251
|
Oh HN, Lee MH, Kim E, Kwak AW, Seo JH, Yoon G, Cho SS, Choi JS, Lee SM, Seo KS, Chae JI, Shim JH. Dual inhibition of EGFR and MET by Echinatin retards cell growth and induces apoptosis of lung cancer cells sensitive or resistant to gefitinib. Phytother Res 2019; 34:388-400. [PMID: 31698509 DOI: 10.1002/ptr.6530] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 10/04/2019] [Accepted: 10/04/2019] [Indexed: 12/14/2022]
Abstract
Patients with non-small-cell lung cancer (NSCLC) containing epidermal growth factor receptor (EGFR) amplification or sensitive mutations initially respond to tyrosine kinase inhibitor gefitinib; however, the treatment is less effective over time. Gefitinib resistance mechanisms include MET gene amplification. A therapeutic strategy targeting MET as well as EGFR can overcome resistance to gefitinib. In the present study we identified Echinatin (Ecn), a characteristic chalcone in licorice, which inhibited both EGFR and MET and strongly altered NSCLC cell growth. The antitumor efficacy of Ecn against gefitinib-sensitive or -resistant NSCLC cells with EGFR mutations and MET amplification was confirmed by suppressing cell proliferation and anchorage-independent colony growth. During the targeting of EGFR and MET, Ecn significantly blocked the kinase activity, which was validated with competitive ATP binding. Inhibition of EGFR and MET by Ecn decreases the phosphorylation of downstream target proteins ERBB3, AKT and ERK compared with total protein expression or control. Ecn induced the G2/M cell cycle arrest, and apoptosis via the intrinsic pathway of caspase-dependent activation. Ecn induced ROS production and GRP78, CHOP, DR5 and DR4 expression as well as depolarized the mitochondria membrane potential. Therefore, our results suggest that Ecn is a promising therapeutic agent in NSCLC therapy.
Collapse
Affiliation(s)
- Ha-Na Oh
- Department of Pharmacy, Mokpo National University, Jeonnam, 58554, Republic of Korea
| | - Mee-Hyun Lee
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450008, P.R. China.,Basic Medical College, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Eunae Kim
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Ah-Won Kwak
- Department of Pharmacy, Mokpo National University, Jeonnam, 58554, Republic of Korea
| | - Ji-Hye Seo
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 Plus, Jeonbuk National University, Jeonju, 54896, Republic of Korea
| | - Goo Yoon
- Department of Pharmacy, Mokpo National University, Jeonnam, 58554, Republic of Korea
| | - Seung-Sik Cho
- Department of Pharmacy, Mokpo National University, Jeonnam, 58554, Republic of Korea
| | - Joon-Seok Choi
- College of Pharmacy, Daegu Catholic University, Gyeongbuk, 38430, Republic of Korea
| | - Sang-Myeong Lee
- Division of Biotechnology, College of Environmental and Bioresources, Jeonbuk National University, Iksan, 54596, Korea
| | - Kang-Seok Seo
- Department of Animal Science and Technology, Sunchon National University, Suncheon, 57922, Republic of Korea
| | - Jung-Il Chae
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 Plus, Jeonbuk National University, Jeonju, 54896, Republic of Korea
| | - Jung-Hyun Shim
- Department of Pharmacy, Mokpo National University, Jeonnam, 58554, Republic of Korea.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450008, P.R. China
| |
Collapse
|
252
|
Kgk D, Kumari S, G S, Malla RR. Marine natural compound cyclo(L-leucyl-L-prolyl) peptide inhibits migration of triple negative breast cancer cells by disrupting interaction of CD151 and EGFR signaling. Chem Biol Interact 2019; 315:108872. [PMID: 31669320 DOI: 10.1016/j.cbi.2019.108872] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/30/2019] [Accepted: 10/21/2019] [Indexed: 12/14/2022]
Abstract
Cyclo (L-Leucyl-L-Prolyl) peptide/CLP is a marine natural metabolite and well recognized as an antimicrobial and antioxidant agent with limited studies on anticancer activity. The current study aims to determine the effect of CLP on migration and growth of triple negative breast cancer cell lines. The anti-growth potential was evaluated by MTT, BrdU and TUNEL assays; DNA damage by γH2AX and Dead green assays; antimigration activity by Boyden chamber invasion and wound healing assays. Interaction of CLP with CD151 was resolved by PatchDock. Effect of CLP on the expression of transmembrane CD151 was evaluated by cell-based ELISA assay. The interaction between CD151 and EGFR was predicted by using FireDoc Web server. Impact of CLP on the interaction of CD151 with EGFR was evaluated by co-immunoprecipitation assay. The effect of CLP on the cell cycle and its controlling proteins was determined by Western blotting. CLP reduced the viability of MDA-MB-231 and MDA-MB-468 TNBC cell lines but not human breast healthy epithelial cell line (MCF-12A) similar to eribulin, standard. CLP also inhibited proliferation; cell cycle and migration. It induced DNA strand breaks, DNA damage, and cell death. It showed the most favorable interactions with CD151 in in silico docking and significantly reduced the expression of membrane-bound CD151 proteins. FireDoc Web study predicted the association between CD151 and EGFR with -29.13 kcal/mol of binding energy. CLP reduced the interaction of CD151 with EGFR along with the expression of cyclin D, CDK4, PAK, RAC1, and P27kiP1. This study concludes that CLP suppresses growth and migration by attenuating cell cycle of TNBC cell lines via EGFR and CD151 signaling. Thus, exploring the EGFR and CD151 signaling pathway targeted by CLP may provide a new approach in the treatment of TNBC.
Collapse
Affiliation(s)
- Deepak Kgk
- Cancer Biology Lab, Department of Biochemistry, GIS, GITAM (Deemed to be University), Visakhapatnam, 530045, Andhra Pradesh, India
| | - Seema Kumari
- Cancer Biology Lab, Department of Biochemistry, GIS, GITAM (Deemed to be University), Visakhapatnam, 530045, Andhra Pradesh, India
| | - Shailender G
- Cancer Biology Lab, Department of Biochemistry, GIS, GITAM (Deemed to be University), Visakhapatnam, 530045, Andhra Pradesh, India
| | - Rama Rao Malla
- Cancer Biology Lab, Department of Biochemistry, GIS, GITAM (Deemed to be University), Visakhapatnam, 530045, Andhra Pradesh, India.
| |
Collapse
|
253
|
Chan ZCK, Leong KH, Kareem HS, Norazit A, Noor SM, Ariffin A. Activation of death receptor, DR5 and mitochondria-mediated apoptosis by a 3,4,5-trimethoxybenzyloxy derivative in wild-type and p53 mutant colorectal cancer cell lines. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2019; 393:405-417. [PMID: 31641820 DOI: 10.1007/s00210-019-01730-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/06/2019] [Indexed: 10/25/2022]
Abstract
The rationale of designing compounds containing a (3,4,5-trimethoxybenzyloxy) phenyl moiety is largely due to its potential antioxidant and cytotoxic activities. A previous study focused on its antioxidant mechanism, whereas in this study, we investigated the cytotoxicity of a series of 28 analogues and the mechanism of apoptosis of the most cytotoxic compound against wild-type (HCT-116) and p53 mutant (HT-29) colorectal cancer cell lines. The series of analogues comprise of different families, namely hydrazone, oxadiazole, thiosemicarbazides and triazoles. In the initial cytotoxicity screening, N-(3,4,5-trimethoxybenzylidene)-4-(3,4,5-trimethoxybenzyloxy) benzohydrazide, henceforth known as, P5H, was found to be most cytotoxic against human colorectal cancer cell lines (IC50 for HCT-116 = 11.79 μM and HT-29 = 18.52 μM). Additionally, P5H was found to have some degree of selectivity towards cancer cells compared to normal human colon cells (CCD-112 CoN). Subsequent investigation had brought insight on P5H ability to induce apoptosis in both HCT-116 and HT-29 cell lines. Cell cycle analysis showed both cell lines were arrested at the G2/M phase upon treatment. Our study concluded that P5H induced the death receptor, DR5 in HCT-116 and mitochondria-mediated apoptosis pathway in HT-29. Therefore, P5H may be a promising candidate as a chemotherapy agent against colon cancer. Graphical abstract The apoptotic pathways induced in HT-29 and HCT-116 cells upon P5H treatment.
Collapse
Affiliation(s)
- Zachariah Chee Ken Chan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Kok Hoong Leong
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Malaya, 50603, Kuala Lumpur, Malaysia. .,Center for Natural Product and Drug Discovery (CENAR), Department of Chemistry, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Huda Salah Kareem
- General Directorate of Curricular, Ministry of Education, Baghdad, 3310, Iraq
| | - Anwar Norazit
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Suzita Mohd Noor
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Azhar Ariffin
- Department of Chemistry, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
254
|
Galvis D, Walsh D, Harries LW, Latorre E, Rankin J. A dynamical systems model for the measurement of cellular senescence. J R Soc Interface 2019; 16:20190311. [PMID: 31594522 PMCID: PMC6833332 DOI: 10.1098/rsif.2019.0311] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Senescent cells provide a good in vitro model to study ageing. However, cultures of ‘senescent’ cells consist of a mix of cell subtypes (proliferative, senescent, growth-arrested and apoptotic). Determining the proportion of senescent cells is crucial for studying ageing and developing new anti-degenerative therapies. Commonly used markers such as doubling population, senescence-associated β-galactosidase, Ki-67, γH2AX and TUNEL assays capture diverse and overlapping cellular populations and are not purely specific to senescence. A newly developed dynamical systems model follows the transition of an initial culture to senescence tracking population doubling, and the proportion of cells in proliferating, growth-arrested, apoptotic and senescent states. Our model provides a parsimonious description of transitions between these states accruing towards a predominantly senescent population. Using a genetic algorithm, these model parameters are well constrained by an in vitro human primary fibroblast dataset recording five markers at 16 time points. The computational model accurately fits to the data and translates these joint markers into the first complete description of the proportion of cells in different states over the lifetime. The high temporal resolution of the dataset demonstrates the efficacy of strategies for reconstructing the trajectory towards replicative senescence with a minimal number of experimental recordings.
Collapse
Affiliation(s)
- Daniel Galvis
- Living Systems Institute, University of Exeter, Exeter, UK.,Translational Research Exchange at Exeter, University of Exeter, Exeter, UK
| | - Darren Walsh
- Institute of Biomedical and Clinical Science, University of Exeter, Medical School, RILD Building, Barrack Road, Exeter EX2 5DW, UK
| | - Lorna W Harries
- Institute of Biomedical and Clinical Science, University of Exeter, Medical School, RILD Building, Barrack Road, Exeter EX2 5DW, UK
| | - Eva Latorre
- Institute of Biomedical and Clinical Science, University of Exeter, Medical School, RILD Building, Barrack Road, Exeter EX2 5DW, UK.,Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, Zaragoza, Spain
| | - James Rankin
- Department of Mathematics, College of Engineering, Mathematics and Physical Sciences, University of Exeter, Harrison Building, North Park Road, Exeter EX4 4QF, UK.,EPSRC Centre for Predictive Modelling in Healthcare, University of Exeter, Exeter EX4 4QJ, UK
| |
Collapse
|
255
|
Ulieme RE, Awer S, Stagg JC, Yau W, Gato WE. The hepatic effects in dams that ingested 2-aminoanthracene during gestation and lactation. Toxicol Ind Health 2019; 35:568-576. [PMID: 31558114 DOI: 10.1177/0748233719875018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Diabetes mellitus has been on a continual rise as one of the top chronic diseases to affect individuals worldwide. The goal of this study was to determine how exposure from a well-known toxicant, a polycyclic aromatic hydrocarbon called 2-aminoanthracene (2AA), could potentially lead to diabetes, damage the liver, and have negative effects to the offspring. Humans are exposed to 2AA from foods cooked in high heat and tobacco smoke, among others. To analyze the effects of 2AA, three groups of Sprague Dawley dams consumed an adulterated 2AA diet from gestation to their postnatal period. Timed-pregnant dams ingested 0 mg/kg (control group (C)), 50 mg/kg (low dose group (LD)), and 100 mg/kg (high dose group (HD)) 2AA. Hepatic gene expressions of Adam8, Bax, Ccng1, CD68, CD93, Cdkn1c, and Ddit4 indicated a significant overexpression of Bax, Ccng1, CD68, CD93, and Cdkn1c in treated groups. Although there was no significant difference in the damage to the liver architecture by 2AA, the positively stained CD68+ cells were slightly increased in treated rats. Significant decreases in the albumin and aspartate aminotransferase levels might indicate an inflammatory response from 2AA exposure in dams. Immunoglobulin A (IgA) concentration was also decreased, in contrast to studies of liver cirrhosis that reported increased serum IgA concentration. Overexpression of genes Ddit4, Cdkn1c, Ccng1, Bax, CD93, and CD68 point to hepatic inflammation and apoptosis. Overall results suggest a link between environmental 2AA exposure and adverse liver effects, which has potential to increase susceptibility to type 2 diabetes and other diseases.
Collapse
Affiliation(s)
- Raven E Ulieme
- Department of Chemistry and Biochemistry, Georgia Southern University, Statesboro, GA, USA
| | - Surjania Awer
- Department of Chemistry and Biochemistry, Georgia Southern University, Statesboro, GA, USA
| | - John C Stagg
- Department of Chemistry and Biochemistry, Georgia Southern University, Statesboro, GA, USA
| | - Wilson Yau
- Department of Pathology, College of Veterinary Medicine, The University of Georgia, Athens, GA, USA
| | - Worlanyo E Gato
- Department of Chemistry and Biochemistry, Georgia Southern University, Statesboro, GA, USA
| |
Collapse
|
256
|
Accumulated cytotoxicity of CDK inhibitor dinaciclib with first-line chemotherapy drugs in salivary adenoid cystic carcinoma cells. Odontology 2019; 108:300-311. [PMID: 31529315 DOI: 10.1007/s10266-019-00451-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 08/15/2019] [Indexed: 01/31/2023]
Abstract
Adenoid cystic carcinoma (ACC) is one of the most common salivary gland malignant tumors. Its treatment failure is partly due to the limitations of chemotherapeutic agents and their adverse effects. The objective of this study was to determine the potential additive anti-cancer effect of a novel CDK inhibitor dinaciclib with first-line chemotherapy drugs in ACC. Protein expression of phosphorylated CDK2 (p-CDK2) in paraffin-embedded tissue specimens of ACC from 17 patients was investigated by immunohistochemistry (IHC). Cell Counting Kit (CCK-8), clone formation assay, and flow cytometry were used to test the proliferation and apoptosis of ACC-2 cells treated with dinaciclib with or without other first-line chemotherapy drugs. Protein expression was also determined by Western blot. Interestingly, we discovered that p-CDK2 protein was expressed in both cytoplasmic and nucleus in salivary ACC tissues, which was higher than that in normal salivary tissues, indicating that agents targeting CDK2 may be potential therapeutic strategies against this type of tumor. As expected, CDK inhibitor dinaciclib significantly induced ACC-2 cells apoptosis. Moreover, it sensitized cells to the chemotherapeutic agents such as cisplatin, pemetrexed, and etoposide (VP-16), and this effect by dinaciclib may induce cell cycle arrest via abrogating CDK2 activity. Therefore, combinational therapy of CDK inhibitor dinaciclib with first-line chemotherapy drugs may be a promising strategy in the treatment of salivary ACC.
Collapse
|
257
|
Cho HD, Min HJ, Won YS, Ahn HY, Cho YS, Seo KI. Solid state fermentation process with Aspergillus kawachii enhances the cancer-suppressive potential of silkworm larva in hepatocellular carcinoma cells. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:241. [PMID: 31488109 PMCID: PMC6727413 DOI: 10.1186/s12906-019-2649-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 08/21/2019] [Indexed: 12/14/2022]
Abstract
Background Mulberry silkworm larvae (Bombyx mori) are known as the oldest resource of food and traditional medicine. Although silkworm larvae have been reported to treat various chronic diseases, the effect of fermentation by microorganisms improving the biological activities of silkworm larvae was not reported. In the present study, fermented silkworm larvae was developed via solid-state fermentation with Aspergillus kawachii and investigated its anti-cancer activity in human hepatocellular carcinoma cells. Methods We investigated the anti-cancer effects of unfermented (SEE) and fermented silkworm larva ethanol extract (FSEE) on HepG2 human hepatocellular carcinoma cells as well as compared changes in free amino acid, fatty acid, and mineral contents. Anti-cancer activities were evaluated by SRB staining, cell cycle analysis, Annexin V staining, Hoechst staining, DNA fragmentation analysis and western blot analysis. Fatty acid, free amino acid and mineral contents of SEE and FSEE were determined by gas chromatography, amino acid analyzer and flame atomic absorption spectrophotometer, respectively. Results Compared with SEE, treatment with FSEE resulted in apoptotic cell death in HepG2 cells characterized by G0/G1 phase cell cycle arrest, DNA fragmentation, and formation of apoptotic bodies. Furthermore, FSEE significantly up-regulated pro-apoptotic as well as down-regulated anti-apoptotic proteins in HepG2 cells. However, an equivalent concentration of SEE did not induce cell cycle arrest or apoptosis in HepG2 cells. Moreover, fermentation process by Aspergillus kawachii resulted in enhancement of fatty acid contents in silkworm larvae, whereas amino acid and mineral contents were decreased. Conclusion Collectively, this study demonstrates that silkworm larvae solid state-fermented by Aspergillus kawachii strongly potentiates caspase-dependent and -independent apoptosis pathways in human hepatocellular carcinoma cells by regulating secondary metabolites. Electronic supplementary material The online version of this article (10.1186/s12906-019-2649-7) contains supplementary material, which is available to authorized users.
Collapse
|
258
|
Respondek M, Beberok A, Rzepka Z, Rok J, Wrześniok D. MIM1 induces COLO829 melanoma cell death through mitochondrial membrane breakdown, GSH depletion, and DNA damage. Fundam Clin Pharmacol 2019; 34:20-31. [PMID: 31410885 DOI: 10.1111/fcp.12503] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 08/02/2019] [Accepted: 08/09/2019] [Indexed: 12/14/2022]
Abstract
Malignant melanoma is a high aggressive malignancy in humans and causes 60-80% of deaths from skin cancer. Defect in an intrinsic pathway of apoptosis via overexpression of Mcl-1 is responsible for malignant melanoma development and progression, and also for resistance to chemotherapeutic agents. MIM1 is a specific low molecular Mcl-1 protein inhibitor that is able to induce Mcl-1-dependent cancer cells death. Here, we examined the effect of MIM1 as well as MIM1 and dacarbazine (DTIC) mixture on cell viability, apoptosis, and cell cycle progression in COLO829 melanoma cells. Cell viability was performed by the WST-1 assay. Analysis of apoptosis as well as cell cycle progression was determined by fluorescence image cytometer NucleoCounter NC-3000. The obtained results demonstrated that the MIM1 exhibited high cytotoxicity against melanotic melanoma cells and induced mitochondrial membrane breakdown, GSH depletion, and DNA fragmentation. Additionally, MIM1 enhanced the proapoptotic effect of DTIC toward melanoma cells; furthermore, a mixture of these drugs caused cell cycle arrest at G2/M phase in COLO829 cells. Taken together, these data provide, for the first time, evidence that a low molecular weight Mcl-1 inhibitor-MIM1 may be a promising agent with antitumor and proapoptotic properties toward melanoma cells.
Collapse
Affiliation(s)
- Michalina Respondek
- Department of Pharmaceutical Chemistry, School of Pharmacy with the Division of Laboratory Medicine, Medical University of Silesia, Jagiellońska, 441-200, Sosnowiec, Poland
| | - Artur Beberok
- Department of Pharmaceutical Chemistry, School of Pharmacy with the Division of Laboratory Medicine, Medical University of Silesia, Jagiellońska, 441-200, Sosnowiec, Poland
| | - Zuzanna Rzepka
- Department of Pharmaceutical Chemistry, School of Pharmacy with the Division of Laboratory Medicine, Medical University of Silesia, Jagiellońska, 441-200, Sosnowiec, Poland
| | - Jakub Rok
- Department of Pharmaceutical Chemistry, School of Pharmacy with the Division of Laboratory Medicine, Medical University of Silesia, Jagiellońska, 441-200, Sosnowiec, Poland
| | - Dorota Wrześniok
- Department of Pharmaceutical Chemistry, School of Pharmacy with the Division of Laboratory Medicine, Medical University of Silesia, Jagiellońska, 441-200, Sosnowiec, Poland
| |
Collapse
|
259
|
Anaya-Eugenio GD, Rebollar-Ramos D, González MDC, Raja H, Mata R, Carcache de Blanco EJ. Apoptotic activity of xanthoquinodin JBIR-99, from Parengyodontium album MEXU 30054, in PC-3 human prostate cancer cells. Chem Biol Interact 2019; 311:108798. [DOI: 10.1016/j.cbi.2019.108798] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/08/2019] [Accepted: 08/17/2019] [Indexed: 12/15/2022]
|
260
|
Respondek M, Beberok A, Rzepka Z, Rok J, Wrześniok D. Mcl-1 Inhibitor Induces Cells Death in BRAF-Mutant Amelanotic Melanoma Trough GSH Depletion, DNA Damage and Cell Cycle Changes. Pathol Oncol Res 2019; 26:1465-1474. [PMID: 31432325 PMCID: PMC7297871 DOI: 10.1007/s12253-019-00715-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 08/12/2019] [Indexed: 01/10/2023]
Abstract
Mcl-1 is a potent antiapoptotic protein and amplifies frequently in many human cancer. Currently, it is considered that the extensively expressed of Mcl-1 protein in melanoma cells is associated with rapid tumor progression, poor prognosis and low chemosensitivity. Therefore, the antiapoptotic protein Mcl-1 could be considered as a potential target for malignant melanoma treatment. The aim of this study was to assess the effect of MIM1 a specific low molecular Mcl-1 protein inhibitor and mixture of MIM1 and dacarbazine on the viability, cell cycle progression and apoptosis induction in amelanotic C32 melanoma cells. The cytotoxic activity of MIM1 towards C32 melanoma cells was examined by the WST-1 test. The Mcl-1 protein level as a drug target in amelanotic melanoma cells was defined by Western blot analysis. Cell cycle progression, DNA fragmentation as well as GSH depletion were determined by fluorescence image cytometer NucleoCounter NC-3000. The obtained results demonstrate that the specific Mcl-1 protein inhibitor - MIM1 decreases cell viability and induce apoptosis (S-phase arrest, DNA fragmentation and redox imbalance) in amelanotic melanoma cells and intensify the proapoptotic properties of DTIC, as a result of interactions with Mcl-1 protein. Taken together, the presented data suggest that Mcl-1 protein is a an important target in malignant melanoma treatment and provide for the first time convincing evidence that MIM1, which inhibits Mcl-1 antiapoptotic protein is able to induce apoptosis and sensitize melanoma cells to alkylating agent.
Collapse
Affiliation(s)
- Michalina Respondek
- School of Pharmacy with the Division of Laboratory Medicine, Department of Pharmaceutical Chemistry, Medical University of Silesia, Jagiellońska 4, 41-200, Sosnowiec, Poland.
| | - Artur Beberok
- School of Pharmacy with the Division of Laboratory Medicine, Department of Pharmaceutical Chemistry, Medical University of Silesia, Jagiellońska 4, 41-200, Sosnowiec, Poland
| | - Zuzanna Rzepka
- School of Pharmacy with the Division of Laboratory Medicine, Department of Pharmaceutical Chemistry, Medical University of Silesia, Jagiellońska 4, 41-200, Sosnowiec, Poland
| | - Jakub Rok
- School of Pharmacy with the Division of Laboratory Medicine, Department of Pharmaceutical Chemistry, Medical University of Silesia, Jagiellońska 4, 41-200, Sosnowiec, Poland
| | - Dorota Wrześniok
- School of Pharmacy with the Division of Laboratory Medicine, Department of Pharmaceutical Chemistry, Medical University of Silesia, Jagiellońska 4, 41-200, Sosnowiec, Poland
| |
Collapse
|
261
|
Ghosh C, Nandi A, Basu S. Lipid Nanoparticle-Mediated Induction of Endoplasmic Reticulum Stress in Cancer Cells. ACS APPLIED BIO MATERIALS 2019; 2:3992-4001. [DOI: 10.1021/acsabm.9b00532] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Chandramouli Ghosh
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune, Maharashtra 411008, India
| | - Aditi Nandi
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune, Maharashtra 411008, India
| | - Sudipta Basu
- Discipline of Chemistry, Indian Institute of Technology (IIT) Gandhinagar, Palaj, Gandhinagar, Gujarat 382355, India
| |
Collapse
|
262
|
Vázquez-Borrego MC, Fuentes-Fayos AC, Herrera-Martínez AD, L-López F, Ibáñez-Costa A, Moreno-Moreno P, Alhambra-Expósito MR, Barrera-Martín A, Blanco-Acevedo C, Dios E, Venegas-Moreno E, Solivera J, Gahete MD, Soto-Moreno A, Gálvez-Moreno MA, Castaño JP, Luque RM. Biguanides Exert Antitumoral Actions in Pituitary Tumor Cells Through AMPK-Dependent and -Independent Mechanisms. J Clin Endocrinol Metab 2019; 104:3501-3513. [PMID: 30860580 DOI: 10.1210/jc.2019-00056] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/06/2019] [Indexed: 12/17/2022]
Abstract
CONTEXT Pituitary neuroendocrine tumors (PitNETs) are a commonly underestimated pathology in terms of incidence and associated morbimortality. Currently, an appreciable subset of patients are resistant or poorly responsive to the main current medical treatments [i.e., synthetic somatostatin analogs (SSAs) and dopamine agonists]. Thus, development and optimization of novel and available medical therapies is necessary. Biguanides (metformin, buformin, and phenformin) are antidiabetic drugs that exert antitumoral actions in several tumor types, but their pharmacological effects on PitNETs are poorly known. OBJECTIVE We aimed to explore the direct effects of biguanides on key functions (cell viability, hormone release, apoptosis, and signaling pathways) in primary cell cultures from human PitNETs and cell lines. Additionally, we evaluated the effect of combined metformin with SSAs on cell viability and hormone secretion. DESIGN A total of 13 corticotropinomas, 13 somatotropinomas, 13 nonfunctioning PitNETs, 3 prolactinomas, and 2 tumoral pituitary cell lines (AtT-20 and GH3) were used to evaluate the direct effects of biguanides on cell viability, hormone release, apoptosis, and signaling pathways. RESULTS Biguanides reduced cell viability in all PitNETs and cell lines (with phenformin being the most effective biguanide) and increased apoptosis in somatotropinomas. Moreover, buformin and phenformin, but not metformin, reduced hormone secretion in a cell type-specific manner. Combination metformin/SSA therapy did not increase SSA monotherapy effectiveness. Effects of biguanides on PitNETs could involve the modulation of AMP-activated protein kinase-dependent ([Ca2+]i, PI3K/Akt) and independent (MAPK) mechanisms. CONCLUSION Altogether, our data unveil clear antitumoral effects of biguanides on PitNET cells, opening avenues to explore their potential as drugs to treat these pathologies.
Collapse
Affiliation(s)
- Mari C Vázquez-Borrego
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| | - Antonio C Fuentes-Fayos
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| | - Aura D Herrera-Martínez
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Endocrinology and Nutrition, IMIBIC, Reina Sofia University Hospital, Cordoba, Spain
| | - Fernando L-López
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| | - Alejandro Ibáñez-Costa
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| | - Paloma Moreno-Moreno
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Endocrinology and Nutrition, IMIBIC, Reina Sofia University Hospital, Cordoba, Spain
| | - María R Alhambra-Expósito
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Endocrinology and Nutrition, IMIBIC, Reina Sofia University Hospital, Cordoba, Spain
| | - Ana Barrera-Martín
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Endocrinology and Nutrition, IMIBIC, Reina Sofia University Hospital, Cordoba, Spain
| | - Cristóbal Blanco-Acevedo
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Neurosurgery, Reina Sofia University Hospital, Cordoba, Spain
| | - Elena Dios
- Metabolism and Nutrition Unit, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | - Eva Venegas-Moreno
- Metabolism and Nutrition Unit, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | - Juan Solivera
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Neurosurgery, Reina Sofia University Hospital, Cordoba, Spain
| | - Manuel D Gahete
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| | - Alfonso Soto-Moreno
- Metabolism and Nutrition Unit, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | - María A Gálvez-Moreno
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Endocrinology and Nutrition, IMIBIC, Reina Sofia University Hospital, Cordoba, Spain
| | - Justo P Castaño
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| | - Raúl M Luque
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| |
Collapse
|
263
|
Peng X, Tu Y, Fu S, Xia Y, Ma C, Yang Y, Wu H, Liu Y, You P. 14-Deoxycoleon U-induced endoplasmic reticulum stress-mediated apoptosis, autophagy, and cell cycle arrest in lung adenocarcinoma. Onco Targets Ther 2019; 12:5955-5965. [PMID: 31534345 PMCID: PMC6681072 DOI: 10.2147/ott.s211933] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/13/2019] [Indexed: 11/23/2022] Open
Abstract
Objective 14-Deoxycoleon U is a natural abietane-type diterpene and exerts an inhibitory effect on tumor cells proliferation, which suggests that 14-Deoxycoleon U may be a potent anti-cancerous lead compound for lung cancer treatment. This study was to evaluate potential of 14-Deoxycoleon U to treat lung adenocarcinoma in vitro and in vivo. Methods In the present study, the cell viability and apoptosis morphology of 14-Deoxycoleon U-treated A549 and LLC cells were explored using cell counting kit-8 assay and Hoechst 33258 staining. Then, the protein expressions about apoptosis, endoplasmic reticulum (ER) stress, autophagy and cell cycle were measured using Western blot. The autophagosome formation of 14-Deoxycoleon U-treated A549 cells was visualized using a confocal microscopy. LLC lung adenocarcinoma model was established. Results The results indicated that 14-Deoxycoleon U significantly inhibited A549 and LLC cell proliferation in a dose-dependent manner via caspase-dependent apoptosis. Furthermore, apoptosis of both cells was mediated by 14-Deoxycoleon U-induced ER stress. In addition, 14-Deoxycoleon U-induced A549 and LLC cell autophagy, thus promoting their death. Moreover, 14-Deoxycoleon U-induced cell cycle arrest in both cells via inhibition of cyclin D3, cyclin-dependent kinase 6, CDC2 and up-regulation of p21. In vivo results showed that administration of 14-Deoxycoleon U significantly suppressed LLC growth and adverse effects of 14-Deoxycoleon U on organs might be lower than of adriamycin. Conclusion Overall, our results demonstrated that 14-Deoxycoleon U represses in vitro and in vivo growth of lung adenocarcinoma through ER stress-mediated apoptosis accompanied by autophagy and cell cycle arrest.
Collapse
Affiliation(s)
- Xiaozhi Peng
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, People's Republic of China
| | - Yijun Tu
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, People's Republic of China
| | - San Fu
- State Key Laboratory of Natural Medicines, Research Department of Pharmacognosy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Yu Xia
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, People's Republic of China
| | - Chaozhi Ma
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, People's Republic of China
| | - Yanfang Yang
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, People's Republic of China
| | - Hezhen Wu
- Department of Pharmacy, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, People's Republic of China
| | - Yanwen Liu
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, People's Republic of China
| | - Pengtao You
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, People's Republic of China
| |
Collapse
|
264
|
MAPK Pathway Inhibitors Attenuated Hydrogen Peroxide Induced Damage in Neural Cells. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5962014. [PMID: 31355271 PMCID: PMC6637717 DOI: 10.1155/2019/5962014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/03/2019] [Indexed: 12/14/2022]
Abstract
Background Oxidative stress due to reactive oxygen species plays a central role in pathophysiology of neurodegenerative diseases. Inhibition of mitogen-activated protein kinase (MAPK) cascades attenuates the oxidative induced cell stress and behaves as potential neuroprotection agent. Materials and Methods In this study, we evaluate hydrogen peroxide induced neural cell stress and determine how different MAPK inhibitors restore the cell damage. Results The results indicated that oxidative stress induced by neural cell damage commonly exists, and MAPK inhibitors partially and selectively attenuated the cell damage by reducing ROS production and cell apoptosis. The cultured neurons are more susceptible to hydrogen peroxide than subculture cells. Conclusion We conclude that the essential role of different MAPK inhibitors is to attenuate the hydrogen peroxide induced neuronal cell damage. Those data broaden the implication between individual neural cells and different MAPK inhibitors and give clues for oxidative stress induced neural diseases.
Collapse
|
265
|
Caspases interplay with kinases and phosphatases to determine cell fate. Eur J Pharmacol 2019; 855:20-29. [DOI: 10.1016/j.ejphar.2019.04.048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 04/05/2019] [Accepted: 04/29/2019] [Indexed: 12/15/2022]
|
266
|
You MS, Wang WP, Wang JY, Jiang YJ, Chi YH. Sun1 Mediates Interkinetic Nuclear Migration and Notch Signaling in the Neurogenesis of Zebrafish. Stem Cells Dev 2019; 28:1116-1127. [PMID: 31140357 DOI: 10.1089/scd.2019.0063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Interkinetic nuclear migration (INM) is a process by which nuclei oscillate between the basal and apical surfaces of epithelial cells in coordination with the cell cycle. The cytoskeletal machinery including microtubules and actin has been reported to drive apical INM; however, the role of nuclear proteins in this process has yet to be fully elucidated. Here, we investigated the function of a SUN-domain protein, Sun1, in zebrafish. We found that zebrafish sun1 is highly expressed in the ventricular zone of the brain. Knocking down sun1 with antisense morpholino oligonucleotides reduced the abundance of nestin- and gfap-expressing neural stem cells and progenitor cells. The live-cell imaging results showed that sun1 morphant cells migrated toward the basal side during the S phase but failed to migrate apically during the G2 phase. On the contrary, the passive stochastic movement during the G2 phase was unaffected. Furthermore, down regulation of sun1 was shown to reduce the expression of genes associated with the Notch pathway, whereas the expression of genes in the Wnt pathway was less perturbed. Findings from this research suggest that the Sun1-mediated nucleo-cytoskeletal interaction contributes to apical nuclear migration, and may thus affect exposure to Notch signal, thereby altering the composition of the progenitor pool in the embryonic neurogenesis of zebrafish.
Collapse
Affiliation(s)
- May-Su You
- 1Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Wan-Ping Wang
- 2Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Jing-Ya Wang
- 2Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Yun-Jin Jiang
- 1Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Ya-Hui Chi
- 2Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan.,3Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| |
Collapse
|
267
|
Lee J, Jang HJ, Chun H, Pham TH, Bak Y, Shin JW, Jin H, Kim YI, Ryu HW, Oh SR, Yoon DY. Calotropis gigantea extract induces apoptosis through extrinsic/intrinsic pathways and reactive oxygen species generation in A549 and NCI-H1299 non-small cell lung cancer cells. Altern Ther Health Med 2019; 19:134. [PMID: 31215445 PMCID: PMC6582476 DOI: 10.1186/s12906-019-2561-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 06/11/2019] [Indexed: 12/25/2022]
Abstract
Background Calotropis gigantea (CG) is a tall and waxy flower that is used as a traditional remedy for fever, indigestion, rheumatism, leprosy, and leukoderma. However, the precise mechanisms of its anticancer effects have not yet been examined in human non-small cell lung cancer (NSCLC) cells. In this study, we investigated whether CG extract exerted an apoptotic effect in A549 and NCI-H1299 NSCLC cells. Methods The ethanol extract of CG was prepared, and its apoptotic effects on A549 and NCI-H1299 NSCLC cells were assessed by using the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxy methoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay, annexin V-fluorescein isothiocyanate/propidium iodide (PI) staining, cell cycle analysis, real-time polymerase chain reaction (RT-PCR), western blotting, JC-1 staining, and ROS detection assay. Results The CG extract induced apoptosis through the stimulation of intrinsic and extrinsic signaling pathways in A549 and NCI-H1299 lung cancer cells. Cell cycle arrest was induced by the CG extract in both cell lines. Reactive oxygen species (ROS), which can induce cell death, were also generated in the CG-treated A549 and NCI-H1299 cells. Conclusions These data confirmed that CG caused apoptosis through the activation of extrinsic and intrinsic pathways, cell cycle arrest, and ROS generation in A549 and NCI-H1299 lung cancer cells. Thus, CG can be suggested as a potential agent for lung cancer therapy. Electronic supplementary material The online version of this article (10.1186/s12906-019-2561-1) contains supplementary material, which is available to authorized users.
Collapse
|
268
|
Sammons RM, Ghose R, Tsai KY, Dalby KN. Targeting ERK beyond the boundaries of the kinase active site in melanoma. Mol Carcinog 2019; 58:1551-1570. [PMID: 31190430 DOI: 10.1002/mc.23047] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/30/2019] [Accepted: 05/10/2019] [Indexed: 12/14/2022]
Abstract
Extracellular signal-regulated kinase 1/2 (ERK1/2) constitute a point of convergence for complex signaling events that regulate essential cellular processes, including proliferation and survival. As such, dysregulation of the ERK signaling pathway is prevalent in many cancers. In the case of BRAF-V600E mutant melanoma, ERK inhibition has emerged as a viable clinical approach to abrogate signaling through the ERK pathway, even in cases where MEK and Raf inhibitor treatments fail to induce tumor regression due to resistance mechanisms. Several ERK inhibitors that target the active site of ERK have reached clinical trials, however, many critical ERK interactions occur at other potentially druggable sites on the protein. Here we discuss the role of ERK signaling in cell fate, in driving melanoma, and in resistance mechanisms to current BRAF-V600E melanoma treatments. We explore targeting ERK via a distinct site of protein-protein interaction, known as the D-recruitment site (DRS), as an alternative or supplementary mode of ERK pathway inhibition in BRAF-V600E melanoma. Targeting the DRS with inhibitors in melanoma has the potential to not only disrupt the catalytic apparatus of ERK but also its noncatalytic functions, which have significant impacts on spatiotemporal signaling dynamics and cell fate.
Collapse
Affiliation(s)
- Rachel M Sammons
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas.,Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Ranajeet Ghose
- Department of Chemistry and Biochemistry, The City College of New York, New York, New York
| | - Kenneth Y Tsai
- Departments of Anatomic Pathology and Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kevin N Dalby
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin, Austin, Texas.,Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
269
|
Yang HZ, Zhang J, Zeng J, Liu S, Zhou F, Zhang F, Giampieri F, Cianciosi D, Forbes-Hernandez TY, Ansary J, Gil E, Chen R, Battino M. Resveratrol inhibits the proliferation of melanoma cells by modulating cell cycle. Int J Food Sci Nutr 2019; 71:84-93. [PMID: 31154861 DOI: 10.1080/09637486.2019.1614541] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The aim of this study was to evaluate the inhibitory effects of resveratrol (RSV) in A375 and A431 melanoma cells, by assessing cell viability (CCK-8 assay), apoptosis through flow cytometer and cell morphology, cell cycle assay by flow cytometer and western blot (Cyclin D1, Rac1 and PCDH9). Our results demonstrated that RSV strongly inhibited A375 cells proliferation, by decreasing cell viability, promoting apoptosis and arresting cell cycle. Besides, to clarify the main factor - duration or concentration of RSV, the double variance analysis of independent factors was operated after Bartlett's test for homogeneity by R project. According to the outcomes obtained from statistical analyses, Cyclin D1 and PCDH9 were strongly affected by RSV duration while Rac1 was not influenced. In conclusion, RSV can inhibit A375 proliferation and trigger apoptosis by influencing cell cycle proteins; for these effects, treatment duration of RSV played more important role than concentration.
Collapse
Affiliation(s)
- Hui-Zhi Yang
- Department of Dermatology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jiaojiao Zhang
- Department of Clinical Sciences, Faculty of Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Jie Zeng
- Department of Dermatology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shengbo Liu
- Guangdong Medical University, Zhanjiang, China
| | - Fei Zhou
- Department of Dermatology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Fang Zhang
- Department of Dermatology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Francesca Giampieri
- Department of Clinical Sciences, Faculty of Medicine, Università Politecnica delle Marche, Ancona, Italy.,Department of Biochemistry, Genetics and Immunology, Faculty of Biology, Nutrition and Food Science Group, University of Vigo, Vigo, Spain
| | - Danila Cianciosi
- Department of Clinical Sciences, Faculty of Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Tamara Y Forbes-Hernandez
- Department of Biochemistry, Genetics and Immunology, Faculty of Biology, Nutrition and Food Science Group, University of Vigo, Vigo, Spain
| | - Johura Ansary
- Department of Clinical Sciences, Faculty of Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Emilio Gil
- Department of Biochemistry, Genetics and Immunology, Faculty of Biology, Nutrition and Food Science Group, University of Vigo, Vigo, Spain
| | - RongYi Chen
- Department of Dermatology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Maurizio Battino
- Department of Clinical Sciences, Faculty of Medicine, Università Politecnica delle Marche, Ancona, Italy.,Department of Analytical and Food Chemistry, Nutrition and Food Science Group, CITACA, CACTI, University of Vigo - Vigo Campus, Vigo, Spain
| |
Collapse
|
270
|
Ghosh S, Pal A, Ray M. Methylglyoxal in combination with 5-Fluorouracil elicits improved chemosensitivity in breast cancer through apoptosis and cell cycle inhibition. Biomed Pharmacother 2019; 114:108855. [DOI: 10.1016/j.biopha.2019.108855] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 03/27/2019] [Accepted: 04/02/2019] [Indexed: 10/27/2022] Open
|
271
|
Interleukin-17F Has Anti-Tumor Effects in Oral Tongue Cancer. Cancers (Basel) 2019; 11:cancers11050650. [PMID: 31083515 PMCID: PMC6562684 DOI: 10.3390/cancers11050650] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 04/30/2019] [Accepted: 05/08/2019] [Indexed: 12/15/2022] Open
Abstract
We recently showed that extracellular interleukin-17F (IL-17F) correlates with better disease-specific survival in oral tongue squamous cell carcinoma (OTSCC) patients. However, the underlying mechanisms of such effect remain obscure. Here, we used qRT-PCR to assess the expression of IL-17F and its receptors (IL-17RA and IL-17RC) in two OTSCC cell lines (HSC-3 and SCC-25) and in normal human oral keratinocytes (HOKs). IL-17F effects on cancer cell proliferation, migration, and invasion were studied using a live-imaging IncuCyte system, and a Caspase-3/7 reagent was used for testing apoptosis. 3D tumor spheroids were utilized to assess the impact of IL-17F on invasion with or without cancer-associated fibroblasts (CAFs). Tube-formation assays were used to examine the effects of IL-17F on angiogenesis using human umbilical vein endothelial cells (HUVEC). OTSCC cells express low levels of IL-17F, IL-17RA, and IL-17RC mRNA compared with HOKs. IL-17F inhibited cell proliferation and random migration of highly invasive HSC-3 cells. CAFs promoted OTSCC invasion in tumor spheroids, whereas IL-17F eliminated such effect. IL-17F suppressed HUVEC tube formation in a dose-dependent manner. Collectively, we suggest that IL-17F counteracts the pro-tumorigenic activity in OTSCC. Due to its downregulation in tumor cells and inhibitory activity in in vitro cancer models, targeting IL-17F or its regulatory pathways could lead to promising immunotherapeutic strategies against OTSCC.
Collapse
|
272
|
Enhanced cytotoxic and genotoxic effects of gadolinium-doped ZnO nanoparticles on irradiated lung cancer cells at megavoltage radiation energies. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 103:109739. [PMID: 31349426 DOI: 10.1016/j.msec.2019.109739] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 01/10/2023]
Abstract
The purpose of this study was to investigate the radiation dose enhancement effects of gadolinium-doped zinc oxide nanoparticles (Gd-doped ZnO NPs) under the megavoltage (MV) X-ray irradiation. ZnO NPs have preferred photocatalytic properties under UV light for cancer killing. UV light has limited applications in cancer treatment and it is necessary to use X-ray photons with MV energies. In order to increase the absorption of radiation and also to enhance the imaging visualization capabilities of ZnO NPs, gadolinium (Gd) as a high atomic number element was selected for doping into the structure of ZnO NPs. Gd-doped ZnO NPs were synthesized by a chemical precipitation method and characterized by transmission electron microscopy, powder X-ray diffraction, ultraviolet-visible spectroscopy, and energy-dispersive X-ray techniques. Cellular uptake was assessed by TEM and inductively coupled plasma mass spectrometry. NPs cytotoxicity was analyzed by MTT assay and radiation dose enhancement was measured by clonogenic survival assay. Apoptosis induction, cell cycle progression, micronucleus formation and expression of DNA double-strand break repair genes of XRCC2 and XRCC4 were determined by flow cytometry, micronucleus assay, and quantitative real-time polymerase chain reaction. CT and MR imaging were used to analyze the image visualization capabilities of NPs. NPs characterization showed that highly pure crystalline Gd-doped ZnO NPs with a narrow size distribution and grain size of 9 nm were synthesized. Gd-doped ZnO NPs were distributed in the cells and showed dose-dependent toxicity. Combination of Gd-doped ZnO NPs with 6 MV X-rays induced dose-dependent radiosensitivity with sensitizer enhancement ratios (SER) of 1.47 and 1.61 for 10 and 20 μg/mL NPs concentrations. Cancer cells blocked in G1, apoptosis rates, and micronuclei formation was enhanced and inversely, the DNA repair efficiency was impaired by down regulation of the mRNA levels of XRCC2 and XRCC4 genes. Gd-doped ZnO NPs enhanced the contrasts of CT and MR images of cancer cells. Overall, the results of this study provide detailed biological insights on the dose enhancement of Gd-doped ZnO NPs at MV radiations, which would contribute to the further development of this potent theranostic platform for clinical applications.
Collapse
|
273
|
Pereira SS, Monteiro MP, Antonini SR, Pignatelli D. Apoptosis regulation in adrenocortical carcinoma. Endocr Connect 2019; 8:R91-R104. [PMID: 30978697 PMCID: PMC6510712 DOI: 10.1530/ec-19-0114] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 04/11/2019] [Indexed: 12/31/2022]
Abstract
Apoptosis evading is a hallmark of cancer. Tumor cells are characterized by having an impaired apoptosis signaling, a fact that deregulates the balance between cell death and survival, leading to tumor development, invasion and resistance to treatment. In general, patients with adrenocortical carcinomas (ACC) have an extremely bad prognosis, which is related to disease progression and significant resistance to treatments. In this report, we performed an integrative review about the disruption of apoptosis in ACC that may underlie the characteristic poor prognosis in these patients. Although the apoptosis has been scarcely studied in ACC, the majority of the deregulation phenomena already described are anti-apoptotic. Most importantly, in a near future, targeting apoptosis modulation in ACC patients may become a promising therapeutic.
Collapse
Affiliation(s)
- Sofia S Pereira
- Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Endocrine, Cardiovascular & Metabolic Research, Department of Anatomy, Multidisciplinary Unit for Biomedical Research (UMIB), Instituto de Ciências Biomédicas Abel Salazar, University of Porto (ICBAS/UP), Porto, Portugal
| | - Mariana P Monteiro
- Endocrine, Cardiovascular & Metabolic Research, Department of Anatomy, Multidisciplinary Unit for Biomedical Research (UMIB), Instituto de Ciências Biomédicas Abel Salazar, University of Porto (ICBAS/UP), Porto, Portugal
| | - Sonir R Antonini
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Duarte Pignatelli
- Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Department of Endocrinology, Hospital S. João, Porto, Portugal
- Correspondence should be addressed to D Pignatelli:
| |
Collapse
|
274
|
Santhakumaran I, Shanuja SK, Narayanaswamy R, Gnanamani A. Asperyellone prevents HDF cells from UVB irradiation damages: An elaborated study. J Cell Biochem 2019; 120:7560-7572. [PMID: 30548661 DOI: 10.1002/jcb.28030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/15/2018] [Indexed: 01/24/2023]
Abstract
The current study explores the photo-protective effect of asperyellone (AY) (a fungal secondary metabolite), assessed under in vitro condition using human dermal fibroblast cell line. AY was isolated from Aspergillus sp. during the resting phase and purified. The initial cytocompatibility assessment on concentrations of AY and the duration of exposure of UVB irradiations were studied respectively. N-acetyl-L-cysteine (NAC) was used as positive control. Cells were then pretreated with optimized concentration of AY (2.0 μM) and NAC (1 mM) for 1 hour and then UVB irradiated (30 mJ/cm 2 ) for the period of 10 minutes. Results revealed that reactive oxygen species generated upon UVB irradiation found scavenged by the AY pretreatment at a significant level. Furthermore, an appreciable reduction in apoptotic cell count and DNA damages support the scavenging effect of AY. Assessments on the expression of enzymatic and nonenzymatic antioxidants evidently prove the protective role of AY. The reduced expression levels of inflammatory markers (TNF-α and COX-2), collagen degraders (MMP 2 and MMP 9), apoptotic protein expressions (Bax and Bcl-2), and cell-cycle arrest analyses substantiate the photo-protective effect of AY similar to NAC (positive control). Thus, the observations made in the current study indicate the possible role of AY as a photo-protective agent.
Collapse
Affiliation(s)
| | | | - Radhakrishnan Narayanaswamy
- Bio Waste Management Laboratory, Vel Tech Technology Incubator, Veltech Dr. RR & Dr. SR University, Chennai, Tamil Nadu, India
| | | |
Collapse
|
275
|
Çevik D, Kan Y, Kırmızıbekmez H. Mechanisms of action of cytotoxic phenolic compounds from Glycyrrhiza iconica roots. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 58:152872. [PMID: 30826527 DOI: 10.1016/j.phymed.2019.152872] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/01/2019] [Accepted: 02/19/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Glycyrrhiza (licorice) species are rich in bioactive secondary metabolites and their roots are used traditionally for the treatment of several diseases. In recent years, secondary metabolites of licorice are gaining popularity, especially due to their significant cytotoxic and antitumor effects. However, Glycyrrhiza iconica, an endemic species to Turkey, was not investigated in terms of its anticancer secondary metabolites previously. PURPOSE This study aimed to isolate the cytotoxic compounds from G. iconica through bioactivity-guided fractionation and to elucidate mechanisms of action of the most potent compounds. METHODS Total MeOH extract and CHCl3, EtOAc, n-buOH and rH2O subextracts were prepared from G. iconica roots. Sequential chromatographic techniques were conducted for the isolation studies. The chemical structures of the isolates were established based on NMR and HR-MS analysis. Sulforhodamine B assay was used to evaluate the cytotoxic activity of extracts, main fractions as well as isolates against hepatocellular (Huh7), breast (MCF7) and colorectal (HCT116) cancer cell lines. The mechanisms underlying the cytotoxicity of the most active compounds in Huh7 cells were elucidated by using Hoechst staining, Fluorescence-activated cell sorting and Western blot assays. RESULTS A new dihydrochalcone, iconichalcone (1) along with 15 known phenolic compounds were isolated from the active CHCl3, EtOAc and n-buOH subextracts. Compounds 2-5, 7-16 were found to be responsible for the in vitro cytotoxic activity of G. iconica against all tested cancer cell lines with IC50 values ranging from 2.4 to 33 µM. Amongst these compounds, licoricidin (10), dehydroglyasperin C (12), iconisoflaven (13) and 1-methoxyficifolinol (15) were found to be the most active compounds according to SRB and real time bioactivity assays and submitted to further mechanistic investigations in Huh7 cells. Compounds 10, 12, 13 and 15 caused accumulation of cells in different phases of cell cycle. Moreover, 10, 12, 13 and 15 induced apoptosis through caspase activation. Besides, 12 showed activation of p53 expression and thus G2/M arrest as well as a condensed nuclei, established very promising results. CONCLUSION The results demonstrated that the aforementioned compounds, particularly 12 could be potential lead molecules for anticancer drug development that deserve further in vivo and clinical investigations.
Collapse
Affiliation(s)
- Dicle Çevik
- Department of Pharmacognosy, Faculty of Pharmacy, Yeditepe University, Kayışdağı, İstanbul TR-34755, Turkey
| | - Yüksel Kan
- Department of Medicinal Plants, Faculty of Agriculture, Selçuk University, Konya TR-42070, Turkey
| | - Hasan Kırmızıbekmez
- Department of Pharmacognosy, Faculty of Pharmacy, Yeditepe University, Kayışdağı, İstanbul TR-34755, Turkey.
| |
Collapse
|
276
|
Periplanetasin-4, a novel antimicrobial peptide from the cockroach, inhibits communications between mitochondria and vacuoles. Biochem J 2019; 476:1267-1284. [DOI: 10.1042/bcj20180933] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 12/16/2022]
Abstract
Abstract
Communications between various organelle–organelles play an essential role in cell survival. The cross-talk between mitochondria and vacuoles comes up with the vital roles of the intercompartmental process. In this study, we found a couple of cell death features, membrane damage, and apoptosis using antimicrobial peptide from American Cockroach. Periplanetasin-4 (LRHKVYGYCVLGP-NH2) is a 13-mer peptide derived from Periplaneta americana and exhibits phosphatidylserine exposure and caspase activation without DNA fragmentation. Apoptotic features without DNA damage provide evidence that this peptide did not interact with DNA directly and exhibited dysfunction of mitochondria and vacuoles. Superoxide radicals were generated from mitochondria and converted to hydrogen peroxide. Despite the enhancement of catalase and total glutathione contents, oxidative damage disrupted intracellular contents. Periplanetasin-4 induced cell death associated with the production of superoxide radicals, calcium uptake in mitochondria and disorder of vacuoles, such as increased permeability and alkalization. While calcium movement from vacuoles to the mitochondria occurred, the cross-talk with these organelles proceeded and the inherent functionality was impaired. To sum up, periplanetasin-4 stimulates superoxide signal along with undermining the mitochondrial functions and interfering in communication with vacuoles.
Collapse
|
277
|
Kishimoto Y, Yamashita M, Wei A, Toya Y, Ye S, Kendziorski C, Welham NV. Reversal of Vocal Fold Mucosal Fibrosis Using siRNA against the Collagen-Specific Chaperone Serpinh1. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 16:616-625. [PMID: 31100613 PMCID: PMC6520554 DOI: 10.1016/j.omtn.2019.04.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/12/2019] [Accepted: 04/13/2019] [Indexed: 12/11/2022]
Abstract
Vocal fold (VF) mucosal fibrosis results in substantial voice impairment and is recalcitrant to current treatments. To reverse this chronic disorder, anti-fibrotic therapies should target the molecular pathology of aberrant collagen accumulation in the extracellular matrix. We investigated the therapeutic potential of siRNA against Serpinh1, a collagen-specific chaperone that enables cotranslational folding and assembly of procollagens in the endoplasmic reticulum. We implemented a previously validated siRNA construct, conducted transfection experiments using in vitro and in vivo rat models, and measured knockdown efficiency, dose responses, delivery strategies, and therapeutic outcomes. Liposome-mediated delivery of Serpinh1-siRNA downregulated collagen production in naive and scar VF fibroblasts as well as naive VF mucosa; moreover, sustained Serpinh1 knockdown in fibrotic VF mucosa reversed scar-associated collagen accumulation within 4 weeks. Analysis of therapeutic effects at the transcriptome level showed evidence of cell cycle upregulation, catabolism, matrix disassembly, and morphogenesis. These findings indicate that Serpinh1-siRNA holds potential as a molecular therapy for chronic VF mucosal fibrosis.
Collapse
Affiliation(s)
- Yo Kishimoto
- Division of Otolaryngology, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Masaru Yamashita
- Division of Otolaryngology, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Alice Wei
- Division of Otolaryngology, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Yutaka Toya
- Division of Otolaryngology, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Shuyun Ye
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, USA
| | - Christina Kendziorski
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, USA
| | - Nathan V Welham
- Division of Otolaryngology, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA.
| |
Collapse
|
278
|
Combination of clotam and vincristine enhances anti-proliferative effect in medulloblastoma cells. Gene 2019; 705:67-76. [PMID: 30991098 DOI: 10.1016/j.gene.2019.04.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/28/2019] [Accepted: 04/12/2019] [Indexed: 12/20/2022]
Abstract
Medulloblastoma (MB) is characterized by highly invasive embryonal neuro-epithelial tumors that metastasize via cerebrospinal fluid. MB is difficult to treat and the chemotherapy is associated with significant toxicities and potential long-term disabilities. Previously, we showed that small molecule, clotam (tolfenamic acid: TA) inhibited MB cell proliferation and tumor growth in mice by targeting, survivin. Overexpression of survivin is associated with aggressiveness and poor prognosis in several cancers, including MB. The aim of this study was to test combination treatment involving Vincristine® (VCR), a standard chemotherapeutic drug for MB and TA against MB cells. DAOY and D283 MB cells were treated with 10 μg/mL TA or VCR (DAOY: 2 ng/mL; D283: 1 ng/mL) or combination (TA + VCR). These optimized doses were lower than individual IC50 values. The effect of single or combination treatment on cell viability (CellTiterGlo kit), Combination Index (Chou-Talalay method based on median-drug effect analysis), activation of apoptosis and cell cycle modulation (by flow cytometry using Annexin V and propidium iodide respectively) and the expression of associated markers including survivin (Western immunoblot) were determined. Combination Index showed moderate synergistic cytotoxic effect in both cells. When compared to individual agents, the combination of TA and VCR increased MB cell growth inhibition, induced apoptosis and caused cell cycle (G2/M phase) arrest. Survivin expression was also decreased by the combination treatment. TA is effective for inducing the anti-proliferative response of VCR in MB cells. MB has four distinct genetic/molecular subgroups. Experiments were conducted with MB cells representing two subgroups (DAOY: SHH group; D283: group 4/3). TA-induced inhibition of survivin expression potentially destabilizes mitotic microtubule assembly, sensitizing MB cells and enhancing the efficacy of VCR.
Collapse
|
279
|
Abstract
While spontaneous tumours may occasionally develop in inbred and isogenic strains of Xenopus laevis, the South African clawed toad, they are extremely rare in natural and laboratory populations. Only two amphibian neoplasms, the renal adenocarcinoma of Rana pipiens and the lymphosarcoma of Xenopus laevis, have been extensively explored. Amphibians are resistant to the development of neo-plasia, even following exposure to “direct-acting” chemical carcinogens such as N-methyl- N-nitrosourea, that are highly lymphotoxic, thus diminishing immune reactivity. Regenerative capacity in adults, and a dramatic metamorphosis which remodels much of the larval body to produce the adult form, are unique to amphibian vertebrates, and the control mechanisms involved may protect against cancer. For example, naturally rising corticosteroid titres during metamorphosis will impair some T-cell functions, and the removal of T-regulatory (suppressor) functions inhibits the induction of altered-self tolerance. Altered-self tolerance is not as effectively induced in adult Xenopus laevis as in mammals, so cancer cells with new antigenicity are more likely be rejected in amphibians. Amphibian immunocytes tend to undergo apoptosis readily in vitro, and, unlike mammalian immunocytes, undergo apoptosis without entering the cell cycle. Cells not in the cell cycle that die from nuclear damage (apoptosis), will have no opportunity to express genetic instability leading to cell transformation. We suggest that all these factors, rather than any one of them, may reduce susceptibility to cancer in amphibians.
Collapse
Affiliation(s)
| | - Richard H. Clothier
- School of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | | |
Collapse
|
280
|
Jang CH, Moon N, Oh J, Kim JS. Luteolin Shifts Oxaliplatin-Induced Cell Cycle Arrest at G₀/G₁ to Apoptosis in HCT116 Human Colorectal Carcinoma Cells. Nutrients 2019; 11:nu11040770. [PMID: 30987009 PMCID: PMC6521147 DOI: 10.3390/nu11040770] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 03/24/2019] [Accepted: 03/29/2019] [Indexed: 12/13/2022] Open
Abstract
Certain antioxidative flavonoids are known to activate nuclear factor E2-related factor 2 (Nrf2), a transcription factor that regulates cellular antioxidants and detoxifying response and is reportedly highly activated in many types of cancers. Few studies on the potential undesired effects of flavonoid intake during chemotherapy have been conducted, yet Nrf2 activators could favor cancer cell survival by attenuating chemotherapeutic efficiency. This study aimed to examine if luteolin, an Nrf2 activator, hinders chemotherapeutic activity of oxaliplatin, a potent anticancer agent for colorectal cancer, in HCT116 cells. Luteolin treatment strongly increased the transcriptional activity of the antioxidant response element in HCT116 cells and induced the protein expression of heme oxygenase-1, which were indicative of its Nrf2-inducing potential. Intriguingly, 25 μM luteolin reduced cell viability through apoptotic induction, which was intensified in p53-expressing cells while 1 μM oxaliplatin caused cell cycle arrest at G0/G1-phase via the p53/p21-dependent mechanism. Moreover, luteolin treatment was found to reduce oxaliplatin-treated p53-null cell viability and colony counts further, thereby demonstrating an additional effect of luteolin in the killing of human colorectal tumor HCT116 cells not expressing functional p53 protein. The findings suggest that luteolin can induce p53-mediated apoptosis regardless of oxaliplatin treatment and may eliminate oxaliplatin-resistant p53-null colorectal cells.
Collapse
Affiliation(s)
- Chan Ho Jang
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Korea.
| | - Nayoung Moon
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Korea.
| | - Jisun Oh
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Korea.
| | - Jong-Sang Kim
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Korea.
| |
Collapse
|
281
|
Polysaccharides as potential anticancer agents—A review of their progress. Carbohydr Polym 2019; 210:412-428. [PMID: 30732778 DOI: 10.1016/j.carbpol.2019.01.064] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/21/2019] [Accepted: 01/21/2019] [Indexed: 12/17/2022]
|
282
|
Li H, Saucedo-Cuevas L, Yuan L, Ross D, Johansen A, Sands D, Stanley V, Guemez-Gamboa A, Gregor A, Evans T, Chen S, Tan L, Molina H, Sheets N, Shiryaev SA, Terskikh AV, Gladfelter AS, Shresta S, Xu Z, Gleeson JG. Zika Virus Protease Cleavage of Host Protein Septin-2 Mediates Mitotic Defects in Neural Progenitors. Neuron 2019; 101:1089-1098.e4. [PMID: 30713029 PMCID: PMC6690588 DOI: 10.1016/j.neuron.2019.01.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/17/2018] [Accepted: 12/31/2018] [Indexed: 01/06/2023]
Abstract
Zika virus (ZIKV) targets neural progenitor cells in the brain, attenuates cell proliferation, and leads to cell death. Here, we describe a role for the ZIKV protease NS2B-NS3 heterodimer in mediating neurotoxicity through cleavage of a host protein required for neurogenesis. Similar to ZIKV infection, NS2B-NS3 expression led to cytokinesis defects and cell death in a protease activity-dependent fashion. Among binding partners, NS2B-NS3 cleaved Septin-2, a cytoskeletal factor involved in cytokinesis. Cleavage of Septin-2 occurred at residue 306 and forced expression of a non-cleavable Septin-2 restored cytokinesis, suggesting a direct mechanism of ZIKV-induced neural toxicity. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Hongda Li
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Laura Saucedo-Cuevas
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Ling Yuan
- Institute of Genetics & Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Danica Ross
- Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Anide Johansen
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Daniel Sands
- Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Valentina Stanley
- Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Alicia Guemez-Gamboa
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Anne Gregor
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lei Tan
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Henrik Molina
- Proteomics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Nicholas Sheets
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Sergey A Shiryaev
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Alexey V Terskikh
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Amy S Gladfelter
- Department of Biology, Howard Hughes Medical Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sujan Shresta
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Zhiheng Xu
- Institute of Genetics & Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Joseph G Gleeson
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA.
| |
Collapse
|
283
|
Li H, Saucedo-Cuevas L, Yuan L, Ross D, Johansen A, Sands D, Stanley V, Guemez-Gamboa A, Gregor A, Evans T, Chen S, Tan L, Molina H, Sheets N, Shiryaev SA, Terskikh AV, Gladfelter AS, Shresta S, Xu Z, Gleeson JG. Zika Virus Protease Cleavage of Host Protein Septin-2 Mediates Mitotic Defects in Neural Progenitors. Neuron 2019; 101:1089-1098.e4. [PMID: 30713029 PMCID: PMC6690588 DOI: 10.1016/j.neuron.2019.01.010#mmc4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/17/2018] [Accepted: 12/31/2018] [Indexed: 10/03/2024]
Abstract
Zika virus (ZIKV) targets neural progenitor cells in the brain, attenuates cell proliferation, and leads to cell death. Here, we describe a role for the ZIKV protease NS2B-NS3 heterodimer in mediating neurotoxicity through cleavage of a host protein required for neurogenesis. Similar to ZIKV infection, NS2B-NS3 expression led to cytokinesis defects and cell death in a protease activity-dependent fashion. Among binding partners, NS2B-NS3 cleaved Septin-2, a cytoskeletal factor involved in cytokinesis. Cleavage of Septin-2 occurred at residue 306 and forced expression of a non-cleavable Septin-2 restored cytokinesis, suggesting a direct mechanism of ZIKV-induced neural toxicity. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Hongda Li
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Laura Saucedo-Cuevas
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Ling Yuan
- Institute of Genetics & Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Danica Ross
- Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Anide Johansen
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Daniel Sands
- Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Valentina Stanley
- Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Alicia Guemez-Gamboa
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Anne Gregor
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lei Tan
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Henrik Molina
- Proteomics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Nicholas Sheets
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Sergey A Shiryaev
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Alexey V Terskikh
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Amy S Gladfelter
- Department of Biology, Howard Hughes Medical Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sujan Shresta
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Zhiheng Xu
- Institute of Genetics & Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Joseph G Gleeson
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA.
| |
Collapse
|
284
|
Zhang W, Liang Y, Li L, Wang X, Yan Z, Dong C, Zeng M, Zhong Q, Liu X, Yu J, Sun S, Liu X, Kang J, Zhao H, Jeong LS, Zhang Y, Jia L. The Nedd8-activating enzyme inhibitor MLN4924 (TAK-924/Pevonedistat) induces apoptosis via c-Myc-Noxa axis in head and neck squamous cell carcinoma. Cell Prolif 2019; 52:e12536. [PMID: 30341788 PMCID: PMC6496207 DOI: 10.1111/cpr.12536] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/08/2018] [Accepted: 07/25/2018] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES The present study aimed to reveal expression status of the neddylation enzymes in HNSCC and to elucidate the anticancer efficacy and the underlying mechanisms of inhibiting neddylation pathway. MATERIALS AND METHODS The expression levels of neddylation enzymes were estimated by Western blotting in human HNSCC specimens and bioinformatics analysis of the cancer genome atlas (TCGA) database. Cell apoptosis was evaluated by Annexin V fluorescein isothiocyanate/propidium iodide (Annexin V-FITC/PI) stain and fluorescence-activated cell sorting (FACS). Small interfering RNA (siRNA) and the CRISPR-Cas9 system were used to elucidate the underlying molecular mechanism of MLN4924-induced HNSCC apoptosis. RESULTS Expression levels of NAE1 and UBC12 were prominently higher in HNSCC tissues than that in normal tissues. Inactivation of the neddylation pathway significantly inhibited malignant phenotypes of HNSCC cells. Mechanistic studies revealed that MLN4924 induced the accumulation of CRL ligase substrate c-Myc that transcriptionally activated pro-apoptotic protein Noxa, which triggered apoptosis in HNSCC. CONCLUSIONS These findings determined the over-expression levels of neddylation enzymes in HNSCC and revealed novel mechanisms underlying neddylation inhibition induced growth suppression in HNSCC cells, which provided preclinical evidence for further clinical evaluation of neddylation inhibitors (eg, MLN4924) for the treatment of HNSCC.
Collapse
Affiliation(s)
- Wenjuan Zhang
- Cancer InstituteFudan University Shanghai Cancer CenterShanghaiChina
- Cancer InstituteLonghua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Yupei Liang
- Cancer InstituteFudan University Shanghai Cancer CenterShanghaiChina
| | - Lihui Li
- Cancer InstituteFudan University Shanghai Cancer CenterShanghaiChina
| | - Xiaofang Wang
- Cancer InstituteFudan University Shanghai Cancer CenterShanghaiChina
| | - Zi Yan
- Cancer InstituteFudan University Shanghai Cancer CenterShanghaiChina
| | - Changsheng Dong
- Cancer InstituteLonghua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Mu‐Sheng Zeng
- Department of Experimental ResearchSun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Qian Zhong
- Department of Experimental ResearchSun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Xue‐Kui Liu
- Department of Head & Neck CancerSun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Jinha Yu
- College of PharmacySeoul National UniversitySeoulKorea
| | - Shuyang Sun
- Department of Oral and Maxillofacial‐Head Neck OncologyNinth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiaojun Liu
- Cancer InstituteFudan University Shanghai Cancer CenterShanghaiChina
| | - Jihui Kang
- Cancer InstituteLonghua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Hu Zhao
- Department of Clinical LaboratoryHuadong HospitalShanghai Key Laboratory of Clinical Geriatric MedicineResearch Center on Aging and MedicineFudan UniversityShanghaiChina
| | | | - Yanmei Zhang
- Department of Clinical LaboratoryHuadong HospitalShanghai Key Laboratory of Clinical Geriatric MedicineResearch Center on Aging and MedicineFudan UniversityShanghaiChina
| | - Lijun Jia
- Cancer InstituteFudan University Shanghai Cancer CenterShanghaiChina
- Cancer InstituteLonghua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
285
|
Bioactive glycoalkaloides isolated from Solanum melongena fruit peels with potential anticancer properties against hepatocellular carcinoma cells. Sci Rep 2019; 9:1746. [PMID: 30741973 PMCID: PMC6370831 DOI: 10.1038/s41598-018-36089-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 11/10/2018] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is progressively increasing tumor with lack of accurate prognosis and inadequate systemic treatment approaches. Solanum sp. (such as Solanum melongena) is a folk herb which is reported to possess anticancer properties. In a continuity for our interest in pursuing the anticancer activity of compounds isolated from the fruit peels of Solanum melongena, the HPLC profiling and ESI-MS assessment for the methanolic extract evidenced the presence of bioactive glycoalkaloids (solasonine, solasodine and solamargine). These glycoalkaloids were isolated, purified and proved to possess in vitro cytotoxicity against human liver cancer cell lines (Huh7 and HepG2). Herein, we investigated the potential mechanism of action of these compounds using DNA content flow-cytometry and apoptosis/necrosis differential anaylsis using annexin-V/FITC staining. Solasonine, solasodine and solamargine inducd significant antiproliferative effect against liver cancer cells (Huh7 and HepG2) which was attributed to cell cycle arrest at S-phase. Solamargine, solasodine and solasonine induced significant apoptosis in Huh7 cells. Only solamargine-induced cell cycle arrest, was reflected as apoptotic cell killing effect against HepG2 cells. In conclusion, glycoalkaloids derived from Solanum melongena and particularly, solamargine are promising antiproliferative agents with potential anticancer effects.
Collapse
|
286
|
Jiménez–Charris E, Lopes DS, Gimenes SNC, Teixeira SC, Montealegre–Sánchez L, Solano–Redondo L, Fierro–Pérez L, Rodrigues Ávila VDM. Antitumor potential of Pllans–II, an acidic Asp49–PLA2 from Porthidium lansbergii lansbergii snake venom on human cervical carcinoma HeLa cells. Int J Biol Macromol 2019; 122:1053-1061. [DOI: 10.1016/j.ijbiomac.2018.09.053] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/05/2018] [Accepted: 09/10/2018] [Indexed: 10/28/2022]
|
287
|
Hydrogen sulfide releasing enmein-type diterpenoid derivatives as apoptosis inducers through mitochondria-related pathways. Bioorg Chem 2019; 82:192-203. [DOI: 10.1016/j.bioorg.2018.10.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/21/2018] [Accepted: 10/04/2018] [Indexed: 01/20/2023]
|
288
|
Guo J, Ozaki I, Xia J, Kuwashiro T, Kojima M, Takahashi H, Ashida K, Anzai K, Matsuhashi S. PDCD4 Knockdown Induces Senescence in Hepatoma Cells by Up-Regulating the p21 Expression. Front Oncol 2019; 8:661. [PMID: 30687637 PMCID: PMC6334536 DOI: 10.3389/fonc.2018.00661] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 12/13/2018] [Indexed: 12/26/2022] Open
Abstract
While the over-expression of tumor suppressor programmed cell death 4 (PDCD4) induces apoptosis, it was recently shown that PDCD4 knockdown also induced apoptosis. In this study, we examined the cell cycle regulators whose activation is affected by PDCD4 knockdown to investigate the contribution of PDCD4 to cell cycle regulation in three types of hepatoma cells: HepG2, Huh7 (mutant p53 and p16-deficient), and Hep3B (p53- and Rb-deficient). PDCD4 knockdown suppressed cell growth in all three cell lines by inhibiting Rb phosphorylation via down-regulating the expression of Rb itself and CDKs, which phosphorylate Rb, and up-regulating the expression of the CDK inhibitor p21 through a p53-independent pathway. We also found that apoptosis was induced in a p53-dependent manner in PDCD4 knockdown HepG2 cells (p53+), although the mechanism of cell death in PDCD4 knockdown Hep3B cells (p53-) was different. Furthermore, PDCD4 knockdown induced cellular senescence characterized by β-galactosidase staining, and p21 knockdown rescued the senescence and cell death as well as the inhibition of Rb phosphorylation induced by PDCD4 knockdown. Thus, PDCD4 is an important cell cycle regulator of hepatoma cells and may be a promising therapeutic target for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Jing Guo
- Division of Hepatology, Diabetology and Endocrinology, Department of Internal Medicine, Saga Medical School, Saga University, Saga, Japan
| | - Iwata Ozaki
- Division of Hepatology, Diabetology and Endocrinology, Department of Internal Medicine, Saga Medical School, Saga University, Saga, Japan.,Health Administration Centre, Saga Medical School, Saga University, Saga, Japan
| | - Jinghe Xia
- Division of Hepatology, Diabetology and Endocrinology, Department of Internal Medicine, Saga Medical School, Saga University, Saga, Japan
| | - Takuya Kuwashiro
- Division of Hepatology, Diabetology and Endocrinology, Department of Internal Medicine, Saga Medical School, Saga University, Saga, Japan
| | - Motoyasu Kojima
- Division of Hepatology, Diabetology and Endocrinology, Department of Internal Medicine, Saga Medical School, Saga University, Saga, Japan
| | - Hirokazu Takahashi
- Division of Hepatology, Diabetology and Endocrinology, Department of Internal Medicine, Saga Medical School, Saga University, Saga, Japan
| | - Kenji Ashida
- Division of Hepatology, Diabetology and Endocrinology, Department of Internal Medicine, Saga Medical School, Saga University, Saga, Japan
| | - Keizo Anzai
- Division of Hepatology, Diabetology and Endocrinology, Department of Internal Medicine, Saga Medical School, Saga University, Saga, Japan
| | - Sachiko Matsuhashi
- Division of Hepatology, Diabetology and Endocrinology, Department of Internal Medicine, Saga Medical School, Saga University, Saga, Japan
| |
Collapse
|
289
|
Ray A, Jena S, Dash B, Sahoo A, Kar B, Patnaik J, Panda PC, Nayak S, Mahapatra N. Hedychium coronarium extract arrests cell cycle progression, induces apoptosis, and impairs migration and invasion in HeLa cervical cancer cells. Cancer Manag Res 2019; 11:483-500. [PMID: 30655700 PMCID: PMC6322495 DOI: 10.2147/cmar.s190004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Hedychium coronarium Koen. (Zingiberaceae) is traditionally used as medicine in countries such as India, China, and Vietnam for treatment of various ailments including cancer. However, in spite of its implied significance in cancer treatment regimes, there are no reports so far involving the anticancerous attributes of H, coronarium ethanol extract (HCEE) on cancer cells and a more comprehensive study on its mechanism is still lacking. Materials and methods The cytotoxicity of HCEE was evaluated by MTT and clonogenic survival assay. Annexin V/propidium iodide (PI), Hoechst 33342 staining, and TUNEL assay were performed to detect apoptosis. Cell cycle analysis was performed using PI staining. JC-1 and 2′,7′-dichlorodihydrofluorescein diacetate assay were used to check the levels of MMP and ROS, respectively. Western blot analysis was carried out to measure the expression levels of proteins. Migration and invasion activity were assessed by wound healing and Transwell membrane assay, respectively. Results Antiproliferative effect of HCEE was investigated in various cancerous and normal cell lines. Among these, HCEE significantly inhibited the survival of HeLa cells without affecting the viability of normal human umbilical vein endothelial cells. Annexin V/PI, Hoechst staining, and TUNEL assay showed HCEE induced apoptosis in HeLa cells in a dose-dependent manner. HCEE promoted cell cycle arrest at G1 phase in HeLa cells by upregulating the levels of p53 and p21 and downregulating the levels of cyclin D1, CDK-4, and CDK-6. Moreover, HCEE treatment upregulated the expression of Bax and downregulated the expression of Bcl-2. Additionally, HCEE activated the caspase cascade by increasing the activities of caspase-9, caspase-8, and caspase-3. The expression levels of Fas ligand and Fas were also upregulated. Further, HCEE inhibited the migratory potential of HeLa cells by downregulating MMP-2 and MMP-9 expression levels. Conclusion Our results indicate H. coronarium exerts antiproliferative and apoptotic effects against HeLa cells, and therefore may be used for treatment against cervical cancer.
Collapse
Affiliation(s)
- Asit Ray
- Regional Medical Research Centre (Indian Council of Medical Research), Chandrasekharpur, Bhubaneswar 751023, Odisha, India.,Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha O Anusandhan University, Kalinganagar, Ghatikia, Bhubaneswar 751003, Odisha, India,
| | - Sudipta Jena
- Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha O Anusandhan University, Kalinganagar, Ghatikia, Bhubaneswar 751003, Odisha, India,
| | - Biswabhusan Dash
- Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha O Anusandhan University, Kalinganagar, Ghatikia, Bhubaneswar 751003, Odisha, India,
| | - Ambika Sahoo
- Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha O Anusandhan University, Kalinganagar, Ghatikia, Bhubaneswar 751003, Odisha, India,
| | - Basudeba Kar
- Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha O Anusandhan University, Kalinganagar, Ghatikia, Bhubaneswar 751003, Odisha, India,
| | - Jeetendranath Patnaik
- Department of Botany, Sri Krushna Chandra Gajapati College, Paralakhemundi 761200, Odisha, India
| | - Pratap Chandra Panda
- Taxonomy and Conservation Division, Regional Plant Resource Centre, Nayapalli, Bhubaneswar 751015, Odisha, India
| | - Sanghamitra Nayak
- Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha O Anusandhan University, Kalinganagar, Ghatikia, Bhubaneswar 751003, Odisha, India,
| | - Namita Mahapatra
- Regional Medical Research Centre (Indian Council of Medical Research), Chandrasekharpur, Bhubaneswar 751023, Odisha, India
| |
Collapse
|
290
|
Mahmoud N, Sabbah DA, Abu-Dahab R, Abuarqoub D, Abdallah M, (Hasan Ibrahim) A, Khalil EA. Cholesterol-coated gold nanorods as an efficient nano-carrier for chemotherapeutic delivery and potential treatment of breast cancer: in vitro studies using the MCF-7 cell line. RSC Adv 2019; 9:12718-12731. [PMID: 35515852 PMCID: PMC9063807 DOI: 10.1039/c9ra01041f] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/10/2019] [Indexed: 01/15/2023] Open
Abstract
Gold nanorods (GNRs) have a recognized role in treatment of cancers as efficient nanocarriers for chemotherapeutic drug delivery. In this study, GNRs modified with cholesterol-PEG were employed as a nanocarrier for a hydrophobic compound having a promising phosphatidylinositol 3-kinase (PI3Kα) inhibitory activity. The acquired nanocomplex was characterized by optical and infra-red (IR) absorption spectroscopies, in addition to hydrodynamic size and zeta potential. Glide docking and superposing of docked poses of the hydrophobic ligand and cholesterol moiety demonstrated that hydrophobic interactions drive the conjugation and attachment of the ligand to the cholesterol moiety of the nanocarrier. In vitro release study under a cellular environment indicates that the presence of cells has enhanced the release and the cellular uptake of the conjugated ligand. Furthermore, the anti-proliferative assay of the nanocomplex revealed potent cytotoxicity over a low concentration range of the nanocomplex against MCF-7 breast cancer cells compared to the free compound or the nanocarrier alone. Analysis of cellular death modality by flow cytometry showed that the nanocomplex has a rapid effect on cell death, as cells went toward the late apoptotic/necrotic stage rapidly and proportionally to the increase of the nanocomplex concentration. The overall results propose that cholesterol-decorated GNRs could be considered as a promising nanocarrier for hydrophobic drugs to achieve efficient delivery and potential therapy against breast cancer cells. Gold nanorods demonstrate a recognized role in the treatment of breast cancer cell lines as an efficient nanocarrier for chemotherapeutic drug delivery.![]()
Collapse
Affiliation(s)
- Nouf N. Mahmoud
- Faculty of Pharmacy
- Al-Zaytoonah University of Jordan
- Amman 11733
- Jordan
| | - Dima A. Sabbah
- Faculty of Pharmacy
- Al-Zaytoonah University of Jordan
- Amman 11733
- Jordan
| | - Rana Abu-Dahab
- School of Pharmacy
- The University of Jordan
- Amman 11942
- Jordan
| | - Duaa Abuarqoub
- Cell Therapy Center
- The University of Jordan
- Amman 11942
- Jordan
| | - Maha Abdallah
- School of Pharmacy
- The University of Jordan
- Amman 11942
- Jordan
| | | | - Enam A. Khalil
- School of Pharmacy
- The University of Jordan
- Amman 11942
- Jordan
| |
Collapse
|
291
|
Biswas S, Reddy ND, Jayashree BS, Rao CM. Evaluation of Novel 3-Hydroxyflavone Analogues as HDAC Inhibitors against Colorectal Cancer. Adv Pharmacol Sci 2018; 2018:4751806. [PMID: 30687400 PMCID: PMC6327263 DOI: 10.1155/2018/4751806] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/15/2018] [Accepted: 11/21/2018] [Indexed: 12/18/2022] Open
Abstract
Alteration of epigenetic enzymes is associated with the pathophysiology of colon cancer with an overexpression of histone deacetylase 8 (HDAC8) enzyme in this tissue. Numerous reports suggest that targeting HDAC8 is a viable strategy for developing new anticancer drugs. Flavonols provide a rich source of molecules that are effective against cancer; however, their clinical use is limited. The present study investigated the potential of quercetin and synthetic 3-hydroxyflavone analogues to inhibit HDAC8 enzyme and evaluated their anticancer property. Synthesis of the analogues was carried out, and cytotoxicity was determined using MTT assay. Nonspecific and specific HDAC enzyme inhibition assays were performed followed by the expression studies of target proteins. Induction of apoptosis was studied through annexin V and caspase 3/7 activation assay. Furthermore, the analogues were assessed against in vivo colorectal cancer. Among the synthesized analogues, QMJ-2 and QMJ-5 were cytotoxic against HCT116 cells with an IC50 value of 68 ± 2.3 and 27.4 ± 1.8 µM, respectively. They inhibited HDAC enzyme in HCT116 cells at an IC50 value of 181.7 ± 22.04 and 70.2 ± 4.3 µM, respectively, and inhibited human HDAC8 and 1 enzyme at an IC50 value of <50 µM with QMJ-5 having greater specificity towards HDAC8. A reduction in the expression of HDAC8 and an increase in acetyl H3K9 expression were observed with the synthesized analogues. Both QMJ-2 and QMJ-5 treatment induced apoptosis through the activation of caspase 3/7 evident from 55.70% and 83.55% apoptotic cells, respectively. In vivo studies revealed a significant decrease in colon weight to length ratio in QMJ-2 and QMJ-5 treatment groups compared to DMH control. Furthermore, a reduction in aberrant crypt foci formation was observed in the treatment groups. The present study demonstrated the potential of novel 3-hydroxyflavone analogues as HDAC8 inhibitors with anticancer property against colorectal cancer.
Collapse
Affiliation(s)
- Subhankar Biswas
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Neetinkumar D. Reddy
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - B. S. Jayashree
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - C. Mallikarjuna Rao
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| |
Collapse
|
292
|
Huang IT, Dhungel B, Shrestha R, Bridle KR, Crawford DHG, Jayachandran A, Steel JC. Spotlight on Bortezomib: potential in the treatment of hepatocellular carcinoma. Expert Opin Investig Drugs 2018; 28:7-18. [PMID: 30474444 DOI: 10.1080/13543784.2019.1551359] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION This study reviews the evidence for the use of Bortezomib (BZB), a first-in-class proteasome inhibitor in advanced Hepatocellular carcinoma (HCC). This review aims to delineate the role of BZB within the management of non-surgical and metastatic HCC, either as an alternative or as an adjunct to the current treatment paradigm. AREAS COVERED In addition to BZB pharmacology and mechanism of action, safety and tolerance profiles of the drug obtained from clinical trials are explored. The utility of BZB as a therapeutic agent either alone or in combination with other therapies against HCC, including its application in both preclinical and clinical settings has been reviewed. In particular, we highlight the importance of preclinical evaluation of BZB as a combinatorial agent in synergism with other therapies for the use in the management of HCC. EXPERT OPINION There has been much interest surrounding the use of BZB, a first-in-class proteasome inhibitor for HCC therapy. The discernment of outcomes of BZB clinical trials for HCC need to take into consideration the disease-specific factors that can affect survival outcomes including patient selection and aetiological differences. Further preclinical testing of BZB in combination with other therapeutic modalities can be important for eliciting enhanced anti-HCC effects.
Collapse
Affiliation(s)
- I-Tao Huang
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Bijay Dhungel
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Ritu Shrestha
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Kim R Bridle
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Darrell H G Crawford
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Aparna Jayachandran
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Jason C Steel
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,c School of Health, Medical and Applied Sciences , CQ University , Rockhampton , Australia
| |
Collapse
|
293
|
Fang J, Zheng Z, Yang Z, Peng X, Zuo Z, Cui H, Ouyang P, Shu G, Chen Z, Huang C. Ameliorative effects of selenium on the excess apoptosis of the jejunum caused by AFB 1 through death receptor and endoplasmic reticulum pathways. Toxicol Res (Camb) 2018; 7:1108-1119. [PMID: 30510680 PMCID: PMC6220717 DOI: 10.1039/c8tx00068a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 07/03/2018] [Indexed: 12/18/2022] Open
Abstract
Aflatoxin B1 (AFB1), one of most potent and common mycotoxins in human food and animal feed, has hepatotoxic and carcinogenic effects on humans and poultry. Recent studies indicated that selenium (Se) has a protective effect on apoptosis induced by toxin poisoning. The present study was designed to reveal the ameliorative effects of selenium on the expression of apoptosis related molecules in the jejunum of broilers exposed to an AFB1 diet for 3 weeks. A total of 216 one-day-old healthy Cobb broilers were randomly divided into the control group (0 mg kg-1 AFB1), AFB1 group (0.6 mg kg-1 AFB1), AFB1 + Se group (0.6 mg kg-1 AFB1 + 0.4 mg kg-1 supplement Se) and Se group (0.4 mg kg-1 supplement Se), respectively. TUNEL and flow cytometry assays both indicated that 0.4 mg kg-1 selenium could ameliorate excess apoptosis caused by AFB1 in jejunal cells. Moreover, the expressions of FAS, FASL, TNF-α, TNF-R1, CASPASE-3, CASPASE-8, CASPASE-10, GRP78 and GRP94 analyzed by qRT-PCR demonstrated that 0.4 mg kg-1 selenium restored these parameters to be close to those in the control group. In summary, supplementation of selenium at a concentration of 0.4 mg kg-1 selenium could protect the chicken's jejunum from excess apoptosis caused by 0.6 mg kg-1 AFB1 through down-regulating the expression of death receptor pathway and endoplasmic reticulum pathway related molecules. According to this conclusion, this study may contribute to a better understanding of selenium's protective role against AFB1 poisoning.
Collapse
Affiliation(s)
- Jing Fang
- College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , Sichuan 611130 , PR China .
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province , College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , Sichuan 611130 , PR China
| | - Zhixiang Zheng
- College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , Sichuan 611130 , PR China .
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province , College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , Sichuan 611130 , PR China
| | - Zhuangzhi Yang
- Chengdu Academy of Agriculture and Forestry Sciences , Chengdu 611130 , Sichuan , PR China
| | - Xi Peng
- College of Life Sciences , China West Normal University , Nanchong , Sichuan 637002 , PR China .
| | - Zhicai Zuo
- College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , Sichuan 611130 , PR China .
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province , College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , Sichuan 611130 , PR China
| | - Hengmin Cui
- College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , Sichuan 611130 , PR China .
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province , College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , Sichuan 611130 , PR China
| | - Ping Ouyang
- College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , Sichuan 611130 , PR China .
| | - Gang Shu
- College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , Sichuan 611130 , PR China .
| | - Zhengli Chen
- College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , Sichuan 611130 , PR China .
| | - Chao Huang
- College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , Sichuan 611130 , PR China .
| |
Collapse
|
294
|
Cell cycle modulatory effects of Amaryllidaceae alkaloids. Life Sci 2018; 213:94-101. [DOI: 10.1016/j.lfs.2018.08.073] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 01/13/2023]
|
295
|
Matsui T, Ito C, Kato A, Wu TS, Itoigawa M. Acrofolione A and B, acetophenone dimers from Acronychia pendunculata, induce an apoptotic effect on human NALM-6 pre-B cell leukaemia cells. J Pharm Pharmacol 2018; 71:348-361. [PMID: 30362134 DOI: 10.1111/jphp.13035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/29/2018] [Indexed: 12/16/2022]
Abstract
OBJECTIVES We investigated the apoptotic activities of acrofolione A (1) and B (2) isolated from Acronychia pedunculata against a human pre-B cell leukaemia cell line (NALM-6) to explore the apoptosis-related signalling molecules targeted by 1 and 2. METHODS The apoptosis effects of 1 and 2 in NALM-6 cells were investigated by TUNEL staining, annexin V, mitochondria membrane potential and caspase 3/7 activity. We carried out a protein array to explore the signalling molecules involved in apoptosis comprehensively. KEY FINDINGS Acrofolione A (1) suppressed the growth of NALM-6, K562 and HPB-ALL cells (IC50 16.7 ± 1.9, 17.9 ± 0.3 and 10.1 ± 0.2 μm, respectively) more effectively than acrofolione B (2). Both compounds time-dependently increased the number of NALM-6 cells with abnormal nuclei, and increased the number of annexin V-positive cells and decreased the mitochondrial membrane potential of NALM-6 cells. Acrofolione A (1) markedly elevated caspase 3/7 activity and increased the number of TUNEL-positive cells. Cells treated with either compound showed enhanced expression of cleaved PARP and cleaved caspase 3 and 7, and reduced survivin protein levels. CONCLUSIONS Acrofolione A (1) and B (2) may be useful in the treatment of various types of leukaemia.
Collapse
Affiliation(s)
- Takuya Matsui
- Department of Physiology, Aichi Medical University, Nagakute, Aichi, Japan.,Faculty of Pharmacy, Meijo University, Nagoya, Aichi, Japan
| | - Chihiro Ito
- Faculty of Pharmacy, Meijo University, Nagoya, Aichi, Japan
| | - Ayumi Kato
- Faculty of Pharmacy, Meijo University, Nagoya, Aichi, Japan
| | - Tian-Shung Wu
- Department of Chemistry, National Cheng Kung University, Tainan, Taiwan
| | - Masataka Itoigawa
- School of Sports and Health Science, Tokai Gakuen University, Miyoshi, Aichi, Japan
| |
Collapse
|
296
|
Mitochondrial enzyme GLUD2 plays a critical role in glioblastoma progression. EBioMedicine 2018; 37:56-67. [PMID: 30314897 PMCID: PMC6284416 DOI: 10.1016/j.ebiom.2018.10.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/25/2018] [Accepted: 10/02/2018] [Indexed: 01/06/2023] Open
Abstract
Background Glioblastoma (GBM) is the most frequent and malignant primary brain tumor in adults and despite the progress in surgical procedures and therapy options, the overall survival remains very poor. Glutamate and α-KG are fundamental elements necessary to support the growth and proliferation of GBM cells. Glutamate oxidative deamination, catalyzed by GLUD2, is the predominant pathway for the production of α-KG. Methods GLUD2 emerged from the RNA-seq analysis of 13 GBM patients, performed in our laboratory and a microarray analysis of 77 high-grade gliomas available on the Geo database. Thereafter, we investigated GLUD2 relevance in cancer cell behavior by GLUD2 overexpression and silencing in two different human GBM cell lines. Finally, we overexpressed GLUD2 in-vivo by using zebrafish embryos and monitored the developing central nervous system. Findings GLUD2 expression was found associated to the histopathological classification, prognosis and survival of GBM patients. Moreover, through in-vitro functional studies, we showed that differences in GLUD2 expression level affected cell proliferation, migration, invasion, colony formation abilities, cell cycle phases, mitochondrial function and ROS production. In support of these findings, we also demonstrated, with in-vivo studies, that GLUD2 overexpression affects glial cell proliferation without affecting neuronal development in zebrafish embryos. Interpretation We concluded that GLUD2 overexpression inhibited GBM cell growth suggesting a novel potential drug target for control of GBM progression. The possibility to enhance GLUD2 activity in GBM could result in a blocked/reduced proliferation of GBM cells without affecting the survival of the surrounding neurons.
Collapse
|
297
|
Mohammadi-Motlagh HR, Yarani R, Sadeghalvad M, Adham E, Rasouli H, Mostafaie A. 2-Methylpyridine-1-ium-1-sulfonate as an Inducer of Apoptosis and Cell Cycle Arrest: A comparative in vitro and Computational Study. Nutr Cancer 2018; 71:643-656. [PMID: 30273005 DOI: 10.1080/01635581.2018.1506495] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
"Let food be thy medicine and thy medicine be thy food" was expressed by Hippocrates and the health benefits of medicinal plants and natural products have been considered by humans since historic times. The current study aims to investigate the anti-cancer activity of 2-Methylpyridine-1-ium-1-sulfonate (MPS) isolated from bulbs of Allium hirtifolium. The MPS compound (in a dose-dependent manner) induced arrest the AGS cells in G1 and G2/M phases, and Caco-2 cells in G1 and S phases. These findings were associated with the down-regulation of cyclin D1, CDK4, and up-regulation of p21, p27 and p53. According to the morphological observations and DNA fragmentation assay, the MPS compound induced apoptosis in both cell lines, and also cause a significant increase in the expression of Bax/Bcl-2. In this context, our molecular docking results unveiled that the MPS compound has considerable affinity to interact with the minor groove of ctDNA and also with cell cycle kinases. To approve and find the accurate MPS mode of action against cancer cell lines (especially in gastrointestinal cancer) further studies is highly recommended.
Collapse
Affiliation(s)
| | - Reza Yarani
- a Medical Biology Research Center , Kermanshah University of Medical Sciences , Kermanshah , Iran.,b Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences , The Panum Institute, University of Copenhagen , Copenhagen N , Denmark
| | - Mona Sadeghalvad
- a Medical Biology Research Center , Kermanshah University of Medical Sciences , Kermanshah , Iran
| | - Elham Adham
- a Medical Biology Research Center , Kermanshah University of Medical Sciences , Kermanshah , Iran
| | - Hassan Rasouli
- a Medical Biology Research Center , Kermanshah University of Medical Sciences , Kermanshah , Iran
| | - Ali Mostafaie
- a Medical Biology Research Center , Kermanshah University of Medical Sciences , Kermanshah , Iran
| |
Collapse
|
298
|
Kim SA, Kang OH, Kwon DY. Cryptotanshinone Induces Cell Cycle Arrest and Apoptosis of NSCLC Cells through the PI3K/Akt/GSK-3β Pathway. Int J Mol Sci 2018; 19:E2739. [PMID: 30217003 PMCID: PMC6163873 DOI: 10.3390/ijms19092739] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/06/2018] [Accepted: 09/06/2018] [Indexed: 12/18/2022] Open
Abstract
Cryptotanshinone (CTT) is a natural product and a quinoid diterpene isolated from the root of the Asian medicinal plant, Salvia miltiorrhizabunge. Notably, CTT has a variety of anti-cancer actions, including the activation of apoptosis, anti-proliferation, and reduction in angiogenesis. We further investigated the anti-cancer effects of CTT using MTS, LDH, and Annexin V assay, DAPI staining, cell cycle arrest, and Western blot analysis in NSCLC cell lines. NSCLC cells treated with CTT reduced cell growth through PI3K/Akt/GSK3β pathway inhibition, G0/G1 cell cycle arrest, and the activation of apoptosis. CTT induced an increase of caspase-3, caspase-9, poly-ADP-ribose polymerase (PARP), and Bax, as well as inhibition of Bcl-2, survivin, and cellular-inhibitor of apoptosis protein 1 and 2 (cIAP-1 and -2). It also induced G0/G1 phase cell cycle arrest by decreasing the expression of the cyclin A, cyclin D, cyclin E, Cdk 2, and Cdk 4. These results highlight anti-proliferation the latent of CTT as natural therapeutic agent for NSCLC. Therefore, we investigated the possibility of CTT as an anti-cancer agent by comparing with GF, which is a representative anti-cancer drug.
Collapse
Affiliation(s)
- Sang-A Kim
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang Oriental Medicines Research Institute, Wonkwang University, Iksan, Jeonbuk 570-749, Korea.
| | - Ok-Hwa Kang
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang Oriental Medicines Research Institute, Wonkwang University, Iksan, Jeonbuk 570-749, Korea.
| | - Dong-Yeul Kwon
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang Oriental Medicines Research Institute, Wonkwang University, Iksan, Jeonbuk 570-749, Korea.
| |
Collapse
|
299
|
Shahin S, Singh SP, Chaturvedi CM. 2.45 GHz microwave radiation induced oxidative and nitrosative stress mediated testicular apoptosis: Involvement of a p53 dependent bax-caspase-3 mediated pathway. ENVIRONMENTAL TOXICOLOGY 2018; 33:931-945. [PMID: 29968967 DOI: 10.1002/tox.22578] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 05/03/2018] [Accepted: 05/06/2018] [Indexed: 06/08/2023]
Abstract
Deleterious effects of MW radiation on the male reproduction are well studied. Previous reports although suggest that 2.45 GHz MW irradiation induced oxidative and nitrosative stress adversely affects the male reproductive function but the detailed molecular mechanism occurring behind it has yet to be elucidated. The aim of present study was to investigate the underlying detailed pathway of the testicular apoptosis induced by free radical load and redox imbalance due to 2.45 GHz MW radiation exposure and the degree of severity along with the increased exposure duration. Twelve-week old male mice were exposed to 2.45 GHz MW radiation [continuous-wave (CW) with overall average Power density of 0.0248 mW/cm2 and overall average whole body SAR value of 0.0146 W/kg] for 2 hr/day over a period of 15, 30, and 60 days. Testicular histology, serum testosterone, ROS, NO, MDA level, activity of antioxidant enzymes, expression of pro-apoptotic proteins (p53 and Bax), anti-apoptotic proteins (Bcl-2 and Bcl-xL ), cytochrome-c, inactive/active caspase-3, and uncleaved PARP-1 were evaluated. Findings suggest that 2.45 GHz MW radiation exposure induced testicular redox imbalance not only leads to enhanced testicular apoptosis via p53 dependent Bax-caspase-3 mediated pathway, but also increases the degree of apoptotic severity in a duration dependent manner.
Collapse
Affiliation(s)
- Saba Shahin
- Department of Zoology, Banaras Hindu University, Varanasi, 221005, India
| | - Surya Pal Singh
- Department of Electronics Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, 221005, India
| | | |
Collapse
|
300
|
Kee JY, Han YH, Mun JG, Park SH, Jeon HD, Hong SH. Gomisin A Suppresses Colorectal Lung Metastasis by Inducing AMPK/p38-Mediated Apoptosis and Decreasing Metastatic Abilities of Colorectal Cancer Cells. Front Pharmacol 2018; 9:986. [PMID: 30210348 PMCID: PMC6124511 DOI: 10.3389/fphar.2018.00986] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 08/10/2018] [Indexed: 01/20/2023] Open
Abstract
Gomisin A (G.A) is a dietary lignan compound from Schisandra chinensis. In this study, the effect of G.A on the proliferation and metastasis of colorectal cancer (CRC) cells was investigated using several CRC cell lines and a lung metastasis mouse model. Both oral and intraperitoneal administration of G.A (50 mg/kg) inhibited lung metastasis of CT26 cells. Various concentrations of G.A were incubated with CRC cell lines and their viability was determined using a cell counting kit-8 assay. G.A significantly decreased the viability of various CRC cell lines, whereas it did not change the proliferation of normal colon cells. G.A induced G0/G1 phase arrest and apoptosis of CT26 and HT29 cells by regulating cyclin D1/cyclin-dependent kinase 4 (CDK4) expression and apoptotic proteins such as caspases and B-cell lymphoma-2 (Bcl-2) family proteins, respectively. G.A-induced apoptosis was mediated by AMPK/p38 activation in CRC cells. A non-cytotoxic concentration of G.A inhibited epithelial–mesenchymal transition of CRC cells by modulating E-cadherin and N-cadherin expression levels. Moreover, the migration and invasion of CRC cells were reduced by G.A treatment. Especially, G.A decreased matrix metalloproteinase (MMP)-2 and MMP-9 expressions and activities. G.A ameliorated lung metastasis of CRC cells by decreasing cell survival and metastatic abilities of CRC cells. Thus, G.A might be a potential novel therapeutic agent for metastatic CRC.
Collapse
Affiliation(s)
- Ji-Ye Kee
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan, South Korea
| | - Yo-Han Han
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan, South Korea
| | - Jeong-Geon Mun
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan, South Korea
| | - Seong-Hwan Park
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan, South Korea
| | - Hee D Jeon
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan, South Korea
| | - Seung-Heon Hong
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan, South Korea
| |
Collapse
|