1
|
PUMA overexpression dissociates thioredoxin from ASK1 to activate the JNK/BCL-2/BCL-XL pathway augmenting apoptosis in ovarian cancer. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166553. [PMID: 36122664 DOI: 10.1016/j.bbadis.2022.166553] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 09/13/2022] [Accepted: 09/13/2022] [Indexed: 11/23/2022]
Abstract
ASK1-JNK signaling promotes mitochondrial dysfunction-mediated apoptosis, but the bridge between JNK and apoptosis is not fully understood. PUMA induces apoptosis through BAX/BAK. Our previous study suggests a therapeutic potential of PUMA for ovarian cancer. However, whether and how PUMA activates ASK1 remains unclear. Here, we found for the first time that PUMA activated ASK1 by dissociating thioredoxin (TRX) from ASK1, however, it neither interacted with ASK1 nor TRX. Furthermore, PUMA overexpression caused ROS release from mitochondrial. H2O2 significantly impaired the interaction of ASK1 with TRX, whereas ROS scavenger NAC effectively abrogated the H2O2 effect, partly rescued PUMA-interfered interaction of ASK1 with TRX, and also abolished ASK1 phosphorylation. Interestingly, PUMA could not impair the association of ASK1 with TRX-C32S or TRX-C35S, two TRX mutants which are no longer oxidized in response to ROS. We further showed that PUMA activated ASK1-JNK axis to phosphorylate BCL-2 and BCL-XL, further augmenting apoptosis of ovarian cancer cells. In vivo, PUMA adenovirus combined with paclitaxel significantly inhibited intrinsically cisplatin-resistant ovarian cancer growth, and caused phosphorylation of BCL-2 and BCL-XL. Our results from human ovarian cancer TMA chips also revealed a positive correlation between PUMA expression and the phosphorylation of BCL-2 and BCL-XL. More importantly, all patients had no distal metastasis, implying a possibly clinical significance. Collectively, our results reveal a new pro-apoptotic signal amplification mechanism for PUMA by which PUMA overexpression first induces ROS-mediated dissociation of TRX from ASK1, and then causes JNK activation-triggering BCL-2/BCL-XL phosphorylation, ultimately augmenting apoptosis in ovarian cancer.
Collapse
|
2
|
Wei L, An T, An Y, He Z, Jia T, Li B, Lun Y. Transcriptome analysis of the effect of a novel human serine protease inhibitor SPINK13 on gene expression in MHCC97-H cells. Transl Cancer Res 2021; 10:4464-4477. [PMID: 35116303 PMCID: PMC8798559 DOI: 10.21037/tcr-21-1928] [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: 08/11/2021] [Accepted: 10/21/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Serine peptidase inhibitor, Kazal type 13 (SPINK13) (also known as hespinter) is a low-molecular-weight inhibitor of uPA that was discovered in 2006. It was detected in prokaryotic cells in 2013 for the first time and preliminarily shown to inhibit hepG2 liver cancer cells growth in vitro in 2015. In this study, the differentially transcribed genes of MHCC97-H cells caused by SPINK13 treatment were studied by transcriptomics and the molecular mechanism of SPINK13 suppressing tumor cells was proposed using bioinformatics. METHODS Preliminary study of the molecular mechanism of SPINK13's anti-cancer effect was performed by identifying potential target sites and signal pathways of SPINK13 through transcriptomics and bioinformatics analysis. RESULTS The results of the transcriptome study showed that there were 446 significantly differentially expressed genes between the experimental group and the blank control group, of which 347 genes were up-regulated and 99 genes were down-regulated. The Gene Ontology (GO) analysis showed that differentially expressed genes were enriched in cell growth regulation and cell division. They were enriched in the signal pathways of tumor transcription and cell cycle by Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis; there were 6 classical tumor signaling pathways (P<0.001): MAPK, apoptosis, tumor necrosis factor (TNF), cell cycle, p53, and transcriptional misregulation in cancer. There were 8 genes in 2 or more classical tumor signaling pathways at the same time: JUN, GADD45A, GADD45B, TNFRSF1A, FOS, CDKN1B, NFKBIA, and BBC3. The interaction analysis of the proteins encoded by the differentially expressed genes showed that there were 35 interaction nodes in the up-regulated genes and 2 interaction nodes in the down-regulated genes. CONCLUSIONS This study showed that SPINK13 inhibits hepatocellular carcinoma cell development by regulating the JNK, p53, and the IKK/NF-κB pathways, its potential targets for antitumor drugs may be JUN, GADD45A, GADD45B, TNFRSF1A, FOS, CDKN1B, NFKBIA, and BBC3.
Collapse
Affiliation(s)
- Ling Wei
- Department of Biotechnology, Beijing Centre for Physical and Chemical Analysis, Beijing, China
| | - Tao An
- Child Health Clinic of Beijing Yayuncun Amcare Women’s and Children’s Hospital, Beijing, China
| | - Yunhe An
- Department of Biotechnology, Beijing Centre for Physical and Chemical Analysis, Beijing, China
| | - Zheng He
- Clinical Laboratory Center, The First Medical Center of PLA General Hospital, Beijing, China
| | - Tingting Jia
- Department of Oral and Maxillofacial Surgery, The First Medical Center of PLA General Hospital, Haidian District, Beijing, China
| | - Baoming Li
- Department of Biotechnology, Beijing Centre for Physical and Chemical Analysis, Beijing, China
| | - Yongzhi Lun
- Department of Laboratory Medicine, School of Pharmacy and Medical Technology, Putian University, Putian, China
| |
Collapse
|
3
|
Lin J. Comments on "Dihydroartemisinin induces pyroptosis by promoting the AIM2/caspase-3/DFNA5 axis in breast cancer cells.". Chem Biol Interact 2021; 345:109551. [PMID: 34119495 DOI: 10.1016/j.cbi.2021.109551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/18/2021] [Accepted: 06/08/2021] [Indexed: 11/30/2022]
Abstract
We read the article "Dihydroartemisinin induces pyroptosis by promoting the AIM2/caspase-3/DFNA5 axis in breast cancer cells" published in Chemico-Biological Interactions. Authors revealed that dihydroartemisinin induced pyroptosis through activating the AIM2/caspase-3/DFNA5 pathway in breast cancer cells. However, some issues in this paper need to be commented. Authors suggested that dihydroartemisinin activated AIM2/caspase-3/DFNA5 axis in MCF-7 cell line. However, previous studies have confirmed that MCF-7 cell line does not express the caspase-3 protein. This makes us confused.
Collapse
Affiliation(s)
- Jiong Lin
- Department of Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| |
Collapse
|
4
|
Abbaspour A, Esmaeilzadeh A, Sharafi A. Suicide gene therapy-mediated purine nucleoside phosphorylase/fludarabine system for in vitro breast cancer model with emphasis on evaluation of vascular endothelial growth factor promoter efficacy. 3 Biotech 2021; 11:140. [PMID: 33708463 DOI: 10.1007/s13205-021-02692-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 02/12/2021] [Indexed: 02/07/2023] Open
Abstract
In this study, a suicide gene therapy approach was optimized by a non-viral polyplex system based on pEGFP-N1 vector harboring purine nucleoside phosphorylase gene conducted by vascular endothelial growth factor promoter for an in vitro breast cancer model (4T1 cell line). The VEGF promoter and purine nucleoside phosphorylase gene were cloned into the vector from the source of 4T1 and E. coli genomic DNA, respectively. A gene construct was developed by replacing VEGF promoter instead of CMV promoter in pEGFP-N1vector. PNP gene was integrated in to the multiple cloning site of the obtained vector. On the other hand, a construct from pEGFP-N1 harboring PNP gene under the control of the original CMV promoter was developed. The transfection method using cationic polymer was optimized based on N/P ratio, cell cytotoxicity, polyplex size, zeta potential and the green fluorescent protein (GFP) expression by fluorescent microscopy and flowcytometry. Also, the effect of hypoxia condition induced by 0.5 mM H2O2 on the promoter efficiency was investigated. The results showed that the performed gene delivery system is capable of the gene transfection to more than 30% of the cancer cells with both VEGF-PNP-pEGFP-N1 and PNP-pEGFP-N1 plasmids. The hypoxia condition did not show a significant effect on the VEGF promoter. But, it revealed that bystander effect can improve the efficacy of this system and reduce drug IC50 to 2 and fourfold for plasmids VEGF-PNP-pEGFP-N1 and PNP-pEGFP-N1, respectively. These results showed that the bystander effect could almost compensate the low efficiency of non-viral gene delivery systems. We suggest that the tumor-specific gene expression system mediated by the VEGF promoter can be especially useful in the present model of breast cancer gene therapy.
Collapse
Affiliation(s)
- Akbar Abbaspour
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Abdolreza Esmaeilzadeh
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Sharafi
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
5
|
Karyopherin-β1 Regulates Radioresistance and Radiation-Increased Programmed Death-Ligand 1 Expression in Human Head and Neck Squamous Cell Carcinoma Cell Lines. Cancers (Basel) 2020; 12:cancers12040908. [PMID: 32276424 PMCID: PMC7226044 DOI: 10.3390/cancers12040908] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/27/2020] [Accepted: 04/04/2020] [Indexed: 12/13/2022] Open
Abstract
Nuclear transport receptors, such as karyopherin-β1 (KPNB1), play important roles in the nuclear-cytoplasmic transport of macromolecules. Recent evidence indicates the involvement of nuclear transport receptors in the progression of cancer, making these receptors promising targets for the treatment of cancer. Here, we investigated the anticancer effects of KPNB1 blockage or in combination with ionizing radiation on human head and neck squamous cell carcinoma (HNSCC). HNSCC cell line SAS and Ca9-22 cells were used in this study. Importazole, an inhibitor of KPNB1, or knockdown of KPNB1 by siRNA transfection were applied for the blockage of KPNB1 functions. The roles of KPNB1 on apoptosis induction and cell surface expression levels of programmed death-ligand 1 (PD-L1) in irradiated HNSCC cells were investigated. The major findings of this study are that (i) blockage of KPNB1 specifically enhanced the radiation-induced apoptosis and radiosensitivity of HNSCC cells; (ii) importazole elevated p53-upregulated modulator of apoptosis (PUMA) expression via blocking the nuclear import of SCC-specific oncogene ΔNp63 in HNSCC cells; and (iii) blockage of KPNB1 attenuated the upregulation of cell surface PD-L1 expression on irradiated HNSCC cells. Taken together, these results suggest that co-treatment with KPNB1 blockage and ionizing radiation is a promising strategy for the treatment of HNSCC.
Collapse
|
6
|
Yang H, Zhang G, Che X, Yu S. Slug inhibition increases radiosensitivity of nasopharyngeal carcinoma cell line C666-1. Exp Ther Med 2018; 15:3477-3482. [PMID: 29545871 DOI: 10.3892/etm.2018.5844] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 10/20/2017] [Indexed: 02/06/2023] Open
Abstract
Slug is associated with the radioresistance of nasopharyngeal carcinoma (NPC) and the main current approach of treatment for NPC is radiotherapy. Hence, the aim of the current study was to determine the effect of Slug silencing on the radiosensitivity of NPC cells. Lentiviral-mediated transfection of Slug RNA interference (RNAi) in NPC cell line C666-1 was performed in vitro. Following Slug inhibition, its expression was detected using western blotting. A clonogenic survival assay and flow cytometry were then performed to evaluate the clonogenic cell survival, cell cycle distribution and apoptosis of C666-1 cells following irradiation. The results indicated that Slug RNAi decreased cell proliferation, and increased cell apoptosis and G0/G1 arrest. Thus, lentiviral-mediated transfection of Slug RNAi enhanced the radiosensitivity of the NPC cell line C666-1, and Slug may therefore be a potential target to improve radiotherapy in treatment of NPC and reduce the radioresistance of NPC.
Collapse
Affiliation(s)
- Hongxia Yang
- Department of Otorhinolaryngology, Maternal and Child Health Hospital of Tai'an, Tai'an, Shandong 271000, P.R. China
| | - Gang Zhang
- Department of Otolaryngology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, P.R. China
| | - Xiaolin Che
- Department of Otolaryngology, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250011, P.R. China
| | - Shudong Yu
- Department of Otolaryngology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
7
|
Yang J, Zhao X, Tang M, Li L, Lei Y, Cheng P, Guo W, Zheng Y, Wang W, Luo N, Peng Y, Tong A, Wei Y, Nie C, Yuan Z. The role of ROS and subsequent DNA-damage response in PUMA-induced apoptosis of ovarian cancer cells. Oncotarget 2017; 8:23492-23506. [PMID: 28423586 PMCID: PMC5410321 DOI: 10.18632/oncotarget.15626] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 02/14/2017] [Indexed: 02/05/2023] Open
Abstract
PUMA is a member of the "BH3-only" branch of the BCL-2 family. Our previous study suggests a therapeutic potential of PUMA in treating ovarian cancer, however, the action mechanism of PUMA remains elusive. In this work, we found that in PUMA adenovirus-infected A2780s ovarian cancer cells, exogenous PUMA was partially accumulated in the cytosol and mainly located to the mitochondria. We further showed that PUMA induces mitochondrial dysfunction-mediated apoptosis and ROS generation through functional BAX in a ROS generating enzyme- and caspase-independent manner irrespective of their p53 status, and results in activation of Nrf2/HO-1 pathway. Furthermore, PUMA induces DNA breaks in γ-H2AX staining, and causes activation of DNA damage-related kinases including ATM, ATR, DNA-PKcs, Chk1 and Chk2, which are correlated with the apoptosis. PUMA also results in ROS-triggered JNK activation. Intriguingly, JNK plays a dual role in both DNA damage response and apoptosis, and has an additional contribution to apoptosis. Taken together, we have provided new insight into the action mechanism by which elevated PUMA first induces ROS generation then results in DNA damage response and JNK activation, ultimately contributing to apoptosis in ovarian cancer cells.
Collapse
Affiliation(s)
- Jun Yang
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinyu Zhao
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mei Tang
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lei Li
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yi Lei
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ping Cheng
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wenhao Guo
- 2 Department of Abdominal Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yu Zheng
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Wang
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Na Luo
- 3 Nankai University, School of Medicine/Collaborative Innovation Center of Biotherapy, Tianjin 300071, China
| | - Yong Peng
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Aiping Tong
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuquan Wei
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chunlai Nie
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhu Yuan
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
8
|
Zhou Z, Zhou L, Jiang F, Zeng B, Wei C, Zhao W, Yu D. Downregulation of miR-222 Induces Apoptosis and Cellular Migration in Adenoid Cystic Carcinoma Cells. Oncol Res 2017; 25:207-214. [PMID: 28277192 PMCID: PMC7840837 DOI: 10.3727/096504016x14732772150460] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Previous studies have shown that miR-222 targets the p53 upregulated modulator of apoptosis (PUMA) to regulate cell biological behavior in some human malignancies. We hypothesized that there was a negative regulation, which might induce apoptosis, between miR-222 and PUMA in adenoid cystic carcinoma (ACC). In this study, the expression levels of miR-222 and the PUMA gene after transfection with antisense miR-222 (As-miR-222) were evaluated by RT-PCR and Western blot assays. Cell proliferation and migratory abilities were detected by CCK-8 and Transwell assays. Cell cycle and apoptosis were analyzed by flow cytometry. Our results showed that, when compared with the control and scramble-transfected groups, the expression of miR-222 in the As-miR-222 group was downregulated, while the expression of PUMA at both mRNA and protein levels was upregulated, cell proliferation and migratory abilities were inhibited, and apoptosis was increased. Our results suggested that As-miR-222 transfection could upregulate the expression of PUMA to induce apoptosis in ACC, providing a new concept for the treatment of ACC.
Collapse
Affiliation(s)
- Ziliang Zhou
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, P.R. China
| | | | | | | | | | | | | |
Collapse
|
9
|
Jiang F, Zhou L, Wei C, Zhao W, Yu D. Slug inhibition increases radiosensitivity of oral squamous cell carcinoma cells by upregulating PUMA. Int J Oncol 2016; 49:709-19. [PMID: 27277529 DOI: 10.3892/ijo.2016.3570] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/23/2016] [Indexed: 11/05/2022] Open
Abstract
As a new strategy, radio-gene therapy was widely used for the treatment of cancer patients in recent few years. Slug was involved in the radioresistance of various cancers and has been found to have an anti-apoptotic effect. This study aims to investigate whether the modulation of Slug expression by siRNA affects oral squamous cell carcinoma sensitivity to X-ray irradiation through upregulating PUMA. Two oral squamous cell carcinoma cell lines (HSC3 and HSC6) were transfected with small interfering RNA (siRNA) targeting Slug and subjected to radiotherapy in vitro. After transfection with Slug siRNA, both HSC3 and HSC6 cells showed relatively lower expression of Slug and higher expression of PUMA. The Slug siRNA transfected cells showed decreased survival and proliferation rates, an increased apoptosis rate and enhanced radiosensitivity to X-ray irradiation. Our results revealed that Slug siRNA transfection in combination with radiation increased the expression of PUMA, which contributed to radiosensitivity of oral squamous cell carcinoma cells. Thus, controlling the expression of Slug might contribute to enhance sensitivity of HSC3 and HSC6 cells toward X-ray irradiation in vitro by upregulating PUMA.
Collapse
Affiliation(s)
- Fangfang Jiang
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Lijie Zhou
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Changbo Wei
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Wei Zhao
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Dongsheng Yu
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, P.R. China
| |
Collapse
|
10
|
RANJBAR S, HASHEMZADEH MS, KHOSHTINAT NIKKHOI S, FARASAT A, TAT M, GHALAVAND M, DOROSTKAR R. Selective suppression of tumor cells by a tumor-specific bicistronic lentiviral vector. Turk J Biol 2016. [DOI: 10.3906/biy-1512-53] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
11
|
Lin B, Gao A, Zhang R, Ma H, Shen H, Hu Q, Zhang H, Zhao M, Lan X, Liu K. Use of a Novel Integrase-Deficient Lentivirus for Targeted Anti-Cancer Therapy With Survivin Promoter-Driven Diphtheria Toxin A. Medicine (Baltimore) 2015; 94:e1301. [PMID: 26252309 PMCID: PMC4616595 DOI: 10.1097/md.0000000000001301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
As an immunotoxin, diphtheria toxin has been widely used in gene therapy and gene function assays for its roles in protein synthesis inhibition, and the aim of our study is to set up a nonintegrating lentiviral system for specific expression of diphtheria toxin A (DTA) used in cancer gene therapy.Here, we established a lentiviral system that could coordinately express fluorescent protein and DTA driven by the cytomegalovirus (CMV) promoter, which is convenient for us to precisely trace the expression of DTA and monitor the process of lentivirus packaging. To achieve safer cancer therapy, we replaced the CMV promoter with the Survivin promoter, a specific promoter that is dramatically activated in cancer tissues and cells, but not in normal tissues and cells, and that will impose greater therapeutic potential because a significant expression difference occurred between these 2 groups. Meanwhile, we obtained integrase-deficient lentivirus (IDLV) after packaging with the integrase mutant, which expresses defective integrase RRK262263264AAH, to minimize the side effects that derived from the insertional mutagenesis of the host genome.Our results suggest that the IDLV system that we generated possesses therapeutic potential in cancers in vitro and in vivo.
Collapse
Affiliation(s)
- Baoshun Lin
- From the Institute for Laboratory Medicine, Fuzhou General Hospital, PLA, Fuzhou, Fujian, P.R. China (BL, AG, RZ, HM, MZ, XL, KL); Dong Fang Hospital, Xiamen University, Fuzhou, Fujian, P.R. China (BL, AG, QH, MZ, XL, KL); Institute for Laboratory Medicine, Fuzhou General Hospital, Second Military Medical University, Fuzhou, Fujian, P.R. China (HM); School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian, P.R. China (HS); Agriculture and Animal Husbandry College, Tibet University, Nyingchi, P.R. China (HZ); and Fuzhou General Hospital Clinical Medical School, Fujian Medical University, Fuzhou, P.R. China (RZ, XL, KL)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Carpenter RL, Han W, Paw I, Lo HW. HER2 phosphorylates and destabilizes pro-apoptotic PUMA, leading to antagonized apoptosis in cancer cells. PLoS One 2013; 8:e78836. [PMID: 24236056 PMCID: PMC3827261 DOI: 10.1371/journal.pone.0078836] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 09/24/2013] [Indexed: 12/29/2022] Open
Abstract
HER2 is overexpressed in 15–20% of breast cancers. HER2 overexpression is known to reduce apoptosis but the underlying mechanisms for this association remain unclear. To elucidate the mechanisms for HER2-mediated survival, we investigated the relationship between HER2 and p53 upregulated modulator of apoptosis (PUMA), a potent apoptosis inducer. Our results showed that HER2 interacts with PUMA, which was independent of HER2 activation. In addition, we observed that HER2 interacted with PUMA in both mitochondrial and non-mitochondrial compartments. We next examined whether HER2 phosphorylates PUMA. Notably, PUMA tyrosine phosphorylation has never been reported. Using an intracellular assay, we found PUMA to be phosphorylated in breast cancer cells with activated HER2. Via cell-free HER2 kinase assay, we observed that PUMA was directly phosphorylated by HER2. Activation of HER2 decreased PUMA protein half-life. To identify which of the three tyrosines within PUMA are targeted by HER2, we generated three PUMA non-phosphorylation mutants each with a single Tyr→Phe substitution. Results indicated that each PUMA single mutant had lost some, but not all phosphorylation by HER2 indicating that HER2 targets all three tyrosines. Consequently, we created an additional PUMA mutant with all three tyrosines mutated (TM-PUMA) that could not be phosphorylated by HER2. Importantly, TM-PUMA was found to have a longer half-life than PUMA. An inverse association was observed between HER2 and PUMA in 93 invasive breast carcinoma samples. We further found that TM-PUMA suppressed growth of breast cancer cells to a greater degree than PUMA. Also, TM-PUMA had a stronger propensity to induce apoptosis than PUMA. Together, our results demonstrate, for the first time, that PUMA can be tyrosine phosphorylated and that HER2-mediated phosphorylation destabilizes PUMA protein. The HER2-PUMA interplay represents a novel mechanism by which PUMA is regulated and a new molecular basis for HER2-mediated growth and survival of cancer cells.
Collapse
Affiliation(s)
- Richard L. Carpenter
- Division of Surgical Sciences, Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Woody Han
- Division of Surgical Sciences, Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Ivy Paw
- Division of Surgical Sciences, Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Hui-Wen Lo
- Division of Surgical Sciences, Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Center for RNA Biology, Duke University School of Medicine, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
13
|
Enhancement of expression of survivin promoter-driven CD/TK double suicide genes by the nuclear matrix attachment region in transgenic gastric cancer cells. Gene 2013; 534:177-82. [PMID: 24220851 DOI: 10.1016/j.gene.2013.10.064] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 10/27/2013] [Accepted: 10/29/2013] [Indexed: 01/10/2023]
Abstract
This work aimed to study a novel transgenic expression system of the CD/TK double suicide genes enhanced by the nuclear matrix attachment region (MAR) for gene therapy. The recombinant vector pMS-CD/TK containing the MAR-survivin promoter-CD/TK cassette was developed and transfected into human gastric cancer SGC-7901 cells. Expression of the CD/TK genes was detected by quantitative real-time PCR (qPCR) and Western blot. Cell viability and apoptosis were measured using the methyl thiazolyl tetrazolium (MTT) assay and flow cytometry. When the MAR fragment was inserted into the upstream of the survivin promoter, the qPCR result showed that the expression of the CD/TK genes significantly increased 7.7-fold in the transgenic SGC-7901 cells with plasmid pMS-CD/TK compared with that without MAR. MTT and flow cytometry analyses indicated that treatment with the prodrugs (5-FC+GCV) significantly decreased the cellular survival rate and enhanced the cellular apoptosis in the SGC-7901 cells. The expression of the CD/TK double suicide genes driven by the survivin promoter can be enhanced by the MAR fragment in human gastric cancer cells.
Collapse
|
14
|
Wang C, Zhou Y, Li S, Li H, Tian L, Wang H, Shang D. Anticancer mechanisms of temporin-1CEa, an amphipathic α-helical antimicrobial peptide, in Bcap-37 human breast cancer cells. Life Sci 2013; 92:1004-14. [PMID: 23583573 DOI: 10.1016/j.lfs.2013.03.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 03/12/2013] [Accepted: 03/27/2013] [Indexed: 12/21/2022]
Abstract
AIMS Temporin-1CEa, a 17-residue antimicrobial peptide, is known to exert broad-spectrum anticancer activity that acts preferentially on cancer cells instead of normal cells. However, the mechanism of cancer cell death induced by temporin-1CEa is weakly understood. MAIN METHODS Here, we investigated the cytotoxic and membrane-disrupting effects of temporin-1CEa on human breast cancer cell line Bcap-37, using MTT assay, electronic microscope observation, fluorescence imaging and flow cytometry analysis. KEY FINDINGS The MTT assay indicated that one-hour temporin-1CEa treatment led to rapid cell death in either caspase-dependent or -independent manner. The electronic microscope observation suggested that temporin-1CEa exposure resulted in profound morphological changes in Bcap-37 cells. The fluorescence imaging and flow cytometry analysis demonstrated that temporin-1CEa exhibited membrane-disrupting property characterized by induction of cell-surface phosphatidylserine exposure, elevation of plasma membrane permeability, and rapid transmembrane potential depolarization. Moreover, temporin-1CEa might also induce rapid cell death through mitochondria-involved mechanisms, including rapid intracellular Ca(2+) leakage, collapse of mitochondrial membrane potential (Δφm) and over-generation of reactive oxygen species (ROS). SIGNIFICANCE In summary, the present study indicates that temporin-1CEa triggers a rapid cytotoxicity in Bcap-37 cells through membrane-destruction and intracellular mechanisms involving mitochondria. These intracellular mechanisms and direct membrane-destruction effect were evaluated helping to understand the detail action of antimicrobial peptides in mammalian cancer cells.
Collapse
Affiliation(s)
- Che Wang
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian 116029, China
| | | | | | | | | | | | | |
Collapse
|
15
|
Ma ZY, Qiao X, Xie CZ, Shao J, Xu JY, Qiang ZY, Lou JS. Activities of a novel Schiff Base copper(II) complex on growth inhibition and apoptosis induction toward MCF-7 human breast cancer cells via mitochondrial pathway. J Inorg Biochem 2012; 117:1-9. [DOI: 10.1016/j.jinorgbio.2012.08.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Revised: 08/16/2012] [Accepted: 08/17/2012] [Indexed: 01/03/2023]
|
16
|
Liu D, Shi P, Yin X, Chen Z, Zhang X. Effect of norcantharidin on the human breast cancer Bcap-37 cells. Connect Tissue Res 2012; 53:508-12. [PMID: 22606958 DOI: 10.3109/03008207.2012.694928] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Norcantharidin (NCTD), a chemically modified form of cantharidin, is a potential anticancer drug. In this study, the effects of NCTD on the cellular viability, reactive oxygen species (ROS), mitochondrial membrane potential (MMP), and DNA damage in the human breast cancer cell line Bcap-37 were investigated with confocal and fluorescence microscopy. The cell cycle was further analyzed using the CellQuest software of a Becton-Dickinson FACS flow cytometer. The results indicated that the cellular viability was decreased with the growing concentrations of NCTD and time exposure. Moreover, the fluorescence intensity of ROS was increased, whereas the MMP was decreased in Bcap-37 cells with the growing concentrations of NCTD. NCTD induced a dose-dependent DNA damage and reduced the G1 peak in Bcap-37 cells. The G2/M peak of Bcap-37 was also decreased by the higher concentration of NCTD.
Collapse
Affiliation(s)
- Dongwu Liu
- School of Life Sciences, Shandong University of Technology, Zibo, China
| | | | | | | | | |
Collapse
|
17
|
Wang H, Pei W, Luan Q, Ma F, Zhou S, Zhao Z, Meng X, Zhang X, Liang X, Chen Y, Zhan Q, Lin C, Qian H, Zhao P. A feasibility study on gene therapy of pancreatic carcinoma with Ad-PUMA. Cancer Biol Ther 2012; 13:712-9. [PMID: 22617775 DOI: 10.4161/cbt.20552] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer is one of the most malignant tumors with high mortality and poor prognosis even with the aggressive conventional therapies. Biotherapy based on the understanding of tumorigenesis mechanism is ongoing to improve the outcomes of cancer patients. We sought here to evaluate the therapeutic potential of a proapoptotic gene, PUMA, in pancreatic cancer. We found that PUMA was differently expressed in a series of pancreatic ductal adenocarcinoma cancer cell lines, and adenovirus-mediated expression of PUMA (Ad-PUMA) in these cells resulted in massive apoptosis. PUMA was more potent than p53 in suppressing growth of cancer cells. RT-PCR and Western Blot revealed that exogenous PUMA was expressed 6 h after Ad-PUMA infection. Furthermore, we assessed the efficacy of Ad-PUMA combining anticancer drugs (5-fluorouracil, cisplatin, gemcitabine hydrochloride, respectively) in these pancreatic cancer cell lines. Data revealed that PUMA significantly sensitized pancreatic carcinoma cell lines to chemotherapeutics, which may be resulted from abundant apoptosis induction. In nude mice with PANC-1 xenografts, Ad-PUMA treatment significantly inhibited the tumor growth. These results suggest that PUMA is a potent molecular tool in suppressing tumor growth sensitizing pancreatic carcinoma cells to chemical drugs. PUMA plays roles in negatively regulating cancer cell growth and may be a promising tool for cancer biotherapy, with or without combination with chemotherapeutic agents.
Collapse
Affiliation(s)
- Haijuan Wang
- State Key Laboratory of Molecular Oncology, Cancer Institute/Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Jha K, Shukla M, Pandey M. Survivin expression and targeting in breast cancer. Surg Oncol 2011; 21:125-31. [PMID: 21334875 DOI: 10.1016/j.suronc.2011.01.001] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 12/27/2010] [Accepted: 01/08/2011] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Survivin a multifunctional protein that controls cell division, inhibition of apoptosis and promotion of angiogenesis. It is expressed in most human neoplasm, but is absent in normal and differentiated tissues. The purpose of this article is to overview the expression of survivin, effect of its expression in response to treatment, correlation with other markers and newer advancement in targeting survivin. METHODS A detailed search of Medline was carried out using the following search strategy: "((survivin) OR ((apoptosis) AND (inhibitor OR inhibitors))) AND ((breast) AND (neoplasm OR neoplasms OR tumor OR tumor OR cancer OR carcinoma))". Abstract of all articles thus identified were reviewed to identify the relevant studies, full articles of studies thus identified were then obtained and reviewed. All relevant data was extracted and tabulated. RESULTS Survivin expression by Immunohistochemistry was identified in 65.3% (55.2-90.0%) of the breast cancer patients among the identified studies while survivin mRNA by RT-PCR was identified in 93.6% (90-97%). Survivin expression has been reported to be associated with over expression of HER 2, vascular endothelial growth factor (VEGF), urokinase plasminogen activator (uPA)/PAI-1. CONCLUSION Survivin is over expressed in majority of breast cancers. The over expression of survivin is found to correlate with HER 2 and EGFR expression. Survivin expression has been found to confer resistance to chemotherapy and radiation. Targeting survivin in experimental models improves survival. More studies are needed on the role of survivin in multi drug resistance (MDR) in the presence of Pgp/uPA/PAI-1 and the impact of survivin over expression in triple negative breast cancer.
Collapse
Affiliation(s)
- Kumkum Jha
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | | | | |
Collapse
|
19
|
Yuan Z, Cao K, Lin C, Li L, Liu HY, Zhao XY, Liu L, Deng HX, Li J, Nie CL, Wei YQ. The p53 upregulated modulator of apoptosis (PUMA) chemosensitizes intrinsically resistant ovarian cancer cells to cisplatin by lowering the threshold set by Bcl-x(L) and Mcl-1. Mol Med 2011; 17:1262-74. [PMID: 21863213 PMCID: PMC3321807 DOI: 10.2119/molmed.2011.00176] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2011] [Accepted: 08/17/2011] [Indexed: 02/05/2023] Open
Abstract
Ovarian cancer is the number one cause of death from gynecologic malignancy. A defective p53 pathway is a hallmark of ovarian carcinoma. The p53 mutation correlates significantly with resistance to platinum-based chemotherapy, early relapse and shortened overall survival in ovarian cancer patients. PUMA (p53 upregulated modulator of apoptosis), a BH3-only Bcl-2 family protein, was recently identified as a transcriptional target of p53 and a potent apoptosis inducer in various cancer cells. In this study, we showed that the induction of PUMA by cisplatin was abolished in p53-deficient SKOV3 cells. Elevated expression of PUMA-induced apoptosis and sensitized A2780s and SKOV3 ovarian cancer cells to cisplatin, and the combination of PUMA and low-dose cisplatin, significantly suppressed xenograft tumor growth in vivo through enhanced induction of apoptosis compared with treatment with PUMA or cisplatin alone. The effects of PUMA were mediated by enhanced caspase activation and release of cytochrome c and Smac (second mitochondria-derived activator of caspase) into the cytosol. Furthermore, PUMA chemosensitized intrinsically resistant SKOV3 cells to cisplatin through downregulation of B-cell lymphoma-extra large (Bcl-x(L)) and myeloid cell leukemia sequence 1 (Mcl-1). PUMA-mediated Bcl-x(L) downregulation mainly happened at the transcription level, whereas PUMA-induced Mcl-1 down-regulation was associated with caspase-dependent cleavage and proteasome-mediated degradation. To our knowledge, these data suggest a new mechanism by which overexpression of PUMA enhances sensitivity of SKOV3 cells to cisplatin by lowering the threshold set simultaneously by Bcl-x(L) and Mcl-1. Taken together, our findings indicate that PUMA is an important modulator of therapeutic responses of ovarian cancer cells and is potentially useful as a chemosensitizer in ovarian cancer therapy.
Collapse
Affiliation(s)
- Zhu Yuan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
- Address correspondence and reprint requests to Zhu Yuan or Chun-lai Nie, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, #1 Keyuan Road 4, Gaopeng, 610041, China. Phone: +86-28-85164063; Fax: +86-28-85164060; E-mails: ,
| | - Kang Cao
- Department of Pathogen Biology, Chengdu Medical College, Chengdu, China
| | - Chao Lin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Lei Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Huan-yi Liu
- Cancer Center, Chengdu Military General Hospital, Chengdu, China
| | - Xin-yu Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Lei Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Hong-xin Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Jiong Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Chun-lai Nie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
- Address correspondence and reprint requests to Zhu Yuan or Chun-lai Nie, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, #1 Keyuan Road 4, Gaopeng, 610041, China. Phone: +86-28-85164063; Fax: +86-28-85164060; E-mails: ,
| | - Yu-quan Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Stimulation of the endosomal TLR pathway enhances autophagy-induced cell death in radiotherapy of breast cancer. Genes Genomics 2010. [DOI: 10.1007/s13258-010-0139-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
21
|
Ryan BM, O'Donovan N, Duffy MJ. Survivin: a new target for anti-cancer therapy. Cancer Treat Rev 2009; 35:553-62. [PMID: 19559538 DOI: 10.1016/j.ctrv.2009.05.003] [Citation(s) in RCA: 299] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2009] [Revised: 05/13/2009] [Accepted: 05/15/2009] [Indexed: 12/22/2022]
Abstract
Survivin is one of the most cancer-specific proteins identified to date, being upregulated in almost all human tumors. Biologically, survivin has been shown to inhibit apoptosis, enhance proliferation and promote angiogenesis. Because of its upregulation in malignancy and its key role in apoptosis, proliferation and angiogenesis, survivin is currently attracting considerable attention as a new target for anti-cancer therapies. In several animal model systems, downregulation of survivin or inactivation of its function has been shown to inhibit tumor growth. Strategies under investigation to target survivin include antisense oligonucleotides, siRNA, ribozymes, immunotherapy and small molecular weight molecules. The translation of these findings to the clinic is currently ongoing with a number of phase I/II clinical trials targeting survivin in progress. These include use of the antisense oligonucleotide LY2181308, the low molecular weight molecule inhibitor YM155 and survivin-directed autologous cytotoxic T lymphocytes. The optimum use of survivin antagonists in the treatment of cancer is likely to be in combination with conventional cancer therapies.
Collapse
Affiliation(s)
- Bríd M Ryan
- Cancer Prevention Fellowship Program, Office of Preventive Oncology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4258, USA.
| | | | | |
Collapse
|
22
|
Abstract
PUMA (p53 upregulated modulator of apoptosis) is a Bcl-2 homology 3 (BH3)-only Bcl-2 family member and a critical mediator of p53-dependent and -independent apoptosis induced by a wide variety of stimuli, including genotoxic stress, deregulated oncogene expression, toxins, altered redox status, growth factor/cytokine withdrawal and infection. It serves as a proximal signaling molecule whose expression is regulated by transcription factors in response to these stimuli. PUMA transduces death signals primarily to the mitochondria, where it acts indirectly on the Bcl-2 family members Bax and/or Bak by relieving the inhibition imposed by antiapoptotic members. It directly binds and antagonizes all known antiapoptotic Bcl-2 family members to induce mitochondrial dysfunction and caspase activation. PUMA ablation or inhibition leads to apoptosis deficiency underlying increased risks for cancer development and therapeutic resistance. Although elevated PUMA expression elicits profound chemo- and radiosensitization in cancer cells, inhibition of PUMA expression may be useful for curbing excessive cell death associated with tissue injury and degenerative diseases. Therefore, PUMA is a general sensor of cell death stimuli and a promising drug target for cancer therapy and tissue damage.
Collapse
Affiliation(s)
- J Yu
- Department of Pathology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - L Zhang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|