1
|
Kaushal S, Gupta S, Shefrin S, Vora DS, Kaul SC, Sundar D, Wadhwa R, Dhanjal JK. Synthetic and Natural Inhibitors of Mortalin for Cancer Therapy. Cancers (Basel) 2024; 16:3470. [PMID: 39456564 PMCID: PMC11506508 DOI: 10.3390/cancers16203470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Upregulation of stress chaperone Mortalin has been closely linked to the malignant transformation of cells, tumorigenesis, the progression of tumors to highly aggressive stages, metastasis, drug resistance, and relapse. Various in vitro and in vivo assays have provided evidence of the critical role of Mortalin upregulation in promoting cancer cell characteristics, including proliferation, migration, invasion, and the inhibition of apoptosis, a consistent feature of most cancers. Given its critical role in several steps in oncogenesis and multi-modes of action, Mortalin presents a promising target for cancer therapy. Consequently, Mortalin inhibitors are emerging as potential anti-cancer drugs. In this review, we discuss various inhibitors of Mortalin (peptides, small RNAs, natural and synthetic compounds, and antibodies), elucidating their anti-cancer potentials.
Collapse
Affiliation(s)
- Shruti Kaushal
- Department of Computational Biology, Indraprastha Institute of Information Technology (IIIT) Delhi, Okhla Industrial Estate, Phase III, New Delhi 110020, India; (S.K.); (S.G.); (D.S.V.)
| | - Samriddhi Gupta
- Department of Computational Biology, Indraprastha Institute of Information Technology (IIIT) Delhi, Okhla Industrial Estate, Phase III, New Delhi 110020, India; (S.K.); (S.G.); (D.S.V.)
| | - Seyad Shefrin
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology (IIT) Delhi, New Delhi 110016, India; (S.S.); (D.S.)
| | - Dhvani Sandip Vora
- Department of Computational Biology, Indraprastha Institute of Information Technology (IIIT) Delhi, Okhla Industrial Estate, Phase III, New Delhi 110020, India; (S.K.); (S.G.); (D.S.V.)
| | - Sunil C. Kaul
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Central 4-1, Tsukuba 305-8565, Japan;
| | - Durai Sundar
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology (IIT) Delhi, New Delhi 110016, India; (S.S.); (D.S.)
- Institute of Bioinformatics and Applied Biotechnology (IBAB), Bengaluru 560100, India
| | - Renu Wadhwa
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Central 4-1, Tsukuba 305-8565, Japan;
| | - Jaspreet Kaur Dhanjal
- Department of Computational Biology, Indraprastha Institute of Information Technology (IIIT) Delhi, Okhla Industrial Estate, Phase III, New Delhi 110020, India; (S.K.); (S.G.); (D.S.V.)
| |
Collapse
|
2
|
Rai R, Lightfoot S, Benbrook DM. Manipulation of metabolic responses enhances SHetA2 efficacy without toxicity in cervical cancer cell lines and xenografts. Gynecol Oncol 2024; 180:44-54. [PMID: 38052108 PMCID: PMC10922646 DOI: 10.1016/j.ygyno.2023.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/10/2023] [Accepted: 11/12/2023] [Indexed: 12/07/2023]
Abstract
OBJECTIVE The high frequency of cervical cancer recurrence after primary therapy necessitates alternative treatments. High-risk human papillomavirus (HR-HPV) causes cervical cancer and it's continued presence supports elevated metabolism, proliferation and survival of cancer cells. The low-to-no toxicity new investigational drug, SHetA2, counteracts high-risk human papillomavirus (HR-HPV) effects on cell proliferation and survival in cervical cancer cells and xenograft tumors by disrupting heat shock protein 70 chaperone protection of oncogenic proteins. Our objective was to study the involvement of metabolism in SHetA2 effects on cervical cancer cells and tumors. METHODS SHetA2-mediated proteomic and metabolic effects were measured in HR-HPV-positive CaSKi and SiHa and HR-HPV-negative C-33 A cervical cancer cell lines. Combined treatment with 2-deoxyglucose (2-DG) was evaluated in cell culture and SiHa xenografts. RESULTS SHetA2 inhibited oxidative phosphorylation (OxPhos) and altered levels of proteins involved in metabolism, protein synthesis, and DNA replication and repair. Cervical cancer cells responded by elevating glycolysis. Inhibition of the glycolytic responses using galactose media or 2-DG increased SHetA2 sensitivity of two HR-HPV-positive, but not an HR-HPV-negative cervical cancer cell line. Interaction of 2-DG and SHetA2 was synergistic in HR-HPV positive cell lines in association with augmentation of SHetA2 ATP reduction, but not SHetA2 DNA damage induction. These results were verified in a SiHa xenograft tumor model without evidence of toxicity. CONCLUSIONS Compensatory glycolysis counteracts OxPhos inhibition in SHetA2-treated HR-HPV-positive cervical cancer cell lines. Prevention of compensatory glycolysis with 2-DG or another glycolysis inhibitor has the potential to improve SHetA2 therapy without toxicity.
Collapse
Affiliation(s)
- Rajani Rai
- Section of Gynecologic Oncology, Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, USA
| | - Stanley Lightfoot
- Department of Pathology, University of Oklahoma Health Sciences Center, USA
| | - Doris Mangiaracina Benbrook
- Section of Gynecologic Oncology, Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, USA.
| |
Collapse
|
3
|
Khaksar S, Aliabadi A, Panjehpour A, Abdolmaleki S. Effect of the extra-nuclear cation on the cytotoxicity and mechanism of action of pyridine-2,6-dicarboxylate Ga(III) complexes. Toxicology 2023; 495:153609. [PMID: 37541566 DOI: 10.1016/j.tox.2023.153609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/21/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023]
Abstract
Two Ga(III) complexes (C1) and (C2) were prepared by the one-pot reaction of pyridine-2,6-dicarboxylic acid and aminopyridine derivatives with gallium(III) nitrate octahydrate. The compounds were characterized by single-crystal X-ray diffraction. The distorted octahedral geometry was confirmed by crystallographic data for both complexes. The study of the in vitro cytotoxicity of the compounds showed that the presence of different extra-nuclear cations can affect the cytotoxicity of the same anionic complexes. The most significant antiproliferative activity was observed for C1 (IC50 = 0.69 μM, MAE = 73.96%) and C2 (IC50 = 3.78 μM, MAE = 60.35%) (where MAE represents the maximal antiproliferative effect) against A431 cell line. The mechanistic study evidenced the same pathway for the death of A431 cells treated with the complexes, although the results for C2 were obtained at approximately five times the concentration of C1. According to the study, both complexes induced cell cycle arrest in G2/M phase in A431 cells by upregulating the levels of p21, p27, p-cdc25C, and p-cdc2 and downregulating the levels of cdc25C, cdc2, and cyclin B1. In addition, apoptosis via a caspase-dependent mitochondrial pathway was confirmed by a decrease in Bcl-2 family proteins and an increase in the expression of procaspase-9 and 3. Also, the complexes induced autophagic cell death by activating the RAGE /PI3KC3/Beclin 1 pathway in A431 cells. DATA AVAILABILITY: CCDC 874052 and 874055 contain the supplementary crystallographic data for C1 and C2, respectively. These data can be obtained free of charge via http://www.ccdc.cam.ac.uk/services/structures?pid=ccdc:874052,874055&sid=CCDCManual, or from the Cambridge Crystallographic Data Centre, 12 Union Road, Cambridge CB2 1EZ, UK; fax: (+44) 1223-336-033; or e-mail: deposit@ccdc.cam.ac.uk.
Collapse
Affiliation(s)
- Samad Khaksar
- School of Science and Technology, The University of Georgia, Tbilisi, Georgia.
| | - Alireza Aliabadi
- Pharmaceutical Sciences Research Center, Health Institute, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Akram Panjehpour
- Department of Chemistry, Faculty of Sciences, Tarbiat Modares University, P.O. Box 14115-175, Tehran, Iran
| | - Sara Abdolmaleki
- School of Science and Technology, The University of Georgia, Tbilisi, Georgia.
| |
Collapse
|
4
|
Mitochondrial Dysfunction Pathway Alterations Offer Potential Biomarkers and Therapeutic Targets for Ovarian Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5634724. [PMID: 35498135 PMCID: PMC9045977 DOI: 10.1155/2022/5634724] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/24/2021] [Accepted: 04/02/2022] [Indexed: 11/29/2022]
Abstract
The mitochondrion is a very versatile organelle that participates in some important cancer-associated biological processes, including energy metabolism, oxidative stress, mitochondrial DNA (mtDNA) mutation, cell apoptosis, mitochondria-nuclear communication, dynamics, autophagy, calcium overload, immunity, and drug resistance in ovarian cancer. Multiomics studies have found that mitochondrial dysfunction, oxidative stress, and apoptosis signaling pathways act in human ovarian cancer, which demonstrates that mitochondria play critical roles in ovarian cancer. Many molecular targeted drugs have been developed against mitochondrial dysfunction pathways in ovarian cancer, including olive leaf extract, nilotinib, salinomycin, Sambucus nigra agglutinin, tigecycline, and eupatilin. This review article focuses on the underlying biological roles of mitochondrial dysfunction in ovarian cancer progression based on omics data, potential molecular relationship between mitochondrial dysfunction and oxidative stress, and future perspectives of promising biomarkers and therapeutic targets based on the mitochondrial dysfunction pathway for ovarian cancer.
Collapse
|
5
|
Benbrook DM. SHetA2 Attack on Mortalin and Colleagues in Cancer Therapy and Prevention. Front Cell Dev Biol 2022; 10:848682. [PMID: 35281109 PMCID: PMC8906462 DOI: 10.3389/fcell.2022.848682] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Heat Shock Proteins of the 70-kDa family (HSP70s) do not cause cancer by themselves, but instead protect cells as they transform into cancer. These molecular chaperones bind numerous client proteins and utilize ATP hydrolysis to facilitate proper protein folding, formation of functional complexes and cellular localizations, or degradation of irreparably damaged proteins. Their transient upregulation by stressful situations avoids induction of programmed cell death. Continued upregulation of the mortalin, heat shock cognate (hsc70) and glucose regulated protein 78 (Grp78) support cancer development and progression by supporting pro-proliferative and metabolic functions and repressing pro-death functions of oncoproteins and tumor suppressor proteins. This review describes the discovery and development of a lead anti-cancer compound, sulfur heteroarotinoid A2 (SHetA2, NSC726189), which was originally developed to bind retinoic acid receptors, but was subsequently found to work independently of these receptors. The discovery and validation of mortalin, hsc70 and Grp78 as SHetA2 target proteins is summarized. The documented and hypothesized roles of these HSP70 proteins and their clients in the mechanism of SHetA2 inhibition of cancer without toxicity are discussed. Use of this mechanistic data to evaluate drug action in a cancer clinical trial and develop synergistic drug combinations is explained. Knowledge needed to optimize SHetA2 analogs for use in cancer therapy and prevention is proposed as future directions.
Collapse
|
6
|
Novel Tl(III) complexes containing pyridine-2,6-dicarboxylate derivatives with selective anticancer activity through inducing mitochondria-mediated apoptosis in A375 cells. Sci Rep 2021; 11:15699. [PMID: 34344980 PMCID: PMC8333620 DOI: 10.1038/s41598-021-95278-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/23/2021] [Indexed: 02/07/2023] Open
Abstract
Three novel Tl(III) complexes (C1), (C2) and (C3) were synthesized using the one-pot reactions of pyridine dicarboxylic acid derivatives, 2-aminobenzimidazole and/or 4-aminopyridine, and also thallium(III) nitrate trihydrate metal salt. The structure of all three complexes was determined by the single-crystal X-ray diffraction. C1 and C2 were realized to be isostructural with disordered square anti-prismatic geometry and for C3 arrangement of the distorted tricapped triangular prism was proposed. Cyclic voltammetry measurements on the complexes exhibited that formal potential values are more positive for C1 (E0' 0.109 V) and C3 (E0' 0.244 V) compared to C2 (E0' -0.051 V), versus Ag/AgCl under argon. Moreover, cytotoxicity of the compounds was evaluated in vitro against two cancer cell lines including a human melanoma (A375), a human colon adenocarcinoma (HT29), and also one normal cell human foreskin fibroblast (HFF). The selective and potent cytotoxicity effect was exhibited by C1 and C3 on cancer cell lines. The apoptosis through a caspase-dependent mitochondrion pathway was confirmed by ROS production, MMP reduction, p53 activation, Bax up-regulation, and Bcl-2 down-regulation, cytochrome c release, procaspase-9, and 3 expression, for A375 cells treated to C1 and C3. According to similar cellular uptake of the complexes in A375 cell line, the generation of ROS was considered as an effective agent to justify the inhibition effect C1 and C3 on mentioned cells. Furthermore, arresting the cell cycle in the G2-M phase and inducing apoptosis were indicated by these two complexes.
Collapse
|
7
|
Rajput M, Bithel N, Vijayakumar S. Antimicrobial, antibiofilm, antioxidant, anticancer, and phytochemical composition of the seed extract of Pongamia pinnata. Arch Microbiol 2021; 203:4005-4024. [PMID: 34037822 DOI: 10.1007/s00203-021-02365-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 10/21/2022]
Abstract
Ethyl acetate seed extract of Pongamia pinnata displayed the highest antimicrobial potential against all test pathogens and Staphylococcus epidermidis was reported as the most sensitive strain with MIC/MBC 1.56/3.12 mg ml-1. It inhibited S. epidermidis biofilm 97.43% at MIC and LM as well as FE-SEM micrographs displayed extensive disintegration in biofilm. It showed the highest TPC (1.23 ± 0.04 g GAE g-1), TFC (0.95 ± 0.05 g CE g-1), and antioxidant activity with IC50 18.47 ± 0.33 μg ml-1. MTT assay displayed concentration-dependent strong cytotoxicity on K562 cells on the treatment of ethyl acetate extract with an IC50 value of 84.41 μg ml-1. On the other hand, it showed minute cytotoxicity on normal PBMCs with an IC50 value of 410.14 μg ml-1. GC-MS analysis revealed that Hexadecanoic acid (35.97%); 2-(1,3-Benzodioxol-5-yl)furo[2,3-h]chromen-4-one (Pongaglabrone) (22.82%); 2,2-Dimethylindane-1,3-dione- (13.05%) were the three major components in ethyl acetate extract. The present investigation showcases ethyl acetate extract as a potent antimicrobial, antibiofilm, antioxidant, and anticancer agent that opens a new avenue for its phytochemicals as a therapeutic agent.
Collapse
Affiliation(s)
- Minakshi Rajput
- Department of Botany and Microbiology, Gurukula Kangri Vishwavidyalaya, Haridwar, 249404, Uttarakhand, India.
| | - Navneet Bithel
- Department of Botany and Microbiology, Gurukula Kangri Vishwavidyalaya, Haridwar, 249404, Uttarakhand, India
| | - Sekar Vijayakumar
- Marine College, Shandong University, Weihai, 264209, People's Republic of China
| |
Collapse
|
8
|
Abstract
Flow cytometry (FCM) is a sophisticated technique that works on the principle of light scattering and fluorescence emission by the specific fluorescent probe-labeled cells as they pass through a laser beam. It offers several unique advantages as it allows fast, relatively quantitative, multiparametric analysis of cell populations at the single cell level. In addition, it also enables physical sorting of the cells to separate the subpopulations based on different parameters. In this constantly evolving field, innovative technologies such as imaging FCM, mass cytometry and Raman FCM are being developed in order to address limitations of traditional FCM. This review explains the general principles, main applications and recent advances in the field of FCM.
Collapse
|
9
|
Weng CH, Wu CS, Wu JC, Kung ML, Wu MH, Tai MH. Cisplatin-Induced Giant Cells Formation Is Involved in Chemoresistance of Melanoma Cells. Int J Mol Sci 2020; 21:ijms21217892. [PMID: 33114317 PMCID: PMC7660656 DOI: 10.3390/ijms21217892] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/17/2020] [Accepted: 10/19/2020] [Indexed: 01/03/2023] Open
Abstract
Melanoma is notoriously resistant to current cancer therapy. However, the chemoresistance mechanism of melanoma remains unclear. The present study unveiled that chemotherapy drug cisplatin induced the formation of giant cells, which exhibited enlargement in cell diameter and nucleus in mice and human melanoma cells. Giant cells were positive with melanoma maker S100 and cancer stem cell markers including ABCB5 and CD133 in vitro and in vivo. Moreover, giant cells retained the mitotic ability with expression of proliferation marker Ki-67 and exhibited multiple drug resistance to doxorubicin and actinomycin D. The mitochondria genesis/activities and cellular ATP level were significantly elevated in giant cells, implicating the demand for energy supply. Application of metabolic blockers such as sodium azide or 2-deoxy glucose abolished the cisplatin-induced giant cells formation and expression of cancer stemness markers. The present study unveils a novel chemoresistance mechanism of melanoma cells via size alteration and the anti-neoplastic strategy by targeting giant cells.
Collapse
Affiliation(s)
- Chien-Hui Weng
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan;
| | - Chieh-Shan Wu
- Department of Dermatology, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan;
- Department of Dermatology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Jian-Ching Wu
- Biobank and Tissue Bank, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
| | - Mei-Lang Kung
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan;
| | - Ming-Hsiu Wu
- Department of Nutrition and Health Science, Fooyin University, Kaohsiung 83102, Taiwan
- Correspondence: (M.-H.W.); (M.-H.T.)
| | - Ming-Hong Tai
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan;
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
- Correspondence: (M.-H.W.); (M.-H.T.)
| |
Collapse
|
10
|
Han SY, Lee EM, Kim S, Kwon AM, Baek EJ. Role of Plasma Gelsolin Protein in the Final Stage of Erythropoiesis and in Correction of Erythroid Dysplasia In Vitro. Int J Mol Sci 2020; 21:ijms21197132. [PMID: 32992584 PMCID: PMC7583768 DOI: 10.3390/ijms21197132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/23/2020] [Accepted: 09/25/2020] [Indexed: 11/16/2022] Open
Abstract
Gelsolin, an actin-remodeling protein, is involved in cell motility, cytoskeletal remodeling, and cytokinesis and is abnormally expressed in many cancers. Recently, human recombinant plasma gelsolin protein (pGSN) was reported to have important roles in cell cycle and maturation of primary erythroblasts. However, the role of human plasma gelsolin in late stage erythroblasts prior to enucleation and putative clinical relevance in patients with myelodysplastic syndrome (MDS) and hemato-oncologic diseases have not been reported. Polychromatic and orthochromatic erythroblasts differentiated from human cord blood CD34+ cells, and human bone marrow (BM) cells derived from patients with MDS, were cultured in serum-free medium containing pGSN. Effects of pGSN on mitochondria, erythroid dysplasia, and enucleation were assessed in cellular and transcriptional levels. With pGSN treatment, terminal maturation at the stage of poly- and ortho-chromatic erythroblasts was enhanced, with higher numbers of orthochromatic erythroblasts and enucleated red blood cells (RBCs). pGSN also significantly decreased dysplastic features of cell morphology. Moreover, we found that patients with MDS with multi-lineage dysplasia or with excess blasts-1 showed significantly decreased expression of gelsolin mRNA (GSN) in their peripheral blood. When BM erythroblasts of MDS patients were cultured with pGSN, levels of mRNA transcripts related to terminal erythropoiesis and enucleation were markedly increased, with significantly decreased erythroid dysplasia. Moreover, pGSN treatment enhanced mitochondrial transmembrane potential that is unregulated in MDS and cultured cells. Our findings demonstrate a key role for plasma gelsolin in erythropoiesis and in gelsolin-depleted MDS patients, and raises the possibility that pGSN administration may promote erythropoiesis in erythroid dysplasia.
Collapse
Affiliation(s)
- So Yeon Han
- Department of Laboratory Medicine, College of Medicine, Hanyang University, Seoul 04763, Korea; (S.Y.H.); (S.K.)
- Department of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea;
| | - Eun Mi Lee
- Department of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea;
| | - Suyeon Kim
- Department of Laboratory Medicine, College of Medicine, Hanyang University, Seoul 04763, Korea; (S.Y.H.); (S.K.)
| | - Amy M. Kwon
- Biostatistical Consulting and Research Laboratory, Medical Research Collaborating Center, Industry-University Cooperation Foundation, Hanyang University, Seoul 04763, Korea;
| | - Eun Jung Baek
- Department of Laboratory Medicine, College of Medicine, Hanyang University, Seoul 04763, Korea; (S.Y.H.); (S.K.)
- Department of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea;
- Correspondence: ; Tel.: +82-31-560-2485; Fax: +82-31-560-2489
| |
Collapse
|
11
|
Li N, Zhan X. MASS SPECTROMETRY-BASED MITOCHONDRIAL PROTEOMICS IN HUMAN OVARIAN CANCERS. MASS SPECTROMETRY REVIEWS 2020; 39:471-498. [PMID: 32020673 DOI: 10.1002/mas.21618] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 01/22/2020] [Indexed: 06/10/2023]
Abstract
The prominent characteristics of mitochondria are highly dynamic and regulatory, which have crucial roles in cell metabolism, biosynthetic, senescence, apoptosis, and signaling pathways. Mitochondrial dysfunction might lead to multiple serious diseases, including cancer. Therefore, identification of mitochondrial proteins in cancer could provide a global view of tumorigenesis and progression. Mass spectrometry-based quantitative mitochondrial proteomics fulfils this task by enabling systems-wide, accurate, and quantitative analysis of mitochondrial protein abundance, and mitochondrial protein posttranslational modifications (PTMs). Multiple quantitative proteomics techniques, including isotope-coded affinity tag, stable isotope labeling with amino acids in cell culture, isobaric tags for relative and absolute quantification, tandem mass tags, and label-free quantification, in combination with different PTM-peptide enrichment methods such as TiO2 enrichment of tryptic phosphopeptides and antibody enrichment of other PTM-peptides, increase flexibility for researchers to study mitochondrial proteomes. This article reviews isolation and purification of mitochondria, quantitative mitochondrial proteomics, quantitative mitochondrial phosphoproteomics, mitochondrial protein-involved signaling pathway networks, mitochondrial phosphoprotein-involved signaling pathway networks, integration of mitochondrial proteomic and phosphoproteomic data with whole tissue proteomic and transcriptomic data and clinical information in ovarian cancers (OC) to in-depth understand its molecular mechanisms, and discover effective mitochondrial biomarkers and therapeutic targets for predictive, preventive, and personalized treatment of OC. This proof-of-principle model about OC mitochondrial proteomics is easily implementable to other cancer types. © 2020 John Wiley & Sons Ltd. Mass Spec Rev.
Collapse
Affiliation(s)
- Na Li
- University Creative Research Initiatives Center, Shandong First Medical University, Shandong, 250062, P. R. China
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, P. R. China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, P. R. China
| | - Xianquan Zhan
- University Creative Research Initiatives Center, Shandong First Medical University, Shandong, 250062, P. R. China
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, P. R. China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, P. R. China
- Department of Oncology, Xiangya Hospital, Central South University, 88 Xiangya Road, Changsha, Hunan, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 88 Xiangya Road, Changsha, Hunan, 410008, P. R. China
| |
Collapse
|
12
|
Kennedy AL, Rai R, Isingizwe ZR, Zhao YD, Lightfoot SA, Benbrook DM. Complementary Targeting of Rb Phosphorylation and Growth in Cervical Cancer Cell Cultures and a Xenograft Mouse Model by SHetA2 and Palbociclib. Cancers (Basel) 2020; 12:cancers12051269. [PMID: 32429557 PMCID: PMC7281234 DOI: 10.3390/cancers12051269] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/08/2020] [Accepted: 05/15/2020] [Indexed: 12/11/2022] Open
Abstract
Cervical cancer is caused by high-risk human papillomavirus (HPV) types and treated with conventional chemotherapy with surgery and/or radiation. HPV E6 and E7 proteins increase phosphorylation of retinoblastoma (Rb) by cyclin D1/cyclin dependent kinase (CDK)4/6 complexes. We hypothesized that cyclin D1 degradation by the SHetA2 drug in combination with palbociclib inhibition of CDK4/6 activity synergistically reduces phosphorylated Rb (phospho-Rb) and inhibits cervical cancer growth. The effects of these drugs, alone, and in combination, were evaluated in SiHa and CaSki HPV-positive and C33A HPV-negative cervical cancer cell lines using cell culture, western blots and ELISA, and in a SiHa xenograft model. Endpoints were compared by isobolograms, ANOVA, and Chi-Square. In all cell lines, combination indexes documented synergistic interaction of SHetA2 and palbociclib in association SHetA2 reduction of cyclin D1 and phospho-Rb, palbociclib reduction of phospho-Rb, and enhanced phospho-Rb reduction upon drug combination. Both drugs significantly reduced phospho-Rb and growth of SiHa xenograft tumors as single agents and acted additively when combined, with no evidence of toxicity. Dilated CD31-negative blood vessels adjacent to, or within, areas of necrosis and apoptosis were observed in all drug-treated tumors. These results justify development of the SHetA2 and palbociclib combination for targeting phospho-Rb in cervical cancer treatment.
Collapse
Affiliation(s)
- Amy L. Kennedy
- Department of Pathology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Rajani Rai
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Zitha Redempta Isingizwe
- Department of Pharmaceutical Sciences, College of Pharmacy University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Yan Daniel Zhao
- Department of Biostatistics and Epidemiology, College of Public Health University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Stanley A. Lightfoot
- Center for Cancer Prevention and Drug Development, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Doris M. Benbrook
- Department of Pathology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Department of Pharmaceutical Sciences, College of Pharmacy University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Department of Biostatistics and Epidemiology, College of Public Health University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Center for Cancer Prevention and Drug Development, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Correspondence:
| |
Collapse
|
13
|
Huang H, Chen AY, Ye X, Guan R, Rankin GO, Chen YC. Galangin, a Flavonoid from Lesser Galangal, Induced Apoptosis via p53-Dependent Pathway in Ovarian Cancer Cells. Molecules 2020; 25:molecules25071579. [PMID: 32235536 PMCID: PMC7180956 DOI: 10.3390/molecules25071579] [Citation(s) in RCA: 20] [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: 02/24/2020] [Revised: 03/23/2020] [Accepted: 03/27/2020] [Indexed: 12/12/2022] Open
Abstract
Among women worldwide, ovarian cancer is one of the most dangerous cancers. Patients undergoing platinum-based chemotherapy might get adverse side effects and develop resistance to drugs. In recent years, natural compounds have aroused growing attention in cancer treatment. Galangin inhibited the growth of two cell lines, A2780/CP70 and OVCAR-3, more strongly than the growth of a normal ovarian cell line, IOSE 364. The IC50 values of galangin on proliferation of A2780/CP70, OVCAR-3 and IOSE 364 cells were 42.3, 34.5, and 131.3 μM, respectively. Flow cytometry analysis indicated that galangin preferentially induced apoptosis in both ovarian cancer cells with respect to normal ovarian cells. Galangin treatment increased the level of cleaved caspase-3 and -7 via the p53-dependent intrinsic apoptotic pathway by up-regulating Bax protein and via the p53-dependent extrinsic apoptotic pathway by up-regulating DR5 protein. By down-regulating the level of p53 with 20 μM pifithrin-α (PFT-α), the apoptotic rates of OVCAR-3 cells induced by galangin treatment (40 μM) were significantly decreased from 18.2% to 10.2%, indicating that p53 is a key regulatory protein in galangin-induced apoptosis in ovarian cancer cells. Although galangin up-regulated the expression of p21, it had little effect on the cell cycle of the two ovarian cancer cell lines. Furthermore, the levels of phosphorylated Akt and phosphorylated p70S6K were decreased through galangin treatment, suggesting that the Akt/p70S6K pathways might be involved in the apoptosis. Our results suggested that galangin is selective against cancer cells and can be used for the treatment of platinum-resistant ovarian cancers in humans.
Collapse
Affiliation(s)
- Haizhi Huang
- College of Life Sciences, China Jiliang University, Hangzhou 310018, China;
- College of Science, Technology & Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA
| | - Allen Y. Chen
- Department of Pharmacy Informatics, Seattle Children’s Hospital, Seattle, WA 98101, USA;
| | - Xingqian Ye
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang University, Hangzhou 310058, China;
| | - Rongfa Guan
- College of Food Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China;
| | - Gary O. Rankin
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA;
| | - Yi Charlie Chen
- College of Science, Technology & Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA
- Correspondence: ; Tel.: +1-304-457-6277; Fax: +1-304-457-6239
| |
Collapse
|
14
|
Sharma A, Li M, Thavathiru E, Ibrahim M, Garcia-Contreras L, Benbrook DM, Woo S. Physiologically Based Pharmacokinetic Modeling and Tissue Distribution Characteristics of SHetA2 in Tumor-Bearing Mice. AAPS JOURNAL 2020; 22:51. [PMID: 32086622 DOI: 10.1208/s12248-020-0421-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/08/2020] [Indexed: 12/11/2022]
Abstract
The orally available novel small molecule SHetA2 is the lead sulfur-containing heteroarotinoid that selectively inhibits cancer cells over normal cells, and is currently under clinical development for anticancer treatment and cancer prevention. The objective of this study was to assess and characterize the tissue distribution of SHetA2 in tumor-bearing mice by developing a physiologically based pharmacokinetic (PBPK) model. An orthotopic SKOV3 ovarian cancer xenograft mouse model was used to most accurately mimic the ovarian cancer tumor microenvironment in the peritoneal cavity. SHetA2 concentrations in plasma and 14 different tissues were measured at various time points after a single intravenous dose of 10 mg/kg and oral dose of 60 mg/kg, and these data were used to develop a whole-body PBPK model. SHetA2 exhibited a multi-exponential plasma concentration decline with an elimination half-life of 4.5 h. Rapid and extensive tissue distribution, which was best described by a perfusion rate-limited model, was observed with the tissue-to-plasma partition coefficients (kp = 1.4-21.2). The PBPK modeling estimated the systemic clearance (76.4 mL/h) from circulation as a main elimination pathway of SHetA2. It also indicated that the amount absorbed into intestine was the major determining factor for the oral bioavailability (22.3%), while the first-pass loss from liver and intestine contributed minimally (< 1%). Our results provide an insight into SHetA2 tissue distribution characteristics. The developed PBPK model can be used to predict the drug exposure at tumors or local sites of action for different dosing regimens and scaled up to humans to correlate with efficacy.
Collapse
Affiliation(s)
- Ankur Sharma
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Mengjie Li
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Elangovan Thavathiru
- Department of Obstetrics and Gynecology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Mariam Ibrahim
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Lucila Garcia-Contreras
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Doris M Benbrook
- Department of Obstetrics and Gynecology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA.,Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Sukyung Woo
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA. .,Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA.
| |
Collapse
|
15
|
Ramraj SK, Elayapillai SP, Pelikan RC, Zhao YD, Isingizwe ZR, Kennedy AL, Lightfoot SA, Benbrook DM. Novel ovarian cancer maintenance therapy targeted at mortalin and mutant p53. Int J Cancer 2020; 147:1086-1097. [PMID: 31845320 DOI: 10.1002/ijc.32830] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 11/26/2019] [Accepted: 12/06/2019] [Indexed: 12/22/2022]
Abstract
Current ovarian cancer maintenance therapy is limited by toxicity and no proven impact on overall survival. To study a maintenance strategy targeted at missense mutant p53, we hypothesized that the release of mutant p53 from mortalin inhibition by the SHetA2 drug combined with reactivation of mutant p53 with the PRIMA-1MET drug inhibits growth and tumor establishment synergistically in a mutant-p53 dependent manner. The Cancer Genome Atlas (TCGA) data and serous ovarian tumors were evaluated for TP53 and HSPA9/mortalin status. SHetA2 and PRIMA-1MET were tested in ovarian cancer cell lines and fallopian tube secretory epithelial cells using isobolograms, fluorescent cytometry, Western blots and ELISAs. Drugs were administered to mice after peritoneal injection of MESOV mutant p53 ovarian cancer cells and prior to tumor establishment, which was evaluated by logistic regression. Fifty-eight percent of TP53 mutations were missense and there were no mortalin mutations in TCGA high-grade serous ovarian cancers. Mortalin levels were sequentially increased in serous benign, borderline and carcinoma tumors. SHetA2 caused p53 nuclear and mitochondrial accumulation in cancer, but not in healthy, cells. Endogenous or exogenous mutant p53 increased SHetA2 resistance. PRIMA-1MET decreased this resistance and interacted synergistically with SHetA2 in mutant and wild type p53-expressing cell lines in association with elevated reactive oxygen species/ATP ratios. Tumor-free rates in animals were 0% (controls), 25% (PRIMA1MET ), 42% (SHetA2) and 67% (combination). SHetA2 (p = 0.004) and PRIMA1MET (p = 0.048) functioned additively in preventing tumor development with no observed toxicity. These results justify the development of SHetA2 and PRIMA-1MET alone and in combination for ovarian cancer maintenance therapy.
Collapse
Affiliation(s)
- Satish K Ramraj
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Sugantha P Elayapillai
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Richard C Pelikan
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Yan D Zhao
- Biostatistics & Epidemiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Zitha R Isingizwe
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Amy L Kennedy
- Department of Pathology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Stanley A Lightfoot
- Center for Cancer Prevention and Drug Development, Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Doris M Benbrook
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK.,Center for Cancer Prevention and Drug Development, Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK.,Obstetrics and Gynecologic, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
16
|
Tetrahydroquinoline units in flexible heteroarotinoids (Flex-Hets) convey anti-cancer properties in A2780 ovarian cancer cells. Bioorg Med Chem 2019; 28:115244. [PMID: 31831296 DOI: 10.1016/j.bmc.2019.115244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 11/26/2019] [Accepted: 11/26/2019] [Indexed: 01/25/2023]
Abstract
SHetA2 (NSC 721689), our lead Flex-Het anti-cancer agent, consists of a thiochroman (Ring A) and a 4-nitrophenyl (Ring B) linked by a thiourea bridge. In this work, several series of new analogs having a tetrahydroquinoline (THQ, Ring A) unit connected by a urea or thiourea linker to a 4-substituted phenyl (Ring B) have been prepared and evaluated relative to SHetA2 in terms of binding affinity with mortalin and inhibition of A2780 ovarian cancer cells. Six of the derivatives equaled or exceeded the efficacy shown by SHetA2. Compounds 1a-d (series 1), lacking a methyl on the Ring A nitrogen and the gem-dimethyls on the adjacent carbon, showed only weak activity. Salt 2, the quaternized N,N-dimethyl iodide salt analog of 1a, also possessed very modest growth inhibition in the cell line studied. Series 3 compounds, which had a C3 ketone and an N-methyl replacing the sulfur in Ring A, were most successful. Compound 3a [Ring A = 1,2,2,4,4-pentamethyl-3-oxo-1,2,3,4-tetrahydroquinolin-6-yl; urea linker; Ring B = 4-nitrophenyl] had slightly lower potency (IC50 3.8 μM), but better efficacy (94.8%) than SHetA2 (IC50 3.17 μM, efficacy 84.3%). In addition, 3c and 3d [urea and thiourea linkers, respectively; Ring B = 4-(trifluoromethyl)phenyl] and 3e and 3f [urea and thiourea linkers, respectively; Ring B = 4-(trifluoromethoxy)phenyl] were also evaluated since these agents possessed electron-withdrawing groups with H-bonding capability. All displayed good activity. Compounds 3c and 3e showed improvement in both potency and efficacy compared to SHetA2. In general, when the linker group between Rings A and B was a urea, efficacy values slightly exceeded those with a thiourea linker in the carbonyl-containing THQ systems 3a-g. In contrast, when Ring A possessed the 1,2,2,4,4-pentamethyl-3-hydroxytetrahydroquinolin-6-yl unit (4a-f, series 4), very modest potency and efficacy was observed. Model compound 5, an exact N-methyl THQ analog of SHetA2, demonstrated less potency (IC50 4.5 μM), but improved efficacy (91.7%). Modeling studies were performed to rationalize the observed results.
Collapse
|
17
|
Fong SY, Wimalasiri D, Piva T, Dekiwadia C, Urban S, Huynh T. Evaluation of cytotoxic and apoptotic activities of Clinacanthus nutans (Burm. f.) Lindau leaves against D24 human melanoma cells. J Herb Med 2019. [DOI: 10.1016/j.hermed.2019.100285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
18
|
Zou H, Sevigny MB, Liu S, Madden DT, Louie MC. Novel flexible heteroarotinoid, SL-1-39, inhibits HER2-positive breast cancer cell proliferation by promoting lysosomal degradation of HER2. Cancer Lett 2019; 443:157-166. [PMID: 30503556 DOI: 10.1016/j.canlet.2018.11.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/21/2018] [Accepted: 11/24/2018] [Indexed: 10/27/2022]
Abstract
SL-1-39 [1-(4-chloro-3-methylphenyl)-3-(4-nitrophenyl)thiourea] is a new flexible heteroarotinoid (Flex-Het) analog derived from the parental compound, SHetA2, previously shown to inhibit cell growth across multiple cancer types. The current study aims to determine growth inhibitory effects of SL-1-39 across the different subtypes of breast cancer cells and delineate its molecular mechanism. Our results demonstrate that while SL-1-39 blocks cell proliferation of all breast cancer subtypes tested, it has the highest efficacy against HER2+ breast cancer cells. Molecular analyses suggest that SL-1-39 prevents S phase progression of HER2+ breast cancer cells (SKBR3 and MDA-MB-453), which is consistent with reduced expression of key cell-cycle regulators at both the protein and transcriptional levels. SL-1-39 treatment also decreases the protein levels of HER2 and pHER2 as well as its downstream effectors, pMAPK and pAKT. Reduction of HER2 and pHER2 at the protein level is attributed to increased lysosomal degradation of total HER2 levels. This is the first study to show that a flexible heteroarotinoid analog modulates the HER2 signaling pathway through lysosomal degradation, and thus further warrants the development of SL-1-39 as a therapeutic option for HER2+ breast cancer.
Collapse
Affiliation(s)
- Hongye Zou
- Department of Natural Sciences and Mathematics, Dominican University of California, 50 Acacia Avenue, San Rafael, CA, 94901, USA.
| | - Mary B Sevigny
- Department of Natural Sciences and Mathematics, Dominican University of California, 50 Acacia Avenue, San Rafael, CA, 94901, USA.
| | - Shengquan Liu
- College of Pharmacy, Touro University California, 1310 Club Drive, Vallejo, CA, 94594, USA.
| | - David T Madden
- College of Pharmacy, Touro University California, 1310 Club Drive, Vallejo, CA, 94594, USA; Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, 94945, USA.
| | - Maggie C Louie
- Department of Natural Sciences and Mathematics, Dominican University of California, 50 Acacia Avenue, San Rafael, CA, 94901, USA; College of Pharmacy, Touro University California, 1310 Club Drive, Vallejo, CA, 94594, USA.
| |
Collapse
|
19
|
Zhang X, Zhuang R. Dione-thiophene conjugate inhibits proliferation and metastasis of nasopharyngeal carcinoma cells through calcium binding protein-P down-regulation. Eur J Med Chem 2019; 168:199-206. [PMID: 30822709 DOI: 10.1016/j.ejmech.2019.01.051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/17/2019] [Accepted: 01/18/2019] [Indexed: 11/30/2022]
Abstract
In the present study a series of dione-thiophenol conjugates was prepared and evaluated against nasopharyngeal carcinoma (NPC) cells. MTT assay showed that compound 4a reduced proliferation of C666-1 and CNE-1 cells to 26 and 24%, respectively at 10 μmol/l concentration. Flow cytometry revealed that increasing the concentration of compound 4a from 2 to 10 μmol/l increased the proportion of early apoptotic C666-1 cells from 2.76 to 69.43%. A significant (P < 0.001) decrease in the expression of S100P was caused by compound 4a. In compound 4a treated C666-1 cells the expression of RAGE, EGFR, CD44, MMP2 and MMP9 was markedly decreased. In summary, compound 4a inhibits nasopharyngeal cancer cell proliferation and induces apoptosis through down-regulation of S100P. Moreover, compound 4a also decreases MMP-2, MMP-9, EGFR, CD44 and RAGE expression in nasopharyngeal cancer cells. Thus, compound 4a can be investigated further as a drug candidate for the treatment of nasopharyngeal cancer.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Oral and Maxillofacial Surgery, Hanzhong Central Hospital, Hanzhong, Shaanxi, 723000, China
| | - Rui Zhuang
- Department of Oral and Maxillofacial Surgery, Hanzhong Central Hospital, Hanzhong, Shaanxi, 723000, China.
| |
Collapse
|
20
|
Ibrahim M, Hatipoglu MK, Garcia-Contreras L. Cryogenic Fabrication of Dry Powders to Enhance the Solubility of a Promising Anticancer Drug, SHetA2, for Oral Administration. AAPS PharmSciTech 2019; 20:20. [PMID: 30604109 DOI: 10.1208/s12249-018-1204-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/31/2018] [Indexed: 01/29/2023] Open
Abstract
SHetA2 is a novel anticancer drug with poor aqueous solubility. In formal toxicological studies, Kolliphor HS 15 was used as a solubilizing agent to increase the oral bioavailability of SHetA2. The purpose of this study was to formulate SHetA2 and Kolliphor HS 15 as solid powders to facilitate their filling in hard gelatin capsules for clinical trials. Two manufacturing processes, ultra-rapid freeze-drying (URFD) and spray freeze drying (SFD), were employed to fabricate solid powders of SHetA2-Kolliphor HS 15 and trehalose. The morphology, size, flowability, and compressibility of URFD-SHetA2 and SFD-SHetA2 powders were characterized. The crystallinity and apparent maximum solubility of SHetA2 in both powders were also determined. SFD-SHetA2 powders were spherical in shape, small, and with a wide size distribution while the URFD-SHetA2 powders were irregularly shaped and big but with a narrower distribution. DSC and XRD analyses indicated that SHetA2 was mostly amorphous in both powders. The flow of both powders was categorized as "good" (angle of repose < 35°). The uniformity of drug content in URFD-SHetA2 powders was more variable than that in SFD-SHetA2 powders. The solubility profile of SHetA2 in both powders SGF exhibited a transient supersaturation "spring effect" due to the drug's amorphousness followed by extended supersaturation "parachute effect" at approximately 6 μg/ml for both powders compared to 0.02 ± 0.01 μg/ml for unprocessed drug. In conclusion, both URFD and SFD formed solid SHetA2 Kolliphor powders that are possible formulation candidates to be filled in hard gelatin capsules for clinical trials.
Collapse
|
21
|
Mahjabeen S, Hatipoglu MK, Benbrook DM, Garcia-Contreras L. Pharmacokinetics and Pharmacodynamics of Escalating Doses of SHetA2 After Vaginal Administration to Mice. J Pharm Sci 2018; 107:3179-3186. [PMID: 30196041 PMCID: PMC6342475 DOI: 10.1016/j.xphs.2018.08.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 11/25/2022]
Abstract
SHetA2 is a novel compound with strong potential to treat cervical dysplasia, but its low aqueous solubility limits its oral bioavailability. A vaginal suppository achieved SHetA2 cervix concentrations that were severalfold above the predicted therapeutic levels. Thus, we aimed at determining the minimum dose that would achieve SHetA2 therapeutic levels while reducing cyclin D1 levels, the pharmacodynamic end point. The disposition of SHetA2 after vaginal administration of escalating SHetA2 doses and the corresponding reduction in cyclin D1 levels was compared to that after the conventional oral treatment. Vaginal administration of a 15-mg/kg dose achieved an area under the cervix concentration versus time curve (AUCcervix) that was ∼120 times larger than that after a 60 mg/kg administered orally. AUCcervix and Cmax-cervix did not increase proportionally with respect to the dose, with the 30-mg/kg dose resulting in higher AUCcervix and Cmax-cervix (1368.53 μg.mL/h and 155.38 μg/g, respectively) compared to the 15 mg/kg (334.98 μg.mL/h and 121.78 μg/g, respectively) or 60 mg/kg (1178.55 μg.mL/h and 410.38 μg/g, respectively). Likewise, the 30-mg/kg dose caused a larger reduction in cyclin D1 levels than the other doses. Thus, the 30-mg/kg dose was selected for future efficacy studies in a mouse model of cervical neoplasia.
Collapse
Affiliation(s)
- Sanjida Mahjabeen
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73126-0901
| | - Manolya Kukut Hatipoglu
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73126-0901
| | - Doris M Benbrook
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73126-0901; Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73126-0901
| | - Lucila Garcia-Contreras
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73126-0901.
| |
Collapse
|
22
|
Activity of oxygen-versus sulfur-containing analogs of the Flex-Het anticancer agent SHetA2. Eur J Med Chem 2018; 158:720-732. [DOI: 10.1016/j.ejmech.2018.09.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 09/03/2018] [Accepted: 09/13/2018] [Indexed: 11/17/2022]
|
23
|
Zhong L, Tran T, Baguley TD, Lee SJ, Henke A, To A, Li S, Yu S, Grieco FA, Roland J, Schultz PG, Eizirik DL, Rogers N, Chartterjee AK, Tremblay MS, Shen W. A novel inhibitor of inducible NOS dimerization protects against cytokine-induced rat beta cell dysfunction. Br J Pharmacol 2018; 175:3470-3485. [PMID: 29888783 PMCID: PMC6086989 DOI: 10.1111/bph.14388] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 05/14/2018] [Accepted: 05/28/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE Beta cell apoptosis is a major feature of type 1 diabetes, and pro-inflammatory cytokines are key drivers of the deterioration of beta cell mass through induction of apoptosis. Mitochondrial stress plays a critical role in mediating apoptosis by releasing cytochrome C into the cytoplasm, directly activating caspase-9 and its downstream signalling cascade. We aimed to identify new compounds that protect beta cells from cytokine-induced activation of the intrinsic (mitochondrial) pathway of apoptosis. EXPERIMENTAL APPROACH Diabetogenic media, composed of IL-1β, IFN-γ and high glucose, were used to induce mitochondrial stress in rat insulin-producing INS1E cells, and a high-content image-based screen of small molecule modulators of Casp9 pathway was performed. KEY RESULTS A novel small molecule, ATV399, was identified from a high-content image-based screen for compounds that inhibit cleaved caspase-9 activation and subsequent beta cell apoptosis induced by a combination of IL-1β, IFN-γ and high glucose, which together mimic the pathogenic diabetic milieu. Through medicinal chemistry optimization, potency was markedly improved (6-30 fold), with reduced inhibitory effects on CYP3A4. Improved analogues, such as CAT639, improved beta cell viability and insulin secretion in cytokine-treated rat insulin-producing INS1E cells and primary dispersed islet cells. Mechanistically, CAT639 reduced the production of NO by allosterically inhibiting dimerization of inducible NOS (iNOS) without affecting its mRNA levels. CONCLUSION AND IMPLICATIONS Taken together, these studies demonstrate a successful phenotypic screening campaign resulting in identification of an inhibitor of iNOS dimerization that protects beta cell viability and function through modulation of mitochondrial stress induced by cytokines.
Collapse
Affiliation(s)
- Linlin Zhong
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Tuan Tran
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Tyler D Baguley
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Sang Jun Lee
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Adam Henke
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Andrew To
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Sijia Li
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Shan Yu
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Fabio A Grieco
- ULB Center for Diabetes ResearchUniversite´ Libre de Bruxelles (ULB)Brussels1070Belgium
| | - Jason Roland
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Peter G Schultz
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
- Department of ChemistryThe Scripps Research InstituteLa JollaCA92037USA
| | - Decio L Eizirik
- ULB Center for Diabetes ResearchUniversite´ Libre de Bruxelles (ULB)Brussels1070Belgium
| | - Nikki Rogers
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | | | | | - Weijun Shen
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| |
Collapse
|
24
|
Sharma A, Benbrook DM, Woo S. Pharmacokinetics and interspecies scaling of a novel, orally-bioavailable anti-cancer drug, SHetA2. PLoS One 2018; 13:e0194046. [PMID: 29634717 PMCID: PMC5892888 DOI: 10.1371/journal.pone.0194046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 02/25/2018] [Indexed: 01/08/2023] Open
Abstract
SHetA2 is a small molecule drug with promising cancer prevention and therapeutic activity and a high preclinical safety profile. The study objectives were to perform interspecies scaling and pharmacokinetic (PK) modeling of SHetA2 for human PK prediction. The PK data obtained from mice, rats, and dogs after intravenous and oral doses were used for simultaneous fitting to PK models. The disposition of SHetA2 was best described by a two-compartment model. The absorption kinetics was well characterized with a first-order absorption model for mice and rats, and a gastrointestinal transit model for dogs. Oral administration of SHetA2 showed a relatively fast absorption in mice, prolonged absorption (i.e., flip-flop kinetics) toward high doses in rats, and an early peak followed by a secondary peak at high doses in dogs. The oral bioavailability was 17.7-19.5% at 20-60 mg/kg doses in mice, <1.6% at 100-2000 mg/kg in rats, and 11.2% at 100 mg/kg decreasing to 3.45% at 400 mg/kg and 1.11% at 1500 mg/kg in dogs. The disposition parameters were well correlated with the body weight for all species using the allometric equation, which predicted values of CL (17.3 L/h), V1 (36.2 L), V2 (68.5 L) and CLD (15.2 L/h) for a 70-kg human. The oral absorption rate and bioavailability of SHetA2 was highly dependent on species, doses, formulations, and possibly other factors. The limited bioavailability at high doses was taken into consideration for the suggested first-in-human dose, which was much lower than the dose estimated based on toxicology studies. In summary, the present study provided the PK model for SHetA2 that depicted the disposition and absorption kinetics in preclinical species, and computational tools for human PK prediction.
Collapse
Affiliation(s)
- Ankur Sharma
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Doris Mangiaracina Benbrook
- Department of Obstetrics and Gynecology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Sukyung Woo
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| |
Collapse
|
25
|
Benbrook DM, Janakiram NB, Chandra V, Pathuri G, Madka V, Stratton NC, Masamha CP, Farnsworth CN, Garcia-Contreras L, Hatipoglu MK, Lighfoot S, Rao CV. Development of a dietary formulation of the SHetA2 chemoprevention drug for mice. Invest New Drugs 2017; 36:561-570. [PMID: 29273857 PMCID: PMC6014882 DOI: 10.1007/s10637-017-0550-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 12/07/2017] [Indexed: 12/17/2022]
Abstract
Development of cancer chemoprevention compounds requires enhanced consideration for toxicity and route of administration because the target population is healthy. The small molecule drug, SHetA2 (NSC 726189), exhibited in vivo chemoprevention activity and lack of toxicity when administered by oral gavage. Our objective was to determine if a dietary formulation of SHetA2 could achieve effective tissue drug levels without toxicity. C57bl/6 J mice were monitored on modified American Institute of Nutrition (AIN)76A diet mixed with SHetA2 in a 3:1 ratio with Kolliphor HS15, a self-emulsifying drug delivery system (SEDDS) to deliver 37.5, 62.5, 125, 187 or 250 mg SHetA2/kg/day. Blood and tissues were evaluated after 1, 3 and 6 weeks. The 187 mg/kg/day dose was identified as optimal based on achievement of maximum blood and tissue drug levels in the effective micromolar range without evidence of toxicity. The 250 mg/kg/day group exhibited lower drug levels and the highest intestinal drug content suggesting that an upper limit of intestinal absorption had been surpassed. Only this highest dose resulted in liver and kidney function tests that were outside of the normal range, and significant reduction of cyclin D1 protein in normal cervical tissue. SHetA2 reduced cyclin D1 to greater extents in cancer compared to non-cancer cell cultures. Given this differential effect, optimal chemoprevention without toxicity would be expected to occur at doses that reduced cyclin D1 in neoplastic, but not in normal tissues. These findings support further development of SHetA2 as a chemoprevention agent and potential food additive.
Collapse
Affiliation(s)
- Doris M Benbrook
- Section of Gynecologic Oncology, Department of Obstetrics and Gynecology, College of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th St., BRC 1217A, Oklahoma City, OK, 73104, USA. .,Center for Cancer Prevention and Drug Development, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th St., Oklahoma City, OK, 73104, USA.
| | - Naveena B Janakiram
- Center for Cancer Prevention and Drug Development, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th St., Oklahoma City, OK, 73104, USA.,Hematologic Oncology Section, College of Medicine, Center for Cancer Prevention and Drug Development, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th St., Oklahoma City, OK, 73104, USA.,VA Medical Center, Oklahoma City, OK, 73104, USA
| | - Vishal Chandra
- Section of Gynecologic Oncology, Department of Obstetrics and Gynecology, College of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th St., BRC 1217A, Oklahoma City, OK, 73104, USA.,Center for Cancer Prevention and Drug Development, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th St., Oklahoma City, OK, 73104, USA
| | - Gopal Pathuri
- Center for Cancer Prevention and Drug Development, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th St., Oklahoma City, OK, 73104, USA.,Hematologic Oncology Section, College of Medicine, Center for Cancer Prevention and Drug Development, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th St., Oklahoma City, OK, 73104, USA
| | - Venkateshwar Madka
- Center for Cancer Prevention and Drug Development, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th St., Oklahoma City, OK, 73104, USA.,Hematologic Oncology Section, College of Medicine, Center for Cancer Prevention and Drug Development, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th St., Oklahoma City, OK, 73104, USA
| | - Nicole C Stratton
- Center for Cancer Prevention and Drug Development, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th St., Oklahoma City, OK, 73104, USA.,Hematologic Oncology Section, College of Medicine, Center for Cancer Prevention and Drug Development, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th St., Oklahoma City, OK, 73104, USA
| | - Chioniso P Masamha
- Section of Gynecologic Oncology, Department of Obstetrics and Gynecology, College of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th St., BRC 1217A, Oklahoma City, OK, 73104, USA.,Butler University, 4600 Sunset Avenue, Indianapolis, IN, 46208, USA
| | | | - Lucila Garcia-Contreras
- Department of Pharmaceutical Sciences, College of Pharmacy, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 1110 N. Stonewall, Oklahoma City, OK, 73117, USA
| | - Manolya Kukut Hatipoglu
- Department of Pharmaceutical Sciences, College of Pharmacy, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 1110 N. Stonewall, Oklahoma City, OK, 73117, USA
| | - Stan Lighfoot
- Center for Cancer Prevention and Drug Development, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th St., Oklahoma City, OK, 73104, USA
| | - Chinthalapally V Rao
- Center for Cancer Prevention and Drug Development, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th St., Oklahoma City, OK, 73104, USA.,Hematologic Oncology Section, College of Medicine, Center for Cancer Prevention and Drug Development, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th St., Oklahoma City, OK, 73104, USA.,VA Medical Center, Oklahoma City, OK, 73104, USA
| |
Collapse
|
26
|
Wright KD, Staruschenko A, Sorokin A. Role of adaptor protein p66Shc in renal pathologies. Am J Physiol Renal Physiol 2017; 314:F143-F153. [PMID: 28978535 DOI: 10.1152/ajprenal.00414.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
p66Shc is one of the three adaptor proteins encoded by the Shc1 gene, which are expressed in many organs, including the kidney. Recent studies shed new light on several key questions concerning the signaling mechanisms mediated by p66Shc. The central goal of this review article is to summarize recent findings on p66Shc and the role it plays in kidney physiology and pathology. This article provides a review of the various mechanisms whereby p66Shc has been shown to function within the kidney through a wide range of actions. The mitochondrial and cytoplasmic signaling of p66Shc, as it relates to production of reactive oxygen species (ROS) and renal pathologies, is further discussed.
Collapse
Affiliation(s)
- Kevin D Wright
- Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin.,Department of Medicine, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Alexander Staruschenko
- Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin.,Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Andrey Sorokin
- Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin.,Department of Medicine, Medical College of Wisconsin , Milwaukee, Wisconsin
| |
Collapse
|
27
|
Fallatah MM, Liu S, Sevigny MB, Zou H, Louie MC. Novel flexible heteroarotinoid, SL-1-18, promotes ERα degradation to inhibit breast cancer cell growth. Cancer Lett 2017; 408:82-91. [PMID: 28844711 DOI: 10.1016/j.canlet.2017.08.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 08/11/2017] [Accepted: 08/17/2017] [Indexed: 12/16/2022]
Abstract
SL-1-18 (1-(chrysen-6-yl)-3-(4-nitrophenyl)thiourea) is new flexible heteroarotinoid (Flex-Het) analog derived from the parent compound, SHetA2, and our previous study showed comparable activity to SHetA2 in terms of inhibiting ER+ breast cancer cell growth. This current study aims to determine the molecular mechanism underlying SL-1-18's effect on breast cancer cell growth. Our results indicate that SL-1-18 inhibits cell proliferation of ER+ breast cancer cells (MCF-7 and T-47D) by preventing cell cycle progression. SL-1-18 treatment correlated positively with decreased expression of key cell-cycle regulators, such as cyclin D1, as well as other ERα-target genes at both the transcript and protein levels. Interestingly, decreased expression of ERα was also observed, with a significant reduction at the protein level within 2 h of SL-1-18 treatment, while the decrease in mRNA occurred at a later time point. ERα degradation was shown to be mediated by the ubiquitination-proteasome pathway. In summary, this is the first study to show that a Flex-Het- SL-1-18- can promote the degradation of ERα via the ubiquitin-proteasome pathway and should be further developed as a therapeutic option for ER+ breast cancer.
Collapse
Affiliation(s)
- Maryam M Fallatah
- Department of Natural Sciences and Mathematics, Dominican University of California, 50 Acacia Avenue, San Rafael, CA 94901, USA
| | - Shengquan Liu
- College of Pharmacy, Touro University of California, 1310 Club Drive, Vallejo, CA 94594, USA
| | - Mary B Sevigny
- Department of Natural Sciences and Mathematics, Dominican University of California, 50 Acacia Avenue, San Rafael, CA 94901, USA
| | - Hongye Zou
- Department of Natural Sciences and Mathematics, Dominican University of California, 50 Acacia Avenue, San Rafael, CA 94901, USA
| | - Maggie C Louie
- Department of Natural Sciences and Mathematics, Dominican University of California, 50 Acacia Avenue, San Rafael, CA 94901, USA; College of Pharmacy, Touro University of California, 1310 Club Drive, Vallejo, CA 94594, USA.
| |
Collapse
|
28
|
Marín-Prida J, Pardo Andreu GL, Rossignoli CP, Durruthy MG, Rodríguez EO, Reyes YV, Acosta RF, Uyemura SA, Alberici LC. The cytotoxic effects of VE-3N, a novel 1,4-dihydropyridine derivative, involve the mitochondrial bioenergetic disruption via uncoupling mechanisms. Toxicol In Vitro 2017; 42:21-30. [PMID: 28363597 DOI: 10.1016/j.tiv.2017.03.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 03/03/2017] [Accepted: 03/27/2017] [Indexed: 01/21/2023]
Abstract
Several 1,4-dihydropyridine derivatives overcome the multidrug resistance in tumors, but their intrinsic cytotoxic mechanisms remain unclear. Here we addressed if mitochondria are involved in the cytotoxicity of the novel 1,4-dihydropyridine derivative VE-3N [ethyl 6-chloro-5-formyl-2-methyl-4-(3-nitrophenyl)-1,4-dihydropyridine-3-carboxylate] towards cancer cells by employing hepatic carcinoma (HepG2) cells and isolated rat liver mitochondria. In HepG2 cells, VE-3N induced mitochondrial membrane potential dissipation, ATP depletion, annexin V/propidium iodide double labeling, and Hoechst staining; events indicating apoptosis induction. In isolated rat liver mitochondria, VE-3N promoted mitochondrial uncoupling by exerting protonophoric actions and by increasing membrane fluidity. Mitochondrial uncoupling was evidenced by an increase in resting respiration, dissipation of mitochondrial membrane potential, inhibition of Ca2+ uptake, stimulation of Ca2+ release, decrease in ATP synthesis, and swelling of valinomycin-treated organelles in hyposmotic potassium acetate media. Furthermore, uncoupling concentrations of VE-3N in the presence of Ca2+ plus ruthenium red induced the mitochondrial permeability transition process. These results indicate that mitochondrial uncoupling is potentially involved in the VE-3N cytotoxic actions towards HepG2 cells. Considering that hepatocellular carcinoma is the most common form of liver cancer, our findings may open a new avenue for the development of VE-3N-based cancer therapies, and help to unravel the cytotoxic mechanisms of 1,4-dihydropyridines towards cancer cells.
Collapse
Affiliation(s)
- Javier Marín-Prida
- Center for Research and Biological Evaluations, Institute of Pharmacy and Food, University of Havana, 222 St. # 2317, La Coronela, La Lisa, Havana, Cuba
| | - Gilberto L Pardo Andreu
- Center for Research and Biological Evaluations, Institute of Pharmacy and Food, University of Havana, 222 St. # 2317, La Coronela, La Lisa, Havana, Cuba.
| | - Camila Pederiva Rossignoli
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. Café s/n, 14040-903, Ribeirão Preto, São Paulo, Brazil
| | - Michael González Durruthy
- Center for Research and Biological Evaluations, Institute of Pharmacy and Food, University of Havana, 222 St. # 2317, La Coronela, La Lisa, Havana, Cuba
| | - Estael Ochoa Rodríguez
- Laboratory of Organic Synthesis, Faculty of Chemistry, University of Havana, Zapata st./G and Carlitos Aguirre, Vedado Plaza de la Revolución, PO 10400, Havana, Cuba
| | - Yamila Verdecia Reyes
- Laboratory of Organic Synthesis, Faculty of Chemistry, University of Havana, Zapata st./G and Carlitos Aguirre, Vedado Plaza de la Revolución, PO 10400, Havana, Cuba
| | - Roberto Fernández Acosta
- Department of Pharmacy, Institute of Pharmacy and Food, University of Havana, 222 St. # 2317, La Coronela, La Lisa, Havana, Cuba
| | - Sergio A Uyemura
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. Café s/n, 14040-903 Ribeirão Preto, São Paulo, Brazil
| | - Luciane C Alberici
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. Café s/n, 14040-903, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
29
|
Sharma A, Thavathiru E, Benbrook DM, Woo S. Bioanalytical method development and validation of HPLCUV assay for the quantification of SHetA2 in mouse and human plasma: Application to pharmacokinetics study. ACTA ACUST UNITED AC 2017; 6. [PMID: 29708233 PMCID: PMC5922436 DOI: 10.7243/2050-120x-6-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background SHetA2 is an oral anticancer agent being investigated for cancer
treatment and prevention. The aim of this study was to develop and validate
a simple, cost-effective, and sensitive HPLC-UV method for the
quantification of SHetA2 in biological samples and to apply the method to
pharmacokinetic studies of the drug. Methods Sample preparation for mouse and human plasmas involved liquid-liquid
precipitation and extraction using chilled acetonitrile with 2,
3-Diphenylquinoxaline as an internal standard. The separation of SHetA2 and
internal standard was achieved via Waters XBridge™ BEH 130 C18 (3.5
μm, 2.1×150 mm) column coupled with a Waters
XBridge™ C-18 (3.5 μm, 2.1×10 mm) guard column using
65% v/v acetonitrile: distilled water as a mobile phase in an
isocratic mode with a flow rate of 0.18 ml/min. The analytes were eluted at
a detection wavelength of 341 nm at a column temperature of
25°C. Results The method was validated across a range of 5-1000 ng/ml for SHetA2 in
plasma, with a lower limit of quantification of 5 ng/ml. The method showed
high recovery in human (79.9-81.8%) and mouse (95.4-109.2%)
plasma with no matrix effect. The intra- and inter-day accuracy and
precision studies demonstrated that the method was specific, sensitive, and
reliable. Stability studies showed that SHetA2 is stable for 20 h
postoperatively in the auto sampler, and for six weeks at -80°C in
plasma. Repetitive freezing and thawing may be avoided by preparing the
aliquots and storing them at -80°C. The developed method was
successfully applied to study the plasma pharmacokinetics of SHetA2 in
tumor-bearing nude mice after intravenous and oral administration. Conclusion A novel method for quantifying SHetA2 in mouse and human plasmas has
been validated and is being applied for pharmacokinetic evaluation of SHetA2
in tumor-bearing mice. The developed method will be utilized for the
quantification of SHetA2 in clinical studies.
Collapse
Affiliation(s)
- Ankur Sharma
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave. CPB331, Oklahoma City, Oklahoma 73117-1200, USA
| | - Elangovan Thavathiru
- Department of Obstetrics and Gynecology, Stephenson Cancer Center (SCC), University of Oklahoma Health Sciences Center, 975 NE 10th St, BRC 1217A, Oklahoma City, Oklahoma 73104, USA
| | - Doris Mangiaracina Benbrook
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave. CPB331, Oklahoma City, Oklahoma 73117-1200, USA.,Department of Obstetrics and Gynecology, Stephenson Cancer Center (SCC), University of Oklahoma Health Sciences Center, 975 NE 10th St, BRC 1217A, Oklahoma City, Oklahoma 73104, USA
| | - Sukyung Woo
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave. CPB331, Oklahoma City, Oklahoma 73117-1200, USA
| |
Collapse
|
30
|
Vejselova D, Kutlu HM, Kuş G. Examining impacts of ceranib-2 on the proliferation, morphology and ultrastructure of human breast cancer cells. Cytotechnology 2016; 68:2721-2728. [PMID: 27380965 PMCID: PMC5101343 DOI: 10.1007/s10616-016-9997-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 06/10/2016] [Indexed: 02/02/2023] Open
Abstract
Acid ceramidases are enzymes with a vital role in metabolizing ceramide to sphingosine-1-phosphate that is an antiproliferative metabolite in the ceramide pathway. Inhibition of exogenous ceramides with ceramidase inhibitors lead to augmented ceramide levels in cells and in turn lead to cell cycle arrest and apoptosis. Our study aimed at targeting ceramide metabolic pathway to induce apoptosis in human breast cancer cell line (MCF7) and we examined the antiproliferative and apoptotic activities of ceranib-2, an inhibitor of human ceramidase, on this cell line as well ultrastructural and mophological changes. Methods used for our examinations in this study were the colorimetric MTT assay, Annexin V/Propidium iodide and JC-1 staining, transmission electron microscopy and confocal microscopy. Ceranib-2 effectively inhibited the viability of MCF7 cells in 24 h in a dose dependent manner leading to apoptosis via the mitochondrial pathway by reducing the potential of mitochondrial membrane. Additionally, significant changes on cell morphology and ultrastructure were observed on MCF7 cells exposed to ceranib-2 indicating apoptotic cell death. Collectively, our data demonstrate that ceranib-2 exerts a great potential to be an antineoplastic compound and that the mechanism of its action rely on its apoptosis inducing ability.
Collapse
Affiliation(s)
- Djanan Vejselova
- Department of Biology, Faculty of Science, Anadolu University, Yunusemre Campus, Tepebasi, 26470, Eskisehir, Turkey.
| | - Hatice Mehtap Kutlu
- Department of Biology, Faculty of Science, Anadolu University, Yunusemre Campus, Tepebasi, 26470, Eskisehir, Turkey
| | - Gökhan Kuş
- Department of Health, Faculty of Open Education, Anadolu University, Eskisehir, Turkey
| |
Collapse
|
31
|
Comparison of cytotoxicity between extracts of Clinacanthus nutans (Burm. f.) Lindau leaves from different locations and the induction of apoptosis by the crude methanol leaf extract in D24 human melanoma cells. Altern Ther Health Med 2016; 16:368. [PMID: 27646974 PMCID: PMC5029048 DOI: 10.1186/s12906-016-1348-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 09/10/2016] [Indexed: 01/15/2023]
Abstract
BACKGROUND Clinacanthus nutans (Burm. f.) Lindau leaves are widely used by cancer patients and the leaf extracts possess cytotoxic and antiproliferative effects on several human cancer cell lines. However, the effect of C. nutans leaf extract on human melanoma, which is the least common but most fatal form of skin cancer and one of the most common cancers diagnosed in both sexes worldwide, is unknown. There is also limited information on whether the bioactivity of extracts differs between C. nutans leaves grown in different geographical locations with varying environmental conditions. METHODS The present study, for the first time, compared and demonstrated the cytotoxicity of the crude methanol extracts of C. nutans leaves from 11 different locations in Malaysia, Thailand and Vietnam, with diverse environmental conditions against D24 melanoma cells through WST-8 assay. The percentage of apoptotic cells following treatment with the most active extract was determined in a dose- and time-dependent manner by a cytofluorometric double staining technique. Biochemical and morphological changes in the treated and untreated cells were examined by fluorescence and transmission electron microscopy techniques, respectively, to further affirm the induction of apoptosis. RESULTS The leaves of plants grown at higher elevations and lower air temperatures were more cytotoxic to the D24 melanoma cells than those grown at lower elevations and higher air temperatures, with the leaf extract from Chiang Dao, Chiang Mai, Thailand exhibited the highest cytotoxicity (24 h EC50: 0.95 mg/mL and 72 h EC50: 0.77 mg/mL). This most active crude extract induced apoptotic cell death in the D24 cells in a dose- and time-dependent manner. Typical biochemical and morphological characteristics of apoptosis were also observed in the treated D24 cells. CONCLUSIONS The results, showing the cytotoxicity of C. nutans and the induction of apoptosis in D24 cells, are significant and useful to facilitate the development of C. nutans as a potential novel chemotherapeutic agent for the management of skin melanoma.
Collapse
|
32
|
Evaluation of cytotoxic potential and apoptotic effect of a methanolic extract of Bauhinia racemosa Lam. against a human cancer cell line, HeLa. Eur J Integr Med 2016. [DOI: 10.1016/j.eujim.2016.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
33
|
Tu Y, Kim E, Gao Y, Rankin GO, Li B, Chen YC. Theaflavin-3, 3'-digallate induces apoptosis and G2 cell cycle arrest through the Akt/MDM2/p53 pathway in cisplatin-resistant ovarian cancer A2780/CP70 cells. Int J Oncol 2016; 48:2657-65. [PMID: 27082635 PMCID: PMC4863729 DOI: 10.3892/ijo.2016.3472] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 02/11/2016] [Indexed: 01/07/2023] Open
Abstract
Ovarian cancer is the most lethal gynecological cancer among women worldwide. Adverse side effects and acquired resistance to conventional platinum based chemotherapy are major impediments in ovarian cancer treatment, and drive the development of more selective anticancer drugs that target cancer-specific defects. In this study, theaflavin-3, 3′-digallate (TF3), the major theaflavin monomer in black tea, exhibited a potent growth inhibitory effect on the cisplatin-resistant ovarian cancer A2780/CP70 cells (IC50, 23.81 μM), and was less cytotoxic to a normal ovarian IOSE-364 cells (IC50, 59.58 μM) than to the cancer cells. Flow cytometry analysis indicated that TF3 induced preferential apoptosis and G2 cell cycle arrest in A2780/CP70 cells with respect to IOSE-364 cells. TF3 induced apoptosis through both the intrinsic and extrinsic apoptotic pathways, and caused G2 cell cycle arrest via cyclin B1 in A2780/CP70 cells. The p53 protein played an important role in TF3-induced apoptosis and G2 cell cycle arrest. TF3 might upregulate the p53 expression via the Akt/MDM2 pathway. Our findings help elucidate the mechanisms by which TF3 may contribute to the prevention and treatment of platinum-resistant ovarian cancer.
Collapse
Affiliation(s)
- Youying Tu
- Department of Tea Science, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Eunhye Kim
- Department of Tea Science, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Ying Gao
- Department of Tea Science, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Gary O Rankin
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Bo Li
- Department of Tea Science, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Yi Charlie Chen
- Department of Tea Science, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| |
Collapse
|
34
|
Juarez AV, Sosa LDV, De Paul AL, Costa AP, Farina M, Leal RB, Torres AI, Pons P. Riboflavin acetate induces apoptosis in squamous carcinoma cells after photodynamic therapy. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2015; 153:445-54. [DOI: 10.1016/j.jphotobiol.2015.10.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 10/26/2015] [Accepted: 10/31/2015] [Indexed: 12/24/2022]
|
35
|
Cockbill LMR, Murk K, Love S, Hanley JG. Protein interacting with C kinase 1 suppresses invasion and anchorage-independent growth of astrocytic tumor cells. Mol Biol Cell 2015; 26:4552-61. [PMID: 26466675 PMCID: PMC4678014 DOI: 10.1091/mbc.e15-05-0270] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 10/07/2015] [Indexed: 12/11/2022] Open
Abstract
Astrocytic tumors are the most common form of primary brain tumor. Astrocytic tumor cells infiltrate the surrounding CNS tissue, allowing them to evade removal upon surgical resection of the primary tumor. Dynamic changes to the actin cytoskeleton are crucial to cancer cell invasion, but the specific mechanisms that underlie the particularly invasive phenotype of astrocytic tumor cells are unclear. Protein interacting with C kinase 1 (PICK1) is a PDZ and BAR domain-containing protein that inhibits actin-related protein 2/3 (Arp2/3)-dependent actin polymerization and is involved in regulating the trafficking of a number of cell-surface receptors. Here we report that, in contrast to other cancers, PICK1 expression is down-regulated in grade IV astrocytic tumor cell lines and also in clinical cases of the disease in which grade IV tumors have progressed from lower-grade tumors. Exogenous expression of PICK1 in the grade IV astrocytic cell line U251 reduces their capacity for anchorage-independent growth, two-dimensional migration, and invasion through a three-dimensional matrix, strongly suggesting that low PICK1 expression plays an important role in astrocytic tumorigenesis. We propose that PICK1 negatively regulates neoplastic infiltration of astrocytic tumors and that manipulation of PICK1 is an attractive possibility for therapeutic intervention.
Collapse
Affiliation(s)
- Louisa M R Cockbill
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Kai Murk
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Seth Love
- School of Clinical Sciences, University of Bristol, Bristol BS10 5NB, United Kingdom
| | - Jonathan G Hanley
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, United Kingdom
| |
Collapse
|
36
|
Huang H, Chen AY, Ye X, Li B, Rojanasakul Y, Rankin GO, Chen YC. Myricetin inhibits proliferation of cisplatin-resistant cancer cells through a p53-dependent apoptotic pathway. Int J Oncol 2015; 47:1494-502. [PMID: 26315556 PMCID: PMC4583523 DOI: 10.3892/ijo.2015.3133] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 07/30/2015] [Indexed: 12/24/2022] Open
Abstract
Cisplatin is a commonly used drug for cancer treatment by crosslinking DNA, leading to apoptosis of cancer cells, resistance to cisplatin treatment often occurs, leading to relapse. Therefore, there is a need for the development of more effective treatment strategies that can overcome chemoresistance. Myricetin is a flavonoid from fruits and vegetables, showing anticancer activity in various cancer cells. In this study, we found myricetin exhibited greater cytotoxicity than cisplatin in two cisplatin-resistant ovarian cancer cell lines, OVCAR-3 and A2780/CP70, and it was less cytotoxic to the normal ovarian cell line IOSE-364. Myricetin selectively induced apoptosis in both cisplatin-resistant cancer cell lines, but did not induce apoptosis in the normal ovarian cell line. It induced both Bcl-2 family-dependent intrinsic and DR5 dependent extrinsic apoptosis in OVCAR-3 cells. P53, a multifunctional tumor suppressor, regulated apoptosis in OVCAR-3 cells through a Bcl-2 family protein-dependent pathway. Myricetin did not induce cell cycle arrest in either ovarian cancer cell line. Because of its potency and selectivity against cisplatin-resistant cancer cells, myricetin could potentially be used to overcome cancer chemoresistance against platinum-based therapy.
Collapse
Affiliation(s)
- Haizhi Huang
- College of Science, Technology and Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA
| | - Allen Y Chen
- Department of Pharmaceutical Science, West Virginia University, Morgantown, WV 26506, USA
| | - Xingqian Ye
- College of Biosystems Engineering and Food Science, Fuli Institute of Food Science, Zhejiang University, Hangzhou, Zhejiang 310027, P.R. China
| | - Bingyun Li
- Department of Orthopaedics, School of Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Yon Rojanasakul
- Department of Pharmaceutical Science, West Virginia University, Morgantown, WV 26506, USA
| | - Gary O Rankin
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Yi Charlie Chen
- College of Science, Technology and Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA
| |
Collapse
|
37
|
Choi YM, Kim HK, Shim W, Anwar MA, Kwon JW, Kwon HK, Kim HJ, Jeong H, Kim HM, Hwang D, Kim HS, Choi S. Mechanism of Cisplatin-Induced Cytotoxicity Is Correlated to Impaired Metabolism Due to Mitochondrial ROS Generation. PLoS One 2015; 10:e0135083. [PMID: 26247588 PMCID: PMC4527592 DOI: 10.1371/journal.pone.0135083] [Citation(s) in RCA: 189] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 07/16/2015] [Indexed: 12/18/2022] Open
Abstract
The chemotherapeutic use of cisplatin is limited by its severe side effects. In this study, by conducting different omics data analyses, we demonstrated that cisplatin induces cell death in a proximal tubular cell line by suppressing glycolysis- and tricarboxylic acid (TCA)/mitochondria-related genes. Furthermore, analysis of the urine from cisplatin-treated rats revealed the lower expression levels of enzymes involved in glycolysis, TCA cycle, and genes related to mitochondrial stability and confirmed the cisplatin-related metabolic abnormalities. Additionally, an increase in the level of p53, which directly inhibits glycolysis, has been observed. Inhibition of p53 restored glycolysis and significantly reduced the rate of cell death at 24 h and 48 h due to p53 inhibition. The foremost reason of cisplatin-related cytotoxicity has been correlated to the generation of mitochondrial reactive oxygen species (ROS) that influence multiple pathways. Abnormalities in these pathways resulted in the collapse of mitochondrial energy production, which in turn sensitized the cells to death. The quenching of ROS led to the amelioration of the affected pathways. Considering these observations, it can be concluded that there is a significant correlation between cisplatin and metabolic dysfunctions involving mROS as the major player.
Collapse
Affiliation(s)
- Yong-Min Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, 443–749, Korea
| | - Han-Kyul Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 443–749, Korea
| | - Wooyoung Shim
- Department of Molecular Science and Technology, Ajou University, Suwon, 443–749, Korea
| | - Muhammad Ayaz Anwar
- Department of Molecular Science and Technology, Ajou University, Suwon, 443–749, Korea
| | - Ji-Woong Kwon
- Department of Molecular Science and Technology, Ajou University, Suwon, 443–749, Korea
| | - Hyuk-Kwon Kwon
- Department of Molecular Science and Technology, Ajou University, Suwon, 443–749, Korea
| | - Hyung Joong Kim
- Division of Energy Systems Research, Ajou University, Suwon, 443–749, Korea
| | - Hyobin Jeong
- School of Interdisciplinary Bioscience and Bioengineering, POSTECH, Pohang, 790–784, Korea
| | - Hwan Myung Kim
- Division of Energy Systems Research, Ajou University, Suwon, 443–749, Korea
| | - Daehee Hwang
- School of Interdisciplinary Bioscience and Bioengineering, POSTECH, Pohang, 790–784, Korea
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 440–746, Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, 443–749, Korea
- * E-mail:
| |
Collapse
|
38
|
Zhou Y, Wei L, Zhang H, Dai Q, Li Z, Yu B, Guo Q, Lu N. FV-429 Induced Apoptosis Through ROS-Mediated ERK2 Nuclear Translocation and p53 Activation in Gastric Cancer Cells. J Cell Biochem 2015; 116:1624-37. [PMID: 25650185 DOI: 10.1002/jcb.25118] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 01/23/2015] [Indexed: 01/30/2023]
Abstract
Following our previous finding which revealed that FV-429 induces apoptosis in human hepatocellular carcinoma HepG2 cells, in this study, we found that FV-429 could also induce apoptosis in human gastric cancer cells. Firstly, FV-429 inhibited the viability of BGC-823 and MGC-803 cells with IC50 values in the range of 38.10 ± 6.28 and 31.53 ± 6.84 µM for 24 h treatment by MTT-assay. Secondly, FV-429 induced apoptosis in BGC-823 and MGC-803 cells through the mitochondrial-mediated pathway, showing an increase in Bax/Bcl-2 ratios, and caspase-9 activation, without change in caspase-8. Further research revealed that the mitogen-activated protein kinases, including c-Jun N-terminal kinase, extracellular regulated kinase, and p38 mitogen-activated protein kinase, could be activated by FV-429-induced high level ROS. Moreover, FV-429 also promoted the ERK2 nuclear translocation, resulting in the co-translocation of p53 to the nucleus and increased transcription of p53-regulated proapoptotic genes. FV-429 significantly inhibited the nude mice xenograft tumors growth of BGC-823 or MGC-803 cells in vivo.
Collapse
Affiliation(s)
- Yuxin Zhou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, JiangSu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, P.R. China
| | - Libin Wei
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, JiangSu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, P.R. China
| | - Haiwei Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, JiangSu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, P.R. China
| | - Qinsheng Dai
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, JiangSu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, P.R. China
| | - Zhiyu Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, JiangSu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, P.R. China
| | - Boyang Yu
- Department of Complex Prescription of TCM, China Pharmaceutical University, Nanjing, P.R. China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, JiangSu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, P.R. China
| | - Na Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, JiangSu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, P.R. China
| |
Collapse
|
39
|
Banerji B, Pramanik SK. Synthesis and cytotoxicity studies of 1-propenyl-1,3-dihydro-benzimidazol-2-one. J Chem Biol 2015; 8:73-8. [PMID: 26101549 DOI: 10.1007/s12154-015-0130-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/24/2015] [Indexed: 01/04/2023] Open
Abstract
A heterocyclic compound 1-propenyl-1,3-dihydro-benzimidazol-2-one was synthesized by a palladium-catalyzed rearrangement reaction. Anticancer activities were confirmed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay against Neura 2a (neuroblastoma cell), HEK 293 (kidney cancer) and MCF-7 (breast cancer) cell lines at low micromolar range. Furthermore, clear images from phase-contrast and fluorescence microscopes and confocal images unambiguously confirm the cancer cell death. The single X-ray crystal structure of the compound unambiguously proves the structure of the benzimidazolone compound.
Collapse
Affiliation(s)
- Biswadip Banerji
- Department of Chemistry, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata, 700032 India
| | - Sumit Kumar Pramanik
- Department of Chemistry, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata, 700032 India
| |
Collapse
|
40
|
Sun J, Li F, Zhao Y, Zhao L, Qiao C, Li Z, Guo Q, Lu N. LZ-207, a Newly Synthesized Flavonoid, Induces Apoptosis and Suppresses Inflammation-Related Colon Cancer by Inhibiting the NF-κB Signaling Pathway. PLoS One 2015; 10:e0127282. [PMID: 26023926 PMCID: PMC4449173 DOI: 10.1371/journal.pone.0127282] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 04/13/2015] [Indexed: 12/19/2022] Open
Abstract
Flavonoids and flavonoid derivatives, which have significant biological and pharmacological activities, including antitumor and anti-inflammatory activities, have been widely used in human healthcare. To design a more effective flavonoid antitumor agent, we altered the flavonoid backbone with substitutions of piperazine and methoxy groups to synthesize a novel flavonoid derivative, LZ-207. The anticancer effect of LZ-207 against HCT116 colon cancer cells and the underlying mechanism of this effect were explored in this study. Specifically, LZ-207 exhibited inhibitory effects on growth and viability in several human colon cancer cell lines and induced apoptosis in HCT116 cells both in vitro and in vivo. LZ-207 treatment also suppressed the nuclear translocation of NF-κB and the phosphorylation of IκB and IKKα/β in a dose-dependent manner in both HCT116 cells and human acute monocytic leukemia THP-1 cells. Moreover, LZ-207 also reduced the secretion of the pro-inflammatory cytokine interleukin-6 (IL-6) in LPS-induced THP-1 cells, and this effect was confirmed at the transcriptional level. Furthermore, LZ-207 significantly inhibited HCT116 cell proliferation that was elicited by LPS-induced THP-1 cells in a co-culture system. These findings elucidated some potential molecular mechanisms for preventing inflammation-driven colon cancer using the newly synthesized flavonoid LZ-207 and suggested the possibility of further developing novel therapeutic agents derived from flavonoids.
Collapse
Affiliation(s)
- Jie Sun
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People’s Republic of China
| | - Fanni Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People’s Republic of China
| | - Yue Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People’s Republic of China
| | - Li Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People’s Republic of China
| | - Chen Qiao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People’s Republic of China
| | - Zhiyu Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, People’s Republic of China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People’s Republic of China
- * E-mail: (QG); (NL)
| | - Na Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People’s Republic of China
- * E-mail: (QG); (NL)
| |
Collapse
|
41
|
Gnanasekaran KK, Benbrook DM, Nammalwar B, Thavathiru E, Bunce RA, Berlin KD. Synthesis and evaluation of second generation Flex-Het scaffolds against the human ovarian cancer A2780 cell line. Eur J Med Chem 2015; 96:209-17. [DOI: 10.1016/j.ejmech.2015.03.070] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 03/10/2015] [Accepted: 03/31/2015] [Indexed: 11/26/2022]
|
42
|
Meinig JM, Peterson BR. Anticancer/antiviral agent Akt inhibitor-IV massively accumulates in mitochondria and potently disrupts cellular bioenergetics. ACS Chem Biol 2015; 10:570-6. [PMID: 25415586 PMCID: PMC4340353 DOI: 10.1021/cb500856c] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
Inhibitors
of the PI3-kinase/Akt (protein kinase B) pathway are
under investigation as anticancer and antiviral agents. Akt inhibitor-IV
(ChemBridge 5233705, CAS 681281-88-9, AKTIV), a small molecule reported
to inhibit this pathway, exhibits potent anticancer and broad-spectrum
antiviral activity. However, depending on concentration, this cationic
benzimidazole derivative exhibits paradoxical positive or negative
effects on the phosphorylation of Akt that are not well understood.
To elucidate its mechanism of action, we investigated its spectroscopic
properties. This compound proved to be sufficiently fluorescent (excitation
λmax = 388 nm, emission λmax = 460
nm) to enable examination of its uptake and distribution in living
mammalian cells. Despite a low quantum yield of 0.0016, imaging of
HeLa cells treated with AKTIV (1 μM, 5 min) by confocal laser
scanning microscopy, with excitation at 405 nm, revealed extensive
accumulation in mitochondria. Treatment of Jurkat lymphocytes with
1 μM AKTIV for 15 min caused accumulation to over 250 μM
in these organelles, whereas treatment with 5 μM AKTIV yielded
concentrations of over 1 mM in mitochondria, as analyzed by flow cytometry.
This massive loading resulted in swelling of these organelles, followed
by their apparent disintegration. These effects were associated with
profound disruption of cellular bioenergetics including mitochondrial
depolarization, diminished mitochondrial respiration, and release
of reactive oxygen species. Because mitochondria play key roles in
both cancer proliferation and viral replication, we conclude that
the anticancer and antiviral activities of AKTIV predominantly result
from its direct and immediate effects on the structure and function
of mitochondria.
Collapse
Affiliation(s)
- J. Matthew Meinig
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Blake R. Peterson
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
| |
Collapse
|
43
|
La Maestra S, De Flora S, Micale RT. Effect of cigarette smoke on DNA damage, oxidative stress, and morphological alterations in mouse testis and spermatozoa. Int J Hyg Environ Health 2015; 218:117-22. [DOI: 10.1016/j.ijheh.2014.08.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 08/19/2014] [Accepted: 08/29/2014] [Indexed: 12/17/2022]
|
44
|
Li B, Gao Y, Rankin GO, Rojanasakul Y, Cutler SJ, Tu Y, Chen YC. Chaetoglobosin K induces apoptosis and G2 cell cycle arrest through p53-dependent pathway in cisplatin-resistant ovarian cancer cells. Cancer Lett 2014; 356:418-33. [PMID: 25304379 DOI: 10.1016/j.canlet.2014.09.023] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 07/26/2014] [Accepted: 09/15/2014] [Indexed: 11/16/2022]
Abstract
Adverse side effects and acquired resistance to conventional platinum based chemotherapy have become major impediments in ovarian cancer treatment, and drive the development of more selective anticancer drugs. Chaetoglobosin K (ChK) was shown to have a more potent growth inhibitory effect than cisplatin on two cisplatin-resistant ovarian cancer cell lines, OVCAR-3 and A2780/CP70, and was less cytotoxic to a normal ovarian cell line, IOSE-364, than to the cancer cell lines. Hoechst 33342 staining and Flow cytometry analysis indicated that ChK induced preferential apoptosis and G2 cell cycle arrest in both ovarian cancer cells with respect to the normal ovarian cells. ChK induced apoptosis through a p53-dependent caspase-8 activation extrinsic pathway, and caused G2 cell cycle arrest via cyclin B1 by increasing p53 expression and p38 phosphorylation in OVCAR-3 and A2780/CP70 cells. DR5 and p21 might play an important role in determining the sensitivity of normal and malignant ovarian cells to ChK. Based on these results, ChK would be a potential compound for treating platinum-resistant ovarian cancer.
Collapse
Affiliation(s)
- Bo Li
- College of Science, Technology and Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA; Department of Tea Science, Zhejiang University, Hangzhou 310058, China
| | - Ying Gao
- College of Science, Technology and Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA; Department of Tea Science, Zhejiang University, Hangzhou 310058, China
| | - Gary O Rankin
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Yon Rojanasakul
- Department of Pharmaceutical Science, West Virginia University, Morgantown, WV 26506, USA
| | - Stephen J Cutler
- Department of Medicinal Chemistry, University of Mississippi, University, MS 38677, USA
| | - Youying Tu
- Department of Tea Science, Zhejiang University, Hangzhou 310058, China.
| | - Yi Charlie Chen
- College of Science, Technology and Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA.
| |
Collapse
|
45
|
Abstract
Mutations in the ataxia telangiectasia mutated (ATM) gene, which encodes a kinase critical for the normal DNA damage response, cause the neurodegenerative disorder ataxia-telangiectasia (AT). The substrates of ATM in the brain are poorly understood. Here we demonstrate that ATM phosphorylates and activates the transcription factor myocyte enhancer factor 2D (MEF2D), which plays a critical role in promoting survival of cerebellar granule cells. ATM associates with MEF2D after DNA damage and phosphorylates the transcription factor at four ATM consensus sites. Knockdown of endogenous MEF2D with a short-hairpin RNA (shRNA) increases sensitivity to etoposide-induced DNA damage and neuronal cell death. Interestingly, substitution of endogenous MEF2D with an shRNA-resistant phosphomimetic MEF2D mutant protects cerebellar granule cells from cell death after DNA damage, whereas an shRNA-resistant nonphosphorylatable MEF2D mutant does not. In vivo, cerebella in Mef2d knock-out mice manifest increased susceptibility to DNA damage. Together, our results show that MEF2D is a substrate for phosphorylation by ATM, thus promoting survival in response to DNA damage. Moreover, dysregulation of the ATM-MEF2D pathway may contribute to neurodegeneration in AT.
Collapse
|
46
|
Celastrol induces apoptosis in gefitinib-resistant non-small cell lung cancer cells via caspases-dependent pathways and Hsp90 client protein degradation. Molecules 2014; 19:3508-22. [PMID: 24662070 PMCID: PMC6271537 DOI: 10.3390/molecules19033508] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 02/26/2014] [Accepted: 03/12/2014] [Indexed: 12/17/2022] Open
Abstract
Celastrol, a triterpene extracted from the Chinese herb Tripterygium wilfordii, has been shown to have multiple bioactivities. Although among these activities, its anti-cancer effects have attracted the most attention, the effect of celastrol on gefitinib-resistant non-small cell lung cancer (NSCLC) cells is not clearly known. Here, we examined the potency of celastrol in three different NSCLC cell lines. We explored its treatment mechanism in two gefitinib-resistant NSCLC cell lines (H1650 and H1975). Our data demonstrated that celastrol exerted its apoptotic effect in a dose- and time-dependent manner. Also, the mitochondria membrane potential was gradually lost and the ratio of Bax/Bcl-2 increased after the treatment of celastrol, both of which are indicators of mitochondria membrane integrity. Although the caspases were activated, the treatment with pan-caspase inhibitor could partially inhibit the level of apoptosis. Moreover, the protein level of Hsp90 client proteins, EGFR and AKT, was measured. Interestingly, both client proteins were remarkably down-regulated after the treatment of celastrol. Taken together, our data showed that celastrol may be developed as a promising agent for treating gefitinib-resistant NSCLCs by inducing apoptosis through caspase-dependent pathways and Hsp90 client protein degradation.
Collapse
|
47
|
Clusianone, a naturally occurring nemorosone regioisomer, uncouples rat liver mitochondria and induces HepG2 cell death. Chem Biol Interact 2014; 212:20-9. [PMID: 24491676 DOI: 10.1016/j.cbi.2014.01.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 01/15/2014] [Accepted: 01/23/2014] [Indexed: 12/22/2022]
Abstract
Clusianone is a member of the polycyclic polyprenylated acylphloroglucinol family of natural products; its cytotoxic mechanism is unknown. Clusianone is a structural isomer of nemorosone, which is a mitochondrial uncoupler and a well-known cytotoxic anti-cancer agent; thus, we addressed clusianone action at the mitochondria and its potential cytotoxic effects on cancer cells. In the HepG2 hepatocarcinoma cell line, clusianone induced mitochondrial membrane potential dissipation, ATP depletion and phosphatidyl serine externalization; this later event is indicative of apoptosis induction. In isolated mitochondria from rat liver, clusianone promoted protonophoric mitochondrial uncoupling. This was evidenced by the dissipation of mitochondrial membrane potential, an increase in resting respiration, an inhibition of Ca(2+) influx, stimulation of Ca(2+) efflux in Ca(2+)-loaded mitochondria, a decrease in ATP and NAD(P)H levels, generation of ROS, and swelling of valinomycin-treated organelles in hyposmotic potassium acetate media. The cytotoxic and uncoupling actions of clusianone were appreciably less than those of nemorosone, likely due to the presence of an intra-molecular hydrogen bond with the juxtaposed carbonyl group at the C15 position. Therefore, clusianone is capable of pharmacologically increasing the leakage of protons from the mitochondria and with favorable cytotoxicity in relation to nemorosone.
Collapse
|
48
|
Benbrook DM, Nammalwar B, Long A, Matsumoto H, Singh A, Bunce RA, Berlin KD. SHetA2 interference with mortalin binding to p66shc and p53 identified using drug-conjugated magnetic microspheres. Invest New Drugs 2013; 32:412-23. [PMID: 24254390 PMCID: PMC4045313 DOI: 10.1007/s10637-013-0041-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 10/09/2013] [Indexed: 11/03/2022]
Abstract
SHetA2 is a small molecule flexible heteroarotinoid (Flex-Het) with promising cancer prevention and therapeutic activity. Extensive preclinical testing documented lack of SHetA2 toxicity at doses 25 to 150 fold above effective doses. Knowledge of the SHetA2 molecular target(s) that mediate(s) the mechanism of SHetA2 action is critical to appropriate design of clinical trials and improved analogs. The aim of this study was to develop a method to identify SHetA2 binding proteins in cancer cells. A known metabolite of SHetA2 that has a hydroxyl group available for attachment was synthesized and conjugated to a linker for attachment to a magnetic microsphere. SHetA2-conjugated magnetic microspheres and unconjugated magnetic microspheres were separately incubated with aliquots of a whole cell protein extract from the A2780 human ovarian cancer cell line. After washing away non-specifically bound proteins with the protein extraction buffer, SHetA2-binding proteins were eluted with an excess of free SHetA2. In two independent experiments, an SDS gel band of about 72 kDa was present at differential levels in wells of eluent from SHetA2-microspheres in comparison to wells of eluent from unconjugated microspheres. Mass spectrometry analysis of the bands (QStar) and straight eluents (Orbitrap) identified mortalin (HSPA9) to be present in the eluent from SHetA2-microspheres and not in eluent from unconjugated microspheres. Co-immunoprecipitation experiments demonstrated that SHetA2 interfered with mortalin binding to p53 and p66 Src homologous-collagen homologue (p66shc) inside cancer cells. Mortalin and SHetA2 conflictingly regulate the same molecules involved in mitochondria-mediated intrinsic apoptosis. The results validate the power of this protocol for revealing drug targets.
Collapse
Affiliation(s)
- Doris Mangiaracina Benbrook
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Room 1372, Oklahoma City, OK, USA,
| | | | | | | | | | | | | |
Collapse
|
49
|
Wang J, Shi X, Johnson RH, Kelbauskas L, Zhang W, Meldrum DR. Single-cell analysis reveals early manifestation of cancerous phenotype in pre-malignant esophageal cells. PLoS One 2013; 8:e75365. [PMID: 24116039 PMCID: PMC3792915 DOI: 10.1371/journal.pone.0075365] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 08/12/2013] [Indexed: 01/03/2023] Open
Abstract
Cellular heterogeneity plays a pivotal role in a variety of functional processes in vivo including carcinogenesis. However, our knowledge about cell-to-cell diversity and how differences in individual cells manifest in alterations at the population level remains very limited mainly due to the lack of appropriate tools enabling studies at the single-cell level. We present a study on changes in cellular heterogeneity in the context of pre-malignant progression in response to hypoxic stress. Utilizing pre-malignant progression of Barrett's esophagus (BE) as a disease model system we studied molecular mechanisms underlying the progression from metaplastic to dysplastic (pre-cancerous) stage. We used newly developed methods enabling measurements of cell-to-cell differences in copy numbers of mitochondrial DNA, expression levels of a set of mitochondrial and nuclear genes involved in hypoxia response pathways, and mitochondrial membrane potential. In contrast to bulk cell studies reported earlier, our study shows significant differences between metaplastic and dysplastic BE cells in both average values and single-cell parameter distributions of mtDNA copy numbers, mitochondrial function, and mRNA expression levels of studied genes. Based on single-cell data analysis, we propose that mitochondria may be one of the key factors in pre-malignant progression in BE.
Collapse
Affiliation(s)
- Jiangxin Wang
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Xu Shi
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Roger H. Johnson
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Laimonas Kelbauskas
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Weiwen Zhang
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Deirdre R. Meldrum
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| |
Collapse
|
50
|
Banerji B, Pramanik SK, Sanphui P, Nikhar S, Biswas SC. Synthesis and cytotoxicity studies of novel triazolo-benzoxazepine as new anticancer agents. Chem Biol Drug Des 2013; 82:401-9. [PMID: 23672315 DOI: 10.1111/cbdd.12164] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Revised: 04/25/2013] [Accepted: 05/03/2013] [Indexed: 12/16/2022]
Abstract
Cancer continues to be one of the biggest threats to the human civilization because there is no cure of it. Small heterocyclic molecule with low molecular weight and novel structural feature is therapeutically highly demanding. These molecules have the capability to disrupt signaling pathways leading to anticancer activities. Therefore, the search for new anticancer agents continues to draw attention to the research community. In this study, a small triazolo-benzoxazepine scaffolds was synthesized using a one-pot four-step synthetic methodology involving click reaction. Small libraries of 12 compounds were successfully synthesized and screened them against different cancer cell lines. Low micromolar anticancer activity was recorded using MTT assay, and further confirmation of cell death was obtained by phase contrast, fluorescent, and confocal images.
Collapse
Affiliation(s)
- Biswadip Banerji
- Department of Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata, 700032, India
| | | | | | | | | |
Collapse
|