1
|
Wu S, Luo T, Lei X, Yang X. Emerging role of competing endogenous RNA in lung cancer drug resistance. J Chemother 2024; 36:546-565. [PMID: 38124356 DOI: 10.1080/1120009x.2023.2294582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 12/03/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023]
Abstract
Lung cancer remains one of the most common malignant cancers worldwide, and its survival rate is extremely low. Chemotherapy, the mainstay of lung cancer treatment, is not as effective as it could be due to the development of cellular resistance. The molecular mechanisms of drug resistance in lung cancer remain to be elucidated. Accumulating evidence suggests that ceRNAs are involved in various carcinogenesis and development. CeRNA is a transcript that regulates each other through competition with miRNA. However, the relationship between ceRNAs and chemoresistance in lung cancer remains unclear. In this narrative review, we provided a summary of treatment approaches that focus on ceRNA networks to overcome drug resistance.
Collapse
Affiliation(s)
- Shijie Wu
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang, People's Republic of China
| | - Ting Luo
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang, People's Republic of China
| | - Xiaoyong Lei
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang, People's Republic of China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, People's Republic of China
| | - Xiaoyan Yang
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang, People's Republic of China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, People's Republic of China
| |
Collapse
|
2
|
Huang YK, Chang KC, Li CY, Lieu AS, Lin CL. AKR1B1 Represses Glioma Cell Proliferation through p38 MAPK-Mediated Bcl-2/BAX/Caspase-3 Apoptotic Signaling Pathways. Curr Issues Mol Biol 2023; 45:3391-3405. [PMID: 37185746 PMCID: PMC10136867 DOI: 10.3390/cimb45040222] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
This study aimed to investigate the regulatory role of Aldo-keto reductase family 1 member B1 (AKR1B1) in glioma cell proliferation through p38 MAPK activation to control Bcl-2/BAX/caspase-3 apoptosis signaling. AKR1B1 expression was quantified in normal human astrocytes, glioblastoma multiforme (GBM) cell lines, and normal tissues by using quantitative real-time polymerase chain reaction. The effects of AKR1B1 overexpression or knockdown and those of AKR1B1-induced p38 MAPK phosphorylation and a p38 MAPK inhibitor (SB203580) on glioma cell proliferation were determined using an MTT assay and Western blot, respectively. Furthermore, the AKR1B1 effect on BAX and Bcl-2 expression was examined in real-time by Western blot. A luminescence detection reagent was also utilized to identify the effect of AKR1B1 on caspase-3/7 activity. The early and late stages of AKR1B1-induced apoptosis were assessed by performing Annexin V-FITC/PI double-staining assays. AKR1B1 expression was significantly downregulated in glioma tissues and GBM cell lines (T98G and 8401). Glioma cell proliferation was inhibited by AKR1B1 overexpression but was slightly increased by AKR1B1 knockdown. Additionally, AKR1B1-induced p38 MAPK phosphorylation and SB203580 reversed AKR1B1's inhibitory effect on glioma cell proliferation. AKR1B1 overexpression also inhibited Bcl-2 expression but increased BAX expression, whereas treatment with SB203580 reversed this phenomenon. Furthermore, AKR1B1 induced caspase-3/7 activity. The induction of early and late apoptosis by AKR1B1 was confirmed using an Annexin V-FITC/PI double-staining assay. In conclusion, AKR1B1 regulated glioma cell proliferation through the involvement of p38 MAPK-induced BAX/Bcl-2/caspase-3 apoptosis signaling. Therefore, AKR1B1 may serve as a new therapeutic target for glioma therapy development.
Collapse
Affiliation(s)
- Yu-Kai Huang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Division of Neurosurgery, Department of Surgery, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung 80145, Taiwan
| | - Kun-Che Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Neurobiology, Center of Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ann-Shung Lieu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Chih-Lung Lin
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| |
Collapse
|
3
|
Thummuri D, Khan S, Underwood PW, Zhang P, Wiegand J, Zhang X, Budamagunta V, Sobh A, Tagmount A, Loguinov A, Riner AN, Akki AS, Williamson E, Hromas R, Vulpe CD, Zheng G, Trevino JG, Zhou D. Overcoming Gemcitabine Resistance in Pancreatic Cancer Using the BCL-X L-Specific Degrader DT2216. Mol Cancer Ther 2022; 21:184-192. [PMID: 34667112 PMCID: PMC8742767 DOI: 10.1158/1535-7163.mct-21-0474] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/16/2021] [Accepted: 10/14/2021] [Indexed: 01/09/2023]
Abstract
Pancreatic cancer is the third most common cause of cancer-related deaths in the United States. Although gemcitabine is the standard of care for most patients with pancreatic cancer, its efficacy is limited by the development of resistance. This resistance may be attributable to the evasion of apoptosis caused by the overexpression of BCL-2 family antiapoptotic proteins. In this study, we investigated the role of BCL-XL in gemcitabine resistance to identify a combination therapy to more effectively treat pancreatic cancer. We used CRISPR-Cas9 screening to identify the key genes involved in gemcitabine resistance in pancreatic cancer. Pancreatic cancer cell dependencies on different BCL-2 family proteins and the efficacy of the combination of gemcitabine and DT2216 (a BCL-XL proteolysis targeting chimera or PROTAC) were determined by MTS, Annexin-V/PI, colony formation, and 3D tumor spheroid assays. The therapeutic efficacy of the combination was investigated in several patient-derived xenograft (PDX) mouse models of pancreatic cancer. We identified BCL-XL as a key mediator of gemcitabine resistance. The combination of gemcitabine and DT2216 synergistically induced cell death in multiple pancreatic cancer cell lines in vitro In vivo, the combination significantly inhibited tumor growth and prolonged the survival of tumor-bearing mice compared with the individual agents in pancreatic cancer PDX models. Their synergistic antitumor activity is attributable to DT2216-induced degradation of BCL-XL and concomitant suppression of MCL-1 by gemcitabine. Our results suggest that DT2216-mediated BCL-XL degradation augments the antitumor activity of gemcitabine and their combination could be more effective for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Dinesh Thummuri
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida
| | - Sajid Khan
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida
| | - Patrick W Underwood
- Department of Surgery, College of Medicine, University of Florida, Gainesville, Florida
| | - Peiyi Zhang
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida
| | - Janet Wiegand
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida
| | - Xuan Zhang
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida
| | - Vivekananda Budamagunta
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida
| | - Amin Sobh
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Abderrahmane Tagmount
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Alexander Loguinov
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Andrea N Riner
- Department of Surgery, College of Medicine, University of Florida, Gainesville, Florida
| | - Ashwin S Akki
- Department of Pathology, College of Medicine, University of Florida, Gainesville, Florida
| | - Elizabeth Williamson
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Robert Hromas
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Christopher D Vulpe
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida
| | - Jose G Trevino
- Department of Surgery, College of Medicine, University of Florida, Gainesville, Florida
- Division of Surgical Oncology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Daohong Zhou
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida.
| |
Collapse
|
4
|
Siddig LA, Khasawneh MA, Samadi A, Saadeh H, Abutaha N, Wadaan MA. Synthesis of novel thiourea-/urea-benzimidazole derivatives as anticancer agents. OPEN CHEM 2021. [DOI: 10.1515/chem-2021-0093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Abstract
A new series of urea and thiourea derivatives containing benzimidazole group as potential anticancer agents have been designed and synthesized. The structures of the synthesized compounds were characterized and confirmed by spectroscopic techniques such as 1H NMR, 13C NMR, and mass spectrometry. In vitro anticancer assay against two breast cancer (BC) cell lines, MDA-MB-231ER(−)/PR(−) and MCF-7ER(+)/PR(+), revealed that the cytotoxicity of 1-(2-(1H-benzo[d]imidazol-2-ylamino)ethyl)-3-p-tolylthiourea (7b) and 4-(1H-benzo[d]imidazol-2-yl)-N-(3-chlorophenyl)piperazine-1-carboxamide (5d) were higher in MCF-7 with IC50 values of 25.8 and 48.3 µM, respectively, as compared with MDA-MB-231 cells. Furthermore, 7b and 5d were assessed for their apoptotic potential using 4′,6-diamidino-2-phenylindole, acridine orange/ethidium bromide staining, and Caspase-3/7. After incubation with MCF-7, the compounds 7b and 5d induced apoptosis through caspase-3/7 activation. In conclusion, the compounds 7b and 5d are potential candidates for inducing apoptosis in different genotypic BC cell lines.
Collapse
Affiliation(s)
- Lamia A. Siddig
- Department of Chemistry, College of Science, United Arab Emirates University , P.O. Box 15551 , Al Ain , United Arab Emirates
| | - Mohammad A. Khasawneh
- Department of Chemistry, College of Science, United Arab Emirates University , P.O. Box 15551 , Al Ain , United Arab Emirates
| | - Abdelouahid Samadi
- Department of Chemistry, College of Science, United Arab Emirates University , P.O. Box 15551 , Al Ain , United Arab Emirates
| | - Haythem Saadeh
- Department of Chemistry, College of Science, United Arab Emirates University , P.O. Box 15551 , Al Ain , United Arab Emirates
- Department of Chemistry, School of Science, The University of Jordan , Amman 11942 , Jordan
| | - Nael Abutaha
- Bioproducts Research Chair, Department of Zoology, College of Science, King Saud University , P.O. Box 2455 , Riyadh 11461 , Saudi Arabia
| | - Mohammad Ahmed Wadaan
- Bioproducts Research Chair, Department of Zoology, College of Science, King Saud University , P.O. Box 2455 , Riyadh 11461 , Saudi Arabia
| |
Collapse
|
5
|
Alam M, Ali S, Mohammad T, Hasan GM, Yadav DK, Hassan MI. B Cell Lymphoma 2: A Potential Therapeutic Target for Cancer Therapy. Int J Mol Sci 2021; 22:ijms221910442. [PMID: 34638779 PMCID: PMC8509036 DOI: 10.3390/ijms221910442] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/15/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022] Open
Abstract
Defects in the apoptosis mechanism stimulate cancer cell growth and survival. B cell lymphoma 2 (Bcl-2) is an anti-apoptotic molecule that plays a central role in apoptosis. Bcl-2 is the founding constituent of the Bcl-2 protein family of apoptosis controllers, the primary apoptosis regulators linked with cancer. Bcl-2 has been identified as being over-expressed in several cancers. Bcl-2 is induced by protein kinases and several signaling molecules which stimulate cancer development. Identifying the important function played by Bcl-2 in cancer progression and development, and treatment made it a target related to therapy for multiple cancers. Among the various strategies that have been proposed to block Bcl-2, BH3-mimetics have appeared as a novel group of compounds thanks to their favorable effects on many cancers within several clinical settings. Because of the fundamental function of Bcl-2 in the regulation of apoptosis, the Bcl-2 protein is a potent target for the development of novel anti-tumor treatments. Bcl-2 inhibitors have been used against several cancers and provide a pre-clinical platform for testing novel therapeutic drugs. Clinical trials of multiple investigational agents targeting Bcl-2 are ongoing. This review discusses the role of Bcl-2 in cancer development; it could be exploited as a potential target for developing novel therapeutic strategies to combat various types of cancers. We further highlight the therapeutic activity of Bcl-2 inhibitors and their implications for the therapeutic management of cancer.
Collapse
Affiliation(s)
- Manzar Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (M.A.); (S.A.); (T.M.)
| | - Sabeeha Ali
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (M.A.); (S.A.); (T.M.)
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (M.A.); (S.A.); (T.M.)
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia;
| | - Dharmendra Kumar Yadav
- Department of Pharmacy and Gachon Institute of Pharmaceutical Science, College of Pharmacy, Gachon University, Hambakmoeiro 191, Yeonsu-gu, Incheon 21924, Korea
- Correspondence: (D.K.Y.); (M.I.H.)
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (M.A.); (S.A.); (T.M.)
- Correspondence: (D.K.Y.); (M.I.H.)
| |
Collapse
|
6
|
Crosstalk between miRNAs and signaling pathways involved in pancreatic cancer and pancreatic ductal adenocarcinoma. Eur J Pharmacol 2021; 901:174006. [PMID: 33711308 DOI: 10.1016/j.ejphar.2021.174006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 01/19/2021] [Accepted: 03/02/2021] [Indexed: 02/08/2023]
Abstract
Pancreatic cancer (PC) is the seventh leading cause of cancer-related deaths worldwide with 5-year survival rates below 8%. Most patients with PC and pancreatic ductal adenocarcinoma (PDAC) die after relapse and cancer progression as well as resistance to treatment. Pancreatic tumors contain a high desmoplastic stroma that forms a rigid mass and has a potential role in tumor growth and metastasis. PC initiates from intraepithelial neoplasia lesions leading to invasive cancer through various pathways. These lesions harbor particular changes in signaling pathways involved in the tumorigenesis process. These events affect both the epithelial cells, including the tumor and the surrounding stroma, and eventually lead to the formation of complex signaling networks. Genetic studies of PC have revealed common molecular features such as the presence of mutations in KRAS gene in more than 90% of patients, as well as the inactivation or deletion mutations of some tumor suppressor genes including TP53, CDKN2A, and SMAD4. In recent years, studies have also identified different roles of microRNAs in PC pathogenesis as well as their importance in PC diagnosis and treatment, and their involvement in various signaling pathways. In this study, we discussed the most common pathways involved in PC and PDAC as well as their role in tumorigenesis and progression. Furthermore, the miRNAs participating in the regulation of these signaling pathways in PC progression are summarized in this study. Therefore, understanding more about pathways involved in PC can help with the development of new and effective therapies in the future.
Collapse
|
7
|
Zhang X, Lu T, Ma Y, Li R, Pang Y, Mao H, Liu P. Novel Nanocomplexes Targeting STAT3 Demonstrate Promising Anti-Ovarian Cancer Effects in vivo. Onco Targets Ther 2020; 13:5069-5082. [PMID: 32606729 PMCID: PMC7292488 DOI: 10.2147/ott.s247398] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 05/06/2020] [Indexed: 12/16/2022] Open
Abstract
Background Cationic solid lipid nanoparticles (SLN) have attracted intensive interest as an effective gene delivery system for its high biocompatibility, stability and low cytotoxicity. In our previous study, we successfully prepared SLN-STAT3 decoy ODN complexes and made a primary study on its antitumor behavior in ovarian cancer cells in vitro. However, there is little information available so far about the effect of SLN-STAT3 decoy ODN complexes on ovarian cancer in vivo, either little information about the pharmacological toxicology in vivo. Material and Methods We applied nanotechnology to improve the gene delivery system and synthesize SLN-STAT3 decoy ODN complexes. Xenograft mouse models were established to assess the antitumor effects of SLN-STAT3 decoy ODN on the tumor growth of ovarian cancer in vivo. To analyze the mechanisms of SLN-STAT3 decoy ODN, we investigated apoptosis, autophagy, epithelial–mesenchymal transition (EMT) in tumor tissues of nude mice and investigated the effects and toxicology of SLN-STAT3 decoy ODN complexes on the vital organs of nude mice. Results The results showed that SLN-STAT3 decoy ODN complexes markedly inhibited tumor growth in vivo. SLN-STAT3 decoy ODN complexes could induce cell apoptosis through downregulating Bcl-2, survivin and pro caspase 3, but upregulating Bax and cleaved caspase 3. These complexes could also regulate autophagy through upregulating LC3A-II, LC3B-II and beclin-1, but downregulating p-Akt and p-mTOR. Moreover, these complexes could inhibit cancer cell invasion through reversing EMT. Besides, SLN-STAT3 decoy ODN complexes showed no obvious toxicity on vital organs and hematological parameters of nude mice. Conclusion The molecular mechanisms that SLN-STAT3 decoy ODN complexes inhibit tumor growth involved activating the apoptotic cascade, regulating autophagy, and reversing EMT program; and these complexes showed no obvious toxicity on nude mice. Our study indicated that the nanocomplexes SLN-STAT3 decoy ODN might be a promising therapeutic approach for ovarian cancer treatment.
Collapse
Affiliation(s)
- Xiaolei Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| | - Tao Lu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| | - Yanhui Ma
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| | - Rui Li
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| | - Yingxin Pang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| | - Hongluan Mao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| | - Peishu Liu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| |
Collapse
|
8
|
Huang Y, Huang H, Wang S, Chen F, Zheng G. Dehydrocorydaline inhibits the tumorigenesis of breast cancer MDA‑MB‑231 cells. Mol Med Rep 2020; 22:43-50. [PMID: 32377708 PMCID: PMC7248526 DOI: 10.3892/mmr.2020.11122] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 12/16/2019] [Indexed: 11/16/2022] Open
Abstract
Dehydrocorydaline (DHC) is an alkaloid isolated from Corydali syanhusuo that exhibits antitumor properties. It has been reported that DHC can inhibit the proliferation of breast cancer cells, however the underlying molecular mechanism remains elusive. Therefore, the main objective of this study was to evaluate the antitumor activity of DHC, and gain further insights into its mechanism of action. The viability of MDA-MB-231 cells was determined through a Cell Counting Kit-8 assay. The effect of DHC on the proliferation of MDA-MB-231 cells was detected by flow cytometry and 5-ethynyl-2′-deoxyuridine staining. Apoptosis was evaluated by Annexin V-FITC and PI staining through flow cytometry. The impact of DHC treatment on the colony-forming ability of breast cancer cells was assessed. The expression levels of proliferation-associated genes cyclin-dependent kinases 1 (CDK1) and cyclin D1 (CCND1) and apoptosis-related genes BCL2 and caspases 3/8/9 were quantified by real-time PCR. Western blot analysis was performed to evaluate the production of cleaved caspase 3/9 and matrix metalloproteinase (MMP)2/9. DHC-treated MDA-MB-231 cells were subcutaneously injected into mice. Subsequent immunohistochemical analyses were performed. DHC inhibited the viability, proliferation, colony-forming ability and migration of MDA-MB-231 cells; in addition, DHC treatment promoted their apoptosis. DHC inhibited the production of proliferation- and anti-apoptosis-associated proteins CDK1, CCND1, BCL2 as well as that of the metastasis-associated proteins MMP2 and MMP9. However, it promoted the expression of the pro-apoptotic caspases 3/8/9. Moreover, DHC inhibited the growth of MDA-MB-231 tumor xenografts in SCID mice, and decreased cell proliferation in newly formed tumors in vivo. DHC exerted anticancer effects by downregulating cell proliferation, antiapoptosis, metastasis-associated proteins CDK1, CCND1, BCL2 and metastasis-associated proteins MMP2 and MMP9, and by upregulating the expression of proapoptotic proteins caspase 3/8/9.
Collapse
Affiliation(s)
- Ying Huang
- Department of Oncology, The Fifth Hospital of Wuhan, Wuhan, Hubei 430050, P.R. China
| | - Hui Huang
- Department of Oncology, The Fifth Hospital of Wuhan, Wuhan, Hubei 430050, P.R. China
| | - Shiying Wang
- Department of Anesthesiology, The Fifth Hospital of Wuhan, Wuhan, Hubei 430050, P.R. China
| | - Feixiang Chen
- Department of General Surgery, The Fifth Hospital of Wuhan, Wuhan, Hubei 430050, P.R. China
| | - Gang Zheng
- Department of General Surgery, The Fifth Hospital of Wuhan, Wuhan, Hubei 430050, P.R. China
| |
Collapse
|
9
|
Bhattacharjee S, Jaiswal RK, Yadava PK. Measles virus phosphoprotein inhibits apoptosis and enhances clonogenic and migratory properties in HeLa cells. J Biosci 2019; 44:10. [PMID: 30837361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Measles virus is the causative agent of measles, a major cause of child mortality in developing countries. Two major proteins, coded by the viral genome, are nucleocapsid protein (N) and phosphoprotein (P). The N protein protects the viral genomic RNA and forms ribonucleoprotein complex (RNP) together with P protein. MeV-P protein recruits the large protein (L), i.e. viral RNA-depended RNA polymerase (RdRp), to ensure viral replication in host cell. Apoptogenic properties of N protein of Edmonston vaccine strain have been established in our lab previously. We investigated the role of MeV-P protein of Edmonston vaccine strain as modulator of apoptosis in cervical cancer cell line (HeLa) and found that MeV-P protein is anti-apoptotic and enhances cell proliferation. Measles virus is considered to be innately oncotropic virus. However, the anti-apoptotic property of MeV-P protein raises important concerns while adopting this virus as an anti-cancer therapeutic tool.
Collapse
Affiliation(s)
- Sankhajit Bhattacharjee
- Applied Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110 067, India
| | | | | |
Collapse
|
10
|
Measles virus phosphoprotein inhibits apoptosis and enhances clonogenic and migratory properties in HeLa cells. J Biosci 2019. [DOI: 10.1007/s12038-018-9834-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
11
|
Wu CW, Lin PJ, Tsai JS, Lin CY, Lin LY. Arsenite-induced apoptosis can be attenuated via depletion of mTOR activity to restore autophagy. Toxicol Res (Camb) 2018; 8:101-111. [PMID: 30713663 DOI: 10.1039/c8tx00238j] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/29/2018] [Indexed: 12/17/2022] Open
Abstract
Arsenic and its compounds are toxic environmental pollutants and known carcinogens. We investigated here the mechanism of arsenite-induced damage in renal cells. Treating human embryonic kidney cells (HEK293) with sodium arsenite reduces cell viability in a dose- and time-dependent manner. The decline of cell viability is due to apoptotic death since arsenite treatment reduces Akt activity and the Bcl2 level but increases caspase 3 activity and the cytochrome c level. These effects can be reverted by the addition of an apoptosis inhibitor. PTEN, the upstream negative regulator of Akt activity, was also reduced with arsenite treatment. Noticeably, PTEN markedly increased in the insoluble fraction of the cells, suggesting a cell failure in removing the damaged proteins. Arsenite treatment activates a variety of signaling factors. Among them, ERK and JNK are associated with autophagy via regulating the levels of LC3 and p62. With arsenite administration, the LC3 and p62 levels increased. However, lysosomal activity was decreased and led to the decline of autophagic activity. The addition of rapamycin, the mTOR inhibitor, activated the autophagic pathway that accelerated the removal of damaged proteins. The recovery of autophagy increased the viability of arsenite-treated cells. Similar to rapamycin treatment, the knockdown of mTOR expression also enhanced the viability of arsenite-treated cells. Both rapamycin treatment and mTOR knockdown enhanced ERK activity further, but reduced JNK activity and the p62 level in arsenite-treated cells. Lysosomal activity increased with the depletion of mTOR, indicating an increase of autophagic activity. These results reveal the critical role of mTOR in regulating the cell fate of arsenite-exposed renal cells.
Collapse
Affiliation(s)
- Chien-Wei Wu
- Institute of Molecular and Cellular Biology and Department of Life Science , National Tsing Hua University , Hsinchu , Taiwan . ; Tel: +886-3-5742693
| | - Pei-Jung Lin
- Institute of Molecular and Cellular Biology and Department of Life Science , National Tsing Hua University , Hsinchu , Taiwan . ; Tel: +886-3-5742693
| | - Jia-Shiuan Tsai
- Institute of Molecular and Cellular Biology and Department of Life Science , National Tsing Hua University , Hsinchu , Taiwan . ; Tel: +886-3-5742693
| | - Chih-Ying Lin
- Institute of Molecular and Cellular Biology and Department of Life Science , National Tsing Hua University , Hsinchu , Taiwan . ; Tel: +886-3-5742693
| | - Lih-Yuan Lin
- Institute of Molecular and Cellular Biology and Department of Life Science , National Tsing Hua University , Hsinchu , Taiwan . ; Tel: +886-3-5742693
| |
Collapse
|
12
|
Discovery of a small-molecule inhibitor of specific serine residue BAD phosphorylation. Proc Natl Acad Sci U S A 2018; 115:E10505-E10514. [PMID: 30309962 DOI: 10.1073/pnas.1804897115] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Human BCL-2-associated death promoter (hBAD) is an apoptosis-regulatory protein mediating survival signals to carcinoma cells upon phosphorylation of Ser99, among other residues. Herein, we screened multiple small-molecule databases queried in a Laplacian-modified naive Bayesian-based cheminformatics platform and identified a Petasis reaction product as a site-specific inhibitor for hBAD phosphorylation. Based on apoptotic efficacy against mammary carcinoma cells, N-cyclopentyl-3-((4-(2,3-dichlorophenyl) piperazin-1-yl) (2-hydroxyphenyl) methyl) benzamide (NPB) was identified as a potential lead compound. In vitro biochemical analyses demonstrated that NPB inhibited the phosphorylation of hBAD specifically on Ser99. NPB was observed to exert this effect independently of AKT and other kinase activities despite the demonstration of AKT-mediated BAD-Ser99 phosphorylation. Using a structure-based bioinformatics platform, we observed that NPB exhibited predicted interactions with hBAD in silico and verified the same by direct binding kinetics. NPB reduced phosphorylation of BAD-Ser99 and enhanced caspase 3/7 activity with associated loss of cell viability in various human cancer cell lines derived from mammary, endometrial, ovarian, hepatocellular, colon, prostatic, and pancreatic carcinoma. Furthermore, by use of a xenograft model, it was observed that NPB, as a single agent, markedly diminished BAD phosphorylation in tumor tissue and significantly inhibited tumor growth. Similar doses of NPB utilized in acute toxicity studies in mice did not exhibit significant effects. Hence, we report a site-specific inhibitor of BAD phosphorylation with efficacy in tumor models.
Collapse
|
13
|
Modi S, Kir D, Banerjee S, Saluja A. Control of Apoptosis in Treatment and Biology of Pancreatic Cancer. J Cell Biochem 2016. [PMID: 26206252 DOI: 10.1002/jcb.25284] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Pancreatic cancer is estimated to be the 12th most common cancer in the United States in 2014 and yet this malignancy is the fourth leading cause of cancer-related death in the United States. Late detection and resistance to therapy are the major causes for its dismal prognosis. Apoptosis is an actively orchestrated cell death mechanism that serves to maintain tissue homoeostasis. Cancer develops from normal cells by accruing significant changes through one or more mechanisms, leading to DNA damage and mutations, which in a normal cell would induce this programmed cell death pathway. As a result, evasion of apoptosis is one of the hallmarks of cancer cells. PDAC is notoriously resistant to apoptosis, thereby explaining its aggressive nature and resistance to conventional treatment modalities. The current review is focus on understanding different intrinsic and extrinsic pathways in pancreatic cancer that may affect apoptosis in this disease.
Collapse
Affiliation(s)
- Shrey Modi
- Division of Basic and Translational Research, Department of Surgery, Minneapolis, Minnesota
| | - Devika Kir
- Division of Basic and Translational Research, Department of Surgery, Minneapolis, Minnesota
| | - Sulagna Banerjee
- Division of Basic and Translational Research, Department of Surgery, Minneapolis, Minnesota
| | - Ashok Saluja
- Division of Basic and Translational Research, Department of Surgery, Minneapolis, Minnesota
| |
Collapse
|
14
|
Momtahen S, Curtin J, Mittal K. Current Chemotherapy and Potential New Targets in Uterine Leiomyosarcoma. J Clin Med Res 2016; 8:181-9. [PMID: 26858789 PMCID: PMC4737027 DOI: 10.14740/jocmr2419w] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2015] [Indexed: 01/12/2023] Open
Abstract
A variety of chemotherapeutic agents have been used for treating recurrent or advanced stage uterine leiomyosarcoma (ULMS). The response rates of these current agents are disappointing, with partial response rates varying from 0% to 33%, and complete response rates varying from 0% to 8%. Recent studies have documented many molecular changes in ULMSs. Prominent amongst these are gains of growth factors C-MYC, Bcl-2, K-ras, and Ki-67, and losses in tumor suppressors p16, p53, Rb1, ING2 and D14S267. Various techniques that have been used to target these molecules are presented. Targeting specific therapies at these underlying molecular changes could potentially yield better response rates with fewer side effects.
Collapse
Affiliation(s)
- Shabnam Momtahen
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - John Curtin
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Langone Medical Center, New York University School of Medicine, New York, NY, USA
| | - Khush Mittal
- Department of Pathology, Langone Medical Center, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
15
|
Muqbil I, Mohammad RM. Selecting efficacious Bcl-2 family inhibitors for optimal clinical outcome. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:312. [PMID: 26697472 DOI: 10.3978/j.issn.2305-5839.2015.09.15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Irfana Muqbil
- 1 Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA ; 2 Interim translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Ramzi M Mohammad
- 1 Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA ; 2 Interim translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
16
|
Park SH, Choi YP, Park J, Share A, Francesconi O, Nativi C, Namkung W, Sessler JL, Roelens S, Shin I. Synthetic aminopyrrolic receptors have apoptosis inducing activity. Chem Sci 2015; 6:7284-7292. [PMID: 28757987 PMCID: PMC5512143 DOI: 10.1039/c5sc03200h] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 09/25/2015] [Indexed: 12/18/2022] Open
Abstract
We report two synthetic aminopyrrolic compounds that induce apoptotic cell death. These compounds have been previously shown to act as receptors for mannosides. The extent of receptor-induced cell death is greater in cells expressing a high level of high-mannose oligosaccharides than in cells producing lower levels of high-mannose glycans. The ability of synthetic receptors to induce cell death is attenuated in the presence of external mannosides. The present results provide support for the suggestion that the observed cell death reflects an ability of the receptors to bind mannose displayed on the cell surface. Signaling pathway studies indicate that the synthetic receptors of the present study promote JNK activation, induce Bax translocation to the mitochondria, and cause cytochrome c release from the mitochondria into the cytosol, thus promoting caspase-dependent apoptosis. Such effects are also observed in cells treated with mannose-binding ConA. The present results thus serve to highlight what may be an attractive new approach to triggering apoptosis via modes of action that differ from those normally used to promote apoptosis.
Collapse
Affiliation(s)
- Seong-Hyun Park
- Center for Biofunctional Molecules , Department of Chemistry , Yonsei University , 03722 Seoul , Korea .
| | - Yoon Pyo Choi
- Center for Biofunctional Molecules , Department of Chemistry , Yonsei University , 03722 Seoul , Korea .
| | - Jinhong Park
- College of Pharmacy , Yonsei Institute of Pharmaceutical Sciences , Yonsei University , 21983 Incheon , Korea
| | - Andrew Share
- Department of Chemistry , The University of Texas at Austin , 78712-1224 Austin , Texas , USA
| | - Oscar Francesconi
- Department of Chemistry and INSTM , University of Florence , Polo Scientifico e Tecnologico, 50019 Sesto Fiorentino , Firenze , Italy
| | - Cristina Nativi
- Department of Chemistry and INSTM , University of Florence , Polo Scientifico e Tecnologico, 50019 Sesto Fiorentino , Firenze , Italy
| | - Wan Namkung
- College of Pharmacy , Yonsei Institute of Pharmaceutical Sciences , Yonsei University , 21983 Incheon , Korea
| | - Jonathan L Sessler
- Department of Chemistry , The University of Texas at Austin , 78712-1224 Austin , Texas , USA
| | - Stefano Roelens
- Istituto di Metodologie Chimiche (IMC) , Consiglio Nazionale delle Ricerche (CNR) , Department of Chemistry and INSTM , University of Florence , Polo Scientifico e Tecnologico, 50019 Sesto Fiorentino , Firenze , Italy
| | - Injae Shin
- Center for Biofunctional Molecules , Department of Chemistry , Yonsei University , 03722 Seoul , Korea .
| |
Collapse
|
17
|
Mohammad RM, Muqbil I, Lowe L, Yedjou C, Hsu HY, Lin LT, Siegelin MD, Fimognari C, Kumar NB, Dou QP, Yang H, Samadi AK, Russo GL, Spagnuolo C, Ray SK, Chakrabarti M, Morre JD, Coley HM, Honoki K, Fujii H, Georgakilas AG, Amedei A, Niccolai E, Amin A, Ashraf SS, Helferich WG, Yang X, Boosani CS, Guha G, Bhakta D, Ciriolo MR, Aquilano K, Chen S, Mohammed SI, Keith WN, Bilsland A, Halicka D, Nowsheen S, Azmi AS. Broad targeting of resistance to apoptosis in cancer. Semin Cancer Biol 2015; 35 Suppl:S78-S103. [PMID: 25936818 PMCID: PMC4720504 DOI: 10.1016/j.semcancer.2015.03.001] [Citation(s) in RCA: 588] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 03/04/2015] [Accepted: 03/04/2015] [Indexed: 12/15/2022]
Abstract
Apoptosis or programmed cell death is natural way of removing aged cells from the body. Most of the anti-cancer therapies trigger apoptosis induction and related cell death networks to eliminate malignant cells. However, in cancer, de-regulated apoptotic signaling, particularly the activation of an anti-apoptotic systems, allows cancer cells to escape this program leading to uncontrolled proliferation resulting in tumor survival, therapeutic resistance and recurrence of cancer. This resistance is a complicated phenomenon that emanates from the interactions of various molecules and signaling pathways. In this comprehensive review we discuss the various factors contributing to apoptosis resistance in cancers. The key resistance targets that are discussed include (1) Bcl-2 and Mcl-1 proteins; (2) autophagy processes; (3) necrosis and necroptosis; (4) heat shock protein signaling; (5) the proteasome pathway; (6) epigenetic mechanisms; and (7) aberrant nuclear export signaling. The shortcomings of current therapeutic modalities are highlighted and a broad spectrum strategy using approaches including (a) gossypol; (b) epigallocatechin-3-gallate; (c) UMI-77 (d) triptolide and (e) selinexor that can be used to overcome cell death resistance is presented. This review provides a roadmap for the design of successful anti-cancer strategies that overcome resistance to apoptosis for better therapeutic outcome in patients with cancer.
Collapse
Affiliation(s)
- Ramzi M Mohammad
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States; Interim translational Research Institute, Hamad Medical Corporation, Doha, Qatar.
| | - Irfana Muqbil
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Leroy Lowe
- Getting to Know Cancer, Truro, Nova Scotia, Canada
| | - Clement Yedjou
- C-SET, [Jackson, #229] State University, Jackson, MS, United States
| | - Hsue-Yin Hsu
- Department of Life Sciences, Tzu-Chi University, Hualien, Taiwan
| | - Liang-Tzung Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Markus David Siegelin
- Department of Pathology and Cell Biology, Columbia University, New York City, NY, United States
| | - Carmela Fimognari
- Dipartimento di Scienze per la Qualità della Vita Alma Mater Studiorum-Università di Bologna, Italy
| | - Nagi B Kumar
- Moffit Cancer Center, University of South Florida College of Medicine, Tampa, FL, United States
| | - Q Ping Dou
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States; Departments of Pharmacology and Pathology, Karmanos Cancer Institute, Detroit MI, United States
| | - Huanjie Yang
- The School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | | | - Gian Luigi Russo
- Institute of Food Sciences National Research Council, Avellino, Italy
| | - Carmela Spagnuolo
- Institute of Food Sciences National Research Council, Avellino, Italy
| | - Swapan K Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Mrinmay Chakrabarti
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - James D Morre
- Mor-NuCo, Inc, Purdue Research Park, West Lafayette, IN, United States
| | - Helen M Coley
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, United Kingdom
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Japan
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Japan
| | - Alexandros G Georgakilas
- Department of Physics, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Zografou 15780, Athens, Greece
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, university of florence, Italy
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, university of florence, Italy
| | - Amr Amin
- Department of Biology, College of Science, UAE University, United Arab Emirates; Faculty of Science, Cairo University, Egypt
| | - S Salman Ashraf
- Department of Chemistry, College of Science, UAE University, United Arab Emirates
| | - William G Helferich
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Xujuan Yang
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Chandra S Boosani
- Department of BioMedical Sciences, School of Medicine Creighton University, Omaha NE, United States
| | - Gunjan Guha
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | - Dipita Bhakta
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | | | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Italy
| | - Sophie Chen
- Ovarian and Prostate Cancer Research Trust Laboratory, Guildford, Surrey, United Kingdom
| | - Sulma I Mohammed
- Department of Comparative Pathobiology and Purdue University Center for Cancer Research, Purdue, West Lafayette, IN, United States
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Ireland
| | - Alan Bilsland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Ireland
| | - Dorota Halicka
- Department of Pathology, New York Medical College, Valhalla, NY, United States
| | - Somaira Nowsheen
- Mayo Graduate School, Mayo Medical School, Mayo Clinic Medical Scientist Training Program, Rochester, MN, United States
| | - Asfar S Azmi
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| |
Collapse
|
18
|
Wang G, Wang Y, Wang L, Han L, Hou X, Fu H, Fang H. Design, synthesis and preliminary bioactivity studies of imidazolidine-2,4-dione derivatives as Bcl-2 inhibitors. Bioorg Med Chem 2015; 23:7359-65. [PMID: 26558516 DOI: 10.1016/j.bmc.2015.10.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 10/14/2015] [Accepted: 10/16/2015] [Indexed: 01/07/2023]
Abstract
Anti-apoptotic B-cell lymphoma-2 (Bcl-2) proteins are promising targets for cancer therapy. In the present study, a series of imidazolidine-2,4-dione derivatives were designed and synthesized to test their inhibitory activities against anti-apoptotic Bcl-2 proteins. Among them, compound 8k had better growth inhibitory effects on K562 and PC-3 cell lines compared to lead compound WL-276.
Collapse
Affiliation(s)
- Gang Wang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmacy, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Yutao Wang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmacy, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Lei Wang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmacy, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Leiqiang Han
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmacy, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Xuben Hou
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmacy, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Huansheng Fu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmacy, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Hao Fang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmacy, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China.
| |
Collapse
|
19
|
Li L, Leung PS. Use of herbal medicines and natural products: an alternative approach to overcoming the apoptotic resistance of pancreatic cancer. Int J Biochem Cell Biol 2014; 53:224-36. [PMID: 24875648 DOI: 10.1016/j.biocel.2014.05.021] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 05/08/2014] [Accepted: 05/16/2014] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer has a poor prognosis with a 5-year survival rate of <5%. It does not respond well to either chemotherapy or radiotherapy, due partly to apoptotic resistance (AR) of the cancer cells. AR has been attributed to certain genetic abnormalities or defects in apoptotic signaling pathways. In pancreatic cancer, significant mutations of K-ras and p53, constitutive activation of NFκB, over-expression of heat shock proteins (Hsp90, Hsp70), histone deacetylase (HDACs) and the activities of other proteins (COX-2, Nrf2 and bcl-2 family members) are closely linked with resistance to apoptosis and invasion. AR has also been associated with aberrant signaling of MAPK, PI3K-AKT, JAK/STAT, SHH, Notch, and Wnt/β-catenin pathways. Strategies targeting these signaling molecules and pathways provide an alternative for overcoming AR in pancreatic cancer. The use of herbal medicines or natural products (HM/NPs) alone or in combination with conventional anti-cancer agents has been shown to produce beneficial effects through actions upon multiple molecular pathways involved in AR. The current standard first-line chemotherapeutic agents for pancreatic cancer are gemcitabine (Gem) or Gem-containing combinations; however, the efficacy is dissatisfied and this limitation is largely attributed to AR. Meanwhile, emerging data have pointed to a combination of HM/NPs that may augment the sensitivity of pancreatic cancer cells to Gem. Greater understanding of how these compounds affect the molecular mechanisms of apoptosis may propel development of HM/NPs as anti-cancer agents and/or adjuvant therapies forward. In this review, we give a critical appraisal of the use of HM/NPs alone and in combination with anti-cancer drugs. We also discuss the potential regulatory mechanisms whereby AR is involved in these protective pathways.
Collapse
Affiliation(s)
- Lin Li
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Po Sing Leung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong.
| |
Collapse
|
20
|
Azam SS, Abro A, Tanvir F, Parvaiz N. Identification of unique binding site and molecular docking studies for structurally diverse Bcl-xL inhibitors. Med Chem Res 2014. [DOI: 10.1007/s00044-014-0957-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
21
|
Song SS, Lee WK, Aluvila S, Oh KJ, Yu YG. Identification of Inhibitors Against BAK Pore Formation using an Improved in vitro Assay System. B KOREAN CHEM SOC 2014. [DOI: 10.5012/bkcs.2014.35.2.419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
22
|
Ilizaliturri-Flores I, Correa-Basurto J, Benítez-Cardoza CG, Zamorano-Carrillo A. A study of the structural properties and thermal stability of human Bcl-2 by molecular dynamics simulations. J Biomol Struct Dyn 2013; 32:1707-19. [PMID: 24028527 DOI: 10.1080/07391102.2013.833858] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The anti-apoptotic B-cell lymphoma 2 (Bcl-2) protein interacts with several proteins that regulate the apoptotic properties of cells. In this research, we conduct several all-atom molecular dynamics (MD) simulations under high-temperature unfolding conditions, from 400 to 800 K, for 25 ns. These simulations were performed using a model of an engineered Bcl-2 human protein (Bcl-2-Δ22Σ3), which lacks 22 C-terminal residues of the transmembrane domain. The aim of this study is to gain insight into the structural behavior of Bcl-2-Δ22Σ3 by mapping the conformational movements involved in Bcl-2 stability and its biological function. To build a Bcl-2-Δ22Σ3 three-dimensional model, the protein core was built by homology modeling and the flexible loop domain (FLD, residues 33-91) by ab initio methods. Further, the entire protein model was refined by MD simulations. Afterwards, the production MD simulations showed that the FLD at 400 and 500 K has several conformations reaching into the protein core, whereas at 600 K some of the alpha-helices were lost. At 800 K, the Bcl-2 core is destabilized suggesting a possible mechanism for protein unfolding, where the alpha helices 1 and 6 were the most stable, and a reduction in the number of hydrogen bonds initially occurs. In conclusion, the structural changes and the internal protein interactions suggest that the core and the FLD are crucial components of Bcl-2 in its function of regulate ng access to the recognition sites of kinases and caspases.
Collapse
Affiliation(s)
- Ian Ilizaliturri-Flores
- a Laboratorio de Investigación Bioquímica, Doctorado en Ciencias en Biotecnología , ENMH, Instituto Politécnico Nacional , Guillermo Massieu Helguera #239 Fracc. "La Escalera" Ticoman, C.P. 07320, D.F. México , Mexico
| | | | | | | |
Collapse
|
23
|
Cao X, Yap JL, Newell-Rogers MK, Peddaboina C, Jiang W, Papaconstantinou HT, Jupitor D, Rai A, Jung KY, Tubin RP, Yu W, Vanommeslaeghe K, Wilder PT, MacKerell AD, Fletcher S, Smythe RW. The novel BH3 α-helix mimetic JY-1-106 induces apoptosis in a subset of cancer cells (lung cancer, colon cancer and mesothelioma) by disrupting Bcl-xL and Mcl-1 protein-protein interactions with Bak. Mol Cancer 2013; 12:42. [PMID: 23680104 PMCID: PMC3663763 DOI: 10.1186/1476-4598-12-42] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 05/02/2013] [Indexed: 11/10/2022] Open
Abstract
Background It has been shown in many solid tumors that the overexpression of the pro-survival Bcl-2 family members Bcl-2/Bcl-xL and Mcl-1 confers resistance to a variety of chemotherapeutic agents. We designed the BH3 α-helix mimetic JY-1-106 to engage the hydrophobic BH3-binding grooves on the surfaces of both Bcl-xL and Mcl-1. Methods JY-1-106–protein complexes were studied using molecular dynamics (MD) simulations and the SILCS methodology. We have evaluated the in vitro effects of JY-1-106 by using a fluorescence polarization (FP) assay, an XTT assay, apoptosis assays, and immunoprecipitation and western-blot assays. A preclinical human cancer xenograft model was used to test the efficacy of JY-1-106 in vivo. Results MD and SILCS simulations of the JY-1-106–protein complexes indicated the importance of the aliphatic side chains of JY-1-106 to binding and successfully predicted the improved affinity of the ligand for Bcl-xL over Mcl-1. Ligand binding affinities were measured via an FP assay using a fluorescently labeled Bak-BH3 peptide in vitro. Apoptosis induction via JY-1-106 was evidenced by TUNEL assay and PARP cleavage as well as by Bax–Bax dimerization. Release of multi-domain Bak from its inhibitory binding to Bcl-2/Bcl-xL and Mcl-1 using JY-1-106 was detected via immunoprecipitation (IP) western blotting. At the cellular level, we compared the growth proliferation IC50s of JY-1-106 and ABT-737 in multiple cancer cell lines with various Bcl-xL and Mcl-1 expression levels. JY-1-106 effectively induced cell death regardless of the Mcl-1 expression level in ABT-737 resistant solid tumor cells, whilst toxicity toward normal human endothelial cells was limited. Furthermore, synergistic effects were observed in A549 cells using a combination of JY-1-106 and multiple chemotherapeutic agents. We also observed that JY-1-106 was a very effective agent in inducing apoptosis in metabolically stressed tumors. Finally, JY-1-106 was evaluated in a tumor-bearing nude mouse model, and was found to effectively repress tumor growth. Strong TUNEL signals in the tumor cells demonstrated the effectiveness of JY-1-106 in this animal model. No significant side effects were observed in mouse organs after multiple injections. Conclusions Taken together, these observations demonstrate that JY-1-106 is an effective pan-Bcl-2 inhibitor with very promising clinical potential.
Collapse
Affiliation(s)
- Xiaobo Cao
- Department of Surgery, Scott & White Memorial Hospital and Clinic, The Texas A&M University System, Health Science Center, College of Medicine, 702 SW HK Dodgen Loop, Temple, Texas 76504, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Dua P, Kang HS, Hong SM, Tsao MS, Kim S, Lee DK. Alkaline phosphatase ALPPL-2 is a novel pancreatic carcinoma-associated protein. Cancer Res 2013; 73:1934-45. [PMID: 23467613 DOI: 10.1158/0008-5472.can-12-3682] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy with a very low median survival rate. The lack of early sensitive diagnostic markers is one of the main causes of PDAC-associated lethality. Therefore, to identify novel pancreatic cancer biomarkers that can facilitate early diagnosis and also help in the development of effective therapeutics, we developed RNA aptamers targeting pancreatic cancer by Cell-systematic evolution of ligands by exponential enrichment (SELEX) approach. Using a selection strategy that could generate aptamers for 2 pancreatic cancer cell lines in one selection scheme, we identified an aptamer SQ-2 that could recognize pancreatic cancer cells with high specificity. Next, by applying 2 alternative approaches: (i) aptamer-based target pull-down and (ii) genome-wide microarray-based identification of differentially expressed mRNAs in aptamer-positive and -negative cells, we identified alkaline phosphatase placental-like 2 (ALPPL-2), an oncofetal protein, as the target of SQ-2. ALPPL-2 was found to be ectopically expressed in many pancreatic cancer cell lines at both mRNA and protein levels. RNA interference-mediated ALPPL-2 knockdown identified novel tumor-associated functions of this protein in pancreatic cancer cell growth and invasion. In addition, the aptamer-mediated identification of ALPPL-2 on the cell surface and cell secretions of pancreatic cancer cells supports its potential use in the serum- and membrane-based diagnosis of PDAC.
Collapse
Affiliation(s)
- Pooja Dua
- Department of Medical Biotechnology, Dongguk University, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
25
|
Mamaghani S, Simpson CD, Cao PM, Cheung M, Chow S, Bandarchi B, Schimmer AD, Hedley DW. Glycogen synthase kinase-3 inhibition sensitizes pancreatic cancer cells to TRAIL-induced apoptosis. PLoS One 2012; 7:e41102. [PMID: 22829912 PMCID: PMC3400624 DOI: 10.1371/journal.pone.0041102] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 06/21/2012] [Indexed: 12/16/2022] Open
Abstract
Tumor necrosis factor-related apoptosis inducing ligand (TRAIL) induces apoptosis in a variety of cancer cell lines with little or no effect on normal cells. However, its effect is limited as some cancers including pancreatic cancer show de novo resistance to TRAIL induced apoptosis. In this study we report that GSK-3 inhibition using the pharmacologic agent AR-18, enhanced TRAIL sensitivity in a range of pancreatic and prostate cancer cell lines. This sensitization was found to be caspase-dependent, and both pharmacological and genetic knock-down of GSK-3 isoforms resulted in apoptotic features as shown by cleavage of PARP and caspase-3. Elevated levels of reactive oxygen intermediates and disturbance of mitochondrial membrane potential point to a mitochondrial amplification loop for TRAIL-induced apoptosis after GSK-3 inhibition. Consistent with this, overexpression of anti-apoptotic mitochondrial targets such as Bcl-XL, Mcl-1, and Bcl-2 rescued PANC-1 and PPC-1 cells from TRAIL sensitization. However, overexpression of the caspase-8 inhibitor CrmA also inhibited the sensitizing effects of GSK-3 inhibitor, suggesting an additional role for GSK-3 that inhibits death receptor signaling. Acute treatment of mice bearing PANC-1 xenografts with a combination of AR-18 and TRAIL also resulted in a significant increase in apoptosis, as measured by caspase-3 cleavage. Sensitization to TRAIL occurred despite an increase in β-catenin due to GSK-3 inhibition, suggesting that the approach might be effective even in cancers with dysregulated β-catenin. These results suggest that GSK-3 inhibitors might be effectively combined with TRAIL for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Shadi Mamaghani
- Division of Applied Molecular Oncology, University Health Network, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Craig D. Simpson
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Pinjiang M. Cao
- Division of Applied Molecular Oncology, University Health Network, Toronto, Ontario, Canada
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - May Cheung
- Division of Applied Molecular Oncology, University Health Network, Toronto, Ontario, Canada
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Sue Chow
- Division of Applied Molecular Oncology, University Health Network, Toronto, Ontario, Canada
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Bizhan Bandarchi
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Aaron D. Schimmer
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
- Department of Medical Oncology and Hematology, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - David W. Hedley
- Division of Applied Molecular Oncology, University Health Network, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
- Department of Medical Oncology and Hematology, Princess Margaret Hospital, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
26
|
Abstract
Celecoxib is a multifaceted drug with promising anticancer properties. A number of studies have been conducted that implicate the compound in modulating the expression of Bcl-2 family members and mitochondria-mediated apoptosis. The growing data surrounding the role of celecoxib in the regulation of the mitochondrial death pathway provides a platform for ongoing debate. Studies that describe celecoxib's properties as a BH3 mimic or as a direct inhibitor of Bcl-2 are not available. The motivations for this review are: to provide the basis for the development of novel compounds that modulate Bcl-2 expression using celecoxib as a structural starting point and to encourage additional biological studies (such as binding and enzymatic assays) that would provide information regarding celecoxib's role as a Bcl-2 antagonist. The current review summarizes work that identifies the role of celecoxib in blocking the activity of Bcl-2.
Collapse
|