1
|
Tan S, Fu L, Dong Q. AATF is Overexpressed in Human Bladder Cancer and Regulates Chemo-Sensitivity Through Survivin. Onco Targets Ther 2022; 14:5493-5505. [PMID: 35002255 PMCID: PMC8721289 DOI: 10.2147/ott.s319734] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 10/06/2021] [Indexed: 01/17/2023] Open
Abstract
Objective Dysregulation of apoptosis antagonizing transcription factor (AATF) has been reported to be closely associated with human cancers. However, its involvement in human bladder cancer (BC) remains unexplored. This study aimed to investigate the clinical significance and biological roles of AATF in human bladder cancers. Methods AATF protein expression was examined in 107 cases of bladder cancer tissues using immunohistochemistry. AATF plasmid transfection and small interfering RNA (siRNA) knockdown were performed in T24 and 5637 cell lines. CCK-8, colony formation, annexin V/PI, JC-1 staining, and Western blotting were carried out to investigate the biological roles and underlying mechanisms of AATF in bladder cancer cells. Results Our results showed that AATF expression was upregulated in human bladder cancer specimens and correlated with T stage. Analysis of the Oncomine database showed elevation of AATF mRNA in BC tissues. The Cancer Genome Atlas (TCGA) data suggested that high AATF expression correlated with poor patient survival. Western blotting showed that AATF protein expression was higher in BC cell lines compared to normal bladder transitional epithelial cell line SV-HUC-1. CCK-8 and colony assays showed that ectopic AATF expression upregulated cell growth rate and colony numbers. CCK-8, annexin V/propidium iodide (PI), JC-1 assays and Western blotting showed that AATF overexpression decreased cisplatin sensitivity, downregulated cisplatin-induced apoptosis and upregulated mitochondrial membrane potential, with decreased cytochrome c and cleaved-PARP expression. AATF siRNA knockdown showed the opposite effects. Mechanistically, AATF overexpression upregulated cyclin E and Survivin at both mRNA and protein levels. The decreased cisplatin sensitivity/apoptosis induced by ectopic AATF were reversed after treatment with Survivin inhibitor YM155. Conclusion Our results showed that AATF was overexpressed in human bladder cancers and promoted malignant behavior by regulating cyclin E and Survivin, indicating AATF could serve as a malignant biomarker and potential therapeutic target in BC.
Collapse
Affiliation(s)
- Shutao Tan
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Lin Fu
- Department of Pathology, College of Basic Medical Science, China Medical University and Department of Pathology, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Qianze Dong
- Department of Pathology, College of Basic Medical Science, China Medical University and Department of Pathology, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
2
|
Zhang Y, Bai X, Zhang Y, Li Y. Daam1 Overexpression Promotes Gastric Cancer Progression and Regulates ERK and AKT Signaling Pathways. Onco Targets Ther 2021; 14:4609-4619. [PMID: 34475767 PMCID: PMC8408046 DOI: 10.2147/ott.s316157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Objective The dishevelled-associated activator of morphogenesis 1 (DAAM1) has been reported to be closely associated with human cancers. However, its involvement in human gastric cancer (GC) remains largely unexplored. This study aimed to investigate the clinical significance and biological roles of Daam1 in human GC. Methods Daam1 protein expression was examined in 124 cases of gastric adenocarcinomas using immunohistochemistry. Daam1 plasmid and siRNA transfection were carried out in SGC7901 and AGS cell lines. CCK-8, colony formation, Annexin V/PI, JC-1 staining, and Western blotting were used to explore the biological functions and potential underlying mechanisms of Daam1 in GC cells. Results Our results showed that Daam1 was overexpressed in GC specimens. A high Daam1 level was associated with tumor-node-metastasis (TNM) stage, T status, nodal metastasis, and poor patient survival. Analysis of the Oncomine dataset revealed upregulation of Daam1 mRNA in GC tissues. Western blot showed that Daam1 protein expression was higher in GC cell lines compared to the normal GES-1 cell line. CCK-8 and colony formation assays showed that ectopic Daam1 expression upregulated the cell growth rate and colony number in SGC-7901 cells, while Daam1 siRNA knockdown downregulated the growth rate and colony number in AGS cells. CCK-8 and Annexin V/PI apoptosis assays demonstrated that Daam1 overexpression decreased cisplatin sensitivity and downregulated cisplatin-induced apoptosis. JC1 staining showed that Daam1 overexpression upregulated, while Daam1 depletion downregulated mitochondrial membrane potential. Mechanistically, Daam1 overexpression downregulated p21 and upregulated p-ERK and p-AKT. The increased proliferation rate and decreased cisplatin sensitivity/apoptosis induced by ectopic Daam1 were reversed after treatment with AKT and ERK inhibitors. Conclusion Taken together, our results showed that Daam1 overexpression was associated with poor prognosis and promoted malignant activity via regulation of ERK and AKT pathways in GC cells, indicating Daam1 is a malignant biomarker and potential therapeutic target in GC.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Xue Bai
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yi Zhang
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yan Li
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
3
|
Wang H, Wang F, Ouyang W, Jiang X, Wang Y. BCAT1 overexpression regulates proliferation and c‑Myc/GLUT1 signaling in head and neck squamous cell carcinoma. Oncol Rep 2021; 45:52. [PMID: 33760210 PMCID: PMC7962101 DOI: 10.3892/or.2021.8003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 11/27/2020] [Indexed: 12/26/2022] Open
Abstract
Branched chain amino acid transaminase 1 (BCAT1) overexpression has been reported in various cancers; however, at present, its significance and biological role in head and neck squamous cell carcinoma (HNSCC) remain unknown. BCAT1 protein expression was upregulated in 56/106 (52.8%) cases of HNSCC. BCAT1 overexpression was associated with tumor-node-metastasis stage, tumor stage and nodal metastasis. The Cancer Genome Atlas data suggested that high BCAT1 expression was associated with poor patient survival. Oncomine data suggested that BCAT1 expression was increased in HNSCC. Functionally, BCAT1 overexpression promoted cell proliferation, colony formation, invasion and cisplatin resistance in FaDu cells. BCAT1 overexpression also upregulated the mitochondrial membrane potential, and increased ATP production, glucose consumption and glucose uptake. Western blotting demonstrated that BCAT1 overexpression upregulated c-Myc and glucose transporter 1 (GLUT1) protein levels. Depletion of c-Myc using small interfering RNA abolished the influence of BCAT1 on GLUT1. Chromatin immunoprecipitation assays demonstrated that c-Myc has binding sites in the GLUT1 promoter. Collectively, the present findings suggested that BCAT1 is upregulated in human HNSCC and regulates HNSCC cell proliferation, invasion, cisplatin sensitivity and c-Myc/GLUT1 signaling.
Collapse
Affiliation(s)
- Hongming Wang
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Fei Wang
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Wenyu Ouyang
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xuejun Jiang
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yan Wang
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
4
|
Rab11a Is Overexpressed in Gastric Cancer and Regulates FAK/AKT Signaling. JOURNAL OF ONCOLOGY 2020; 2020:3494396. [PMID: 33178272 PMCID: PMC7648696 DOI: 10.1155/2020/3494396] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/15/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023]
Abstract
Dysregulation of Rab11a has been implicated in the progression of several cancers. However, there have been no such studies for human gastric cancers. In the current study, we examined Rab11a protein expression and found it was upregulated in 49 of 108 gastric cancer tissues and correlated with local invasion, nodal metastasis, and advanced stage. Rab11a protein was higher in gastric cancer cell lines than normal gastric cell line. We transfected Rab11a plasmid and siRNA in both MGC803 and AGS cell lines. Rab11a overexpression increased the cell growth rate, colony numbers, and invasion ability in both MGC803 and AGS cell lines. Downregulation of Rab11a using siRNA decreased the cell proliferation rate, colony numbers, and inhibited invasion. Rab11a overexpression also conferred cisplatin resistance. Annexin V/PI staining showed that Rab11a overexpression suppressed cisplatin-induced apoptosis, while Rab11a depletion promoted cell apoptosis. We also showed that Rab11a overexpression maintained mitochondrial membrane potential. Western blot analysis revealed that Rab11a increased protein expression of MMP2, cyclin D1, Bcl-2, p-FAK, and p-AKT, while Rab11a depletion showed the opposite effects. Blockage of FAK using inhibitor downregulated Bcl-2, cyclin D1, MMP2, and p-AKT expression and abolished the effects of Rab11a on these proteins. In summary, our data demonstrated that Rab11a is upregulated in human gastric cancers. Rab11a facilitated cell proliferation and invasion, as well as cisplatin sensitivity and mitochondrial membrane potential, possibly via the FAK/AKT signaling pathway.
Collapse
|
5
|
Ji X, Guo X, Wang Y, Li X, Li H. Rab18 Regulates Proliferation, Invasion and Cisplatin Sensitivity Through STAT3 Signaling in Head and Neck Squamous Cell Carcinoma. Onco Targets Ther 2020; 13:4123-4134. [PMID: 32494165 PMCID: PMC7231766 DOI: 10.2147/ott.s238503] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 04/14/2020] [Indexed: 12/23/2022] Open
Abstract
Introduction The clinical significance, biological roles and potential mechanism of Rab18 remain unknown in most human cancers, including head and neck squamous cell carcinoma (HNSCC). Methods We used immunohistochemistry to examine Rab18 protein expression in 112 cases of HNSCC specimens. We overexpressed and knockdown Rab18 in FaDu and Detroit562 cancer cell lines. Biological roles and mechanisms of Rab18 were examined using MTT, colony formation, Matrigel invasion assay, Western blotting, Annexin V and JC1 staining. Results Rab18 was upregulated in 45/112 (40.2%) cases of HNSCC tissues, which correlated with advanced T classification, positive nodal metastasis and tumor node metastasis (TNM) stage. The Oncomine and The Cancer Genome Atlas (TCGA) analyses indicated that Rab18 was elevated in human HNSCC tissues and correlated with poor patient survival. Functionally, Rab18 overexpression increased growth rate, colony numbers, cell cycle progression and invading ability in FaDu cells. Rab18 downregulated cisplatin-induced apoptosis and upregulated the mitochondrial membrane potential (Δψm). Western blot revealed that Rab18 overexpression induced epithelial-to-mesenchymal transition, with downregulation of E-cadherin and upregulation of N-cadherin, Vimentin and Twist. Rab18 overexpression also upregulated Survivin protein and Rab18 knockdown showed the opposite effects on these proteins. Treatment of STAT3 inhibitor, SH-4-54, inhibited cell invasion, increased E-cadherin and downregulated N-cadherin, Twist and Survivin. SH-4-54 also abolished the effects of BCAT1 on these proteins, as well as cell invasion. Conclusion In summary, our data showed that Rab18 was overexpressed in human HNSCC and functioned as an oncoprotein. Rab18 regulated HNSCC cell proliferation, invasion and cisplatin sensitivity through STAT3 signaling in HNSCC.
Collapse
Affiliation(s)
- Xu Ji
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Xing Guo
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yan Wang
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Xiaotian Li
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Hong Li
- Department of Otolaryngology, The Fourth Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
6
|
Sirt5 Attenuates Cisplatin-Induced Acute Kidney Injury through Regulation of Nrf2/HO-1 and Bcl-2. BIOMED RESEARCH INTERNATIONAL 2019; 2019:4745132. [PMID: 31815138 PMCID: PMC6878818 DOI: 10.1155/2019/4745132] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/11/2019] [Accepted: 08/29/2019] [Indexed: 12/30/2022]
Abstract
Cisplatin- (CDDP) induced acute kidney injury (AKI) limits the clinical use of cisplatin. Several sirtuin (SIRT) family proteins are involved in AKI, while the roles of Sirt5 in cisplatin-induced AKI remain unknown. In the present study, we characterized the role and mechanism of Sirt5 in cisplatin-induced apoptosis using the human kidney 2 (HK-2) cell line. CDDP treatment decreased Sirt5 expression of HK-2 cells in a dose-dependent manner. In addition, Sirt5 overexpression enhanced the metabolic activity in CDDP-treated HK-2 cells while Sirt5 siRNA attenuated it. Forced expression of Sirt5 inhibited CDDP-induced apoptosis while Sirt5 siRNA showed the opposite effects. Accordingly, Sirt5 overexpression inhibited the level of caspase 3 cleavage and cytochrome c levels. Furthermore, we found that Sirt5 increased mitochondrial membrane potentials and ameliorated intracellular ROS production. Mitotracker Red staining indicated that Sirt5 overexpression was able to maintain the mitochondrial density during CDDP treatment. We also investigated possible downstream targets of Sirt5 and found that Sirt5 increased Nrf2, HO-1, and Bcl-2 while it decreased Bax protein expression. Sirt5 siRNA showed the opposite effect on these proteins. The levels of Nrf2, HO-1, and Bcl-2 proteins in HK-2 cells were also decreased after CDDP treatment. Moreover, Nrf2 and Bcl-2 siRNA partly abolished the protecting effect of Sirt5 on CDDP-induced apoptosis and cytochrome c release. Catalase inhibitor 3-AT also abolished the cytoprotective effect of Sirt5. Together, the results demonstrated that Sirt5 attenuated cisplatin-induced apoptosis and mitochondrial injury in human kidney HK-2 cells, possibly through the regulation of Nrf2/HO-1 and Bcl-2.
Collapse
|
7
|
Morin Hydrate Reverses Cisplatin Resistance by Impairing PARP1/HMGB1-Dependent Autophagy in Hepatocellular Carcinoma. Cancers (Basel) 2019; 11:cancers11070986. [PMID: 31311167 PMCID: PMC6678360 DOI: 10.3390/cancers11070986] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/05/2019] [Accepted: 07/12/2019] [Indexed: 12/24/2022] Open
Abstract
Chemoresistance is a major obstacle that limits the benefits of cisplatin-based chemotherapy in various cancers, including hepatocellular carcinoma. De-regulation of the poly(ADP-ribose) polymerase 1 (PARP1)/high-mobility group box 1 (HMGB1) signaling pathway has been proposed as an important mechanism involved in cisplatin-resistance. In this study, we investigated therapeutic potential of a natural flavonoid Morin hydrate against cisplatin-induced toxicity using the HepG2DR multi-drug resistant cell line, which is derived from the HepG2 human hepatocellular carcinoma cell line. HepG2DR cells were exposed to cisplatin and Morin hydrate alone or together after which autophagy and apoptotic signaling pathways were monitored by fluorometric assay and Western blot analysis. Xenograft mouse models were performed to confirm the in vitro effect of Morin hydrate. PARP1 was hyper activated in cisplatin-resistant HepG2DR cells. Cisplatin-induced PARP1 activation resulted in chemoresistance via increased autophagy. The cisplatin/Morin hydrate combination was effective in the reversal of the HepG2DR cell resistance via suppression of PARP1-mediated autophagy by regulating the HMGB1 and microtubule-associated protein 1A/1B light chain 3B (LC3) I/II. Moreover, PARP1 inhibition by 4-amino-1,8-naphthalimide or autophagy inhibition by a knockdown of the autophagy-related 5 (ATG5) gene resulted in sensitizing the HepG2DR cells to cisplatin (CP) through activation of the c-Jun N-terminal kinase (JNK) pathway. In a mouse xenograft model, the treatment of cisplatin with Morin hydrate reversed the increased expression of PARP and HMGB1 and significantly suppressed tumor growth. These findings indicate dysregulated expression of PARP1 confers cisplatin-resistance via autophagy activation in HepG2DR cells. Morin hydrate inhibits cisplatin-mediated autophagy induction, resulting in increased susceptibility of HepG2DR cells to cisplatin cytotoxicity. The combination of Morin hydrate with cisplatin may be a promising therapeutic strategy to enhance the efficacy of conventional chemotherapeutic drugs.
Collapse
|
8
|
Li H, Fu L, Liu B, Lin X, Dong Q, Wang E. Ajuba overexpression regulates mitochondrial potential and glucose uptake through YAP/Bcl-xL/GLUT1 in human gastric cancer. Gene 2019; 693:16-24. [DOI: 10.1016/j.gene.2019.01.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 12/28/2018] [Accepted: 01/22/2019] [Indexed: 02/06/2023]
|
9
|
Du Y, Zhang W, Du B, Zang S, Wang X, Mao X, Hu Z. TRIM32 overexpression improves chemoresistance through regulation of mitochondrial function in non-small-cell lung cancers. Onco Targets Ther 2018; 11:7841-7852. [PMID: 30464531 PMCID: PMC6225845 DOI: 10.2147/ott.s176689] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background TRIM32 is overexpressed in several human cancers. However, its expression pattern, biological characteristics and mechanisms in human non-small cell lung cancer (NSCLC) have not been reported. Methods We examined TRIM32 protein in 115 cases of NSCLC specimens. TRIM32 plasmid transfection and siRNA knockdown was carried out in NSCLC cell lines. AnnexinV/PI and JC-1 staining were performed to examine the change of apoptosis and mitochondrial membrane potential. Western blot was used to detect change of downstream proteins. Results We found that TRIM32 protein was upregulated in 69 cases and positively correlated with advanced TNM stage. TRIM32 overexpression also correlated with poor survival of NSCLC patients. Biological assays demonstrated that TRIM32 overexpression promoted while it depletion inhibited cell growth, colony formation and invasion. In addition, TRIM32 maintained NSCLC cell viability and reduced apoptosis when treated with cisplatin. JC-1 and CellRox staining demonstrated that TRIM32 could maintain mitochondrial membrane potential and reduce Reactive Oxygen Species (ROS) production after cisplatin treatment. Western blot analysis showed that TRIM32 overexpression downregulated caspase 3 cleavage and cytochrome c release. TRIM32 also positively regulated Bcl-2 protein expression and NF-κB signaling. Inhibition of NF-κB abolished the effects of TRIM32 on Bcl-2. Conclusion Taken together, our results indicated that TRIM32 is overexpressed in NSCLC and regulates cisplatin resistance, possibly through NF-κB and Bcl-2.
Collapse
Affiliation(s)
- Yaming Du
- Department of Vascular Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Wei Zhang
- Department of Vascular Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Binghui Du
- Department of Vascular Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Sheng Zang
- Department of Vascular Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Xinpeng Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Xin Mao
- Department of Vascular Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Zhansheng Hu
- Department of Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China,
| |
Collapse
|
10
|
Wu B, Qi R, Liu X, Qian L, Wu Z. Rab18 overexpression promotes proliferation and chemoresistance through regulation of mitochondrial function in human gastric cancer. Onco Targets Ther 2018; 11:7805-7820. [PMID: 30464528 PMCID: PMC6225851 DOI: 10.2147/ott.s170829] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Dysregulation of Rab18 has been implicated in human cancers. However, its clinical significance and biological function in gastric cancer have not been investigated. Methods We examined Rab18 expression in gastric cancer tissues using immunohistochemistry. We used SNU-1 and AGS cell lines for plasmid and siRNA transfection respectively. MTT, colony formation assay, cell cycle analysis, matrigel invasion, wound healing assay, AnnexinV/PI analysis and western blotting were used to examine the biological effect and mechanism of Rab18 in gastric cancer cell lines. Results Rab18 protein expression was upregulated in gastric cancer tissues and this correlated with advanced stage and poor prognosis. Rab18 overexpression promoted proliferation in vitro and in vivo. Cell cycle analysis showed that Rab18 overexpression upregulated, while its depletion downregulated S phase percentage. Matrigel invasion and wound healing assays indicated that Rab18 positively regulated SNU-1 cell invasion and migration while its knockdown inhibited AGS cell invasion and migration. Rab18 maintained cell viability and downregulated apoptosis after cisplatin treatment, with upregulated mitochondrial membrane potential and downregulated mitochondrial reactive oxygen species (ROS) production. Rab18 overexpression upregulated p-Rb, survivin while downregulated cytochrome c, cleaved caspase-3 and cleaved PARP. Conclusion In conclusion, our results indicate that Rab18 promoted gastric cancer growth and chemoresistance, possibly through regulation of mitochondrial function and survivin.
Collapse
Affiliation(s)
- Binge Wu
- Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China,
| | - Rui Qi
- Department of Ophthalmology, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Xu Liu
- Department of Ophthalmology, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Lehua Qian
- Department of Ophthalmology, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Zhongjun Wu
- Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China,
| |
Collapse
|
11
|
Lupini L, Pepe F, Ferracin M, Braconi C, Callegari E, Pagotto S, Spizzo R, Zagatti B, Lanuti P, Fornari F, Ghasemi R, Mariani-Costantini R, Bolondi L, Gramantieri L, Calin GA, Sabbioni S, Visone R, Veronese A, Negrini M. Over-expression of the miR-483-3p overcomes the miR-145/TP53 pro-apoptotic loop in hepatocellular carcinoma. Oncotarget 2017; 7:31361-71. [PMID: 27120784 PMCID: PMC5058762 DOI: 10.18632/oncotarget.8913] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 04/10/2016] [Indexed: 01/01/2023] Open
Abstract
The miR-145-5p, which induces TP53-dependent apoptosis, is down-regulated in several tumors, including hepatocellular carcinomas (HCCs), but some HCCs show physiological expression of this miR. Here we demonstrate that in HCC cells carrying wild-type TP53 the steady activation of the miR-145 signaling selects clones resistant to apoptosis via up-regulation of the oncogenic miR-483-3p. Expression of the miR-145-5p and of the miR-483-3p correlated negatively in non-neoplastic liver (n=41; ρ=−0.342, P=0.028), but positively in HCCs (n=21; ρ=0.791, P<0.0001), which we hypothesized to be due to impaired glucose metabolism in HCCs versus normal liver. In fact, when liver cancer cells were grown in low glucose, miR-145-5p lowered miR-483-3p expression, allowing apoptosis, whereas when cells were grown in high glucose the levels of miR-483-3p increased, reducing the apoptotic rate. This indicates that depending on glucose availability the miR-145-5p has double effects on the miR-483-3p, either inhibitory or stimulatory. Moreover, resistance to apoptosis in clones overexpressing both miR-145-5p and miR-483-3p was abrogated by silencing the miR-483-3p. Our data highlight a novel mechanism of resistance to apoptosis in liver cancer cells harbouring wild type TP53 and suggest a potential role of miR-145-5p and miR-483-3p as druggable targets in a subset of HCCs.
Collapse
Affiliation(s)
- Laura Lupini
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Felice Pepe
- Unit of General Pathology, Aging Research Center (Ce.S.I.), G. d'Annunzio University Foundation, Chieti, Italy.,Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University, Chieti, Italy
| | - Manuela Ferracin
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Chiara Braconi
- Division of Cancer Therapeutics, Institute of Cancer Research, London, UK
| | - Elisa Callegari
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Sara Pagotto
- Unit of General Pathology, Aging Research Center (Ce.S.I.), G. d'Annunzio University Foundation, Chieti, Italy.,Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University, Chieti, Italy
| | | | - Barbara Zagatti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Paola Lanuti
- Department of Medicine and Aging Science, G. d'Annunzio University, Chieti, Italy
| | | | - Reza Ghasemi
- Unit of General Pathology, Aging Research Center (Ce.S.I.), G. d'Annunzio University Foundation, Chieti, Italy
| | - Renato Mariani-Costantini
- Unit of General Pathology, Aging Research Center (Ce.S.I.), G. d'Annunzio University Foundation, Chieti, Italy.,Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University, Chieti, Italy
| | - Luigi Bolondi
- S.Orsola-Malpighi University Hospital, Bologna, Italy
| | | | - George A Calin
- Department of Experimental Therapeutics, MD Anderson Medical Centre, Houston, TX, USA
| | - Silvia Sabbioni
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Rosa Visone
- Unit of General Pathology, Aging Research Center (Ce.S.I.), G. d'Annunzio University Foundation, Chieti, Italy.,Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University, Chieti, Italy
| | - Angelo Veronese
- Unit of General Pathology, Aging Research Center (Ce.S.I.), G. d'Annunzio University Foundation, Chieti, Italy.,Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University, Chieti, Italy
| | - Massimo Negrini
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
12
|
Changes of essential element content and prevention of cisplatin induced oxidative stress in rat liver by CV247 administration. Eur J Integr Med 2016. [DOI: 10.1016/j.eujim.2016.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
13
|
Szentmihályi K, May Z, Szénási G, Máthé C, Sebestény A, Albert M, Blázovics A. Cisplatin administration influences on toxic and non-essential element metabolism in rats. J Trace Elem Med Biol 2014; 28:317-21. [PMID: 24928713 DOI: 10.1016/j.jtemb.2014.02.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 01/13/2014] [Accepted: 02/15/2014] [Indexed: 12/21/2022]
Abstract
Nowadays several papers deal with the effectiveness and side effects of metal complexes, especially cisplatin, in cancer therapy. The excretion of essential metal elements from the body is a serious problem in the treatment, but there are no data concerning the distribution and metabolism of toxic and nonessential elements. Therefore our aim was to study the concentration of some of these elements after treatment with cisplatin. Male Wistar rats (n=20, 175-190 g) were randomly divided into 2 groups (n=10/group). The control group received 1% (w/v) methyl cellulose at 10 mL/kg body weight, p.o. by gastric gavage twice daily for 14 days, while cisplatin was injected i.p. in a single dose of 6.5 mg/kg body weight. Inductively coupled plasma optical emission spectrometry (ICP-OES) was used for measuring Al, B, Ba, Cr, Li, Ni, Pb, Pt, Sb, Si, Sn, Sr and V content in plasma, liver and kidney. Liver total scavenger capacity, diene conjugate content and malondialdehyde concentration were also determined. Cisplatin elevated the free radical reactions in the liver, although redox balance did not change significantly. According to the study it seems that the metabolism of Al, Ba, Cr, Ni, Pb, Sr were changed by the effect of cisplatin, and the most notable alterations were found for Al and Pb. Therefore, besides the toxic effect of and free radical induction by Pt, the side effects of increased levels of other toxic and non-essential elements have to be taken into consideration.
Collapse
Affiliation(s)
- Klára Szentmihályi
- Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences of the HAS, Magyar tudósok körútja 2, 1117 Budapest, Hungary.
| | - Zoltán May
- Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences of the HAS, Magyar tudósok körútja 2, 1117 Budapest, Hungary
| | - Gábor Szénási
- Institute of Pathophysiology, Semmelweis University, H-1089 Budapest, Hungary.
| | - Csaba Máthé
- Department of Pulmonology, Semmelweis University, H-1125 Budapest, Hungary.
| | - Andor Sebestény
- Laboratory Animal Science Unit, Faculty of Veterinary Science, Szent István University, H-1078 Budapest, Hungary.
| | | | - Anna Blázovics
- Department of Pharmacognosy, Semmelweis University, H-1085 Budapest, Hungary.
| |
Collapse
|
14
|
Ari F, Aztopal N, Icsel C, Yilmaz VT, Guney E, Buyukgungor O, Ulukaya E. Synthesis, structural characterization and cell death-inducing effect of novel palladium(II) and platinum(II) saccharinate complexes with 2-(hydroxymethyl)pyridine and 2-(2-hydroxyethyl)pyridine on cancer cells in vitro. Bioorg Med Chem 2013; 21:6427-34. [DOI: 10.1016/j.bmc.2013.08.050] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 08/16/2013] [Accepted: 08/23/2013] [Indexed: 12/15/2022]
|
15
|
Yeh CH, Yang YY, Huang YF, Chow KC, Chen MF. Induction of apoptosis in human Hep3B hepatoma cells by norcantharidin through a p53 independent pathway via TRAIL/DR5 signal transduction. Chin J Integr Med 2012; 18:676-82. [DOI: 10.1007/s11655-012-1206-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Indexed: 11/30/2022]
|
16
|
Nuclear Factor-kappa B as a Resistance Factor to Platinum-Based Antineoplasic Drugs. Met Based Drugs 2011; 2008:576104. [PMID: 18414584 PMCID: PMC2291150 DOI: 10.1155/2008/576104] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Accepted: 12/18/2007] [Indexed: 11/27/2022] Open
Abstract
Platinum drugs continue to be major chemotherapy drugs for cancer treatment. Nevertheless, acquired or intrinsic resistance to these compounds is common in human tumors. One important mechanism for this resistance is the avoidance of cells entering the apoptotic pathway. Nuclear factor-kappa B (NF-kappa B, NF-κB) is a pleiotropic transcription factor key in determining the death threshold of human cells. This factor is important in the final response of cells to platinum drugs, as exemplified by in vitro and in vivo models showing that inhibition of NF-κB sensitizes cancer cells to the effects of these drugs. New approaches focusing on the inhibition of NF-κB could help to minimize or even eliminate intrinsic or acquired resistance to platinum drugs.
Collapse
|
17
|
Liu W, Li X, Chu ESH, Go MYY, Xu L, Zhao G, Li L, Dai N, Si J, Tao Q, Sung JJY, Yu J. Paired box gene 5 is a novel tumor suppressor in hepatocellular carcinoma through interaction with p53 signaling pathway. Hepatology 2011; 53:843-53. [PMID: 21319196 DOI: 10.1002/hep.24124] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Accepted: 11/16/2010] [Indexed: 12/21/2022]
Abstract
UNLABELLED The paired box 5 (PAX5) is a member of PAX transcription factors family involved in the regulation of embryonic development. However, the role of PAX5 in carcinogenesis is largely unclear. We identified that PAX5 is involved in human cancer by methylation-sensitive representational difference analysis. We examined the biological functions and related molecular mechanisms of PAX5 in hepatocellular carcinoma (HCC). Promoter methylation of PAX5 was evaluated by methylation-specific polymerase chain reaction (PCR) and bisulfite genomic sequencing (BGS). The functions of ectopic PAX5 expression were determined by viability assay, colony formation, and cell cycle analyses, along with in vivo tumorigenicity assays. The PAX5 target signal pathway was identified by promoter luciferase assay, chromosome immunoprecipitation (ChIP), and pathway PCR array. PAX5 is expressed in normal human liver tissue, but silenced or down-regulated in 83% (10/12) of HCC cell lines. The mean expression level of PAX5 was significantly lower in primary HCCs as compared to their adjacent normal tissues (P < 0.0001). The promoter methylation contributes to the inactivation of PAX5. Restoring PAX5 expression in silenced HCC cell lines suppressed cell proliferation, induced apoptosis in vitro, and inhibited tumor growth in nude mice (P < 0.0001). The pathway luciferase reporter assay indicated that PAX5 activated p53 and p21 signaling. ChIP analysis demonstrated that PAX5 directly bound to the p53 promoter. The antitumorigenic function of PAX5 was at least up-regulated by p53 and its downstream targets including tumor necrosis factor, Fas ligand, leucine-rich repeats, and death domain-containing, poly(rC) binding protein 4, p21, and growth arrest and DNA-damage-inducible alpha. CONCLUSION PAX5 is frequently inactivated by promoter methylation in HCC. PAX5 appears to be a functional tumor suppressor involved in liver carcinogenesis through direct regulation of the p53 signaling pathway.
Collapse
Affiliation(s)
- Weili Liu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Zheng T, Wang J, Song X, Meng X, Pan S, Jiang H, Liu L. Nutlin-3 cooperates with doxorubicin to induce apoptosis of human hepatocellular carcinoma cells through p53 or p73 signaling pathways. J Cancer Res Clin Oncol 2010; 136:1597-604. [PMID: 20174822 DOI: 10.1007/s00432-010-0817-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2009] [Accepted: 02/01/2010] [Indexed: 11/30/2022]
Abstract
PURPOSE Despite recent advances in chemotherapeutic agents for Hepatocellular carcinoma (HCC) treatment, the results of chemotherapy remain unsatisfactory. Doxorubicin (DOX) still represents the cornerstone in HCC chemotherapy, but resistance and toxicity to normal cells are major obstacles to successful chemotherapy. Therefore, new active agents in HCC chemotherapy and agents that increase the chemosensitivity of HCC cells to DOX are still urgently required. Nutlin-3 is a small-molecule inhibitor that acts to inhibit murine double minute-2 (MDM2) binding to p53 or p73, and subsequently activates p53- or p73-dependent apoptosis signaling pathway. This study was designed to investigate whether Nutlin-3 alters cell toxicity to HCC cells following DNA damage and to assess the suitability of DOX/Nutlin-3 as a chemotherapeutic combination in HCC chemotherapy. METHODS Four human HCC cells were analyzed using cell proliferation assay, apoptosis assay, western blotting, co-immunoprecipitation and siRNA experiments. Anti-tumoral effects of Nutlin-3/DOX targeting the p53/MDM2 and p73/MDM2 pathways were evaluated in HCC cell lines. RESULTS Nutlin-3 enhances the growth inhibition by DOX and potentates the apoptotic effect in all HCC cell lines with different p53 types. Nutlin-3 acts through the disruption of p53-MDM2 binding in HepG2, and the disruption of p73-MDM2 in Huh-7 and Hep3B cell lines with subsequent activation of the apoptotic pathway, which leads to the increase in chemosensitivity to DOX in HCC cells. CONCLUSIONS Taken together, our findings suggest that Nutlin-3 will be active in the treatment of HCC and offers new prospects for overcoming DOX resistance.
Collapse
Affiliation(s)
- Tongsen Zheng
- Department of General Surgery, First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin 150001, Heilongjiang Province, China
| | | | | | | | | | | | | |
Collapse
|
19
|
Singh S, Okamura T, Ali-Osman F. Serine phosphorylation of glutathione S-transferase P1 (GSTP1) by PKCα enhances GSTP1-dependent cisplatin metabolism and resistance in human glioma cells. Biochem Pharmacol 2010; 80:1343-55. [PMID: 20654585 DOI: 10.1016/j.bcp.2010.07.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Revised: 07/09/2010] [Accepted: 07/12/2010] [Indexed: 01/20/2023]
Abstract
Recently, we reported that the human GSTP1 is phosphorylated and functionally activated by the PKC class of serine/threonine kinases. In this study, we investigated the contribution of this post-translational modification of GSTP1 to tumor cisplatin resistance. Using two malignant glioma cell lines, MGR1 and MGR3, the ability of PKCα-phosphorylated GSTP1 to catalyze the conjugation of cisplatin to glutathione was assessed and correlated with cisplatin sensitivity and cisplatin-induced DNA interstrand cross-links and apoptosis of the cells. The results showed PKCα activation and associated phosphorylation of GSTP1 to correlate significantly with increased glutathionylplatinum formation, decreased DNA interstrand cross-link formation and increased cisplatin resistance. Following PKC activation, the IC(50) of cisplatin increased from 13.63μM to 36.49μM in MGR1 and from 20.75μM to 38.45μM in MGR3. In both cell lines, siRNA-mediated GSTP1 or PKCα transcriptional suppression similarly decreased cisplatin IC(50) and was associated with decreased intracellular levels of glutathionylplatinum metabolite. Combined inhibition/transcriptional suppression of both PKCα and GSTP1 was synergistic in enhancing cisplatin sensitivity. Although, cisplatin-induced apoptosis was associated with the translocation of Bax to mitochondria, release of cytochrome c and caspase-3/7 activation, the levels of relocalized Bax and cytochrome c were significantly greater following GSTP1 knockdown. These results support a mechanism of cisplatin resistance mediated by the PKCα-dependent serine phosphorylation of GSTP1 and its associated increased cisplatin metabolism, and suggest the potential of simultaneous targeting of GSTP1 and PKCα to improve the efficacy of cisplatin therapy.
Collapse
Affiliation(s)
- Simendra Singh
- Department of Surgery, The Preston Robert Tisch Brain Tumor Center, Durham, NC 27710, USA.
| | | | | |
Collapse
|
20
|
Kim JS, Ahn KJ, Kim JA, Kim HM, Lee JD, Lee JM, Kim SJ, Park JH. Role of reactive oxygen species-mediated mitochondrial dysregulation in 3-bromopyruvate induced cell death in hepatoma cells : ROS-mediated cell death by 3-BrPA. J Bioenerg Biomembr 2008; 40:607-18. [PMID: 19067133 DOI: 10.1007/s10863-008-9188-0] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2008] [Accepted: 11/04/2008] [Indexed: 01/04/2023]
Abstract
Hexokinase type II (HK II) is the key enzyme for maintaining increased glycolysis in cancer cells where it is overexpressed. 3-bromopyruvate (3-BrPA), an inhibitor of HK II, induces cell death in cancer cells. To elucidate the molecular mechanism of 3-BrPA-induced cell death, we used the hepatoma cell lines SNU449 (low expression of HKII) and Hep3B (high expression of HKII). 3-BrPA induced ATP depletion-dependent necrosis and apoptosis in both cell lines. 3-BrPA increased intracellular reactive oxygen species (ROS) leading to mitochondrial dysregulation. NAC (N-acetyl-L: -cysteine), an antioxidant, blocked 3-BrPA-induced ROS production, loss of mitochondrial membrane potential and cell death. 3-BrPA-mediated oxidative stress not only activated poly-ADP-ribose (PAR) but also translocated AIF from the mitochondria to the nucleus. Taken together, 3-BrPA induced ATP depletion-dependent necrosis and apoptosis and mitochondrial dysregulation due to ROS production are involved in 3-BrPA-induced cell death in hepatoma cells.
Collapse
Affiliation(s)
- Ji Su Kim
- Department of Microbiology and Brain Korea 21 Project for Medical Sciences, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, 134, Shinchon-dong, Seodaemoon-gu, 120-752, Seoul, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Ma Y, Yu WD, Hershberger PA, Flynn G, Kong RX, Trump DL, Johnson CS. 1alpha,25-Dihydroxyvitamin D3 potentiates cisplatin antitumor activity by p73 induction in a squamous cell carcinoma model. Mol Cancer Ther 2008; 7:3047-55. [PMID: 18790784 DOI: 10.1158/1535-7163.mct-08-0243] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
1alpha,25-Dihydroxyvitamin D3 (1,25D3) exhibits antitumor activity in a variety of cancers including squamous cell carcinoma (SCC). Intrinsic resistance of SCC cells to cisplatin was observed and led to the investigation into whether 1,25D3 sensitizes SCC cells to cisplatin. Pretreatment with 1,25D3 followed by cisplatin enhanced growth inhibition in SCC cells compared with 1,25D3 alone as assessed by cytotoxicity and in vitro clonogenic assays. In addition, 1,25D3 sensitized SCC cells to cisplatin-mediated apoptosis. Treatment of tumor-bearing C3H mice with 1,25D3 before cisplatin reduced clonogenic survival using in vivo excision clonogenic assay. These results were not observed in a 1,25D3-resistant SCC variant, indicating the critical role of 1,25D3 in sensitizing SCC cells to cisplatin. Further, a marked decrease in fractional tumor volume was observed when SCC tumor-bearing mice were treated with 1,25D3 before cisplatin compared with either agent administered alone. Cisplatin has been shown to modulate p73 protein level in certain cancer cells. Our data showed that p73 level was not affected by cisplatin but increased by 1,25D3 in SCC cells. Knocking down p73 by small interfering RNA protected SCC cells against 1,25D3 and cisplatin-mediated clonogenic cell kill and apoptosis. Increasing p73 protein level by knocking down UFD2a, which mediates p73 degradation, promoted 1,25D3 and cisplatin-mediated clonogenic cell kill. These results suggest that 1,25D3 potentiates cisplatin antitumor activity in vitro and in vivo in a SCC model system possibly through p73 induction and apoptosis. The combination treatment may provide a more effective therapeutic regimen in cancer treatment.
Collapse
Affiliation(s)
- Yingyu Ma
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Brenes O, Arce F, Gätjens-Boniche O, Díaz C. Characterization of cell death events induced by anti-neoplastic drugs cisplatin, paclitaxel and 5-fluorouracil on human hepatoma cell lines: Possible mechanisms of cell resistance. Biomed Pharmacother 2007; 61:347-55. [PMID: 17399942 DOI: 10.1016/j.biopha.2007.02.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2007] [Accepted: 02/12/2007] [Indexed: 11/30/2022] Open
Abstract
Two different hepatoma cell lines were incubated for 48h with chemotherapeutic drugs cisplatin, paclitaxel and 5-FU to determine their ability to induce cytotoxicity and DNA fragmentation as well as to modify the expression of some cell death-related genes that could be involved in the resistance to therapy. We observed that cisplatin and paclitaxel induced cytotoxicity, but significant differences between both cell lines, were found only in the case of paclitaxel. At 48h, apoptosis was clearly present in Hep3B cells treated with cisplatin and HepG2 cells treated with paclitaxel. 5-FU induced cytotoxicity in both cell lines but only at higher concentrations than the other two drugs, triggering apoptosis and necrosis in HepG2 cells and only necrosis in Hep3B. When a time course was performed for the first 8h of treatment to elucidate the initial mechanism of cell death responsible for DNA fragmentation, we observed that 5-FU in Hep3B, and cisplatin in both cell lines, induces primary necrosis, whereas at the concentration tested here, paclitaxel clearly triggers apoptosis in both cell lines. HepG2 cells were weakly sensitive to 5-FU in the first 8h of treatment, so the primary mechanism of cell death was not clear, but results seem to indicate that it could be apoptosis. At 48h, Bax was not up-regulated with any of the treatments, whereas cisplatin was able to induce Bcl-xL down-regulation in both cell lines. Treatment with 5-FU also down-regulated Bcl-xL in HepG2 cells. We also measured variations in the expression of survivin, an inhibitor of apoptosis that has also been involved in mitototic catastrophe. Hep3B cells seem to show an increase in protein levels with all treatments. Exposure to paclitaxel resulted in the highest effect. In the case of HepG2 cells, there was a decrease in survivin expression when cells were treated with 5FU and paclitaxel, both treatments showing complete loss of the protein. Using an antibody that recognizes unprocessed caspase-3, we observed that the enzyme was assumingly activated in HepG2 cells treated with 5FU and paclitaxel, but only weakly after treatment with cisplatin. Hep3B cells did not show activation since the levels of the pro-enzyme remained the same as that in the control. In conclusion, the three drugs tested in this study could induce cell death, with paclitaxel being more effective inducing apoptosis. 5FU was only effective at high doses and its mechanism seems to be primarily related to necrosis in Hep3B and probably apoptosis in HepG2. Cisplatin mechanism of cell death is probably mediated by the decrease in anti-apoptotic protein Bcl-xL whereas paclitaxel and 5FU are decreasing the apoptosis inhibitor survivin. According to pro-enzyme levels, caspase-3 was only activated in HepG2 cells, whereas in the case of Hep3B cells the mechanisms of toxicity appear to be caspase-3-independent at the time and concentrations tested in this study. The resistance of Hep3B cells to death induced by chemotherapy could be related to an increase in the expression of IAP survivin, which can decrease cell response to the treatment or even switch the type of death from apoptosis to another kind, making therapy less efficient.
Collapse
Affiliation(s)
- O Brenes
- Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | | | | | | |
Collapse
|
23
|
Kikuchi H, Ozaki T, Furuya K, Hanamoto T, Nakanishi M, Yamamoto H, Yoshida K, Todo S, Nakagawara A. NF-kappaB regulates the stability and activity of p73 by inducing its proteolytic degradation through a ubiquitin-dependent proteasome pathway. Oncogene 2006; 25:7608-17. [PMID: 16953234 DOI: 10.1038/sj.onc.1209748] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nuclear factor kappa B (NF-kappaB), which exists as heterodimeric complexes composed of p50 and p65, has been shown to play an important role in cell survival processes. In the present study, we found for the first time that NF-kappaB has an ability to induce the ubiquitin-dependent proteasomal degradation of proapoptotic p73alpha. The activation of NF-kappaB in tumor necrosis factor alpha (TNF-alpha)-stimulated H1299 cells resulted in a significant reduction in the amounts of the endogenous p73alpha. Consistent with these results, TNF-alpha-mediated downregulation of p73alpha was observed in wild-type (WT) mouse embryonic fibroblasts (MEFs) but not in p65-deficient MEFs. Ectopic expression of NF-kappaB decreased a half-life of p73alpha by increasing its ubiquitination levels, and thereby inhibiting the transcriptional activity as well as proapoptotic function of p73alpha, whereas NF-kappaB had undetectable effects on p53. Immunoprecipitation experiments demonstrated that, under our experimental conditions, NF-kappaB does not bind to p73alpha in mammalian cultured cells. In contrast to WT p65, the COOH-terminal deletion mutant of p65 (p65DeltaC) failed to reduce the expression levels of p73alpha, suggesting that NF-kappaB-mediated proteolytic degradation of p73alpha requires the transcriptional activity of NF-kappaB. Taken together, our present results imply that NF-kappaB-mediated degradation of proapoptotic p73 is a novel inhibitory mechanism of p73 that regulates cell survival and death.
Collapse
Affiliation(s)
- H Kikuchi
- Division of Biochemistry, Chiba Cancer Center Research Institute, Chiba, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Lasfer M, Davenne L, Vadrot N, Alexia C, Sadji-Ouatas Z, Bringuier AF, Feldmann G, Pessayre D, Reyl-Desmars F. Protein kinase PKC delta and c-Abl are required for mitochondrial apoptosis induction by genotoxic stress in the absence of p53, p73 and Fas receptor. FEBS Lett 2006; 580:2547-52. [PMID: 16631755 DOI: 10.1016/j.febslet.2006.03.089] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2005] [Revised: 03/20/2006] [Accepted: 03/29/2006] [Indexed: 01/30/2023]
Abstract
Doxorubicin, cis-diamminedichloroplatinum (II) and 5-fluorouracil used in chemotherapy induce apoptosis in Hep3B cells in the absence of p53, p73, and functional Fas. Since mediators remain unknown, the requirement of PKC delta (PKCdelta) and c-Abl was investigated. Suppression of c-Abl or PKCdelta expression using SiRNAs impaired PARP cleavage, Gleevec and/or rottlerin inhibited the induction of the subG1 phase and the increase of reactive oxygen species level. Co-precipitations and phosphorylations to mitochondria of c-Abl, PKCdelta and Bcl-X(L/s) were induced. A depolarization of the mitochondrial membrane and activations of caspase-2 and -9 were observed. We propose that, in the absence of p53, p73 and Fas, genotoxic drugs could require both PKCdelta and c-Abl to induce apoptosis through the mitochondrial pathway.
Collapse
Affiliation(s)
- Malika Lasfer
- INSERM, U773, Equipe 5, Centre de Recherche Bichat Beaujon CRB3, BP416, F-75018 Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Xie J, Shaikh ZA. Cadmium-Induced Apoptosis in Rat Kidney Epithelial Cells Involves Decrease in Nuclear Factor-Kappa B Activity. Toxicol Sci 2006; 91:299-308. [PMID: 16478757 DOI: 10.1093/toxsci/kfj131] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Renal epithelial cells undergo apoptosis upon exposure to cadmium (Cd). Transcription factors, such as nuclear factor-kappa B (NF-kappaB), mediate the expression of a number of genes involved in apoptosis. The present study was designed to examine the involvement of this transcription factor in Cd-induced apoptosis. Rat kidney proximal tubular epithelial cells, NRK-52E, were incubated with up to 20 microM CdCl2 in serum-free medium for 5 h followed by incubation in serum-containing medium (without Cd) for an additional 12 h. The cells accumulated 582 +/- 19 ng Cd/mg protein after 5-h exposure to 20 microM Cd. As a result of Cd exposure, the DNA-binding activity of the p65 subunit of NF-kappaB was decreased in a concentration- and time-dependent manner. The activity of tumor necrosis factor-alpha-induced inhibitor of kappa B (IkappaB) kinase alpha was also inhibited by Cd. In addition, the phosphorylation of IkappaB-alpha and NF-kappaB p65, as well as the levels of NF-kappaB target gene products, cIAP-1 and cIAP-2, were reduced. Pretreatment of the cells with the antioxidant U83836E or butylated hydroxytoluene preserved the DNA-binding activity and blocked the Cd-induced decease in IkappaB-alpha phosphorylation. Cd exposure caused the activation of caspase-3, -7, and -9 and DNA fragmentation. By flow cytometry, 14.6 and 30.5% apoptosis was detected at 6 and 12 h after stopping the Cd exposure. Overexpression of NF-kappaB p65 by transient transfection protected the cells from the Cd-induced apoptosis. Conversely, attenuation of NF-kappaB activity by pretreatment with SN50, an NF-kappaB nuclear translocation inhibitor, potentiated apoptosis. These results suggest that Cd-induced apoptosis involves suppression of NF-kappaB activity which may be mediated by oxidative stress.
Collapse
Affiliation(s)
- Jianxun Xie
- Department of Biomedical and Pharmaceutical Sciences and Center for Molecular Toxicology, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island 02881, USA
| | | |
Collapse
|
26
|
Campbell KJ, Witty JM, Rocha S, Perkins ND. Cisplatin Mimics ARF Tumor Suppressor Regulation of RelA (p65) Nuclear Factor-κB Transactivation. Cancer Res 2006; 66:929-35. [PMID: 16424027 DOI: 10.1158/0008-5472.can-05-2234] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The RelA (p65) nuclear factor-kappaB (NF-kappaB) subunit can contribute towards tumor cell survival through inducing the expression of a variety of antiapoptotic genes. However, the NF-kappaB response can show great diversity and is not always antiapoptotic. Here, we find that cisplatin, a DNA cross-linking agent and commonly used anticancer compound, does not affect RelA nuclear translocation but modulates its transcriptional activity. Similar to other genotoxic agents, such as daunorubicin and UV light, cisplatin treatment in the U-2 OS osteosarcoma cell line represses RelA activity and inhibits expression of the NF-kappaB antiapoptotic target gene Bcl-x(L). The mechanism through which cisplatin achieves these effects is different to daunorubicin and UV light but shows great similarity to the RelA regulatory pathway induced by the ARF tumor suppressor: cisplatin regulation of RelA requires ATR/Chk1 activity, represses Bcl-x(L) but not XIAP expression, and results in phosphorylation of RelA at Thr(505). In contrast to these results, another chemotherapeutic drug etoposide activates NF-kappaB and induces expression of these target genes. Thus, within a single tumor cell line, there is great heterogeneity in the NF-kappaB response to different, commonly used chemotherapeutic drugs. These observations suggest that it might be possible to minimize the ability of RelA to inhibit cancer therapy by diagnostically predicting the type of chemotherapeutic drug most compatible with NF-kappaB functionality in a tumor cell type. Moreover, our data indicate that at least with respect to RelA, cisplatin functions as an ARF mimic. Other drugs capable of mimicking this aspect of ARF function might therefore have therapeutic potential.
Collapse
Affiliation(s)
- Kirsteen J Campbell
- School of Life Sciences, Division of Gene Regulation and Expression, University of Dundee, Dow Street, Dundee DD1 5EH, Scotland, United Kingdom
| | | | | | | |
Collapse
|
27
|
Wang Y, Perchellet EM, Ward MM, Lou K, Hua DH, Perchellet JPH. Rapid collapse of mitochondrial transmembrane potential in HL-60 cells and isolated mitochondria treated with anti-tumor 1,4-anthracenediones. Anticancer Drugs 2005; 16:953-67. [PMID: 16162972 DOI: 10.1097/01.cad.0000180123.24031.5a] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Since synthetic analogs of 1,4-anthraquinone (AQ code number), such as AQ8, AQ9 and AQ10, can trigger cytochrome c release without caspase activation and retain their ability to induce apoptosis in multidrug-resistant (MDR) tumor cells, fluorescent probes of transmembrane potential have been used to determine whether these anti-tumor compounds might directly target mitochondria in cell and cell-free systems to cause the collapse of mitochondrial membrane potential (/Deltapsim) that is linked to permeability transition pore (PTP) opening. Using JC-1 dye, the abilities of various AQ analogs to induce the /Deltapsim in wild-type and MDR HL-60 cells are rapid (within 2.5-10 min), irreversible after drug removal, concentration dependent in the 0.256-10 micromol/l range and generally related to their anti-tumor activities in vitro. The /Deltapsim caused by AQ9 and AQ10, which are more potent than mitoxantrone, staurosporine and the reference depolarizing agent carbonyl cyanide m-chlorophenylhydrazone (CCCP) in HL-60 cells, are not prevented by caspase-2 or -8 inhibitors, suggesting that activations of these apical caspases upstream of mitochondria are not involved in this process. Antitumor AQ analogs (0.256-10 micromol/l) also mimic the abilities of the known depolarizing agents CCCP, alamethicin, gramicidin A and 100 micromol/l CaCl2 to directly induce within 15 min the /Deltapsim in isolated mitochondria prepared from mouse liver and loaded with rhodamine 123 dye. The fact that 20 micromol/l Ca2+, which is insufficient to trigger depolarization on its own, is required to reveal the depolarizing effect of AQ9 in isolated mitochondria suggests that anti-tumor AQ analogs might interact with the PTP to alter its conformation and increase its Ca2+ sensitivity. Indeed, such Ca2+-dependent /Deltapsim of isolated mitochondria treated with 1.6 micromol/l AQ9 or 100 micromol/l Ca2+ are blocked by ruthenium red. Daunorubicin (DAU) is unable to mimic the rapid /Deltapsim caused by anti-tumor AQ analogs within 2.5-40 min of treatment in HL-60 cells or isolated mitochondria. Moreover, the /Deltapsim caused by 1.6 micromol/l AQ9 or 100 micromol/l Ca2+ in isolated mitochondria are similarly blocked by cyclosporin A (CsA), bongkrekic acid and decylubiquinone, which prevent PTP opening, suggesting that, in contrast to DAU, anti-tumor AQ analogs that directly target mitochondria to trigger the Ca2+-dependent and CsA-sensitive /Deltapsim, might induce PTP opening and the mitochondrial pathway of apoptosis even in the absence of nuclear signals.
Collapse
Affiliation(s)
- Yang Wang
- Anti-Cancer Drug Laboratory, Division of Biology, Ackert Hall, Kansas State University, Manhattan, Kansas 66506-4901, USA
| | | | | | | | | | | |
Collapse
|
28
|
Venkatraman M, Anto RJ, Nair A, Varghese M, Karunagaran D. Biological and chemical inhibitors of NF-kappaB sensitize SiHa cells to cisplatin-induced apoptosis. Mol Carcinog 2005; 44:51-9. [PMID: 16044419 DOI: 10.1002/mc.20116] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Cisplatin, a chemotherapeutic agent, is known to induce apoptosis of cancer cells. We examined the role of NF-kappaB during cisplatin-induced apoptosis in two human cervical cancer cell lines, HeLa and SiHa, known to differ in their response to cisplatin treatment. We found that SiHa cells were relatively more resistant than HeLa cells to the cytotoxic effects induced by cisplatin as measured by MTT assays. HeLa cells were more sensitive to the apoptotic effects induced by cisplatin as shown by increases in annexin staining, DNA fragmentation, and loss of mitochondrial membrane potential. Similarly the activities of caspases 3, 8, and 9 and cleavage of PARP induced by cisplatin were more in HeLa than SiHa cells. Cisplatin induced NF-kappaB DNA binding activity in HeLa and SiHa cells but not in primary cervical cells and the active DNA binding complex in SiHa cells consists of p50 and RelA heterodimers. However, when NF-kappaB DNA binding activity was blocked by chemical (curcumin, PDTC, or salicylic acid) or biological inhibitors (NIK-KM or IKK-beta DN), the cell viability was less in SiHa cells with cisplatin treatment, but these effects were not observed in HeLa cells. Similarly upon treatment with cisplatin SiHa cells had more activation of caspases compared to that seen in HeLa cells under conditions of NF-kappaB inhibition by biological or chemical inhibitors. These results suggest that NF-kappaB may contribute to the resistance of human cervical cancer cells to cisplatin and highlight the potential use of combination therapy involving cisplatin and NF-kappaB inhibitors.
Collapse
Affiliation(s)
- Manickam Venkatraman
- Division of Cancer Biology, Rajiv Gandhi Center for Biotechnology, Thiruvananthapuram, Kerala, India
| | | | | | | | | |
Collapse
|
29
|
Seth R, Yang C, Kaushal V, Shah SV, Kaushal GP. p53-dependent caspase-2 activation in mitochondrial release of apoptosis-inducing factor and its role in renal tubular epithelial cell injury. J Biol Chem 2005; 280:31230-9. [PMID: 15983031 DOI: 10.1074/jbc.m503305200] [Citation(s) in RCA: 149] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We demonstrate the role of p53-mediated caspase-2 activation in the mitochondrial release of apoptosis-inducing factor (AIF) in cisplatin-treated renal tubular epithelial cells. Gene silencing of AIF with its small interfering RNA (siRNA) suppressed cisplatin-induced AIF expression and provided a marked protection against cell death. Subcellular fractionation and immunofluorescence studies revealed cisplatin-induced translocation of AIF from the mitochondria to the nuclei. Pancaspase inhibitor benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone or p53 inhibitor pifithrin-alpha markedly prevented mitochondrial release of AIF, suggesting that caspases and p53 are involved in this release. Caspase-2 and -3 that were predominantly activated in response to cisplatin provided a unique model to study the role of these caspases in AIF release. Cisplatin-treated caspase-3 (+/+) and caspase-3 (-/-) cells exhibited similar AIF translocation to the nuclei, suggesting that caspase-3 does not affect AIF translocation, and thus, caspase-2 may be involved in the translocation. Caspase-2 inhibitor benzyloxycarbonyl-Val-Asp-Val-Ala-Asp-fluoromethylketone or down-regulation of caspase-2 by its siRNA significantly prevented translocation of AIF. Caspase-2 activation was a critical response from p53, which was markedly induced and phosphorylated in cisplatin-treated cells. Overexpression of p53 not only resulted in caspase-2 activation but also mitochondrial release of AIF. The p53 inhibitor pifithrin-alpha or p53 siRNA prevented both cisplatin-induced caspase-2 activation and mitochondrial release of AIF. Caspase-2 activation was dependent on the p53-responsive gene, PIDD, a death domain-containing protein that was induced by cisplatin in a p53-dependent manner. These results suggest that caspase-2 activation mediated by p53 is an important pathway involved in the mitochondrial release of AIF in response to cisplatin injury.
Collapse
Affiliation(s)
- Rohit Seth
- Department of Medicine, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, Arkansas 72205, USA
| | | | | | | | | |
Collapse
|
30
|
Arce F, Gätjens-Boniche O, Vargas E, Valverde B, Díaz C. Apoptotic events induced by naturally occurring retinoids ATRA and 13-cis retinoic acid on human hepatoma cell lines Hep3B and HepG2. Cancer Lett 2005; 229:271-81. [PMID: 16135400 DOI: 10.1016/j.canlet.2005.06.047] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2005] [Revised: 06/10/2005] [Accepted: 06/26/2005] [Indexed: 12/11/2022]
Abstract
Two hepatoma cell lines were incubated for 72 h with ATRA and its analog 13cisRA and according to MTT assay, Hep3B cells were highly susceptible whereas HepG2 cells were more resistant to the treatment. At the high concentration of 166 microM, retinoids were able to induce apoptosis in both cell lines and the highest effect was observed in HepG2 cells treated with ATRA. TUNEL-based photometric ELISA showed that at the same retinoid concentration tested by flow cytometry, both cell lines showed apoptosis whereas plasma membranes were not significantly disrupted. Inhibitors of apoptosis Bcl-xL and survivin were downregulated in Hep3B cells by treatment with both retinoids. Bax, a pro-apoptotic protein, was not significantly upregulated in Hep3B cells, but was slightly increased in HepG2 cells treated with 13cisRA. Both procaspase-3 and procaspase-8 were cleaved in Hep3B cells, suggesting apoptosis could be triggered through the extrinsic pathway. In the case of HepG2 cells, lack of caspase activation suggests a mechanism dependent on other kind of proteases.
Collapse
Affiliation(s)
- Frederick Arce
- Instituto Clodomiro Picado, Universidad de Costa Rica, San José, Costa Rica
| | | | | | | | | |
Collapse
|
31
|
Velthuis JHL, Gavric Z, de Bont HJGM, Nagelkerke JF. Impaired activation of caspases and prevention of mitochondrial dysfunction in the metastatic colon carcinoma CC531s-m2 cell line. Biochem Pharmacol 2004; 69:463-71. [PMID: 15652237 DOI: 10.1016/j.bcp.2004.10.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2004] [Accepted: 10/27/2004] [Indexed: 11/28/2022]
Abstract
In a previous paper we described the properties of a rapidly metastasizing cell line CC531s-m2 derived from the poorly metastasizing CC531s cell. The m2-cell line was relatively resistant to killing by NK cells. Both CD95L and TRAIL mediated apoptosis was decreased in the m2-cell line. Now, by flow cytometrical analysis of intra- and extra-cellular expressed receptors, we show that the localization of the receptors for CD95L and TRAIL was not altered in the CC531s-m2 cells as compared to the parental cell line. Subsequently caspase-activation and mitochondrial function were studied by enzymatic cleavage of fluorescent caspase-substrates and retention of the mitochondrial dye rhodamine-123, respectively. The activation of caspases as well as the loss of the mitochondrial membrane potential (MMP) was less in the CC531s-m2 cell line upon CD95L- and TRAIL-signalling. Furthermore, the sensitivity of the CC531-m2 towards cisplatin-induced apoptosis was strongly decreased. This was consistent with less mitochondrial damage, delayed caspase cleavage and decreased caspase activity. Altogether, we conclude that an Natural Killer-cell insensitive cell is less sensitive to CD95L- and TRAIL-induced apoptosis as well as anti-cancer drug induced apoptosis by prevention of mitochondrial damage and activation of caspases.
Collapse
Affiliation(s)
- Jurjen H L Velthuis
- Division of Toxicology, Leiden/Amsterdam Center for Drug Research, Leiden University, Gorlaeus Laboratrory, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | | | | | | |
Collapse
|
32
|
Perchellet EM, Wang Y, Weber RL, Lou K, Hua DH, Perchellet JPH. Antitumor triptycene bisquinones induce a caspase-independent release of mitochondrial cytochrome c and a caspase-2-mediated activation of initiator caspase-8 and -9 in HL-60 cells by a mechanism which does not involve Fas signaling. Anticancer Drugs 2004; 15:929-46. [PMID: 15514562 DOI: 10.1097/00001813-200411000-00002] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Synthetic triptycene analogs (TT code number) mimic the antitumor effects of daunorubicin (DAU) in vitro, but have the advantage of blocking nucleoside transport, inhibiting both DNA topoisomerase I and II activities, and retaining their efficacy in multidrug-resistant (MDR) tumor cells. Since TT bisquinones induce poly(ADP-ribose) polymerase-1 (PARP-1) cleavage at 6 h and internucleosomal DNA fragmentation at 24 h, which are, respectively, early and late markers of apoptosis, these antitumor drugs were tested for their ability to trigger the release of mitochondrial cytochrome c (Cyt c) and the caspase activation cascade in the HL-60 cell system. Based on their ability to reduce the viability of wild-type, drug-sensitive HL-60-S cells in the nanomolar range, six lead antitumor TT bisquinones have been identified so far: TT2, TT13, TT16, TT19, TT24 and TT26. In accord with the fact that effector caspase-3 is responsible for PARP-1 cleavage, 4 microM concentrations of DAU and these TT bisquinones all maximally induce caspase-3 activity at 6 h in HL-60-S cells, an effect which persists when the drugs are removed after a 1-h pulse treatment. Since caspase-3 may be activated by initiator caspase-9 and -8, it is significant to show that such caspase activation cascade is induced by 4 microM DAU and TT bisquinones at 6 h in HL-60-S cells. Although the relationship is not perfect, the ability of TT analogs to induce caspase-3, -8 and -9 activities may be linked to their quinone functionality and cytotoxicity. Interestingly, 4 microM concentrations of TT bisquinones retain their ability to induce caspase-3, -8 and -9 activities at 6 h in the MDR HL-60-RV cell line where 4 microM DAU becomes totally ineffective. The release of mitochondrial Cyt c is also detected within 6 h in HL-60-S cells treated with 4 microM DAU or TT bisquinones, a finding consistent with the fact that Cyt c is the apoptotic trigger that activates caspase-9. Caspase-2 and -8 may both act upstream of mitochondria to promote Cyt c release, but caspase-2 is already maximally activated 6 h after 4 microM DAU or TT13 treatments, whereas DAU- or TT-induced caspase-8 and -9 activities peak at 9 h. Pre-treatments with 15 microM of the caspase-2 inhibitor benzyloxycarbonyl (z)-Val-Asp-Val-Ala-Asp (VDVAD)-fluoromethyl ketone (fmk) totally block DAU- and TT13-induced caspase-2, -8 and -9 activities, whereas pre-treatments with 15 microM of the caspase-8 inhibitor z-Ile-Glu-Thr-Asp (IETD)-fmk prevent DAU and TT13 from inducing caspase-8 activities without affecting their caspase-2- and -9-inducing activities, suggesting that the induction of apical caspase-2 activity by these drugs may be a critical upstream event required for the activation of other downstream caspases, including caspase-9 and the mitochondrial amplification loop through caspase-8. However, the mechanisms by which DAU and TT13 induce the release of mitochondrial Cyt c appear to be caspase-independent since they are both insensitive to similar pre-treatments with 100 microM of these specific caspase-2 and -8 inhibitors. Moreover, pre-treatments with 10 microg/ml of the antagonistic anti-Fas DX2 and ZB4 monoclonal antibodies (mAbs), and the neutralizing anti-Fas ligand (FasL) NOK-1 mAb are all unable to prevent DAU and TT13 from inducing Cyt c release and caspase-2, -8 and -9 activities, suggesting that the Fas-FasL signaling pathway is not involved in the mechanism by which these quinone antitumor drugs trigger apoptosis in HL-60 cells.
Collapse
Affiliation(s)
- Elisabeth M Perchellet
- Anti-Cancer Drug Laboratory, Division of Biology, Ackert Hall; Department of Chemistry, Kansas State University, Manhattan, KS 66506-4901, USA
| | | | | | | | | | | |
Collapse
|