1
|
Hink F, Aduriz-Arrizabalaga J, Lopez X, Suga H, De Sancho D, Rogers JM. Mixed Stereochemistry Macrocycle Acts as a Helix-Stabilizing Peptide N-Cap. J Am Chem Soc 2024; 146:24348-24357. [PMID: 39182188 DOI: 10.1021/jacs.4c05378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Interactions between proteins and α-helical peptides have been the focus of drug discovery campaigns. However, the large interfaces formed between multiple turns of an α-helix and a binding protein represent a significant challenge to inhibitor discovery. Modified peptides featuring helix-stabilizing macrocycles have shown promise as inhibitors of these interactions. Here, we tested the ability of N-terminal to side-chain thioether-cyclized peptides to inhibit the α-helix binding protein Mcl-1, by screening a trillion-scale library. The enriched peptides were lariats featuring a small, four-amino-acid N-terminal macrocycle followed by a short linear sequence that resembled the natural α-helical Mcl-1 ligands. These "Heliats" (helical lariats) bound Mcl-1 with tens of nM affinity, and inhibited the interaction between Mcl-1 and a natural peptide ligand. Macrocyclization was found to stabilize α-helical structures and significantly contribute to affinity and potency. Yet, the 2nd and 3rd positions within the macrocycle were permissible to sequence variation, so that a minimal macrocyclic motif, of an N-acetylated d-phenylalanine at the 1st position thioether connected to a cysteine at the 4th, could be grafted into a range of peptides and stabilize helical conformations. We found that d-stereochemistry is more helix-stabilizing than l- at the 1st position in the motif, as the d-amino acid can utilize polyproline II torsional angles that allow for more optimal intrachain hydrogen bonding. This mixed stereochemistry macrocyclic N-cap is synthetically accessible, requiring only minor modifications to standard solid-phase peptide synthesis, and its compatibility with peptide screening can provide ready access to helix-focused peptide libraries for de novo inhibitor discovery.
Collapse
Affiliation(s)
- Fabian Hink
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| | - Julen Aduriz-Arrizabalaga
- Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, UPV/EHU & Donostia International Physics Center (DIPC), PK 1072, Donostia-San Sebastian, Euskadi 20018, Spain
| | - Xabier Lopez
- Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, UPV/EHU & Donostia International Physics Center (DIPC), PK 1072, Donostia-San Sebastian, Euskadi 20018, Spain
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, Bunkyo-ku 113-0033, Japan
| | - David De Sancho
- Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, UPV/EHU & Donostia International Physics Center (DIPC), PK 1072, Donostia-San Sebastian, Euskadi 20018, Spain
| | - Joseph M Rogers
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| |
Collapse
|
2
|
Zhang S, Jin S, Zhang S, Li YY, Wang H, Chen Y, Lu H. Vitexin protects against high glucose-induced endothelial cell apoptosis and oxidative stress via Wnt/β-catenin and Nrf2 signalling pathway. Arch Physiol Biochem 2024; 130:275-284. [PMID: 35254859 DOI: 10.1080/13813455.2022.2028845] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/13/2021] [Accepted: 01/07/2022] [Indexed: 01/07/2023]
Abstract
Vitexin, a polyphenolic flavonoid, has been reported to be traditionally applied in the treatment of diabetes, cancer and cardiovascular diseases. OBJECTIVE The aim of this study was to investigate the anti-apoptosis and anti-oxidation effect and the potential mechanism of vitexin on high glucose-induced HUVECs. MATERIALS AND METHODS A high dose of glucose was added to HUVECs to establish an in vitro model. The cell viability was detected by CCK8 and flow cytometry assays. 2,7-dichlorodihydrofluorescein diacetate, colorimetry, and enzyme-linked immunosorbent assay were performed to detect oxidative stress. Besides, top flash and western blotting were employed to evaluate the effect of vitexin on Wnt/β-catenin. Furthermore, a Wnt/β-catenin inhibitor (KYA1797K) was used to confirm whether Wnt/β-catenin is involved in the protection of vitexin. At the same time, RT-PCR and western blot were performed to determine the effect of vitexin on Nrf2, while immunofluorescence assays were employed for the assessment of Nrf2 localisation. Then, in order to validate that Nrf2 plays an important role in the anti-oxidant effect of vitexin, methods were utilised to silence Nrf2 gene. RESULTS Herein, vitexin inhibited the proliferation and apoptosis of HG-mediated HUVECs. Mechanically, vitexin disrupted Wnt/β-catenin signalling pathway, thus resulting in the decrease of apoptosis in HG-induced HUVECs. A Wnt/β-catenin inhibitor (KYA1797K), was used for reverse verification. In the meantime, vitexin administration decreased reactive oxygen species (ROS) production and malondialdehyde (MDA) content and increased superoxide dismutase (SOD) activity in HG-induced HUVECs. Further investigations have revealed vitexin activated Nrf2 in HUVEC under high glucose, which was involved in its anti-oxidant effects. CONCLUSION Our investigation demonstrated that vitexin protected HUVECs from high glucose-induced injury via up-regulation of Wnt/β-catenin and Nrf2 signalling pathway. These results suggested that vitexin might serve as a potential drug for atherosclerosis and cardiovascular complications of diabetes.
Collapse
Affiliation(s)
- Sheng Zhang
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Shenyi Jin
- Department of Endocrinology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Shunxiao Zhang
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Yuan-Yuan Li
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Hua Wang
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Yue Chen
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
- Department of Endocrinology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Hao Lu
- Department of Endocrinology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| |
Collapse
|
3
|
Musa M, Abid M, Bradshaw TD, Boocock DJ, Coveney C, Argent SP, Woodward S. Probing the Mechanism of Action of Bis(phenolato) Amine (ONO Donor Set) Titanium(IV) Anticancer Agents. J Med Chem 2024; 67:2732-2744. [PMID: 38331433 PMCID: PMC10895680 DOI: 10.1021/acs.jmedchem.3c01874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 02/10/2024]
Abstract
The need for anticancer therapies that overcome metallodrug resistance while minimizing adverse toxicities is targeted, herein, using titanium coordination complexes. Octahedral titanium(IV) trans,mer-[Ti{R1N(CH2-2-MeO-4-R1-C6H2)2}2] [R1 = Et, allyl, n-Pr, CHO, F, CH2(morpholino), the latter from the formyl derivative; R2 = Me, Et; not all combinations] are attained from Mannich reactions of commercial 2-methoxyphenols (27-74% overall yield, 2 steps). These crystalline (four X-ray structures) Ti(IV)-complexes are active against MCF-7, HCT-116, HT-29, PANC-1, and MDA-MB-468 cancer cell lines (GI50 = 0.5-38 μM). Their activity and cancer selectivity (vs nontumor MRC-5 cells) typically exceeds that of cisplatin (up to 16-fold). Proteomic analysis (in MCF-7) supported by other studies (G2/M cell cycle arrest, ROS generation, γH2AX production, caspase activation, annexin positivity, western blot, and kinase screens in MCF-7 and HCT-116) suggest apoptosis elicited by more than one mechanism of action. Comparison of these data to the modes of action proposed for salan Ti(IV) complexes is made.
Collapse
Affiliation(s)
- Mustapha Musa
- GSK
Carbon Neutral Laboratories for Sustainable Chemistry, University of Nottingham, Triumph Road, Nottingham NG7 2TU, U.K.
| | - Mohammed Abid
- Department
of Chemistry, College of Science, University
of Anbar, Anbarshire 31001, Iraq
| | - Tracey D. Bradshaw
- BDI,
School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| | - David J. Boocock
- School
of Science and Technology, Nottingham Trent
University, Clifton, Nottingham NG11 8NS, U.K.
| | - Clare Coveney
- School
of Science and Technology, Nottingham Trent
University, Clifton, Nottingham NG11 8NS, U.K.
| | - Stephen P. Argent
- School
of Chemistry, University of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| | - Simon Woodward
- GSK
Carbon Neutral Laboratories for Sustainable Chemistry, University of Nottingham, Triumph Road, Nottingham NG7 2TU, U.K.
| |
Collapse
|
4
|
Montinaro A, Walczak H. Harnessing TRAIL-induced cell death for cancer therapy: a long walk with thrilling discoveries. Cell Death Differ 2023; 30:237-249. [PMID: 36195672 PMCID: PMC9950482 DOI: 10.1038/s41418-022-01059-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 02/10/2023] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL) can induce apoptosis in a wide variety of cancer cells, both in vitro and in vivo, importantly without killing any essential normal cells. These findings formed the basis for the development of TRAIL-receptor agonists (TRAs) for cancer therapy. However, clinical trials conducted with different types of TRAs have, thus far, afforded only limited therapeutic benefit, as either the respectively chosen agonist showed insufficient anticancer activity or signs of toxicity, or the right TRAIL-comprising combination therapy was not employed. Therefore, in this review we will discuss molecular determinants of TRAIL resistance, the most promising TRAIL-sensitizing agents discovered to date and, importantly, whether any of these could also prove therapeutically efficacious upon cancer relapse following conventional first-line therapies. We will also discuss the more recent progress made with regards to the clinical development of highly active non-immunogenic next generation TRAs. Based thereupon, we next propose how TRAIL resistance might be successfully overcome, leading to the possible future development of highly potent, cancer-selective combination therapies that are based on our current understanding of biology TRAIL-induced cell death. It is possible that such therapies may offer the opportunity to tackle one of the major current obstacles to effective cancer therapy, namely overcoming chemo- and/or targeted-therapy resistance. Even if this were achievable only for certain types of therapy resistance and only for particular types of cancer, this would be a significant and meaningful achievement.
Collapse
Affiliation(s)
- Antonella Montinaro
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK.
| | - Henning Walczak
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK.
- CECAD Cluster of Excellence, University of Cologne, 50931, Cologne, Germany.
- Center for Biochemistry, Medical Faculty, Joseph-Stelzmann-Str. 52, University of Cologne, 50931, Cologne, Germany.
| |
Collapse
|
5
|
Blockage of Autophagy Increases Timosaponin AIII-Induced Apoptosis of Glioma Cells In Vitro and In Vivo. Cells 2022; 12:cells12010168. [PMID: 36611961 PMCID: PMC9818637 DOI: 10.3390/cells12010168] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Timosaponin AIII (TSAIII), a saponin isolated from Anemarrhena asphodeloides and used in traditional Chinese medicine, exerts antitumor, anti-inflammatory, anti-angiogenesis, and pro-apoptotic activity on a variety of tumor cells. This study investigated the antitumor effects of TSAIII and the underlying mechanisms in human glioma cells in vitro and in vivo. TSAIII significantly inhibited glioma cell viability in a dose- and time-dependent manner but did not affect the growth of normal astrocytes. We also observed that in both glioma cell lines, TSAIII induces cell death and mitochondrial dysfunction, consistent with observed increases in the protein expression of cleaved-caspase-3, cleaved-caspase-9, cleaved-PARP, cytochrome c, and Mcl-1. TSAIII also activated autophagy, as indicated by increased accumulation of the autophagosome markers p62 and LC3-II and the autolysosome marker LAMP1. LC3 silencing, as well as TSAIII combined with the autophagy inhibitor 3-methyladenine (3MA), increased apoptosis in GBM8401 cells. TSAIII inhibited tumor growth in xenografts and in an orthotopic GBM8401 mice model in vivo. These results demonstrate that TSAIII exhibits antitumor effects and may hold potential as a therapy for glioma.
Collapse
|
6
|
Kim H, Choi JH, Moon CM, Kang JL, Woo M, Kim M. Shrimp miR-965 transfers tumoricidal mitochondria. Biol Proced Online 2022; 24:16. [PMID: 36289539 PMCID: PMC9598032 DOI: 10.1186/s12575-022-00178-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 10/19/2022] [Indexed: 11/21/2022] Open
Abstract
Background Micro RNA of Marsupenaeus japonicas has been known to promote apoptosis of tumor cells. However, the detailed mechanisms are not well understood. Results Using tomographic microscope, which can detect the internal structure of cells, we observed breast tumor cells following treatment of the miRNA. Intriguingly, we found that mitochondria migrate to an adjacent tumor cells through a tunneling nanotube. To recapitulate this process, we engineered a microfluidic device through which mitochondria were transferred. We show that this mitochondrial transfer process released endonuclease G (Endo G) into tumor cells, which we referred to herein as unsealed mitochondria. Importantly, Endo G depleted mitochondria alone did not have tumoricidal effects. Moreover, unsealed mitochondria had synergistic apoptotic effects with subtoxic dose of doxorubicin thereby mitigating cardiotoxicity. Conclusions Together, we show that the mitochondrial transfer through microfluidics can provide potential novel strategies towards tumor cell death. Supplementary Information The online version contains supplementary material available at 10.1186/s12575-022-00178-8.
Collapse
Affiliation(s)
- Hyueyun Kim
- grid.255649.90000 0001 2171 7754Department of Pharmacology, College of Medicine, Ewha Womans University, Magokdong-Ro 2-Gil, Gangseogu, Seoul, 07804 Republic of Korea
| | - Ji Ha Choi
- grid.255649.90000 0001 2171 7754Department of Pharmacology, College of Medicine, Ewha Womans University, Magokdong-Ro 2-Gil, Gangseogu, Seoul, 07804 Republic of Korea
| | - Chang Mo Moon
- grid.255649.90000 0001 2171 7754Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Jihee Lee Kang
- grid.255649.90000 0001 2171 7754Department of Physiology and Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Minna Woo
- grid.17063.330000 0001 2157 2938Toronto General Hospital Research Institute and Division of Endocrinology and Metabolism, Department of Medicine, University Health Network, University of Toronto, Toronto, ON Canada
| | - Minsuk Kim
- grid.255649.90000 0001 2171 7754Department of Pharmacology, College of Medicine, Ewha Womans University, Magokdong-Ro 2-Gil, Gangseogu, Seoul, 07804 Republic of Korea
| |
Collapse
|
7
|
Kumar R, Harilal S, Parambi DGT, Narayanan SE, Uddin MS, Marathakam A, Jose J, Mathew GE, Mathew B. Fascinating Chemopreventive Story of Wogonin: A Chance to Hit on the Head in Cancer Treatment. Curr Pharm Des 2021; 27:467-478. [PMID: 32338206 DOI: 10.2174/1385272824999200427083040] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/30/2020] [Indexed: 11/22/2022]
Abstract
Cancer, global havoc, is a group of debilitating diseases that strikes family as well as society. Cancer cases are drastically increasing these days. Despite many therapies and surgical procedures available, cancer is still difficult to control due to limited effective therapies or targeted therapies. Natural products can produce lesser side effects to the normal cells, which are the major demerit of chemotherapies and radiation. Wogonin, a natural product extracted from the plant, Scutellaria baicalensis has been widely studied and found with a high caliber to tackle most of the cancers via several mechanisms that include intrinsic as well as extrinsic apoptosis signaling pathways, carcinogenesis diminution, telomerase activity inhibition, metastasis inhibition in the inflammatory microenvironment, anti-angiogenesis, cell growth inhibition and arrest of the cell cycle, increased generation of H2O2 and accumulation of Ca2+ and also as an adjuvant along with anticancer drugs. This article discusses the role of wogonin in various cancers, its synergism with various drugs, and the mechanism by which wogonin controls tumor growth.
Collapse
Affiliation(s)
- Rajesh Kumar
- Department of Pharmacy, Kerala University of Health Sciences, Thrissur, Kerala, India
| | - Seetha Harilal
- Department of Pharmacy, Kerala University of Health Sciences, Thrissur, Kerala, India
| | - Della G T Parambi
- College of Pharmacy, Department of Pharmaceutical Chemistry, Jouf University, Sakaka, Al Jouf, 2014, Saudi Arabia
| | - Siju E Narayanan
- P.G. Department of Pharmacology, College of Pharmaceutical Sciences, Government Medical College, Kannur-670503, India
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | - Akash Marathakam
- Department of Pharmaceutical Chemistry, National College of Pharmacy, Calicut, India
| | - Jobin Jose
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Science, NITTE Deemed to be University, Manglore, 575018, India
| | - Githa E Mathew
- Department of Pharmacology, Grace College of Pharmacy, Palakkad, India
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, 678557, Kerala, India
| |
Collapse
|
8
|
Chi RPA, van der Watt P, Wei W, Birrer MJ, Leaner VD. Inhibition of Kpnβ1 mediated nuclear import enhances cisplatin chemosensitivity in cervical cancer. BMC Cancer 2021; 21:106. [PMID: 33530952 PMCID: PMC7852134 DOI: 10.1186/s12885-021-07819-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 01/19/2021] [Indexed: 02/07/2023] Open
Abstract
Background Inhibition of nuclear import via Karyopherin beta 1 (Kpnβ1) shows potential as an anti-cancer approach. This study investigated the use of nuclear import inhibitor, INI-43, in combination with cisplatin. Methods Cervical cancer cells were pre-treated with INI-43 before treatment with cisplatin, and MTT cell viability and apoptosis assays performed. Activity and localisation of p53 and NFκB was determined after co-treatment of cells. Results Pre-treatment of cervical cancer cells with INI-43 at sublethal concentrations enhanced cisplatin sensitivity, evident through decreased cell viability and enhanced apoptosis. Kpnβ1 knock-down cells similarly displayed increased sensitivity to cisplatin. Combination index determination using the Chou-Talalay method revealed that INI-43 and cisplatin engaged in synergistic interactions. p53 was found to be involved in the cell death response to combination treatment as its inhibition abolished the enhanced cell death observed. INI-43 pre-treatment resulted in moderately stabilized p53 and induced p53 reporter activity, which translated to increased p21 and decreased Mcl-1 upon cisplatin combination treatment. Furthermore, cisplatin treatment led to nuclear import of NFκB, which was diminished upon pre-treatment with INI-43. NFκB reporter activity and expression of NFκB transcriptional targets, cyclin D1, c-Myc and XIAP, showed decreased levels after combination treatment compared to single cisplatin treatment and this associated with enhanced DNA damage. Conclusions Taken together, this study shows that INI-43 pre-treatment significantly enhances cisplatin sensitivity in cervical cancer cells, mediated through stabilization of p53 and decreased nuclear import of NFκB. Hence this study suggests the possible synergistic use of nuclear import inhibition and cisplatin to treat cervical cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-07819-3.
Collapse
Affiliation(s)
- Ru-Pin Alicia Chi
- Division of Medical Biochemistry & Structural Biology, Department of Integrative Biomedical Sciences, SAMRC/UCT Gynaecological Cancer Research Centre, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, Cape Town, 7925, South Africa
| | - Pauline van der Watt
- Division of Medical Biochemistry & Structural Biology, Department of Integrative Biomedical Sciences, SAMRC/UCT Gynaecological Cancer Research Centre, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, Cape Town, 7925, South Africa
| | - Wei Wei
- Pfizer, Andover, MA, 01810, USA
| | - Michael J Birrer
- University of Arkansas Medical Sciences, D Winthrop P. Rockefeller Cancer Institute, Little Rock, AR, USA
| | - Virna D Leaner
- Division of Medical Biochemistry & Structural Biology, Department of Integrative Biomedical Sciences, SAMRC/UCT Gynaecological Cancer Research Centre, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, Cape Town, 7925, South Africa.
| |
Collapse
|
9
|
Alcon C, Manzano-Muñoz A, Prada E, Mora J, Soriano A, Guillén G, Gallego S, Roma J, Samitier J, Villanueva A, Montero J. Sequential combinations of chemotherapeutic agents with BH3 mimetics to treat rhabdomyosarcoma and avoid resistance. Cell Death Dis 2020; 11:634. [PMID: 32801295 PMCID: PMC7429859 DOI: 10.1038/s41419-020-02887-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/04/2020] [Accepted: 08/04/2020] [Indexed: 01/30/2023]
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in childhood and adolescence. Refractory/relapsed RMS patients present a bad prognosis that combined with the lack of specific biomarkers impairs the development of new therapies. Here, we utilize dynamic BH3 profiling (DBP), a functional predictive biomarker that measures net changes in mitochondrial apoptotic signaling, to identify anti-apoptotic adaptations upon treatment. We employ this information to guide the use of BH3 mimetics to specifically inhibit BCL-2 pro-survival proteins, defeat resistance and avoid relapse. Indeed, we found that BH3 mimetics that selectively target anti-apoptotic BCL-xL and MCL-1, synergistically enhance the effect of clinically used chemotherapeutic agents vincristine and doxorubicin in RMS cells. We validated this strategy in vivo using a RMS patient-derived xenograft model and observed a reduction in tumor growth with a tendency to stabilization with the sequential combination of vincristine and the MCL-1 inhibitor S63845. We identified the molecular mechanism by which RMS cells acquire resistance to vincristine: an enhanced binding of BID and BAK to MCL-1 after drug exposure, which is suppressed by subsequently adding S63845. Our findings validate the use of DBP as a functional assay to predict treatment effectiveness in RMS and provide a rationale for combining BH3 mimetics with chemotherapeutic agents to avoid tumor resistance, improve treatment efficiency, and decrease undesired secondary effects.
Collapse
Affiliation(s)
- Clara Alcon
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028, Barcelona, Spain
| | - Albert Manzano-Muñoz
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028, Barcelona, Spain
| | - Estela Prada
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, 08950, Esplugues de Llobregat, Spain
- Department of Haematology and Oncology, Hospital Sant Joan de Déu Barcelona, 08950, Esplugues de Llobregat, Spain
| | - Jaume Mora
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, 08950, Esplugues de Llobregat, Spain
- Department of Haematology and Oncology, Hospital Sant Joan de Déu Barcelona, 08950, Esplugues de Llobregat, Spain
| | - Aroa Soriano
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain
| | - Gabriela Guillén
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain
- Department of Surgery, Universitat Autònoma de Barcelona (UAB), 08193, Barcelona, Spain
| | - Soledad Gallego
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain
| | - Josep Roma
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain
| | - Josep Samitier
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028, Barcelona, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona (UB), 08028, Barcelona, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain
| | - Alberto Villanueva
- Program against Cancer Therapeutic Resistance (ProCURE), IDIBELL, Catalan Institute of Oncology, l'Hospitalet del Llobregat, 08907, Barcelona, Spain
- Xenopat S.L., Business Bioincubator, Bellvitge Health Science Campus, l'Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Joan Montero
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028, Barcelona, Spain.
| |
Collapse
|
10
|
Moyzis AG, Lally NS, Liang W, Leon LJ, Najor RH, Orogo AM, Gustafsson ÅB. Mcl-1-mediated mitochondrial fission protects against stress but impairs cardiac adaptation to exercise. J Mol Cell Cardiol 2020; 146:109-120. [PMID: 32717194 DOI: 10.1016/j.yjmcc.2020.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/30/2020] [Accepted: 07/19/2020] [Indexed: 12/26/2022]
Abstract
Myeloid cell leukemia-1 (Mcl-1) is a structurally and functionally unique anti-apoptotic Bcl-2 protein. While elevated levels of Mcl-1 contribute to tumor cell survival and drug resistance, loss of Mcl-1 in cardiac myocytes leads to rapid mitochondrial dysfunction and heart failure development. Although Mcl-1 is an anti-apoptotic protein, previous studies indicate that its functions extend beyond regulating apoptosis. Mcl-1 is localized to both the mitochondrial outer membrane and matrix. Here, we have identified that Mcl-1 in the outer mitochondrial membrane mediates mitochondrial fission, which is independent of its anti-apoptotic function. We demonstrate that Mcl-1 interacts with Drp1 to promote mitochondrial fission in response to various challenges known to perturb mitochondria morphology. Induction of fission by Mcl-1 reduces nutrient deprivation-induced cell death and the protection is independent of its BH3 domain. Finally, cardiac-specific overexpression of Mcl-1OM, but not Mcl-1Matrix, contributes to a shift in the balance towards fission and leads to reduced exercise capacity, suggesting that a pre-existing fragmented mitochondrial network leads to decreased ability to adapt to an acute increase in workload and energy demand. Overall, these findings highlight the importance of Mcl-1 in maintaining mitochondrial health in cells.
Collapse
Affiliation(s)
- Alexandra G Moyzis
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States of America
| | - Navraj S Lally
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States of America
| | - Wenjing Liang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States of America
| | - Leonardo J Leon
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States of America
| | - Rita H Najor
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States of America
| | - Amabel M Orogo
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States of America
| | - Åsa B Gustafsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States of America.
| |
Collapse
|
11
|
Wang Y, Su W, Mai Z, Du M, Yu S, Liu Y, Wang X, Chen T. Co-expression of Mcl-1 and Bak induces mitochondrial swelling. Biochem Biophys Res Commun 2020; 527:866-873. [PMID: 32430171 DOI: 10.1016/j.bbrc.2020.04.154] [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: 04/04/2020] [Accepted: 04/30/2020] [Indexed: 10/24/2022]
Abstract
We here used fluorescence imaging to explore the effect of co-overexpression of Mcl-1 and Bak/BH3-only proteins on mitochondrial morphology. The cells co-expressing CFP-Mcl-1 and YFP-Bak/BimL/Puma/tBid showed co-localization of Mcl-1 with Bak/Puma/BimL/tBid and also showed the inhibitory action of Mcl-1 on the Bak-, BimL-, Puma- or tBid-mediated cell death. Co-expression of Mcl-1 and Bak but not BH3-only proteins induced time-dependent mitochondrial swelling. Fluorescence resonance energy transfer (FRET) imaging proved the direct binding of Mcl-1 to Bak, BimL, Puma and tBid, respectively. In addition, Mcl-1 prevented Bak oligomerization by retrotranslocating Bak from mitochondria into cytoplasm. Moreover, Mcl-1-Bak complex exhibited a good co-localization with mitochondria, and co-expression of Mcl-1 and Bak for more than 24 h not only induced mitochondrial swelling but also impaired mitochondrial membrane potential. Collectively, co-expression of Mcl-1 and Bak but not BH3-only proteins significantly induced mitochondrial swelling and subsequent loss of mitochondrial membrane potential.
Collapse
Affiliation(s)
- Yong Wang
- MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Wenhua Su
- MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Zihao Mai
- MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Mengyan Du
- MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Si Yu
- MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Yangpei Liu
- MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Xiaopin Wang
- Department of Pain Management, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China.
| | - Tongshen Chen
- MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
12
|
Sebastian-Valverde M, Pasinetti GM. The NLRP3 Inflammasome as a Critical Actor in the Inflammaging Process. Cells 2020; 9:cells9061552. [PMID: 32604771 PMCID: PMC7348816 DOI: 10.3390/cells9061552] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/12/2020] [Accepted: 06/23/2020] [Indexed: 02/06/2023] Open
Abstract
As a consequence of the considerable increase in the human lifespan over the last century, we are experiencing the appearance and impact of new age-related diseases. The causal relationships between aging and an enhanced susceptibility of suffering from a broad spectrum of diseases need to be better understood. However, one specific shared feature seems to be of capital relevance for most of these conditions: the low-grade chronic inflammatory state inherently associated with aging, i.e., inflammaging. Here, we review the molecular and cellular mechanisms that link aging and inflammaging, focusing on the role of the innate immunity and more concretely on the nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, as well as how the chronic activation of this inflammasome has a detrimental effect on different age-related disorders.
Collapse
Affiliation(s)
| | - Giulio M. Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- James J. Peters Veterans Affairs Medical Center, Bronx, New York, NY 10468, USA
- Correspondence: ; Tel.: +1-212-241-1952
| |
Collapse
|
13
|
Li M, Li J, Guo X, Pan H, Zhou Q. Absence of HTATIP2 Expression in A549 Lung Adenocarcinoma Cells Promotes Tumor Plasticity in Response to Hypoxic Stress. Cancers (Basel) 2020; 12:cancers12061538. [PMID: 32545251 PMCID: PMC7352940 DOI: 10.3390/cancers12061538] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 12/15/2022] Open
Abstract
HIV-1 Tat Interactive Protein 2 (HTATIP2) is a tumor suppressor, of which reduced or absent expression is associated with increased susceptibility to tumorigenesis and enhanced tumor invasion and metastasis. However, whether the absent expression of HTATIP2 is a tumor-promoting factor that acts through improving tumor adaptation to hypoxia is unclear. Here, we established a stable HTATIP2-knockdown A549 human lung adenocarcinoma cell line (A549shHTATIP2) using lentiviral-delivered HTATIP2-targeting short hairpin RNA (shRNA), employed a double subcutaneous xenograft model and incorporated photoacoustic imaging and metabolomics approaches to elucidate the impact of the absent HTATIP2 expression on tumor response to hypoxic stress. Results from the in vivo study showed that A549shHTATIP2 tumors exhibited accelerated growth but decreased intratumoral oxygenation and angiogenesis and reduced sensitivity to sorafenib treatment as compared with their parental counterparts. Moreover, results of the immunoblot and real-time PCR analyses revealed that the HIF2α protein and mRNA levels in vehicle-treated A549shHTATIP2 tumors were significantly increased (p < 0.01 compared with the parental control tumors). Despite the strong HIF2α-c-Myc protein interaction indicated by our co-immunoprecipitation data, the increase in the c-Myc protein and mRNA levels was not significant in the A549shHTATIP2 tumors. Nonetheless, MCL-1 and β-catenin protein levels in A549shHTATIP2 tumors were significantly increased (p < 0.05 compared with the parental control tumors), suggesting an enhanced β-catenin/c-Myc/MCL-1 pathway in the absence of HTATIP2 expression. The finding of significantly decreased E-cadherin (p < 0.01 compared with vehicle-treated A549shHTATIP2 tumors) and increased vimentin (p < 0.05 compared with sorafenib-treated A549 tumors) protein levels in A549shHTATIP2 tumors implicates that the absence of HTATIP2 expression increases the susceptibility of A549 tumors to sorafenib-activated epithelial-mesenchymal transition (EMT) process. Comparison of the metabolomic profiles between A549 and A549shHTATIP2 tumors demonstrated that the absence of HTATIP2 expression resulted in increased tumor metabolic plasticity that enabled tumor cells to exploit alternative metabolic pathways for survival and proliferation rather than relying on glutamine and fatty acids as a carbon source to replenish TCA cycle intermediates. Our data suggest a mechanism by which the absent HTATIP2 expression modulates tumor adaptation to hypoxia and promotes an aggressive tumor phenotype by enhancing the HIF2α-regulated β-catenin/c-Myc/MCL-1 signaling, increasing the susceptibility of tumors to sorafenib treatment-activated EMT process, and improving tumor metabolic plasticity.
Collapse
Affiliation(s)
- Minghua Li
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA; (M.L.); (X.G.)
| | - Jing Li
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA;
| | - Xiaofang Guo
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA; (M.L.); (X.G.)
| | - Hua Pan
- Division of Cardiovascular Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Qingyu Zhou
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA; (M.L.); (X.G.)
- Correspondence: ; Tel.: +1-813-974-7081
| |
Collapse
|
14
|
Han JM, Hong KO, Yang IH, Ahn CH, Jin B, Lee W, Jung YC, Kim KA, Shin JA, Cho SD, Hong SD. Oridonin induces the apoptosis of mucoepidermoid carcinoma cell lines in a myeloid cell leukemia‑1‑dependent manner. Int J Oncol 2020; 57:377-385. [PMID: 32467983 DOI: 10.3892/ijo.2020.5061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/24/2020] [Indexed: 11/06/2022] Open
Abstract
Oridonin, an active diterpenoid isolated from Rabdosia rubescens, has been reported to exhibit anticancer activities in several tumors. The aim of the present study was to investigate the anticancer effects and molecular mechanisms of oridonin in mucoepidermoid carcinoma (MEC). Treatment with oridonin induced the apoptosis of MC‑3 and YD‑15 cell and inhibited the expression of myeloid cell leukemia‑1 (MCL‑1) through the regulation of the protein level through post‑translational regulation in these cell lines. Oridonin significantly increased the expression level of truncated Bid (t‑Bid) as a downstream target of MCL‑1 and subsequently decreased the mitochondrial membrane potential. The ectopic expression of MCL‑1 protein was sufficient to reverse the induction of apoptosis and the increased t‑Bid expression induced by oridonin in both cell lines. Taken together, these results suggest that oridonin exerts an apoptotic effect through the modulation of MCL‑1 and t‑Bid in human MEC cell lines and may thus be a potential anticancer drug candidate for the treatment of human MEC.
Collapse
Affiliation(s)
- Jung-Min Han
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Kyoung-Ok Hong
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - In-Hyoung Yang
- Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Chi-Hyun Ahn
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Bohwan Jin
- Laboratory Animal Center, CHA University, CHA Biocomplex, Seongnam, Gyeonggi 13488, Republic of Korea
| | - Wonwoo Lee
- Laboratory Animal Center, CHA University, CHA Biocomplex, Seongnam, Gyeonggi 13488, Republic of Korea
| | | | - Kyung-A Kim
- Department of Dentistry, School of Medicine, Eulji University, Daejeon 34824, Republic of Korea
| | - Ji-Ae Shin
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Sung-Dae Cho
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Seong-Doo Hong
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| |
Collapse
|
15
|
Wang Y, Su W, Mai Z, Yu S, Wang X, Chen T. Anti-apoptotic capacity of Mcl-1Δ127. Biochem Biophys Res Commun 2020; 526:1042-1048. [PMID: 32331834 DOI: 10.1016/j.bbrc.2020.03.181] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 03/30/2020] [Indexed: 11/29/2022]
Abstract
The anti-apoptotic ability of Mcl-1Δ127, a caspase cleavage product of Mcl-1, is debated. We here used fluorescence imaging to assess the anti-apoptotic capacity of Mcl-1Δ127 in living cells. Fluorescence imaging of living cells expressing CFP-Mcl-1Δ127 showed that Mcl-1Δ127 existed mainly in cytoplasm. Fluorescence imaging of living cells co-expressing CFP-Mcl-1Δ127 and YFP-Bak, CFP-Mcl-1Δ127 and YFP-BimL, CFP-Mcl-1Δ127 and YFP-Puma or CFP-Mcl-1Δ127 and YFP-tBid showed that Mcl-1Δ127 markedly inhibited the oligomerization of Bak, BimL, Puma and tBid on mitochondria and also inhibited the Bak-, BimL-, Puma- or tBid-mediated cell death, resulting in their partial localization in cytoplasm. Fluorescence resonance energy transfer (FRET) imaging proved that Mcl-1Δ127 bound to Bak, BimL, Puma and tBid, respectively. Fluorescence loss in photobleaching (FLIP) analyses showed that Mcl-1Δ127 did prevent Bak oligomerization by retrotranslocating Bak from mitochondria into cytoplasm. Collectively, Mcl-1Δ127 has the same anti-apoptotic capacity as Mcl-1, and prevents apoptosis by sequestering BH3-only or Bak proteins, thus inhibiting their oligomerization on mitochondria.
Collapse
Affiliation(s)
- Yong Wang
- MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Wenhua Su
- MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Zihao Mai
- MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Si Yu
- MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Xiaoping Wang
- Department of Pain Management, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China.
| | - Tongsheng Chen
- MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
16
|
Han B, Wu J, Huang L. Induction of Apoptosis in Lung Cancer Cells by Viburnum grandiflorum via Mitochondrial Pathway. Med Sci Monit 2020; 26:e920265. [PMID: 31900380 PMCID: PMC6977709 DOI: 10.12659/msm.920265] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background Lung cancer is one of the leading causes of mortality and morbidity. Viburnum grandiflorum is a medicinal herb known for its wide spectrum of pharmacological activities, but its anti-cancer properties against lung cancer cells have not been previously investigated. The present study elucidated the antitumor effect and associated mechanism of methanol extract of Viburnum grandiflorum extract (VGE) against lung cancer cells. Material/Methods The viability of H1650, HCC827, and H1299 cells was measured using MTT assay. Apoptosis and cell cycle progression were determined by flow cytometry using annexin-V/PI and JC-1 stains, respectively. The Lipofectamine Plus reagent (Invitrogen) was used for transfection of caspase-9 plasmid to H1650 and H1299 cells. Results The results showed decreased H1650, HCC827, and H1299 cell viability by VGE, which occurred in a concentration- and time-dependent manner. The VGE treatment significantly increased the rate of apoptosis in H1650 (P<0.05) and H1299 (P<0.02) cells at 48 and 72 h. Treatment of H1650 and H1299 cells with 10 μM of VGE significantly enhanced the number of cells in sub-G1 phase. The VGE treatment cleaved pro-caspase-8/-9 and-3 in H1650 and HCC827 cells at 72 h. The VGE treatment of H1650 and HCC827 cells reduced Mcl-1 protein expression. Treatment of H1650 and HCC827 cells with VGE markedly decreased the level of p-Akt. However, dominant-negative caspase-9 (caspase-9 dN) plasmid transfection prevented the viability-inhibitory effect of VGE on H1650 and HCC827 cells. Treatment of H1650 and HCC827 cells with VGE increased levels of cytochrome c in the cytosol. Conclusions VGE inhibited lung carcinoma cell viability by apoptosis activation through a caspase-dependent pathway. Therefore, VGE is a potent anti-cancer agent against lung cancer cells.
Collapse
Affiliation(s)
- Bing Han
- Department of Cardio-Thoracic Surgery, People's Hospital of Deyang City, Deyang, Sichuan, China (mainland)
| | - Jianqiang Wu
- Department of Cardio-Thoracic Surgery, People's Hospital of Deyang City, Deyang, Sichuan, China (mainland)
| | - Lei Huang
- Department of Cardio-Thoracic Surgery, People's Hospital of Deyang City, Deyang, Sichuan, China (mainland)
| |
Collapse
|
17
|
Fontaine MAC, Westra MM, Bot I, Jin H, Franssen AJPM, Bot M, de Jager SCA, Dzhagalov I, He YW, van Vlijmen BJM, Gijbels MJJ, Reutelingsperger CP, van Berkel TJC, Sluimer JC, Temmerman L, Biessen EAL. Low human and murine Mcl-1 expression leads to a pro-apoptotic plaque phenotype enriched in giant-cells. Sci Rep 2019; 9:14547. [PMID: 31601924 PMCID: PMC6787218 DOI: 10.1038/s41598-019-51020-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 09/23/2019] [Indexed: 12/21/2022] Open
Abstract
The anti-apoptotic protein myeloid cell leukemia 1 (Mcl-1) plays an important role in survival and differentiation of leukocytes, more specifically of neutrophils. Here, we investigated the impact of myeloid Mcl-1 deletion in atherosclerosis. Western type diet fed LDL receptor-deficient mice were transplanted with either wild-type (WT) or LysMCre Mcl-1fl/fl (Mcl-1−/−) bone marrow. Mcl-1 myeloid deletion resulted in enhanced apoptosis and lipid accumulation in atherosclerotic plaques. In vitro, Mcl-1 deficient macrophages also showed increased lipid accumulation, resulting in increased sensitivity to lipid-induced cell death. However, plaque size, necrotic core and macrophage content were similar in Mcl-1−/− compared to WT mice, most likely due to decreased circulating and plaque-residing neutrophils. Interestingly, Mcl-1−/− peritoneal foam cells formed up to 45% more multinucleated giant cells (MGCs) in vitro compared to WT, which concurred with an increased MGC presence in atherosclerotic lesions of Mcl-1−/− mice. Moreover, analysis of human unstable atherosclerotic lesions also revealed a significant inverse correlation between MGC lesion content and Mcl-1 gene expression, coinciding with the mouse data. Taken together, these findings suggest that myeloid Mcl-1 deletion leads to a more apoptotic, lipid and MGC-enriched phenotype. These potentially pro-atherogenic effects are however counteracted by neutropenia in circulation and plaque.
Collapse
Affiliation(s)
- Margaux A C Fontaine
- Experimental Vascular Pathology Group, Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Marijke M Westra
- Division of BioTherapeutics, Leiden Amsterdam Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Ilze Bot
- Division of BioTherapeutics, Leiden Amsterdam Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Han Jin
- Experimental Vascular Pathology Group, Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Aimée J P M Franssen
- Experimental Vascular Pathology Group, Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Martine Bot
- Division of BioTherapeutics, Leiden Amsterdam Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Saskia C A de Jager
- Division of BioTherapeutics, Leiden Amsterdam Centre for Drug Research, Leiden University, Leiden, the Netherlands.,Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ivan Dzhagalov
- Institue of Microbiology and Immunology, National Yang-Ming University, Taipei, 112, Taiwan
| | - You-Wen He
- Institue of Microbiology and Immunology, National Yang-Ming University, Taipei, 112, Taiwan
| | - Bart J M van Vlijmen
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, the Netherlands
| | - Marion J J Gijbels
- Experimental Vascular Pathology Group, Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands.,Department of Molecular Genetics, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Chris P Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Theo J C van Berkel
- Division of BioTherapeutics, Leiden Amsterdam Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Judith C Sluimer
- Experimental Vascular Pathology Group, Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands.,Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Lieve Temmerman
- Experimental Vascular Pathology Group, Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands.
| | - Erik A L Biessen
- Experimental Vascular Pathology Group, Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
18
|
Zhang Y, Li J, Zhang X, Gao G, Lu J, Zhang Q, Ding Y. Inhibition of U87 Glioma Cell Growth by Baicalein Through Apoptosis Induction and Cell Cycle Arrest. INT J PHARMACOL 2019. [DOI: 10.3923/ijp.2019.844.850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
19
|
Mahalingam D, Carew JS, Espitia CM, Cool RH, Giles FJ, de Jong S, Nawrocki ST. Heightened JNK Activation and Reduced XIAP Levels Promote TRAIL and Sunitinib-Mediated Apoptosis in Colon Cancer Models. Cancers (Basel) 2019; 11:E895. [PMID: 31248045 PMCID: PMC6678293 DOI: 10.3390/cancers11070895] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/20/2019] [Accepted: 06/22/2019] [Indexed: 12/11/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a potent inducer of apoptosis that may be a promising agent in cancer therapy due to its selectivity toward tumor cells. However, many cancer cells are resistant to TRAIL due to defects in apoptosis signaling or activation of survival pathways. We hypothesized that a disruption of pro-survival signaling cascades with the multi-tyrosine kinase inhibitor sunitinib and would be an effective strategy to enhance TRAIL-mediated apoptosis. Here we demonstrate that sunitinib significantly augments the anticancer activity of TRAIL in models of colon cancer. The therapeutic benefit of the TRAIL/sunitinib combination was associated with increased apoptosis marked by enhanced caspase-3 cleavage and DNA fragmentation. Overexpression of the anti-apoptotic factor B-cell lymphoma 2 (BCL-2) in HCT116 cells reduced TRAIL/sunitinib-mediated apoptosis, further supporting that sunitinib enhances the anticancer activity of TRAIL via augmented apoptosis. Analysis of pro-survival factors identified that the combination of TRAIL and sunitinib significantly downregulated the anti-apoptotic protein X-linked inhibitor of apoptosis protein (XIAP) through a c-Jun N-terminal kinase (JNK)-mediated mechanism. Short hairpin RNA (shRNA)-mediated knockdown of JNK confirmed its key role in the regulation of sensitivity to this combination as cells with suppressed JNK expression exhibited significantly reduced TRAIL/sunitinib-mediated apoptosis. Importantly, the therapeutic benefit of the TRAIL/sunitinib combination was validated in the HCT116-Luc and HCT15 colon cancer xenograft models, which both demonstrated significant anti-tumor activity in response to combination treatment. Collectively, our data demonstrate that sunitinib enhances TRAIL-mediated apoptosis by heightened JNK activation, diminished XIAP levels, and augmented apoptosis.
Collapse
Affiliation(s)
- Devalingam Mahalingam
- Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA.
| | - Jennifer S Carew
- Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA.
| | - Claudia M Espitia
- Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA.
| | - Robbert H Cool
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 GZ Groningen, The Netherlands.
| | - Francis J Giles
- Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA.
| | - Steven de Jong
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands.
| | - Steffan T Nawrocki
- Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA.
| |
Collapse
|
20
|
Petsri K, Chamni S, Suwanborirux K, Saito N, Chanvorachote P. Renieramycin T Induces Lung Cancer Cell Apoptosis by Targeting Mcl-1 Degradation: A New Insight in the Mechanism of Action. Mar Drugs 2019; 17:md17050301. [PMID: 31117253 PMCID: PMC6562878 DOI: 10.3390/md17050301] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/16/2022] Open
Abstract
Among malignancies, lung cancer is the major cause of cancer death. Despite the advance in lung cancer therapy, the five-year survival rate is extremely restricted due to therapeutic failure and disease relapse. Targeted therapies selectively inhibiting certain molecules in cancer cells have been accepted as promising ways to control cancer. In lung cancer, evidence has suggested that the myeloid cell leukemia 1 (Mcl-1) protein, an anti-apoptotic member of the Bcl-2 family, is a target for drug action. Herein, we report the Mcl-1 targeting activity of renieramycin T (RT), a marine-derived tetrahydroisoquinoline alkaloid that was isolated from the Thai blue sponge Xestospongia sp. RT was shown to be dominantly toxic to lung cancer cells compared to the normal cells in the lung. The cytotoxicity of this compound toward lung cancer cells was mainly exerted through apoptosis induction. For the mechanism of action, we found that RT mediated activation of p53 protein and caspase-9 and -3 activations. While others Bcl-2 family proteins (Bcl-2, Bak, and Bax) were minimally changed in response to RT, Mcl-1 protein was dramatically diminished. We further performed the cycloheximide experiment and found that the half-life of Mcl-1 was significantly shortened by RT treatment. When MG132, a potent selective proteasome inhibitor, was utilized, it could restore the Mcl-1 level. Furthermore, immunoprecipitation analysis revealed that RT significantly increased the formation of Mcl-1-ubiquitin complex compared to the non-treated control. In conclusion, we report the potential apoptosis induction of RT with a mechanism of action involving the targeting of Mcl-1 for ubiquitin-proteasomal degradation. As Mcl-1 is critical for cancer cell survival and chemotherapeutic failure, this novel information regarding the Mcl-1-targeted compound would be beneficial for the development of efficient anti-cancer strategies or targeted therapies.
Collapse
Affiliation(s)
- Korrakod Petsri
- Cell-Based Drug and Health Products Development Research Unit, Chulalongkorn University, Bangkok 10330, Thailand.
- Doctor of Philosophy Program in Interdisciplinary Pharmacology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand.
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Supakarn Chamni
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Khanit Suwanborirux
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Naoki Saito
- Graduate School of Pharmaceutical Sciences, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan.
| | - Pithi Chanvorachote
- Cell-Based Drug and Health Products Development Research Unit, Chulalongkorn University, Bangkok 10330, Thailand.
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
21
|
Hsp90 Inhibitor SNX-2112 Enhances TRAIL-Induced Apoptosis of Human Cervical Cancer Cells via the ROS-Mediated JNK-p53-Autophagy-DR5 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9675450. [PMID: 31019655 PMCID: PMC6452544 DOI: 10.1155/2019/9675450] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 11/08/2018] [Accepted: 11/19/2018] [Indexed: 12/28/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a potent cancer cell apoptosis-inducing factor that can induce apoptosis in a variety of cancer cells. However, resistance to TRAIL in cancer cells is a huge obstacle in creating effective TRAIL-targeted clinical therapies. Thus, agents that can either enhance the effect of TRAIL or overcome its resistance are needed. In this study, we combined TRAIL with SNX-2112, an Hsp90 inhibitor we previously developed, to explore the effect and mechanism that SNX-2112 enhanced TRAIL-induced apoptosis in cervical cancer cells. Our results showed that SNX-2112 markedly enhanced TRAIL-induced cytotoxicity in HeLa cells, and this combination was found to be synergistic. Additionally, we found that SNX-2112 sensitized TRAIL-mediated apoptosis caspase-dependently in TRAIL-resistant HeLa cells. Mechanismly, SNX-2112 downregulated antiapoptosis proteins, including Bcl-2, Bcl-XL, and FLIP, promoted the accumulation of reactive oxygen species (ROS), and increased the expression levels of p-JNK and p53. ROS scavenger NAC rescued SNX-2112/TRAIL-induced apoptosis and suppressed SNX-2112-induced p-JNK and p53. Moreover, SNX-2112 induced the upregulation of death-receptor DR5 in HeLa cells. The silencing of DR5 by siRNA significantly decreased cell apoptosis by the combined effect of SNX-2112 and TRAIL. In addition, SNX-2112 inhibited the Akt/mTOR signaling pathway and induced autophagy in HeLa cells. The blockage of autophagy by bafilomycin A1 or Atg7 siRNA abolished SNX-2112-induced upregulation of DR5. Meanwhile, ROS scavenger NAC, JNK inhibitor SP600125, and p53 inhibitor PFTα were used to verify that autophagy-mediated upregulation of DR5 was regulated by the SNX-2112-stimulated activation of the ROS-JNK-p53 signaling pathway. Thus, the combination of SNX-2112 and TRAIL may provide a novel strategy for the treatment of human cervical cancer by overcoming cellular mechanisms of apoptosis resistance.
Collapse
|
22
|
Jeena LM, Kumar D, Rahangdale S, Singh AP, Sarkhel BC. Effect of cumulus cells of cumulus-oocyte complexes on in vitro maturation, embryonic developmental and expression pattern of apoptotic genes after in vitro fertilization in water buffalo (Bubalus bubalis). Anim Biotechnol 2019; 31:135-141. [PMID: 30905313 DOI: 10.1080/10495398.2018.1554580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
In the present study, the potential of different grades of cumulus-oocyte complexes (COCs) for in vitro maturation (IVM) and embryonic development was assessed. Further, the association of the expression pattern of anti-apoptotic Mcl-1 and pro-apoptotic Bax genes in embryonic development was analyzed. Abattoir derived oocytes were graded into grade A and B based on surrounding cumulus rings. Out of 1050 ovaries, a total number of 770 and 1360, were of grade A and B COCs, respectively, were aspirated. After IVM, grade A COCs had a significantly higher number of polar bodies (92.04 ± 0.60%) as compared to grade B (85.88 ± 0.46%). On IVF and embryo culture, grade A COCs produced the significantly higher rate of cleavage and blastocyst (90.44 ± 0.71% and 41.55 ± 0.96%) as compared to grade B COCs (79.77 ± 0.76% and 30.44 ± 0.96%). The transcriptional analysis of apoptotic genes expression by Real-time PCR revealed a significantly higher expression of Mcl-1 gene in embryos of grade A as compared to grade B, whereas, the relative expression of Bax gene was down-regulated in grade A than grade B embryos. Thus it was concluded that the pattern of apoptotic genes expression in early-stage embryos can be used as a marker gene to predict the developmental competence of COCs.
Collapse
Affiliation(s)
- Lalit Mohan Jeena
- Animal Biotechnology Centre, Nanaji Deshmukh Veterinary Science University, Jabalpur, Madhya Pradesh, India
| | - Dharmendra Kumar
- Animal Biotechnology Centre, Nanaji Deshmukh Veterinary Science University, Jabalpur, Madhya Pradesh, India
| | - Sandeep Rahangdale
- Animal Biotechnology Centre, Nanaji Deshmukh Veterinary Science University, Jabalpur, Madhya Pradesh, India
| | - Ajit Pratap Singh
- Animal Biotechnology Centre, Nanaji Deshmukh Veterinary Science University, Jabalpur, Madhya Pradesh, India
| | - Bikash Chandra Sarkhel
- Animal Biotechnology Centre, Nanaji Deshmukh Veterinary Science University, Jabalpur, Madhya Pradesh, India
| |
Collapse
|
23
|
Annunziato S, de Ruiter JR, Henneman L, Brambillasca CS, Lutz C, Vaillant F, Ferrante F, Drenth AP, van der Burg E, Siteur B, van Gerwen B, de Bruijn R, van Miltenburg MH, Huijbers IJ, van de Ven M, Visvader JE, Lindeman GJ, Wessels LFA, Jonkers J. Comparative oncogenomics identifies combinations of driver genes and drug targets in BRCA1-mutated breast cancer. Nat Commun 2019; 10:397. [PMID: 30674894 PMCID: PMC6344487 DOI: 10.1038/s41467-019-08301-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 12/21/2018] [Indexed: 01/22/2023] Open
Abstract
BRCA1-mutated breast cancer is primarily driven by DNA copy-number alterations (CNAs) containing large numbers of candidate driver genes. Validation of these candidates requires novel approaches for high-throughput in vivo perturbation of gene function. Here we develop genetically engineered mouse models (GEMMs) of BRCA1-deficient breast cancer that permit rapid introduction of putative drivers by either retargeting of GEMM-derived embryonic stem cells, lentivirus-mediated somatic overexpression or in situ CRISPR/Cas9-mediated gene disruption. We use these approaches to validate Myc, Met, Pten and Rb1 as bona fide drivers in BRCA1-associated mammary tumorigenesis. Iterative mouse modeling and comparative oncogenomics analysis show that MYC-overexpression strongly reshapes the CNA landscape of BRCA1-deficient mammary tumors and identify MCL1 as a collaborating driver in these tumors. Moreover, MCL1 inhibition potentiates the in vivo efficacy of PARP inhibition (PARPi), underscoring the therapeutic potential of this combination for treatment of BRCA1-mutated cancer patients with poor response to PARPi monotherapy.
Collapse
Affiliation(s)
- Stefano Annunziato
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Oncode Institute, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Julian R de Ruiter
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Oncode Institute, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Linda Henneman
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Transgenic Core Facility, Mouse Clinic for Cancer and Aging (MCCA), The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Chiara S Brambillasca
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Oncode Institute, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Catrin Lutz
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Oncode Institute, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - François Vaillant
- ACRF Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Federica Ferrante
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Oncode Institute, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Anne Paulien Drenth
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Oncode Institute, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Eline van der Burg
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Oncode Institute, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Bjørn Siteur
- Preclinical Intervention Unit, Mouse Clinic for Cancer and Aging (MCCA), The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Bas van Gerwen
- Preclinical Intervention Unit, Mouse Clinic for Cancer and Aging (MCCA), The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Roebi de Bruijn
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Oncode Institute, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Martine H van Miltenburg
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.,Oncode Institute, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Ivo J Huijbers
- Transgenic Core Facility, Mouse Clinic for Cancer and Aging (MCCA), The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Marieke van de Ven
- Preclinical Intervention Unit, Mouse Clinic for Cancer and Aging (MCCA), The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | - Jane E Visvader
- ACRF Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Geoffrey J Lindeman
- ACRF Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.,Department of Medicine, University of Medicine, Parkville, VIC, 3010, Australia.,Parkville Familial Cancer Centre, Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Parkville, VIC, 3050, Australia
| | - Lodewyk F A Wessels
- Cancer Genomics Netherlands, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands. .,Oncode Institute, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands. .,Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.
| | - Jos Jonkers
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands. .,Cancer Genomics Netherlands, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands. .,Oncode Institute, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands.
| |
Collapse
|
24
|
Salisbury-Ruf CT, Bertram CC, Vergeade A, Lark DS, Shi Q, Heberling ML, Fortune NL, Okoye GD, Jerome WG, Wells QS, Fessel J, Moslehi J, Chen H, Roberts LJ, Boutaud O, Gamazon ER, Zinkel SS. Bid maintains mitochondrial cristae structure and function and protects against cardiac disease in an integrative genomics study. eLife 2018; 7:40907. [PMID: 30281024 PMCID: PMC6234033 DOI: 10.7554/elife.40907] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/27/2018] [Indexed: 01/07/2023] Open
Abstract
Bcl-2 family proteins reorganize mitochondrial membranes during apoptosis, to form pores and rearrange cristae. In vitro and in vivo analysis integrated with human genetics reveals a novel homeostatic mitochondrial function for Bcl-2 family protein Bid. Loss of full-length Bid results in apoptosis-independent, irregular cristae with decreased respiration. Bid-/- mice display stress-induced myocardial dysfunction and damage. A gene-based approach applied to a biobank, validated in two independent GWAS studies, reveals that decreased genetically determined BID expression associates with myocardial infarction (MI) susceptibility. Patients in the bottom 5% of the expression distribution exhibit >4 fold increased MI risk. Carrier status with nonsynonymous variation in Bid’s membrane binding domain, BidM148T, associates with MI predisposition. Furthermore, Bid but not BidM148T associates with Mcl-1Matrix, previously implicated in cristae stability; decreased MCL-1 expression associates with MI. Our results identify a role for Bid in homeostatic mitochondrial cristae reorganization, that we link to human cardiac disease. Cells contain specialized structures called mitochondria, which help to convert fuel into energy. These tiny energy factories have a unique double membrane, with a smooth outer and a folded inner lining. The folds, called cristae, provide a scaffold for the molecular machinery that produces chemical energy that the cell can use. The cristae are dynamic, and can change shape, condensing to increase energy output. Mitochondria also play a role in cell death. In certain situations, cristae can widen and release the proteins held within their folds. This can trigger a program of self-destruction in the cell. A family of proteins called Bcl-2 control such a ‘programmed cell death’ through the release of mitochondrial proteins. Some family members, including a protein called Bid, can reorganize cristae to regulate this cell-death program. When cells die, Bid proteins that had been split move to the mitochondria. But, even when cells are healthy, Bid molecules that are intact are always there, suggesting that this form of the protein may have another purpose. To investigate this further, Salisbury-Ruf, Bertram et al. used mice with Bid, and mice that lacked the protein. Without Bid, cells – including heart cells – struggled to work properly and used less oxygen than their normal counterparts. A closer look using electron microscopy revealed abnormalities in the cristae. However, adding ‘intact’ Bid proteins back in to the deficient cells restored them to normal. Moreover, without Bid, the mice hearts were less able to respond to an increased demand for energy. This decreased their performance and caused the formation of scars in the heart muscle called fibrosis, similar to a pattern observed in human patients following a heart attack. DNA data from an electronic health record database revealed a link between low levels of Bid genes and heart attack in humans, which was confirmed in further studies. In addition, a specific mutation in the Bid gene was found to affect its ability to regulate the formation of proper cristae. Combining evidence from mice with human genetics revealed new information about heart diseases. Mitochondrial health may be affected by a combination of specific variations in genes and changes in the Bid protein, which could affect heart attack risk. Understanding more about this association could help to identify and potentially reduce certain risk factors for heart attack.
Collapse
Affiliation(s)
- Christi T Salisbury-Ruf
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Clinton C Bertram
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Aurelia Vergeade
- Department of Pharmacology, Vanderbilt University, Nashville, United States
| | - Daniel S Lark
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, United States
| | - Qiong Shi
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States
| | - Marlene L Heberling
- Department of Biological Sciences, Vanderbilt University, Nashville, United States
| | - Niki L Fortune
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States
| | - G Donald Okoye
- Division of Cardiovascular Medicine and Cardio-oncology Program, Vanderbilt University Medical Center, Nashville, United States
| | - W Gray Jerome
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, United States
| | - Quinn S Wells
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States
| | - Josh Fessel
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States
| | - Javid Moslehi
- Division of Cardiovascular Medicine and Cardio-oncology Program, Vanderbilt University Medical Center, Nashville, United States
| | - Heidi Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, United States
| | - L Jackson Roberts
- Department of Pharmacology, Vanderbilt University, Nashville, United States.,Department of Medicine, Vanderbilt University Medical Center, Nashville, United States
| | - Olivier Boutaud
- Department of Pharmacology, Vanderbilt University, Nashville, United States
| | - Eric R Gamazon
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, United States.,Clare Hall, University of Cambridge, Cambridge, United Kingdom
| | - Sandra S Zinkel
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States.,Department of Medicine, Vanderbilt University Medical Center, Nashville, United States
| |
Collapse
|
25
|
Ralff MD, El-Deiry WS. TRAIL pathway targeting therapeutics. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2018; 3:197-204. [PMID: 30740527 DOI: 10.1080/23808993.2018.1476062] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Introduction Despite decades of focused research efforts, cancer remains a significant cause of morbidity and mortality. Tumor necrosis factor(TNF)-related apoptosis-inducing ligand (TRAIL) is capable of inducing cell death selectively in cancer cells while sparing normal cells. Areas covered In this review, the authors cover TRA therapy and strategies that have been undertaken to improve their efficacy, as well as unconventional approaches to TRAIL pathway activation including TRAIL-inducing small molecules. They also discuss mechanisms of resistance to TRAIL and the use of combination strategies to overcome it. Expert commentary Targeting the TRAIL pathway has been of interest in oncology, and although initial clinical trials of TRAIL receptor agonists (TRAs) showed limitations, novel approaches represent the future of TRAIL-based therapy.
Collapse
Affiliation(s)
- Marie D Ralff
- MD/PhD Program, Lewis Katz School of Medicine, Temple University, Philadelphia, PA.,Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Hematology/Oncology and Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA
| | - Wafik S El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Hematology/Oncology and Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA
| |
Collapse
|
26
|
Hsu CL, Chang HY, Chang JY, Hsu WM, Huang HC, Juan HF. Unveiling MYCN regulatory networks in neuroblastoma via integrative analysis of heterogeneous genomics data. Oncotarget 2017; 7:36293-36310. [PMID: 27167114 PMCID: PMC5095001 DOI: 10.18632/oncotarget.9202] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 04/19/2016] [Indexed: 12/31/2022] Open
Abstract
MYCN, an oncogenic transcription factor of the Myc family, is a major driver of neuroblastoma tumorigenesis. Due to the difficulty in drugging MYCN directly, revealing the molecules in MYCN regulatory networks will help to identify effective therapeutic targets for neuroblastoma therapy. Here we perform ChIP-sequencing and small RNA-sequencing of neuroblastoma cells to determine the MYCN-binding sites and MYCN-associated microRNAs, and integrate various types of genomic data to construct MYCN regulatory networks. The overall analysis indicated that MYCN-regulated genes were involved in a wide range of biological processes and could be used as signatures to identify poor-prognosis MYCN-non-amplified patients. Analysis of the MYCN binding sites showed that MYCN principally served as an activator. Using a computational approach, we identified 32 MYCN co-regulators, and some of these findings are supported by previous studies. Moreover, we investigated the interplay between MYCN transcriptional and microRNA post-transcriptional regulations and identified several microRNAs, such as miR-124-3p and miR-93-5p, which may significantly contribute to neuroblastoma pathogenesis. We also found MYCN and its regulated microRNAs acted together to repress the tumor suppressor genes. This work provides a comprehensive view of MYCN regulations for exploring therapeutic targets in neuroblastoma, as well as insights into the mechanism of neuroblastoma tumorigenesis.
Collapse
Affiliation(s)
- Chia-Lang Hsu
- Department of Life Science, Institute of Molecular and Cellular Biology, Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 106, Taiwan
| | - Hsin-Yi Chang
- Department of Life Science, Institute of Molecular and Cellular Biology, Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 106, Taiwan
| | - Jen-Yun Chang
- Department of Life Science, Institute of Molecular and Cellular Biology, Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 106, Taiwan
| | - Wen-Ming Hsu
- Department of Surgery, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Hsuan-Cheng Huang
- Institute of Biomedical Informatics, Center for Systems and Synthetic Biology, National Yang-Ming University, Taipei 112, Taiwan
| | - Hsueh-Fen Juan
- Department of Life Science, Institute of Molecular and Cellular Biology, Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 106, Taiwan
| |
Collapse
|
27
|
Extra-mitochondrial prosurvival BCL-2 proteins regulate gene transcription by inhibiting the SUFU tumour suppressor. Nat Cell Biol 2017; 19:1226-1236. [PMID: 28945232 PMCID: PMC5657599 DOI: 10.1038/ncb3616] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 08/17/2017] [Indexed: 02/07/2023]
Abstract
Direct interactions between pro- and anti-apoptotic BCL-2 family members form the basis of cell death decision-making at the outer mitochondrial membrane (OMM). Here we report that three antiapoptotic BCL-2 proteins (MCL-1, BCL-2, and BCL-XL) found untethered from the OMM function as transcriptional regulators of a prosurvival and growth program. Antiapoptotic BCL-2 proteins engage a BCL-2 homology (BH) domain sequence found in Suppressor of Fused (SUFU), a tumor suppressor and antagonist of the GLI DNA binding proteins. BCL-2 proteins directly promote SUFU turnover, inhibit SUFU-GLI interaction, and induce the expression of the GLI target genes BCL-2, MCL-1, and BCL-XL. Antiapoptotic BCL-2 protein/SUFU feedforward signaling promotes cancer cell survival and growth and can be disabled with BH3 mimetics – small molecules that target antiapoptotic BCL-2 proteins. Our findings delineate a chemical strategy for countering drug resistance in GLI-associated tumors and reveal unanticipated functions for BCL-2 proteins as transcriptional regulators.
Collapse
|
28
|
Holbeck SL, Camalier R, Crowell JA, Govindharajulu JP, Hollingshead M, Anderson LW, Polley E, Rubinstein L, Srivastava A, Wilsker D, Collins JM, Doroshow JH. The National Cancer Institute ALMANAC: A Comprehensive Screening Resource for the Detection of Anticancer Drug Pairs with Enhanced Therapeutic Activity. Cancer Res 2017; 77:3564-3576. [PMID: 28446463 PMCID: PMC5499996 DOI: 10.1158/0008-5472.can-17-0489] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/13/2017] [Accepted: 04/24/2017] [Indexed: 12/22/2022]
Abstract
To date, over 100 small-molecule oncology drugs have been approved by the FDA. Because of the inherent heterogeneity of tumors, these small molecules are often administered in combination to prevent emergence of resistant cell subpopulations. Therefore, new combination strategies to overcome drug resistance in patients with advanced cancer are needed. In this study, we performed a systematic evaluation of the therapeutic activity of over 5,000 pairs of FDA-approved cancer drugs against a panel of 60 well-characterized human tumor cell lines (NCI-60) to uncover combinations with greater than additive growth-inhibitory activity. Screening results were compiled into a database, termed the NCI-ALMANAC (A Large Matrix of Anti-Neoplastic Agent Combinations), publicly available at https://dtp.cancer.gov/ncialmanac Subsequent in vivo experiments in mouse xenograft models of human cancer confirmed combinations with greater than single-agent efficacy. Concomitant detection of mechanistic biomarkers for these combinations in vivo supported the initiation of two phase I clinical trials at the NCI to evaluate clofarabine with bortezomib and nilotinib with paclitaxel in patients with advanced cancer. Consequently, the hypothesis-generating NCI-ALMANAC web-based resource has demonstrated value in identifying promising combinations of approved drugs with potent anticancer activity for further mechanistic study and translation to clinical trials. Cancer Res; 77(13); 3564-76. ©2017 AACR.
Collapse
Affiliation(s)
- Susan L Holbeck
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, Maryland
| | - Richard Camalier
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, Maryland
| | - James A Crowell
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, Maryland
| | - Jeevan Prasaad Govindharajulu
- Clinical Pharmacodynamics Program, Applied/Developmental Research Directorate, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Melinda Hollingshead
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, Maryland
| | - Lawrence W Anderson
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, Maryland
| | - Eric Polley
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, Maryland
| | - Larry Rubinstein
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, Maryland
| | - Apurva Srivastava
- Clinical Pharmacodynamics Program, Applied/Developmental Research Directorate, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Deborah Wilsker
- Clinical Pharmacodynamics Program, Applied/Developmental Research Directorate, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Jerry M Collins
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, Maryland
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, Maryland.
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| |
Collapse
|
29
|
Haimovici A, Humbert M, Federzoni EA, Shan-Krauer D, Brunner T, Frese S, Kaufmann T, Torbett BE, Tschan MP. PU.1 supports TRAIL-induced cell death by inhibiting NF-κB-mediated cell survival and inducing DR5 expression. Cell Death Differ 2017; 24:866-877. [PMID: 28362429 PMCID: PMC5423115 DOI: 10.1038/cdd.2017.40] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 02/28/2017] [Accepted: 03/02/2017] [Indexed: 12/17/2022] Open
Abstract
The hematopoietic Ets-domain transcription factor PU.1/SPI1 orchestrates myeloid, B- and T-cell development, and also supports hematopoietic stem cell maintenance. Although PU.1 is a renowned tumor suppressor in acute myeloid leukemia (AML), a disease characterized by an accumulation of immature blast cells, comprehensive studies analyzing the role of PU.1 during cell death responses in AML treatment are missing. Modulating PU.1 expression in AML cells, we found that PU.1 supports tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis via two mechanisms: (a) by repressing NF-κB activity via a novel direct PU.1-RelA/p65 protein-protein interaction, and (b) by directly inducing TRAIL receptor DR5 expression. Thus, expression of NF-κB-regulated antiapoptotic genes was sustained in PU.1-depleted AML cells upon TRAIL treatment and DR5 levels were decreased. Last, PU.1 deficiency significantly increased AML cell resistance to anthracycline treatment. Altogether, these results reveal a new facet of PU.1's tumor suppressor function during antileukemic therapies.
Collapse
Affiliation(s)
- Aladin Haimovici
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Freiestrasse 1, Bern, Switzerland
| | - Magali Humbert
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Elena A Federzoni
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Deborah Shan-Krauer
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Thomas Brunner
- Chair of Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Steffen Frese
- Department of Thoracic Surgery, ELK Berlin Chest Hospital, Berlin, Germany
| | - Thomas Kaufmann
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Bruce E Torbett
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Mario P Tschan
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Freiestrasse 1, Bern, Switzerland
| |
Collapse
|
30
|
Anti-tumor activity of wogonin, an extract from Scutellaria baicalensis, through regulating different signaling pathways. Chin J Nat Med 2017; 15:15-40. [PMID: 28259249 DOI: 10.1016/s1875-5364(17)30005-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/06/2016] [Indexed: 12/14/2022]
Abstract
Wogonin is a plant flavonoid compound extracted from Scutellaria baicalensis (Huang-Qin or Chinese skullcap) and has been studied thoroughly by many researchers till date for its anti-viral, anti-oxidant, anti-cancerous and neuro-protective properties. Numerous experiments conducted in vitro and in vivo have demonstrated wogonin's excellent tumor inhibitory properties. The anti-cancer mechanism of wogonin has been ascribed to modulation of various cell signaling pathways, including serine-threonine kinase Akt (also known as protein kinase B) and AMP-activated protein kinase (AMPK) pathways, p53-dependent/independent apoptosis, and inhibition of telomerase activity. Furthermore, wogonin also decreases DNA adduct formation with a carcinogenic compound 2-Aminofluorene and inhibits growth of drug resistant malignant cells and their migration and metastasis, without any side effects. Recently, newly synthesized wogonin derivatives have been developed with impressive anti-tumor activity. This review is the succinct appraisal of the pertinent articles on the mechanisms of anti-tumor properties of wogonin. We also summarize the potential of wogonin and its derivatives used alone or as an adjunct therapy for cancer treatment. Furthermore, pharmacokinetics and side effects of wogonin and its analogues have also been discussed.
Collapse
|
31
|
Ginsenoside compound K sensitizes human colon cancer cells to TRAIL-induced apoptosis via autophagy-dependent and -independent DR5 upregulation. Cell Death Dis 2016; 7:e2334. [PMID: 27512955 PMCID: PMC5108320 DOI: 10.1038/cddis.2016.234] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 06/24/2016] [Accepted: 07/07/2016] [Indexed: 11/08/2022]
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a potent cancer cell-specific apoptosis-inducing cytokine with little toxicity to most normal cells. However, acquired resistance of cancer cells to TRAIL is a roadblock. Agents that can either potentiate the effect of TRAIL or overcome resistance to TRAIL are urgently needed. This article reports that ginsenoside compound K (CK) potentiates TRAIL-induced apoptosis in HCT116 colon cancer cells and sensitizes TRAIL-resistant colon cancer HT-29 cells to TRAIL. On a cellular mechanistic level, CK downregulated cell survival proteins including Mcl-1, Bcl-2, surviving, X-linked inhibitor of apoptosis protein and Fas-associated death domain-like IL-1-converting enzyme-inhibitory protein, upregulated cell pro-apoptotic proteins including Bax, tBid and cytochrome c, and induced the cell surface expression of TRAIL death receptor DR5. Reduction of DR5 levels by siRNAs significantly decreases CK- and TRAIL-mediated apoptosis. Importantly, our results indicate, for the first time, that DR5 upregulation is mediated by autophagy, as blockade of CK-induced autophagy by 3-MA, LY294002 or Atg7 siRNAs substantially decreases DR5 upregulation and reduces the synergistic effect. Furthermore, CK-stimulated autophagy is mediated by the reactive oxygen species–c-Jun NH2-terminal kinase pathway. Moreover, we found that p53 and the C/EBP homologous (CHOP) protein is also required for DR5 upregulation but not related with autophagy. Our findings contribute significantly to the understanding of the mechanism accounted for the synergistic anticancer activity of CK and TRAIL, and showed a novel mechanism related with DR5 upregulation.
Collapse
|
32
|
Ong PS, Wang L, Chia DMH, Seah JYX, Kong LR, Thuya WL, Chinnathambi A, Lau JYA, Wong ALA, Yong WP, Yang D, Ho PCL, Sethi G, Goh BC. A novel combinatorial strategy using Seliciclib(®) and Belinostat(®) for eradication of non-small cell lung cancer via apoptosis induction and BID activation. Cancer Lett 2016; 381:49-57. [PMID: 27461583 DOI: 10.1016/j.canlet.2016.07.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 07/04/2016] [Accepted: 07/19/2016] [Indexed: 01/22/2023]
Abstract
With conventional anticancer agents for non-small cell lung cancer (NSCLC) reaching therapeutic ceiling, the novel combination using histone deacetylase inhibitor, PXD101 (Belinostat(®)), and CDK inhibitor, CYC202 (Seliciclib(®)), was investigated as an alternative anticancer strategy. At clinically achievable concentration of CYC202 (15 µM), combination therapy resulted in significant reduction in cell proliferation (IC50 = 3.67 ± 0.80 µM, p < 0.05) compared with PXD101 alone (IC50 = 6.56 ± 0.42 µM) in p53 wild-type A549 cells. Significant increase in apoptosis that occurred independently of cell cycle arrest was observed after concurrent treatment. This result was corroborated by greater formation of cleaved caspase-8, caspase-3 and PARP. Up-regulation of p53 and truncated BID protein levels was seen while Mcl-1 and XIAP protein levels were down-regulated upon combined treatment. Further analysis of apoptotic pathways revealed that caspase inhibitors, but not p53 silencing, significantly abrogated the cytotoxic enhancement. Moreover, the enhanced efficacy of this combination was additionally confirmed in p53 null H2444 cells, suggesting the potential of this combination for treatment of NSCLC that are not amenable to effects of conventional p53-inducing agents.
Collapse
Affiliation(s)
- Pei-Shi Ong
- Department of Pharmacy, National University of Singapore, Singapore
| | - Lingzhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, National University of Singapore, Singapore
| | | | | | - Li-Ren Kong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Win-Lwin Thuya
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Jie-Ying Amelia Lau
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Andrea Li-Ann Wong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Hematology & Oncology, National University Health System, Singapore
| | - Wei-Peng Yong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Hematology & Oncology, National University Health System, Singapore
| | - Daiwen Yang
- Department of Biological Science, National University of Singapore, Singapore
| | - Paul Chi-Lui Ho
- Department of Pharmacy, National University of Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, National University of Singapore, Singapore.
| | - Boon-Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, National University of Singapore, Singapore; Department of Hematology & Oncology, National University Health System, Singapore.
| |
Collapse
|
33
|
Ko H, Jeong MH, Jeon H, Sung GJ, So Y, Kim I, Son J, Lee SW, Yoon HG, Choi KC. Delphinidin sensitizes prostate cancer cells to TRAIL-induced apoptosis, by inducing DR5 and causing caspase-mediated HDAC3 cleavage. Oncotarget 2016; 6:9970-84. [PMID: 25991668 PMCID: PMC4496411 DOI: 10.18632/oncotarget.3667] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 02/12/2015] [Indexed: 12/31/2022] Open
Abstract
TRAIL can induce apoptosis in some cancer cells and is an immune effector in the surveillance and elimination of developing tumors. Yes, some cancers are resistant to TRAIL. Delphinidin, a polyphenolic compound contained in brightly colored fruits and vegetables, has anti-inflammatory, anti-oxidant, and anti-tumorigenic activities. Here we showed that delphinidin sensitized TRAIL-resistant human prostate cancer cells to undergo apoptosis. Cells treated with delphinidin and TRAIL activated the extrinsic and intrinsic pathways of caspase activation. TRAIL-induced apoptosis in prostate cancer cells pretreated with delphinidin was dependent on death receptor 5 (DR5) and downstream cleavage of histone deacetylase 3 (HDAC3). In conclusion, delphinidin sensitizes prostate cancer cells to TRAIL-induced apoptosis by inducing DR5, thus causing caspase-mediated HDAC3 cleavage. Our data reveal a potential way of chemoprevention of prostate cancer by enabling TRAIL-mediated apoptosis.
Collapse
Affiliation(s)
- Hyeonseok Ko
- Laboratory of Molecular Oncology, Cheil General Hospital & Women's Healthcare Center, College of Medicine, Dankook University, Seoul, South Korea
| | - Mi-Hyeon Jeong
- Department of Biomedical Sciences and Department of Pharmacology, University of Ulsan College of Medicine, Seoul, Korea.,Department of Biochemistry and Molecular Biology, Brain Korea PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Hyelin Jeon
- Department of Biomedical Sciences and Department of Pharmacology, University of Ulsan College of Medicine, Seoul, Korea
| | - Gi-Jun Sung
- Department of Biomedical Sciences and Department of Pharmacology, University of Ulsan College of Medicine, Seoul, Korea
| | - Youngsin So
- Department of Biomedical Sciences and Department of Pharmacology, University of Ulsan College of Medicine, Seoul, Korea
| | - InKi Kim
- Asan Institute for Medical Research, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - JaeKyoung Son
- Department of Biomedical Sciences and Department of Pharmacology, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang-wook Lee
- Department of Radiation Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Ho-Geun Yoon
- Department of Biochemistry and Molecular Biology, Brain Korea PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Kyung-Chul Choi
- Department of Biomedical Sciences and Department of Pharmacology, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
34
|
Morciano G, Giorgi C, Balestra D, Marchi S, Perrone D, Pinotti M, Pinton P. Mcl-1 involvement in mitochondrial dynamics is associated with apoptotic cell death. Mol Biol Cell 2015; 27:20-34. [PMID: 26538029 PMCID: PMC4694758 DOI: 10.1091/mbc.e15-01-0028] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 10/29/2015] [Indexed: 01/04/2023] Open
Abstract
The B-cell lymphoma-2 (Bcl-2) family proteins are critical regulators of apoptosis and consist of both proapoptotic and antiapoptotic factors. Within this family, the myeloid cell leukemia factor 1 (Mcl-1) protein exists in two forms as the result of alternative splicing. The long variant (Mcl-1L) acts as an antiapoptotic factor, whereas the short isoform (Mcl-1S) displays proapoptotic activity. In this study, using splice-switching antisense oligonucleotides (ASOs), we increased the synthesis of Mcl-1S, which induced a concurrent reduction of Mcl-1L, resulting in increased sensitivity of cancer cells to apoptotic stimuli. The Mcl-1 ASOs also induced mitochondrial hyperpolarization and a consequent increase in mitochondrial calcium (Ca(2+)) accumulation. The high Mcl-1S/L ratio correlated with significant hyperfusion of the entire mitochondrial network, which occurred in a dynamin-related protein (Drp1)-dependent manner. Our data indicate that the balance between the long and short variants of the Mcl-1 gene represents a key aspect of the regulation of mitochondrial physiology. We propose that the Mcl-1L/S balance is a novel regulatory factor controlling the mitochondrial fusion and fission machinery.
Collapse
Affiliation(s)
- Giampaolo Morciano
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies
| | - Carlotta Giorgi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies
| | - Dario Balestra
- Department of Life Sciences and Biotechnology, University of Ferrara, FE 44121 Ferrara, Italy
| | - Saverio Marchi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies
| | - Daniela Perrone
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, FE 44121 Ferrara, Italy
| | - Mirko Pinotti
- Department of Life Sciences and Biotechnology, University of Ferrara, FE 44121 Ferrara, Italy
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies
| |
Collapse
|
35
|
SOPHONNITHIPRASERT THANET, NILWARANGKOON SIRINUN, NAKAMURA YUKIO, WATANAPOKASIN RAMIDA. Goniothalamin enhances TRAIL-induced apoptosis in colorectal cancer cells through DR5 upregulation and cFLIP downregulation. Int J Oncol 2015; 47:2188-96. [DOI: 10.3892/ijo.2015.3204] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/24/2015] [Indexed: 11/05/2022] Open
|
36
|
Lee HE, Nam JS, Shin JA, Hong IS, Yang IH, You MJ, Cho SD. Convallaria keiskei as a novel therapeutic alternative for salivary gland cancer treatment by targeting myeloid cell leukemia-1. Head Neck 2015; 38 Suppl 1:E761-70. [PMID: 25914292 DOI: 10.1002/hed.24096] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Various chemotherapeutic agents have been used largely for the treatment of salivary gland cancer. However, results are disappointing, and these agents can cause some serious side effects. Therefore, recent studies have focused on the possible roles of natural products to overcome these limitations. METHODS Salivary gland cancer cells treated with or without Convallaria keiskei (MECK) for 24 hours. Apoptotic changes were evaluated by live/dead assay, immunoblotting, and expression levels of caspase-3 and B-cell lymphoma-2 family member. RESULTS MECK significantly inhibited salivary gland cancer growth. At the molecular level, MECK dramatically reduced myeloid cell leukemia-1 (Mcl-1) in a translation-dependent manner and thereby induced apoptosis through Bax/Bid. Furthermore, we found that Mcl-1 could be a potential therapeutic target of MECK-induced apoptosis and its stability is regulated by extracellular signal-regulated kinases 1/2 (ERK1/2) signaling CONCLUSION MECK can be used as a safe and efficient therapeutic alternative for the treatment of salivary gland cancer. © 2015 Wiley Periodicals, Inc. Head Neck 38: E761-E770, 2016.
Collapse
Affiliation(s)
- Haeng-Eun Lee
- Department of Oral Pathology, School of Dentistry and Institute of Biodegradable Material, Institute of Oral Bioscience, Brain Korea 21 Project, Chonbuk National University, Jeonju, Republic of Korea
| | - Jeong-Seok Nam
- Laboratory of Tumor Suppressor, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, South Korea.,Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, South Korea
| | - Ji-Ae Shin
- Department of Oral Pathology, School of Dentistry and Institute of Biodegradable Material, Institute of Oral Bioscience, Brain Korea 21 Project, Chonbuk National University, Jeonju, Republic of Korea
| | - In-Sun Hong
- Laboratory of Tumor Suppressor, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, South Korea.,Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, South Korea
| | - In-Hyoung Yang
- Department of Oral Pathology, School of Dentistry and Institute of Biodegradable Material, Institute of Oral Bioscience, Brain Korea 21 Project, Chonbuk National University, Jeonju, Republic of Korea
| | - Myung-Jo You
- Laboratory of Veterinary Parasitology, College of Veterinary Medicine and Bio-Safety Research Centre, Chonbuk National University, Jeonju, Republic of Korea
| | - Sung-Dae Cho
- Department of Oral Pathology, School of Dentistry and Institute of Biodegradable Material, Institute of Oral Bioscience, Brain Korea 21 Project, Chonbuk National University, Jeonju, Republic of Korea
| |
Collapse
|
37
|
Inoue T, Aoyama-Ishikawa M, Kamoshida S, Nishino S, Sasano M, Oka N, Yamashita H, Kai M, Nakao A, Kotani J, Usami M. Endogenous interleukin 18 regulates testicular germ cell apoptosis during endotoxemia. Reproduction 2015; 150:105-14. [PMID: 25934945 DOI: 10.1530/rep-14-0427] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 05/01/2015] [Indexed: 01/26/2023]
Abstract
Orchitis (testicular swelling) often occurs during systemic inflammatory conditions, such as sepsis. Interleukin 18 (IL18) is a proinflammatory cytokine and is an apoptotic mediator during endotoxemia, but the role of IL18 in response to inflammation in the testes was unclear. WT and IL18 knockout (KO) mice were injected lipopolysaccharide (LPS) to induce endotoxemia and examined 12 and 48 h after LPS administration to model the acute and recovery phases of endotoxemia. Caspase activation was assessed using immunohistochemistry. Protein and mRNA expression were examined by western blot and quantitative real-time RT-PCR respectively. During the acute phase of endotoxemia, apoptosis (as indicated by caspase-3 cleavage) was increased in WT mice but not in IL18 KO mice. The death receptor-mediated and mitochondrial-mediated apoptotic pathways were both activated in the WT mice but not in the KO mice. During the recovery phase of endotoxemia, apoptosis was observed in the IL18 KO mice but not in the WT mice. Activation of the death-receptor mediated apoptotic pathway could be seen in the IL18 KO mice but not the WT mice. These results suggested that endogenous IL18 induces germ cell apoptosis via death receptor mediated- and mitochondrial-mediated pathways during the acute phase of endotoxemia and suppresses germ cell apoptosis via death-receptor mediated pathways during recovery from endotoxemia. Taken together, IL18 could be a new therapeutic target to prevent orchitis during endotoxemia.
Collapse
Affiliation(s)
- Taketo Inoue
- Department of BiophysicsKobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, 654-0142 Hyogo, JapanOno Ladies Clinic538-3, Nishihonmachi, Ono, 675-1375 Hyogo, JapanDepartment of EmergencyDisaster and Critical Care Medicine, Hyogo College of Medicine, 1-1, Mukogawa-cho, Nishinomiya, 663-8501 Hyogo, Japan Department of BiophysicsKobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, 654-0142 Hyogo, JapanOno Ladies Clinic538-3, Nishihonmachi, Ono, 675-1375 Hyogo, JapanDepartment of EmergencyDisaster and Critical Care Medicine, Hyogo College of Medicine, 1-1, Mukogawa-cho, Nishinomiya, 663-8501 Hyogo, Japan
| | - Michiko Aoyama-Ishikawa
- Department of BiophysicsKobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, 654-0142 Hyogo, JapanOno Ladies Clinic538-3, Nishihonmachi, Ono, 675-1375 Hyogo, JapanDepartment of EmergencyDisaster and Critical Care Medicine, Hyogo College of Medicine, 1-1, Mukogawa-cho, Nishinomiya, 663-8501 Hyogo, Japan Department of BiophysicsKobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, 654-0142 Hyogo, JapanOno Ladies Clinic538-3, Nishihonmachi, Ono, 675-1375 Hyogo, JapanDepartment of EmergencyDisaster and Critical Care Medicine, Hyogo College of Medicine, 1-1, Mukogawa-cho, Nishinomiya, 663-8501 Hyogo, Japan
| | - Shingo Kamoshida
- Department of BiophysicsKobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, 654-0142 Hyogo, JapanOno Ladies Clinic538-3, Nishihonmachi, Ono, 675-1375 Hyogo, JapanDepartment of EmergencyDisaster and Critical Care Medicine, Hyogo College of Medicine, 1-1, Mukogawa-cho, Nishinomiya, 663-8501 Hyogo, Japan
| | - Satoshi Nishino
- Department of BiophysicsKobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, 654-0142 Hyogo, JapanOno Ladies Clinic538-3, Nishihonmachi, Ono, 675-1375 Hyogo, JapanDepartment of EmergencyDisaster and Critical Care Medicine, Hyogo College of Medicine, 1-1, Mukogawa-cho, Nishinomiya, 663-8501 Hyogo, Japan
| | - Maki Sasano
- Department of BiophysicsKobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, 654-0142 Hyogo, JapanOno Ladies Clinic538-3, Nishihonmachi, Ono, 675-1375 Hyogo, JapanDepartment of EmergencyDisaster and Critical Care Medicine, Hyogo College of Medicine, 1-1, Mukogawa-cho, Nishinomiya, 663-8501 Hyogo, Japan
| | - Nobuki Oka
- Department of BiophysicsKobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, 654-0142 Hyogo, JapanOno Ladies Clinic538-3, Nishihonmachi, Ono, 675-1375 Hyogo, JapanDepartment of EmergencyDisaster and Critical Care Medicine, Hyogo College of Medicine, 1-1, Mukogawa-cho, Nishinomiya, 663-8501 Hyogo, Japan
| | - Hayato Yamashita
- Department of BiophysicsKobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, 654-0142 Hyogo, JapanOno Ladies Clinic538-3, Nishihonmachi, Ono, 675-1375 Hyogo, JapanDepartment of EmergencyDisaster and Critical Care Medicine, Hyogo College of Medicine, 1-1, Mukogawa-cho, Nishinomiya, 663-8501 Hyogo, Japan
| | - Motoki Kai
- Department of BiophysicsKobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, 654-0142 Hyogo, JapanOno Ladies Clinic538-3, Nishihonmachi, Ono, 675-1375 Hyogo, JapanDepartment of EmergencyDisaster and Critical Care Medicine, Hyogo College of Medicine, 1-1, Mukogawa-cho, Nishinomiya, 663-8501 Hyogo, Japan
| | - Atsunori Nakao
- Department of BiophysicsKobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, 654-0142 Hyogo, JapanOno Ladies Clinic538-3, Nishihonmachi, Ono, 675-1375 Hyogo, JapanDepartment of EmergencyDisaster and Critical Care Medicine, Hyogo College of Medicine, 1-1, Mukogawa-cho, Nishinomiya, 663-8501 Hyogo, Japan
| | - Joji Kotani
- Department of BiophysicsKobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, 654-0142 Hyogo, JapanOno Ladies Clinic538-3, Nishihonmachi, Ono, 675-1375 Hyogo, JapanDepartment of EmergencyDisaster and Critical Care Medicine, Hyogo College of Medicine, 1-1, Mukogawa-cho, Nishinomiya, 663-8501 Hyogo, Japan
| | - Makoto Usami
- Department of BiophysicsKobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, 654-0142 Hyogo, JapanOno Ladies Clinic538-3, Nishihonmachi, Ono, 675-1375 Hyogo, JapanDepartment of EmergencyDisaster and Critical Care Medicine, Hyogo College of Medicine, 1-1, Mukogawa-cho, Nishinomiya, 663-8501 Hyogo, Japan
| |
Collapse
|
38
|
Shin JA, Hong IS, Oh S, Cho SD, Lee KE. Direct effect of streptozotocin on periodontal ligament cells through myeloid cell leukemia-1. J Periodontal Res 2015; 50:807-13. [PMID: 25824804 DOI: 10.1111/jre.12268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2015] [Indexed: 01/07/2023]
Abstract
OBJECTIVE We hypothesized that streptozotocin (STZ) has a direct impact on periodontal ligament cell (PDL) damage as a potential direct inducer of periodontitis. BACKGROUND Since diabetes was accepted as one of the risk factors for the development of periodontal disease, various scientific studies have been undertaken in the STZ-induced periodontal disease models. STZ induces β-cell damage and subsequent diabetes development in vivo. Until now, assessment of the impacts of STZ-induced experimental diabetes on periodontitis has generally been conducted on the fundamental assumption that STZ have no direct action on PDL and its function. However, several recent studies suggest that STZ also directly affect many different biological functions in various tissues or organs. MATERIAL AND METHODS To assess the apoptotic effects of STZ on PDLs, they were treated with or without STZ at different concentrations. Qualitative estimation of apoptotic cell death was obtained by live/dead assay. The expression levels of apoptosis-related proteins were evaluated by western blot analysis. RESULTS STZ inhibits growth and induces apoptosis in PDLs in a dose-dependent manner. Furthermore, STZ dramatically induced Mcl-1 downregulation in a proteasome-dependent manner and thereby induced apoptosis of PDLs through the Bak/Bax apoptotic signaling pathway. CONCLUSION Our results support the hypothesis that suppression of the cellular Mcl-1 levels by STZ may be at least partly attributed to the development of periodontitis in STZ-induced diabetic animal models.
Collapse
Affiliation(s)
- J-A Shin
- Department of Oral Pathology, School of Dentistry and Institute of Biodegradable Material, Institute of Oral Bioscience, Brain Korea 21 Project, Chonbuk National University, Jeon-ju, Korea
| | - I-S Hong
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, South Korea.,Lee Gil Ya Cancer and Diabetes Institute, Incheon, South Korea
| | - S Oh
- Department of Oral Pathology, School of Dentistry and Institute of Biodegradable Material, Institute of Oral Bioscience, Brain Korea 21 Project, Chonbuk National University, Jeon-ju, Korea
| | - S-D Cho
- Department of Oral Pathology, School of Dentistry and Institute of Biodegradable Material, Institute of Oral Bioscience, Brain Korea 21 Project, Chonbuk National University, Jeon-ju, Korea
| | - K-E Lee
- Department of Oral Medicine, School of Dentistry and Research Institute of Clinical Medicine of Chonbuk, National University, Biomedical Research Institute, Chonbuk National University Hospital, Jeonju, Korea
| |
Collapse
|
39
|
ZHANG YANG, ZHU XIAOBO, HOU KUN, ZHAO JINCHUAN, HAN ZHIGUO, ZHANG XIAONA. Mcl-1 downregulation sensitizes glioma to bortezomib-induced apoptosis. Oncol Rep 2015; 33:2277-84. [DOI: 10.3892/or.2015.3875] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 02/10/2015] [Indexed: 11/06/2022] Open
|
40
|
Zhu X, Zhang K, Wang Q, Chen S, Gou Y, Cui Y, Li Q. Cisplatin-mediated c-myc overexpression and cytochrome c (cyt c) release result in the up-regulation of the death receptors DR4 and DR5 and the activation of caspase 3 and caspase 9, likely responsible for the TRAIL-sensitizing effect of cisplatin. Med Oncol 2015; 32:133. [PMID: 25796504 DOI: 10.1007/s12032-015-0588-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 03/14/2015] [Indexed: 01/25/2023]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) reverses multidrug resistance (MDR) and induces apoptosis in MDR gastric carcinoma cells. In our previous study, cisplatin proved to be a sensitizing agent for TRAIL. To study the synergistic effects of cisplatin and TRAIL, we investigated the mechanism by which TRAIL reverses multidrug resistance, the role of c-myc in modulating the death receptors DR4 and DR5 and the relationship between cisplatin and cytochrome c (cyt c) release in SGC7901/VCR and SGC7901/DDP cells. We found that after treatment with TRAIL, the DNA-PKcs/Akt/GSK-3β pathway, which is positively correlated with the levels of MDR1 and MRP1, was significantly inhibited and that this tendency can be abolished by Z-DEVD-FMK (a specific caspase 3 inhibitor). We also found that suppression of c-myc by siRNA reduced the expression of DR4 and DR5 and that transfection with a pAVV-c-myc expression vector increased the expression of DR4 and DR5. Moreover, cisplatin increased the expression of c-myc in the presence of TRAIL, and there is a clear increase in cyt c release from mitochondria with the increasing concentrations of cisplatin. Meanwhile, the intrinsic death receptor pathway of caspase 9, as well as the common intrinsic and extrinsic downstream target, caspase 3, was potently activated by the release of cyt c. Together, we conclude that in TRAIL-treated MDR gastric carcinoma cells, cisplatin induces the death receptors DR4 and DR5 through the up-regulation of c-myc and strengthens the activation of caspases via promoting the release of cyt c. These effects would then be responsible for the TRAIL sensitization effect of cisplatin.
Collapse
Affiliation(s)
- Xingchao Zhu
- Department of Gastroenterology, Affiliated Provincial Hospital of Anhui Medical University, 17 Lu Jiang Road, Hefei, 230001, Anhui Province, China
| | | | | | | | | | | | | |
Collapse
|
41
|
Murphy ÁC, Weyhenmeyer B, Noonan J, Kilbride SM, Schimansky S, Loh KP, Kögel D, Letai AG, Prehn JHM, Murphy BM. Modulation of Mcl-1 sensitizes glioblastoma to TRAIL-induced apoptosis. Apoptosis 2015; 19:629-42. [PMID: 24213561 PMCID: PMC3938842 DOI: 10.1007/s10495-013-0935-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Glioblastoma (GBM) is the most aggressive form of primary brain tumour, with dismal patient outcome. Treatment failure is associated with intrinsic or acquired apoptosis resistance and the presence of a highly tumourigenic subpopulation of cancer cells called GBM stem cells. Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) has emerged as a promising novel therapy for some treatment-resistant tumours but unfortunately GBM can be completely resistant to TRAIL monotherapy. In this study, we identified Mcl-1, an anti-apoptotic Bcl-2 family member, as a critical player involved in determining the sensitivity of GBM to TRAIL-induced apoptosis. Effective targeting of Mcl-1 in TRAIL resistant GBM cells, either by gene silencing technology or by treatment with R-roscovitine, a cyclin-dependent kinase inhibitor that targets Mcl-1, was demonstrated to augment sensitivity to TRAIL, both within GBM cells grown as monolayers and in a 3D tumour model. Finally, we highlight that two separate pathways are activated during the apoptotic death of GBM cells treated with a combination of TRAIL and R-roscovitine, one which leads to caspase-8 and caspase-3 activation and a second pathway, involving a Mcl-1:Noxa axis. In conclusion, our study demonstrates that R-roscovitine in combination with TRAIL presents a promising novel strategy to trigger cell death pathways in glioblastoma.
Collapse
Affiliation(s)
- Á C Murphy
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, York House, St. Stephen's Green, Dublin, 2, Ireland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
From nature to bedside: Pro-survival and cell death mechanisms as therapeutic targets in cancer treatment. Biotechnol Adv 2014; 32:1111-22. [DOI: 10.1016/j.biotechadv.2014.03.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 03/04/2014] [Accepted: 03/04/2014] [Indexed: 12/11/2022]
|
43
|
Yu HJ, Shin JA, Jung JY, Nam JS, Hong IS, Cho NP, Cho SD. Inhibition of myeloid cell leukemia-1: Association with sorafenib-induced apoptosis in human mucoepidermoid carcinoma cells and tumor xenograft. Head Neck 2014; 37:1326-35. [PMID: 25043125 DOI: 10.1002/hed.23749] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 03/24/2014] [Accepted: 05/07/2014] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The purpose of our study was to investigate the anticancer effect of sorafenib on mucoepidermoid carcinoma (MEC) and find its new molecular mechanism. METHODS The apoptotic effects of sorafenib were performed using MTS assay, diamidino-phenylindole (DAPI) staining, Western blotting, reverse transcription-polymerase chain reaction (RT-PCR), siRNA, and xenograft. RESULTS Sorafenib had apoptotic effects on MC-3 and YD15 cells and decreased myeloid cell leukemia-1 (Mcl-1) through proteasome-dependent protein degradation and the inhibition of protein synthesis. Sorafenib significantly affected truncated bid (t-Bid) and siMcl-1 resulting in the upregulation of t-Bid to induce apoptosis. Signal transducer and activator of transcription 3 (STAT3) phosphorylation was also blocked by sorafenib and a potent STAT3 inhibitor, cryptotanshinone clearly induced poly ADP-ribose polymerase (PARP) cleavage by inhibiting Mcl-1 and increasing t-Bid. Finally, administration of sorafenib significantly suppressed tumor growth and induced apoptosis in tumor xenograft model in association with downregulation of Mcl-1 without any side effects. CONCLUSION Taken together, these findings suggest that sorafenib can be a good anticancer drug candidate for the treatment of MEC.
Collapse
Affiliation(s)
- Hyun-Ju Yu
- Department of Oral Pathology, School of Dentistry, and Institute of Oral Bioscience, Chonbuk National University, Jeonju, Republic of Korea
| | - Ji-Ae Shin
- Department of Oral Pathology, School of Dentistry, and Institute of Oral Bioscience, Chonbuk National University, Jeonju, Republic of Korea
| | - Ji-Youn Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Republic of Korea
| | - Jeong-Seok Nam
- Lee Gil Ya Cancer and Diabetes Institute, Inchon, Republic of Korea
| | - In-Sun Hong
- Department of Molecular Medicine, Gachon University, Incheon, Republic of Korea
| | - Nam-Pyo Cho
- Department of Oral Pathology, School of Dentistry, and Institute of Oral Bioscience, Chonbuk National University, Jeonju, Republic of Korea
| | - Sung-Dae Cho
- Department of Oral Pathology, School of Dentistry, and Institute of Oral Bioscience, Chonbuk National University, Jeonju, Republic of Korea
| |
Collapse
|
44
|
Zhao RN, Fan S, Han JG, Liu G. Molecular dynamics study of segment peptides of Bax, Bim, and Mcl-1 BH3 domain of the apoptosis-regulating proteins bound to the anti-apoptotic Mcl-1 protein. J Biomol Struct Dyn 2014; 33:1067-81. [PMID: 24865469 DOI: 10.1080/07391102.2014.929028] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Mcl-1 has emerged as a potential therapeutic target in the treatment of several malignancies. Peptides representing BH3 region of pro-apoptotic proteins have been shown to bind the hydrophobic cleft of anti-apoptotic Mcl-1 and this segment is responsible for modulating the apoptotic pathways in living cells. Understanding the molecular basis of protein-peptide interaction is required to develop potent inhibitors specific for Mcl-1. Molecular dynamics simulations were performed for Mcl-1 in complex with three different BH3 peptides derived from Mcl-1, Bax, and Bim. Accordingly, the calculated binding free energies using MM-PBSA method are obtained and comparison with the experimentally determined binding free energies is made. The interactions involving two conserved charged residues (Aspi, and Arg/Lysi-4) and three upstream conserved hydrophobic residues (Leui-5, Ile/Vali-2, and Glyi-1, respectively) of BH3 peptides play the important roles in the structural stability of the complexes. The calculated results exhibit that the interactions of Bim BH3 peptides to Mcl-1 is stronger than the complex with Bax 19BH3 peptides. The hydrophobic residues (position i - 9, i - 8 and i + 2) of BH3 peptides can be involved in their inhibitory specificity. The calculated results can be used for designing more effective MCL-1 inhibitors.
Collapse
Affiliation(s)
- Run-Ning Zhao
- a Institute of Applied Mathematics and Physics , Shanghai Dianji University , Shanghai 201306 , P.R. China
| | | | | | | |
Collapse
|
45
|
Lemke J, von Karstedt S, Zinngrebe J, Walczak H. Getting TRAIL back on track for cancer therapy. Cell Death Differ 2014; 21:1350-64. [PMID: 24948009 PMCID: PMC4131183 DOI: 10.1038/cdd.2014.81] [Citation(s) in RCA: 369] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/14/2014] [Accepted: 04/15/2014] [Indexed: 02/07/2023] Open
Abstract
Unlike other members of the TNF superfamily, the TNF-related apoptosis-inducing ligand (TRAIL, also known as Apo2L) possesses the unique capacity to induce apoptosis selectively in cancer cells in vitro and in vivo. This exciting discovery provided the basis for the development of TRAIL-receptor agonists (TRAs), which have demonstrated robust anticancer activity in a number of preclinical studies. Subsequently initiated clinical trials testing TRAs demonstrated, on the one hand, broad tolerability but revealed, on the other, that therapeutic benefit was rather limited. Several factors that are likely to account for TRAs' sobering clinical performance have since been identified. First, because of initial concerns over potential hepatotoxicity, TRAs with relatively weak agonistic activity were selected to enter clinical trials. Second, although TRAIL can induce apoptosis in several cancer cell lines, it has now emerged that many others, and importantly, most primary cancer cells are resistant to TRAIL monotherapy. Third, so far patients enrolled in TRA-employing clinical trials were not selected for likelihood of benefitting from a TRA-comprising therapy on the basis of a valid(ated) biomarker. This review summarizes and discusses the results achieved so far in TRA-employing clinical trials in the light of these three shortcomings. By integrating recent insight on apoptotic and non-apoptotic TRAIL signaling in cancer cells, we propose approaches to introduce novel, revised TRAIL-based therapeutic concepts into the cancer clinic. These include (i) the use of recently developed highly active TRAs, (ii) the addition of efficient, but cancer-cell-selective TRAIL-sensitizing agents to overcome TRAIL resistance and (iii) employing proteomic profiling to uncover resistance mechanisms. We envisage that this shall enable the design of effective TRA-comprising therapeutic concepts for individual cancer patients in the future.
Collapse
Affiliation(s)
- J Lemke
- 1] Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK [2] Clinic of General and Visceral Surgery, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - S von Karstedt
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - J Zinngrebe
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - H Walczak
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| |
Collapse
|
46
|
Abstract
Members of the Bcl-2 protein family are frequently deregulated in tumors as they critically control cell death induction in mammalian cells. Alterations of these proteins may cause resistance to chemotherapy-induced cell death and immune responses. By serendipity we cloned a variant of the anti-apoptotic Bcl2-family member Myeloid cell leukemia-1 (Mcl1) from human neuroblastoma and leukemia cells. This Mcl1L variant lacks a 45 bp sequence that codes for 15 highly conserved amino acids ranging from Gly158 to Asp172. This region is part of the so called PEST-sequence of Mcl1L and contains two phosphorylation sites (Ser159 and Thr163) that regulate Mcl1L stability. A caspase 3/caspase 8 cleavage site at Asp157 which has been reported to be critical for death-receptor-induced apoptosis and for the conversion of Mcl1L into a pro-apoptotic protein is also missing in this novel variant. Importantly, Mcl1LdelGly158-Asp172 bound significantly more pro-apoptotic Bim compared to Mcl1L and showed increased anti-proliferative and anti-apoptotic activity compared to Mcl1L during death receptor-induced cell death. This suggests that this novel Mcl1L variant efficiently protects tumor cells against extrinsic death signalling and therefore may provide a survival advantage for highly aggressive tumors.
Collapse
|
47
|
Evodiamine induces apoptosis and enhances TRAIL-induced apoptosis in human bladder cancer cells through mTOR/S6K1-mediated downregulation of Mcl-1. Int J Mol Sci 2014; 15:3154-71. [PMID: 24566141 PMCID: PMC3958903 DOI: 10.3390/ijms15023154] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 02/13/2014] [Accepted: 02/14/2014] [Indexed: 12/25/2022] Open
Abstract
The tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), either alone or in combination with other anti-cancer agents, has been considered as a new strategy for anti-cancer therapy. In this study, we demonstrated that evodiamine, a quinolone alkaloid isolated from the fruit of Evodia fructus, induced apoptosis and enhanced TRAIL-induced apoptosis in human bladder cancer cells. To elucidate the underlying mechanism, we found that evodiamine significantly reduced the protein levels of Mcl-1 in 253J and T24 bladder cancer cells, and overexpression of this molecule attenuated the apoptosis induced by evodiamine alone, or in combination with TRAIL. Further experiments revealed that evodiamine did not affect the mRNA level, proteasomal degradation and protein stability of Mcl-1. On the other hand, evodiamine inhibited the mTOR/S6K1 pathway, which usually regulates protein translation; moreover, knockdown of S6K1 with small interfering RNA (siRNA) effectively reduced Mcl-1 levels, indicating evodiamine downregulates c-FLIP through inhibition of mTOR/S6K1 pathway. Taken together, our results indicate that evodiamine induces apoptosis and enhances TRAIL-induced apoptosis possibly through mTOR/S6K1-mediated downregulation of Mcl-1; furthermore, these findings provide a rationale for the combined application of evodiamine with TRAIL in the treatment of bladder cancer.
Collapse
|
48
|
Ionizing radiation-inducible microRNA miR-193a-3p induces apoptosis by directly targeting Mcl-1. Apoptosis 2013; 18:896-909. [PMID: 23546867 DOI: 10.1007/s10495-013-0841-7] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The functions of microRNAs (miRNAs) as either oncogenes or tumor suppressors in regulating cancer-related events have been established. We analyzed the alterations in the miRNA expression profile of the glioma cell line U-251 caused by ionizing radiation (IR) by using an miRNA array and identified several miRNAs whose expression was significantly affected by IR. Among the IR-responsive miRNAs, we further examined the function of miR-193a-3p, which exhibited the most significant growth-inhibiting effect. miR-193a-3p was observed to induce apoptosis in both U-251 and HeLa cells. We also demonstrated that miR-193a-3p induces the accumulation of intracellular reactive oxygen species (ROS) and DNA damage as determined by the level of γH2AX and by performing the comet assay. The induction of both apoptosis and DNA damage by miR-193a-3p was blocked by antioxidant treatment, indicating the crucial role of ROS in the action of miR-193a-3p. Among the putative target proteins, the expression of Mcl-1, an anti-apoptotic Bcl-2 family member, decreased because of miR-193a-3p transfection. A reporter assay using a luciferase construct containing the 3'-untranslated region of Mcl-1 confirmed that Mcl-1 is a direct target of miR-193a-3p. Down-regulation of Mcl-1 by siRNA transfection closely mimicked the outcome of miR-193a-3p transfection showing increased ROS, DNA damage, cytochrome c release, and apoptosis. Ectopic expression of Mcl-1 suppressed the pro-apoptotic action of miR-193a-3p, suggesting that Mcl-1 depletion is critical for miR-193a-3p induced apoptosis. Collectively, our results suggest a novel function for miR-193a-3p and its potential application in cancer therapy.
Collapse
|
49
|
Chan SH, Leu WJ, Hsu LC, Chang HS, Hwang TL, Chen IS, Chen CS, Guh JH. Reevesioside F induces potent and efficient anti-proliferative and apoptotic activities through Na⁺/K⁺-ATPase α3 subunit-involved mitochondrial stress and amplification of caspase cascades. Biochem Pharmacol 2013; 86:1564-75. [PMID: 24099795 DOI: 10.1016/j.bcp.2013.09.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 09/26/2013] [Accepted: 09/27/2013] [Indexed: 11/28/2022]
Abstract
Reevesioside F, isolated from Reevesia formosana, induced anti-proliferative activity that was highly correlated with the expression of Na⁺/K⁺-ATPase α₃ subunit in several cell lines, including human leukemia HL-60 and Jurkat cells, and some other cell lines. Knockdown of α₃ subunit significantly inhibited cell apoptosis suggesting a crucial role of the α₃ subunit. Reevesioside F induced a rapid down-regulation of survivin protein, followed by release of cytochrome c from mitochondria and loss of mitochondrial membrane potential (ΔΨm). Further examination demonstrated the mitochondrial damage in leukemic cells through Mcl-1 down-regulation, Noxa up-regulation and an increase of the formation of truncated Bid, tBim and a 23-kDa cleaved Bcl-2 fragment. Furthermore, reevesioside F induced an increase of mitochondria-associated acetyl α-tubulin that may also contribute to apoptosis. The caspase cascade was profoundly activated by reevesioside F. Notably, the specific caspase-3 inhibitor z-DEVD-fmk significantly blunted reevesioside F-induced loss of ΔΨm and apoptosis, suggesting that caspase-3 activation may further amplify mitochondrial damage and apoptotic signaling cascade. In spite of being a cardiac glycoside, reevesioside F did not increase the intracellular Ca²⁺ levels. Moreover, CGP-37157 which blocked Na⁺/Ca²⁺ exchanger on plasma membrane and mitochondria did not modify reevesioside F-mediated effect. In summary, the data suggest that reevesioside F induces apoptosis through the down-regulation of survivin and Mcl-1, and the formation of pro-apoptotic fragments from Bcl-2 family members. The loss of ΔΨm and mitochondrial damage are responsible for the activation of caspases. Moreover, the amplification of caspase-3-mediated signaling pathway contributes largely to the execution of apoptosis in leukemic cells.
Collapse
Affiliation(s)
- She-Hung Chan
- School of Pharmacy, National Taiwan University, No.1, Sect. 1, Jen-Ai Rd, Taipei 100, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Kumar D, Gopalakrishna R, Singh AP, Ranjan R, Pandey SK, Sarkhel BC. Developmental potency of pre-implant parthenogenetic goat embryos: effect of activation protocols and culture media. In Vitro Cell Dev Biol Anim 2013; 50:1-6. [PMID: 23982915 DOI: 10.1007/s11626-013-9680-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 08/02/2013] [Indexed: 11/30/2022]
Abstract
The developmental potency of pre-implant parthenogentic goat embryos were compared under two chemical activation protocols in three different culture media groups. The in vitro matured oocytes were chemically activated by two protocols viz. P1 (CB-CHX-6DMAP) and P2 (Ca-CHX-6DMAP). The activated oocytes under both the protocols were developed in three culture media, viz. modified synthetic oviductal fluid (mSOF), research vitro cleave medium (RVCL), and RVCL-Blast. While comparing the developmental potential of activated oocytes, it was observed that the oocytes activated under P2 protocol pooled over three culture media group producing significantly higher mean cleavage rate (43.2±0.9 vs 40.6±1.5), blastocyst development (16.4±1.1 vs 12.6±0.8), and blastomere count (120.7±4.7 vs 113.2±4.1) as compared to P1 protocol. The comparison of effect of culture media pooled over protocol groups revealed that the mean cleavage rate observed under RVCL-Blast (44.8±1.3) and RVCL (45.3±0.5) were significantly higher (P≤0.01) than mSOF (35.8±1.2). However, the mean blastocyst development observed under RVCL-Blast group (18.8±3.2) was significantly higher than RVCL (14.0±0.8) and mSOF (10.8±0.4). Similarly, the mean blastomere count under RVCL-Blast group (136.0±3.7) was significantly higher (P≤0.01) than RVCL (114.7±1.0) and mSOF (100.2±0.5) groups. The semiquantitative RT PCR analysis showed the expression of pro-apoptotic caspase 3 gene in P1 and anti-apoptotic Mcl-1 gene in P2. This study concludes that the activation protocol P2 and embryo cultured under RVCL-Blast group were optimum for chemical activation and culture medium, respectively.
Collapse
Affiliation(s)
- Dharmendra Kumar
- Nanaji Deshmukh Veterinary Sciences University, Jabalpur, Madhya Pradesh, India
| | | | | | | | | | | |
Collapse
|