1
|
Sheta YS, Sarg MT, Abdulrahman FG, Nossier ES, Husseiny EM. Novel imidazolone derivatives as potential dual inhibitors of checkpoint kinases 1 and 2: Design, synthesis, cytotoxicity evaluation, and mechanistic insights. Bioorg Chem 2024; 149:107471. [PMID: 38823311 DOI: 10.1016/j.bioorg.2024.107471] [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: 04/04/2024] [Revised: 05/03/2024] [Accepted: 05/15/2024] [Indexed: 06/03/2024]
Abstract
Applying various drug design strategies including ring variation, substituents variation, and ring fusion, two series of 2-(alkylthio)-5-(arylidene/heteroarylidene)imidazolones and imidazo[1,2-a]thieno[2,3-d]pyrimidines were designed and prepared as dual potential Chk1 and Chk2 inhibitors. The newly synthesized hybrids were screened in NCI 60 cell line panel where the most active derivatives 4b, d-f, and 6a were further estimated for their five dose antiproliferative activity against the most sensitive tumor cells including breast MCF-7 and MDA-MB-468 and non-small cell lung cancer EKVX as well as normal WI-38 cell. Noticeably, increasing the carbon chain attached to thiol moiety at C-2 of imidazolone scaffold elevated the cytotoxic activity. Hence, compounds 4e and 4f, containing S-butyl fragment, exhibited the most antiproliferative activity against the tested cells where 4f showed extremely potent selectivity toward them. As well, compound 6a, containing imidazothienopyrimidine core, exerted significant cytotoxic activity and selectivity toward the examined cells. The mechanistic investigation of the most active cytotoxic analogs was achieved through the evaluation of their inhibitory activity against Chk1 and Chk2. Results revealed that 4f displayed potent dual inhibition of both Chk1 and Chk2 with IC50 equal 0.137 and 0.25 μM, respectively. It also promoted its antiproliferative and Chk suppression activity via EKVX cell cycle arrest at S phase through stimulating the apoptotic approach. The apoptosis induction was also emphasized by elevating the expression of Caspase-3 and Bax, that are accompanied by Bcl-2 diminution. The in silico molecular docking and ADMET profiles of the most active analogs have been carried out to evaluate their potential as significant anticancer drug candidates.
Collapse
Affiliation(s)
- Yasmin S Sheta
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt
| | - Marwa T Sarg
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt
| | - Fatma G Abdulrahman
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt
| | - Eman S Nossier
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11754, Egypt; The National Committee of Drugs, Academy of Scientific Research and Technology, Cairo 11516, Egypt
| | - Ebtehal M Husseiny
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt.
| |
Collapse
|
2
|
Anisenko AN, Nefedova AA, Kireev II, Gottikh MB. Post-Integrational DNA Repair of HIV-1 Is Associated with Activation of the DNA-PK and ATM Cellular Protein Kinases and Phosphorylation of Their Targets. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1122-1132. [PMID: 38981705 DOI: 10.1134/s0006297924060117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/18/2024] [Accepted: 04/25/2024] [Indexed: 07/11/2024]
Abstract
Integration of the DNA copy of HIV-1 genome into the cellular genome results in series of damages, repair of which is critical for successful replication of the virus. We have previously demonstrated that the ATM and DNA-PK kinases, normally responsible for repairing double-strand breaks in the cellular DNA, are required to initiate the HIV-1 DNA postintegrational repair, even though integration does not result in DNA double-strand breaks. In this study, we analyzed changes in phosphorylation status of ATM (pSer1981), DNA-PK (pSer2056), and their related kinase ATR (pSer428), as well as their targets: Chk1 (pSer345), Chk2 (pThr68), H2AX (pSer139), and p53 (pSer15) during the HIV-1 DNA postintegrational repair. We have shown that ATM and DNA-PK, but not ATR, undergo autophosphorylation during postintegrational DNA repair and phosphorylate their target proteins Chk2 and H2AX. These data indicate common signaling mechanisms between the double-strand DNA break repair and postintegrational repair of HIV-1 DNA.
Collapse
Affiliation(s)
- Andrey N Anisenko
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russia.
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Anastasiia A Nefedova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Igor I Kireev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Marina B Gottikh
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
3
|
Thapa R, Afzal O, Bhat AA, Goyal A, Alfawaz Altamimi AS, Almalki WH, Alzarea SI, Kazmi I, Singh SK, Dua K, Thangavelu L, Gupta G. New horizons in lung cancer management through ATR/CHK1 pathway modulation. Future Med Chem 2023; 15:1807-1818. [PMID: 37877252 DOI: 10.4155/fmc-2023-0164] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. Molecular profiling has contributed to a new classification of lung cancer, driving advancements in research and therapy. The ataxia telangiectasia and rad3/checkpoint kinase 1 (ATR/CHK1) pathway plays a crucial role in maintaining genomic stability, and its activation has been linked to the development of lung cancer, drug resistance and poor prognosis. Clinical and preclinical studies have demonstrated promising results in targeting this pathway. ATR and CHK1 are proteins that collaborate to repair DNA damage caused by radiation or chemotherapy. ATR/CHK1 inhibitors are currently under investigation in preclinical and clinical trials. This article explores the ATR/CHK1 pathway and its potential for treating lung cancer.
Collapse
Affiliation(s)
- Riya Thapa
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, 11942, Saudi Arabia
| | - Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, U.P., India
| | | | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India
- Faculty of Health, Australian Research Centre in Complementary & Integrative Medicine, University of Technology, Sydney, Ultimo, NSW, 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary & Integrative Medicine, University of Technology, Sydney, Ultimo, NSW, 2007, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology, Sydney, Ultimo-NSW, 2007, Australia
| | - Lakshmi Thangavelu
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical & Technical Sciences, Saveetha University, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
- School of Pharmacy, Graphic Era Hill University Dehradun, 248007, India
| |
Collapse
|
4
|
PUMA overexpression dissociates thioredoxin from ASK1 to activate the JNK/BCL-2/BCL-XL pathway augmenting apoptosis in ovarian cancer. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166553. [PMID: 36122664 DOI: 10.1016/j.bbadis.2022.166553] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 09/13/2022] [Accepted: 09/13/2022] [Indexed: 11/23/2022]
Abstract
ASK1-JNK signaling promotes mitochondrial dysfunction-mediated apoptosis, but the bridge between JNK and apoptosis is not fully understood. PUMA induces apoptosis through BAX/BAK. Our previous study suggests a therapeutic potential of PUMA for ovarian cancer. However, whether and how PUMA activates ASK1 remains unclear. Here, we found for the first time that PUMA activated ASK1 by dissociating thioredoxin (TRX) from ASK1, however, it neither interacted with ASK1 nor TRX. Furthermore, PUMA overexpression caused ROS release from mitochondrial. H2O2 significantly impaired the interaction of ASK1 with TRX, whereas ROS scavenger NAC effectively abrogated the H2O2 effect, partly rescued PUMA-interfered interaction of ASK1 with TRX, and also abolished ASK1 phosphorylation. Interestingly, PUMA could not impair the association of ASK1 with TRX-C32S or TRX-C35S, two TRX mutants which are no longer oxidized in response to ROS. We further showed that PUMA activated ASK1-JNK axis to phosphorylate BCL-2 and BCL-XL, further augmenting apoptosis of ovarian cancer cells. In vivo, PUMA adenovirus combined with paclitaxel significantly inhibited intrinsically cisplatin-resistant ovarian cancer growth, and caused phosphorylation of BCL-2 and BCL-XL. Our results from human ovarian cancer TMA chips also revealed a positive correlation between PUMA expression and the phosphorylation of BCL-2 and BCL-XL. More importantly, all patients had no distal metastasis, implying a possibly clinical significance. Collectively, our results reveal a new pro-apoptotic signal amplification mechanism for PUMA by which PUMA overexpression first induces ROS-mediated dissociation of TRX from ASK1, and then causes JNK activation-triggering BCL-2/BCL-XL phosphorylation, ultimately augmenting apoptosis in ovarian cancer.
Collapse
|
5
|
DCZ0014, a novel compound in the therapy of diffuse large B-cell lymphoma via the B cell receptor signaling pathway. Neoplasia 2021; 24:50-61. [PMID: 34890905 DOI: 10.1016/j.neo.2021.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 11/24/2022]
Abstract
Diffuse large B cell lymphoma (DLBCL) is a clinical and genetically heterogeneous lymphoid malignancy. Although R-CHOP (rituximab plus cyclophosphamide, vincristine, doxorubicin, and prednisone) treatment can improve the survival rate of patients with DLBCL, more than 30% of patients exhibit treatment failure, relapse, or refractory disease. Therefore, novel drugs or targeted therapies are needed to improve the survival of patients with DLBCL. The compound DCZ0014 is a novel chemical similar to berberine. In this study, we found that DCZ0014 significantly inhibited the proliferation and activity of DLBCL cells, and induced cell apoptosis. Following treatment with DCZ0014, DLBCL cells accumulated in G0/G1-phase of the cell cycle and showed decreased mitochondrial membrane potential. Additionally, DCZ0014 inhibited DNA synthesis, enhanced DNA damage in DLBCL cells, as well as inhibited Lyn/Syk in B cell receptor signaling pathway. Further experiments demonstrated that DCZ0014 did not significantly affect peripheral blood mononuclear cells. Tumor xenograft model showed that DCZ0014 not only inhibited tumor growth but also extended the survival time of mice. Thus, DCZ0014 showed potential for clinical application in the treatment of patients with DLBCL.
Collapse
|
6
|
Huang B, Chen Q, Wang L, Gao X, Zhu W, Mu P, Deng Y. Aflatoxin B1 Induces Neurotoxicity through Reactive Oxygen Species Generation, DNA Damage, Apoptosis, and S-Phase Cell Cycle Arrest. Int J Mol Sci 2020; 21:ijms21186517. [PMID: 32899983 PMCID: PMC7554769 DOI: 10.3390/ijms21186517] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 12/21/2022] Open
Abstract
Aflatoxin B1 (AFB1) is a mycotoxin widely distributed in a variety of food commodities and exhibits strong toxicity toward multiple tissues and organs. However, little is known about its neurotoxicity and the associated mechanism. In this study, we observed that brain integrity was markedly damaged in mice after intragastric administration of AFB1 (300 μg/kg/day for 30 days). The toxicity of AFB1 on neuronal cells and the underlying mechanisms were then investigated in the neuroblastoma cell line IMR-32. A cell viability assay showed that the IC50 values of AFB1 on IMR-32 cells were 6.18 μg/mL and 5.87 μg/mL after treatment for 24 h and 48 h, respectively. ROS levels in IMR-32 cells increased significantly in a time- and AFB1 concentration-dependent manner, which was associated with the upregulation of NOX2, and downregulation of OXR1, SOD1, and SOD2. Substantial DNA damage associated with the downregulation of PARP1, BRCA2, and RAD51 was also observed. Furthermore, AFB1 significantly induced S-phase arrest, which is associated with the upregulation of CDKN1A, CDKN2C, and CDKN2D. Finally, AFB1 induced apoptosis involving CASP3 and BAX. Taken together, AFB1 manifests a wide range of cytotoxicity on neuronal cells including ROS accumulation, DNA damage, S-phase arrest, and apoptosis-all of which are key factors for understanding the neurotoxicology of AFB1.
Collapse
Affiliation(s)
- Boyan Huang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Tianhe District, Guangzhou 510642, China; (B.H.); (Q.C.); (L.W.); (X.G.); (W.Z.)
- Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China
| | - Qingmei Chen
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Tianhe District, Guangzhou 510642, China; (B.H.); (Q.C.); (L.W.); (X.G.); (W.Z.)
- Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China
| | - Lingling Wang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Tianhe District, Guangzhou 510642, China; (B.H.); (Q.C.); (L.W.); (X.G.); (W.Z.)
- Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China
| | - Xiaojuan Gao
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Tianhe District, Guangzhou 510642, China; (B.H.); (Q.C.); (L.W.); (X.G.); (W.Z.)
- Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China
| | - Wenya Zhu
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Tianhe District, Guangzhou 510642, China; (B.H.); (Q.C.); (L.W.); (X.G.); (W.Z.)
- Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China
| | - Peiqiang Mu
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Tianhe District, Guangzhou 510642, China; (B.H.); (Q.C.); (L.W.); (X.G.); (W.Z.)
- Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China
- Correspondence: (P.M.); (Y.D.); Tel./Fax: +86-20-3860-4967 (Y.D.)
| | - Yiqun Deng
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Tianhe District, Guangzhou 510642, China; (B.H.); (Q.C.); (L.W.); (X.G.); (W.Z.)
- Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China
- Correspondence: (P.M.); (Y.D.); Tel./Fax: +86-20-3860-4967 (Y.D.)
| |
Collapse
|
7
|
Huang B, Mu P, Chen X, Tang S, Ye W, Zhu W, Deng Y. Aflatoxin B 1 induces S phase arrest by upregulating the expression of p21 via MYC, PLK1 and PLD1. Biochem Pharmacol 2019; 166:108-119. [PMID: 31075264 DOI: 10.1016/j.bcp.2019.05.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 05/06/2019] [Indexed: 02/06/2023]
Abstract
Aflatoxin B1 (AFB1), a member of the aflatoxin family, is a common contaminant in foods and feeds, and AFB1 exposure is associated with various clinical conditions. Thus far, research on the toxicity of AFB1 has mainly focused on its induction of liver cancer, but little research has been reported on renal toxicity, especially with regards to the underlying molecular mechanisms. In this study, we found that AFB1 treatment significantly induced kidney damage and reduced kidney weight. The human kidney cell line HEK293T was used to further study the molecular mechanism of the toxicity of AFB1 to kidney cells. We found that AFB1 significantly and dose-dependently induced S phase arrest and upregulated p21 mRNA and protein expression. Upstream of p21, three negative regulators, PLK1, MYC, and PLD1, were significantly downregulated under AFB1 treatment. Consistently, p21 was upregulated, and PLK1, MYC and PLD1 were downregulated in mouse kidney after AFB1 treatment. Interestingly, AFB1 also decreased the physical interaction between PLK1 and MYC and weakened the stability of the MYC protein. Importantly, overexpression of PLK1, MYC and PLD1 significantly blocked the upregulation of p21 and attenuated the S phase arrest caused by AFB1. In summary, AFB1 markedly induces kidney damage and strongly induces S phase arrest by upregulating the expression of p21 via PLK1, PLD1 and MYC, which represents a noval mechanism of the renal toxicity of AFB1.
Collapse
Affiliation(s)
- Boyan Huang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Tianhe District, Guangzhou, Guangdong 510642, PR China; Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong 510642, PR China
| | - Peiqiang Mu
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Tianhe District, Guangzhou, Guangdong 510642, PR China; Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong 510642, PR China
| | - Xiaoxuan Chen
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Tianhe District, Guangzhou, Guangdong 510642, PR China; Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong 510642, PR China
| | - Shulin Tang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Tianhe District, Guangzhou, Guangdong 510642, PR China; Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong 510642, PR China
| | - Wenchu Ye
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Tianhe District, Guangzhou, Guangdong 510642, PR China; Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong 510642, PR China
| | - Wenya Zhu
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Tianhe District, Guangzhou, Guangdong 510642, PR China; Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong 510642, PR China
| | - Yiqun Deng
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Tianhe District, Guangzhou, Guangdong 510642, PR China; Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong 510642, PR China.
| |
Collapse
|
8
|
Wang H, Luo YH, Shen GN, Piao XJ, Xu WT, Zhang Y, Wang JR, Feng YC, Li JQ, Zhang Y, Zhang T, Wang SN, Xue H, Wang HX, Wang CY, Jin CH. Two novel 1,4‑naphthoquinone derivatives induce human gastric cancer cell apoptosis and cell cycle arrest by regulating reactive oxygen species‑mediated MAPK/Akt/STAT3 signaling pathways. Mol Med Rep 2019; 20:2571-2582. [PMID: 31322207 PMCID: PMC6691246 DOI: 10.3892/mmr.2019.10500] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 04/12/2019] [Indexed: 12/15/2022] Open
Abstract
1,4-Naphthoquinone derivatives have superior anticancer effects, but their use has been severely limited in clinical practice due to adverse side effects. To reduce the side effects and extend the anticancer effects of 1,4-naphthoquinone derivatives, 2-(butane-1-sulfinyl)-1,4-naphthoquinone (BQ) and 2-(octane-1-sulfinyl)-1,4-naphthoquinone (OQ) were synthesized, and their anticancer activities were investigated. The anti-proliferation effects, determined by MTT assays, showed that BQ and OQ significantly inhibited the viability of gastric cancer cells and had no significant cytotoxic effect on normal cell lines. The apoptotic effect was determined by flow cytometry, and the results showed that BQ and OQ induced cell apoptosis by regulating the mitochondrial pathway and cell cycle arrest at the G2/M phase via inhibition of the Akt signaling pathway in AGS cells. Furthermore, BQ and OQ significantly increased the levels of reactive oxygen species (ROS) and this effect was blocked by the ROS scavenger NAC in AGS cells. BQ and OQ induced apoptosis by upregulating the protein expression of p38 and JNK and downregulating the levels of ERK and STAT3. Furthermore, expression levels of these proteins were also blocked after NAC treatment. These results demonstrated that BQ and OQ induced apoptosis and cell cycle arrest at the G2/M phase in AGS cells by stimulating ROS generation, which caused subsequent activation of MAPK, Akt and STAT3 signaling pathways. Thus, BQ and OQ may serve as potential therapeutic agents for the treatment of human gastric cancer.
Collapse
Affiliation(s)
- Hao Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Ying-Hua Luo
- Department of Grass Science, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Gui-Nan Shen
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Xian-Ji Piao
- Department of Gynaecology and Obstetrics, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, Heilongjiang 163316, P.R. China
| | - Wan-Ting Xu
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yi Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Jia-Ru Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yu-Chao Feng
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Jin-Qian Li
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yu Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Tong Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Shi-Nong Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Hui Xue
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Hong-Xing Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Chang-Yuan Wang
- Department of Food Science and Engineering, College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Cheng-Hao Jin
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| |
Collapse
|
9
|
Rafoxanide, an organohalogen drug, triggers apoptosis and cell cycle arrest in multiple myeloma by enhancing DNA damage responses and suppressing the p38 MAPK pathway. Cancer Lett 2018; 444:45-59. [PMID: 30583070 DOI: 10.1016/j.canlet.2018.12.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 12/04/2018] [Accepted: 12/11/2018] [Indexed: 12/31/2022]
Abstract
Rafoxanide is used in veterinary medicine for the treatment of fascioliasis. We previously repositioned the drug as the inhibitor of B-Raf V600E, but its anti-tumor effect in human cancer has never been reported. In this study, we investigated the effects of rafoxanide in multiple myeloma (MM) in vitro and in vivo. We found that rafoxanide inhibited cell proliferation and overcame the protective effect of the bone marrow (BM) microenvironment on MM cells. Rafoxanide induced cell apoptosis by reducing mitochondrial membrane potential (MMP) and regulating the caspase pathway, while having no apparent toxic effect on normal cells. Rafoxanide also inhibited DNA synthesis and caused cell cycle arrest by regulating the cdc25A-degradation pathway. In addition, rafoxanide enhanced the DNA damage response by up-regulating the expression of γ-H2AX, and suppressed activation of the p38 MAPK pathway by down-regulating p38 MAPK phosphorylation and Stat1 phosphorylation. Rafoxanide treatment inhibited tumor growth, with no significant side effects, in an MM mouse xenograft model. Combination of rafoxanide with bortezomib or lenalidomide significantly induced synergistic cytotoxicity in MM cells. Finally, rafoxanide had anti-proliferation effect on both wild type and B-Raf V600E mutated MM cells. And the weaker anti-MM activity of rafoxanide than vemurafenib may indicate other potential mechanisms besides targeting B-Raf V600E mutation. Collectively, our results provide a rationale for use of this drug in MM treatment.
Collapse
|
10
|
Ou X, Zhang GT, Xu Z, Chen JS, Xie Y, Liu JK, Liu XP. Desumoylating Isopeptidase 2 (DESI2) Inhibits Proliferation and Promotes Apoptosis of Pancreatic Cancer Cells through Regulating PI3K/AKT/mTOR Signaling Pathway. Pathol Oncol Res 2018; 25:635-646. [DOI: 10.1007/s12253-018-0487-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/09/2018] [Indexed: 12/24/2022]
|
11
|
Yan H, Guo W, Li K, Tang M, Zhao X, Lei Y, Nie CL, Yuan Z. Combination of DESI2 and endostatin gene therapy significantly improves antitumor efficacy by accumulating DNA lesions, inducing apoptosis and inhibiting angiogenesis. Exp Cell Res 2018; 371:50-62. [DOI: 10.1016/j.yexcr.2018.07.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/11/2018] [Accepted: 07/24/2018] [Indexed: 10/28/2022]
|
12
|
Martin B, Coutard B, Guez T, Paesen GC, Canard B, Debart F, Vasseur JJ, Grimes JM, Decroly E. The methyltransferase domain of the Sudan ebolavirus L protein specifically targets internal adenosines of RNA substrates, in addition to the cap structure. Nucleic Acids Res 2018; 46:7902-7912. [PMID: 30192980 PMCID: PMC6125687 DOI: 10.1093/nar/gky637] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 06/29/2018] [Accepted: 07/04/2018] [Indexed: 01/08/2023] Open
Abstract
Mononegaviruses, such as Ebola virus, encode an L (large) protein that bears all the catalytic activities for replication/transcription and RNA capping. The C-terminal conserved region VI (CRVI) of L protein contains a K-D-K-E catalytic tetrad typical for 2'O methyltransferases (MTase). In mononegaviruses, cap-MTase activities have been involved in the 2'O methylation and N7 methylation of the RNA cap structure. These activities play a critical role in the viral life cycle as N7 methylation ensures efficient viral mRNA translation and 2'O methylation hampers the detection of viral RNA by the host innate immunity. The functional characterization of the MTase+CTD domain of Sudan ebolavirus (SUDV) revealed cap-independent methyltransferase activities targeting internal adenosine residues. Besides this, the MTase+CTD also methylates, the N7 position of the cap guanosine and the 2'O position of the n1 guanosine provided that the RNA is sufficiently long. Altogether, these results suggest that the filovirus MTases evolved towards a dual activity with distinct substrate specificities. Whereas it has been well established that cap-dependent methylations promote protein translation and help to mimic host RNA, the characterization of an original cap-independent methylation opens new research opportunities to elucidate the role of RNA internal methylations in the viral replication.
Collapse
Affiliation(s)
- Baptiste Martin
- AFMB, CNRS, Aix-Marseille Université, UMR 7257, Case 925, 163 Avenue de Luminy, 13288 Marseille Cedex 09, France
| | - Bruno Coutard
- AFMB, CNRS, Aix-Marseille Université, UMR 7257, Case 925, 163 Avenue de Luminy, 13288 Marseille Cedex 09, France
| | - Théo Guez
- IBMM, University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Guido C Paesen
- Division of Structural Biology, Wellcome Centre for Human Genetics, Oxford OX3 7BN, UK
| | - Bruno Canard
- AFMB, CNRS, Aix-Marseille Université, UMR 7257, Case 925, 163 Avenue de Luminy, 13288 Marseille Cedex 09, France
| | - Françoise Debart
- IBMM, University of Montpellier, CNRS, ENSCM, Montpellier, France
| | | | - Jonathan M Grimes
- Division of Structural Biology, Wellcome Centre for Human Genetics, Oxford OX3 7BN, UK
- Diamond Light Source Limited, Harwell Science and Innovation Campus, Didcot OX11 0DE, UK
| | - Etienne Decroly
- AFMB, CNRS, Aix-Marseille Université, UMR 7257, Case 925, 163 Avenue de Luminy, 13288 Marseille Cedex 09, France
| |
Collapse
|
13
|
Li JM, Tseng CW, Lin CC, Law CH, Chien YA, Kuo WH, Chou HC, Wang WC, Chan HL. Upregulation of LGALS1 is associated with oral cancer metastasis. Ther Adv Med Oncol 2018; 10:1758835918794622. [PMID: 30159048 PMCID: PMC6109855 DOI: 10.1177/1758835918794622] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 07/04/2018] [Indexed: 12/12/2022] Open
Abstract
Background Oral cancer metastasis is a devastating process that contributes to poor prognosis and high mortality, yet its detailed underlying mechanisms remain unclear. Here, we aimed to evaluate metastasis-specific markers in oral cancer and to provide comprehensive recognition concerning functional roles of the specific target in oral cancer metastasis. Methods Lectin, galactoside-binding, soluble, 1 (LGALS1) was identified by secretomic analysis. LGALS1 expression of patient samples with oral cancer on the tissue microarray were examined by immunochemical (IHC) staining. Small interfering RNA (siRNA)-mediated knockdown of LGALS1 revealed the role of LGALS1 in oral cancer metastasis in vitro and in vivo. Results LGALS1 was observed to be upregulated in highly invasive oral cancer cells, and elevated LGALS1 expression was correlated with cancer progression and lymph node metastasis in oral cancer tissue specimens. Functionally, silencing LGALS1 resulted in suppressed cell growth, wound healing, cell migration, and cell invasion in oral cancer cells in vitro. Knockdown of LGALS1 in highly invasive oral cancer cells dramatically inhibited lung metastasis in an in vivo mouse model. Mechanistic studies suggested p38 mitogen-activated protein kinase (MAPK) phosphorylation, upregulated MMP-9, and mesenchymal phenotypes of epithelial-mesenchymal transition (EMT) in highly invasive oral cancer cells, whereas siRNA against LGALS1 resulted in the inactivation of p38 MAPK pathway, downregulated MMP-9, and EMT inhibition. Conclusions These findings demonstrate that elevated LGALS1 is strongly correlated with oral cancer progression and metastasis, and that it could potentially serve as a prognostic biomarker and an innovative target for oral cancer therapy.
Collapse
Affiliation(s)
- Ji-Min Li
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Chien-Wei Tseng
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Chi-Chen Lin
- Department of Life Sciences, Institute of Biomedical Science, National Chung Hsing University, Taichung, Taiwan
| | - Ching-Hsuan Law
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-An Chien
- Department of Applied Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Wen-Hung Kuo
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsiu-Chuan Chou
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Wen-Ching Wang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Hong-Lin Chan
- Institute of Bioinformatics and Structural Biology and Department of Medical Sciences, National Tsing Hua University, No. 101, Kuang-Fu Rd. Sec. 2, Hsinchu, 30013, Taiwan
| |
Collapse
|
14
|
Navarro SD, Pessatto LR, Meza A, de Oliveira EJT, Auharek SA, Vilela LC, de Lima DP, de Azevedo RB, Kassuya CAL, Cáceres OIA, da Silva Gomes R, Beatriz A, Oliveira RJ, Martines MAU. Resorcinolic lipid 3-heptyl-3,4,6-trimethoxy-3H-isobenzofuran-1-one is a strategy for melanoma treatment. Life Sci 2018; 209:300-312. [PMID: 30102904 DOI: 10.1016/j.lfs.2018.08.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 08/02/2018] [Accepted: 08/08/2018] [Indexed: 12/12/2022]
Abstract
AIMS Previous studies performed by our research group indicated that cytosporone analogues are capable of prevent or repair DNA damages. This work presents the evaluation of the activity of AMS35AA for metastatic murine melanoma cells (B16F10) in experimental model in vitro and, in pre-clinic assay of metastatic melanoma in vivo, using mice lineage C57BL/6. MAIN METHODS In vitro assays were performed: MTT and comet assay, flow cytometry evaluation, gene expression assay by RT-PCR, qualitative evaluation of cell death using B16F10 cells. In vivo assays: micronucleus and comet assay, splenic phagocytosis, melanoma murine model and histopathological analysis, using mice lineage C57BL/6 (n = 20). KEY FINDINGS In vitro results performed by MTT assay showed that AMS35AA is cytotoxic for B16F10 cells (p < 0.05). Based on comet assay the genotoxicity of the IC50 was determined (95.83 μg/mL) (p < 0.05). These data were corroborated by flow cytometry analysis after the treatment with AMS35AA, which indicates the cellular death by apoptosis (p < 0.05) and increasing of ATR, p53, p21 and GADD45 gene expressions verified using RT-PCR. With respect to in vivo results, it was observed that AMS35AA did not show genotoxic activity. Data of tumor volume ex vivo indicate reduction of tumor for the treated animals with AMS35AA up to 15.84×, which is superior to Dacarbazina (50 mg/Kg, p.c.; i.p.). SIGNIFICANCE In summary, the study showed that AMS35AA reveals relevant results regarding to cytotoxicity of B16F10 murine melanoma cells, inducing death by apoptosis via mitochondrial and/or mediated by DNA damages.
Collapse
Affiliation(s)
- Stephanie Dynczuki Navarro
- Graduate Program in Biotechnology and Biodiversity, Pro Midwest Network, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Lucas Roberto Pessatto
- Research Center in Stem Cells, Cell Therapy and Genetic Toxicology (CeTroGen), "Maria Aparecida Pedrossian" University Hospital, Campo Grande, Mato Grosso do Sul, Brazil; Graduate Program in Genetics and Molecular Biology, General Biology Department, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Alisson Meza
- Graduate Program in Chemistry, Chemistry Institute, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Edwin José Torres de Oliveira
- Research Center in Stem Cells, Cell Therapy and Genetic Toxicology (CeTroGen), "Maria Aparecida Pedrossian" University Hospital, Campo Grande, Mato Grosso do Sul, Brazil; Graduate Program in Genetics and Molecular Biology, General Biology Department, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Sarah Alves Auharek
- Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Teófilo Otoni, Minas Gerais, Brazil
| | - Lizia Colares Vilela
- Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Teófilo Otoni, Minas Gerais, Brazil
| | - Dênis Pires de Lima
- Graduate Program in Chemistry, Chemistry Institute, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Ricardo Bentes de Azevedo
- Genetics and Morphology Department, Biosciences Institute, Brasilia University, Brasilia, Federal District, Brazil
| | | | - Osmar Ignacio Ayala Cáceres
- Graduate Program in Chemistry, Chemistry Institute, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Roberto da Silva Gomes
- Graduate Program in Chemistry, Chemistry Institute, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil; Synthesis and Molecular Modification Laboratory, Faculty of Exact Sciences and Technology, Federal University of Grande Dourados, Dourados, Mato Grosso do Sul, Brazil
| | - Adilson Beatriz
- Graduate Program in Chemistry, Chemistry Institute, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Rodrigo Juliano Oliveira
- Research Center in Stem Cells, Cell Therapy and Genetic Toxicology (CeTroGen), "Maria Aparecida Pedrossian" University Hospital, Campo Grande, Mato Grosso do Sul, Brazil; Graduate Program in Genetics and Molecular Biology, General Biology Department, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil; Graduate Program in Health and Development of Midwest Region, Faculty of Medicine "Dr Hélio Mandetta", Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil.
| | - Marco Antonio Utrera Martines
- Graduate Program in Biotechnology and Biodiversity, Pro Midwest Network, Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil; Graduate Program in Chemistry, Chemistry Institute, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil.
| |
Collapse
|
15
|
ATM inhibition induces synthetic lethality and enhances sensitivity of PTEN-deficient breast cancer cells to cisplatin. Exp Cell Res 2018. [PMID: 29522753 DOI: 10.1016/j.yexcr.2018.03.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
PTEN deficiency often causes defects in DNA damage repair. Currently, effective therapies for breast cancer are lacking. ATM is an attractive target for cancer treatment. Previous studies suggested a synthetic lethality between PTEN and PARP. However, the synthetically lethal interaction between PTEN and ATM in breast cancer has not been reported. Moreover, the mechanism remains elusive. Here, using KU-60019, an ATM kinase inhibitor, we investigated ATM inhibition as a synthetically lethal strategy to target breast cancer cells with PTEN defects. We found that KU-60019 preferentially sensitizes PTEN-deficient MDA-MB-468 breast cancer cells to cisplatin, though it also slightly enhances sensitivity of PTEN wild-type breast cancer cells. The increased cytotoxic sensitivity is associated with apoptosis, as evidenced by flow cytometry and PARP cleavage. Additionally, the increase of DNA damage accumulation due to the decreased capability of DNA repair, as indicated by γ-H2AX and Rad51 foci, also contributed to this selective cytotoxicity. Mechanistically, compared with PTEN wild-type MDA-MB-231 cells, PTEN-deficient MDA-MB-468 cells have lower level of Rad51, higher ATM kinase activity, and display the elevated level of DNA damage. Moreover, these differences could be further enlarged by cisplatin. Our findings suggest that ATM is a promising target for PTEN-defective breast cancer.
Collapse
|
16
|
Handayani S, Susidarti RA, Jenie RI, Meiyanto E. Two Active Compounds from Caesalpinia sappan L. in Combination with Cisplatin Synergistically Induce Apoptosis and Cell Cycle Arrest on WiDr Cells. Adv Pharm Bull 2017; 7:375-380. [PMID: 29071219 PMCID: PMC5651058 DOI: 10.15171/apb.2017.045] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 07/17/2017] [Accepted: 07/19/2017] [Indexed: 12/25/2022] Open
Abstract
Purpose: The aim of this study is to observe the synergistic effect of two active compounds of secang, brazilin and brazilein, combined with cisplatin on WiDr colon cancer cells. Methods: Cytotoxic activities of brazilin (Bi) and brazilein (Be) in single and in combination with cisplatin (Cisp) were examined by MTT assay. Synergistic effect was analyzed by combination index (CI) parameter. Apoptosis and cell cycle profiles were observed by using flow cytometry. Results: The result of MTT assay showed that IC50 value of brazilin and brazilein on WiDr cancer cells were 41 µM and 52 µM respectively. The combination of ½ IC50 of Bi-Cisp reduced cells viability up to 64% and showed synergistic effect with CI value less than 1 (CI = 0.8). The combinations of ½ IC50 of Be-Cisp also reduced cells viability up to 78% and showed synergistic effect (CI=0.65). Combination of Bi-Cisp and Be-Cisp induced apoptosis higher than the single treatments. Further analysis on the cell cycle progression showed that single treatment of ½ IC50 of Be and Bi induced S-phase and G2/M-phase accumulation, while combination of Be-Cisp and Bi-Cisp enhanced S-phase accumulation. Conclusion: Both combination of Bi-Cisp and Be-Cisp showed synergistic effect on WiDr cells through induction of apoptosis and halted the cell cycle progression, thus, WiDr cells growth were significantly reduced.
Collapse
Affiliation(s)
- Sri Handayani
- Research Center for Chemistry, Indonesian Institute of Sciences (LIPI), Serpong, Indonesia.,Cancer Chemoprevention Research Center, Faculty of Pharmacy,Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Ratna Asmah Susidarti
- Cancer Chemoprevention Research Center, Faculty of Pharmacy,Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Riris Istighfari Jenie
- Cancer Chemoprevention Research Center, Faculty of Pharmacy,Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Edy Meiyanto
- Cancer Chemoprevention Research Center, Faculty of Pharmacy,Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
17
|
Lin C, Yan H, Yang J, Li L, Tang M, Zhao X, Nie C, Luo N, Wei Y, Yuan Z. Combination of DESI2 and IP10 gene therapy significantly improves therapeutic efficacy against murine carcinoma. Oncotarget 2017; 8:56281-56295. [PMID: 28915590 PMCID: PMC5593561 DOI: 10.18632/oncotarget.17623] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 04/20/2017] [Indexed: 02/05/2023] Open
Abstract
DESI2 (also known as PNAS-4) is a novel pro-apoptotic gene activated during the early response to DNA damage. We previously reported that overexpression of DESI2 induces S phase arrest and apoptosis by activating checkpoint kinases. The present study was designed to test whether combination of DESI2 and IP10 could improve the therapy efficacy in vitro and in vivo. The recombinant plasmid co-expressing DESI2 and IP10 was encapsulated with DOTAP/Cholesterol nanoparticle. Immunocompetent mice bearing CT26 colon carcinoma and LL2 lung cancer were treated with the complex. We found that, in vitro, the combination of DESI2 and IP10 more efficiently inhibited proliferation of CT26, LL2, SKOV3 and A549 cancer cells via apoptosis. In vivo, the combined gene therapy more significantly inhibited tumor growth and efficiently prolonged the survival of tumor bearing mice. Mechanistically, the augmented antitumor activity in vivo was associated with induction of apoptosis and inhibition of angiogenesis. The anti-angiogenesis was further mimicked by inhibiting proliferation of immortalized HUVEC cells in vitro. Meanwhile, the infiltration of lymphocytes also contributed to the enhanced antitumor effects. Depletion of CD8+ T lymphocytes significantly abrogated the antitumor activity, whereas depletion of CD4+ T cells or NK cells showed partial abrogation. Our data suggest that the combined gene therapy of DESI2 and IP10 can significantly enhance the antitumor activity as apoptosis inducer, angiogenesis inhibitor and immune response initiator. The present study may provide a novel and effective method for treating cancer.
Collapse
Affiliation(s)
- Chao Lin
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - HuaYing Yan
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
- Department of Functional Imaging, Sichuan Provincial Women's and Children's Hospital, Chengdu, 610031, China
| | - Jun Yang
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - Lei Li
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - Mei Tang
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - Xinyu Zhao
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - Chunlai Nie
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - Na Luo
- Nankai University School of Medicine, Collaborative Innovation Center of Biotherapy, Tianjin, 300071, China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - Zhu Yuan
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
18
|
Wang Y, Ren N, Rankin GO, Li B, Rojanasakul Y, Tu Y, Chen YC. Anti-proliferative effect and cell cycle arrest induced by saponins extracted from tea ( Camellia sinensis) flower in human ovarian cancer cells. J Funct Foods 2017; 37:310-321. [PMID: 32719725 DOI: 10.1016/j.jff.2017.08.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Tea (Camellia sinensis) flower saponins (TFS) have various biological properties. However, the anti-cancer effects of TFS have not been investigated in any detail. Here, we evaluated the anti-cancer effects of TFS using human ovarian cancer cell lines. TFS (1.5 μg/ml) produced significant antiproliferative effects against A2780/CP70 and OVCAR-3 cells by inducing p53-dependent apoptosis and S phase arrest. Further study showed that TFS decreased mitochondrial membrane potential, activated Caspase-3/7, Caspase-8 and Caspase-9 activities, and that the p53 inhibitor PFT-α reversed the TFS-induced cell growth inhibition and apoptosis. In addition, TFS inhibited the expression of Cdc25A, Cdk2, and CyclinD1 and upregulated Cyclin E and Cyclin A, suggesting that the Cdc25A-Cdk2-Cyclin E/A pathway was involved in TFS-induced S phase arrest. Furthermore, the S phase arrest was associated with a Chk2-Cdc25A DNA damage response. These results demonstrated that TFS has promising potential serving as functional food components for prevention of ovarian cancer.
Collapse
Affiliation(s)
- Yaomin Wang
- Department of Tea Science, Zhejiang University, Hangzhou 310058, P.R. China.,College of Science, Technology and Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA
| | - Ning Ren
- Department of Tea Science, Zhejiang University, Hangzhou 310058, P.R. China
| | - Gary O Rankin
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV25755, USA
| | - Bo Li
- Department of Tea Science, Zhejiang University, Hangzhou 310058, P.R. China
| | - Yon Rojanasakul
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV 26506, USA
| | - Youying Tu
- Department of Tea Science, Zhejiang University, Hangzhou 310058, P.R. China
| | - Yi Charlie Chen
- College of Science, Technology and Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA
| |
Collapse
|
19
|
Lu TN, Ganganna B, Pham TT, Vo AV, Lu TP, Nguyen HGT, Nguyen MNT, Huynh PN, Truong NT, Lee J. Antitumor effect of the integrin α4 signaling inhibitor JK273 in non-small cell lung cancer NCI-H460 cells. Biochem Biophys Res Commun 2017; 491:355-360. [PMID: 28728840 DOI: 10.1016/j.bbrc.2017.07.096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 07/16/2017] [Indexed: 01/14/2023]
Abstract
Lung cancer accounts for the highest death rate among cancers worldwide, with most patients being diagnosed with non-small cell lung cancer (NSCLC), urging more effective therapies. We report that JK273, a pyrrolo[2,3-d]pyrimidine analog, which inhibits α4 integrin signaling, showed a selective cytotoxic effect against HCI-H460 NSCLC cells, with an IC50 of 0.98 ± 0.15 μM, but showed less sensitivity to fibroblasts with a selectivity index (SI) greater than 30. This effect was attributed to cell cycle arrest at S phase by JK273 treatment, resulting in the apoptosis of NCI-H460 cells, further confirmed by exposing phosphatidylserine and morphological changes. Taken together with the previous study of JK273 inhibiting cell migration, we propose that JK273 could serve as an antitumor compound to specifically target cancer cells but not non-cancerous cells by triggering programmed cell death, in addition to anti-metastatic effects in cancer therapy.
Collapse
Affiliation(s)
- Thien Nhan Lu
- College of Pharmacy, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon-do 24341, Republic of Korea; Department of Organic Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, 41 Dinh Tien Hoang Street, District 1, Ho Chi Minh City 701000, Viet Nam.
| | - Bogonda Ganganna
- College of Pharmacy, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon-do 24341, Republic of Korea
| | - Thuy Trang Pham
- College of Pharmacy, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon-do 24341, Republic of Korea
| | - Anh Van Vo
- Department of Immunology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennohdai, Tsukuba, Ibaraki 305-8575, Japan; Human Biology Program, School of Integrative and Global Majors, University of Tsukuba, 1-1-1, Tennohdai, Tsukuba, Ibaraki 305-8575, Japan
| | - Thien Phuc Lu
- Department of Analytical Chemistry- Toxicology- Drug Quality Control, Faculty of Pharmacy, Can Tho University of Medicine and Pharmacy, 179 Nguyen Van Cu Street, Ninh Kieu District, Can Tho City 900000, Viet Nam
| | - Huong-Giang Thi Nguyen
- Department of Organic Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, 41 Dinh Tien Hoang Street, District 1, Ho Chi Minh City 701000, Viet Nam
| | - My-Nuong Thi Nguyen
- Department of Genetics, Faculty of Biology, University of Science, 227 Nguyen Van Cu Street, District 5, Ho Chi Minh City 703000, Viet Nam
| | - Phuong Nguyen Huynh
- Department of Organic Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, 41 Dinh Tien Hoang Street, District 1, Ho Chi Minh City 701000, Viet Nam
| | - Ngoc Tuyen Truong
- Department of Organic Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, 41 Dinh Tien Hoang Street, District 1, Ho Chi Minh City 701000, Viet Nam
| | - Jongkook Lee
- College of Pharmacy, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon-do 24341, Republic of Korea
| |
Collapse
|
20
|
Yang J, Zhao X, Tang M, Li L, Lei Y, Cheng P, Guo W, Zheng Y, Wang W, Luo N, Peng Y, Tong A, Wei Y, Nie C, Yuan Z. The role of ROS and subsequent DNA-damage response in PUMA-induced apoptosis of ovarian cancer cells. Oncotarget 2017; 8:23492-23506. [PMID: 28423586 PMCID: PMC5410321 DOI: 10.18632/oncotarget.15626] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 02/14/2017] [Indexed: 02/05/2023] Open
Abstract
PUMA is a member of the "BH3-only" branch of the BCL-2 family. Our previous study suggests a therapeutic potential of PUMA in treating ovarian cancer, however, the action mechanism of PUMA remains elusive. In this work, we found that in PUMA adenovirus-infected A2780s ovarian cancer cells, exogenous PUMA was partially accumulated in the cytosol and mainly located to the mitochondria. We further showed that PUMA induces mitochondrial dysfunction-mediated apoptosis and ROS generation through functional BAX in a ROS generating enzyme- and caspase-independent manner irrespective of their p53 status, and results in activation of Nrf2/HO-1 pathway. Furthermore, PUMA induces DNA breaks in γ-H2AX staining, and causes activation of DNA damage-related kinases including ATM, ATR, DNA-PKcs, Chk1 and Chk2, which are correlated with the apoptosis. PUMA also results in ROS-triggered JNK activation. Intriguingly, JNK plays a dual role in both DNA damage response and apoptosis, and has an additional contribution to apoptosis. Taken together, we have provided new insight into the action mechanism by which elevated PUMA first induces ROS generation then results in DNA damage response and JNK activation, ultimately contributing to apoptosis in ovarian cancer cells.
Collapse
Affiliation(s)
- Jun Yang
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinyu Zhao
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mei Tang
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lei Li
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yi Lei
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ping Cheng
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wenhao Guo
- 2 Department of Abdominal Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yu Zheng
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Wang
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Na Luo
- 3 Nankai University, School of Medicine/Collaborative Innovation Center of Biotherapy, Tianjin 300071, China
| | - Yong Peng
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Aiping Tong
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuquan Wei
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chunlai Nie
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhu Yuan
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
21
|
Wei R, Guo J, Li M, Yang X, Zhu R, Huang H, Li K, Zhang L, Gao R. Smurf1 controls S phase progression and tumorigenesis through Wee1 degradation. FEBS Lett 2017; 591:1150-1158. [DOI: 10.1002/1873-3468.12624] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 02/28/2017] [Accepted: 03/07/2017] [Indexed: 12/24/2022]
Affiliation(s)
- Rongfei Wei
- Key Laboratory of Human Disease Comparative Medicine; Ministry of Health; Institute of Laboratory Animal Science; Chinese Academy of Medical Sciences & Comparative Medical Center; Peking Union Medical College; Beijing China
| | - Jing Guo
- Key Laboratory of Human Disease Comparative Medicine; Ministry of Health; Institute of Laboratory Animal Science; Chinese Academy of Medical Sciences & Comparative Medical Center; Peking Union Medical College; Beijing China
- Department of Inorganic Non-metallic Materials; School of Materials Science and Engineering; University of Science and Technology Beijing; China
| | - Mengyuan Li
- Key Laboratory of Human Disease Comparative Medicine; Ministry of Health; Institute of Laboratory Animal Science; Chinese Academy of Medical Sciences & Comparative Medical Center; Peking Union Medical College; Beijing China
| | - Xingjiu Yang
- Key Laboratory of Human Disease Comparative Medicine; Ministry of Health; Institute of Laboratory Animal Science; Chinese Academy of Medical Sciences & Comparative Medical Center; Peking Union Medical College; Beijing China
| | - Ruimin Zhu
- Key Laboratory of Human Disease Comparative Medicine; Ministry of Health; Institute of Laboratory Animal Science; Chinese Academy of Medical Sciences & Comparative Medical Center; Peking Union Medical College; Beijing China
| | - Hao Huang
- Key Laboratory of Human Disease Comparative Medicine; Ministry of Health; Institute of Laboratory Animal Science; Chinese Academy of Medical Sciences & Comparative Medical Center; Peking Union Medical College; Beijing China
| | - Kejuan Li
- Key Laboratory of Human Disease Comparative Medicine; Ministry of Health; Institute of Laboratory Animal Science; Chinese Academy of Medical Sciences & Comparative Medical Center; Peking Union Medical College; Beijing China
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics; Beijing Proteome Research Center; Beijing Institute of Radiation Medicine; Collaborative Innovation Center for Cancer Medicine; Beijing China
| | - Ran Gao
- Key Laboratory of Human Disease Comparative Medicine; Ministry of Health; Institute of Laboratory Animal Science; Chinese Academy of Medical Sciences & Comparative Medical Center; Peking Union Medical College; Beijing China
| |
Collapse
|
22
|
Wang Y, Compton C, Rankin GO, Cutler SJ, Rojanasakul Y, Tu Y, Chen YC. 3-Hydroxyterphenyllin, a natural fungal metabolite, induces apoptosis and S phase arrest in human ovarian carcinoma cells. Int J Oncol 2017; 50:1392-1402. [PMID: 28259974 PMCID: PMC5363874 DOI: 10.3892/ijo.2017.3894] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 02/13/2017] [Indexed: 01/20/2023] Open
Abstract
In the present study, we evaluated 3-Hydroxyter-phenyllin (3-HT) as a potential anticancer agent using the human ovarian cancer cells A2780/CP70 and OVCAR-3, and normal human epithelial ovarian cells IOSE-364 as an in vitro model. 3-HT suppressed proliferation and caused cytotoxicity against A2780/CP70 and OVCAR-3 cells, while it exhibited lower cytotoxicity in IOSE-364 cells. Subsequently, we found that 3-HT induced S phase arrest and apoptosis in a dose-independent manner. Further investigation revealed that S phase arrest was related with DNA damage which mediated the ATM/p53/Chk2 pathway. Downregulation of cyclin D1, cyclin A2, cyclin E1, CDK2, CDK4 and Cdc25C, and the upregulation of Cdc25A and cyclin B1 led to the accumulation of cells in S phase. The apoptotic effect was confirmed by Hoechst 33342 staining, depolarization of mitochondrial membrane potential and activation of cleaved caspase-3 and PARP1. Additional results revealed both intrinsic and extrinsic apoptotic pathways were involved. The intrinsic apoptotic pathway was activated through decreasing the protein levels of Bcl2, Bcl-xL and procaspase-9 and increasing the protein level of Puma. The induction of DR5 and DR4 indicated that the extrinsic apoptotic pathway was also activated. Induction of ROS and activation of ERK were observed in ovarian cancer cells. We therefore concluded that 3-HT possessed anti-proliferative effect on A2780/CP70 and OVCAR-3 cells, induced S phase arrest and caused apoptosis. Taken together, we propose that 3-HT shows promise as a therapeutic candidate for treating ovarian cancer.
Collapse
Affiliation(s)
- Yaomin Wang
- Department of Tea Science, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Casey Compton
- College of Science, Technology and Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA
| | - Gary O Rankin
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Stephen J Cutler
- Department of BioMolecular Sciences, University of Mississippi, University, MS 38677, USA
| | - Yon Rojanasakul
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV 26506, USA
| | - Youying Tu
- Department of Tea Science, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Yi Charlie Chen
- College of Science, Technology and Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA
| |
Collapse
|
23
|
Choi HK, Ryu H, Son AR, Seo B, Hwang SG, Song JY, Ahn J. The novel anthraquinone derivative IMP1338 induces death of human cancer cells by p53-independent S and G2/M cell cycle arrest. Biomed Pharmacother 2016; 79:308-14. [PMID: 27044842 DOI: 10.1016/j.biopha.2016.02.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/25/2016] [Accepted: 02/25/2016] [Indexed: 10/22/2022] Open
Abstract
To identify novel small molecules that induce selective cancer cell death, we screened a chemical library containing 1040 compounds in HT29 colon cancer and CCD18-Co normal colon cells, using a phenotypic cell-based viability assay system with the Cell Counting Kit-8 (CCK-8). We discovered a novel anthraquinone derivative, N-(4-[{(9,10-dioxo-9,10-dihydro-1-anthracenyl)sulfonyl}amino]phenyl)-N-methylacetamide (IMP1338), which was cytotoxic against the human colon cancer cells tested. The MTT cell viability assay showed that treatment with IMP1338 selectively inhibited HCT116, HCT116 p53(-/-), HT29, and A549 cancer cell proliferation compared to that of Beas2B normal epithelial cells. To elucidate the cellular mechanism underlying the cytotoxicity of IMP1338, we examined the effect of IMP1338 on the cell cycle distribution and death of cancer cells. IMP1338 treatment significantly arrested the cell cycle at S and G2/M phases by DNA damage and led to apoptotic cell death, which was determined using FACS analysis with Annexin V/PI double staining. Furthermore, IMP1338 increased caspase-3 cleavage in wild-type p53, p53 knockout HCT116, and HT29 cells as determined using immunoblotting. In addition, IMP1338 markedly induced the phosphorylation of histone H2AX and Chk1 in both cell lines while the combination of 5-fluorouracil (5-FU) and radiation inhibited the viability of HCT116, HCT116 p53(-/-), and HT29 cells compared to 5-FU or radiation alone. Our findings indicated that IMP1338 induced p53-independent cell death through S and G2/M phase arrest as well as DNA damage. These results provide a basis for future investigations assessing the promising anticancer properties of IMP1338.
Collapse
Affiliation(s)
- Hyun Kyung Choi
- Department of Medicinal Chemistry, Jungwon University, 85 Munmuro, Goesan 28024, South Korea
| | - Hwani Ryu
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, 75 Nowonro Nowongu, Seoul 01812, South Korea
| | - A-Rang Son
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, 75 Nowonro Nowongu, Seoul 01812, South Korea
| | - Bitna Seo
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, 75 Nowonro Nowongu, Seoul 01812, South Korea
| | - Sang-Gu Hwang
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, 75 Nowonro Nowongu, Seoul 01812, South Korea
| | - Jie-Young Song
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, 75 Nowonro Nowongu, Seoul 01812, South Korea.
| | - Jiyeon Ahn
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, 75 Nowonro Nowongu, Seoul 01812, South Korea.
| |
Collapse
|
24
|
Gu L, Hitzel J, Moll F, Kruse C, Malik RA, Preussner J, Looso M, Leisegang MS, Steinhilber D, Brandes RP, Fork C. The Histone Demethylase PHF8 Is Essential for Endothelial Cell Migration. PLoS One 2016; 11:e0146645. [PMID: 26751588 PMCID: PMC4713448 DOI: 10.1371/journal.pone.0146645] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 12/20/2015] [Indexed: 01/26/2023] Open
Abstract
Epigenetic marks critically control gene expression and thus the cellular activity state. The functions of many epigenetic modifiers in the vascular system have not yet been studied. We screened for histone modifiers in endothelial cells and observed a fairly high expression of the histone plant homeodomain finger protein 8 (PHF8). Given its high expression, we hypothesize that this histone demethylase is important for endothelial cell function. Overexpression of PHF8 catalyzed the removal of methyl-groups from histone 3 lysine 9 (H3K9) and H4K20, whereas knockdown of the enzyme increased H3K9 methylation. Knockdown of PHF8 by RNAi also attenuated endothelial proliferation and survival. As a functional readout endothelial migration and tube formation was studied. PHF8 siRNA attenuated the capacity for migration and developing of capillary-like structures. Given the impact of PHF8 on cell cycle genes, endothelial E2F transcription factors were screened, which led to the identification of the gene repressor E2F4 to be controlled by PHF8. Importantly, PHF8 maintains E2F4 but not E2F1 expression in endothelial cells. Consistently, chromatin immunoprecipitation revealed that PHF8 reduces the H3K9me2 level at the E2F4 transcriptional start site, demonstrating a direct function of PHF8 in endothelial E2F4 gene regulation. Conclusion: PHF8 by controlling E2F4 expression maintains endothelial function.
Collapse
Affiliation(s)
- Lunda Gu
- Institute for Cardiovascular Physiology, Medical Faculty, Goethe-University Frankfurt, Germany
| | - Juliane Hitzel
- Institute for Cardiovascular Physiology, Medical Faculty, Goethe-University Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Franziska Moll
- Institute for Cardiovascular Physiology, Medical Faculty, Goethe-University Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Christoph Kruse
- Institute for Cardiovascular Physiology, Medical Faculty, Goethe-University Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Randa Abdel Malik
- Institute of Vascular Signalling, Centre for Molecular Medicine, Goethe-University Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Jens Preussner
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Mario Looso
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Matthias S. Leisegang
- Institute for Cardiovascular Physiology, Medical Faculty, Goethe-University Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Dieter Steinhilber
- Institute of Pharmaceutical Chemistry/ZAFES, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Ralf P. Brandes
- Institute for Cardiovascular Physiology, Medical Faculty, Goethe-University Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Christian Fork
- Institute for Cardiovascular Physiology, Medical Faculty, Goethe-University Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
- * E-mail:
| |
Collapse
|
25
|
Ruiz de Sabando A, Wang C, He Y, García-Barros M, Kim J, Shroyer KR, Bannister TD, Yang VW, Bialkowska AB. ML264, A Novel Small-Molecule Compound That Potently Inhibits Growth of Colorectal Cancer. Mol Cancer Ther 2015; 15:72-83. [PMID: 26621868 DOI: 10.1158/1535-7163.mct-15-0600] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/14/2015] [Indexed: 12/25/2022]
Abstract
Colorectal cancer is one of the leading causes of cancer mortality in Western civilization. Studies have shown that colorectal cancer arises as a consequence of the modification of genes that regulate important cellular functions. Deregulation of the WNT and RAS/MAPK/PI3K signaling pathways has been shown to be important in the early stages of colorectal cancer development and progression. Krüppel-like factor 5 (KLF5) is a transcription factor that is highly expressed in the proliferating intestinal crypt epithelial cells. Previously, we showed that KLF5 is a mediator of RAS/MAPK and WNT signaling pathways under homeostatic conditions and that it promotes their tumorigenic functions during the development and progression of intestinal adenomas. Recently, using an ultrahigh-throughput screening approach we identified a number of novel small molecules that have the potential to provide therapeutic benefits for colorectal cancer by targeting KLF5 expression. In the current study, we show that an improved analogue of one of these screening hits, ML264, potently inhibits proliferation of colorectal cancer cells in vitro through modifications of the cell-cycle profile. Moreover, in an established xenograft mouse model of colon cancer, we demonstrate that ML264 efficiently inhibits growth of the tumor within 5 days of treatment. We show that this effect is caused by a significant reduction in proliferation and that ML264 potently inhibits the expression of KLF5 and EGR1, a transcriptional activator of KLF5. These findings demonstrate that ML264, or an analogue, may hold a promise as a novel therapeutic agent to curb the development and progression of colorectal cancer.
Collapse
Affiliation(s)
- Ainara Ruiz de Sabando
- Department of Medicine, Stony Brook University School of Medicine, Stony Brook, New York
| | - Chao Wang
- Department of Chemistry, The Scripps Research Institute, Jupiter, Florida
| | - Yuanjun He
- Department of Chemistry, The Scripps Research Institute, Jupiter, Florida
| | | | - Julie Kim
- Department of Medicine, Stony Brook University School of Medicine, Stony Brook, New York
| | - Kenneth R Shroyer
- Department of Pathology, Stony Brook University School of Medicine, Stony Brook, New York
| | - Thomas D Bannister
- Department of Chemistry, The Scripps Research Institute, Jupiter, Florida
| | - Vincent W Yang
- Department of Medicine, Stony Brook University School of Medicine, Stony Brook, New York. Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, New York.
| | - Agnieszka B Bialkowska
- Department of Medicine, Stony Brook University School of Medicine, Stony Brook, New York.
| |
Collapse
|
26
|
Spryszyńska S, Smok-Pieniążek A, Ferlińska M, Roszak J, Nocuń M, Stępnik M. The influence of ATM, ATR, DNA-PK inhibitors on the cytotoxic and genotoxic effects of dibenzo[def,p]chrysene on human hepatocellular cancer cell line HepG2. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2015; 791:12-24. [PMID: 26338538 DOI: 10.1016/j.mrgentox.2015.07.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 07/09/2015] [Accepted: 07/21/2015] [Indexed: 02/06/2023]
Abstract
The effect of inhibitors of phosphatidylinositol-3-kinase related kinases (PIKK): ataxia-telangiectasia mutated (ATM), ATM- and Rad3-related (ATR) and DNA-dependent protein kinase (DNA-PK) on the response of HepG2 human liver cancer cells to dibenzo[def,p]chrysene (DBC) was investigated. High cytotoxicity of DBC (IC50=0.1μM) was observed after 72h incubation. PIKK inhibitors: KU55933 (5μM), NU7026 (10μM) or caffeine (1 and 2mM) when used alone did not significantly influence the cytotoxicity. However, two combinations: KU55933/NU7026 and caffeine/NU7026 significantly increased HepG2 viability (by 25%) after treatment with DBC at 0.5μM. The cytoprotective effect was confirmed by cell cycle and apoptosis/necrosis analysis. DNA damage level after exposure to DBC assessed by comet assay (single strand breaks) showed a long persistence and significant decrease after incubation of the cells in the presence the inhibitors (the combination of KU55933+NU7026 showed the strongest effect). Weak induction of reactive oxygen species (ROS) by DBC (0.5μM) was observed. Although, KU55933 and NU7026 when used alone did not increase ROS levels in the cells, their combination induced the ROS increase and moderately enhanced ROS generation by DBC. We propose a mechanism how cells with damaged DNA after exposure to DBC and under the condition of PIKK inhibition, may be at higher risk of undergoing malignant transformation.
Collapse
Affiliation(s)
- Sylwia Spryszyńska
- Department of Toxicology and Carcinogenesis, Nofer Institute of Occupational Medicine, Łódź, Poland.
| | - Anna Smok-Pieniążek
- Department of Toxicology and Carcinogenesis, Nofer Institute of Occupational Medicine, Łódź, Poland.
| | - Magdalena Ferlińska
- Department of Toxicology and Carcinogenesis, Nofer Institute of Occupational Medicine, Łódź, Poland.
| | - Joanna Roszak
- Department of Toxicology and Carcinogenesis, Nofer Institute of Occupational Medicine, Łódź, Poland.
| | - Marek Nocuń
- Department of Toxicology and Carcinogenesis, Nofer Institute of Occupational Medicine, Łódź, Poland.
| | - Maciej Stępnik
- Department of Toxicology and Carcinogenesis, Nofer Institute of Occupational Medicine, Łódź, Poland.
| |
Collapse
|