of csGRP78 represents a novel neoantigen leading to the production of anti-GRP78 autoantibodies that can bind csGRP78 and further amplify these cellular pathways to enhance cell growth and mitigate apoptotic cell death. This review examines the current body of literature that delineates the mechanisms by which ER-resident GRP78 localizes to the cell surface and its consequences, as well as potential therapeutics that target csGRP78 and block its interaction with anti-GRP78 autoantibodies, thereby inhibiting further amplification of cancer cell progression.
Collapse
Affiliation(s)
- Jack Chen
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
| | - Edward G. Lynn
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
| | - Tamana R. Yousof
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
| | - Hitesh Sharma
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
| | - Melissa E. MacDonald
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
| | - Jae Hyun Byun
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
| | - Bobby Shayegan
- Department of Surgery, Division of Urology, The Research Institute of St. Joe′s Hamilton, McMaster University, ON L8N 4A6, Canada;
| | - Richard C. Austin
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
- Correspondence: ; Tel.: +1-905-522-1155 (ext. 35175)
| |
Collapse
|
4
|
Farshbaf M, Khosroushahi AY, Mojarad-Jabali S, Zarebkohan A, Valizadeh H, Walker PR. Cell surface GRP78: An emerging imaging marker and therapeutic target for cancer. J Control Release 2020; 328:932-941. [DOI: 10.1016/j.jconrel.2020.10.055] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 12/12/2022]
|
5
|
Limso C, Ngo JM, Nguyen P, Leal S, Husain A, Sahoo D, Ghosh P, Bhandari D. The Gα-interacting vesicle-associated protein interacts with and promotes cell surface localization of GRP78 during endoplasmic reticulum stress. FEBS Lett 2019; 594:1088-1100. [PMID: 31736058 DOI: 10.1002/1873-3468.13685] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 11/07/2019] [Accepted: 11/12/2019] [Indexed: 12/18/2022]
Abstract
Cell surface translocation of the chaperone glucose-regulated protein 78 kDa (GRP78) is a key event that promotes cancer cell survival during endoplasmic reticulum (ER) stress. Here, we identify Gα-interacting vesicle-associated protein (GIV) - an enhancer of prosurvival signaling during ER stress - as a binding partner of GRP78. We show that GIV and GRP78 interact in an ER stress-dependent manner through their respective carboxyl terminal domains and that GIV aids in the localization of GRP78 to the plasma membrane. Kaplan-Meier analysis of disease-free survival in cancer patients shows poor prognosis for patients with high expression of both GIV and GRP78, further suggesting a vital role for these two proteins in enhancing cancer cell viability.
Collapse
Affiliation(s)
- Clariss Limso
- Department of Chemistry and Biochemistry, California State University Long Beach, CA, USA
| | - Jordan Matthew Ngo
- Department of Chemistry and Biochemistry, California State University Long Beach, CA, USA
| | - Peter Nguyen
- Department of Chemistry and Biochemistry, California State University Long Beach, CA, USA
| | - Stephanie Leal
- Department of Chemistry and Biochemistry, California State University Long Beach, CA, USA
| | - Aida Husain
- Department of Chemistry and Biochemistry, California State University Long Beach, CA, USA
| | - Debashis Sahoo
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA.,Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Pradipta Ghosh
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.,Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Deepali Bhandari
- Department of Chemistry and Biochemistry, California State University Long Beach, CA, USA
| |
Collapse
|
6
|
Lu T, Wang Y, Xu K, Zhou Z, Gong J, Zhang Y, Gong H, Dai Q, Yang J, Xiong B, Song Z, Yang G. Co-downregulation of GRP78 and GRP94 Induces Apoptosis and Inhibits Migration in Prostate Cancer Cells. Open Life Sci 2019; 14:384-391. [PMID: 33817173 PMCID: PMC7874808 DOI: 10.1515/biol-2019-0043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 05/27/2019] [Indexed: 12/13/2022] Open
Abstract
Background Both glucose-regulated protein 78 kDa (GRP78) and glucose-regulated protein 94 kDa (GRP94) are important molecular chaperones that play critical roles in maintaining tumor survival and progression. This study investigated the effects in prostate cancer cells following the downregulation of GRP78 and GRP94. Methods RNA interference was used to downregulate GRP78 and GRP94 expression in the prostate cancer cell line, PC-3. The effects on apoptosis and cell migration was examined along with expression of these related proteins. Results Small interfering RNAs targeting GRP78 and GRP94 successfully down-regulated their expression. This resulted in the induction of apoptosis and inhibition of cell migration. Preliminary mechanistic studies indicated that caspase-9 (cleaved) and Bax expression levels were upregulated while Bcl-2 and vimentin expression levels were downregulated. Conclusion Co-downregulation of GRP78 and GRP94 expression induces apoptosis and inhibits migration in prostate cancer cells.
Collapse
Affiliation(s)
- Tong Lu
- Department of Urology, The First People's Hospital of Tianmen City, East No.1, Renmin Avenue, Tianmen City, Hubei 431700, P.R.China
| | - Yue Wang
- Sinopharm Wuhan Plasma-derived Biotherapies Co. Ltd., No.1 attached No.1, Zhengdian Gold Industrial Park Road, Jiangxia District, Wuhan, Hubei 430070, P.R.China
| | - Kang Xu
- Department of Urology, The First People's Hospital of Tianmen City, East No.1, Renmin Avenue, Tianmen City, Hubei 431700, P.R.China
| | - Zhijun Zhou
- Department of Urology, The First People's Hospital of Tianmen City, East No.1, Renmin Avenue, Tianmen City, Hubei 431700, P.R.China
| | - Juan Gong
- Department of Urology, The First People's Hospital of Tianmen City, East No.1, Renmin Avenue, Tianmen City, Hubei 431700, P.R.China
| | - Yingang Zhang
- Department of Urology, The First People's Hospital of Tianmen City, East No.1, Renmin Avenue, Tianmen City, Hubei 431700, P.R.China
| | - Hua Gong
- Department of Urology, The First People's Hospital of Tianmen City, East No.1, Renmin Avenue, Tianmen City, Hubei 431700, P.R.China
| | - Qiang Dai
- Department of Urology, The First People's Hospital of Tianmen City, East No.1, Renmin Avenue, Tianmen City, Hubei 431700, P.R.China
| | - Jun Yang
- Department of Urology, The First People's Hospital of Tianmen City, East No.1, Renmin Avenue, Tianmen City, Hubei 431700, P.R.China
| | - Biao Xiong
- Department of Urology, The First People's Hospital of Tianmen City, East No.1, Renmin Avenue, Tianmen City, Hubei 431700, P.R.China
| | - Ze Song
- Department of Urology, The First People's Hospital of Tianmen City, East No.1, Renmin Avenue, Tianmen City, Hubei 431700, P.R.China
| | - Gang Yang
- Department of Urology, The First People's Hospital of Tianmen City, East No.1, Renmin Avenue, Tianmen City, Hubei 431700, P.R.China
| |
Collapse
|
7
|
Shen K, Johnson DW, Vesey DA, McGuckin MA, Gobe GC. Role of the unfolded protein response in determining the fate of tumor cells and the promise of multi-targeted therapies. Cell Stress Chaperones 2018; 23:317-334. [PMID: 28952072 PMCID: PMC5904077 DOI: 10.1007/s12192-017-0844-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 09/13/2017] [Indexed: 02/06/2023] Open
Abstract
Although there have been advances in our understanding of carcinogenesis and development of new treatments, cancer remains a common cause of death. Many regulatory pathways are incompletely understood in cancer development and progression, with a prime example being those related to the endoplasmic reticulum (ER). The pathological sequelae that arise from disruption of ER homeostasis are not well defined. The ER is an organelle that is responsible for secretory protein biosynthesis and the quality control of protein folding. The ER triggers an unfolded protein response (UPR) when misfolded proteins accumulate, and while the UPR acts to restore protein folding and ER homeostasis, this response can work as a switch to determine the death or survival of cells. The treatment of cancer with agents that target the UPR has shown promising outcomes. The UPR has wide crosstalk with other signaling pathways. Multi-targeted cancer therapies which target the intersections within signaling networks have shown synergistic tumoricidal effects. In the present review, the basic cellular and signaling pathways of the ER and UPR are introduced; then the crosstalk between the ER and other signaling pathways is summarized; and ultimately, the evidence that the UPR is a potential target for cancer therapy is discussed. Regulation of the UPR downstream signaling is a common therapeutic target for different tumor types. Tumoricidal effects achieved from modulating the UPR downstream signaling could be enhanced by phosphodiesterase 5 (PDE5) inhibitors. Largely untapped by Western medicine for cancer therapies are Chinese herbal medicines. This review explores and discusses the value of some Chinese herbal extracts as PDE5 inhibitors.
Collapse
Affiliation(s)
- Kunyu Shen
- Kidney Disease Research Group, UQ Diamantina Institute, Translational Research Institute, The University of Queensland, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia
| | - David W Johnson
- Kidney Disease Research Group, UQ Diamantina Institute, Translational Research Institute, The University of Queensland, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia
- Department of Nephrology, Princess Alexandra Hospital, Woolloongabba, Brisbane, Australia
- Centre for Health Services Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - David A Vesey
- Department of Nephrology, Princess Alexandra Hospital, Woolloongabba, Brisbane, Australia
- Centre for Health Services Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Michael A McGuckin
- Mucosal Disease Inflammatory Disease Biology and Therapeutics Group, UQ Mater Research Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Glenda C Gobe
- Kidney Disease Research Group, UQ Diamantina Institute, Translational Research Institute, The University of Queensland, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia.
- Centre for Health Services Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
8
|
Endoplasmic reticulum stress activates SRC, relocating chaperones to the cell surface where GRP78/CD109 blocks TGF-β signaling. Proc Natl Acad Sci U S A 2018; 115:E4245-E4254. [PMID: 29654145 DOI: 10.1073/pnas.1714866115] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The discovery that endoplasmic reticulum (ER) luminal chaperones such as GRP78/BiP can escape to the cell surface upon ER stress where they regulate cell signaling, proliferation, apoptosis, and immunity represents a paradigm shift. Toward deciphering the mechanisms, we report here that, upon ER stress, IRE1α binds to and triggers tyrosine kinase SRC activation, leading to ASAP1 phosphorylation and Golgi accumulation of ASAP1 and Arf1-GTP, resulting in KDEL receptor dispersion from the Golgi and suppression of retrograde transport. At the cell surface, GRP78 binds to and acts in concert with a glycosylphosphatidylinositol-anchored protein, CD109, in blocking TGF-β signaling by promoting the routing of the TGF-β receptor to the caveolae, thereby disrupting its binding to and activation of Smad2. Collectively, we uncover a SRC-mediated signaling cascade that leads to the relocalization of ER chaperones to the cell surface and a mechanism whereby GRP78 counteracts the tumor-suppressor effect of TGF-β.
Collapse
|
9
|
Knockdown of GRP78 enhances cell death by cisplatin and radiotherapy in nasopharyngeal cells. Anticancer Drugs 2017; 27:726-33. [PMID: 27254284 DOI: 10.1097/cad.0000000000000377] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Radiotherapy and adjuvant cisplatin chemotherapy are the mainstream approaches in the treatment of nasopharyngeal carcinoma (NPC). These have been shown to effectively improve the outcome and reduce tumor recurrence. However, radiotherapy and chemotherapy resistance during the course of treatment has become more common recently, resulting in the failure of NPC therapy. Therefore, new therapeutic strategies or adjuvant drugs are urgently needed. The current study was designed to look for new treatment strategies or auxiliary drugs in the treatment of NPC. Two human NPC cell lines, HNE1 and HNE1/DDP, were used to examine the relationship between endoplasmic reticulum stress and cell resistance to ionizing radiation (IR) and cisplatin (DDP). Cell proliferation was assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Meanwhile, propidium iodide (PI) staining and PI/Annexin V staining were used to observe cell apoptosis. Finally, western blot was used to detect the endogenous expression of glucose-regulated protein 78 (GRP78) and other apoptosis-related proteins. GRP78 small interference RNA was transfected using Lipofectamine 2000. Compared with HNE1/DDP, IR and DDP increased the cell apoptosis and inhibited the cell proliferation of HNE1. Inhibition of GRP78 can reverse IR and DDP resistance in NPC cells by PI/Annexin V staining. Knockdown of GRP78 upregulates the expression of pro-apoptotic proteins and downregulates the expression of antiapoptotic proteins. These results indicate that HNE1 is more sensitive to DDP and IR than HNE1/DDP. Knockdown GRP78 can reverse IR and DDP resistance in NPC cells. Inhibition of GRP78 gives us a new target to overcome resistance to radiotherapy and chemotherapy of NPC cells. Thus, this study should be further explored in vivo and assessed for possible clinical applications.
Collapse
|
10
|
Al-Hashimi AA, Lebeau P, Majeed F, Polena E, Lhotak Š, Collins CAF, Pinthus JH, Gonzalez-Gronow M, Hoogenes J, Pizzo SV, Crowther M, Kapoor A, Rak J, Gyulay G, D'Angelo S, Marchiò S, Pasqualini R, Arap W, Shayegan B, Austin RC. Autoantibodies against the cell surface-associated chaperone GRP78 stimulate tumor growth via tissue factor. J Biol Chem 2017; 292:21180-21192. [PMID: 29066620 PMCID: PMC5743090 DOI: 10.1074/jbc.m117.799908] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 09/05/2017] [Indexed: 12/24/2022] Open
Abstract
Tumor cells display on their surface several molecular chaperones that normally reside in the endoplasmic reticulum. Because this display is unique to cancer cells, these chaperones are attractive targets for drug development. Previous epitope-mapping of autoantibodies (AutoAbs) from prostate cancer patients identified the 78-kDa glucose-regulated protein (GRP78) as one such target. Although we previously showed that anti-GRP78 AutoAbs increase tissue factor (TF) procoagulant activity on the surface of tumor cells, the direct effect of TF activation on tumor growth was not examined. In this study, we explore the interplay between the AutoAbs against cell surface-associated GRP78, TF expression/activity, and prostate cancer progression. First, we show that tumor GRP78 expression correlates with disease stage and that anti-GRP78 AutoAb levels parallel prostate-specific antigen concentrations in patient-derived serum samples. Second, we demonstrate that these anti-GRP78 AutoAbs target cell-surface GRP78, activating the unfolded protein response and inducing tumor cell proliferation through a TF-dependent mechanism, a specific effect reversed by neutralization or immunodepletion of the AutoAb pool. Finally, these AutoAbs enhance tumor growth in mice bearing human prostate cancer xenografts, and heparin derivatives specifically abrogate this effect by blocking AutoAb binding to cell-surface GRP78 and decreasing TF expression/activity. Together, these results establish a molecular mechanism in which AutoAbs against cell-surface GRP78 drive TF-mediated tumor progression in an experimental model of prostate cancer. Heparin derivatives counteract this mechanism and, as such, represent potentially appealing compounds to be evaluated in well-designed translational clinical trials.
Collapse
Affiliation(s)
- Ali A Al-Hashimi
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
- the Department of Surgery, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Paul Lebeau
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Fadwa Majeed
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Enio Polena
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Šárka Lhotak
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Celeste A F Collins
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Jehonathan H Pinthus
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
- the Department of Surgery, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Mario Gonzalez-Gronow
- the Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710
| | - Jen Hoogenes
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
- the Department of Surgery, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Salvatore V Pizzo
- the Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710
| | - Mark Crowther
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Anil Kapoor
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
- the Department of Surgery, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Janusz Rak
- the Department of Pediatrics, Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Gabriel Gyulay
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Sara D'Angelo
- the University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico 87106
- the Divisions of Molecular Medicine and
| | - Serena Marchiò
- the University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico 87106
- the Divisions of Molecular Medicine and
- the Department of Oncology, University of Turin, 10124 Turin, Italy, and
- the Candiolo Cancer Institute-Fondazione del Piemonte per l'Oncologia (FPO)-Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), 10060 Candiolo, Italy
| | - Renata Pasqualini
- the University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico 87106
- the Divisions of Molecular Medicine and
| | - Wadih Arap
- the University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico 87106
- Hematology/Oncology, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico 87131
| | - Bobby Shayegan
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
- the Department of Surgery, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Richard C Austin
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada,
| |
Collapse
|
11
|
Van Hoesen K, Meynier S, Ribaux P, Petignat P, Delie F, Cohen M. Circulating GRP78 antibodies from ovarian cancer patients: a promising tool for cancer cell targeting drug delivery system? Oncotarget 2017; 8:107176-107187. [PMID: 29291021 PMCID: PMC5739806 DOI: 10.18632/oncotarget.22412] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 09/04/2017] [Indexed: 11/25/2022] Open
Abstract
Glucose-regulated protein 78 (GRP78) is a chaperone protein that has a high frequency in tumor cells. Normally it is found in the endoplasmic reticulum to assist in protein folding, but under cellular stress, GRP78 influences proliferative signaling pathways at the cell surface. The increased expression elicits autoantibody production, providing a biomarker of ovarian cancer, as well as other types of cancer. This study aims to determine the epitope recognition of GRP78 autoantibodies isolated from serum of ovarian cancer patients and use the identified antibodies to design new drug delivery systems to specifically target cancer cells. We first confirmed that the membrane GRP78 levels are increased in ovarian cancer cells and positively correlate with proliferation. However, the level of circulating GRP78 autoantibodies did not correlate with membrane GRP78 expression in ovarian cancer cells and was lower, although not significantly, compared to control patients. We then determined the epitope recognition of GRP78 autoantibodies and showed that treatment with paclitaxel-loaded nanoparticles coated with anti-GRP78 antibodies significantly decreased tumor development in chick embryo culture of ovarian cancer cell tumors compared to paclitaxel treatment alone. This evidence suggests that nanoparticle drug delivery systems coupled with antibodies against GRP78 has potential as a powerful therapy against ovarian cancer.
Collapse
Affiliation(s)
- Kylie Van Hoesen
- Department of Gynecology Obstetrics, University of Geneva, 1205 Geneva, Switzerland
| | - Sonia Meynier
- Department of Gynecology Obstetrics, University of Geneva, 1205 Geneva, Switzerland
| | - Pascale Ribaux
- Department of Gynecology Obstetrics, University of Geneva, 1205 Geneva, Switzerland
| | - Patrick Petignat
- Department of Gynecology Obstetrics, University of Geneva, 1205 Geneva, Switzerland
| | - Florence Delie
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 1205 Geneva, Switzerland
| | - Marie Cohen
- Department of Gynecology Obstetrics, University of Geneva, 1205 Geneva, Switzerland
| |
Collapse
|
12
|
Zhao S, Li H, Wang Q, Su C, Wang G, Song H, Zhao L, Luan Z, Su R. The role of c-Src in the invasion and metastasis of hepatocellular carcinoma cells induced by association of cell surface GRP78 with activated α2M. BMC Cancer 2015; 15:389. [PMID: 25958313 PMCID: PMC4455704 DOI: 10.1186/s12885-015-1401-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 04/29/2015] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Emerging data have suggested that cell surface GRP78 is a multifunctional receptor and has been linked to proliferative and antiapoptotic signaling cascades. Activated α2-macroglobin (α2M*) is a natural circulating ligand of cell surface GRP78. Association of cell surface GRP78 with α2M* is involved in the regulation of cell proliferation, survival and apoptosis in human cancers. METHODS The invasion and metastasis of HCC cells were examined using transwell and wound healing assay; Cell surface expression of GRP78 was detected by in cell western assay. Translocation of GRP78 from cytosol to cell surface was observed by transfection of GRP78-EGFP plus TRIRC-WGA staining. The levels of Src, phosphor-Src, FAK, phospho-FAK, EGFR, phospho-EGFR, phospho-Cortactin, phospho-Paxillin were determined by western blot. Cell surface expression of GRP78 in HCC tissue samples was observed by immunofluorescence. The distribution of Paxillin and Cortactin in HCC cells was also observed by immunofluorescence. The interaction between GRP78 and Src were detected by far-western blot, co-immunoprecipitation and GST pulldown. GRP78 mRNA was detected by RT-PCR. RESULTS In the current study, we showed that association of cell surface GRP78 with α2M* stimulated the invasion and metastasis of HCC. Cell surface GRP78 could interact directly with c-Src, promoted the phosphorylation of c-Src at Y416. Inhibition of the tyrosine kinase activity of c-Src with PP2 reverted the stimulatory effect caused by association of cell surface GRP78 with α2M*. Moreover, association of cell surface GRP78 with α2M* facilitates the interaction between EGFR and c-Src and consequently phosphorylated EGFR at Y1101 and Y845, promoting the invasion and metastasis of HCCs. However, inhibition of the tyrosine kinase of c-Src do not affect the interaction between EGFR and Src. CONCLUSION c-Src plays a critical role in the invasion and metastasis of HCC induced by association of cell surface GRP78 with α2M*. Cell surface GRP78 directly binds and phosphorylates c-Src. As a consequence, c-Src phosphorylated EGFR, promoting the invasion and metastasis of HCCs.
Collapse
Affiliation(s)
- Song Zhao
- Central laboratory, Liaoning Medical College, No 40 Songpo Road, Jinzhou, 121001, China.
| | - Hongdan Li
- Central laboratory, Liaoning Medical College, No 40 Songpo Road, Jinzhou, 121001, China.
| | - Qingjun Wang
- Oncology Department, the First Affiliated Hospital of Liaoning Medical College, No 40 Songpo Road, Jinzhou, 121001, China.
| | - Chang Su
- Veterinary Medicine Department, Liaoning Medical College, No 40 Songpo Road, Jinzhou, 121001, China.
| | - Guan Wang
- Central laboratory, Liaoning Medical College, No 40 Songpo Road, Jinzhou, 121001, China.
| | - Huijuan Song
- Central laboratory, Liaoning Medical College, No 40 Songpo Road, Jinzhou, 121001, China.
| | - Liang Zhao
- Pharmacy Department, Liaoning Medical College, No 40 Songpo Road, Jinzhou, 121000, China.
| | - Zhidong Luan
- Development Department, Liaoning Medical College, No 40 Songpo Road, Jinzhou, 121000, China.
| | - Rongjian Su
- Central laboratory, Liaoning Medical College, No 40 Songpo Road, Jinzhou, 121001, China. .,Cell Biology AND Genetic Department, Liaoning Medical College, No 40 Songpo Road, Jinzhou, 121000, China.
| |
Collapse
|
13
|
Misra UK, Pizzo SV. Activated α2-macroglobulin binding to human prostate cancer cells triggers insulin-like responses. J Biol Chem 2015; 290:9571-87. [PMID: 25720493 DOI: 10.1074/jbc.m114.617837] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Indexed: 12/21/2022] Open
Abstract
Ligation of cell surface GRP78 by activated α2-macroglobulin (α2M*) promotes cell proliferation and suppresses apoptosis. α2M*-treated human prostate cancer cells exhibit a 2-3-fold increase in glucose uptake and lactate secretion, an effect similar to insulin treatment. In both α2M* and insulin-treated cells, the mRNA levels of SREBP1-c, SREBP2, fatty-acid synthase, acetyl-CoA carboxylase, ATP citrate lyase, and Glut-1 were significantly increased together with their protein levels, except for SREBP2. Pretreatment of cells with α2M* antagonist antibody directed against the carboxyl-terminal domain of GRP78 blocks these α2M*-mediated effects, and silencing GRP78 expression by RNAi inhibits up-regulation of ATP citrate lyase and fatty-acid synthase. α2M* induces a 2-3-fold increase in lipogenesis as determined by 6-[(14)C]glucose or 1-[(14)C]acetate incorporation into free cholesterol, cholesterol esters, triglycerides, free fatty acids, and phosphatidylcholine, which is blocked by inhibitors of fatty-acid synthase, PI 3-kinase, mTORC, or an antibody against the carboxyl-terminal domain of GRP78. We also assessed the incorporation of [(14)CH3]choline into phosphatidylcholine and observed similar effects. Lipogenesis is significantly affected by pretreatment of prostate cancer cells with fatostatin A, which blocks sterol regulatory element-binding protein proteolytic cleavage and activation. This study demonstrates that α2M* functions as a growth factor, leading to proliferation of prostate cancer cells by promoting insulin-like responses. An antibody against the carboxyl-terminal domain of GRP78 may have important applications in prostate cancer therapy.
Collapse
Affiliation(s)
- Uma Kant Misra
- From the Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710
| | - Salvatore Vincent Pizzo
- From the Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710
| |
Collapse
|
14
|
Mo L, Bachelder RE, Kennedy M, Chen PH, Chi JT, Berchuck A, Cianciolo G, Pizzo SV. Syngeneic Murine Ovarian Cancer Model Reveals That Ascites Enriches for Ovarian Cancer Stem-Like Cells Expressing Membrane GRP78. Mol Cancer Ther 2015; 14:747-56. [PMID: 25589495 DOI: 10.1158/1535-7163.mct-14-0579] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 12/29/2014] [Indexed: 12/21/2022]
Abstract
Patients with ovarian cancer are generally diagnosed at FIGO (International Federation of Gynecology and Obstetrics) stage III/IV, when ascites is common. The volume of ascites correlates positively with the extent of metastasis and negatively with prognosis. Membrane GRP78, a stress-inducible endoplasmic reticulum chaperone that is also expressed on the plasma membrane ((mem)GRP78) of aggressive cancer cells, plays a crucial role in the embryonic stem cell maintenance. We studied the effects of ascites on ovarian cancer stem-like cells using a syngeneic mouse model. Our study demonstrates that ascites-derived tumor cells from mice injected intraperitoneally with murine ovarian cancer cells (ID8) express increased (mem)GRP78 levels compared with ID8 cells from normal culture. We hypothesized that these ascites-associated (mem)GRP78(+) cells are cancer stem-like cells (CSC). Supporting this hypothesis, we show that (mem)GRP78(+) cells isolated from murine ascites exhibit increased sphere forming and tumor initiating abilities compared with (mem)GRP78(-) cells. When the tumor microenvironment is recapitulated by adding ascites fluid to cell culture, ID8 cells express more (mem)GRP78 and increased self-renewing ability compared with those cultured in medium alone. Moreover, compared with their counterparts cultured in normal medium, ID8 cells cultured in ascites, or isolated from ascites, show increased stem cell marker expression. Antibodies directed against the carboxy-terminal domain of GRP78: (i) reduce self-renewing ability of murine and human ovarian cancer cells preincubated with ascites and (ii) suppress a GSK3α-AKT/SNAI1 signaling axis in these cells. Based on these data, we suggest that (mem)GRP78 is a logical therapeutic target for late-stage ovarian cancer.
Collapse
Affiliation(s)
- Lihong Mo
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Robin E Bachelder
- Department of Pathology, Duke University Medical Center, Durham, North Carolina.
| | - Margaret Kennedy
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Po-Han Chen
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina. Center for Genomic and Computational Biology, Duke University Medical Center, Durham, North Carolina
| | - Jen-Tsan Chi
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina. Center for Genomic and Computational Biology, Duke University Medical Center, Durham, North Carolina
| | - Andrew Berchuck
- Department of Obstetrics/Gynecology, Division of Gynecologic Oncology, Duke University Medical Center, Durham, North Carolina
| | - George Cianciolo
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Salvatore V Pizzo
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
15
|
Abstract
The glucose-regulated proteins (GRPs) are stress-inducible chaperones that mostly reside in the endoplasmic reticulum or the mitochondria. Recent advances show that the GRPs have functions that are distinct from those of the related heat shock proteins, and they can be actively translocated to other cellular locations and assume novel functions that control signalling, proliferation, invasion, apoptosis, inflammation and immunity. Mouse models further identified their specific roles in development, tumorigenesis, metastasis and angiogenesis. This Review describes their discovery and regulation, as well as their biological functions in cancer. Promising agents that use or target the GRPs are being developed, and their efficacy as anticancer therapeutics is also discussed.
Collapse
Affiliation(s)
- Amy S Lee
- Department of Biochemistry and Molecular Biology, University of Southern California Norris Comprehensive Cancer Center, 1441 Eastlake Avenue, Room 5308, Los Angeles, California 900899176, USA
| |
Collapse
|
16
|
Peng Y, Li Z, Li Z. GRP78 secreted by tumor cells stimulates differentiation of bone marrow mesenchymal stem cells to cancer-associated fibroblasts. Biochem Biophys Res Commun 2013; 440:558-63. [PMID: 24113381 DOI: 10.1016/j.bbrc.2013.09.108] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 09/20/2013] [Indexed: 01/09/2023]
Abstract
Cancer-associated fibroblasts (CAFs), one type of tumor-associated stromal cells, have been shown to provide a favorable environment for the malignant tumor progression. Extensive reports have demonstrated that mesenchymal stem cells (MSCs) can function as precursors for CAFs. However, the mechanisms by which tumor cells induce the transition of MSCs to CAFs have not been well established. GRP78, traditionally known as an endoplasmic reticulum (ER) chaperone, has been identified to overexpress in a variety of tumor entities and be involved in promoting survival and chemoresistance of tumor cells. Here, we interrogated the role of GRP78 in the generation of CAFs from MSCs. The results showed that GRP78 treatment induced expression of α-smooth muscle actin (α-SMA), a marker for CAFs, in human bone marrow mesenchymal stem cells (HBMSCs) as well as murine bone marrow mesenchymal stem cells (BMMSCs). This phenomenon was correlated with the stimulated phosphorylation of Smad2/3. Furthermore, the GRP78-induced α-SMA expression in HBMSCs was obviously attenuated by SB431542, a TGF-β type I receptor kinase inhibitor. Taken together, the present data suggested that tumor-derived secreted GRP78 elicited the differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) to CAFs through activating TGF-β/Smad signaling pathway, which may represent a novel mechanism for transition of BMSCs to CAFs and a hitherto unknown function of GRP78 in the tumor microenvironment.
Collapse
Affiliation(s)
- Yanan Peng
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | | | | |
Collapse
|
17
|
Misra UK, Payne S, Pizzo SV. The Monomeric Receptor Binding Domain of Tetrameric α2-Macroglobulin Binds to Cell Surface GRP78 Triggering Equivalent Activation of Signaling Cascades. Biochemistry 2013; 52:4014-25. [DOI: 10.1021/bi400376s] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Uma Kant Misra
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710, United
States
| | - Sturgis Payne
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710, United
States
| | - Salvatore Vincent Pizzo
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710, United
States
| |
Collapse
|
18
|
Schönthal AH. Endoplasmic reticulum stress: its role in disease and novel prospects for therapy. SCIENTIFICA 2012; 2012:857516. [PMID: 24278747 PMCID: PMC3820435 DOI: 10.6064/2012/857516] [Citation(s) in RCA: 218] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 11/12/2012] [Indexed: 05/19/2023]
Abstract
The endoplasmic reticulum (ER) is a multifunctional organelle required for lipid biosynthesis, calcium storage, and protein folding and processing. A number of physiological and pathological conditions, as well as a variety of pharmacological agents, are able to disturb proper ER function and thereby cause ER stress, which severely impairs protein folding and therefore poses the risk of proteotoxicity. Specific triggers for ER stress include, for example, particular intracellular alterations (e.g., calcium or redox imbalances), certain microenvironmental conditions (e.g., hypoglycemia, hypoxia, and acidosis), high-fat and high-sugar diet, a variety of natural compounds (e.g., thapsigargin, tunicamycin, and geldanamycin), and several prescription drugs (e.g., bortezomib/Velcade, celecoxib/Celebrex, and nelfinavir/Viracept). The cell reacts to ER stress by initiating a defensive process, called the unfolded protein response (UPR), which is comprised of cellular mechanisms aimed at adaptation and safeguarding cellular survival or, in cases of excessively severe stress, at initiation of apoptosis and elimination of the faulty cell. In recent years, this dichotomic stress response system has been linked to several human diseases, and efforts are underway to develop approaches to exploit ER stress mechanisms for therapy. For example, obesity and type 2 diabetes have been linked to ER stress-induced failure of insulin-producing pancreatic beta cells, and current research efforts are aimed at developing drugs that ameliorate cellular stress and thereby protect beta cell function. Other studies seek to pharmacologically aggravate chronic ER stress in cancer cells in order to enhance apoptosis and achieve tumor cell death. In the following, these principles will be presented and discussed.
Collapse
Affiliation(s)
- Axel H. Schönthal
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 2011 Zonal Avenue, HMR-405, Los Angeles, CA 90033, USA
| |
Collapse
|
19
|
A murine monoclonal antibody directed against the carboxyl-terminal domain of GRP78 suppresses melanoma growth in mice. Melanoma Res 2012; 22:225-35. [PMID: 22495669 DOI: 10.1097/cmr.0b013e32835312fd] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The HSP70 family member GRP78 is a selective tumor marker upregulated on the surface of many tumor cell types, including melanoma, where it acts as a growth factor receptor-like protein. Receptor-recognized forms of the proteinase inhibitor α2-macroglobulin (α2M*) are the best-characterized ligands for GRP78, but in melanoma and other cancer patients, autoantibodies arise against the NH2-terminal domain of GRP78 that react with tumor cell-surface GRP78. This causes the activation of signaling cascades that are proproliferative and antiapoptotic. Antibodies directed against the COOH-terminal domain of GRP78, however, upregulate p53-mediated proapoptotic signaling, leading to cell death. Here, we describe the binding characteristics, cell signaling properties, and downstream cellular effects of three novel murine monoclonal antibodies. The NH2-terminal domain-reactive antibody, N88, mimics α2M* as a ligand and drives PI 3-kinase-dependent activation of Akt and the subsequent stimulation of cellular proliferation in vitro. The COOH-terminal domain-reactive antibody, C38, acts as an antagonist of both α2M* and N88, whereas another, C107, directly induces apoptosis in vitro. In a murine B16F1 melanoma flank tumor model, we demonstrate the acceleration of tumor growth by treatment with N88, whereas C107 significantly slowed tumor growth whether administered before (P<0.005) or after (P<0.05) tumor implantation.
Collapse
|
20
|
Ray R, de Ridder GG, Eu JP, Paton AW, Paton JC, Pizzo SV. The Escherichia coli subtilase cytotoxin A subunit specifically cleaves cell-surface GRP78 protein and abolishes COOH-terminal-dependent signaling. J Biol Chem 2012; 287:32755-69. [PMID: 22851173 DOI: 10.1074/jbc.m112.399808] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
GRP78, a molecular chaperone with critical endoplasmic reticulum functions, is aberrantly expressed on the surface of cancer cells, including prostate and melanoma. Here it functions as a pro-proliferative and anti-apoptotic signaling receptor via NH(2)-terminal domain ligation. Auto-antibodies to this domain may appear in cancer patient serum where they are a poor prognostic indicator. Conversely, GRP78 COOH-terminal domain ligation is pro-apoptotic and anti-proliferative. There is no method to disrupt cell-surface GRP78 without compromising the total GRP78 pool, making it difficult to study cell-surface GRP78 function. We studied six cell lines representing three cancer types. One cell line per group expresses high levels of cell-surface GRP78, and the other expresses low levels (human hepatoma: Hep3B and HepG2; human prostate cancer: PC3 and 1-LN; murine melanoma: B16F0 and B16F1). We investigated the effect of Escherichia coli subtilase cytoxin catalytic subunit (SubA) on GRP78. We report that SubA specifically cleaves cell-surface GRP78 on HepG2, 1-LN, and B16F1 cells without affecting intracellular GRP78. B16F0 cells (GRP78(low)) have lower amounts of cleaved cell-surface GRP78. SubA has no effect on Hep3B and PC3 cells. The predicted 28-kDa GRP78 COOH-terminal fragment is released into the culture medium by SubA treatment, and COOH-terminal domain signal transduction is abrogated, whereas pro-proliferative signaling mediated through NH(2)-terminal domain ligation is unaffected. These experiments clarify cell-surface GRP78 topology and demonstrate that the COOH-terminal domain is necessary for pro-apoptotic signal transduction occurring upon COOH-terminal antibody ligation. SubA is a powerful tool to specifically probe the functions of cell-surface GRP78.
Collapse
Affiliation(s)
- Rupa Ray
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | |
Collapse
|
21
|
Yang M, Yan Y, Fang M, Wan M, Wu X, Zhang X, Zhao T, Wei H, Song D, Wang L, Yu Y. MF59 formulated with CpG ODN as a potent adjuvant of recombinant HSP65-MUC1 for inducing anti-MUC1+ tumor immunity in mice. Int Immunopharmacol 2012; 13:408-16. [PMID: 22595192 PMCID: PMC7106219 DOI: 10.1016/j.intimp.2012.05.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 04/23/2012] [Accepted: 05/03/2012] [Indexed: 12/31/2022]
Abstract
MF59 is an oil-in-water emulsion adjuvant approved for influenza vaccines for human use in Europe. Due to its Th2 inducing properties, MF59 is seldom tested for cancer vaccines. In this study, MF59 formulated with a C-type CpG oligodeoxynucleotide (YW002) was tested for its Th1 adjuvant activity to induce immune responses to HSP65-MUC1, a recombinant fusion protein incorporating a mycobacterial heat shock protein (HSP65) and mucin 1, cell surface associated (MUC1) derived peptide. Combination of YW002 with MF59 (MF59-YW002) could confer a potent Th1 biasing property to the adjuvant, which enhanced the immunogenicity of HSP65-MUC1 to induce significantly higher levels of specific IgG2c, increased IFN-γ mRNA expression in splenocytes and the generation of antigen-specific cytotoxic T lymphocytes in mice. When prophylactically applied, MF59-YW002 adjuvant containing HSP65-MUC1 inhibited the growth of MUC1+ B16 melanoma and prolonged the survival of tumor-bearing mice. In contrast, adjuvant containing MF59 with HSP65-MUC1 in the absence of YW002, promoted the growth of MUC1+ B16 melanoma in mice. These results suggest that MF59 plus CpG oligodeoxynucleotide might be developed as an efficient adjuvant for tumor vaccines against melanoma, and possibly other tumors.
Collapse
Affiliation(s)
- Ming Yang
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
GRP78 Protein Expression in Ovarian Cancer Patients and Perspectives for a Drug-Targeting Approach. JOURNAL OF ONCOLOGY 2012; 2012:468615. [PMID: 22481929 PMCID: PMC3317113 DOI: 10.1155/2012/468615] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Revised: 01/03/2012] [Accepted: 01/12/2012] [Indexed: 11/18/2022]
Abstract
Glucose-regulated protein of 78 kD (GRP78) is a chaperone protein mainly located in the endoplasmic reticulum (ER). This protein is normally present at low levels in adult cells but its expression is triggered by ER stress including glucose deprivation and hypoxia. In tumor cells, it is overexpressed with fraction of protein found at the cell surface. This paper presents the physiology of GRP78 in the context of ovarian cancer and its potential use as drug delivery systems targeting ovarian cancer cell.
Collapse
|
23
|
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
|
24
|
Changes in oligosaccharide chains of autoantibodies to GRP78 expressed during progression of malignant melanoma stimulate melanoma cell growth and survival. Melanoma Res 2011; 21:323-34. [PMID: 21597391 DOI: 10.1097/cmr.0b013e3283471042] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A correlation between expression of the glucose-regulated protein of 78 kDa (GRP78) in malignant melanoma tumors and poor patient survival is well established. In this study, in addition to demonstrating the expression of GRP78 in tumor tissue, we investigated the immune response against GRP78 in a group of patients with different progression stages of malignant melanoma. Furthermore, we analyzed the glycosylation status of GRP78 immunoglobulin (Ig) G autoantibodies at these stages and evaluated their capacities to affect the protein B-dependent protein kinase signaling pathway and unfolded protein response signaling mechanisms, all known to promote malignant melanoma cell proliferation and survival. We found that progression of disease correlates not only with enhanced expression of GRP78 in the tumor but also with an increase in GRP78 autoantibody serum titers in these patients. We also found that the glycosylation status of anti-GRP78 IgG changes as the disease progresses. The anti-GRP78 IgG is abnormally glycosylated in the Fc region and asymmetrically glycosylated in the Fab region. We demonstrate that hyperglycosylated anti-GRP78 IgGs stimulate cell proliferation through protein B-dependent protein kinase signaling pathways. They also mimic the effects of α2-macroglobulin on the upregulation of GRP78 and X-box binding protein 1, activating transcription factor 6 α, and serine/threonine-protein kinase/endoribonuclease precursor α as endoplasmic reticulum stress biomarkers and show no effect on expression or activation of caspases 3, 9, or 12. In conclusion, the anti-GRP78 IgG autoantibodies downregulate apoptosis and activate unfolded protein response mechanisms, which are essential to promote melanoma cell growth and survival.
Collapse
|