1
|
Balamurli G, Liew AQX, Tee WW, Pervaiz S. Interplay between epigenetics, senescence and cellular redox metabolism in cancer and its therapeutic implications. Redox Biol 2024; 78:103441. [PMID: 39612910 PMCID: PMC11629570 DOI: 10.1016/j.redox.2024.103441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024] Open
Abstract
There is accumulating evidence indicating a close crosstalk between key molecular events regulating cell growth and proliferation, which could profoundly impact carcinogenesis and its progression. Here we focus on reviewing observations highlighting the interplay between epigenetic modifications, irreversible cell cycle arrest or senescence, and cellular redox metabolism. Epigenetic alterations, such as DNA methylation and histone modifications, dynamically influence tumour transcriptome, thereby impacting tumour phenotype, survival, growth and spread. Interestingly, the acquisition of senescent phenotype can be triggered by epigenetic changes, acting as a double-edged sword via its ability to suppress tumorigenesis or by facilitating an inflammatory milieu conducive for cancer progression. Concurrently, an aberrant redox metabolism, which is a function of the balance between reactive oxygen species (ROS) generation and intracellular anti-oxidant defences, influences signalling cascades and genomic stability in cancer cells by serving as a critical link between epigenetics and senescence. Recognizing this intricate interconnection offers a nuanced perspective for therapeutic intervention by simultaneously targeting specific epigenetic modifications, modulating senescence dynamics, and restoring redox homeostasis.
Collapse
Affiliation(s)
- Geoffrey Balamurli
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, NUS, Singapore; Chromatin Dynamics and Disease Epigenetics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Angeline Qiu Xia Liew
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore; Integrative Science and Engineering Programme (ISEP), NUS Graduate School (NUSGS), NUS, Singapore
| | - Wee Wei Tee
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, NUS, Singapore; Chromatin Dynamics and Disease Epigenetics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, NUS, Singapore; Integrative Science and Engineering Programme (ISEP), NUS Graduate School (NUSGS), NUS, Singapore; NUS Medicine Healthy Longevity Program, NUS, Singapore; National University Cancer Institute, National University Health System, Singapore.
| |
Collapse
|
2
|
Wu Z, Ma Y, Chen S, Liu Y, Liu X, Cao H, Jin T, Li L, Huang M, Yang F, Dong W. Arginine Biosynthesis Mediates Wulingzhi Extract Resistance to Busulfan-Induced Male Reproductive Toxicity. Int J Mol Sci 2024; 25:6320. [PMID: 38928028 PMCID: PMC11203605 DOI: 10.3390/ijms25126320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/01/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Busulfan, an indispensable medicine in cancer treatment, can cause serious reproductive system damage to males as a side effect of its otherwise excellent therapeutic results. Its widespread use has also caused its accumulation in the environment and subsequent ecotoxicology effects. As a Chinese medicine, Wulingzhi (WLZ) has the effects of promoting blood circulation and improving female reproductive function. However, the potential effects of WLZ in male reproduction and in counteracting busulfan-induced testis damage, as well as its probable mechanisms, are still ambiguous. In this study, busulfan was introduced in a mouse model to evaluate its production of the testicular damage. The components of different WLZ extracts were compared using an untargeted metabolome to select extracts with greater efficacy, which were further confirmed in vivo. Here, we demonstrate abnormal spermatogenesis and low sperm quality in busulfan-injured testes. The WLZ extracts showed a strong potential to rehabilitate the male reproductive system; this effect was more prominent in room-temperature extracts. Additionally, both water and ethanol WLZ extracts at room temperature alleviated various busulfan-induced adverse effects. In particular, WLZ recovered spermatogenesis, re-activated arginine biosynthesis, and alleviated the increased oxidative stress and inflammation in the testis, ultimately reversing the busulfan-induced testicular injury. Collectively, these results suggest a promising approach to protecting the male reproductive system from busulfan-induced adverse side effects, as well as those of other similar anti-cancer drugs.
Collapse
Affiliation(s)
- Zifang Wu
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (Z.W.); (Y.M.); (S.C.); (Y.L.); (H.C.); (T.J.); (L.L.); (M.H.)
| | - Yuxuan Ma
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (Z.W.); (Y.M.); (S.C.); (Y.L.); (H.C.); (T.J.); (L.L.); (M.H.)
| | - Shaoxian Chen
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (Z.W.); (Y.M.); (S.C.); (Y.L.); (H.C.); (T.J.); (L.L.); (M.H.)
| | - Yuyan Liu
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (Z.W.); (Y.M.); (S.C.); (Y.L.); (H.C.); (T.J.); (L.L.); (M.H.)
| | - Xianglin Liu
- College of Forestry, Northwest A&F University, Xianyang 712100, China;
| | - Heran Cao
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (Z.W.); (Y.M.); (S.C.); (Y.L.); (H.C.); (T.J.); (L.L.); (M.H.)
| | - Tianqi Jin
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (Z.W.); (Y.M.); (S.C.); (Y.L.); (H.C.); (T.J.); (L.L.); (M.H.)
| | - Long Li
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (Z.W.); (Y.M.); (S.C.); (Y.L.); (H.C.); (T.J.); (L.L.); (M.H.)
| | - Mengqi Huang
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (Z.W.); (Y.M.); (S.C.); (Y.L.); (H.C.); (T.J.); (L.L.); (M.H.)
| | - Fangxia Yang
- College of Forestry, Northwest A&F University, Xianyang 712100, China;
| | - Wuzi Dong
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (Z.W.); (Y.M.); (S.C.); (Y.L.); (H.C.); (T.J.); (L.L.); (M.H.)
- College of Forestry, Northwest A&F University, Xianyang 712100, China;
| |
Collapse
|
3
|
Iskandar K, Foo J, Liew AQX, Zhu H, Raman D, Hirpara JL, Leong YY, Babak MV, Kirsanova AA, Armand AS, Oury F, Bellot G, Pervaiz S. A novel MTORC2-AKT-ROS axis triggers mitofission and mitophagy-associated execution of colorectal cancer cells upon drug-induced activation of mutant KRAS. Autophagy 2024; 20:1418-1441. [PMID: 38261660 PMCID: PMC11210925 DOI: 10.1080/15548627.2024.2307224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/02/2024] [Accepted: 01/13/2024] [Indexed: 01/25/2024] Open
Abstract
RAS is one of the most commonly mutated oncogenes associated with multiple cancer hallmarks. Notably, RAS activation induces intracellular reactive oxygen species (ROS) generation, which we previously demonstrated as a trigger for autophagy-associated execution of mutant KRAS-expressing cancer cells. Here we report that drug (merodantoin; C1)-induced activation of mutant KRAS promotes phospho-AKT S473-dependent ROS-mediated S616 phosphorylation and mitochondrial localization of DNM1L/DRP1 (dynamin 1 like) and cleavage of the fusion-associated protein OPA1 (OPA1 mitochondrial dynamin like GTPase). Interestingly, accumulation of the outer mitochondrial membrane protein VDAC1 (voltage dependent anion channel 1) is observed in mutant KRAS-expressing cells upon exposure to C1. Conversely, silencing VDAC1 abolishes C1-induced mitophagy, and gene knockdown of either KRAS, AKT or DNM1L rescues ROS-dependent VDAC1 accumulation and stability, thus suggesting an axis of mutant active KRAS-phospho-AKT S473-ROS-DNM1L-VDAC1 in mitochondrial morphology change and cancer cell execution. Importantly, we identified MTOR (mechanistic target of rapamycin kinsase) complex 2 (MTORC2) as the upstream mediator of AKT phosphorylation at S473 in our model. Pharmacological or genetic inhibition of MTORC2 abrogated C1-induced phosphorylation of AKT S473, ROS generation and mitophagy induction, as well as rescued tumor colony forming ability and migratory capacity. Finally, increase in thermal stability of KRAS, AKT and DNM1L were observed upon exposure to C1 only in mutant KRAS-expressing cells. Taken together, our work has unraveled a novel mechanism of selective targeting of mutant KRAS-expressing cancers via MTORC2-mediated AKT activation and ROS-dependent mitofission, which could have potential therapeutic implications given the relative lack of direct RAS-targeting strategies in cancer.Abbreviations: ACTB/ß-actin: actin beta; AKT: AKT serine/threonine kinase; C1/merodantoin: 1,3-dibutyl-2-thiooxo-imidazoldine-4,5-dione; CAT: catalase; CETSA: cellular thermal shift assay; CHX: cycloheximide; DKO: double knockout; DNM1L/DRP1: dynamin 1 like; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; H2O2: hydrogen peroxide; HSPA1A/HSP70-1: heat shock protein family A (Hsp70) member 1A; HSP90AA1/HSP90: heat shock protein 90 alpha family class A member 1; KRAS: KRAS proto-oncogene, GTPase; MAP1LC3B/LC3B, microtubule associated protein 1 light chain 3 beta; LC3B-I: unlipidated form of LC3B; LC3B-II: phosphatidylethanolamine-conjugated form of LC3B; MAPKAP1/SIN1: MAPK associated protein 1; MAPK1/ERK2: mitogen-activated protein kinase 1; MAPK3/ERK1: mitogen-activated protein kinase 3; MFI: mean fluorescence intensity; MiNA: Mitochondrial Network Analysis; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin kinase complex 1; MTORC2: mechanistic target of rapamycin kinase complex 2; O2.-: superoxide; OMA1: OMA1 zinc metallopeptidase; OPA1: OPA1 mitochondrial dynamin like GTPase; RICTOR: RPTOR independent companion of MTOR complex 2; ROS: reactive oxygen species; RPTOR/raptor: regulatory associated protein of MTOR complex 1; SOD1: superoxide dismutase 1; SOD2: superoxide dismutase 2; SQSTM1/p62: sequestosome 1; VDAC1: voltage dependent anion channel 1; VDAC2: voltage dependent anion channel 2.
Collapse
Affiliation(s)
- Kartini Iskandar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jonathan Foo
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Integrative Science and Engineering Programme (ISEP), NUS Graduate School (NUSGS), National University of Singapore, Singapore
| | - Angeline Qiu Xia Liew
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Integrative Science and Engineering Programme (ISEP), NUS Graduate School (NUSGS), National University of Singapore, Singapore
| | - Haiyuxin Zhu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Deepika Raman
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Yan Yi Leong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Maria V. Babak
- Drug Discovery Laboratory, Department of Chemistry, City University of Hong Kong, Hong Kong, SAR, China
| | - Anna A. Kirsanova
- Drug Discovery Laboratory, Department of Chemistry, City University of Hong Kong, Hong Kong, SAR, China
| | - Anne-Sophie Armand
- Institut Necker Enfants Malades (INEM), INSERM U1151, Université Paris Cité, Paris, France
| | - Franck Oury
- Institut Necker Enfants Malades (INEM), INSERM U1151, Université Paris Cité, Paris, France
| | - Gregory Bellot
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Integrative Science and Engineering Programme (ISEP), NUS Graduate School (NUSGS), National University of Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- NUS Medicine Healthy Longevity Program, National University of Singapore, Singapore
- National University Cancer Institute, National University Health System, Singapore
| |
Collapse
|
4
|
Noè R, Inglese N, Romani P, Serafini T, Paoli C, Calciolari B, Fantuz M, Zamborlin A, Surdo NC, Spada V, Spacci M, Volta S, Ermini ML, Di Benedetto G, Frusca V, Santi C, Lefkimmiatis K, Dupont S, Voliani V, Sancineto L, Carrer A. Organic Selenium induces ferroptosis in pancreatic cancer cells. Redox Biol 2023; 68:102962. [PMID: 38029455 PMCID: PMC10698006 DOI: 10.1016/j.redox.2023.102962] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/10/2023] [Indexed: 12/01/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) cells reprogram both mitochondrial and lysosomal functions to support growth. At the same time, this causes significant dishomeostasis of free radicals. While this is compensated by the upregulation of detoxification mechanisms, it also represents a potential vulnerability. Here we demonstrate that PDA cells are sensitive to the inhibition of the mevalonate pathway (MVP), which supports the biosynthesis of critical antioxidant intermediates and protect from ferroptosis. We attacked the susceptibility of PDA cells to ferroptotic death with selenorganic compounds, including dibenzyl diselenide (DBDS) that exhibits potent pro-oxidant properties and inhibits tumor growth in vitro and in vivo. DBDS treatment induces the mobilization of iron from mitochondria enabling uncontrolled lipid peroxidation. Finally, we showed that DBDS and statins act synergistically to promote ferroptosis and provide evidence that combined treatment is a viable strategy to combat PDA.
Collapse
Affiliation(s)
- Roberta Noè
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy; Department of Biology, University of Padova, 35126, Padova, Italy
| | - Noemi Inglese
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy; Department of Biology, University of Padova, 35126, Padova, Italy
| | - Patrizia Romani
- Department of Molecular Medicine, University of Padova, 35126, Padova, Italy
| | - Thauan Serafini
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy
| | - Carlotta Paoli
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy; Department of Biology, University of Padova, 35126, Padova, Italy
| | - Beatrice Calciolari
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy; Department of Biology, University of Padova, 35126, Padova, Italy
| | - Marco Fantuz
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy; Department of Biology, University of Padova, 35126, Padova, Italy
| | - Agata Zamborlin
- NEST-Scuola Normale Superiore, 56127, Pisa, Italy; Center for Nanotechnology Innovation, Istituto Italiano di Tecnologia, 56127, Pisa, Italy
| | - Nicoletta C Surdo
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy; Neuroscience Institute, National Research Council (CNR), 35121, Padova, Italy
| | - Vittoria Spada
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy
| | - Martina Spacci
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy; Department of Biology, University of Padova, 35126, Padova, Italy
| | - Sara Volta
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy
| | - Maria Laura Ermini
- Center for Nanotechnology Innovation, Istituto Italiano di Tecnologia, 56127, Pisa, Italy
| | - Giulietta Di Benedetto
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy; Neuroscience Institute, National Research Council (CNR), 35121, Padova, Italy
| | - Valentina Frusca
- Center for Nanotechnology Innovation, Istituto Italiano di Tecnologia, 56127, Pisa, Italy; Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127, Pisa, Italy
| | - Claudio Santi
- Group of Catalysis and Green Organic Chemistry, Department of Pharmaceutical Sciences, University of Perugia, 06122, Perugia, PG, Italy
| | - Konstantinos Lefkimmiatis
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy; Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Sirio Dupont
- Department of Molecular Medicine, University of Padova, 35126, Padova, Italy
| | - Valerio Voliani
- Center for Nanotechnology Innovation, Istituto Italiano di Tecnologia, 56127, Pisa, Italy; Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genova, 16148, Genoa, Italy.
| | - Luca Sancineto
- Group of Catalysis and Green Organic Chemistry, Department of Pharmaceutical Sciences, University of Perugia, 06122, Perugia, PG, Italy.
| | - Alessandro Carrer
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy; Department of Biology, University of Padova, 35126, Padova, Italy.
| |
Collapse
|
5
|
Javali PS, Sekar M, Kumar A, Thirumurugan K. Dynamics of redox signaling in aging via autophagy, inflammation, and senescence. Biogerontology 2023; 24:663-678. [PMID: 37195483 DOI: 10.1007/s10522-023-10040-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 05/09/2023] [Indexed: 05/18/2023]
Abstract
Review paper attempts to explain the dynamic aspects of redox signaling in aging through autophagy, inflammation, and senescence. It begins with ROS source in the cell, then states redox signaling in autophagy, and regulation of autophagy in aging. Next, we discuss inflammation and redox signaling with various pathways involved: NOX pathway, ROS production via TNF-α, IL-1β, xanthine oxidase pathway, COX pathway, and myeloperoxidase pathway. Also, we emphasize oxidative damage as an aging marker and the contribution of pathophysiological factors to aging. In senescence-associated secretory phenotypes, we link ROS with senescence, aging disorders. Relevant crosstalk between autophagy, inflammation, and senescence using a balanced ROS level might reduce age-related disorders. Transducing the context-dependent signal communication among these three processes at high spatiotemporal resolution demands other tools like multi-omics aging biomarkers, artificial intelligence, machine learning, and deep learning. The bewildering advancement of technology in the above areas might progress age-related disorders diagnostics with precision and accuracy.
Collapse
Affiliation(s)
- Prashanth S Javali
- #412J, Structural Biology Lab, Pearl Research Park, School of Biosciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Mouliganesh Sekar
- #412J, Structural Biology Lab, Pearl Research Park, School of Biosciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Ashish Kumar
- #412J, Structural Biology Lab, Pearl Research Park, School of Biosciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Kavitha Thirumurugan
- #412J, Structural Biology Lab, Pearl Research Park, School of Biosciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
6
|
A Redoxable Mn Porphyrin, MnTnBuOE-2-PyP5+, Synergizes with Carboplatin in Treatment of Chemoresistant Ovarian Cell Line. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9664636. [PMID: 35898616 PMCID: PMC9313984 DOI: 10.1155/2022/9664636] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/29/2022] [Indexed: 12/20/2022]
Abstract
We have employed a redox-active MnP (MnTnBuOE-2-PyP5+, Mn(III) meso-tetrakis (N-n-butoxyethylpyridinium-2-yl) porphyrin) frequently identified as superoxide dismutase mimic or BMX-001, to explore the redox status of normal ovarian cell in relation to two ovarian cancer cell lines: OV90 human serous ovarian cancer cell and chemotherapy-resistant OV90 cell (OVCD). We identified that OVCD cells are under oxidative stress due to high hydrogen peroxide (H2O2) levels and low glutathione peroxidase and thioredoxin 1. Furthermore, OVCD cells have increased glycolysis activity and mitochondrial respiration when compared to immortalized ovarian cells (hTER7) and parental cancer cells (OV90). Our goal was to study how ovarian cell growth depends upon the redox state of the cell; hence, we used MnP (BMX-001), a redox-active MnSOD mimetic, as a molecular tool to alter ovarian cancer redox state. Interestingly, OVCD cells preferentially uptake MnP relative to OV90 cells which led to increased inhibition of cell growth, glycolytic activity, OXPHOS, and ATP, in OVCD cells. These effects were further increased when MnP was combined with carboplatin. The effects were discussed with regard to the elevation in H2O2 levels, increased oxidative stress, and reduced Nrf2 levels and its downstream targets when cells were exposed to either MnP or MnP/carboplatin. It is significant to emphasize that MnP protects normal ovarian cell line, hTER7, against carboplatin toxicity. Our data demonstrate that the addition of MnP-based redox-active drugs may be used (via increasing excessively the oxidative stress of serous ovarian cancer cells) to improve cancer patients' chemotherapy outcomes, which develop resistance to platinum-based drugs.
Collapse
|
7
|
Ferrara MG, Stefani A, Pilotto S, Carbone C, Vita E, Di Salvatore M, D'Argento E, Sparagna I, Monaca F, Valente G, Vitale A, Piro G, Belluomini L, Milella M, Tortora G, Bria E. The Renaissance of KRAS Targeting in Advanced Non-Small-Cell Lung Cancer: New Opportunities Following Old Failures. Front Oncol 2022; 11:792385. [PMID: 35004317 PMCID: PMC8733471 DOI: 10.3389/fonc.2021.792385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/06/2021] [Indexed: 12/14/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) represents the perfect paradigm of ‘precision medicine’ due to its complex intratumoral heterogeneity. It is truly characterized by a range of molecular alterations that can deeply influence the natural history of this disease. Several molecular alterations have been found over time, paving the road to biomarker-driven therapy and radically changing the prognosis of ‘oncogene addicted’ NSCLC patients. Kirsten rat sarcoma (KRAS) mutations are present in up to 30% of NSCLC (especially in adenocarcinoma histotype) and have been identified decades ago. Since its discovery, its molecular characteristics and its marked affinity to a specific substrate have led to define KRAS as an undruggable alteration. Despite that, many attempts have been made to develop drugs capable of targeting KRAS signaling but, until a few years ago, these efforts have been unsuccessful. Comprehensive genomic profiling and wide-spectrum analysis of genetic alterations have only recently allowed to identify different types of KRAS mutations. This tricky step has finally opened new frontiers in the treatment approach of KRAS-mutant patients and might hopefully increase their prognosis and quality of life. In this review, we aim to highlight the most interesting aspects of (epi)genetic KRAS features, hoping to light the way to the state of art of targeting KRAS in NSCLC.
Collapse
Affiliation(s)
- Miriam Grazia Ferrara
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.,Section of Oncology, Department of Translational Medicine, Università Cattolica Del Sacro Cuore, Roma, Italy
| | - Alessio Stefani
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.,Section of Oncology, Department of Translational Medicine, Università Cattolica Del Sacro Cuore, Roma, Italy
| | - Sara Pilotto
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Carmine Carbone
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Emanuele Vita
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.,Section of Oncology, Department of Translational Medicine, Università Cattolica Del Sacro Cuore, Roma, Italy
| | | | - Ettore D'Argento
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Ileana Sparagna
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.,Section of Oncology, Department of Translational Medicine, Università Cattolica Del Sacro Cuore, Roma, Italy
| | - Federico Monaca
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.,Section of Oncology, Department of Translational Medicine, Università Cattolica Del Sacro Cuore, Roma, Italy
| | - Giustina Valente
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.,Section of Oncology, Department of Translational Medicine, Università Cattolica Del Sacro Cuore, Roma, Italy
| | - Antonio Vitale
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.,Section of Oncology, Department of Translational Medicine, Università Cattolica Del Sacro Cuore, Roma, Italy
| | - Geny Piro
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Lorenzo Belluomini
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Michele Milella
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Giampaolo Tortora
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.,Section of Oncology, Department of Translational Medicine, Università Cattolica Del Sacro Cuore, Roma, Italy
| | - Emilio Bria
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.,Section of Oncology, Department of Translational Medicine, Università Cattolica Del Sacro Cuore, Roma, Italy
| |
Collapse
|
8
|
Raman D, Tay P, Hirpara JL, Liu D, Pervaiz S. TRAIL sensitivity of nasopharyngeal cancer cells involves redox dependent upregulation of TMTC2 and its interaction with membrane caspase-3. Redox Biol 2021; 48:102193. [PMID: 34839142 PMCID: PMC8636823 DOI: 10.1016/j.redox.2021.102193] [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: 08/26/2021] [Revised: 11/12/2021] [Accepted: 11/19/2021] [Indexed: 12/09/2022] Open
Abstract
AIMS Preferential expression of receptors for TNF-family related apoptosis inducing ligand (TRAIL), DR4 and DR5 makes TRAIL an attractive anti-cancer therapeutic. However, the efficacy of targeting death receptors has not been extensively studied in nasopharyngeal cancer (NPC). Here we investigated TRAIL sensitivity and its underlying mechanism in NPC cell lines, and assessed the potential of TRAIL as a therapeutic option against NPC. RESULTS Using two established NPC cell lines, we report the expression of DR4 and DR5, which respond to TRAIL ligation by triggering efficient Type II apoptosis. Mechanistically, early activation of caspase-3 and its membrane recruitment is identified in NPC cell lines, which is associated with, hitherto unreported, interaction with transmembrane and tetratricopeptide repeat containing 2 (TMTC2) in the lipid raft domains. TMTC2 expression is induced upon exposure to TRAIL and involves intracellular increase in peroxynitrite (ONOO-) production. While ONOO- increase is downstream of caspase-8 activation, it is involved in the upregulation of TMTC2, gene knockdown of which abrogated TRAIL-induced apoptotic execution. Bioinformatics analyses also provide evidence for a strong correlation between TMTC2 and DR4 or caspase-3 as well as a significantly better disease-free survival in patients with high TMTC2 expression. INNOVATION AND CONCLUSION Collectively, redox-dependent execution of NPC cells upon ligation of TRAIL receptors reintroduces the possible therapeutic use of TRAIL in NPC as well as underscores the potential of using TMTC2 as a biomarker of TRAIL sensitivity.
Collapse
Affiliation(s)
- Deepika Raman
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Patricia Tay
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Dan Liu
- Integrated Science and Engineering Program (ISEP), NUS Graduate School, National University of Singapore, Singapore
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cancer Science Institute, National University of Singapore, Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; National University Cancer Institute, National University Health System, Singapore; Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
9
|
Ngoi NY, Liew AQ, Chong SJF, Davids MS, Clement MV, Pervaiz S. The redox-senescence axis and its therapeutic targeting. Redox Biol 2021; 45:102032. [PMID: 34147844 PMCID: PMC8220395 DOI: 10.1016/j.redox.2021.102032] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 12/14/2022] Open
Abstract
Significance Cellular growth arrest, associated with ‘senescence’, helps to safeguard against the accumulation of DNA damage which is often recognized as the underlying mechanism of a wide variety of age-related pathologies including cancer. Cellular senescence has also been described as a ‘double-edged sword’. In cancer, for example, the creation of an immune-suppressive milieu by senescent tumor cells through the senescence-associated secretory phenotype contributes toward carcinogenesis and cancer progression. Recent advances The potential for cellular senescence to confer multi-faceted effects on tissue fate has led to a rejuvenated interest in its landscape and targeting. Interestingly, redox pathways have been described as both triggers and propagators of cellular senescence, leading to intricate cross-links between both pathways. Critical issues In this review, we describe the mechanisms driving cellular senescence, the interface with cellular redox metabolism as well as the role that chemotherapy-induced senescence plays in secondary carcinogenesis. Notably, the role that anti-apoptotic proteins of the Bcl-2 family play in inducing drug resistance via mechanisms that involve senescence induction. Future directions Though the therapeutic targeting of senescent cells as cancer therapy remains in its infancy, we summarize the current development of senotherapeutics, including recognized senotherapies, as well as the repurposing of drugs as senomorphic/senolytic candidates.
Collapse
Affiliation(s)
- Natalie Yl Ngoi
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Angeline Qx Liew
- Integrative Science and Engineering Programme (ISEP), NUS Graduate School (NUSGS), National University of Singapore, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Stephen J F Chong
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Matthew S Davids
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Marie-Veronique Clement
- Integrative Science and Engineering Programme (ISEP), NUS Graduate School (NUSGS), National University of Singapore, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Medicine Healthy Longevity Program, National University of Singapore, Singapore
| | - Shazib Pervaiz
- Integrative Science and Engineering Programme (ISEP), NUS Graduate School (NUSGS), National University of Singapore, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Medicine Healthy Longevity Program, National University of Singapore, Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; National University Cancer Institute, National University Health System, Singapore; Faculté de Medicine, University of Paris, Paris, France.
| |
Collapse
|
10
|
Lu J, Liu Z, Shu M, Zhang L, Xia W, Tang L, Li J, Huang B, Li H. Human placental mesenchymal stem cells ameliorate chemotherapy-induced damage in the testis by reducing apoptosis/oxidative stress and promoting autophagy. Stem Cell Res Ther 2021; 12:199. [PMID: 33743823 PMCID: PMC7981860 DOI: 10.1186/s13287-021-02275-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/09/2021] [Indexed: 01/18/2023] Open
Abstract
Background The side effects of busulfan on male reproduction are serious, so fertility preservation in children undergoing busulfan treatment is a major worldwide concern. Human placental mesenchymal stem cells (hPMSCs) have advantages such as stable proliferation and lower immunogenicity that make them an ideal material for stimulating tissue repair, especially restoring spermatogenesis. The protective effects of hPMSCs in busulfan-induced Sertoli cells and in busulfan-treated mouse testes have not been determined. Our study aimed to elaborate the protective effect and potential mechanisms of hPMSCs in busulfan-treated testes and Sertoli cells. Methods First, we developed a mouse model of busulfan-induced testicular toxicity in vivo and a mouse Sertoli cell line treated with busulfan in vitro to assess the protective effect and mechanisms of hPMSC treatment on spermatogenesis. Then, the length, width, and weight of the testes were monitored using Vernier calipers. Furthermore, at 1 week and 4 weeks after the transplantation of hPMSCs, histological sections of testes were stained with hematoxylin-eosin, and the seminiferous tubules with fluid-filled cavities were counted. Through ELISA analysis, testosterone levels and MDA, SOD, LDH, and CAT activities, which are associated with ROS, were detected. Markers of ROS, proliferation (Ki67), and apoptosis (Annexin V) were evaluated by FACS. Next, the fluorescence intensity of proliferation markers (BrdU and SCP3), an antioxidant marker (SIRT1), a spermatogenesis marker (PLZF), and autophagy-related genes (P62 and LC3AB) were detected by fluorescence microscopy. The mRNA expression of γ-H2AX, BRCA1, PARP1, PCNA, Ki67, P62, and LC3 was determined by qRT-PCR. Results hPMSCs restored disrupted spermatogenesis, promoted improved semen parameters, and increased testosterone levels, testis size, and autophagy in the testis toxicity mouse model induced by busulfan. hPMSCs suppressed the apoptosis of Sertoli cells and enhanced their rate of proliferation in vitro. Additionally, hPMSCs protected against oxidative stress and decreased oxidative damage in the testis toxicity mouse model induced by busulfan. Furthermore, hPMSCs increased the expression of proliferation genes (PCNA and KI67) and decreased the mRNA levels of apoptotic genes such as γ-H2AX, BRCA1, and PARP1. Conclusions This research showed that hPMSC injection ameliorated busulfan-induced damage in the testis by reducing apoptosis/oxidative stress and promoting autophagy. The present study offers an idea for a new method for clinical treatment of chemotherapy-induced spermatogenesis. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02275-z.
Collapse
Affiliation(s)
- Jiafeng Lu
- Center of Reproduction and Genetics, The affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Suzhou, 215002, China
| | - Zhenxing Liu
- Center of Reproduction and Genetics, The affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Suzhou, 215002, China
| | - Mingkai Shu
- Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, 215123, China
| | - Liya Zhang
- Center of Reproduction and Genetics, The affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Suzhou, 215002, China
| | - Wenjuan Xia
- Center of Reproduction and Genetics, The affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Suzhou, 215002, China
| | - Liuna Tang
- Center of Reproduction and Genetics, The affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Suzhou, 215002, China
| | - Jincheng Li
- Center of Reproduction and Genetics, The affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Suzhou, 215002, China
| | - Boxian Huang
- Center of Reproduction and Genetics, The affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Suzhou, 215002, China.
| | - Hong Li
- Center of Reproduction and Genetics, The affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Suzhou, 215002, China.
| |
Collapse
|
11
|
Liu D, Qiu X, Xiong X, Chen X, Pan F. Current updates on the role of reactive oxygen species in bladder cancer pathogenesis and therapeutics. Clin Transl Oncol 2020; 22:1687-1697. [PMID: 32189139 PMCID: PMC7423792 DOI: 10.1007/s12094-020-02330-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/25/2020] [Indexed: 12/15/2022]
Abstract
Bladder cancer (BCa) is the fourth most common urological malignancy in the world, it has become the costliest cancer to manage due to its high rate of recurrence and lack of effective treatment modalities. As a natural byproduct of cellular metabolism, reactive oxygen species (ROS) have an important role in cell signaling and homeostasis. Although up-regulation of ROS is known to induce tumorigenesis, growing evidence suggests a number of agents that can selectively kill cancer cells through ROS induction. In particular, accumulation of ROS results in oxidative stress-induced apoptosis in cancer cells. So, ROS is a double-edged sword. A modest level of ROS is required for cancer cells to survive, whereas excessive levels kill them. This review summarizes the up-to-date findings of oxidative stress-regulated signaling pathways and transcription factors involved in the etiology and progression of BCa and explores the possible therapeutic implications of ROS regulators as therapeutic agents for BCa.
Collapse
Affiliation(s)
- D Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - X Qiu
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - X Xiong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - X Chen
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Institute of Brain Research, Key Laboratory of Neurological Diseases, Ministry of Education, Hubei Provincial Key Laboratory of Neurological Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - F Pan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
12
|
Ciccarese F, Raimondi V, Sharova E, Silic-Benussi M, Ciminale V. Nanoparticles as Tools to Target Redox Homeostasis in Cancer Cells. Antioxidants (Basel) 2020; 9:antiox9030211. [PMID: 32143322 PMCID: PMC7139659 DOI: 10.3390/antiox9030211] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/20/2020] [Accepted: 02/29/2020] [Indexed: 12/18/2022] Open
Abstract
Reactive oxygen species (ROS) constitute a homeostatic rheostat that modulates signal transduction pathways controlling cell turnover. Most oncogenic pathways activated in cancer cells drive a sustained increase in ROS production, and cancer cells are strongly addicted to the increased activity of scavenging pathways to maintain ROS below levels that produce macromolecular damage and engage cell death pathways. Consistent with this notion, tumor cells are more vulnerable than their normal counterparts to pharmacological treatments that increase ROS production and inhibit ROS scavenging. In the present review, we discuss the recent advances in the development of integrated anticancer therapies based on nanoparticles engineered to kill cancer cells by raising their ROS setpoint. We also examine nanoparticles engineered to exploit the metabolic and redox alterations of cancer cells to promote site-specific drug delivery to cancer cells, thus maximizing anticancer efficacy while minimizing undesired side effects on normal tissues.
Collapse
Affiliation(s)
- Francesco Ciccarese
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (F.C.); (V.R.); (E.S.); (M.S.-B.)
| | - Vittoria Raimondi
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (F.C.); (V.R.); (E.S.); (M.S.-B.)
| | - Evgeniya Sharova
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (F.C.); (V.R.); (E.S.); (M.S.-B.)
| | - Micol Silic-Benussi
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (F.C.); (V.R.); (E.S.); (M.S.-B.)
| | - Vincenzo Ciminale
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (F.C.); (V.R.); (E.S.); (M.S.-B.)
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
- Correspondence:
| |
Collapse
|
13
|
Ngoi NYL, Eu JQ, Hirpara J, Wang L, Lim JSJ, Lee SC, Lim YC, Pervaiz S, Goh BC, Wong ALA. Targeting Cell Metabolism as Cancer Therapy. Antioxid Redox Signal 2020; 32:285-308. [PMID: 31841375 DOI: 10.1089/ars.2019.7947] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significance: Cancer cells exhibit altered metabolic pathways to keep up with biosynthetic and reduction-oxidation needs during tumor proliferation and metastasis. The common induction of metabolic pathways during cancer progression, regardless of cancer histio- or genotype, makes cancer metabolism an attractive target for therapeutic exploitation. Recent Advances: Emerging data suggest that these altered pathways may even result in resistance to anticancer therapies. Identifying specific metabolic dependencies that are unique to cancer cells has proved challenging in this field, limiting the therapeutic window for many candidate drug approaches. Critical Issues: Cancer cells display significant metabolic flexibility in nutrient-limited environments, hampering the longevity of suppressing cancer metabolism through any singular approach. Combinatorial "synthetic lethal" approaches may have a better chance for success and promising strategies are reviewed here. The dynamism of the immune system adds a level of complexity, as various immune populations in the tumor microenvironment often share metabolic pathways with cancer, with successive alterations during immune activation and quiescence. Decoding the reprogramming of metabolic pathways within cancer cells and stem cells, as well as examining metabolic symbiosis between components of the tumor microenvironment, would be essential to further meaningful drug development within the tumor's metabolic ecosystem. Future Directions: In this article, we examine evidence for the therapeutic potential of targeting metabolic alterations in cancer, and we discuss the drawbacks and successes that have stimulated this field.
Collapse
Affiliation(s)
- Natalie Y L Ngoi
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Jie Qing Eu
- Department of Physiology and Medical Science Cluster Cancer Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Cancer Science Institute, Singapore, National University of Singapore, Singapore
| | - Jayshree Hirpara
- Department of Physiology and Medical Science Cluster Cancer Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Cancer Science Institute, Singapore, National University of Singapore, Singapore
| | - Lingzhi Wang
- Cancer Science Institute, Singapore, National University of Singapore, Singapore
| | - Joline S J Lim
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Soo-Chin Lee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute, Singapore, National University of Singapore, Singapore
| | - Yaw-Chyn Lim
- Department of Physiology and Medical Science Cluster Cancer Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shazib Pervaiz
- Department of Physiology and Medical Science Cluster Cancer Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore.,National University Cancer Institute, National University Health System, Singapore
| | - Boon Cher Goh
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute, Singapore, National University of Singapore, Singapore
| | - Andrea L A Wong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute, Singapore, National University of Singapore, Singapore
| |
Collapse
|
14
|
Heslop KA, Rovini A, Hunt EG, Fang D, Morris ME, Christie CF, Gooz MB, DeHart DN, Dang Y, Lemasters JJ, Maldonado EN. JNK activation and translocation to mitochondria mediates mitochondrial dysfunction and cell death induced by VDAC opening and sorafenib in hepatocarcinoma cells. Biochem Pharmacol 2020; 171:113728. [PMID: 31759978 PMCID: PMC7309270 DOI: 10.1016/j.bcp.2019.113728] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/20/2019] [Indexed: 02/07/2023]
Abstract
The multikinase inhibitor sorafenib, and opening of voltage dependent anion channels (VDAC) by the erastin-like compound X1 promotes oxidative stress and mitochondrial dysfunction in hepatocarcinoma cells. Here, we hypothesized that X1 and sorafenib induce mitochondrial dysfunction by increasing reactive oxygen species (ROS) formation and activating c-Jun N-terminal kinases (JNKs), leading to translocation of activated JNK to mitochondria. Both X1 and sorafenib increased production of ROS and activated JNK. X1 and sorafenib caused a drop in mitochondrial membrane potential (ΔΨ), a readout of mitochondrial metabolism, after 60 min. Mitochondrial depolarization after X1 and sorafenib occurred in parallel with JNK activation, increased superoxide (O2•-) production, decreased basal and oligomycin sensitive respiration, and decreased maximal respiratory capacity. Increased production of O2•- after X1 or sorafenib was abrogated by JNK inhibition and antioxidants. S3QEL 2, a specific inhibitor of site IIIQo, at Complex III, prevented depolarization induced by X1. JNK inhibition by JNK inhibitors VIII and SP600125 also prevented mitochondrial depolarization. After X1, activated JNK translocated to mitochondria as assessed by proximity ligation assays. Tat-Sab KIM1, a peptide selectively preventing the binding of JNK to the outer mitochondrial membrane protein Sab, blocked the depolarization induced by X1 and sorafenib. X1 promoted cell death mostly by necroptosis that was partially prevented by JNK inhibition. These results indicate that JNK activation and translocation to mitochondria is a common mechanism of mitochondrial dysfunction induced by both VDAC opening and sorafenib.
Collapse
Affiliation(s)
- K A Heslop
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - A Rovini
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - E G Hunt
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - D Fang
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - M E Morris
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - C F Christie
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - M B Gooz
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - D N DeHart
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Y Dang
- Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - J J Lemasters
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States; Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, United States; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - E N Maldonado
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
15
|
Pervaiz S, Bellot GL, Lemoine A, Brenner C. Redox signaling in the pathogenesis of human disease and the regulatory role of autophagy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 352:189-214. [DOI: 10.1016/bs.ircmb.2020.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
16
|
Raimondi V, Ciccarese F, Ciminale V. Oncogenic pathways and the electron transport chain: a dangeROS liaison. Br J Cancer 2019; 122:168-181. [PMID: 31819197 PMCID: PMC7052168 DOI: 10.1038/s41416-019-0651-y] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 10/30/2019] [Accepted: 11/05/2019] [Indexed: 02/07/2023] Open
Abstract
Driver mutations in oncogenic pathways, rewiring of cellular metabolism and altered ROS homoeostasis are intimately connected hallmarks of cancer. Electrons derived from different metabolic processes are channelled into the mitochondrial electron transport chain (ETC) to fuel the oxidative phosphorylation process. Electrons leaking from the ETC can prematurely react with oxygen, resulting in the generation of reactive oxygen species (ROS). Several signalling pathways are affected by ROS, which act as second messengers controlling cell proliferation and survival. On the other hand, oncogenic pathways hijack the ETC, enhancing its ROS-producing capacity by increasing electron flow or by impinging on the structure and organisation of the ETC. In this review, we focus on the ETC as a source of ROS and its modulation by oncogenic pathways, which generates a vicious cycle that resets ROS levels to a higher homoeostatic set point, sustaining the cancer cell phenotype.
Collapse
Affiliation(s)
| | | | - Vincenzo Ciminale
- Veneto Institute of Oncology IOV - IRCCS, Padua, Italy. .,Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy.
| |
Collapse
|
17
|
Chen F, Alphonse MP, Liu Y, Liu Q. Targeting Mutant KRAS for Anticancer Therapy. Curr Top Med Chem 2019; 19:2098-2113. [DOI: 10.2174/1568026619666190902151307] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/28/2019] [Accepted: 07/01/2019] [Indexed: 12/13/2022]
Abstract
:Over the past decades, designing therapeutic strategies to target KRAS-mutant cancers, which is one of the most frequent mutant oncogenes among all cancer types, have proven unsuccessful regardless of many concerted attempts. There are key challenges for KRAS-mutant anticancer therapy, as the complex cellular processes involved in KRAS signaling has present. Herein, we highlight the emerging therapeutic approaches for inhibiting KRAS signaling and blocking KRAS functions, in hope to serve as a more effective guideline for future development of therapeutics.
Collapse
Affiliation(s)
- Fengqian Chen
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH), Texas Tech University, Lubbock, TX 79416, United States
| | - Martin P. Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Cancer Research Building II, Suite 216, 1550 Orleans Street, Baltimore, MD 21231, United States
| | - Yan Liu
- Western University of Health Sciences, 309 E. Second Street, Pomona, CA 91766, United States
| | - Qi Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Cancer Research Building II, Suite 216, 1550 Orleans Street, Baltimore, MD 21231, United States
| |
Collapse
|
18
|
Hong BJ, Park WY, Kim HR, Moon JW, Lee HY, Park JH, Kim SK, Oh Y, Roe JS, Kim MY. Oncogenic KRAS Sensitizes Lung Adenocarcinoma to GSK-J4-Induced Metabolic and Oxidative Stress. Cancer Res 2019; 79:5849-5859. [PMID: 31506334 DOI: 10.1158/0008-5472.can-18-3511] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 07/29/2019] [Accepted: 09/06/2019] [Indexed: 11/16/2022]
Abstract
Genetic and epigenetic changes (e.g., histone methylation) contribute to cancer development and progression, but our understanding of whether and how specific mutations affect a cancer's sensitivity to histone demethylase (KDM) inhibitors is limited. Here, we evaluated the effects of a panel of KDM inhibitors on lung adenocarcinomas (LuAC) with various mutations. Notably, LuAC lines harboring KRAS mutations showed hypersensitivity to the histone H3K27 demethylase inhibitor GSK-J4. Specifically, GSK-J4 treatment of KRAS mutant-containing LuAC downregulated cell-cycle progression genes with increased H3K27me3. In addition, GSK-J4 upregulated expression of genes involved in glutamine/glutamate transport and metabolism. In line with this, GSK-J4 reduced cellular levels of glutamate, a key source of the TCA cycle intermediate α-ketoglutarate (αKG) and of the antioxidant glutathione, leading to reduced cell viability. Supplementation with an αKG analogue or glutathione protected KRAS-mutant LuAC cells from GSK-J4-mediated reductions in viability, suggesting GSK-J4 exerts its anticancer effects by inducing metabolic and oxidative stress. Importantly, KRAS knockdown in mutant LuAC lines prevented GSK-J4-induced decrease in glutamate levels and reduced their susceptibility to GSK-J4, whereas overexpression of oncogenic KRAS in wild-type LuAC lines sensitized them to GSK-J4. Collectively, our study uncovers a novel association between a genetic mutation and KDM inhibitor sensitivity and identifies the underlying mechanisms. This suggests GSK-J4 as a potential treatment option for cancer patients with KRAS mutations. SIGNIFICANCE: This study not only provides a novel association between KRAS mutation and GSK-J4 sensitivity but also demonstrates the underlying mechanisms, suggesting a potential use of GSK-J4 in cancer patients with KRAS mutations.
Collapse
Affiliation(s)
- Beom-Jin Hong
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Woo-Yong Park
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Hwa-Ryeon Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Jin Woo Moon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | | | - Jun Hyung Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Seon-Kyu Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, South Korea
| | - Youngbin Oh
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Jae-Seok Roe
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea.
| | - Mi-Young Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea. .,KAIST Institute for the BioCentury, Cancer Metastasis Control Center, Daejeon, South Korea
| |
Collapse
|
19
|
Foo CHJ, Pervaiz S. gRASping the redox lever to modulate cancer cell fate signaling. Redox Biol 2019; 25:101094. [PMID: 30638892 PMCID: PMC6859584 DOI: 10.1016/j.redox.2018.101094] [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: 11/29/2018] [Revised: 12/22/2018] [Accepted: 12/26/2018] [Indexed: 01/17/2023] Open
Abstract
RAS proteins are critical regulators of signaling networks controlling diverse cellular functions such as cell proliferation and survival and its mutation are among the most powerful oncogenic drivers in human cancers. Despite intense efforts, direct RAS-targeting strategies remain elusive due to its "undruggable" nature. To that end, bulk of the research efforts has been directed towards targeting upstream and/or downstream of RAS signaling. However, the therapeutic efficacies of these treatments are limited in the long run due to the acquired drug resistance in RAS-driven cancers. Interestingly, recent studies have uncovered a potential role of RAS in redox-regulation as well as the interplay between ROS and RAS-associated signaling networks during process of cancer initiation and progression. More specifically, these studies provide ample evidence to implicate RAS as a redox-rheostat, manipulating ROS levels to provide a redox-milieu conducive for carcinogenesis. Importantly, the understanding of RAS-ROS interplay could provide us with novel targetable vulnerabilities for designing therapeutic strategies. In this review, we provide a brief summary of the advances in the field to illustrate the dual role of RAS in redox-regulation and its implications in RAS signaling outcomes and also emerging redox-based strategies to target RAS-driven cancers.
Collapse
Affiliation(s)
- Chuan Han Jonathan Foo
- Department of Physiology, YLL School of Medicine, National University of Singapore (NUS), Singapore; NUS Graduate School of Integrative Sciences and Engineering, NUS, Singapore
| | - Shazib Pervaiz
- Department of Physiology, YLL School of Medicine, National University of Singapore (NUS), Singapore; Medical Science Cluster Cancer Program, YLL School of Medicine, National University of Singapore (NUS), Singapore; NUS Graduate School of Integrative Sciences and Engineering, NUS, Singapore; National University Cancer Institute, NUHS, Singapore.
| |
Collapse
|
20
|
Abstract
In this Review, we focus on catalytic antioxidant study based on transition metal complexes, organoselenium compounds, supramolecules and protein scaffolds.
Collapse
Affiliation(s)
- Riku Kubota
- Department of Applied Chemistry for Environment
- Tokyo Metropolitan University
- Hachioji
- Japan
| | - Shoichiro Asayama
- Department of Applied Chemistry for Environment
- Tokyo Metropolitan University
- Hachioji
- Japan
| | - Hiroyoshi Kawakami
- Department of Applied Chemistry for Environment
- Tokyo Metropolitan University
- Hachioji
- Japan
| |
Collapse
|
21
|
Zhou L, Yi Y, Yuan Q, Zhang J, Li Y, Wang P, Xu M, Xie S. VAOS, a novel vanadyl complexes of alginate saccharides, inducing apoptosis via activation of AKT-dependent ROS production in NSCLC. Free Radic Biol Med 2018; 129:177-185. [PMID: 30223019 DOI: 10.1016/j.freeradbiomed.2018.09.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/05/2018] [Accepted: 09/14/2018] [Indexed: 02/04/2023]
Abstract
Previous studies have confirmed that protein tyrosine phosphatase 1B (PTP1B) can promote tumour progression in non-small cell lung cancer (NSCLC). Vanadyl alginate oligosaccharides (VAOS) is a new coordination compounds that possesses a good PTP1B inhibitory activity. However, the potent anticancer efficacy of VAOS in human NSCLC requires further study. In this study, VAOS exhibited effective inhibitory effects in NSCLC both in cultured cells and in a xenograft mouse model. VAOS was further identified to induce NSCLC cell apoptosis through activating protein kinase B (AKT) to elevate intracellular reactive oxygen species (ROS) levels by increasing in oxygen consumption and impairing the ROS-scavenging system. Neither silencing of PTP1B by siRNA nor transient overexpression of PTP1B had an effect on the AKT phosphorylation triggered by VAOS, indicating that PTP1B inhibition was not involved in VAOS-induced apoptosis. Through phosphorus colorimetric assay, we demonstrated that VAOS notably inhibited phosphatase and tensin homologue deleted on chromosome 10 (PTEN) dephosphorylation activity, another member of the protein tyrosine phosphatases (PTPases)-upstream factor of AKT. Interestingly, PTEN knockdown sensitized cells to VAOS, whereas ectopic expression of PTEN markedly rescued VAOS-mediated lethality. In vivo, VAOS treatment markedly reduced PTEN activity and tumour cell burden with low systemic toxicity. Thus, our data not only provided a new therapeutic drug candidate for NSCLC, but presented new understanding into the pharmacological research of VAOS.
Collapse
MESH Headings
- A549 Cells
- Alginates/chemical synthesis
- Alginates/pharmacology
- Animals
- Antineoplastic Agents/chemical synthesis
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Apoptosis/genetics
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Cell Survival/drug effects
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- PTEN Phosphohydrolase/antagonists & inhibitors
- PTEN Phosphohydrolase/genetics
- PTEN Phosphohydrolase/metabolism
- Phosphorylation
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Reactive Oxygen Species/agonists
- Reactive Oxygen Species/metabolism
- Signal Transduction
- Tumor Burden/drug effects
- Vanadates/chemical synthesis
- Vanadates/pharmacology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Ling Zhou
- Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong 264003, PR China; The Key Laboratory of Traditional Chinese Medicine Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine, School of Pharmacy, Binzhou Medical University, YanTai, ShanDong 264003, PR China
| | - Yuetao Yi
- Yantai Institute of Coastal Zone Research Chinese Academy of Sciences, Yantai 264003, PR China
| | - Qing Yuan
- Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong 264003, PR China
| | - Jing Zhang
- Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong 264003, PR China
| | - Youjie Li
- Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong 264003, PR China
| | - Pingyu Wang
- Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong 264003, PR China
| | - Maolei Xu
- The Key Laboratory of Traditional Chinese Medicine Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine, School of Pharmacy, Binzhou Medical University, YanTai, ShanDong 264003, PR China.
| | - Shuyang Xie
- Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong 264003, PR China.
| |
Collapse
|
22
|
Batinic-Haberle I, Tovmasyan A, Spasojevic I. Mn Porphyrin-Based Redox-Active Drugs: Differential Effects as Cancer Therapeutics and Protectors of Normal Tissue Against Oxidative Injury. Antioxid Redox Signal 2018; 29:1691-1724. [PMID: 29926755 PMCID: PMC6207162 DOI: 10.1089/ars.2017.7453] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
SIGNIFICANCE After approximatelty three decades of research, two Mn(III) porphyrins (MnPs), MnTE-2-PyP5+ (BMX-010, AEOL10113) and MnTnBuOE-2-PyP5+ (BMX-001), have progressed to five clinical trials. In parallel, another similarly potent metal-based superoxide dismutase (SOD) mimic-Mn(II)pentaaza macrocycle, GC4419-has been tested in clinical trial on application, identical to that of MnTnBuOE-2-PyP5+-radioprotection of normal tissue in head and neck cancer patients. This clearly indicates that Mn complexes that target cellular redox environment have reached sufficient maturity for clinical applications. Recent Advances: While originally developed as SOD mimics, MnPs undergo intricate interactions with numerous redox-sensitive pathways, such as those involving nuclear factor κB (NF-κB) and nuclear factor E2-related factor 2 (Nrf2), thereby impacting cellular transcriptional activity. An increasing amount of data support the notion that MnP/H2O2/glutathione (GSH)-driven catalysis of S-glutathionylation of protein cysteine, associated with modification of protein function, is a major action of MnPs on molecular level. CRITICAL ISSUES Differential effects of MnPs on normal versus tumor cells/tissues, which support their translation into clinic, arise from differences in their accumulation and redox environment of such tissues. This in turn results in different yields of MnP-driven modifications of proteins. Thus far, direct evidence for such modification of NF-κB, mitogen-activated protein kinases (MAPK), phosphatases, Nrf2, and endogenous antioxidative defenses was provided in tumor, while indirect evidence shows the modification of NF-κB and Nrf2 translational activities by MnPs in normal tissue. FUTURE DIRECTIONS Studies that simultaneously explore differential effects in same animal are lacking, while they are essential for understanding of extremely intricate interactions of metal-based drugs with complex cellular networks of normal and cancer cells/tissues.
Collapse
Affiliation(s)
- Ines Batinic-Haberle
- 1 Department of Radiation Oncology, Duke University School of Medicine , Durham, North Carolina
| | - Artak Tovmasyan
- 1 Department of Radiation Oncology, Duke University School of Medicine , Durham, North Carolina
| | - Ivan Spasojevic
- 2 Department of Medicine, Duke University School of Medicine , Durham, North Carolina.,3 PK/PD Core Laboratory, Pharmaceutical Research Shared Resource, Duke Cancer Institute , Durham, North Carolina
| |
Collapse
|
23
|
Chong SJF, Lai JXH, Eu JQ, Bellot GL, Pervaiz S. Reactive Oxygen Species and Oncoprotein Signaling-A Dangerous Liaison. Antioxid Redox Signal 2018; 29:1553-1588. [PMID: 29186971 DOI: 10.1089/ars.2017.7441] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
SIGNIFICANCE There is evidence to implicate reactive oxygen species (ROS) in tumorigenesis and its progression. This has been associated with the interplay between ROS and oncoproteins, resulting in enhanced cellular proliferation and survival. Recent Advances: To date, studies have investigated specific contributions of the crosstalk between ROS and signaling networks in cancer initiation and progression. These investigations have challenged the established dogma of ROS as agents of cell death by demonstrating a secondary function that fuels cell proliferation and survival. Studies have thus identified (onco)proteins (Bcl-2, STAT3/5, RAS, Rac1, and Myc) in manipulating ROS level as well as exploiting an altered redox environment to create a milieu conducive for cancer formation and progression. CRITICAL ISSUES Despite these advances, drug resistance and its association with an altered redox metabolism continue to pose a challenge at the mechanistic and clinical levels. Therefore, identifying specific signatures, altered protein expressions, and modifications as well as protein-protein interplay/function could not only enhance our understanding of the redox networks during cancer initiation and progression but will also provide novel targets for designing specific therapeutic strategies. FUTURE DIRECTIONS Not only a heightened realization is required to unravel various gene/protein networks associated with cancer formation and progression, particularly from the redox standpoint, but there is also a need for developing more sensitive tools for assessing cancer redox metabolism in clinical settings. This review attempts to summarize our current knowledge of the crosstalk between oncoproteins and ROS in promoting cancer cell survival and proliferation and treatment strategies employed against these oncoproteins. Antioxid. Redox Signal.
Collapse
Affiliation(s)
- Stephen Jun Fei Chong
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Jolin Xiao Hui Lai
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Jie Qing Eu
- 2 Cancer Science Institute , Singapore, Singapore
| | - Gregory Lucien Bellot
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,3 Department of Hand and Reconstructive Microsurgery, National University Health System , Singapore, Singapore
| | - Shazib Pervaiz
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,4 NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , Singapore, Singapore .,5 National University Cancer Institute, National University Health System , Singapore, Singapore .,6 School of Biomedical Sciences, Curtin University , Perth, Australia
| |
Collapse
|
24
|
Chaiswing L, St. Clair WH, St. Clair DK. Redox Paradox: A Novel Approach to Therapeutics-Resistant Cancer. Antioxid Redox Signal 2018; 29:1237-1272. [PMID: 29325444 PMCID: PMC6157438 DOI: 10.1089/ars.2017.7485] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 01/05/2018] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Cancer cells that are resistant to radiation and chemotherapy are a major problem limiting the success of cancer therapy. Aggressive cancer cells depend on elevated intracellular levels of reactive oxygen species (ROS) to proliferate, self-renew, and metastasize. As a result, these aggressive cancers maintain high basal levels of ROS compared with normal cells. The prominence of the redox state in cancer cells led us to consider whether increasing the redox state to the condition of oxidative stress could be used as a successful adjuvant therapy for aggressive cancers. Recent Advances: Past attempts using antioxidant compounds to inhibit ROS levels in cancers as redox-based therapy have met with very limited success. However, recent clinical trials using pro-oxidant compounds reveal noteworthy results, which could have a significant impact on the development of strategies for redox-based therapies. CRITICAL ISSUES The major objective of this review is to discuss the role of the redox state in aggressive cancers and how to utilize the shift in redox state to improve cancer therapy. We also discuss the paradox of redox state parameters; that is, hydrogen peroxide (H2O2) as the driver molecule for cancer progression as well as a target for cancer treatment. FUTURE DIRECTIONS Based on the biological significance of the redox state, we postulate that this system could potentially be used to create a new avenue for targeted therapy, including the potential to incorporate personalized redox therapy for cancer treatment.
Collapse
Affiliation(s)
- Luksana Chaiswing
- Department of Toxicology and Cancer Biology, University of Kentucky-Lexington, Lexington, Kentucky
| | - William H. St. Clair
- Department of Radiation Medicine, University of Kentucky-Lexington, Lexington, Kentucky
| | - Daret K. St. Clair
- Department of Toxicology and Cancer Biology, University of Kentucky-Lexington, Lexington, Kentucky
| |
Collapse
|
25
|
Abstract
Cellular redox state is a consequence of the balance between the rates of reactive oxygen species and/or reactive nitrogen species, and their dissipation via enzymatic and nonenzymatic redox buffering systems. While low levels of oscillation are associated with normal cellular metabolism, stimuli that favor a significant shift in the redox microenvironment, through either the increased production and/or compromise of the antioxidant defenses, induce overt oxidative stress. This change in the redox set point triggers a host of cellular responses ranging from modifications in cellular macromolecules, organelle morphology and physiology, amplified cell-to-cell and intracellular signaling, and changes in genome, epigenome, and proteome. The consequence of this dysregulated cellular homeostasis is therefore manifested in the form of a plethora of pathological states such as inflammation, diabetes mellitus, neurodegenerative disorders, atherosclerosis, and cancer. On the backdrop of these observations, this Forum attempts at reviewing the current understanding of how a prooxidant intracellular milieu favors cell survival while overt oxidative stress results in death execution, and the translation of these biological effects in human disease states, in particular cancer. The far-reaching biochemical, biological, and clinical ramifications of an altered redox environment are also discussed from the standpoint of strategic therapeutic design against refractory and aggressive cancers. It is tempting to conjecture if the inherent or acquired redox heterogeneity, at least in the case of cancer, has evolved as an "evasive mechanism," or presents itself as the "Achilles heel" for therapeutic exploitation.
Collapse
Affiliation(s)
- Shazib Pervaiz
- 1 Cancer Biology Program, Medical Science Cluster, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,2 NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , Singapore, Singapore .,3 National University Cancer Institute, National University Health System , Singapore, Singapore .,4 Curtin Health Innovation Research Institute, School of Pharmacy and Biomedical Sciences, Curtin University, Perth, Australia
| |
Collapse
|
26
|
Ediriweera MK, Tennekoon KH, Samarakoon SR. In vitro assays and techniques utilized in anticancer drug discovery. J Appl Toxicol 2018; 39:38-71. [DOI: 10.1002/jat.3658] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 06/01/2018] [Accepted: 06/04/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Meran Keshawa Ediriweera
- Institute of Biochemistry, Molecular Biology and Biotechnology; University of Colombo; Colombo 03 Sri Lanka
| | - Kamani Hemamala Tennekoon
- Institute of Biochemistry, Molecular Biology and Biotechnology; University of Colombo; Colombo 03 Sri Lanka
| | | |
Collapse
|
27
|
Hirpara JL, Loh T, Ng SB, Chng WJ, Pervaiz S. Aberrant localization of apoptosis protease activating factor-1 in lipid raft sub-domains of diffuse large B cell lymphomas. Oncotarget 2018; 7:83964-83975. [PMID: 27863378 PMCID: PMC5356638 DOI: 10.18632/oncotarget.13336] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 11/08/2016] [Indexed: 11/25/2022] Open
Abstract
Resistance to chemotherapy remains a challenge in the clinical management of diffuse B cell lymphomas despite aggressive chemotherapy such as CHOP and monoclonal CD20. Here we provide evidence that the apoptosome adaptor protein, Apaf-1, is mislocalized in primary cells derived from patients with diffuse large B cell lymphomas (DLBCL). Whereas, the total expression of Apaf-1 did not change, its sub-cellular localization was significantly different in DLBCL, compared to T cell lymphomas as well as cells derived from reactive lymphadenopathy biopsies. As expected, Apaf-1 was detected in the cytosolic fractions of non-B cell lymphomas and non-cancerous tissues; however, in B cell derived lymphomas the protein was detected in membrane raft sub-domains rather than the cytosol. Disruption of lipid raft structures resulted in the redistribution of Apaf-1 to the cytosol and restored apoptosis sensitivity of DLBCL. Furthermore, we identified novel small molecule compounds that target DLBCL by promoting Apaf-1 release form lipid rafts via mechanisms that involve an increase in intracellular reactive oxygen species production. Taken together, our results implicate Apaf-1 mislocalization as a potential diagnostic and prognostic marker for DLBCL, and provide a novel therapeutic strategy for circumventing the drug refractory nature of this sub-class of B cell lymphoma.
Collapse
Affiliation(s)
- Jayshree L Hirpara
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Experimental Therapeutics Program, Cancer Science Institute, National University Healthcare System, Singapore
| | - Thomas Loh
- Department of Otolaryngology, National University Healthcare System, Singapore
| | - Siok Bian Ng
- Department of Pathology, National University Healthcare System, Singapore
| | - Wee Joo Chng
- Cancer Science Institute, National University Healthcare System, Singapore
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore.,National University Cancer Institute, National University Healthcare System,.,School of Biomedical Sciences, Curtin University, Perth, Australia
| |
Collapse
|
28
|
Li B, He X, Zhuang M, Niu B, Wu C, Mu H, Tang F, Cui Y, Liu W, Zhao B, Peng S, Li G, Hua J. Melatonin Ameliorates Busulfan-Induced Spermatogonial Stem Cell Oxidative Apoptosis in Mouse Testes. Antioxid Redox Signal 2018; 28:385-400. [PMID: 28027652 DOI: 10.1089/ars.2016.6792] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
AIMS Many men endure immunosuppressive or anticancer treatments that contain alkylating agents before the age of sexual maturity, especially the increasing number of preadolescent males who undergo busulfan treatment for myeloablative conditioning before hematopoietic stem cell transplantation. Before sperm production, there are no sperm available for cryopreservation. Thus, it is necessary to identify a solution to ameliorate the busulfan-induced damage of spermatogonial stem cells (SSCs). RESULTS In this study, we demonstrated that melatonin relieved the previously described SSC loss and apoptosis in mouse testes. Melatonin increased the expression of manganese superoxide dismutase (MnSOD), which regulated the production of busulfan-induced reactive oxygen species (ROS). Moreover, melatonin promoted sirtuin type 1 (SIRT1) expression. SIRT1 participated in the deacetylation of p53, which promotes p53 ubiquitin degradation. Decreased concentrations of deacetylated p53 resulted in spermatogonial cell resistance to apoptosis. Acute T cell leukemia cell assay demonstrated that melatonin does not affect busulfan-induced cancer cell apoptosis and ROS. INNOVATION The current evidence suggests that melatonin may alleviate the side effects of alkylating drugs, such as busulfan. CONCLUSION Melatonin promoted MnSOD and SIRT1 expression, which successfully ameliorated busulfan-induced SSC apoptosis caused by high concentrations of ROS and p53. Antioxid. Redox Signal. 28, 385-400.
Collapse
Affiliation(s)
- Bo Li
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| | - Xin He
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| | - Mengru Zhuang
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| | - Bowen Niu
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| | - Chongyang Wu
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| | - Hailong Mu
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| | - Furong Tang
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| | - Yanhua Cui
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| | - Weishuai Liu
- 3 Department of Pathology, Yangling Demonstration Zone Hospital , Yangling, Shaanxi, China
| | - Baoyu Zhao
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| | - Sha Peng
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| | - Guangpeng Li
- 2 Key Laboratory for Mammalian Reproductive Biology and Biotechnology, Ministry of Education, Inner Mongolia University , Hohhot, China
| | - Jinlian Hua
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| |
Collapse
|
29
|
Choudhury AR, Singh KK. Mitochondrial determinants of cancer health disparities. Semin Cancer Biol 2017; 47:125-146. [PMID: 28487205 PMCID: PMC5673596 DOI: 10.1016/j.semcancer.2017.05.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/25/2017] [Accepted: 05/03/2017] [Indexed: 01/10/2023]
Abstract
Mitochondria, which are multi-functional, have been implicated in cancer initiation, progression, and metastasis due to metabolic alterations in transformed cells. Mitochondria are involved in the generation of energy, cell growth and differentiation, cellular signaling, cell cycle control, and cell death. To date, the mitochondrial basis of cancer disparities is unknown. The goal of this review is to provide an understanding and a framework of mitochondrial determinants that may contribute to cancer disparities in racially different populations. Due to maternal inheritance and ethnic-based diversity, the mitochondrial genome (mtDNA) contributes to inherited racial disparities. In people of African ancestry, several germline, population-specific haplotype variants in mtDNA as well as depletion of mtDNA have been linked to cancer predisposition and cancer disparities. Indeed, depletion of mtDNA and mutations in mtDNA or nuclear genome (nDNA)-encoded mitochondrial proteins lead to mitochondrial dysfunction and promote resistance to apoptosis, the epithelial-to-mesenchymal transition, and metastatic disease, all of which can contribute to cancer disparity and tumor aggressiveness related to racial disparities. Ethnic differences at the level of expression or genetic variations in nDNA encoding the mitochondrial proteome, including mitochondria-localized mtDNA replication and repair proteins, miRNA, transcription factors, kinases and phosphatases, and tumor suppressors and oncogenes may underlie susceptibility to high-risk and aggressive cancers found in African population and other ethnicities. The mitochondrial retrograde signaling that alters the expression profile of nuclear genes in response to dysfunctional mitochondria is a mechanism for tumorigenesis. In ethnic populations, differences in mitochondrial function may alter the cross talk between mitochondria and the nucleus at epigenetic and genetic levels, which can also contribute to cancer health disparities. Targeting mitochondrial determinants and mitochondrial retrograde signaling could provide a promising strategy for the development of selective anticancer therapy for dealing with cancer disparities. Further, agents that restore mitochondrial function to optimal levels should permit sensitivity to anticancer agents for the treatment of aggressive tumors that occur in racially diverse populations and hence help in reducing racial disparities.
Collapse
Affiliation(s)
| | - Keshav K Singh
- Departments of Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA; Departments of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA; Departments of Environmental Health, University of Alabama at Birmingham, Birmingham, AL, 35294, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA; Center for Aging, University of Alabama at Birmingham, Birmingham, AL, 35294, USA; UAB Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA; Birmingham Veterans Affairs Medical Center, Birmingham, AL, 35294, USA.
| |
Collapse
|
30
|
Wu Q, Yao B, Li N, Ma L, Deng Y, Yang Y, Zeng C, Yang Z, Liu B. Nrf2 mediates redox adaptation in NOX4-overexpressed non-small cell lung cancer cells. Exp Cell Res 2017; 352:245-254. [PMID: 28196727 DOI: 10.1016/j.yexcr.2017.02.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 02/08/2017] [Accepted: 02/10/2017] [Indexed: 12/19/2022]
Abstract
The redox adaptation mechanisms in cancer cells are very complex and remain largely unclear. Our previous studies have confirmed that NADPH oxidase 4 (NOX4) is abundantly expressed in non-small cell lung cancer (NSCLC) and confers apoptosis resistance on NSCLC cells. However, the comprehensive mechanisms for NOX4-mediated oxidative resistance of cancer cells remain still undentified. The present study found that NOX4-derived H2O2 enhanced the nuclear factor erythroid 2-related factor 2 (Nrf2) stability via disruption of redox-dependent proteasomal degradation and stimulated its activity through activation of PI3K signaling. Specifically, the results showed that ectopic NOX4 expression did not induce apoptosis of A549 cells; however, inhibition of Nrf2 resulted in obvious apoptotic death of NOX4-overexpressed A549 cells, accompanied by a significant increase in H2O2 level and decrease in GSH content. Besides, inhibition of Nrf2 could suppress cell growth and efficiently reverse the enhancement effect of NOX4 on cell growth. The in vivo data confirmed that inhibition of Nrf2 could interfere apoptosis resistance in NOX4-overexpressed A549 tumors and led to cell growth inhibition. In conclusion, these results reveal that Nrf2 is critically involved in redox adaptation regulation in NOX4-overexpressed NSCLC cells. Therefore, NOX4 and Nrf2 may be promising combination targets against malignant progression of NSCLC.
Collapse
Affiliation(s)
- Qipeng Wu
- Department of Clinical Pharmacy, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Bei Yao
- Department of Clinical Pharmacy, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Ning Li
- Department of Clinical Pharmacy, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lei Ma
- Department of Clinical Pharmacy, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yanchao Deng
- Department of Clinical Pharmacy, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yang Yang
- Department of Clinical Pharmacy, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Cheng Zeng
- Department of Clinical Pharmacy, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhicheng Yang
- Department of Clinical Pharmacy, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Bing Liu
- Department of Clinical Pharmacy, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
31
|
Abstract
SIGNIFICANCE Breast cancer is a unique disease characterized by heterogeneous cell populations causing roadblocks in therapeutic medicine, owing to its complex etiology and primeval understanding of the biology behind its genesis, progression, and sustenance. Globocan statistics indicate over 1.7 million new breast cancer diagnoses in 2012, accounting for 25% of all cancer morbidities. RECENT ADVANCES Despite these dismal statistics, the introduction of molecular gene signature platforms, progressive therapeutic approaches in diagnosis, and management of breast cancer has led to more effective treatment strategies and control measures concurrent with an equally reassuring decline in the mortality rate. CRITICAL ISSUES However, an enormous body of research in this area is requisite as high mortality associated with metastatic and/or drug refractory tumors continues to present a therapeutic challenge. Despite advances in systemic chemotherapy, the median survival of patients harboring metastatic breast cancers continues to be below 2 years. FUTURE DIRECTIONS Hence, a massive effort to scrutinize and evaluate chemotherapeutics on the basis of the molecular classification of these cancers is undertaken with the objective to devise more attractive and feasible approaches to treat breast cancers and improve patients' quality of life. This review aims to summarize the current understanding of the biology of breast cancer as well as challenges faced in combating breast cancer, with special emphasis on the current battery of treatment strategies. We will also try and gain perspective from recent encounters on novel findings responsible for the progression and metastatic transformation of breast cancer cells in an endeavor to develop more targeted treatment options. Antioxid. Redox Signal. 25, 337-370.
Collapse
Affiliation(s)
- Deepika Raman
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Chuan Han Jonathan Foo
- 2 NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , Singapore, Singapore
| | - Marie-Veronique Clement
- 2 NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , Singapore, Singapore .,3 Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Shazib Pervaiz
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,2 NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , Singapore, Singapore .,4 National University Cancer Institute , NUHS, Singapore, Singapore .,5 School of Biomedical Sciences, Curtin University , Perth, Australia
| |
Collapse
|