1
|
Wang H, Zhou R, Xu C, Dai L, Hou R, Zheng L, Fu C, Shi G, Wang J, Li Y, Cen J, Xu X, Yu L, Li Y, Wang J, Du Q, Li Z. GRP78 Nanobody-Directed Immunotoxin Activates Innate Immunity Through STING Pathway to Synergize Tumor Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408086. [PMID: 40135833 PMCID: PMC12097070 DOI: 10.1002/advs.202408086] [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] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 02/27/2025] [Indexed: 03/27/2025]
Abstract
The lack of targetable antigens poses a significant challenge in developing effective cancer-targeted therapies. Cell surface translocation of endoplasmic reticulum (ER) chaperones, such as glucose-regulated protein 78 (GRP78), during malignancy, drug resistance, and ER stress induced by therapies, offers a promising pan-cancer target. To target GRP78, nanobody C5, identified from a phage library and exhibiting high affinity for human and mouse GRP78, is utilized to develop the Pseudomonas exotoxin (PE) immunotoxin C5-PE38. C5-PE38 induced ER stress, apoptosis and immunogenic cell death in targeted cells and showed antitumor efficacy against colorectal cancer and melanoma models without obvious toxicity. Mechanistically, transcriptome profiling showed that C5-PE38 reshaped the tumor immune microenvironment with enhanced innate and adaptive immune response and response to interferon beta. Moreover, C5-PE38-induced cell death could trans-activate STING pathway in dendritic cells and macrophages, promoting CD8+ T cell infiltration. It also sensitizes both primary and metastatic melanomas to anti-PD1 therapy, partly through STING activation. Overall, this study unveils a feasible GRP78 nanobody-directed therapy strategy for single or combinatorial cancer intervention. This work finds that C5-PE38-induced cell death stimulates STING-dependent cytosolic DNA release to promote antitumor immunity, a mechanism not previously reported for PE38, providing valuable insights for its clinical use.
Collapse
Affiliation(s)
- Huifang Wang
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
- Post‐doctoral Scientific Research Station of Basic MedicineJinan UniversityGuangzhou510632China
| | - Runhua Zhou
- Clinical Pharmacy CenterNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Chengchao Xu
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Lingyun Dai
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Rui Hou
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
- Harry Perkins Institute of Medical ResearchQEII Medical Centre and Centre for Medical ResearchThe University of Western AustraliaNedlandsWA6009Australia
| | - Liuhai Zheng
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Chunjin Fu
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Guangwei Shi
- Department of Neurosurgery & Medical Research CenterShunde HospitalSouthern Medical University (The First People's Hospital of Shunde Foshan)Guangzhou510515China
| | - Jingwei Wang
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
- Clinical Pharmacy CenterNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yang Li
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Jinpeng Cen
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Xiaolong Xu
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Le Yu
- Clinical Pharmacy CenterNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
- School of Traditional Chinese Medicine and School of Pharmaceutical SciencesGuangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yilei Li
- Clinical Pharmacy CenterNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Jigang Wang
- School of Traditional Chinese Medicine and School of Pharmaceutical SciencesGuangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di HerbsArtemisinin Research CenterInstitute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijing100700China
- State Key Laboratory of Antiviral DrugsSchool of PharmacyHenan UniversityKaifeng475004China
| | - Qingfeng Du
- School of Traditional Chinese Medicine and School of Pharmaceutical SciencesGuangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Zhijie Li
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| |
Collapse
|
2
|
Su S, Wu L, Huang C, He C, Wang L, Li W, Liu W, Liu L. ATF6 activation promotes tumorigenesis and drug resistance in diffuse large B-cell lymphoma (DLBCL) by regulating the mTOR/S6K signaling pathway. Discov Oncol 2025; 16:499. [PMID: 40205285 PMCID: PMC11982007 DOI: 10.1007/s12672-025-02264-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Abstract
This study analyzes the expression and functional role of activating transcription factor 6 (ATF6) in diffuse large B-cell lymphoma (DLBCL) and its effects on disease progression. ATF6, a core component of the unfolded protein response (UPR) pathway, participates in many cellular activities and notably contributes to tumorigenesis. Through a combination of techniques, including immunohistochemistry (IHC) staining to assess ATF6 and pS6K levels, siRNA-mediated ATF6 knockdown, cytotoxicity assays, flow cytometry, quantitative real-time PCR (qRT-PCR), as well as Western blotting, this study clarified the functioning mechanisms of ATF6 in DLBCL and its potential clinical relevance. Further exploration of ATF6's involvement in the mTORC1 pathway was achieved through RNA sequencing (RNA-seq) and gene set enrichment analysis (GSEA). Our findings demonstrate that ATF6 expression is upregulated in DLBCL and linked to poor prognosis, particularly in people aged over 60 with Ann Arbor stage III-IV disease, B symptoms, non-GCB subtype, an international prognostic index (IPI) score greater than 2, and extranodal involvement. Notably, the ATF6 inhibitor ceapinA7 was shown to suppress ATF6 and mTORC1 activation, leading to less cell proliferation and the induction of apoptosis in DLBCL cells. Additionally, ceapinA7 increased the sensitivity of DLBCL cells to adriamycin. The foregoing results underscore the critical role of ATF6 in DLBCL and lay a theoretical and experimental foundation for future targeted therapies and drug development aimed at improving treatment outcomes for DLBCL.
Collapse
Affiliation(s)
- Shuang Su
- Department of Hematology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Lili Wu
- Department of Hematology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Chen Huang
- Department of Hematology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Cuiying He
- Department of Hematology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Lianjing Wang
- Department of Hematology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Weijing Li
- Department of Hematology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Wei Liu
- Department of Hematology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Lihong Liu
- Department of Hematology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China.
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China.
| |
Collapse
|
3
|
Hilan G, Daniel G, Collak F, Sabatino D, Willmore W. Cancer-Targeting Peptides Functionalized With Polyarginine Enables GRP78-Dependent Cell Uptake and siRNA Delivery Within the DU145 Prostate Cancer Cells. J Pept Sci 2025; 31:e70007. [PMID: 39967318 PMCID: PMC11836551 DOI: 10.1002/psc.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 02/03/2025] [Accepted: 02/06/2025] [Indexed: 02/20/2025]
Abstract
This study investigated a peptide-based GRP78-targeting strategy for short-interfering (si) RNA delivery in cancer cells. Synthetic fluorescein-labeled amphiphilic peptides composed of the hydrophobic cell surface (cs) GRP78-targeting and hydrophilic, polycationic arginine-rich cell penetrating peptides demonstrated GRP78-dependent cell uptake in the DU145 prostate cancer cells, and to a lesser extent in the non-cancerous human lung fibroblast WI-38 cell line. Mechanistic studies revealed energy-dependent GRP78 receptor-mediated endocytosis of the GRP78-targeting peptide with polyarginine (W1-R9). The cytosolic accumulation of this peptide underscored its potential utility in siRNA delivery. Peptide:siRNA complexes formed stably condensed nanoparticles, with calcium functioning as an ionic stabilizer and additive promoting endosomal siRNA escape for RNA interference (RNAi) activity. Preliminary peptide-based siRNA transfections in the DU145 cells demonstrated that GRP78 knockdown led to an interplay in between pro-survival and cell death outcomes under ER stress induction. Thus, the GRP78-targeting polyarginine peptides enables efficient cell uptake for specific siRNA delivery in the DU145 cells. This class of bio-active synthetic peptides is important for the investigation of cancer biology, leading to the innovation of cancer-targeted gene delivery and therapy approaches.
Collapse
Affiliation(s)
- George Hilan
- Department of BiologyCarleton UniversityOttawaONCanada
| | - Grace Daniel
- Department of ChemistryCarleton UniversityOttawaONCanada
- Institute of BiochemistryCarleton UniversityOttawaONCanada
| | - Filiz Collak
- Department of BiologyCarleton UniversityOttawaONCanada
- Department of ChemistryCarleton UniversityOttawaONCanada
| | - David Sabatino
- Department of ChemistryCarleton UniversityOttawaONCanada
- Institute of BiochemistryCarleton UniversityOttawaONCanada
| | - William G. Willmore
- Department of BiologyCarleton UniversityOttawaONCanada
- Department of ChemistryCarleton UniversityOttawaONCanada
- Institute of BiochemistryCarleton UniversityOttawaONCanada
| |
Collapse
|
4
|
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
|
5
|
Liu M, Yuan M, Ma Y, Wang J, Cheng X, Shi Y, Shang J, He M, Bai L, Du L, Tang H. Wild-Type and rtA181T/sW172* Mutant Strains of Hepatitis B Virus Drive Hepatocarcinogenesis via Distinct GRP78-Mediated ER Stress Pathways. J Med Virol 2025; 97:e70151. [PMID: 39749680 DOI: 10.1002/jmv.70151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/14/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025]
Abstract
Glucose-regulated protein 78 kDa (GRP78), a key marker of endoplasmic reticulum stress (ERS), is upregulated in hepatocellular carcinoma (HCC) tissues, but its role in hepatitis B virus (HBV)-induced tumorigenesis remains unclear. This study aimed to investigate the contribution of GRP78 to HBV-associated tumor development and explore the ERS pathways involved. The results showed that increased GRP78 expression in patients with HBV-related HCC was associated with a poor prognosis within the first 2 years following diagnosis. Furthermore, using wild-type HBV strain and the oncogenic HBV rtA181T/sW172* mutant, this study demonstrated that the HBV-induced GRP78 expression correlated with elevated reactive oxygen species (ROS) levels. Moreover, GRP78 expression enhanced hepatocyte proliferation and resistance to apoptosis. In wild-type HBV-infected hepatocytes, GRP78 suppressed apoptosis by inhibiting the PERK/p38 pathway. In contrast, the HBV rtA181T/sW172* mutation led to increased GRP78 expression and inhibition of cell apoptosis through activation of the IRE-1α/XBP1/BCL-2 pathway. In conclusion, GRP78 plays a pivotal role in HBV-induced hepatocarcinogenesis by modulating distinct ERS pathways. Targeting these pathways may aid in the therapeutic management of HBV-associated hepatocarcinogenesis.
Collapse
Affiliation(s)
- Miao Liu
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Man Yuan
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Yuanji Ma
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Jiayi Wang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Xing Cheng
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Ying Shi
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jin Shang
- Liver Transplantation Center and HBP Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Min He
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Lang Bai
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Lingyao Du
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Shen X, Ma Y, Luo H, Abdullah R, Pan Y, Zhang Y, Zhong C, Zhang B, Zhang G. Peptide Aptamer-Paclitaxel Conjugates for Tumor Targeted Therapy. Pharmaceutics 2024; 17:40. [PMID: 39861688 PMCID: PMC11768741 DOI: 10.3390/pharmaceutics17010040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/23/2024] [Accepted: 12/26/2024] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Traditional paclitaxel therapy often results in significant side effects due to its non-specific targeting of cancer cells. Peptide aptamer-paclitaxel conjugates present a promising alternative by covalently attaching paclitaxel to a versatile peptide aptamer via a linker. Compared to antibody-paclitaxel conjugates, peptide aptamer-paclitaxel conjugates offer several advantages, including a smaller size, lower immunogenicity, improved tissue penetration, and easier engineering. Methods: This review provides an in-depth analysis of the multifunctional peptide aptamers in these conjugates, emphasizing their structural features, therapeutic efficacy, and challenges in clinical applications. Results: This analysis highlights the potential of peptide aptamer-paclitaxel conjugates as a novel and effective approach for targeted cancer therapy. By harnessing the unique properties of peptide aptamers, these conjugates demonstrate significant promise in improving drug delivery efficiency while reducing the adverse effects associated with traditional paclitaxel therapy. Conclusions: The incorporation of peptide aptamers into paclitaxel conjugates offers a promising pathway for developing more efficient and targeted cancer therapies. However, further research and clinical studies are essential to fully unlock the therapeutic potential of these innovative conjugates and enhance patient outcomes.
Collapse
Affiliation(s)
- Xinyang Shen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuan Ma
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China; (Y.M.)
- Law Sau Fai Institute for Advancing Translational Medicine in Bone &Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Hang Luo
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China; (Y.M.)
| | - Razack Abdullah
- Law Sau Fai Institute for Advancing Translational Medicine in Bone &Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Yufei Pan
- Law Sau Fai Institute for Advancing Translational Medicine in Bone &Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Yihao Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China; (Y.M.)
| | - Chuanxin Zhong
- Law Sau Fai Institute for Advancing Translational Medicine in Bone &Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Baoting Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China; (Y.M.)
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone &Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| |
Collapse
|
7
|
Chen YJN, Shi RC, Xiang YC, Fan L, Tang H, He G, Zhou M, Feng XZ, Tan JD, Huang P, Ye X, Zhao K, Fu WY, Li LL, Bian XT, Chen H, Wang F, Wang T, Zhang CK, Zhou BH, Chen W, Liang TT, Lv JT, Kang X, Shi YX, Kim E, Qin YH, Hettinghouse A, Wang KD, Zhao XL, Yang MY, Tang YZ, Piao HL, Guo L, Liu CJ, Miao HM, Tang KL. Malate initiates a proton-sensing pathway essential for pH regulation of inflammation. Signal Transduct Target Ther 2024; 9:367. [PMID: 39737965 PMCID: PMC11683149 DOI: 10.1038/s41392-024-02076-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 01/01/2025] Open
Abstract
Metabolites can double as a signaling modality that initiates physiological adaptations. Metabolism, a chemical language encoding biological information, has been recognized as a powerful principle directing inflammatory responses. Cytosolic pH is a regulator of inflammatory response in macrophages. Here, we found that L-malate exerts anti-inflammatory effect via BiP-IRF2BP2 signaling, which is a sensor of cytosolic pH in macrophages. First, L-malate, a TCA intermediate upregulated in pro-inflammatory macrophages, was identified as a potent anti-inflammatory metabolite through initial screening. Subsequent screening with DARTS and MS led to the isolation of L-malate-BiP binding. Further screening through protein‒protein interaction microarrays identified a L-malate-restrained coupling of BiP with IRF2BP2, a known anti-inflammatory protein. Interestingly, pH reduction, which promotes carboxyl protonation of L-malate, facilitates L-malate and carboxylate analogues such as succinate to bind BiP, and disrupt BiP-IRF2BP2 interaction in a carboxyl-dependent manner. Both L-malate and acidification inhibit BiP-IRF2BP2 interaction, and protect IRF2BP2 from BiP-driven degradation in macrophages. Furthermore, both in vitro and in vivo, BiP-IRF2BP2 signal is required for effects of both L-malate and pH on inflammatory responses. These findings reveal a previously unrecognized, proton/carboxylate dual sensing pathway wherein pH and L-malate regulate inflammatory responses, indicating the role of certain carboxylate metabolites as adaptors in the proton biosensing by interactions between macromolecules.
Collapse
Affiliation(s)
- Yu-Jia-Nan Chen
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China.
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA.
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases & Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China.
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China.
| | - Rong-Chen Shi
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
| | - Yuan-Cai Xiang
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Li Fan
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases & Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Hong Tang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Gang He
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Mei Zhou
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Xin-Zhe Feng
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
| | - Jin-Dong Tan
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Pan Huang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Xiao Ye
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Kun Zhao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
| | - Wen-Yu Fu
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
- Department of Orthopedics and Rehabilitations, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Liu-Li Li
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
| | - Xu-Ting Bian
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Huan Chen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Feng Wang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Teng Wang
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
| | - Chen-Ke Zhang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Bing-Hua Zhou
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Wan Chen
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Tao-Tao Liang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Jing-Tong Lv
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Xia Kang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
| | - You-Xing Shi
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Ellen Kim
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
| | - Yin-Hua Qin
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Army Medical University, Chongqing, 400038, China
| | - Aubryanna Hettinghouse
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
| | - Kai-di Wang
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
- Department of Medical Experimental Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266000, China
| | - Xiang-Li Zhao
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
- Department of Orthopedics and Rehabilitations, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Ming-Yu Yang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Yu-Zhen Tang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Hai-Long Piao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Lin Guo
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| | - Chuan-Ju Liu
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA.
- Department of Orthopedics and Rehabilitations, Yale University School of Medicine, New Haven, CT, 06519, USA.
| | - Hong-Ming Miao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China.
- Jinfeng Laboratory, Chongqing, 401329, China.
| | - Kang-Lai Tang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
8
|
Robinson RM, Reyes L, Christopher BN, Duncan RM, Burge RA, Siegel J, Nasarre P, Wang P, O'Bryan JP, Hobbs GA, Klauber-DeMore N, Dolloff NG. A High-Affinity Monoclonal Antibody Against the Pancreatic Ductal Adenocarcinoma Target, Anterior Gradient-2 (AGR2/PDIA17). Antibodies (Basel) 2024; 13:101. [PMID: 39727484 DOI: 10.3390/antib13040101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Anterior Gradient-2 (AGR2/PDIA17) is a member of the protein disulfide isomerase (PDI) family of oxidoreductases. AGR2 is up-regulated in several solid tumors, including pancreatic ductal adenocarcinoma (PDAC). Given the dire need for new therapeutic options for PDAC patients, we investigated the expression and function of AGR2 in PDAC and developed a novel series of affinity-matured AGR2-specific single-chain variable fragments (scFvs) and monoclonal antibodies. RESULTS We found that AGR2 was expressed in approximately 90% of PDAC but not normal pancreas biopsies, and the level of AGR2 expression correlated with increasing disease stage. AGR2 expression was inversely related to SMAD4 status in PDAC and colorectal cancer cell models and was secreted from cells into their media. In normal tissues, a high density of AGR2 was detected in the epithelium of cells in the digestive tract but was lacking in most other normal tissue systems. The addition of recombinant AGR2 to cell culture and genetic overexpression of AGR2 increased the adhesion, motility, and invasiveness of both human and mouse PDAC cells. Human phage display library screening led to the discovery of multiple AGR2-specific scFv clones that were affinity-matured to produce monoclonal antibody (MAb) clones with low picomolar binding affinity (S31R/A53F/Y). These high-affinity MAbs inhibited AGR2-mediated cell adhesion, migration, and binding to LYPD3, which is a putative cell surface binding partner of AGR2. CONCLUSIONS Our study provides novel, high-affinity, fully human, anti-AGR2 MAbs that neutralize the pro-tumor effects of extracellular AGR2 in PDAC.
Collapse
Affiliation(s)
- Reeder M Robinson
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Leticia Reyes
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Benjamin N Christopher
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ravyn M Duncan
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Rachel A Burge
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Julie Siegel
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Patrick Nasarre
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | - John P O'Bryan
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - G Aaron Hobbs
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Nancy Klauber-DeMore
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
- MUSC Hollings Cancer Center, Charleston, SC 29425, USA
| | - Nathan G Dolloff
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
- MUSC Hollings Cancer Center, Charleston, SC 29425, USA
- Zucker Institute for Innovation Commercialization, Charleston, SC 29425, USA
| |
Collapse
|
9
|
Cifric S, Turi M, Folino P, Clericuzio C, Barello F, Maciel T, Anderson KC, Gulla A. DAMPening Tumor Immune Escape: The Role of Endoplasmic Reticulum Chaperones in Immunogenic Chemotherapy. Antioxid Redox Signal 2024; 41:661-674. [PMID: 38366728 DOI: 10.1089/ars.2024.0558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/18/2024]
Abstract
Significance: Preclinical and clinical research in the past two decades has redefined the mechanism of action of some chemotherapeutics that are able to activate the immune system against cancer when cell death is perceived by the immune cells. This immunogenic cell death (ICD) activates antigen-presenting cells (APCs) and T cells to induce immune-mediated tumor clearance. One of the key requirements to achieve this effect is the externalization of the damage-associated molecular patterns (DAMPs), molecules released or exposed by cancer cells during ICD that increase the visibility of the cancer cells by the immune system. Recent Advances: In this review, we focus on the role of calreticulin (CRT) and other endoplasmic reticulum (ER) chaperones, such as the heat-shock proteins (HSPs) and the protein disulfide isomerases (PDIs), as surface-exposed DAMPs. Once exposed on the cell membrane, these proteins shift their role from that of ER chaperone and regulator of Ca2+ and protein homeostasis to act as an immunogenic signal for APCs, driving dendritic cell (DC)-mediated phagocytosis and T-mediated antitumor response. Critical Issues: However, cancer cells exploit several mechanisms of resistance to immune attack, including subverting the exposure of ER chaperones on their surface to avoid immune recognition. Future Directions: Overcoming these mechanisms of resistance represents a potential therapeutic opportunity to improve cancer treatment effectiveness and patient outcomes.
Collapse
Affiliation(s)
- Selma Cifric
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Marcello Turi
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Pietro Folino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Cole Clericuzio
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Tallya Maciel
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Kenneth C Anderson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
10
|
Ha DP, Shin WJ, Liu Z, Doche ME, Lau R, Leli NM, Conn CS, Russo M, Lorenzato A, Koumenis C, Yu M, Mumenthaler SM, Lee AS. Targeting stress induction of GRP78 by cardiac glycoside oleandrin dually suppresses cancer and COVID-19. Cell Biosci 2024; 14:115. [PMID: 39238058 PMCID: PMC11378597 DOI: 10.1186/s13578-024-01297-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Despite recent therapeutic advances, combating cancer resistance remains a formidable challenge. The 78-kilodalton glucose-regulated protein (GRP78), a key stress-inducible endoplasmic reticulum (ER) chaperone, plays a crucial role in both cancer cell survival and stress adaptation. GRP78 is also upregulated during SARS-CoV-2 infection and acts as a critical host factor. Recently, we discovered cardiac glycosides (CGs) as novel suppressors of GRP78 stress induction through a high-throughput screen of clinically relevant compound libraries. This study aims to test the possibility that agents capable of blocking stress induction of GRP78 could dually suppress cancer and COVID-19. RESULTS Here we report that oleandrin (OLN), is the most potent among the CGs in inhibiting acute stress induction of total GRP78, which also results in reduced cell surface and nuclear forms of GRP78 in stressed cells. The inhibition of stress induction of GRP78 is at the post-transcriptional level, independent of protein degradation and autophagy and may involve translational control as OLN blocks stress-induced loading of ribosomes onto GRP78 mRNAs. Moreover, the human Na+/K+-ATPase α3 isoform is critical for OLN suppression of GRP78 stress induction. OLN, in nanomolar range, enhances apoptosis, sensitizes colorectal cancer cells to chemotherapeutic agents, and reduces the viability of patient-derived colon cancer organoids. Likewise, OLN, suppresses GRP78 expression and impedes tumor growth in an orthotopic breast cancer xenograft model. Furthermore, OLN blocks infection by SARS-CoV-2 and its variants and enhances existing anti-viral therapies. Notably, GRP78 overexpression mitigates OLN-mediated cancer cell apoptotic onset and suppression of virus release. CONCLUSION Our findings validate GRP78 as a target of OLN anti-cancer and anti-viral activities. These proof-of-principle studies support further investigation of OLN as a readily accessible compound to dually combat cancer and COVID-19.
Collapse
Affiliation(s)
- Dat P Ha
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Woo-Jin Shin
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, 34987, USA
- Department of Cancer Biology, Infection Biology Program, and Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44106, USA
| | - Ze Liu
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Michael E Doche
- Ellison Institute of Technology, Los Angeles, CA, 90064, USA
| | - Roy Lau
- Ellison Institute of Technology, Los Angeles, CA, 90064, USA
| | - Nektaria Maria Leli
- Department of Radiation Oncology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Crystal S Conn
- Department of Radiation Oncology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mariangela Russo
- Dipartimento di Oncologia, Molecular Biotechnology Center, Università di Torino, Turin, Italy
| | - Annalisa Lorenzato
- Dipartimento di Oncologia, Molecular Biotechnology Center, Università di Torino, Turin, Italy
| | - Constantinos Koumenis
- Department of Radiation Oncology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Min Yu
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Shannon M Mumenthaler
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Ellison Institute of Technology, Los Angeles, CA, 90064, USA
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Amy S Lee
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
11
|
Li W, Liu J, Yu T, Lu F, Miao Q, Meng X, Xiao W, Yang H, Zhang X. ZDHHC9-mediated Bip/GRP78 S-palmitoylation inhibits unfolded protein response and promotes bladder cancer progression. Cancer Lett 2024; 598:217118. [PMID: 39002690 DOI: 10.1016/j.canlet.2024.217118] [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: 01/09/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 07/15/2024]
Abstract
Recent studies have highlighted palmitoylation, a novel protein post-translational modification, as a key player in various signaling pathways that contribute to tumorigenesis and drug resistance. Despite this, its role in bladder cancer (BCa) development remains inadequately understood. In this study, ZDHHC9 emerged as a significantly upregulated oncogene in BCa. Functionally, ZDHHC9 knockdown markedly inhibited tumor proliferation, promoted tumor cell apoptosis, and enhanced the efficacy of gemcitabine (GEM) and cisplatin (CDDP). Mechanistically, SP1 was found to transcriptionally activate ZDHHC9 expression. ZDHHC9 subsequently bound to and palmitoylated the Bip protein at cysteine 420 (Cys420), thereby inhibiting the unfolded protein response (UPR). This palmitoylation at Cys420 enhanced Bip's protein stability and preserved its localization within the endoplasmic reticulum (ER). ZDHHC9 might become a novel therapeutic target for BCa and could also contribute to combination therapy with GEM and CDDP.
Collapse
Affiliation(s)
- Weiquan Li
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China
| | - Jingchong Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China
| | - Tiexi Yu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China
| | - Feiyi Lu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China
| | - Qi Miao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China
| | - Xiangui Meng
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China.
| | - Wen Xiao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China.
| | - Hongmei Yang
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China.
| |
Collapse
|
12
|
Johnson N, Pattinson C, Burgoyne K, Hijazi K, Houssen WE, Milne BF. SARS-CoV-2 Spike Protein-Derived Cyclic Peptides as Modulators of Spike Interaction with GRP78. Chembiochem 2024; 25:e202300789. [PMID: 38613462 PMCID: PMC11497264 DOI: 10.1002/cbic.202300789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 04/12/2024] [Accepted: 04/12/2024] [Indexed: 04/15/2024]
Abstract
The human glucose-regulated protein GRP78 is a human chaperone that translocactes to the cell surface when cells are under stress. Theoretical studies suggested it could be involved in SARS-CoV-2 virus entry to cells. In this work, we used in vitro surface plasmon resonance-based assays to show that human GRP78 indeed binds to SARS-CoV-2 spike protein. We have designed and synthesised cyclic peptides based on the loop structure of amino acids 480-488 of the SARS-CoV-2 spike protein S1 domain from the Wuhan and Omicron variants and showed that both peptides bind to GRP78. Consistent with the greater infectiousness of the Omicron variant, the Omicron-derived peptide displays slower dissociation from the target protein. Both peptides significantly inhibit the binding of wild-type S1 protein to the human protein GRP78 suggesting that further development of these cyclic peptide motifs may provide a viable route to novel anti-SARS-CoV-2 agents.
Collapse
Affiliation(s)
- Nicholas Johnson
- Institute of Medical SciencesUniversity of AberdeenAshgrove Road WestAberdeenAB25 2ZDUK
| | - Craig Pattinson
- School of MedicineMedical Sciences and NutritionUniversity of AberdeenAberdeenAB25 2ZDUK
| | - Kate Burgoyne
- School of MedicineMedical Sciences and NutritionUniversity of AberdeenAberdeenAB25 2ZDUK
| | - Karolin Hijazi
- School of MedicineMedical Sciences and NutritionUniversity of AberdeenAberdeenAB25 2ZDUK
| | - Wael E. Houssen
- Institute of Medical SciencesUniversity of AberdeenAshgrove Road WestAberdeenAB25 2ZDUK
- Department of ChemistryUniversity of AberdeenMeston WalkAberdeenAB24 3UEUK
| | - Bruce F. Milne
- Department of ChemistryUniversity of AberdeenMeston WalkAberdeenAB24 3UEUK
- CFisUCDepartment of PhysicsUniversity of CoimbraRua Larga3004-516CoimbraPortugal
| |
Collapse
|
13
|
Öz-Arslan D, Yavuz M, Kan B. Exploring orphan GPCRs in neurodegenerative diseases. Front Pharmacol 2024; 15:1394516. [PMID: 38895631 PMCID: PMC11183337 DOI: 10.3389/fphar.2024.1394516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/15/2024] [Indexed: 06/21/2024] Open
Abstract
Neurodegenerative disorders represent a significant and growing health burden worldwide. Unfortunately, limited therapeutic options are currently available despite ongoing efforts. Over the past decades, research efforts have increasingly focused on understanding the molecular mechanisms underlying these devastating conditions. Orphan receptors, a class of receptors with no known endogenous ligands, emerge as promising druggable targets for diverse diseases. This review aims to direct attention to a subgroup of orphan GPCRs, in particular class A orphans that have roles in neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and Multiple sclerosis. We highlight the diverse roles orphan receptors play in regulating critical cellular processes such as synaptic transmission, neuronal survival and neuro-inflammation. Moreover, we discuss the therapeutic potential of targeting orphan receptors for the treatment of neurodegenerative disorders, emphasizing recent advances in drug discovery and preclinical studies. Finally, we outline future directions and challenges in orphan receptor research.
Collapse
Affiliation(s)
- Devrim Öz-Arslan
- Department of Biophysics, Acibadem MAA University, School of Medicine, Istanbul, Türkiye
- Department of Neurosciences, Acibadem MAA University, Institute of Health Sciences, İstanbul, Türkiye
| | - Melis Yavuz
- Department of Neurosciences, Acibadem MAA University, Institute of Health Sciences, İstanbul, Türkiye
- Department of Pharmacology, Acibadem MAA University, School of Pharmacy, Istanbul, Türkiye
| | - Beki Kan
- Department of Biophysics, Acibadem MAA University, School of Medicine, Istanbul, Türkiye
- Department of Neurosciences, Acibadem MAA University, Institute of Health Sciences, İstanbul, Türkiye
| |
Collapse
|
14
|
Zhang Y, Gu X, Huang L, Yang Y, He J. Enhancing precision medicine: Bispecific antibody-mediated targeted delivery of lipid nanoparticles for potential cancer therapy. Int J Pharm 2024; 654:123990. [PMID: 38467208 DOI: 10.1016/j.ijpharm.2024.123990] [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: 10/31/2023] [Revised: 03/07/2024] [Accepted: 03/09/2024] [Indexed: 03/13/2024]
Abstract
The precise delivery of therapeutic agents to specific cell populations, including cancer cells, remains a target in modern medicine, to enhance treatment efficacy, while minimizing unintended side effects. This study presents a strategy utilizing bispecific antibodies for the targeted delivery of nucleic acid drugs to the surface of glucose-regulated protein 78 (GRP78)-overexpressing cancer cells. Strong binding affinity of the bispecific antibodies to GRP78-overexpressing cancer cells, including HEPG2 cells, confirmed the tumor-targeting potential of this platform. Functional analyses demonstrated the role of the bispecific antibodies in enhancing lipid nanoparticle (LNP) uptake, causing increased gene expression levels of nucleic acid drugs loaded within LNPs. In vivo imaging confirmed the potency of the bispecific-antibody-modified LNPs in delivering nucleic acid drugs to tumors and sustaining therapeutic expression levels. In vivo therapy results indicated that the bispecific antibodies improved the antitumor activity of PE38-loaded LNPs in tumors overexpressing surface GRP78. This study pioneered a bispecific-antibody-centered platform for the targeted delivery of nucleic acid drugs. The robust antigen-antibody binding affinity, tumor-selective interactions, enhanced cellular uptake, and proficient gene expression promise to advance precision therapeutics in oncology. Continued refinement and translation of this drug delivery strategy are important to unlock its full clinical potential.
Collapse
Affiliation(s)
- Yue Zhang
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, PR China
| | - Xiaoyan Gu
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, PR China
| | - Lili Huang
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, PR China
| | - Yani Yang
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, PR China
| | - Jun He
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, PR China.
| |
Collapse
|
15
|
Daniel G, Hilan G, Ploeg L, Sabatino D. Self-assembly of amphiphilic helical-coiled peptide nanofibers and inhibition of fibril formation with curcumin. Bioorg Med Chem Lett 2024; 102:129682. [PMID: 38432287 DOI: 10.1016/j.bmcl.2024.129682] [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: 12/18/2023] [Revised: 02/06/2024] [Accepted: 02/28/2024] [Indexed: 03/05/2024]
Abstract
Amphiphilic peptide sequences are conducive to secondary structures that self-assemble into higher-ordered peptide nanostructures. A select set of amphiphilic polycationic peptides displayed stable helical-coiled structures that self-assembled into peptide nanofibers. The progression of peptide fibril formation revealed short protofibrils that extended into thin filaments and into an entangled network of nanofibers over an extended (5 days) incubation period. Ligand binding with 8-anilinonaphthalene-1-sulfonic acid (ANS) and Congo Red (CR) confirmed the amphiphilic helical-coiled peptide structure assembly into nanofibers, whereas curcumin treatment led to inhibition of fibril formation. Considering the vast repertoire of fibrous biomaterials and peptide or protein (mis)folding contingent on fibril formation, this work relates the molecular interplay in between sequence composition, structural folding and the ligand binding events impacting peptide self-assembly into nanofibers.
Collapse
Affiliation(s)
- Grace Daniel
- Department of Chemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada; Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| | - George Hilan
- Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| | - Lisa Ploeg
- Department of Chemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| | - David Sabatino
- Department of Chemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada; Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada.
| |
Collapse
|
16
|
Yuan Y, Wang W, Luo J, Tang C, Zheng Y, Yu J, Xu H, Zhu M, Hang T, Wang H, Diao X. Metabolite characterisation of the peptide-drug conjugate LN005 in liver S9s by UHPLC-Orbitrap-HRMS. Xenobiotica 2024; 54:1-9. [PMID: 38044881 DOI: 10.1080/00498254.2023.2289635] [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: 09/19/2023] [Accepted: 11/27/2023] [Indexed: 12/05/2023]
Abstract
LN005 is a peptide-drug conjugate (PDC) targeting glucose-regulated protein 78 (GRP78) to treat several types of cancer, such as breast, colon, and prostate cancer.As a new drug modality, understanding its metabolism and elimination pathways will help us to have a whole picture of it. Currently, there are no metabolic studies on LN005; therefore, this study aimed to investigate the metabolism of LN005, clarify its metabolic profile in the liver S9s of different species, and identify the major metabolic pathways and differences between species.The incubation samples were measured by ultra-high performance liquid chromatography combined with orbitrap tandem mass spectrometry (UHPLC-Orbitrap-HRMS).The results showed that LN005 was metabolised by liver S9s, and four metabolites were identified. The main metabolic pathway of LN005 in liver S9s was oxidative deamination to ketone or hydrolysis. Similar metabolic profiles were observed in mouse, rat, dog, monkey, and human liver S9s, indicating no differences between these four animal species and humans.This study provides information for the structural modification and optimisation of LN005 and affords a reference for subsequent animal experiments and human metabolism of other PDCs.
Collapse
Affiliation(s)
- Yali Yuan
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Weiqiang Wang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Jing Luo
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
- Department of Preparation, Shanghai Whittlong Pharmaceutical Institute Co., Ltd, Shanghai, China
| | - Chongzhuang Tang
- Department of Biotransformation, XenoFinder Co., Ltd, Suzhou, China
| | - Yuandong Zheng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jinghua Yu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Honghong Xu
- Department of Preparation, Shanghai Whittlong Pharmaceutical Institute Co., Ltd, Shanghai, China
| | - Mingshe Zhu
- Department of Biotransformation, XenoFinder Co., Ltd, Suzhou, China
| | - Taijun Hang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Hao Wang
- National Pharmaceutical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Xingxing Diao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
17
|
Guo W, Wang M, Yang Z, Liu D, Ma B, Zhao Y, Chen Y, Hu Y. Recent advances in small molecule and peptide inhibitors of glucose-regulated protein 78 for cancer therapy. Eur J Med Chem 2023; 261:115792. [PMID: 37690265 DOI: 10.1016/j.ejmech.2023.115792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/18/2023] [Accepted: 09/02/2023] [Indexed: 09/12/2023]
Abstract
Glucose-regulated protein 78 (GRP78) is one of key endoplasmic reticulum (ER) chaperone proteins that regulates the unfolded protein response (UPR) to maintain ER homeostasis. As a core factor in the regulation of the UPR, GRP78 takes a critical part in the cellular processes required for tumorigenesis, such as proliferation, metastasis, anti-apoptosis, immune escape and chemoresistance. Overexpression of GRP78 is closely correlated with tumorigenesis and poor prognosis in various malignant tumors. Targeting GRP78 is regarded as a potentially promising therapeutic strategy for cancer therapy. Although none of the GRP78 inhibitors have been approved to date, there have been several studies of GRP78 inhibitors. Herein, we comprehensively review the structure, physiological functions of GRP78 and the recent progress of GRP78 inhibitors, and discuss the structures, in vitro and in vivo efficacies, and merits and demerits of these inhibitors to inspire further research. Additionally, the feasibility of GRP78-targeting proteolysis-targeting chimeras (PROTACs), disrupting GRP78 cochaperone interactions, or covalent inhibition are also discussed as novel strategies for drugs discovery targeting GRP78, with the hope that these strategies can provide new opportunities for targeted GRP78 antitumor therapy.
Collapse
Affiliation(s)
- Weikai Guo
- The Jointed National Laboratory of Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, 475004, China
| | - Manjie Wang
- The Jointed National Laboratory of Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, 475004, China
| | - Zhengfan Yang
- The Jointed National Laboratory of Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, 475004, China
| | - Danyang Liu
- The Jointed National Laboratory of Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, 475004, China
| | - Borui Ma
- The Jointed National Laboratory of Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, 475004, China
| | - Yanqun Zhao
- The Jointed National Laboratory of Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, 475004, China
| | - Yihua Chen
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Yanzhong Hu
- The Jointed National Laboratory of Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
18
|
Bai Y, Wang W, Cheng Y, Yang Y. Research progress on the GRP78 gene in the diagnosis, treatment and immunity of cervical cancer. Eur J Med Res 2023; 28:447. [PMID: 37858217 PMCID: PMC10588224 DOI: 10.1186/s40001-023-01241-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 07/22/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND GRP78 is a molecular chaperone protein in the endoplasmic reticulum that is involved in protein assembly and quality control, and it participates in ER stress regulation of endoplasmic reticulum stress pathways. Studies have confirmed that GRP78 gene is highly expressed in a variety of tumors and is involved in different biological functions. PURPOSE The present review highlights the involvement of the GRP78 gene in regulating the development of cervical cancer by promoting the proliferation and invasion of cervical cancer cells as well as by inhibiting apoptosis and promoting the Warburg effect. High expression of GRP78 is positively correlated with chemotherapy resistance in cervical cancer. GRP78 plays an anticancer role in cervical cancer by regulating autophagy and apoptosis. Mediated immune CD8 + T cells regulate tumor cell immunity and play a role in the application of the HPV vaccine. CONCLUSIONS GRP78 plays a multifunctional role in cervical cancer and has important therapeutic and diagnostic value.
Collapse
Affiliation(s)
- Yingying Bai
- Department of Gynecology and obstetrics, Tangdu Hospital, Air Force Medical University, 569Xinsi Road, Baqiao District, Xian, 710038 China
| | - Wenhua Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Yuemei Cheng
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Yongxiu Yang
- Department of Gynecology and obstetrics, Tangdu Hospital, Air Force Medical University, 569Xinsi Road, Baqiao District, Xian, 710038 China
- Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Lanzhou, Gansu People’s Republic of China
- No.1, Dong gang West Road, Cheng guan District, Lanzhou, Gansu People’s Republic of China
| |
Collapse
|
19
|
Peña MS, Tang FHF, Franco FADL, Rodrigues AT, Carrara GMP, Araujo TLS, Giordano RJ, Palmisano G, de Camargo MM, Uliana SRB, Stolf BS. Leishmania (L.) amazonensis LaLRR17 increases parasite entry in macrophage by a mechanism dependent on GRP78. Parasitology 2023; 150:922-933. [PMID: 37553284 PMCID: PMC10577668 DOI: 10.1017/s0031182023000720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/29/2023] [Accepted: 07/27/2023] [Indexed: 08/10/2023]
Abstract
Leishmaniases affect 12 million people worldwide. They are caused by Leishmania spp., protozoan parasites transmitted to mammals by female phlebotomine flies. During the life cycle, promastigote forms of the parasite live in the gut of infected sandflies and convert into amastigotes inside the vertebrate macrophages. The parasite evades macrophage's microbicidal responses due to virulence factors that affect parasite phagocytosis, survival and/or proliferation. The interaction between Leishmania and macrophage molecules is essential to phagocytosis and parasite survival. Proteins containing leucine-rich repeats (LRRs) are common in several organisms, and these motifs are usually involved in protein–protein interactions. We have identified the LRR17 gene, which encodes a protein with 6 LRR domains, in the genomes of several Leishmania species. We show here that promastigotes of Leishmania (L.) amazonensis overexpressing LaLRR17 are more infective in vitro. We produced recombinant LaLRR17 protein and identified macrophage 78 kDa glucose-regulated protein (GRP78) as a ligand for LaLRR17 employing affinity chromatography followed by mass spectrometry. We showed that GRP78 binds to LaLRR17 and that its blocking precludes the increase of infection conferred by LaLRR17. Our results are the first to report LRR17 gene and protein, and we hope they stimulate further studies on how this protein increases phagocytosis of Leishmania.
Collapse
Affiliation(s)
- Mauricio S. Peña
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Fenny Hui Fen Tang
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | | | | | | | | | - Ricardo José Giordano
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Giuseppe Palmisano
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | - Beatriz Simonsen Stolf
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
20
|
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
|
21
|
Khater I, Nassar A. A computational peptide model induces cancer cells' apoptosis by docking Kringle 5 to GRP78. BMC Mol Cell Biol 2023; 24:25. [PMID: 37553635 PMCID: PMC10408047 DOI: 10.1186/s12860-023-00484-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/23/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Cells can die through a process called apoptosis in both pathological and healthy conditions. Cancer development and progression may result from abnormal apoptosis. The 78-kDa glucose-regulated protein (GRP78) is increased on the surface of cancer cells. Kringle 5, a cell apoptosis agent, is bound to GRP78 to induce cancer cell apoptosis. Kringle 5 was docked to GRP78 using ClusPro 2.0. The interaction between Kringle 5 and GRP78 was investigated. RESULTS The interacting amino acids were found to be localized in three areas of Kringle 5. The proposed peptide is made up of secondary structure amino acids that contain Kringle 5 interaction residues. The 3D structure of the peptide model amino acids was created using the PEP-FOLD3 web tool. CONCLUSIONS The proposed peptide completely binds to the GRP78 binding site on the Kringle 5, signaling that it might be effective in the apoptosis of cancer cells.
Collapse
Affiliation(s)
- Ibrahim Khater
- Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Aaya Nassar
- Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt.
- Department of Clinical Research and Leadership, School of Medicine and Health Sciences, George Washington University, Washington DC, USA.
| |
Collapse
|
22
|
Liu Z, Liu G, Ha DP, Wang J, Xiong M, Lee AS. ER chaperone GRP78/BiP translocates to the nucleus under stress and acts as a transcriptional regulator. Proc Natl Acad Sci U S A 2023; 120:e2303448120. [PMID: 37487081 PMCID: PMC10400976 DOI: 10.1073/pnas.2303448120] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/08/2023] [Indexed: 07/26/2023] Open
Abstract
Cancer cells are commonly subjected to endoplasmic reticulum (ER) stress. To gain survival advantage, cancer cells exploit the adaptive aspects of the unfolded protein response such as upregulation of the ER luminal chaperone GRP78. The finding that when overexpressed, GRP78 can escape to other cellular compartments to gain new functions regulating homeostasis and tumorigenesis represents a paradigm shift. Here, toward deciphering the mechanisms whereby GRP78 knockdown suppresses EGFR transcription, we find that nuclear GRP78 is prominent in cancer and stressed cells and uncover a nuclear localization signal critical for its translocation and nuclear activity. Furthermore, nuclear GRP78 can regulate expression of genes and pathways, notably those important for cell migration and invasion, by interacting with and inhibiting the activity of the transcriptional repressor ID2. Our study reveals a mechanism for cancer cells to respond to ER stress via transcriptional regulation mediated by nuclear GRP78 to adopt an invasive phenotype.
Collapse
Affiliation(s)
- Ze Liu
- Department of Biochemistry and Molecular Medicine, University of Southern California, Keck School of Medicine, Los Angeles, CA90033
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Guanlin Liu
- Department of Biochemistry and Molecular Medicine, University of Southern California, Keck School of Medicine, Los Angeles, CA90033
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Dat P. Ha
- Department of Biochemistry and Molecular Medicine, University of Southern California, Keck School of Medicine, Los Angeles, CA90033
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Justin Wang
- Department of Molecular Medicine, Scripps Research, La Jolla, CA92037
| | - Min Xiong
- Department of System Biology, Beckman Research Institute, City of Hope, Duarte, CA91010
| | - Amy S. Lee
- Department of Biochemistry and Molecular Medicine, University of Southern California, Keck School of Medicine, Los Angeles, CA90033
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| |
Collapse
|
23
|
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
|
24
|
Yao B, Wang L, Xie C, Li M, Peng C, Li Z, Lu W, Chen J. Biological evaluation of a novel stable peptide PET molecular probe [ 18F]AlF-NOTA- DVAP targeting to tumor cell surface GRP78. Nucl Med Biol 2023; 118-119:108330. [PMID: 36889247 DOI: 10.1016/j.nucmedbio.2023.108330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 03/04/2023]
Abstract
BACKGROUNDS Glucose-Regulated Protein 78 (GRP78) is an attractive anticancer target for its selective anchoring on the surface of tumor cells and cancer endothelial cells rather than normal cells. Cell-surface GRP78 overexpression of tumor indicates that GRP78 is a crucial target for relative tumor imaging and clinical treatment. Herein, we report the design and preclinical evaluation of a new D peptide ligand [18F]AlF-NOTA-DVAP recognizing GRP78 expressed on the cell surface of breast cancer. METHODS Radiochemical synthesis of [18F]AlF-NOTA-DVAP was achieved via a one-pot labeling process by heating NOTA-DVAP in the presence of in situ prepared [18F]AlF for 15 min at 110 °C and purified through HPLC. RESULTS The radiotracer showed high in vitro stability in rat serum at 37 °C over 3 h. Both biodistribution studies and in vivo micro-PET/CT imaging studies in BALB/c mice bearing 4 T1 tumor showed [18F]AlF-NOTA-DVAP had a rapid and high uptake in tumor, as well as a long residence time. The high hydrophilicity of the radiotracer enables its fast clearance from most normal tissues and thus improves the tumor-to-normal tissue ratios (4.40 at 60 min) which is better than [18F]FDG (1.31 at 60 min). Pharmacokinetic studies showed the average in vivo mean residence time of the radiotracer was just 0.6432 h and indicated that this hydrophilic radiotracer was quickly eliminated from the body to reduce the distribution of non-target tissues. CONCLUSIONS These results suggest that [18F]AlF-NOTA-DVAP is a very promising PET probe for tumor-specific imaging of cell-surface GRP78-positive tumor.
Collapse
Affiliation(s)
- Bolin Yao
- Radiopharmacy and Molecular Imaging Center, School of Pharmacy, Fudan University, Shanghai 201203, China; Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China.
| | - Luting Wang
- Radiopharmacy and Molecular Imaging Center, School of Pharmacy, Fudan University, Shanghai 201203, China; Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Cao Xie
- Radiopharmacy and Molecular Imaging Center, School of Pharmacy, Fudan University, Shanghai 201203, China; Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Ming Li
- PET Center, Huashan Hospital, Fudan University, Shanghai 200235, China
| | - Chengyuan Peng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhuoyun Li
- Radiopharmacy and Molecular Imaging Center, School of Pharmacy, Fudan University, Shanghai 201203, China; Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China.
| | - Weiyue Lu
- Radiopharmacy and Molecular Imaging Center, School of Pharmacy, Fudan University, Shanghai 201203, China; Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China.
| | - Jian Chen
- Radiopharmacy and Molecular Imaging Center, School of Pharmacy, Fudan University, Shanghai 201203, China; Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China.
| |
Collapse
|
25
|
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
|
26
|
Schneider M, Antes I. Comparison of allosteric signaling in DnaK and BiP using mutual information between simulated residue conformations. Proteins 2023; 91:237-255. [PMID: 36111439 DOI: 10.1002/prot.26425] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/06/2022] [Accepted: 09/13/2022] [Indexed: 01/13/2023]
Abstract
The heat shock protein 70 kDa (Hsp70) chaperone system serves as a critical component of protein quality control across a wide range of prokaryotic and eukaryotic organisms. Divergent evolution and specialization to particular organelles have produced numerous Hsp70 variants which share similarities in structure and general function, but differ substantially in regulatory aspects, including conformational dynamics and activity modulation by cochaperones. The human Hsp70 variant BiP (also known as GRP78 or HSPA5) is of therapeutic interest in the context of cancer, neurodegenerative diseases, and viral infection, including for treatment of the pandemic virus SARS-CoV-2. Due to the complex conformational rearrangements and high sequential variance within the Hsp70 protein family, it is in many cases poorly understood which amino acid mutations are responsible for biochemical differences between protein variants. In this study, we predicted residues associated with conformational regulation of human BiP and Escherichia coli DnaK. Based on protein structure networks obtained from molecular dynamics simulations, we analyzed the shared information between interaction timelines to highlight residue positions with strong conformational coupling to their environment. Our predictions, which focus on the binding processes of the chaperone's substrate and cochaperones, indicate residues filling potential signaling roles specific to either DnaK or BiP. By combining predictions of individual residues into conformationally coupled chains connecting ligand binding sites, we predict a BiP specific secondary signaling pathway associated with substrate binding. Our study sheds light on mechanistic differences in signaling and regulation between Hsp70 variants, which provide insights relevant to therapeutic applications of these proteins.
Collapse
Affiliation(s)
- Markus Schneider
- TUM Center for Functional Protein Assemblies and TUM School of Life Sciences, Technische Universität München, Freising, Bavaria, Germany
| | - Iris Antes
- TUM Center for Functional Protein Assemblies and TUM School of Life Sciences, Technische Universität München, Freising, Bavaria, Germany
| |
Collapse
|
27
|
Zheng S, Wang X, Liu H, Zhao D, Lin Q, Jiang Q, Li L, Hu Y. iASPP suppression mediates terminal UPR and improves BRAF-inhibitor sensitivity of colon cancers. Cell Death Differ 2023; 30:327-340. [PMID: 36380064 PMCID: PMC9950372 DOI: 10.1038/s41418-022-01086-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/23/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022] Open
Abstract
Unfolded protein response (UPR) signaling is activated under endoplasmic reticulum (ER) stress, an emerging cancer hallmark, leading to either adaptive survival or cell death, while the mechanisms underlying adaptation-death switch remain poorly understood. Here, we examined whether oncogene iASPP regulates the switch and how the mechanisms can be used in colon cancer treatment. iASPP is downregulated when cells undergo transition from adaptation to death during therapy-induced ER stress. Blocking iASPP's downregulation attenuates stress-induced cell death. Mechanistically, Hu-antigen R (HuR)-mediated stabilization of iASPP mRNA and subsequent iASPP protein production is significantly impaired with prolonged ER stress, which facilitates the degradation of GRP78, a key regulator of the UPR, in the cytosol. Because iASPP competes with GRP78 in binding the ER-resident E3 ligase RNF185, and tips the balance in favor of cell death. Positive correlation between the levels of HuR, iASPP, and GRP78 are detectable in colon cancer tissues in vivo. Genetic inhibition of iASPP/GRP78 or chemical inhibition of HuR not only inhibits tumor growth, but also sensitizes colon cancer cells' responses to BRAF inhibitor-induced ER stress and cell death. This study provides mechanistic insights into the switch between adaptation and death during ER stress, and also identifies a potential strategy to improve BRAF-inhibitor efficiency in colon cancers.
Collapse
Affiliation(s)
- Shanliang Zheng
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
| | - Xingwen Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
| | - Hao Liu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
| | - Dong Zhao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
| | - Qingyu Lin
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
| | - Qinghua Jiang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
| | - Li Li
- The third affiliated hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150040, China.
| | - Ying Hu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China.
| |
Collapse
|
28
|
Eggleton P, De Alba J, Weinreich M, Calias P, Foulkes R, Corrigall VM. The therapeutic mavericks: Potent immunomodulating chaperones capable of treating human diseases. J Cell Mol Med 2023; 27:322-339. [PMID: 36651415 PMCID: PMC9889696 DOI: 10.1111/jcmm.17669] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/16/2022] [Accepted: 12/28/2022] [Indexed: 01/19/2023] Open
Abstract
Two major chaperones, calreticulin (CRT) and binding immunoglobulin protein (GRP78/BiP) dependent on their location, have immunoregulatory or anti-inflammatory functions respectively. CRT induces pro-inflammatory cytokines, dendritic cell (DC) maturation and activates cytotoxic T cells against tumours. By contrast, GRP78/BiP induces anti-inflammatory cytokines, inhibits DC maturation and heightens T-regulatory cell responses. These latter functions rebalance immune homeostasis in inflammatory diseases, such as rheumatoid arthritis. Both chaperones are therapeutically relevant agents acting primarily on monocytes/DCs. Endogenous exposure of CRT on cancer cell surfaces acts as an 'eat-me' signal and facilitates improved elimination of stressed and dying tumour cells by DCs. Therefore, therapeutics that promote endogenous CRT translocation to the cell surface can improve the removal of cancer cells. However, infused recombinant CRT dampens this cancer cell eradication by binding directly to the DCs. Low levels of endogenous BiP appear as a surface biomarker of endoplasmic reticulum (ER) stress in some types of tumour cells, a reflection of cells undergoing proliferation, in which resulting hypoxia and nutrient deprivation perturb ER homeostasis triggering the unfolded protein response, leading to increased expression of GRP78/BiP and altered cellular location. Conversely, infusion of an analogue of GRP78/BiP (IRL201805) can lead to long-term immune resetting and restoration of immune homeostasis. The therapeutic potential of both chaperones relies on them being relocated from their intracellular ER environment. Ongoing clinical trials are employing therapeutic interventions to either enhance endogenous cell surface CRT or infuse IRL201805, thereby triggering several disease-relevant immune responses leading to a beneficial clinical outcome.
Collapse
Affiliation(s)
- Paul Eggleton
- Revolo BiotherapeuticsNew OrleansLouisianaUSA,University of Exeter Medical SchoolExeterUK
| | | | | | | | | | - Valerie M. Corrigall
- Revolo BiotherapeuticsNew OrleansLouisianaUSA,Centre for Inflammation Biology and Cancer Immunology, King's College London, New Hunts HouseGuy' HospitalLondonUK
| |
Collapse
|
29
|
Parrasia S, Szabò I, Zoratti M, Biasutto L. Peptides as Pharmacological Carriers to the Brain: Promises, Shortcomings and Challenges. Mol Pharm 2022; 19:3700-3729. [PMID: 36174227 DOI: 10.1021/acs.molpharmaceut.2c00523] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Central nervous system (CNS) diseases are among the most difficult to treat, mainly because the vast majority of the drugs fail to cross the blood-brain barrier (BBB) or to reach the brain at concentrations adequate to exert a pharmacological activity. The obstacle posed by the BBB has led to the in-depth study of strategies allowing the brain delivery of CNS-active drugs. Among the most promising strategies is the use of peptides addressed to the BBB. Peptides are versatile molecules that can be used to decorate nanoparticles or can be conjugated to drugs, with either a stable link or as pro-drugs. They have been used to deliver to the brain both small molecules and proteins, with applications in diverse therapeutic areas such as brain cancers, neurodegenerative diseases and imaging. Peptides can be generally classified as receptor-targeted, recognizing membrane proteins expressed by the BBB microvessels (e.g., Angiopep2, CDX, and iRGD), "cell-penetrating peptides" (CPPs; e.g. TAT47-57, SynB1/3, and Penetratin), undergoing transcytosis through unspecific mechanisms, or those exploiting a mixed approach. The advantages of peptides have been extensively pointed out, but so far few studies have focused on the potential negative aspects. Indeed, despite having a generally good safety profile, some peptide conjugates may display toxicological characteristics distinct from those of the peptide itself, causing for instance antigenicity, cardiovascular alterations or hemolysis. Other shortcomings are the often brief lifetime in vivo, caused by the presence of peptidases, the vulnerability to endosomal/lysosomal degradation, and the frequently still insufficient attainable increase of brain drug levels, which remain below the therapeutically useful concentrations. The aim of this review is to analyze not only the successful and promising aspects of the use of peptides in brain targeting but also the problems posed by this strategy for drug delivery.
Collapse
Affiliation(s)
- Sofia Parrasia
- Department of Biology, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| | - Ildikò Szabò
- Department of Biology, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| | - Mario Zoratti
- CNR Neuroscience Institute, Viale G. Colombo 3, 35131 Padova, Italy.,Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| | - Lucia Biasutto
- CNR Neuroscience Institute, Viale G. Colombo 3, 35131 Padova, Italy.,Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| |
Collapse
|
30
|
Hu C, Yang J, Qi Z, Wu H, Wang B, Zou F, Mei H, Liu J, Wang W, Liu Q. Heat shock proteins: Biological functions, pathological roles, and therapeutic opportunities. MedComm (Beijing) 2022; 3:e161. [PMID: 35928554 PMCID: PMC9345296 DOI: 10.1002/mco2.161] [Citation(s) in RCA: 237] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 12/12/2022] Open
Abstract
The heat shock proteins (HSPs) are ubiquitous and conserved protein families in both prokaryotic and eukaryotic organisms, and they maintain cellular proteostasis and protect cells from stresses. HSP protein families are classified based on their molecular weights, mainly including large HSPs, HSP90, HSP70, HSP60, HSP40, and small HSPs. They function as molecular chaperons in cells and work as an integrated network, participating in the folding of newly synthesized polypeptides, refolding metastable proteins, protein complex assembly, dissociating protein aggregate dissociation, and the degradation of misfolded proteins. In addition to their chaperone functions, they also play important roles in cell signaling transduction, cell cycle, and apoptosis regulation. Therefore, malfunction of HSPs is related with many diseases, including cancers, neurodegeneration, and other diseases. In this review, we describe the current understandings about the molecular mechanisms of the major HSP families including HSP90/HSP70/HSP60/HSP110 and small HSPs, how the HSPs keep the protein proteostasis and response to stresses, and we also discuss their roles in diseases and the recent exploration of HSP related therapy and diagnosis to modulate diseases. These research advances offer new prospects of HSPs as potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Chen Hu
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
| | - Jing Yang
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
| | - Ziping Qi
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
| | - Hong Wu
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
| | - Beilei Wang
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
| | - Fengming Zou
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
| | - Husheng Mei
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- University of Science and Technology of ChinaHefeiAnhuiP. R. China
| | - Jing Liu
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
- University of Science and Technology of ChinaHefeiAnhuiP. R. China
| | - Wenchao Wang
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
- University of Science and Technology of ChinaHefeiAnhuiP. R. China
| | - Qingsong Liu
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
- University of Science and Technology of ChinaHefeiAnhuiP. R. China
- Precision Medicine Research Laboratory of Anhui ProvinceHefeiAnhuiP. R. China
| |
Collapse
|
31
|
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: 11] [Impact Index Per Article: 3.7] [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
|
32
|
Decoy receptor 2 mediates the apoptosis-resistant phenotype of senescent renal tubular cells and accelerates renal fibrosis in diabetic nephropathy. Cell Death Dis 2022; 13:522. [PMID: 35661704 PMCID: PMC9166763 DOI: 10.1038/s41419-022-04972-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 05/18/2022] [Accepted: 05/25/2022] [Indexed: 01/21/2023]
Abstract
Apoptotic resistance leads to persistent accumulation of senescent cells and sustained expression of a senescence-associated secretory phenotype, playing an essential role in the progression of tissue fibrosis. However, whether senescent renal tubular epithelial cells (RTECs) exhibit an apoptosis-resistant phenotype, and the role of this phenotype in diabetic nephropathy (DN) remain unclear. Our previous study was the first to demonstrate that decoy receptor 2 (DcR2) is associated with apoptotic resistance in senescent RTECs and renal fibrosis. In this study, we aimed to further explore the mechanism of DcR2 in apoptosis-resistant RTECs and renal fibrosis in DN. DcR2 was co-localized with fibrotic markers (α-SMA, collagen IV, fibronectin), senescent marker p16, and antiapoptotic proteins FLIP and Bcl2 but rarely co-localized with caspase 3 or TUNEL. DcR2 overexpression promoted renal fibrosis in mice with streptozotocin (STZ)-induced DN, as evidenced by augmented Masson staining and upregulated expression of fibrotic markers. DcR2 overexpression also enhanced FLIP expression while reducing the expression of pro-apoptotic proteins (caspases 8 and 3) in senescent RTECs, resulting in apoptotic resistance. In contrast, DcR2 knockdown produced the opposite effects in vitro and in vivo. Moreover, quantitative proteomics and co-immunoprecipitation experiments demonstrated that DcR2 interacted with glucose-related protein 78 kDa (GRP78), which has been shown to promote apoptotic resistance in cancer. GRP78 exhibited co-localization with senescent and antiapoptotic markers but was rarely co-expressed with caspase 3 or TUNEL. Additionally, GRP78 knockdown decreased the apoptosis resistance of HG-induced senescent RTECs with upregulated cleaved caspase 3 and increased the percentage of apoptotic RTECs. Mechanistically, DcR2 mediated apoptotic resistance in senescent RTECs by enhancing GRP78-caspase 7 interactions and promoting Akt phosphorylation. Thus, DcR2 mediated the apoptotic resistance of senescent RTECs and renal fibrosis by interacting with GRP78, indicating that targeting the DcR2-GRP78 axis represents a promising therapeutic strategy for DN.
Collapse
|
33
|
Wen Y, Jing N, Huo F, Yin C. Rational design of a turn-on fluorescent probe for visualization of GRP78 protein in tumor models. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.06.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
34
|
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
|
35
|
Qiao L, Yang H, Shao XX, Yin Q, Fu XJ, Wei Q. Research Progress on Nanoplatforms and Nanotherapeutic Strategies in Treating Glioma. Mol Pharm 2022; 19:1927-1951. [DOI: 10.1021/acs.molpharmaceut.1c00856] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Li Qiao
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Marine Traditional Chinese Medicine Research Center, Qingdao Academy of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Qingdao 266114, China
| | - Huishu Yang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xin-xin Shao
- Marine Traditional Chinese Medicine Research Center, Qingdao Academy of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Qingdao 266114, China
| | - Qiuyan Yin
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xian-Jun Fu
- Marine Traditional Chinese Medicine Research Center, Qingdao Academy of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Qingdao 266114, China
- Shandong Engineering and Technology Research Center of Traditional Chinese Medicine, Jinan 250355, China
| | - Qingcong Wei
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| |
Collapse
|
36
|
Ye JJ, Yu W, Xie BR, Li K, Liu MD, Dong X, Chen ZX, Feng J, Zhang XZ. Self-Reinforced Cancer Targeting (SRCT) Depending on Reciprocally Enhancing Feedback between Targeting and Therapy. ACS NANO 2022; 16:5851-5866. [PMID: 35412799 DOI: 10.1021/acsnano.1c10999] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Conventional cancer targeting methodology needs to be reformed to overcome the intrinsic barriers responsible for poor targeting efficiency. This study describes a concept of self-reinforced cancer targeting (SRCT) by correlating targeting with therapy in a reciprocally enhancing manner. SRCT is achieved on the basis of two prerequisites: (1) target molecules have to be expressed on cancer cell membranes but not on normal cells, and (2) notably, their expression on cancer cells must be actively upregulated in response to cellular attack by cancer treatments. As a proof-of-concept, a GRP78-targeting nanovehicle for chemotherapy was designed. Resultant data showed that chemotherapeutic drugs could effectively elevate GRP78 expression on the plasma membranes of cancer cells while having minimal influence on normal cells. DOX pretreatment of cancer cells and tumor tissues can greatly increase the targeting efficacy and therapeutic performance of the prepared GRP78-targeting nanomedicine while somewhat disfavoring the nontargeting counterpart. In vivo and in vitro results demonstrated that this GRP78-targeting nanomedicine could accurately target cancer cells to not only implement chemotherapy but also induce GRP78 upregulation on cancer cells, eventually benefiting continuous cancer-cell-targeted attack by the nanomedicines remaining in the blood circulation or administered in the next dose. The GRP78-targeting nanomedicine displays much better antitumor performance compared with the nontargeting counterpart. SRCT is expected to advance cancer-targeted therapy based on the positive dependency between targeting and therapeutic modalities.
Collapse
Affiliation(s)
- Jing-Jie Ye
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China
| | - Wuyang Yu
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China
| | - Bo-Ru Xie
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China
| | - Ke Li
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China
| | - Miao-Deng Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China
| | - Xue Dong
- Institute for Advanced Studies, Wuhan University, Wuhan 430072, P.R. China
| | - Zhao-Xia Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China
| | - Jun Feng
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China
| |
Collapse
|
37
|
Shan S, Niu J, Yin R, Shi J, Zhang L, Wu C, Li H, Li Z. Peroxidase from foxtail millet bran exerts anti-colorectal cancer activity via targeting cell-surface GRP78 to inactivate STAT3 pathway. Acta Pharm Sin B 2022; 12:1254-1270. [PMID: 35530132 PMCID: PMC9069399 DOI: 10.1016/j.apsb.2021.10.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 12/24/2022] Open
Abstract
Molecular targeted therapy has become an emerging promising strategy in cancer treatment, and screening the agents targeting at cancer cell specific targets is very desirable for cancer treatment. Our previous study firstly found that a secretory peroxidase of class III derived from foxtail millet bran (FMBP) exhibited excellent targeting anti-colorectal cancer (CRC) activity in vivo and in vitro, whereas its underlying target remains unclear. The highlight of present study focuses on the finding that cell surface glucose-regulated protein 78 (csGRP78) abnormally located on CRC is positively correlated with the anti-CRC effects of FMBP, indicating it serves as a potential target of FMBP against CRC. Further, we demonstrated that the combination of FMBP with the nucleotide binding domain (NBD) of csGRP78 interfered with the downstream activation of signal transducer and activator of transcription 3 (STAT3) in CRC cells, thus promoting the intracellular accumulation of reactive oxygen species (ROS) and cell grown inhibition. These phenomena were further confirmed in nude mice tumor model. Collectively, our study highlights csGRP78 acts as an underlying target of FMBP against CRC, uncovering the clinical potential of FMBP as a targeted agent for CRC in the future.
Collapse
Key Words
- CAC, colitis-associated carcinogenesis
- CDKs, cyclin-dependent kinases
- CETSA, cellular thermal shift assay
- CRC, colorectal cancer
- Co-IP, co-immunoprecipitation
- Colorectal cancer
- DCFH-DA, dichloro-dihydro-fluorescein diacetate
- EGFR, epidermal growth factor receptor
- ER, endoplasmic reticulum
- FDA, U.S. Food and Drug Administration
- FMBP
- FMBP, peroxidase derived from foxtail millet bran
- Foxtail millet bran
- GRP78, glucose-regulated protein 78
- H&E, hematoxylin & eosin
- ISM, isthmin
- MPs, membrane proteins
- NBD, the nucleotide binding domain of csGRP78
- PD-1, programmed death-1
- ROS
- ROS, reactive oxygen species
- SBD, substrate-binding domain of csGRP78
- SPF, specific pathogen free
- STAT3
- STAT3, signal transducer and activator of transcription 3
- TRAIL, tumor necrosis factor-related apoptosis-inducing ligand
- csGRP78
- csGRP78, cell surface glucose-regulated protein 78
- rGRP78, recombinant GRP78
Collapse
|
38
|
Liu S, Li Y, Li Z. Salidroside suppresses the activation of nasopharyngeal carcinoma cells via targeting miR-4262/GRP78 axis. Cell Cycle 2022; 21:720-729. [PMID: 35220889 PMCID: PMC8973335 DOI: 10.1080/15384101.2021.2019976] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
To study the effect of Salidroside on nasopharyngeal carcinoma (NPC) cells and its mechanism. NPC cells were cultured, MTT was used to detect the effect of Salidroside on cell proliferation, apoptosis detected by flow cytometry assay, Western blot was used to detect the related protein expression. MiR-4262 and GRP78 used qRT-PCR for evaluation. Mimics/mimic NC and miR-4262 inhibitor/inhibitor NC were transfected into CNE2 and HONE1 cell lines, and cell viability was detected by MTT. Caspase-3, −8 and −9 activities were detected by caspase colorimetric assay kit. Targetscan predicted that downstream target of miR-4262. Relative luciferase activity was detected by luciferase assay. The effect of Salidroside on the growth of transplanted tumor in nude mice was observed. After Salidroside treatment, cell proliferation decreased and apoptosis increased, Bax protein expression increased and Bcl-2 decreased; miR-4262 expression level in nasopharyngeal carcinoma tissues was lower than that in adjacent tissues. GRP78 was the target of miR-4262 and downregulate the expression of miR-4262 in NPC cells can increase the expression of GRP78, and the expression of GRP78 decreased after upregulating the expression of miR-4262. Salidroside could inhibit the growth of NPC xenografts in nude mice. The level of Bax was increased and Bcl-2 was decreased in Salidroside group. Salidroside can significantly inhibit the proliferation and promote the apoptosis of NPC cells via regulating miR-4262/GRP78 signal axis.
Collapse
Affiliation(s)
- Shaosheng Liu
- Department of Otorhinolaryngology, People’s Hospital of Beilun (Beilun Branch, the First Hospital of Zhejiang University), Ningbo, China
| | - Yuanyuan Li
- Department of Medicine and Equipment, The 988 Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army Kaifeng, Hena, P.R. China
| | - Zhaoxia Li
- Department of Otolaryngology, Jinan Central Hospital. Jinan City, Shangdong, P.R. China
| |
Collapse
|
39
|
Luo D, Fan N, Zhang X, Ngo FY, Zhao J, Zhao W, Huang M, Li D, Wang Y, Rong J. Covalent inhibition of endoplasmic reticulum chaperone GRP78 disconnects the transduction of ER stress signals to inflammation and lipid accumulation in diet-induced obese mice. eLife 2022; 11:72182. [PMID: 35138251 PMCID: PMC8828050 DOI: 10.7554/elife.72182] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 01/12/2022] [Indexed: 12/14/2022] Open
Abstract
Targeting endoplasmic reticulum (ER) stress, inflammation, and metabolic dysfunctions may halt the pathogenesis of obesity and thereby reduce the prevalence of diabetes, cardiovascular disesases, and cancers. The present study was designed to elucidate the mechnaisms by which plant-derived celastrol ameliorated inflammation and lipid accumulation in obesity. The mouse model of diet-induced obesity was induced by feeding high-fat diet for 3 months and subsequently intervented with celastrol for 21 days. Hepatic and adipose tissues were analyzed for lipid accumulation, macrophage activation, and biomarker expression. As result, celastrol effectively reduced body weight, suppressed ER stress, inflammation, and lipogenesis while promoted hepatic lipolysis. RNA-sequencing revealed that celastrol-loaded nanomicelles restored the expression of 49 genes that regulate ER stress, inflammation, and lipid metabolism. On the other hand, celastrol-PEG4-alkyne was synthesized for identifying celastrol-bound proteins in RAW264.7 macrophages. ER chaperone GRP78 (78 kDa glucose-regulated protein) was identified by proteomics approach for celastrol binding to the residue Cys41. Upon binding and conjugation, celastrol diminished the chaperone activity of GRP78 by 130-fold and reduced ER stress in palmitate-challenged cells, while celastrol analog lacking quinone methide failed to exhibit antiobesity effects. Thus, covalent GRP78 inhibition may induce the reprograming of ER signaling, inflammation, and metabolism against diet-induced obesity.
Collapse
Affiliation(s)
- Dan Luo
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Ni Fan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Xiuying Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Fung Yin Ngo
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Jia Zhao
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Wei Zhao
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Ming Huang
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Ding Li
- Colleage of Chemistry and Pharmacy, College of Chemistry & Pharmacy, Northwest A&F University, Shaanxi, China
| | - Yu Wang
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Jianhui Rong
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| |
Collapse
|
40
|
Hernandez I, Cohen M. Linking cell-surface GRP78 to cancer: From basic research to clinical value of GRP78 antibodies. Cancer Lett 2022; 524:1-14. [PMID: 34637844 DOI: 10.1016/j.canlet.2021.10.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/10/2021] [Accepted: 10/05/2021] [Indexed: 01/01/2023]
Abstract
Glucose-related protein 78 (GRP78) is a chaperone protein localized primarily in the endoplasmic reticulum (ER) lumen, where it helps in proper protein folding by targeting misfolded proteins and facilitating protein assembly. In stressed cells, GRP78 is translocated to the cell surface (csGRP78) where it binds to various ligands and triggers different intracellular pathways. Thus, csGRP78 expression is associated with cancer, involved in the maintenance and progression of the disease. Extracellular exposition of csGRP78 leads to the production of autoantibodies as observed in patients with prostate or ovarian cancer, in which the ability to target csGRP78 affects the tumor development. Present on the surface of cancer cells and not normal cells in vivo, csGRP78 represents an interesting target for therapeutic antibody strategies. Here we give an overview of the csGRP78 function in the cell and its role in oncogenesis, thereby providing insight into the clinical value of GRP78 monoclonal antibodies for cancer prognosis and treatment.
Collapse
Affiliation(s)
- Isabelle Hernandez
- Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Translational Research Centre in Oncohaematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Marie Cohen
- Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Translational Research Centre in Oncohaematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
41
|
Kabakov AE, Gabai VL. HSP70s in Breast Cancer: Promoters of Tumorigenesis and Potential Targets/Tools for Therapy. Cells 2021; 10:cells10123446. [PMID: 34943954 PMCID: PMC8700403 DOI: 10.3390/cells10123446] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/25/2021] [Accepted: 12/03/2021] [Indexed: 12/20/2022] Open
Abstract
The high frequency of breast cancer worldwide and the high mortality among women with this malignancy are a serious challenge for modern medicine. A deeper understanding of the mechanisms of carcinogenesis and emergence of metastatic, therapy-resistant breast cancers would help development of novel approaches to better treatment of this disease. The review is dedicated to the role of members of the heat shock protein 70 subfamily (HSP70s or HSPA), mainly inducible HSP70, glucose-regulated protein 78 (GRP78 or HSPA5) and GRP75 (HSPA9 or mortalin), in the development and pathogenesis of breast cancer. Various HSP70-mediated cellular mechanisms and pathways which contribute to the oncogenic transformation of mammary gland epithelium are reviewed, as well as their role in the development of human breast carcinomas with invasive, metastatic traits along with the resistance to host immunity and conventional therapeutics. Additionally, intracellular and cell surface HSP70s are considered as potential targets for therapy or sensitization of breast cancer. We also discuss a clinical implication of Hsp70s and approaches to targeting breast cancer with gene vectors or nanoparticles downregulating HSP70s, natural or synthetic (small molecule) inhibitors of HSP70s, HSP70-binding antibodies, HSP70-derived peptides, and HSP70-based vaccines.
Collapse
Affiliation(s)
- Alexander E. Kabakov
- Department of Radiation Biochemistry, A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Koroleva 4, 249036 Obninsk, Russia;
| | - Vladimir L. Gabai
- CureLab Oncology Inc., Dedham, MA 02026, USA
- Correspondence: ; Tel.: +1-617-319-7314
| |
Collapse
|
42
|
De Maio A, Hightower L. The interaction of heat shock proteins with cellular membranes: a historical perspective. Cell Stress Chaperones 2021; 26:769-783. [PMID: 34478113 PMCID: PMC8413713 DOI: 10.1007/s12192-021-01228-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 01/09/2023] Open
Abstract
The interaction of heat shock proteins (HSP) with cellular membranes has been an enigmatic process, initially observed by morphological studies, inferred during the purification of HSP70s, and confirmed after the detection of these proteins on the surface of cancer cells and their insertion into artificial lipid bilayers. Today, the association of several HSP with lipid membranes is well established. However, the mechanisms for membrane insertion have been elusive. There is conclusive evidence indicating that HSP70s have a great selectivity for negatively charged phospholipids, whereas other HSP have a broader spectrum of lipid specificity. HSP70 also oligomerizes upon membrane insertion, forming ion conductance channels. The functional role of HSP70 lipid interactions appears related to membrane stabilization that may play a role during cell membrane biogenesis. They could also play a role as membrane chaperones as well as during endocytosis, microautophagy, and signal transduction. Moreover, HSP membrane association is a key component in the extracellular export of these proteins. The presence of HSP70 on the surface of cancer cells and its interaction with lysosome membranes have been envisioned as potential therapeutic targets. Thus, the biology and function of HSP membrane association are reaching a new level of excitement. This review is an attempt to preserve the recollection of the pioneering contributions of many investigators that have participated in this endeavor.
Collapse
Affiliation(s)
- Antonio De Maio
- Department of Surgery, Division of Trauma, Critical Care, Burns, and Acute Care Surgery, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
- Center for Investigations of Health and Education Disparities, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Lawrence Hightower
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, 06269, USA
| |
Collapse
|
43
|
Gonzalez-Gronow M, Gopal U, Austin RC, Pizzo SV. Glucose-regulated protein (GRP78) is an important cell surface receptor for viral invasion, cancers, and neurological disorders. IUBMB Life 2021; 73:843-854. [PMID: 33960608 DOI: 10.1002/iub.2502] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/14/2021] [Accepted: 05/01/2021] [Indexed: 12/22/2022]
Abstract
The 78 kDa glucose-regulated protein (GRP78) is an endoplasmic reticulum (ER)-resident molecular chaperone. GRP78 is a member of the 70 kDa heat shock family of proteins involved in correcting and clearing misfolded proteins in the ER. In response to cellular stress, GRP78 escapes from the ER and moves to the plasma membrane where it (a) functions as a receptor for many ligands, and (b) behaves as an autoantigen for autoantibodies that contribute to human disease and cancer. Cell surface GRP78 (csGRP78) associates with the major histocompatibility complex class I (MHC-I), and is the port of entry for several viruses, including the predictive binding of the novel SARS-CoV-2. Furthermore, csGRP78 is found in association with partners as diverse as the teratocarcinoma-derived growth factor 1 (Cripto), the melanocortin-4 receptor (MC4R) and the DnaJ-like protein MTJ-1. CsGRP78 also serves as a receptor for a large variety of ligands including activated α2 -macroglobulin (α2 M*), plasminogen kringle 5 (K5), microplasminogen, the voltage-dependent anion channel (VDAC), tissue factor (TF), and the prostate apoptosis response-4 protein (Par-4). In this review, we discuss the mechanisms involved in the translocation of GRP78 from the ER to the cell surface, and the role of secreted GRP78 and its autoantibodies in cancer and neurological disorders.
Collapse
Affiliation(s)
- Mario Gonzalez-Gronow
- Department of Biological Sciences, Laboratory of Environmental Neurotoxicology, Faculty of Medicine, Universidad Católica del Norte, Coquimbo, Chile.,Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Udhayakumar Gopal
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Richard C Austin
- Department of Medicine, Division of Nephrology, McMaster University and The Research Institute of St. Joseph's Hamilton, Hamilton, Ontario, Canada
| | - Salvatore V Pizzo
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
44
|
Descalzi-Montoya D, Montel RA, Smith K, Dziopa E, Darwich A, Yang Z, Bitsaktsis C, Korngold R, Sabatino D. Synthetic Antibody Mimics Based on Cancer-Targeting Immunostimulatory Peptides. Chembiochem 2021; 22:1589-1596. [PMID: 32964656 PMCID: PMC8191480 DOI: 10.1002/cbic.202000407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/16/2020] [Indexed: 11/08/2022]
Abstract
De novo cancer-targeting immunostimulatory peptides have been designed and developed as synthetic antibody mimics. A series of bifunctional peptides incorporating NKp30-binding and NK-cell-activating domains were synthesized as linear dimers and then extended into branching trimeric peptides by the incorporation of GRP78-targeting and tumor-cell-binding sequences. A selected trimeric peptide from this small set of peptides displayed binding capabilities on GRP78+ HepG2 and A549 target cells. Cell binding diminished in the presence of an anti-GRP78 peptide blocker, thus suggesting GRP78-binding dependence. Similarly, the selected trimeric peptide was also found to exhibit NK cell binding in an NKp30-dependent manner, which translated into NK cell activation as indicated by cytokine secretion. In co-culture, fluorescence microscopy revealed that the target GFP-expressing A549 cells were visibly associated with the effector NK cells when pre-activated with lead trimeric peptide. Accordingly, A549 cells were found to be compromised, as evidenced by the loss of GFP signal and notable detection of early-/late-stage apoptosis. Investigation of the immunological markers related to toxicity revealed detectable secretion of pro-inflammatory cytokines and chemokines, including IFN-γ, TNF-α, and IL-8. Furthermore, administration of peptide-activated NK cells into A549-tumor-bearing mice resulted in a consistent decrease in tumor growth when compared to the untreated control group. Taken together, the identification of a lead trimeric peptide capable of targeting and activating NK cells' immunotoxicity directly towards GRP78+ /B7H6- tumors provides a novel proof-of-concept for the development of cancer-targeting immunostimulatory peptide ligands that mimic antibody-targeting and -activating functions related to cancer immunotherapy applications.
Collapse
MESH Headings
- Adjuvants, Immunologic/chemistry
- Adjuvants, Immunologic/pharmacology
- Adjuvants, Immunologic/therapeutic use
- Animals
- Antibodies/chemistry
- Antibodies/immunology
- Cell Line, Tumor
- Cytokines/metabolism
- Endoplasmic Reticulum Chaperone BiP/immunology
- Female
- Humans
- Immunotherapy/methods
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lymphocyte Activation/drug effects
- Male
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Neoplasms/drug therapy
- Neoplasms/pathology
- Peptides/chemical synthesis
- Peptides/chemistry
- Peptides/pharmacology
- Peptides/therapeutic use
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Dante Descalzi-Montoya
- Center for Discovery and Innovation, Hackensack-Meridian Health, 340 Kingsland Street, Nutley, NJ 07110, USA
| | - Rachel A Montel
- Department of Biological Sciences and Chemistry and Biochemistry, Seton Hall University, 400 South Orange Avenue, South Orange, NJ 07079, USA
| | - Keith Smith
- Department of Biological Sciences and Chemistry and Biochemistry, Seton Hall University, 400 South Orange Avenue, South Orange, NJ 07079, USA
| | - Eugenia Dziopa
- Center for Discovery and Innovation, Hackensack-Meridian Health, 340 Kingsland Street, Nutley, NJ 07110, USA
| | - Andrieh Darwich
- Department of Biological Sciences and Chemistry and Biochemistry, Seton Hall University, 400 South Orange Avenue, South Orange, NJ 07079, USA
| | - Zheng Yang
- Center for Discovery and Innovation, Hackensack-Meridian Health, 340 Kingsland Street, Nutley, NJ 07110, USA
| | - Constantine Bitsaktsis
- Department of Biological Sciences and Chemistry and Biochemistry, Seton Hall University, 400 South Orange Avenue, South Orange, NJ 07079, USA
| | - Robert Korngold
- Center for Discovery and Innovation, Hackensack-Meridian Health, 340 Kingsland Street, Nutley, NJ 07110, USA
| | - David Sabatino
- Department of Biological Sciences and Chemistry and Biochemistry, Seton Hall University, 400 South Orange Avenue, South Orange, NJ 07079, USA
| |
Collapse
|
45
|
Ayo A, Laakkonen P. Peptide-Based Strategies for Targeted Tumor Treatment and Imaging. Pharmaceutics 2021; 13:pharmaceutics13040481. [PMID: 33918106 PMCID: PMC8065807 DOI: 10.3390/pharmaceutics13040481] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 02/03/2023] Open
Abstract
Cancer is one of the leading causes of death worldwide. The development of cancer-specific diagnostic agents and anticancer toxins would improve patient survival. The current and standard types of medical care for cancer patients, including surgery, radiotherapy, and chemotherapy, are not able to treat all cancers. A new treatment strategy utilizing tumor targeting peptides to selectively deliver drugs or applicable active agents to solid tumors is becoming a promising approach. In this review, we discuss the different tumor-homing peptides discovered through combinatorial library screening, as well as native active peptides. The different structure–function relationship data that have been used to improve the peptide’s activity and conjugation strategies are highlighted.
Collapse
Affiliation(s)
- Abiodun Ayo
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
| | - Pirjo Laakkonen
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
- Laboratory Animal Center, HiLIFE—Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
- Correspondence: ; Tel.: +358-50-4489100
| |
Collapse
|
46
|
Abstract
Glucose-regulating protein 78 (GRP78) is a molecular chaperone in the endoplasmic reticulum (ER) that promotes folding and assembly of proteins, controls the quality of proteins, and regulates ER stress signaling through Ca2+ binding to the ER. In tumors, GRP78 is often upregulated, acting as a central stress sensor that senses and adapts to changes in the tumor microenvironment, mediating ER stress of cancer cells under various stimulations of the microenvironment to trigger the folding protein response. Increasing evidence has shown that GRP78 is closely associated with the progression and poor prognosis of lung cancer, and plays an important role in the treatment of lung cancer. Herein, we reviewed for the first time the functions and mechanisms of GRP78 in the pathological processes of lung cancer, including tumorigenesis, apoptosis, autophagy, progression, and drug resistance, giving a comprehensive understanding of the function of GRP78 in lung cancer. In addition, we also discussed the potential role of GRP78 as a prognostic biomarker and therapeutic target for lung cancer, which is conducive to improving the assessment of lung cancer and the development of new therapeutic interventions.
Collapse
Affiliation(s)
- Shengkai Xia
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, No. 467 Zhongshan Road, Dalian, 116023, China
| | - Wenzhe Duan
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, No. 467 Zhongshan Road, Dalian, 116023, China
| | - Wenwen Liu
- Cancer Translational Medicine Research Center, The Second Hospital, Dalian Medical University, Dalian, 116023, China
| | - Xinri Zhang
- Department of Respiratory and Critical Care Medicine, The First Hospital, Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China.
| | - Qi Wang
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, No. 467 Zhongshan Road, Dalian, 116023, China. .,Cancer Translational Medicine Research Center, The Second Hospital, Dalian Medical University, Dalian, 116023, China.
| |
Collapse
|
47
|
Chen X, Cubillos-Ruiz JR. Endoplasmic reticulum stress signals in the tumour and its microenvironment. Nat Rev Cancer 2021; 21:71-88. [PMID: 33214692 PMCID: PMC7927882 DOI: 10.1038/s41568-020-00312-2] [Citation(s) in RCA: 764] [Impact Index Per Article: 191.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/13/2020] [Indexed: 02/06/2023]
Abstract
Protein handling, modification and folding in the endoplasmic reticulum (ER) are tightly regulated processes that determine cell function, fate and survival. In several tumour types, diverse oncogenic, transcriptional and metabolic abnormalities cooperate to generate hostile microenvironments that disrupt ER homeostasis in malignant and stromal cells, as well as infiltrating leukocytes. These changes provoke a state of persistent ER stress that has been demonstrated to govern multiple pro-tumoural attributes in the cancer cell while dynamically reprogramming the function of innate and adaptive immune cells. Aberrant activation of ER stress sensors and their downstream signalling pathways have therefore emerged as key regulators of tumour growth and metastasis as well as response to chemotherapy, targeted therapies and immunotherapy. In this Review, we discuss the physiological inducers of ER stress in the tumour milieu, the interplay between oncogenic signalling and ER stress response pathways in the cancer cell and the profound immunomodulatory effects of sustained ER stress responses in tumours.
Collapse
Affiliation(s)
- Xi Chen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
| | - Juan R Cubillos-Ruiz
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA.
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
48
|
Huang J, Pan H, Wang J, Wang T, Huo X, Ma Y, Lu Z, Sun B, Jiang H. Unfolded protein response in colorectal cancer. Cell Biosci 2021; 11:26. [PMID: 33514437 PMCID: PMC7844992 DOI: 10.1186/s13578-021-00538-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 01/11/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is a gastrointestinal malignancy originating from either the colon or the rectum. A growing number of researches prove that the unfolded protein response (UPR) is closely related to the occurrence and progression of colorectal cancer. The UPR has three canonical endoplasmic reticulum (ER) transmembrane protein sensors: inositol requiring kinase 1 (IRE1), pancreatic ER eIF2α kinase (PERK), and activating transcription factor 6 (ATF6). Each of the three pathways is closely associated with CRC development. The three pathways are relatively independent as well as interrelated. Under ER stress, the activated UPR boosts the protein folding capacity to maximize cell adaptation and survival, whereas sustained or excessive ER triggers cell apoptosis conversely. The UPR involves different stages of CRC pathogenesis, promotes or hinders the progression of CRC, and will pave the way for novel therapeutic and diagnostic approaches. Meanwhile, the correlation between different signal branches in UPR and the switch between the adaptation and apoptosis pathways still need to be further investigated in the future.
Collapse
Affiliation(s)
- Jingjing Huang
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Huayang Pan
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Jinge Wang
- The Second Affiliated Hospital & College of Nursing, Harbin Medical University, Harbin, People's Republic of China
| | - Tong Wang
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Xiaoyan Huo
- Pediatrics Department of The First Affiliated Hospital, Harbin Medical University, Harbin, People's Republic of China
| | - Yong Ma
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Zhaoyang Lu
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Bei Sun
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Hongchi Jiang
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, People's Republic of China.
| |
Collapse
|
49
|
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]
|
50
|
Gopal U, Pizzo SV. Cell surface GRP78 signaling: An emerging role as a transcriptional modulator in cancer. J Cell Physiol 2020; 236:2352-2363. [PMID: 32864780 DOI: 10.1002/jcp.30030] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/07/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022]
Abstract
Cancer cells acquire dysregulated gene expression to establish specific transcriptional dependencies and their underlying mechanisms that are ultimately responsible for this addictions have not been fully elucidated. Glucose-regulated protein 78 (GRP78) is a stress-inducible, multifunctional, prosurvival, endoplasmic reticulum chaperone in the heat shock protein 70 family. Expression of cell surface GRP78 (CS-GRP78) is associated with increased malignant behavior and resistance to chemotherapy and radiotherapy by endowing various cancer cells with increased proliferative ability, altered metabolism, improved survival, and augmented invasive and metastatic potential. Emerging evidence has highlighted an unusual role of CS-GRP78 in regulating transcription factors (TFs) by mediating various signaling pathways involved in malignant transformation, metabolic reprogramming, and tumor progression. During the last decade, we targeted CS-GRP78 with C38 monoclonal antibody (C38 Mab) in numerous studies, which have highlighted the epigenetic interplay between CS-GRP78 and various TFs including c-MYC, Yes-associated protein/transcriptional coactivator with PDZ-binding motif, c-Fos, and histone acetylation to potentiate subsequent modulation of tumorigenesis, invasion, and metastasis. Here, we summarize the current state of knowledge about the role of CS-GRP78 in cancer development and progression, including epigenetic regulation and sheds light on CS-GRP78 as vulnerable target for cancer therapy. Overall, this review focuses on the mechanisms of TFs that are behind the transcriptional dysregulation in cancer and lays the groundwork for rational therapeutic use of C38 Mab based on CS-GRP78 biology.
Collapse
Affiliation(s)
- Udhayakumar Gopal
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Salvatore V Pizzo
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|