1
|
Petrosyan E, Fares J, Fernandez LG, Yeeravalli R, Dmello C, Duffy JT, Zhang P, Lee-Chang C, Miska J, Ahmed AU, Sonabend AM, Balyasnikova IV, Heimberger AB, Lesniak MS. Endoplasmic Reticulum Stress in the Brain Tumor Immune Microenvironment. Mol Cancer Res 2023; 21:389-396. [PMID: 36652630 PMCID: PMC10159901 DOI: 10.1158/1541-7786.mcr-22-0920] [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] [Revised: 01/05/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023]
Abstract
Immunotherapy has emerged as a powerful strategy for halting cancer progression. However, primary malignancies affecting the brain have been exempt to this success. Indeed, brain tumors continue to portend severe morbidity and remain a globally lethal disease. Extensive efforts have been directed at understanding how tumor cells survive and propagate within the unique microenvironment of the central nervous system (CNS). Cancer genetic aberrations and metabolic abnormalities provoke a state of persistent endoplasmic reticulum (ER) stress that in turn promotes tumor growth, invasion, therapeutic resistance, and the dynamic reprogramming of the infiltrating immune cells. Consequently, targeting ER stress is a potential therapeutic approach. In this work, we provide an overview of how ER stress response is advantageous to brain tumor development, discuss the significance of ER stress in governing antitumor immunity, and put forth therapeutic strategies of regulating ER stress to augment the effect of immunotherapy for primary CNS tumors.
Collapse
Affiliation(s)
- Edgar Petrosyan
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Jawad Fares
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Luis G. Fernandez
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Ragini Yeeravalli
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Crismita Dmello
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Joseph T. Duffy
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Peng Zhang
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Catalina Lee-Chang
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Jason Miska
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Atique U. Ahmed
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Adam M. Sonabend
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Irina V. Balyasnikova
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Amy B. Heimberger
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Maciej S. Lesniak
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| |
Collapse
|
2
|
Comparative Transcriptomics and Proteomics of Cancer Cell Lines Cultivated by Physiological and Commercial Media. Biomolecules 2022; 12:biom12111575. [DOI: 10.3390/biom12111575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/21/2022] [Accepted: 10/20/2022] [Indexed: 11/16/2022] Open
Abstract
Aiming to reduce the gap between in vitro and in vivo environment, a complex culture medium, Plasmax, was introduced recently, which includes nutrients and metabolites with concentrations normally found in human plasma. Herein, to study the influence of this medium on cellular behaviors, we utilized Plasmax to cultivate two cancer cell lines, including one breast cancer cell line, MDA-MB-231BR, and one brain cancer cell line, CRL-1620. Cancer cells were harvested and prepared for transcriptomics and proteomics analyses to assess the discrepancies caused by the different nutritional environments of Plasmax and two commercial media: DMEM, and EMEM. Total RNAs of cells were extracted using mammalian total RNA extract kits and analyzed by next-generation RNA sequencing; proteomics analyses were performed using LC-MS/MS. Gene oncology and pathway analysis were employed to study the affected functions. The cellular invasion and cell death were inhibited in MDA-MB-231BR cell line when cultured in Plasmax compared to DMEM and EMEM, whereas the invasion, migration and protein synthesis of CRL-1620 cell line were activated in Plasmax in relative to both commercial media. The expression changes of some proteins were more significant compared to their corresponding transcripts, indicating that Plasmax has more influence upon regulatory processes of proteins after translation. This work provides complementary information to the original study of Plasmax, aiming to facilitate the selection of appropriate media for in vitro cancer cell studies.
Collapse
|
3
|
Ziko L, AbdelRaheem O, Nabil M, Aziz RK, Siam R. Bioprospecting the microbiome of Red Sea Atlantis II brine pool for peptidases and biosynthetic genes with promising antibacterial activity. Microb Cell Fact 2022; 21:109. [PMID: 35655185 PMCID: PMC9161539 DOI: 10.1186/s12934-022-01835-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/05/2022] [Indexed: 11/23/2022] Open
Abstract
Background The search for novel antimicrobial agents is crucial as antibiotic-resistant pathogens continue to emerge, rendering the available antibiotics no longer effective. Likewise, new anti-cancer drugs are needed to combat the emergence of multi-drug resistant tumors. Marine environments are wealthy sources for natural products. Additionally, extreme marine environments are interesting niches to search for bioactive natural compounds. In the current study, a fosmid library of metagenomic DNA isolated from Atlantis II Deep Lower Convective Layer (ATII LCL), was functionally screened for antibacterial activity as well as anticancer effects. Results Two clones exhibited antibacterial effects against the marine Bacillus Cc6 strain, namely clones 102-5A and 88-1G and they were further tested against eleven other challenging strains, including six safe relatives of ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp.), a safe relative to Mycobacterium tuberculosis and four resistant clinical isolates. Clone 88-1G resulted in clear zones of inhibition against eight bacterial strains, while clone 102-5A resulted in zones of inhibition against five bacterial strains. The whole cell lysates of clone 88-1G showed 15% inhibition of Mtb ClpP protease -Mycobacterium tuberculosis drug target-, while whole cell lysates of clone 102-5A showed 19% inhibition of Mtb ClpP protease. Whole cell lysates from the selected clones exhibited anticancer effects against MCF-7 breast cancer cells (cell viability at 50% v/v was 46.2% ± 9.9 for 88-1G clone and 38% ± 7 for 102-5A clone), U2OS osteosarcoma cells (cell viability at 50% v/v was 64.6% ± 12.3 for 88-1G clone and 28.3% ± 1.7 for 102-5A clone) and 1BR hTERT human fibroblast cells (cell viability at 50% v/v was 74.4% ± 5.6 for 88-1G clone and 57.6% ± 8.9 for 102-5A clone). Sequencing of 102-5A and 88-1G clones, and further annotation detected putative proteases and putative biosynthetic genes in clones 102-5A and 88-1G, respectively. Conclusions The ATII LCL metagenome hosts putative peptidases and biosynthetic genes that confer antibiotic and anti-cancer effects. The tested clones exhibited promising antibacterial activities against safe relative strains to ESKAPE pathogens and Mycobacterium tuberculosis. Thus, searching the microbial dark matter of extreme environments is a promising approach to identify new molecules with pharmaceutical potential use. Supplementary Information The online version contains supplementary material available at 10.1186/s12934-022-01835-z.
Collapse
Affiliation(s)
- Laila Ziko
- School of Life and Medical Sciences, University of Hertfordshire, Hosted by Global Academic Foundation, 11865, New Administrative Capital, Egypt.,Department of Biology, School of Sciences and Engineering, The American University in Cairo, 11835, New Cairo, Egypt
| | - Omnia AbdelRaheem
- Graduate Program of Biotechnology, School of Sciences and Engineering, The American University in Cairo, 11835, New Cairo, Egypt
| | - Marina Nabil
- Graduate Program of Biotechnology, School of Sciences and Engineering, The American University in Cairo, 11835, New Cairo, Egypt
| | - Ramy K Aziz
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt.,Microbiology and Immunology Research Program, Children's Cancer Hospital Egypt 57357, 11617, Cairo, Egypt
| | - Rania Siam
- Department of Biology, School of Sciences and Engineering, The American University in Cairo, 11835, New Cairo, Egypt. .,University of Medicine and Health Sciences, Basseterre, West Indies, Saint Kitts and Nevis.
| |
Collapse
|
4
|
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
|
5
|
Zuo Q, Ou Y, Zhong S, Yu H, Zhan F, Zhang M. Targeting GRP78 enhances the sensitivity of HOS osteosarcoma cells to pyropheophorbide-α methyl ester-mediated photodynamic therapy via the Wnt/β-catenin signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2021; 53:1387-1397. [PMID: 34494093 PMCID: PMC8507956 DOI: 10.1093/abbs/gmab115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Indexed: 12/20/2022] Open
Abstract
Photodynamic therapy (PDT), which is a new method for treating tumors, has been used in the treatment of cancer. In-depth research has shown that PDT cannot completely kill tumor cells, indicating that tumor cells are resistant to PDT. Glucose regulatory protein 78 (GRP78), which is a key regulator of endoplasmic reticulum stress, has been confirmed to be related to tumor resistance and recurrence, but there are relatively few studies on the further mechanism of GRP78 in PDT. Our experiment aimed to observe the role of GRP78 in HOS human osteosarcoma cells treated with pyropheophorbide-α methyl ester-mediated photodynamic therapy (MPPα-PDT) and to explore the possible mechanism by which the silencing of GRP78 expression enhances the sensitivity of HOS osteosarcoma cells to MPPα-PDT. HOS osteosarcoma cells were transfected with siRNA-GRP78. Apoptosis and reactive oxygen species (ROS) levels were detected by Hoechst staining and flow cytometry, cell viability was detected by Cell Counting Kit-8 assay, GRP78 protein fluorescence intensity was detected by immunofluorescence, and apoptosis-related proteins, cell proliferation-related proteins, and Wnt pathway-related proteins were detected by western blot. The results showed that MPPα-PDT can induce HOS cell apoptosis and increase GRP78 expression. After successful siRNA-GRP78 transfection, HOS cell proliferation was decreased, and apoptosis-related proteins expressions was increased, Wnt/β-catenin-related proteins expressions was decreased, and ROS levels was increased. In summary, siRNA-GRP78 enhances the sensitivity of HOS cells to MPPα-PDT, the mechanism may be related to inhibiting Wnt pathway activation and increasing ROS levels.
Collapse
Affiliation(s)
- Qiang Zuo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yunsheng Ou
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Shenxi Zhong
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Haoyang Yu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fangbiao Zhan
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Muzi Zhang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Zhang ZJ, Wang KP, Mo JG, Xiong L, Wen Y. Photodynamic therapy regulates fate of cancer stem cells through reactive oxygen species. World J Stem Cells 2020; 12:562-584. [PMID: 32843914 PMCID: PMC7415247 DOI: 10.4252/wjsc.v12.i7.562] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/17/2020] [Accepted: 05/21/2020] [Indexed: 02/06/2023] Open
Abstract
Photodynamic therapy (PDT) is an effective and promising cancer treatment. PDT directly generates reactive oxygen species (ROS) through photochemical reactions. This oxygen-dependent exogenous ROS has anti-cancer stem cell (CSC) effect. In addition, PDT may also increase ROS production by altering metabolism, endoplasmic reticulum stress, or potential of mitochondrial membrane. It is known that the half-life of ROS in PDT is short, with high reactivity and limited diffusion distance. Therefore, the main targeting position of PDT is often the subcellular localization of photosensitizers, which is helpful for us to explain how PDT affects CSC characteristics, including differentiation, self-renewal, apoptosis, autophagy, and immunogenicity. Broadly speaking, excess ROS will damage the redox system and cause oxidative damage to molecules such as DNA, change mitochondrial permeability, activate unfolded protein response, autophagy, and CSC resting state. Therefore, understanding the molecular mechanism by which ROS affect CSCs is beneficial to improve the efficiency of PDT and prevent tumor recurrence and metastasis. In this article, we review the effects of two types of photochemical reactions on PDT, the metabolic processes, and the biological effects of ROS in different subcellular locations on CSCs.
Collapse
Affiliation(s)
- Zi-Jian Zhang
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Kun-Peng Wang
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, Zhejiang Province, China
| | - Jing-Gang Mo
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, Zhejiang Province, China
| | - Li Xiong
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Yu Wen
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China.
| |
Collapse
|
9
|
Nam SM, Jeon YJ. Proteostasis In The Endoplasmic Reticulum: Road to Cure. Cancers (Basel) 2019; 11:E1793. [PMID: 31739582 PMCID: PMC6895847 DOI: 10.3390/cancers11111793] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/04/2019] [Accepted: 11/12/2019] [Indexed: 12/14/2022] Open
Abstract
The endoplasmic reticulum (ER) is an interconnected organelle that is responsible for the biosynthesis, folding, maturation, stabilization, and trafficking of transmembrane and secretory proteins. Therefore, cells evolve protein quality-control equipment of the ER to ensure protein homeostasis, also termed proteostasis. However, disruption in the folding capacity of the ER caused by a large variety of pathophysiological insults leads to the accumulation of unfolded or misfolded proteins in this organelle, known as ER stress. Upon ER stress, unfolded protein response (UPR) of the ER is activated, integrates ER stress signals, and transduces the integrated signals to relive ER stress, thereby leading to the re-establishment of proteostasis. Intriguingly, severe and persistent ER stress and the subsequently sustained unfolded protein response (UPR) are closely associated with tumor development, angiogenesis, aggressiveness, immunosuppression, and therapeutic response of cancer. Additionally, the UPR interconnects various processes in and around the tumor microenvironment. Therefore, it has begun to be delineated that pharmacologically and genetically manipulating strategies directed to target the UPR of the ER might exhibit positive clinical outcome in cancer. In the present review, we summarize recent advances in our understanding of the UPR of the ER and the UPR of the ER-mitochondria interconnection. We also highlight new insights into how the UPR of the ER in response to pathophysiological perturbations is implicated in the pathogenesis of cancer. We provide the concept to target the UPR of the ER, eventually discussing the potential of therapeutic interventions for targeting the UPR of the ER for cancer treatment.
Collapse
Affiliation(s)
- Su Min Nam
- Department of Biochemistry, Chungnam National University College of Medicine, Daejeon 35015, Korea;
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea
| | - Young Joo Jeon
- Department of Biochemistry, Chungnam National University College of Medicine, Daejeon 35015, Korea;
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea
| |
Collapse
|
10
|
Gallardo-Villagrán M, Leger DY, Liagre B, Therrien B. Photosensitizers Used in the Photodynamic Therapy of Rheumatoid Arthritis. Int J Mol Sci 2019; 20:E3339. [PMID: 31284664 PMCID: PMC6651633 DOI: 10.3390/ijms20133339] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 12/29/2022] Open
Abstract
Photodynamic Therapy (PDT) has become one of the most promising treatment against autoimmune diseases, such as rheumatoid arthritis (RA), as well as in the treatment of different types of cancer, since it is a non-invasive method and easy to carry out. The three main ingredients of PDT are light irradiation, oxygen, and a photosensitizer (PS). Light irradiation depends on the type of molecule or compound to be used as a PS. The concentration of O2 fluctuates according to the medium where the target tissue is located and over time, although it is known that it is possible to provide oxygenated species to the treated area through the PS itself. Finally, each PS has its own characteristics, the efficacy of which depends on multiple factors, such as solubility, administration technique, retention time, stability, excitation wavelength, biocompatibility, and clearance, among others. Therefore, it is essential to have a thorough knowledge of the disease to select the best PS for a specific target, such as RA. In this review we will present the PSs used in the last three decades to treat RA under PDT protocol, as well as insights on the relevant strategies.
Collapse
Affiliation(s)
- Manuel Gallardo-Villagrán
- Laboratoire PEIRENE, Faculté de Pharmacie, Université de Limoges, EA 7500, F-87025 Limoges, France
- Institut de Chimie, Université de Neuchâtel, Avenue de Bellevaux 51, CH-2000 Neuchâtel, Switzerland
| | - David Yannick Leger
- Laboratoire PEIRENE, Faculté de Pharmacie, Université de Limoges, EA 7500, F-87025 Limoges, France.
| | - Bertrand Liagre
- Laboratoire PEIRENE, Faculté de Pharmacie, Université de Limoges, EA 7500, F-87025 Limoges, France.
| | - Bruno Therrien
- Institut de Chimie, Université de Neuchâtel, Avenue de Bellevaux 51, CH-2000 Neuchâtel, Switzerland.
| |
Collapse
|
11
|
Ziko L, Saqr AHA, Ouf A, Gimpel M, Aziz RK, Neubauer P, Siam R. Antibacterial and anticancer activities of orphan biosynthetic gene clusters from Atlantis II Red Sea brine pool. Microb Cell Fact 2019; 18:56. [PMID: 30885206 PMCID: PMC6423787 DOI: 10.1186/s12934-019-1103-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 03/06/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cancer and infectious diseases are problematic because of continuous emergence of drug resistance. One way to address this enormous global health threat is bioprospecting the unlikeliest environments, such as extreme marine niches, which have tremendous biodiversity that is barely explored. One such environment is the Red Sea brine pool, Atlantis II Deep (ATII). Here, we functionally screened a fosmid library of metagenomic DNA isolated from the ATII lower convective layer (LCL) for antibacterial and anticancer activities. RESULTS Selected clones, 14-7E and 10-2G, displayed antibacterial effects on the marine strain Bacillus sp. Cc6. Moreover, whole cell lysates from 14-7E and 10-2G exhibited decreased cell viability against MCF-7 (39.1% ± 6.6, 42% ± 8.1 at 50% v/v) and U2OS cells (35.7% ± 1.9, 79.9% ± 5.9 at 50% v/v), respectively. By sequencing the insert DNA from 14-7E and 10-2G, we identified two putative orphan biosynthetic gene clusters. Both clusters harbored putative ATP-binding cassette (ABC) transporter permeases and S-adenosylmethionine-related genes. Interestingly, the biosynthetic gene cluster identified on 14-7E is of archaeal origin and harbors a putative transcription factor. Several identified genes may be responsible for the observed antibacterial and anticancer activities. The 14-7E biosynthetic gene cluster may be encoding enzymes producing a specialized metabolite (effect of detected genes involved in C-C bond formation and glycosylation). The bioactivity may also be due to predicted subtilases encoded by this cluster. The 10-2G cluster harbored putative glycosyltransferase and non-ribosomal peptide synthase genes; thus the observed activity of this clone could be caused by a bioactive peptide. CONCLUSIONS The ATII LCL prokaryotic metagenome hosts putative orphan biosynthetic gene clusters that confer antibiotic and anticancer effects. Further biochemical studies should characterize the detected bioactive components, and the potential use of 14-7E metabolite for antibiosis and 10-2G metabolite as a selective anti-breast cancer drug.
Collapse
Affiliation(s)
- Laila Ziko
- Graduate Program of Biotechnology, School of Sciences and Engineering, The American University in Cairo, New Cairo, Cairo, 11835, Egypt
| | - Al-Hussein A Saqr
- Department of Biology, School of Sciences and Engineering, The American University in Cairo, SSE (Parcel 7), Second Floor, Office: Room 2194, AUC Avenue, New Cairo, Cairo, 11835, Egypt
| | - Amged Ouf
- Graduate Program of Biotechnology, School of Sciences and Engineering, The American University in Cairo, New Cairo, Cairo, 11835, Egypt
- Department of Biology, School of Sciences and Engineering, The American University in Cairo, SSE (Parcel 7), Second Floor, Office: Room 2194, AUC Avenue, New Cairo, Cairo, 11835, Egypt
| | - Matthias Gimpel
- Chair of Bioprocess Engineering, Department of Biotechnology, Technische Universität Berlin, Ackerstrasse 76, ACK24, 13355, Berlin, Germany
| | - Ramy K Aziz
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Peter Neubauer
- Chair of Bioprocess Engineering, Department of Biotechnology, Technische Universität Berlin, Ackerstrasse 76, ACK24, 13355, Berlin, Germany
| | - Rania Siam
- Graduate Program of Biotechnology, School of Sciences and Engineering, The American University in Cairo, New Cairo, Cairo, 11835, Egypt.
- Department of Biology, School of Sciences and Engineering, The American University in Cairo, SSE (Parcel 7), Second Floor, Office: Room 2194, AUC Avenue, New Cairo, Cairo, 11835, Egypt.
| |
Collapse
|
12
|
Dailton Guedes de Oliveira Moraes C, Henrique Godoi B, Chaves Silva Carvalho I, Cristina Pinto J, Carvalho Rossato R, Soares da Silva N, Pacheco Soares C. Genotoxic effects of photodynamic therapy in laryngeal cancer cells - An in vitro study. Exp Biol Med (Maywood) 2019; 244:262-271. [PMID: 30674213 DOI: 10.1177/1535370219826544] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
IMPACT STATEMENT Recently, the use of photodynamic therapy grows as an alternative treatment for cancer, since it has a noninvasive characteristic and affinity to the tumor tissue. Accordingly, understanding the therapy's foci of action is important for the technique improvement. This work aims to understand the genotoxic effect triggered by the therapy action, thus evidencing the permanent changes caused to the genetic material of the tumor cell after the treatment. Therefore, to increase the knowledge in this study field, the methodology of the comet assay and count of micronucleus formed after the therapy was adopted in order to understand if the damage caused to the DNA of tumor cell makes its replication process unfeasible in future generations. The study allows a better therapeutic approach to the cancer treatment, making the process of association between therapies a more effective option during the disease treatment.
Collapse
Affiliation(s)
- Carlos Dailton Guedes de Oliveira Moraes
- 1 Institute of Research and Development - IP&D, Universidade do Vale do Paraíba - UNIVAP, Laboratory Dynamics of Cellular Compartments, Sao Paulo 12244-000, Brazil
| | - Bruno Henrique Godoi
- 1 Institute of Research and Development - IP&D, Universidade do Vale do Paraíba - UNIVAP, Laboratory Dynamics of Cellular Compartments, Sao Paulo 12244-000, Brazil
| | - Isabel Chaves Silva Carvalho
- 1 Institute of Research and Development - IP&D, Universidade do Vale do Paraíba - UNIVAP, Laboratory Dynamics of Cellular Compartments, Sao Paulo 12244-000, Brazil
| | - Jessica Cristina Pinto
- 1 Institute of Research and Development - IP&D, Universidade do Vale do Paraíba - UNIVAP, Laboratory Dynamics of Cellular Compartments, Sao Paulo 12244-000, Brazil
| | - Rafaella Carvalho Rossato
- 1 Institute of Research and Development - IP&D, Universidade do Vale do Paraíba - UNIVAP, Laboratory Dynamics of Cellular Compartments, Sao Paulo 12244-000, Brazil
| | - Newton Soares da Silva
- 2 Institute of Research and Development - IP&D, Universidade do Vale do Paraíba - UNIVAP, Laboratory of Cell Biology and Tissue, Sao Paulo, CEP 12244-000, Brazil
| | - Cristina Pacheco Soares
- 1 Institute of Research and Development - IP&D, Universidade do Vale do Paraíba - UNIVAP, Laboratory Dynamics of Cellular Compartments, Sao Paulo 12244-000, Brazil
| |
Collapse
|
13
|
Clarke WR, Amundadottir L, James MA. CLPTM1L/CRR9 ectodomain interaction with GRP78 at the cell surface signals for survival and chemoresistance upon ER stress in pancreatic adenocarcinoma cells. Int J Cancer 2019; 144:1367-1378. [PMID: 30468251 DOI: 10.1002/ijc.32012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 09/24/2018] [Accepted: 11/09/2018] [Indexed: 12/15/2022]
Abstract
Altered regulation of endoplasmic reticulum (ER) homeostasis has been implicated in many cancers and has recently become a therapeutic and chemosensitization target of interest. We have identified Cleft Lip and Palate Transmembrane 1-Like (CLPTM1L)/Cisplatin Resistance Related Protein 9 (CRR9) as an ER stress related mediator of cytoprotection in pancreatic cancer. We recently demonstrated that CLPTM1L is highly expressed in pancreatic ductal adenocarcinoma and associated with poor outcome. Furthermore, we have discovered that CLPTM1L interacts with phosphoinositol-3-kinase-alpha at the tumor cell surface and causes up-regulation of Bcl-xL and pAkt mediated survival signaling. Here, we demonstrate surface relocalization and survival signaling by CLPTM1L triggered by endoplasmic reticular (ER) stress. We demonstrate the interaction of CLPTM1L with the central ER stress survival mediator, Glucose Regulated Protein 78 (GRP78)/Binding Immunoglobulin Protein (BiP) and PI3K-alpha /p110α. This interaction and surface relocalization of CLPTM1L and GRP78 is induced by ER stress, including that caused by treatment with gemcitabine. We demonstrate that the extracellular loop of CLPTM1L is required for gemcitabine resistance and interaction with GRP78. This interaction and the chemoresistance effect conferred by this pathway is targetable with our recently developed inhibitory CLPTM1L antibodies, which may represent novel modalities of chemosensitization and treatment of pancreatic adenocarcinoma. Anchorage independent growth, GRP78-mediated chemoresistance, and Akt phosphorylation were abrogated by inhibition of CLPTM1L. These findings demonstrate a novel and potentially targetable mechanism of cytoprotection and chemoresistance in pancreatic tumors.
Collapse
Affiliation(s)
- William R Clarke
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Laufey Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Gaithersburg, Maryland, USA
| | - Michael A James
- Department of Surgery, Division of Research, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
14
|
Gkouveris I, Nikitakis NG, Aseervatham J, Ogbureke KUE. Interferon γ suppresses dentin sialophosphoprotein in oral squamous cell carcinoma cells resulting in antitumor effects, via modulation of the endoplasmic reticulum response. Int J Oncol 2018; 53:2423-2432. [PMID: 30320380 PMCID: PMC6203152 DOI: 10.3892/ijo.2018.4590] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 09/19/2018] [Indexed: 12/15/2022] Open
Abstract
The expression of proinflammatory cytokines in various malignant neoplasms is widely considered to represent the host immune response to tumor development. The role of interferon (IFN)γ in head and neck squamous cell carcinoma, and its association with endoplasmic reticulum (ER) stress pathways, remains a subject of ongoing investigation. Dentin sialophosphoprotein (DSPP), which is a member of the small integrin-binding N-linked glycoproteins family, has been implicated in malignant transformation and invasion of oral squamous cell carcinoma (OSCC). Recent studies have established matrix metalloproteinase (MMP)20 as the cognate MMP partner of DSPP. The present study examined the effects of IFNγ treatment on DSPP and MMP20 expression, ER stress, the unfolded protein response (UPR), and calcium (Ca) homeostasis regulatory mechanisms in OSCC cells. The OSC2 OSCC cell line was treated with IFNγ at specific time-points. At each time-point, the mRNA expression levels of DSPP and MMP20, and those of ER-stress-, UPR- and Ca homeostasis-associated proteins [78-kDa glucose-regulated protein (GRP78), sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2b), inositol 1,4,5-trisphosphate receptor (IP3r), protein kinase R-like ER kinase (PERK) and inositol-requiring enzyme 1 (IRE1)], were assessed by reverse transcription-quantitative polymerase chain reaction. The protein expression levels of B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (Bax), proliferating cell nuclear antigen (PCNA) and cytochrome c were analyzed by western blotting. Cell viability, apoptosis and migration were evaluated by MTT, Annexin V-fluorescein isothiocyanate flow cytometry and wound-healing assays, respectively. IFNγ treatment significantly downregulated the mRNA expression levels of the major ER stress regulator GRP78 and, to a lesser extent, the UPR-associated molecule IRE1; however, IFNγ had no significant effect on PERK. With regards to ER Ca homeostasis molecules, treatment with IFNγ downregulated the mRNA expression levels of SERCA2b and upregulated those of IP3r. Furthermore, DSPP and MMP20 mRNA expression levels were significantly reduced following IFNγ treatment. Notably, treatment with IFNγ hampered OSC2 migration, reduced cell viability and PCNA protein expression, enhanced apoptosis, downregulated Bcl-2, and upregulated Bax and cytochrome c. Overall, IFNγ inhibited OSCC cell viability and migration, and increased apoptosis, possibly by regulating ER stress and UPR mechanisms. In addition, IFNγ-induced DSPP and MMP20 downregulation may correspond with alteration in ER Ca homeostasis.
Collapse
Affiliation(s)
- Ioannis Gkouveris
- Division of Diagnostic and Surgical Sciences, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Nikolaos G Nikitakis
- Department of Oral Medicine and Pathology, School of Dentistry, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Jaya Aseervatham
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Kalu U E Ogbureke
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| |
Collapse
|
15
|
Ma B, Zhang H, Wang Y, Zhao A, Zhu Z, Bao X, Sun Y, Li L, Zhang Q. Corosolic acid, a natural triterpenoid, induces ER stress-dependent apoptosis in human castration resistant prostate cancer cells via activation of IRE-1/JNK, PERK/CHOP and TRIB3. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:210. [PMID: 30176898 PMCID: PMC6122202 DOI: 10.1186/s13046-018-0889-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/17/2018] [Indexed: 02/06/2023]
Abstract
Background The development of potent non-toxic chemotherapeutic drugs against castration resistant prostate cancer (CRPC) remains a major challenge. Corosolic acid (CA), a natural triterpenoid, has anti-cancer activity with limited side effects. However, CA anti-prostate cancer activities and mechanisms, particularly in CRPC, are not clearly understood. In this study, we investigated CA anti-tumor ability against human CRPC and its mechanism of action. Methods The cell apoptosis and proliferation effects were evaluated via MTT detection, colony formation assay and flow cytometry. Western blot, gene transfection and immunofluorescence assay were applied to investigate related protein expression of Endoplasmic reticulum stress. A xenograft tumor model was established to investigate the inhibitory effect of CA on castration resistant prostate cancer in vivo. Results The results showed that CA inhibited cell growth and induced apoptosis in human prostate cancer cell (PCa) line PC-3 and DU145, as well as retarded tumor growth in a xenograft model, exerting a limited toxicity to normal cells and tissues. Importantly, CA activated endoplasmic reticulum (ER) stress-associated two pro-apoptotic signaling pathways, as evidenced by increased protein levels of typical ER stress markers including IRE-1/ASK1/JNK and PERK/eIF2α/ATF4/CHOP. IRE-1, PERK or CHOP knockdown partially attenuated CA cytotoxicity against PCa cells. Meanwhile, CHOP induced expression increased Tribbles 3 (TRIB3) level, which lead to AKT inactivation and PCa cell death. CHOP silencing resulted in PCa cells sensitive to CA-induced apoptosis. Conclusion Our data demonstrated, for the first time, that CA might represent a novel drug candidate for the development of an anti-CRPC therapy. Electronic supplementary material The online version of this article (10.1186/s13046-018-0889-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bo Ma
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Hang Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China.,Institute of Advanced Materials (IAM), Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Yu Wang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Ang Zhao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Zhiming Zhu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Xiaowen Bao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Yang Sun
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Lin Li
- Institute of Advanced Materials (IAM), Nanjing Tech University, Nanjing, 210009, People's Republic of China.
| | - Qi Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
16
|
Kim C, Kim B. Anti-Cancer Natural Products and Their Bioactive Compounds Inducing ER Stress-Mediated Apoptosis: A Review. Nutrients 2018; 10:nu10081021. [PMID: 30081573 PMCID: PMC6115829 DOI: 10.3390/nu10081021] [Citation(s) in RCA: 305] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/23/2018] [Accepted: 08/01/2018] [Indexed: 12/14/2022] Open
Abstract
Cancer is the second biggest cause of death worldwide. Despite a number of studies being conducted, the effective mechanism for treating cancer has not yet been fully understood. The tumor-microenvironment such as hypoxia, low nutrients could disturb function of endoplasmic reticulum (ER) to maintain cellular homeostasis, ultimately leading to the accumulation of unfolded proteins in ER, so-called ER stress. The ER stress has a close relation with cancer. ER stress initiates unfolded protein response (UPR) to re-establish ER homeostasis as an adaptive pathway in cancer. However, persistent ER stress triggers the apoptotic pathway. Therefore, blocking the adaptive pathway of ER stress or facilitating the apoptotic pathway could be an anti-cancer strategy. Recently, natural products and their derivatives have been reported to have anti-cancer effects via ER stress. Here, we address mechanisms of ER stress-mediated apoptosis and highlight strategies for cancer therapy by utilizing ER stress. Furthermore, we summarize anti-cancer activity of the natural products via ER stress in six major types of cancers globally (lung, breast, colorectal, gastric, prostate and liver cancer). This review deepens the understanding of ER stress mechanisms in major cancers as well as the suppressive impact of natural products against cancers via ER stress.
Collapse
Affiliation(s)
- Changmin Kim
- Department of Pathology, College of Korean Medicine, Graduate School, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Korea.
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Graduate School, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Korea.
| |
Collapse
|
17
|
Chen J, Huang JH, Wang Z, Song X, Chen Z, Zeng Q, Zhou X, Zuo Z, Zhao S, Chen X, Kang J. Endoplasmic reticulum stress-mediated autophagy contributes to 5-ethylamino-9-diethylaminobenzo[a]phenoselenazinium-mediated photodynamic therapy via the PERK-eIF2α pathway. Onco Targets Ther 2018; 11:4315-4325. [PMID: 30100737 PMCID: PMC6065466 DOI: 10.2147/ott.s163366] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Introduction 5-ethylamino-9-diethylaminobenzo[a]phenoselenazinium (EtNBSe) is a novel synthetic bipolar photosensitizer with many promising applications. This study investigated the impact of EtNBSe-mediated photodynamic therapy (EtNBSe-PDT) on the autophagy and endoplasmic reticulum (ER) stress of squamous carcinoma cells (A-431 cells), as well as the related molecular mechanisms. Methods The potency of EtNBSe-PDT against squamous cell carcinoma was evaluated in BALB/c nude mice. Cell viability was evaluated using MTT. Western blotting and immunofluorescence were used to determine the expression levels of ER stress- and autophagy-related proteins. Results Both morphological and microscopic findings showed that the tumor on the xenograft mice exhibited an apparent reduction in volume and was replaced with fibrosis 20 days after EtNBSe-PDT. Additionally, in an in vitro study using A-431 cells, EtNBSe-PDT was found to inhibit A-431 cell survival in an EtNBSe concentration- and light dose- dependent manner, and to induce ER stress via the PERK-eIF2α signaling pathway. Additionally, EtNBSe-PDT could also induce autophagy of A-431 cells. Furthermore, the ER stress inhibitor 4-PBA and the eIF2α inhibitor salubrinal were found to inhibit the autophagy induced by EtNBSe-PDT. Conclusion This study demonstrated that the PERK-eIF2α signaling pathway was involved in the ER stress induced by EtNBSe-PDT. Meanwhile, the ER stress via the PERK-eIF2α pathway promoted the occurrence of autophagy in A-431 cells.
Collapse
Affiliation(s)
- Jing Chen
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China,
| | - Jin-Hua Huang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China,
| | - Zhen Wang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China,
| | - Xiangzhi Song
- College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, People's Republic of China
| | - Zeyi Chen
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China,
| | - Qinghai Zeng
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China,
| | - Xiping Zhou
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China,
| | - Zhihong Zuo
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China,
| | - Shuang Zhao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China,
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China,
| | - Jian Kang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China,
| |
Collapse
|
18
|
Wang M, Law ME, Castellano RK, Law BK. The unfolded protein response as a target for anticancer therapeutics. Crit Rev Oncol Hematol 2018; 127:66-79. [DOI: 10.1016/j.critrevonc.2018.05.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/22/2018] [Accepted: 05/07/2018] [Indexed: 12/11/2022] Open
|
19
|
Zhu J, Tian S, Li KT, Chen Q, Jiang Y, Lin HD, Yu LH, Bai DQ. Inhibition of breast cancer cell growth by methyl pyropheophenylchlorin photodynamic therapy is mediated though endoplasmic reticulum stress-induced autophagy in vitro and vivo. Cancer Med 2018; 7:1908-1920. [PMID: 29577663 PMCID: PMC5943539 DOI: 10.1002/cam4.1418] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 01/13/2018] [Accepted: 02/04/2018] [Indexed: 12/18/2022] Open
Abstract
Autophagy and ER stress participated in the inhibition of MPPa‐PDT on tumor growth, but the molecular links between them remain undefined. We just explore the molecular mechanism between them in vitro and vivo. CCK‐8 assay and flow cytometer were used to detect the cytotoxicity and mode of cell death after MPPa‐PDT. Furthermore, the role of autophagy was verified in MPPa‐PDT. Confocal microscopy was used to show the intracellular distribution of MPPa. ER stress markers and PERK signaling pathway were detected by western blot. While in vivo, tumor histology and immunohistochemistry were performed to show the effect of MPPa‐PDT in mice. After MPPa‐PDT, cells viability decreased in dose‐dependent manner. Besides, the cell apoptosis increased along with the increasing of Beclin‐1and LC3B II but declining of P62. When pretreated with 3‐MA, LC3B II formation and the cytotoxicity declined. MPPa‐PDT caused increasing of ER stress markers (GRP78, CHOP) as MPPa accumulated in ER. However, pretreatment with ER stress inhibitor 4PBA, the expression of GRP78 and LC3B II was blocked but the PERK signaling pathway activated and the expression of P62 increased. In vivo, the tumor growth was significantly inhibited by MPPa‐PDT. Besides, the appearance of ER stress and autophagy was further demonstrated by immunohistochemistry. Our findings demonstrate that autophagy mediated by MPPa‐PDT was regulated by ER stress, via PERK signaling pathway, to kill MDA‐MB‐231 cells in vitro and vivo.
Collapse
Affiliation(s)
- Jiang Zhu
- Department of Rehabilitation, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Rehabilitation, Southwest University Hospital, Chongqing, China
| | - Si Tian
- Department of Rehabilitation, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kai-Ting Li
- Department of Rehabilitation, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qing Chen
- Department of Rehabilitation, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuan Jiang
- Department of Rehabilitation, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hai-Dan Lin
- Department of Rehabilitation, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Le-Hua Yu
- Department of Rehabilitation, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ding-Qun Bai
- Department of Rehabilitation, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
20
|
Bisdemethoxycurcumin exerts pro-apoptotic effects in human pancreatic adenocarcinoma cells through mitochondrial dysfunction and a GRP78-dependent pathway. Oncotarget 2018; 7:83641-83656. [PMID: 27845899 PMCID: PMC5347794 DOI: 10.18632/oncotarget.13272] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 10/14/2016] [Indexed: 12/27/2022] Open
Abstract
Pancreatic cancer is a highly aggressive malignancy, which is intrinsically resistant to current chemotherapies. Herein, we investigate whether bisdemethoxycurcumin (BDMC), a derivative of curcumin, potentiates gemcitabine in human pancreatic cancer cells. The result suggests that BDMC sensitizes gemcitabine by inducing mitochondrial dysfunctions and apoptosis in PANC-1 and MiaPaCa-2 pancreatic cancer cells. Utilizing two-dimensional gel electrophoresis and mass spectrometry, we identify 13 essential proteins with significantly altered expressions in response to gemcitabine alone or combined with BDMC. Protein-protein interaction network analysis pinpoints glucose-regulated protein 78 (GRP78) as the key hub activated by BDMC. We then reveal that BDMC upregulates GRP78 and facilitates apoptosis through eIF2α/CHOP pathway. Moreover, DJ-1 and prohibitin, two identified markers of chemoresistance, are increased by gemcitabine in PANC-1 cells. This could be meaningfully reversed by BDMC, suggesting that BDMC partially offsets the chemoresistance induced by gemcitabine. In summary, these findings show that BDMC promotes apoptosis through a GRP78-dependent pathway and mitochondrial dysfunctions, and potentiates the antitumor effect of gemcitabine in human pancreatic cancer cells.
Collapse
|
21
|
Li JJ, Zhang JJ, Wang X, Sun ZM. Effects of 17-DMAG on diffuse large B-cell lymphoma cell apoptosis. Exp Ther Med 2017; 14:3727-3731. [PMID: 29042970 PMCID: PMC5639270 DOI: 10.3892/etm.2017.4995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/20/2017] [Indexed: 12/26/2022] Open
Abstract
17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG) is a water soluble, semisynthetic derivative of endotoxin that has anticancer effects. The aim of the present study was to determine whether 17-DMAG enhances the apoptosis of lymphoma cells in diffuse large B-cell lymphoma. Apoptosis was induced in SU-DHL-4 diffuse large B-cell lymphoma cells treated with 17-DMAG, as evaluated by MTT assay and flow cytometry analysis. Apoptosis-associated protein levels were assessed using western blotting, and the results indicated that B-cell lymphoma 2 (Bcl-2)-associated protein X (Bax) was upregulated, whereas heat shock protein family A member 5 (HSPA5) and Bcl-2 were downregulated. Additionally, staining with 5,5',6,6'-Tetrachloro-1,1',3,3'-tetraethyl-imidacarbocyanine iodide revealed that treatment with 17-DMAG decreased mitochondrial membrane potential in SU-DHL-4 diffuse large B-cell lymphoma cells. These results suggested that 17-DMAG is able to inhibit proliferation in diffuse large B-cell lymphoma cells in a concentration-dependent manner. The underlying mechanism may be that 17-DMAG induces oxidative stress, which inhibits the expression of HSPA5 and Bcl-2 and promotes the expression of Bax, leading to the apoptosis of SU-DHL-4 cells. Taken together, these results indicated that 17-DMAG may be an effective novel agent for the treatment of diffuse large B-cell lymphoma.
Collapse
Affiliation(s)
- Jia-Jia Li
- Department of Hematology, Affiliated Provincial Hospital of Anhui Medical University, Hefei, Anhui 230001, P.R. China
| | - Jing-Jing Zhang
- Department of Hematology, Affiliated Provincial Hospital of Anhui Medical University, Hefei, Anhui 230001, P.R. China
| | - Xiu Wang
- Department of Hematology, Affiliated Provincial Hospital of Anhui Medical University, Hefei, Anhui 230001, P.R. China
| | - Zi-Min Sun
- Department of Hematology, Affiliated Provincial Hospital of Anhui Medical University, Hefei, Anhui 230001, P.R. China
| |
Collapse
|
22
|
Machihara K, Tanaka H, Hayashi Y, Murakami I, Namba T. Questiomycin A stimulates sorafenib-induced cell death via suppression of glucose-regulated protein 78. Biochem Biophys Res Commun 2017; 492:33-40. [PMID: 28811106 DOI: 10.1016/j.bbrc.2017.08.042] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 08/12/2017] [Indexed: 12/25/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most difficult cancers to treat owing to the lack of effective chemotherapeutic methods. Sorafenib, the first-line and only available treatment for HCC, extends patient overall survival by several months, with a response rate below 10%. Thus, the identification of an agent that enhances the anticancer effect of sorafenib is critical for the development of therapeutic options for HCC. Endoplasmic reticulum (ER) stress response is one of the methods of sorafenib-induced cell death. Here we report that questiomycin A suppresses expression of GRP78, a cell-protective ER chaperone protein. Analysis of the molecular mechanisms of questiomycin A revealed that this compound stimulated GRP78 protein degradation in an ER stress response-independent manner. Cotreatment with sorafenib and questiomycin A suppressed GRP78 protein expression, which is essential for the stimulation of sorafenib-induced cell death. Moreover, our in vivo study demonstrated that the coadministration of sorafenib and questiomycin A suppressed tumor formation in HCC-induced xenograft models. These results suggest that cotreatment with sorafenib and questiomycin A is a novel therapeutic strategy for HCC by enhancing sorafenib-dependent ER stress-induced cell death, and downregulation of GRP78 is a new target for the stimulation of the therapeutic effects of sorafenib in HCC.
Collapse
Affiliation(s)
- Kayo Machihara
- Science Research Center, Kochi University, Kochi 783-8505, Japan
| | - Hidenori Tanaka
- Science Research Center, Kochi University, Kochi 783-8505, Japan
| | - Yoshihiro Hayashi
- Equipment of Support Planning Office, Kochi University, Kochi 783-8505, Japan
| | - Ichiro Murakami
- Department of Pathology, Kochi Medical School, Kochi University, Kochi 783-8505, Japan
| | - Takushi Namba
- Science Research Center, Kochi University, Kochi 783-8505, Japan.
| |
Collapse
|
23
|
Gao F, Zheng W, Gao L, Cai P, Liu R, Wang Y, Yuan Q, Zhao Y, Gao X. Au Nanoclusters and Photosensitizer Dual Loaded Spatiotemporal Controllable Liposomal Nanocomposites Enhance Tumor Photodynamic Therapy Effect by Inhibiting Thioredoxin Reductase. Adv Healthc Mater 2017; 6. [PMID: 28322024 DOI: 10.1002/adhm.201601453] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/16/2017] [Indexed: 12/16/2022]
Abstract
Photodynamic therapy (PDT) is a minimally invasive therapeutic procedure of tumors with high selectivity and low side effect. However, it is usually not efficient in long-lasting tumor control. One of the main reasons is tumor cells develop some protective mechanisms that help them to deal with oxidative stress in the environment. The thioredoxin system in cancer is an important antioxidant defense system. Au nanoclusters could effectively inhibit thioredoxin reductase (TrxR) in tumor cell cytoplasm. Herein, Au nanoclusters and photosensitizer Chlorine 6 (Ce6) are co-loaded in spatiotemporal controllable liposomal nanocomposites. pH responsive molecule inserted in lipid bilayer greatly contributes to the instability of the lipid membrane in lysosomal at low pH environment. Then the payloads can rapidly release into cytoplasm. Au nanoclusters effectively inhibit TrxR in cytoplasm and enhance the photodynamic-induced intracellular reactive oxygen-free radical concentration, improving the effect of PDT. Breast cancer is chosen as a tumor model and the Au nanoclusters and photosensitizer co-loaded liposomal nanocomposites are studied to improve the effect of PDT both in vitro and in vivo, and its corresponding mechanism is investigated. This study develops a new application of gold nanoclusters and provides a new train of thoughts for enhancing the effect of PDT.
Collapse
Affiliation(s)
- Fuping Gao
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety; Institute of High Energy Physics; Chinese Academy of Sciences; Beijing 100049 P. R. China
| | - Weiping Zheng
- The First Affiliated Hospital of Guangxi University of Chinese Medicine; Nanning 530023 P. R. China
| | - Liang Gao
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety; Institute of High Energy Physics; Chinese Academy of Sciences; Beijing 100049 P. R. China
| | - Pengju Cai
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety; Institute of High Energy Physics; Chinese Academy of Sciences; Beijing 100049 P. R. China
| | - Ru Liu
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety; Institute of High Energy Physics; Chinese Academy of Sciences; Beijing 100049 P. R. China
| | - Yaling Wang
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety; Institute of High Energy Physics; Chinese Academy of Sciences; Beijing 100049 P. R. China
| | - Qing Yuan
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety; Institute of High Energy Physics; Chinese Academy of Sciences; Beijing 100049 P. R. China
| | - Yuliang Zhao
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety; Institute of High Energy Physics; Chinese Academy of Sciences; Beijing 100049 P. R. China
| | - Xueyun Gao
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety; Institute of High Energy Physics; Chinese Academy of Sciences; Beijing 100049 P. R. China
| |
Collapse
|
24
|
Ahmadi A, Khansarinejad B, Hosseinkhani S, Ghanei M, Mowla SJ. miR-199a-5p and miR-495 target GRP78 within UPR pathway of lung cancer. Gene 2017; 620:15-22. [PMID: 28363780 DOI: 10.1016/j.gene.2017.03.032] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 03/22/2017] [Accepted: 03/24/2017] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Non-small cell lung cancer (NSCLC) is the most common subtype of lung cancer. One of the signal transduction pathways related to NSCLC is Unfolded Protein Response (UPR), which is mainly regulated by GRP78 (HSPA5, Gene ID: 3309). The aim of this study was to employ bioinformatics tools to predict microRNAs (miRNAs) affecting GRP78 expression, experimentally validate interaction of these miRNAs with GRP78 and also evaluating the expression correlation of GRP78 and its predicted miRNAs in clinical samples. MATERIALS AND METHODS Various software were used to predict miRNAs that simultaneously target all upstream and downstream components of GRP78 in the UPR, as well as the main components of PI3K/AKT, MAPK, ErbB and calcium pathways. For experimental analysis, 36 pairs of Formalin-Fixed Paraffin-Embedded (FFPE) lung tumor and non-tumor tissue samples were obtained. Additionally, A549 and QU-DB lung cancer cell lines were used for expression determination of GRP78 and its predicted targeting miRNAs. We also employed a luciferase assay to evaluate interactions between candidate miRNAs with the 3'-UTR of GRP78. RESULTS hsa-miR-495 and hsa-miR-199-5p were chosen based on several criteria including thermodynamic binding features of miRNAs to the target transcripts, number of recognition sites, and conservation of binding sites within the 3'-UTR of GRP78. RT-qPCR data revealed a significant up-regulation of GRP78 (3.87 times, P=0.002) and down-regulation of miR-199a-5p (0.13 times, P=0.0001) and miR-495 (0.085 times, P=0.0001) in tumor samples. Luciferase assay confirmed an interaction of hsa-miR-199a-5p and hsa-miR-495 with the 3'-UTR of GRP78 transcript. In addition, over-expression and competitive inhibition of the aforementioned miRNAs, significantly altered the expression of GRP78 and spliced XBP1 level. CONCLUSION Our data revealed a significant up-regulation of GRP78 and a concomitant down-regulation of miR-495 and miR-199a-5p in NSCLC. Accordingly, our data suggest a causative role for miR-199-5p and miR-495 in tumorgenesis of lung and probably other cancer types.
Collapse
Affiliation(s)
- Azam Ahmadi
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Behzad Khansarinejad
- Infectious Diseases Research Center (IDRC), Department of Microbiology and Immunology, Arak University of Medical Sciences, Arak, Iran
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mostafa Ghanei
- Research Center of Chemical Injuries, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Seyed Javad Mowla
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
25
|
Patel MR, Kozuch SD, Cultrara CN, Yadav R, Huang S, Samuni U, Koren J, Chiosis G, Sabatino D. RNAi Screening of the Glucose-Regulated Chaperones in Cancer with Self-Assembled siRNA Nanostructures. NANO LETTERS 2016; 16:6099-6108. [PMID: 27669096 PMCID: PMC5378679 DOI: 10.1021/acs.nanolett.6b02274] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
The emerging field of RNA nanotechnology has been used to design well-programmed, self-assembled nanostructures for applications in chemistry, biology, and medicine. At the forefront of its utility in cancer is the unrestricted ability to self-assemble multiple siRNAs within a single nanostructure formulation for the RNAi screening of a wide range of oncogenes while potentiating the gene therapy of malignant tumors. In our RNAi nanotechnology approach, V- and Y-shape RNA templates were designed and constructed for the self-assembly of discrete, higher-ordered siRNA nanostructures targeting the oncogenic glucose regulated chaperones. The GRP78-targeting siRNAs self-assembled into genetically encoded spheres, triangles, squares, pentagons and hexagons of discrete sizes and shapes according to TEM imaging. Furthermore, gel electrophoresis, thermal denaturation, and CD spectroscopy validated the prerequisite siRNA hybrids for their RNAi application. In a 24 sample siRNA screen conducted within the AN3CA endometrial cancer cells known to overexpress oncogenic GRP78 activity, the self-assembled siRNAs targeting multiple sites of GRP78 expression demonstrated more potent and long-lasting anticancer activity relative to their linear controls. Extending the scope of our RNAi screening approach, the self-assembled siRNA hybrids (5 nM) targeting of GRP-75, 78, and 94 resulted in significant (50-95%) knockdown of the glucose regulated chaperones, which led to synergistic effects in tumor cell cycle arrest (50-80%) and death (50-60%) within endometrial (AN3CA), cervical (HeLa), and breast (MDA-MB-231) cancer cell lines. Interestingly, a nontumorigenic lung (MRC5) cell line displaying normal glucose regulated chaperone levels was found to tolerate siRNA treatment and demonstrated less toxicity (5-20%) relative to the cancer cells that were found to be addicted to glucose regulated chaperones. These remarkable self-assembled siRNA nanostructures may thus encompass a new class of potent siRNAs that may be useful in screening important oncogene targets while improving siRNA therapeutic efficacy and specificity in cancer.
Collapse
Affiliation(s)
- Mayurbhai R. Patel
- Program in Chemical Biology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Department of Chemistry and Biochemistry, Seton Hall University, South Orange, New Jersey 07079, United States
| | - Stephen D. Kozuch
- Department of Chemistry and Biochemistry, Seton Hall University, South Orange, New Jersey 07079, United States
| | - Christopher N. Cultrara
- Department of Chemistry and Biochemistry, Seton Hall University, South Orange, New Jersey 07079, United States
| | - Reeta Yadav
- Department of Chemistry and Biochemistry, Queens College, City University of New York, 65-30 Kissena Blvd., Flushing, New York 11367, United States
- Ph.D. Programs in Chemistry and Biochemistry, The Graduate Center of the City University of New York, New York City, New York 10016, United States
| | - Suiying Huang
- Department of Chemistry and Biochemistry, Queens College, City University of New York, 65-30 Kissena Blvd., Flushing, New York 11367, United States
- Ph.D. Programs in Chemistry and Biochemistry, The Graduate Center of the City University of New York, New York City, New York 10016, United States
| | - Uri Samuni
- Department of Chemistry and Biochemistry, Queens College, City University of New York, 65-30 Kissena Blvd., Flushing, New York 11367, United States
- Ph.D. Programs in Chemistry and Biochemistry, The Graduate Center of the City University of New York, New York City, New York 10016, United States
| | - John Koren
- Program in Chemical Biology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Gabriela Chiosis
- Program in Chemical Biology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - David Sabatino
- Department of Chemistry and Biochemistry, Seton Hall University, South Orange, New Jersey 07079, United States
- Corresponding Author.
| |
Collapse
|
26
|
Li KT, Chen Q, Wang DW, Duan QQ, Tian S, He JW, Ou YS, Bai DQ. Mitochondrial pathway and endoplasmic reticulum stress participate in the photosensitizing effectiveness of AE-PDT in MG63 cells. Cancer Med 2016; 5:3186-3193. [PMID: 27700017 PMCID: PMC5119974 DOI: 10.1002/cam4.895] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 07/18/2016] [Accepted: 08/08/2016] [Indexed: 01/01/2023] Open
Abstract
Photodynamic therapy (PDT) is a promising treatment in cancer therapy, with a photosensitizer activated by visible light. Aloe‐emodin (AE) is a promising photosensitive agent. In this study, the photosensitizing effects and possible mechanisms of AE‐PDT in MG63 cells were evaluated. The efficiency of AE‐PDT was analyzed by MTT assay. The mode of cell death was investigated by Hoechst 33,342 staining and flow cytometer. The intracellular distribution of AE was detected with confocal microscopy. The formation of reactive oxygen species (ROS) was detected by DCFH‐DA. The mitochondrial membrane potential (MMP) was measured by Rhodamine 123. The expression of proteins including cytochrome c, caspase‐3, ‐9, and ‐12, CHOP and GRP78 was detected by western blot. Apoptosis is the primary mode of cell death in our study, which occurs in a manner of depending on AE concentration and irradiation dose. Confocal microscopy showed that AE was primarily localized on the mitochondria and endoplasmic reticulum (ER) of MG63 cells. AE‐PDT resulted in rapid increases of intracellular ROS production, which reached a peak at 2 h, followed by declining of mitochondrial membrane potential, releasing of cytochrome c from mitochondria into the cytoplasm, and up‐regulation of caspase‐3, ‐9, and ‐12, CHOP and GRP78. These results suggest that death of MG63 cells induced by AE‐PDT is triggered by ROS. Meanwhile, Mitochondria and ER serve as the subcellular targets, which are responsible for AE‐PDT‐induced death of MG63 cells.
Collapse
Affiliation(s)
- Kai-Ting Li
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qing Chen
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Da-Wu Wang
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qin-Qin Duan
- Department of Gastroenterology, Chinese Medicine Hospital of Longquan, Chengdu, China
| | - Si Tian
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Juan-Wen He
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yun-Sheng Ou
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ding-Qun Bai
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
27
|
Le Reste PJ, Avril T, Quillien V, Morandi X, Chevet E. Reprint of: Signaling the Unfolded Protein Response in primary brain cancers. Brain Res 2016; 1648:542-552. [PMID: 27362469 DOI: 10.1016/j.brainres.2016.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/16/2016] [Accepted: 03/17/2016] [Indexed: 11/15/2022]
Abstract
The Unfolded Protein Response (UPR) is an adaptive cellular program used by eukaryotic cells to cope with protein misfolding stress in the Endoplasmic Reticulum (ER). During tumor development, cancer cells are facing intrinsic (oncogene activation) and extrinsic (limiting nutrient or oxygen supply; exposure to chemotherapies) challenges, with which they must cope to survive. Primary brain tumors are relatively rare but deadly and present a significant challenge in the determination of risk factors in the population. These tumors are inherently difficult to cure because of their protected location in the brain. As such surgery, radiation and chemotherapy options carry potentially lasting patient morbidity and incomplete tumor cure. Some of these tumors, such as glioblastoma, were reported to present features of ER stress and to depend on UPR activation to sustain growth, but to date there is no clear general representation of the ER stress status in primary brain tumors. In this review, we describe the key molecular mechanisms controlling the UPR and their implication in cancers. Then we extensively review the literature reporting the status of ER stress in various primary brain tumors and discuss the potential impact of such observation on patient stratification and on the possibility of developing appropriate targeted therapies using the UPR as therapeutic target. This article is part of a Special Issue entitled SI:ER stress.
Collapse
Affiliation(s)
- Pierre-Jean Le Reste
- Department of Neurosurgery, University Hospital Pontchaillou, Rennes, France; Inserm ERL440 "Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France
| | - Tony Avril
- Inserm ERL440 "Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France; Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Véronique Quillien
- Inserm ERL440 "Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France; Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Xavier Morandi
- Department of Neurosurgery, University Hospital Pontchaillou, Rennes, France
| | - Eric Chevet
- Inserm ERL440 "Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France; Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France.
| |
Collapse
|
28
|
Investigation of cell death mechanisms in human lymphatic endothelial cells undergoing photodynamic therapy. Photodiagnosis Photodyn Ther 2016; 14:57-65. [DOI: 10.1016/j.pdpdt.2016.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 01/25/2016] [Accepted: 02/05/2016] [Indexed: 12/24/2022]
|
29
|
Zhang X, Zhang R, Yang H, Xiang Q, Jiang Q, He Q, Zhang T, Chen C, Zhu H, Wang Q, Ning Q, Li Y, Lei P, Shen G. Hepatitis B virus enhances cisplatin-induced hepatotoxicity via a mechanism involving suppression of glucose-regulated protein of 78 Kda. Chem Biol Interact 2016; 254:45-53. [PMID: 27234046 DOI: 10.1016/j.cbi.2016.05.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 04/30/2016] [Accepted: 05/23/2016] [Indexed: 01/13/2023]
Abstract
Cisplatin is a classical platinum-based chemotherapeutic drug used in the treatment of many cancer types, including hepatocellular carcinoma (HCC). The application of cisplatin is significantly limited by its toxicity, which may be affected by various biological factors. Persistence of Hepatitis B virus (HBV) infection leads to HCC development and may be associated with higher incidence of severe hepatitis during chemotherapy. However, whether HBV alters the susceptibility of hepatocytes to cisplatin remains poorly understood. Here, we demonstrate that HBV transfection enhanced cisplatin-induced hepatotoxicity via a mechanism involving suppression of glucose-regulated protein of 78 KDa (Grp78), a major stress-induced chaperone that localizes to the endoplasmic reticulum. Silencing Grp78 gene increased the susceptibility of HepG2 to cisplatin by activating caspase-3. Grp78 expression was down-regulated by HBV infection both in vitro and in liver tissues of patients. We compared the cisplatin sensitivity of hepatoma cells either expressing (HepG2.2.15 cells) or not expressing the entire Hepatitis B Virus genome (HepG2). HepG2.2.15 cells showed increased sensitivity to cisplatin and a higher apoptosis rate. Overexpression of Grp78 counteracted the increase of sensitivity of HepG2.215 cells to cisplatin. Furthermore, we found that HBV disrupted Grp78 synthesis in response to cisplatin stimulation, which may trigger severe and prolonged endoplasmic reticulum (ER) stress that can induce cellular apoptosis. Our findings provide new information into the effect of HBV in the modulation of Grp78 expression, and, consequently on cisplatin-induced hepatotoxicity during viral infection.
Collapse
Affiliation(s)
- Xiaoxue Zhang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, PR China; Department of Laboratory Medicine, Affiliated Pu'ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, PR China
| | - Rui Zhang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, PR China
| | - HuiOu Yang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, PR China
| | - Qian Xiang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, PR China
| | - Qing Jiang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, PR China
| | - Qi He
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, PR China
| | - Ting Zhang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, PR China
| | - Chen Chen
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, PR China
| | - Huifen Zhu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, PR China
| | - Qiang Wang
- Department of Immunology, Medical College of Wuhan University of Science and Technology, PR China
| | - Qin Ning
- Department of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, PR China
| | - Yiwu Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, PR China
| | - Ping Lei
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, PR China.
| | - Guanxin Shen
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, PR China.
| |
Collapse
|
30
|
Gabrysiak M, Wachowska M, Barankiewicz J, Pilch Z, Ratajska A, Skrzypek E, Winiarska M, Domagala A, Rygiel TP, Jozkowicz A, Boon L, Golab J, Firczuk M. Low dose of GRP78-targeting subtilase cytotoxin improves the efficacy of photodynamic therapy in vivo. Oncol Rep 2016; 35:3151-8. [PMID: 27035643 PMCID: PMC4872279 DOI: 10.3892/or.2016.4723] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 12/23/2015] [Indexed: 12/22/2022] Open
Abstract
Photodynamic therapy (PDT) exerts direct cytotoxic effects on tumor cells, destroys tumor blood and lymphatic vessels and induces local inflammation. Although PDT triggers the release of immunogenic antigens from tumor cells, the degree of immune stimulation is regimen-dependent. The highest immunogenicity is achieved at sub-lethal doses, which at the same time trigger cytoprotective responses, that include increased expression of glucose-regulated protein 78 (GRP78). To mitigate the cytoprotective effects of GRP78 and preserve the immunoregulatory activity of PDT, we investigated the in vivo efficacy of PDT in combination with EGF-SubA cytotoxin that was shown to potentiate in vitro PDT cytotoxicity by inactivating GRP78. Treatment of immunocompetent BALB/c mice with EGF-SubA improved the efficacy of PDT but only when mice were treated with a dose of EGF-SubA that exerted less pronounced effects on the number of T and B lymphocytes as well as dendritic cells in mouse spleens. The observed antitumor effects were critically dependent on CD8+ T cells and were completely abrogated in immunodeficient SCID mice. All these results suggest that GRP78 targeting improves in vivo PDT efficacy provided intact T-cell immune system.
Collapse
Affiliation(s)
- Magdalena Gabrysiak
- Department of Immunology, Center of Biostructure Research, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland
| | - Malgorzata Wachowska
- Department of Immunology, Center of Biostructure Research, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland
| | - Joanna Barankiewicz
- Department of Immunology, Center of Biostructure Research, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland
| | - Zofia Pilch
- Department of Immunology, Center of Biostructure Research, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland
| | - Anna Ratajska
- Department of Pathology, Center of Biostructure Research, Medical University of Warsaw, 02-004 Warsaw, Poland
| | - Ewa Skrzypek
- Department of Pathology, Center of Biostructure Research, Medical University of Warsaw, 02-004 Warsaw, Poland
| | - Magdalena Winiarska
- Department of Immunology, Center of Biostructure Research, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland
| | - Antoni Domagala
- Department of Immunology, Center of Biostructure Research, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland
| | - Tomasz P Rygiel
- Department of Immunology, Center of Biostructure Research, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kracow, Poland
| | - Louis Boon
- EPIRUS Biopharmaceuticals Netherlands BV, 3584 CM Utrecht, The Netherlands
| | - Jakub Golab
- Department of Immunology, Center of Biostructure Research, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland
| | - Malgorzata Firczuk
- Department of Immunology, Center of Biostructure Research, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland
| |
Collapse
|
31
|
Silva LC, Ferreira-Strixino J, Fontana LC, Rocha Gonsalves AMD, Serra AC, Pineiro M, Canevari RA. Molecular analysis of apoptosis pathway after photodynamic therapy in breast cancer: Animal model study. Photodiagnosis Photodyn Ther 2016; 14:152-8. [PMID: 27018246 DOI: 10.1016/j.pdpdt.2016.03.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 03/06/2016] [Accepted: 03/21/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Molecular investigation of breast tumors has permitted better understanding about interaction of genes and pathways involved in tumor progression. OBJECTIVE The aim of this study was to evaluate the association between genes belonging to the pathway of apoptosis with tumor response to photodynamic therapy. STUDY DESIGN/MATERIALS AND METHODS The mammary tumors were induced in twenty-four Spraguey-Dawley female rats by oral gavage of 7,12-dimethylbenz(a)anthracene (8mg/Kg body weight). Animals were divided into three groups: G1 (normal tissue), G2 (tumors without treatment), G3 (animals euthanized 48h after treatment). The photosensitizer used was a chlorin, 5,15-bis-(2-bromo-5-hydroxyphenyl) chlorin in the dose of 8mg/kg for each animal. Light source of diode laser at a wavelength of 660nm, fluence rate of 100mW/cm, and light dose of 100J/cm was delivery to lesions for treatment. A sample from each animal was investigated by quantitative real time PCR using Rat Apoptosis RT(2) Profiler™ PCR Array platform. RESULTS Pro-apoptotic BAK1, CARD6, CASP8, CIDEA, CIDEB, DAPK1, TNF, TNFRSF10B, FASLG, LOC687813, and TP73 genes showed increased expression, and CD40 anti-apoptotic gene showed decreased expression in the group who underwent PDT (G3) in relation to G2. CONCLUSION The results indicated that these genes are involved more directly with cellular apoptosis induced by PDT using the Chlorin photosensitizer.
Collapse
Affiliation(s)
- Luciana C Silva
- Instituto de Pesquisa e Desenvolvimento, IP&D-Laboratório de Biologia Molecular do Câncer and Laboratório de Espectroscopia Vibracional Biomédica, Universidade do Vale do Paraíba-UNIVAP, São José dos Campos, 12400-000, SP, Brazil
| | - Juliana Ferreira-Strixino
- Instituto de Pesquisa e Desenvolvimento, IP&D-Laboratório de Terapia Fotodinâmica, Universidade do Vale do Paraíba-UNIVAP, São José dos Campos, 12400-000, SP, Brazil.
| | - Letícia C Fontana
- Instituto de Pesquisa e Desenvolvimento, IP&D-Laboratório de Terapia Fotodinâmica, Universidade do Vale do Paraíba-UNIVAP, São José dos Campos, 12400-000, SP, Brazil
| | | | - Arménio C Serra
- Chymiotechnon, Departamento de Química, Universidade de Coimbra, 3049-535, Coimbra, Portugal
| | - Marta Pineiro
- Chymiotechnon, Departamento de Química, Universidade de Coimbra, 3049-535, Coimbra, Portugal
| | - Renata A Canevari
- Instituto de Pesquisa e Desenvolvimento, IP&D-Laboratório de Biologia Molecular do Câncer and Laboratório de Espectroscopia Vibracional Biomédica, Universidade do Vale do Paraíba-UNIVAP, São José dos Campos, 12400-000, SP, Brazil.
| |
Collapse
|
32
|
Le Reste PJ, Avril T, Quillien V, Morandi X, Chevet E. Signaling the Unfolded Protein Response in primary brain cancers. Brain Res 2016; 1642:59-69. [PMID: 27016056 DOI: 10.1016/j.brainres.2016.03.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/16/2016] [Accepted: 03/17/2016] [Indexed: 12/14/2022]
Abstract
The Unfolded Protein Response (UPR) is an adaptive cellular program used by eukaryotic cells to cope with protein misfolding stress in the Endoplasmic Reticulum (ER). During tumor development, cancer cells are facing intrinsic (oncogene activation) and extrinsic (limiting nutrient or oxygen supply; exposure to chemotherapies) challenges, with which they must cope to survive. Primary brain tumors are relatively rare but deadly and present a significant challenge in the determination of risk factors in the population. These tumors are inherently difficult to cure because of their protected location in the brain. As such surgery, radiation and chemotherapy options carry potentially lasting patient morbidity and incomplete tumor cure. Some of these tumors, such as glioblastoma, were reported to present features of ER stress and to depend on UPR activation to sustain growth, but to date there is no clear general representation of the ER stress status in primary brain tumors. In this review, we describe the key molecular mechanisms controlling the UPR and their implication in cancers. Then we extensively review the literature reporting the status of ER stress in various primary brain tumors and discuss the potential impact of such observation on patient stratification and on the possibility of developing appropriate targeted therapies using the UPR as therapeutic target.
Collapse
Affiliation(s)
- Pierre-Jean Le Reste
- Department of Neurosurgery, University Hospital Pontchaillou, Rennes, France; Inserm ERL440 "Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France
| | - Tony Avril
- Inserm ERL440 "Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France; Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Véronique Quillien
- Inserm ERL440 "Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France; Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Xavier Morandi
- Department of Neurosurgery, University Hospital Pontchaillou, Rennes, France
| | - Eric Chevet
- Inserm ERL440 "Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France; Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France.
| |
Collapse
|
33
|
Rodríguez ME, Cogno IS, Milla Sanabria LS, Morán YS, Rivarola VA. Heat shock proteins in the context of photodynamic therapy: autophagy, apoptosis and immunogenic cell death. Photochem Photobiol Sci 2016; 15:1090-1102. [DOI: 10.1039/c6pp00097e] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Heat shock proteins can mediate resistance to photodynamic therapy by inhibiting apoptosis and modulating autophagy which, in turn, prevents apoptosis and immunogenic cell death.
Collapse
Affiliation(s)
- Matías E. Rodríguez
- Departamento de Biología Molecular
- Universidad Nacional de Río Cuarto
- Río Cuarto (5800)
- Argentina
| | - Ingrid S. Cogno
- Departamento de Biología Molecular
- Universidad Nacional de Río Cuarto
- Río Cuarto (5800)
- Argentina
| | - Laura S. Milla Sanabria
- Departamento de Biología Molecular
- Universidad Nacional de Río Cuarto
- Río Cuarto (5800)
- Argentina
| | - Yanina S. Morán
- Departamento de Biología Molecular
- Universidad Nacional de Río Cuarto
- Río Cuarto (5800)
- Argentina
| | - Viviana A. Rivarola
- Departamento de Biología Molecular
- Universidad Nacional de Río Cuarto
- Río Cuarto (5800)
- Argentina
| |
Collapse
|
34
|
Broekgaarden M, Weijer R, van Gulik TM, Hamblin MR, Heger M. Tumor cell survival pathways activated by photodynamic therapy: a molecular basis for pharmacological inhibition strategies. Cancer Metastasis Rev 2015; 34:643-90. [PMID: 26516076 PMCID: PMC4661210 DOI: 10.1007/s10555-015-9588-7] [Citation(s) in RCA: 176] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Photodynamic therapy (PDT) has emerged as a promising alternative to conventional cancer therapies such as surgery, chemotherapy, and radiotherapy. PDT comprises the administration of a photosensitizer, its accumulation in tumor tissue, and subsequent irradiation of the photosensitizer-loaded tumor, leading to the localized photoproduction of reactive oxygen species (ROS). The resulting oxidative damage ultimately culminates in tumor cell death, vascular shutdown, induction of an antitumor immune response, and the consequent destruction of the tumor. However, the ROS produced by PDT also triggers a stress response that, as part of a cell survival mechanism, helps cancer cells to cope with the PDT-induced oxidative stress and cell damage. These survival pathways are mediated by the transcription factors activator protein 1 (AP-1), nuclear factor E2-related factor 2 (NRF2), hypoxia-inducible factor 1 (HIF-1), nuclear factor κB (NF-κB), and those that mediate the proteotoxic stress response. The survival pathways are believed to render some types of cancer recalcitrant to PDT and alter the tumor microenvironment in favor of tumor survival. In this review, the molecular mechanisms are elucidated that occur post-PDT to mediate cancer cell survival, on the basis of which pharmacological interventions are proposed. Specifically, pharmaceutical inhibitors of the molecular regulators of each survival pathway are addressed. The ultimate aim is to facilitate the development of adjuvant intervention strategies to improve PDT efficacy in recalcitrant solid tumors.
Collapse
Affiliation(s)
- Mans Broekgaarden
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Ruud Weijer
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Thomas M van Gulik
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, MA, USA
| | - Michal Heger
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
35
|
The interplay between GRP78 expression and Akt activation in human colon cancer cells under celecoxib treatment. Anticancer Drugs 2015. [PMID: 26225471 DOI: 10.1097/cad.0000000000000273] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
It has been reported previously that celecoxib shows antitumor effects in many types of cancers. Here, we detected its effects on DLD-1 and SW480 (two human colon cancer cell lines) and investigated the dynamic relationship between the 78-kDa glucose-regulatory protein (GRP78) and the phosphoinositide 3-kinase (PI3K)/Akt pathway. Gene expression was detected by real-time PCR and western blot analysis; the cytotoxicity was determined by the MTT assay and flow cytometry. First, the results showed that celecoxib induced cytotoxicity in a dose-dependent and time-dependent manner. Furthermore, we found the celecoxib-triggered unfolded protein response and the bidirectional regulation of Akt activation in both cell lines. Inhibiting the Akt activation by the PI3K inhibitor LY294002 markedly enhanced GRP78 expression. Besides, silencing the GRP78 expression regulated Akt activation in a time-dependent manner and increased the induction of the C/EBP homologous protein (CHOP) as well as considerably promoted celecoxib-induced apoptosis. In conclusion, these findings provide evidence that under the celecoxib treatment, GRP78 plays a protective role by modulating Akt activation and abrogating CHOP expression. However, Akt activation can provide a feedback loop to inhibit GRP78 expression. These studies can lead to novel therapeutic strategies for human colon cancer.
Collapse
|
36
|
Cancer Microenvironment and Endoplasmic Reticulum Stress Response. Mediators Inflamm 2015; 2015:417281. [PMID: 26491226 PMCID: PMC4600498 DOI: 10.1155/2015/417281] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 04/21/2015] [Accepted: 04/24/2015] [Indexed: 12/17/2022] Open
Abstract
Different stressful conditions such as hypoxia, nutrient deprivation, pH changes, or reduced vascularization, potentially able to act as growth-limiting factors for tumor cells, activate the unfolded protein response (UPR). UPR is therefore involved in tumor growth and adaptation to severe environments and is generally cytoprotective in cancer. The present review describes the molecular mechanisms underlying UPR and able to promote survival and proliferation in cancer. The critical role of UPR activation in tumor growth promotion is discussed in detail for a few paradigmatic tumors such as prostate cancer and melanoma.
Collapse
|
37
|
Zhen Z, Tang W, Zhang W, Xie J. Folic acid conjugated ferritins as photosensitizer carriers for photodynamic therapy. NANOSCALE 2015; 7:10330-10333. [PMID: 25998995 PMCID: PMC4885642 DOI: 10.1039/c5nr01833a] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
We coupled folic acid as a tumour targeting ligand to the surface of ferritins and loaded them with ZnF16Pc. The resulting nanoconjugates can efficiently hone in on 4T1 tumours in vivo, and, with photoirradiation, leading to suppressed tumour growth and tumour metastasis.
Collapse
Affiliation(s)
- Zipeng Zhen
- Department of Chemistry, University of Georgia, Athens, Georgia 30602, United States
- Bio-Imaging Research Center, University of Georgia, Athens, Georgia 30602, United States
| | - Wei Tang
- Department of Chemistry, University of Georgia, Athens, Georgia 30602, United States
- Bio-Imaging Research Center, University of Georgia, Athens, Georgia 30602, United States
| | - Weizhong Zhang
- Department of Chemistry, University of Georgia, Athens, Georgia 30602, United States
- Bio-Imaging Research Center, University of Georgia, Athens, Georgia 30602, United States
| | - Jin Xie
- Department of Chemistry, University of Georgia, Athens, Georgia 30602, United States
- Bio-Imaging Research Center, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
38
|
Tameire F, Verginadis II, Koumenis C. Cell intrinsic and extrinsic activators of the unfolded protein response in cancer: Mechanisms and targets for therapy. Semin Cancer Biol 2015; 33:3-15. [PMID: 25920797 DOI: 10.1016/j.semcancer.2015.04.002] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 04/16/2015] [Indexed: 02/07/2023]
Abstract
A variety of cell intrinsic or extrinsic stresses evoke perturbations in the folding environment of the endoplasmic reticulum (ER), collectively known as ER stress. Adaptation to stress and re-establishment of ER homeostasis is achieved by activation of an integrated signal transduction pathway called the unfolded protein response (UPR). Both ER stress and UPR activation have been implicated in a variety of human cancers. Although at early stages or physiological conditions of ER stress, the UPR generally promotes survival, when the stress becomes more stringent or prolonged, its role can switch to a pro-cell death one. Here, we discuss historical and recent evidence supporting an involvement of the UPR in malignancy, describe the main mechanisms by which tumor cells overcome ER stress to promote their survival, tumor progression and metastasis and discuss the current state of efforts to develop therapeutic approaches of targeting the UPR.
Collapse
Affiliation(s)
- Feven Tameire
- Department of Radiation Oncology, Perelman University School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Program in Cell and Molecular Biology, Perelman University School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ioannis I Verginadis
- Department of Radiation Oncology, Perelman University School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman University School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
39
|
Li D, Li L, Li P, Li Y, Chen X. Apoptosis of HeLa cells induced by a new targeting photosensitizer-based PDT via a mitochondrial pathway and ER stress. Onco Targets Ther 2015; 8:703-11. [PMID: 25897245 PMCID: PMC4396590 DOI: 10.2147/ott.s76370] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Photodynamic therapy (PDT) is emerging as a viable treatment for many cancers. To decrease the cutaneous photosensitivity induced by PDT, many attempts have been made to search for a targeting photosensitizer; however, few reports describe the molecular mechanism of PDT mediated by this type of targeting photosensitizer. The present study aimed to investigate the molecular mechanism of PDT induced by a new targeting photosensitizer (PS I), reported previously by us, on HeLa cells. Apoptosis is the primary mode of HeLa cell death in our system, and apoptosis occurs in a manner dependent on concentration, irradiation dose, and drug–light intervals. After endocytosis mediated by the folate receptor, PS I was primarily localized to the mitochondria and the endoplasmic reticulum (ER) of HeLa cells. PS I PDT resulted in rapid increases in intracellular reactive oxygen species (ROS) production and Ca2+ concentration, both of which reached a peak nearly simultaneously at 15 minutes, followed by the loss of mitochondrial membrane potential at 30 minutes, release of cytochrome c from mitochondria into the cytoplasm, downregulation of Bcl-2 expression, and upregulation of Bax expression. Meanwhile, activation of caspase-3, -9, and -12, as well as induction of C/EBP homologous protein (CHOP) and glucose-regulated protein (GRP78), in HeLa cells after PS I PDT was also detected. These results suggest that apoptosis of HeLa cells induced by PS I PDT is not only triggered by ROS but is also regulated by Ca2+ overload. Mitochondria and the ER serve as the subcellular targets of PS I PDT, the effective activation of which is responsible for PS I PDT-induced apoptosis in HeLa cells.
Collapse
Affiliation(s)
- Donghong Li
- State Key Laboratory of Trauma, Burn and Combined Injury, The Second Department of Research Institute of Surgery, Third Military Medical University, Chongqing, People's Republic of China
| | - Lei Li
- The First Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Pengxi Li
- State Key Laboratory of Trauma, Burn and Combined Injury, The Second Department of Research Institute of Surgery, Third Military Medical University, Chongqing, People's Republic of China
| | - Yi Li
- Cancer Center, Daping Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Xiangyun Chen
- State Key Laboratory of Trauma, Burn and Combined Injury, The Second Department of Research Institute of Surgery, Third Military Medical University, Chongqing, People's Republic of China
| |
Collapse
|
40
|
Mihailidou C, Chatzistamou I, Papavassiliou AG, Kiaris H. Improvement of chemotherapeutic drug efficacy by endoplasmic reticulum stress. Endocr Relat Cancer 2015; 22:229-38. [PMID: 25670032 DOI: 10.1530/erc-15-0019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tunicamycin (TUN), an inhibitor of protein glycosylation and therefore a potent stimulator of endoplasmic reticulum (ER) stress, has been used to improve anticancer drug efficacy, but the underlying mechanism remains obscure. In this study, we show that acute administration of TUN in mice induces the unfolded protein response and suppresses the levels of P21, a cell cycle regulator with anti-apoptotic activity. The inhibition of P21 after ER stress appears to be C/EBP homologous protein (CHOP)-dependent because in CHOP-deficient mice, TUN not only failed to suppress, but rather induced the expression of P21. Results of promoter-activity reporter assays using human cancer cells and mouse fibroblasts indicated that the regulation of P21 by CHOP operates at the level of transcription and involves direct binding of CHOP transcription factor to the P21 promoter. The results of cell viability and clonogenic assays indicate that ER-stress-related suppression of P21 expression potentiates caspase activation and sensitizes cells to doxorubicin treatment, while administration of TUN to mice increases the therapeutic efficacy of anticancer therapy for HepG2 liver and A549 lung cancers.
Collapse
Affiliation(s)
- Chrysovalantou Mihailidou
- Department of Biological ChemistryUniversity of Athens Medical School, Athens 11527, GreeceDepartment of Basic SciencesDental School, University of Athens, Athens 11527, GreeceDepartment of PathologyMicrobiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina 29208, USADepartment of Drug Discovery and Biomedical SciencesUniversity of South Carolina, Columbia, South Carolina 29425, USA
| | - Ioulia Chatzistamou
- Department of Biological ChemistryUniversity of Athens Medical School, Athens 11527, GreeceDepartment of Basic SciencesDental School, University of Athens, Athens 11527, GreeceDepartment of PathologyMicrobiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina 29208, USADepartment of Drug Discovery and Biomedical SciencesUniversity of South Carolina, Columbia, South Carolina 29425, USA Department of Biological ChemistryUniversity of Athens Medical School, Athens 11527, GreeceDepartment of Basic SciencesDental School, University of Athens, Athens 11527, GreeceDepartment of PathologyMicrobiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina 29208, USADepartment of Drug Discovery and Biomedical SciencesUniversity of South Carolina, Columbia, South Carolina 29425, USA
| | - Athanasios G Papavassiliou
- Department of Biological ChemistryUniversity of Athens Medical School, Athens 11527, GreeceDepartment of Basic SciencesDental School, University of Athens, Athens 11527, GreeceDepartment of PathologyMicrobiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina 29208, USADepartment of Drug Discovery and Biomedical SciencesUniversity of South Carolina, Columbia, South Carolina 29425, USA
| | - Hippokratis Kiaris
- Department of Biological ChemistryUniversity of Athens Medical School, Athens 11527, GreeceDepartment of Basic SciencesDental School, University of Athens, Athens 11527, GreeceDepartment of PathologyMicrobiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina 29208, USADepartment of Drug Discovery and Biomedical SciencesUniversity of South Carolina, Columbia, South Carolina 29425, USA Department of Biological ChemistryUniversity of Athens Medical School, Athens 11527, GreeceDepartment of Basic SciencesDental School, University of Athens, Athens 11527, GreeceDepartment of PathologyMicrobiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina 29208, USADepartment of Drug Discovery and Biomedical SciencesUniversity of South Carolina, Columbia, South Carolina 29425, USA
| |
Collapse
|
41
|
Garg AD, Maes H, van Vliet AR, Agostinis P. Targeting the hallmarks of cancer with therapy-induced endoplasmic reticulum (ER) stress. Mol Cell Oncol 2014; 2:e975089. [PMID: 27308392 PMCID: PMC4905250 DOI: 10.4161/23723556.2014.975089] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 09/24/2014] [Accepted: 09/25/2014] [Indexed: 12/19/2022]
Abstract
The endoplasmic reticulum (ER) is at the center of a number of vital cellular processes such as cell growth, death, and differentiation, crosstalk with immune or stromal cells, and maintenance of proteostasis or homeostasis, and ER functions have implications for various pathologies including cancer. Recently, a number of major hallmarks of cancer have been delineated that are expected to facilitate the development of anticancer therapies. However, therapeutic induction of ER stress as a strategy to broadly target multiple hallmarks of cancer has been seldom discussed despite the fact that several primary or secondary ER stress-inducing therapies have been found to exhibit positive clinical activity in cancer patients. In the present review we provide a brief historical overview of the major discoveries and milestones in the field of ER stress biology with important implications for anticancer therapy. Furthermore, we comprehensively discuss possible strategies enabling the targeting of multiple hallmarks of cancer with therapy-induced ER stress.
Collapse
Affiliation(s)
- Abhishek D Garg
- Cell Death Research & Therapy (CDRT) Laboratory; Department for Cellular and Molecular Medicine; KU Leuven University of Leuven ; Leuven, Belgium
| | - Hannelore Maes
- Cell Death Research & Therapy (CDRT) Laboratory; Department for Cellular and Molecular Medicine; KU Leuven University of Leuven ; Leuven, Belgium
| | - Alexander R van Vliet
- Cell Death Research & Therapy (CDRT) Laboratory; Department for Cellular and Molecular Medicine; KU Leuven University of Leuven ; Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) Laboratory; Department for Cellular and Molecular Medicine; KU Leuven University of Leuven ; Leuven, Belgium
| |
Collapse
|
42
|
GRP78 regulates sensitivity of human colorectal cancer cells to DNA targeting agents. Cytotechnology 2014; 68:459-67. [PMID: 25399254 DOI: 10.1007/s10616-014-9799-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 10/20/2014] [Indexed: 10/24/2022] Open
Abstract
This study was carried out to investigate the activation status of unfolded protein response (UPR) in colorectal cancer (CRC) and its contribution to CRC resistance to chemotherapy-induced apoptosis. Chemotherapy-induced apoptosis was assessed by the propidium iodide method. Activation of UPR was evaluated in CRC cell lines using immunoblotting technique and in CRC tissues using immunohistochemistry. Findings of the present study revealed that the UPR is constitutively activated in CRC cell lines and CRC tissues isolated from patients, as evidenced by relatively high levels of the 78-kDa glucose-regulated protein (GRP78) and spliced X-box-binding protein 1 mRNA in tissue samples. In addition, CRC cell lines differentially responded to clinically relevant DNA-targeting agents including cisplatin, and 5-flourouracil. Moreover, the levels of GRP78 were inversely associated with sensitivity of CRC cells to chemotherapy-induced apoptosis. Inhibition of GRP78 by siRNA resulted in increased sensitivity of CRC cells to chemotherapeutic agents. Collectively, current results appear to provide novel insights into the role of UPR in determining sensitivity of CRC cells to chemotherapeutic agents and might have important implications for personalized CRC treatment.
Collapse
|
43
|
Wang WA, Groenendyk J, Michalak M. Endoplasmic reticulum stress associated responses in cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2143-9. [DOI: 10.1016/j.bbamcr.2014.01.012] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 01/08/2014] [Accepted: 01/10/2014] [Indexed: 11/29/2022]
|
44
|
Yadav RK, Chae SW, Kim HR, Chae HJ. Endoplasmic reticulum stress and cancer. J Cancer Prev 2014; 19:75-88. [PMID: 25337575 PMCID: PMC4204165 DOI: 10.15430/jcp.2014.19.2.75] [Citation(s) in RCA: 282] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 06/07/2014] [Accepted: 06/07/2014] [Indexed: 12/14/2022] Open
Abstract
The endoplasmic reticulum (ER) is the principal organelle responsible for multiple cellular functions including protein folding and maturation and the maintenance of cellular homeostasis. ER stress is activated by a variety of factors and triggers the unfolded protein response (UPR), which restores homeostasis or activates cell death. Multiple studies have clarified the link between ER stress and cancer, and particularly the involvement of the UPR. The UPR seems to adjust the paradoxical microenvironment of cancer and, as such, is one of resistance mechanisms against cancer therapy. This review describes the activity of different UPRs involved in tumorigenesis and resistance to cancer therapy.
Collapse
Affiliation(s)
- Raj Kumar Yadav
- Department of Pharmacology and Institute of Cardiovascular Research, School of Medicine, Chonbuk National University, Jeonju, Chonbuk, Korea
| | - Soo-Wan Chae
- Department of Pharmacology and Institute of Cardiovascular Research, School of Medicine, Chonbuk National University, Jeonju, Chonbuk, Korea
| | - Hyung-Ryong Kim
- Department of Dental Pharmacology, College of Dentistry, Wonkwang University, Iksan, Chonbuk, Korea
| | - Han Jung Chae
- Department of Pharmacology and Institute of Cardiovascular Research, School of Medicine, Chonbuk National University, Jeonju, Chonbuk, Korea
| |
Collapse
|
45
|
Feng C, He K, Zhang C, Su S, Li B, Li Y, Duan CY, Chen S, Chen R, Liu Y, Li H, Wei M, Xia X, Dai R. JNK contributes to the tumorigenic potential of human cholangiocarcinoma cells through the mTOR pathway regulated GRP78 induction. PLoS One 2014; 9:e90388. [PMID: 24587347 PMCID: PMC3938720 DOI: 10.1371/journal.pone.0090388] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 01/28/2014] [Indexed: 12/14/2022] Open
Abstract
Less is known about the roles of c-Jun N-terminal kinase (JNK) in cholangiocarcinoma (CCA). Here, we report that JNK exerts its oncogenic action in human CCA cells, partially due to the mammalian target of rapamycin (mTOR) pathway regulated glucose-regulated protein 78 (GRP78) induction. In human CCA cells, the phosphorylation of eukaryotic initiation factor alpha (eIF2α) results in the accumulation of activating transcription factor 4 (ATF4) and GRP78 independent of unfolded protein response (UPR). Suppression of GRP78 expression decreases the proliferation and invasion of human CCA cells. It's notable that mTOR is required for eIF2α phosphorylation-induced ATF4 and GRP78 expression. Importantly, JNK promotes eIF2α/ATF4-mediated GRP78 induction through regulating the activity of mTOR. Thus, our study implicates JNK/mTOR signaling plays an important role in cholangiocarcinogenesis, partially through promoting the eIF2α/ATF4/GRP78 pathway.
Collapse
Affiliation(s)
- Chunhong Feng
- Department of Hepatobiliary Surgery of the Affiliated Hospital, Luzhou Medical College, Luzhou, Sichuan, China
| | - Kai He
- Department of Hepatobiliary Surgery of the Affiliated Hospital, Luzhou Medical College, Luzhou, Sichuan, China
| | - Chunyan Zhang
- Department of Biochemistry and Molecular Biology, Luzhou Medical College, Luzhou, Sichuan, China
| | - Song Su
- Department of Hepatobiliary Surgery of the Affiliated Hospital, Luzhou Medical College, Luzhou, Sichuan, China
| | - Bo Li
- Department of Hepatobiliary Surgery of the Affiliated Hospital, Luzhou Medical College, Luzhou, Sichuan, China
| | - Yuxiao Li
- Department of Biochemistry and Molecular Biology, Luzhou Medical College, Luzhou, Sichuan, China
| | - Chun-Yan Duan
- Department of Biochemistry and Molecular Biology, Luzhou Medical College, Luzhou, Sichuan, China
| | - Shaokun Chen
- Department of Biochemistry and Molecular Biology, Luzhou Medical College, Luzhou, Sichuan, China
| | - Run Chen
- Department of Public Health, Luzhou Medical College, Luzhou, Sichuan, China
| | - Youping Liu
- Department of Biochemistry and Molecular Biology, Luzhou Medical College, Luzhou, Sichuan, China
| | - Hong Li
- Department of Biochemistry and Molecular Biology, Luzhou Medical College, Luzhou, Sichuan, China
| | - Mei Wei
- Affiliated Hospital of Chinese Traditional Medicine, Luzhou Medical College, Luzhou, Sichuan, China
| | - Xianming Xia
- Department of Hepatobiliary Surgery of the Affiliated Hospital, Luzhou Medical College, Luzhou, Sichuan, China
| | - Rongyang Dai
- Department of Hepatobiliary Surgery of the Affiliated Hospital, Luzhou Medical College, Luzhou, Sichuan, China
- Department of Biochemistry and Molecular Biology, Luzhou Medical College, Luzhou, Sichuan, China
| |
Collapse
|
46
|
Niforou K, Cheimonidou C, Trougakos IP. Molecular chaperones and proteostasis regulation during redox imbalance. Redox Biol 2014; 2:323-32. [PMID: 24563850 PMCID: PMC3926111 DOI: 10.1016/j.redox.2014.01.017] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/11/2014] [Accepted: 01/18/2014] [Indexed: 02/05/2023] Open
Abstract
Free radicals originate from both exogenous environmental sources and as by-products of the respiratory chain and cellular oxygen metabolism. Sustained accumulation of free radicals, beyond a physiological level, induces oxidative stress that is harmful for the cellular homeodynamics as it promotes the oxidative damage and stochastic modification of all cellular biomolecules including proteins. In relation to proteome stability and maintenance, the increased concentration of oxidants disrupts the functionality of cellular protein machines resulting eventually in proteotoxic stress and the deregulation of the proteostasis (homeostasis of the proteome) network (PN). PN curates the proteome in the various cellular compartments and the extracellular milieu by modulating protein synthesis and protein machines assembly, protein recycling and stress responses, as well as refolding or degradation of damaged proteins. Molecular chaperones are key players of the PN since they facilitate folding of nascent polypeptides, as well as holding, folding, and/or degradation of unfolded, misfolded, or non-native proteins. Therefore, the expression and the activity of the molecular chaperones are tightly regulated at both the transcriptional and post-translational level at organismal states of increased oxidative and, consequently, proteotoxic stress, including ageing and various age-related diseases (e.g. degenerative diseases and cancer). In the current review we present a synopsis of the various classes of intra- and extracellular chaperones, the effects of oxidants on cellular homeodynamics and diseases and the redox regulation of chaperones. Free radicals originate from various sources and at physiological concentrations are essential for the modulation of cell signalling pathways. Abnormally high levels of free radicals induce oxidative stress and damage all cellular biomolecules, including proteins. Molecular chaperones facilitate folding of nascent polypeptides, as well as holding, folding, and/or degradation of damaged proteins. The expression and the activity of chaperones during oxidative stress are regulated at both the transcriptional and post-translational level.
Collapse
Key Words
- AGEs, Advanced Glycation End Products
- ALS, Autophagy Lysosome System
- AP-1, Activator Protein-1
- CLU, apolipoprotein J/Clusterin
- Chaperones
- Diseases
- EPMs, Enzymatic Protein Modifications
- ER, Endoplasmic Reticulum
- ERAD, ER-Associated protein Degradation
- Free radicals
- GPx7, Glutathione Peroxidase 7
- GRP78, Glucose Regulated Protein of 78 kDa
- HSF1, Heat Shock transcription Factor-1
- HSP, Heat Shock Protein
- Hb, Haemoglobin
- Keap1, Kelch-like ECH-associated protein 1
- NADH, Nicotinamide Adenine Dinucleotide
- NEPMs, Non-Enzymatic Protein Modifications
- NOS, Nitric Oxide Synthase
- NOx, NAD(P)H Oxidase
- Nrf2, NF-E2-related factor 2
- Oxidative stress
- PDI, Protein Disulfide Isomerase
- PDR, Proteome Damage Responses
- PN, Proteostasis Network
- Proteome
- RNS, Reactive Nitrogen Species
- ROS, Reactive Oxygen Species
- Redox signalling
- UPR, Unfolded Protein Response
- UPS, Ubiquitin Proteasome System
- α(2)M, α(2)-Macroglobulin
Collapse
Affiliation(s)
- Katerina Niforou
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, Athens 15784, Greece
| | - Christina Cheimonidou
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, Athens 15784, Greece
| | - Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, Athens 15784, Greece
| |
Collapse
|