1
|
Sheng YC, Huang JN, Wu WL, Wan XR, Wang J, Qin ZH, Wang Y. TIGAR plays neuroprotective roles in MPP +/MPTP-induced Parkinson's disease by alleviating ferroptosis. Eur J Pharmacol 2025; 995:177430. [PMID: 40015596 DOI: 10.1016/j.ejphar.2025.177430] [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: 10/28/2024] [Revised: 02/23/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder worldwide, characterized by the loss of dopaminergic (DA) neurons in the substantia nigra and is associated with iron dyshomeostasis. Ferroptosis, a form of programmed cell death, involves iron-dependent lipid peroxidation and serves as a significant regulatory mechanism in PD. This study identified Tp53-induced glycolysis and apoptosis regulator (TIGAR) as a potential regulator of ferroptosis resistance in PD development. In this study, we demonstrated that in HT22 cells, 1-methyl-4-phenylpyridinium (MPP+) increased lipid peroxidation levels and reduced cell viability. These effects were reversed by the ferroptosis inhibitor ferrostatin-1 (Fer-1). MPP+ also induced elevated intracellular iron ion deposition, reactive oxygen species (ROS), and the lipid peroxidation product malondialdehyde (MDA). Meanwhile, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) significantly decreased glutathione (GSH) and nicotinamide adenine dinucleotide phosphate (NADPH) levels, glutathione peroxidase (GPX) activity, and TIGAR expression, all of which were reversible with TIGAR overexpression. In an MPTP-induced in vivo PD model, TIGAR overexpression markedly increased DA neurons and reduced iron deposition. To summarize, TIGAR enhances intracellular NADPH production via the promotion of the pentose phosphate pathway (PPP), reduces intracellular glutathione disulfide (GSSG) to GSH, boosts GPX activity, and inhibits ferroptosis, thus providing neuronal protection.
Collapse
Affiliation(s)
- Yi-Chao Sheng
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China; Jinhua Municipal Central Hospital, Jinhua, Zhejiang, China
| | - Jia-Ni Huang
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Wei-Long Wu
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Xiao-Rui Wan
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Jing Wang
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Zheng-Hong Qin
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Yan Wang
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
2
|
Wu Y, Wei M, Wang M, Guo M, Yu H, Chen Y, Xu T, Zhou Y. Schisandra total lignans ameliorate neuronal ferroptosis in 3xTg-AD mice via regulating NADK/NADPH/GSH pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156612. [PMID: 40088743 DOI: 10.1016/j.phymed.2025.156612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/22/2024] [Accepted: 03/04/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder with limited treatments. Schisandra total lignans (STL), the primary active component of Schisandra chinensis, shows potential in alleviating AD-related symptoms, though the mechanisms remain unclear. PURPOSE Considering the promoting effect of neuronal ferroptosis on AD and the neuroprotective activity of STL, this study aimed to investigate the impact of STL on AD neuronal ferroptosis and elucidate its underlying mechanisms. METHODS This study used 3xTg-AD mice and SH-SY5Y cells overexpressing APPswe as models. UHPLC/Q-TOF-MS was applied for identifying components in STL extract and the plasma of 3xTg-AD mice, as well as to detect cellular endogenous metabolites for one-carbon metabolism analysis. Behavioral tests, including the Y maze, novel object recognition, Morris water maze, and open field, were conducted to assess the cognitive function and emotional state. Histopathological examinations were performed using immunofluorescence, immunohistochemistry, Nissl staining, and transmission electron microscopy. The GSH, GSSG, NAD(H), NADP(H), and MDA levels, as well as GPX and GR activity were measured using assay kits. ROS, Fe2+, and lipid peroxidation levels were detected with probes. Protein expression was evaluated by Western blot. Molecular docking, molecular dynamics simulations and cellular thermal shift assay were performed to analyze the STL-NADK interactions. RESULTS Behavioral tests indicated that STL alleviated cognitive impairments and anxiety in 3xTg-AD mice. Histological analysis showed that STL decreased hippocampal Aβ levels, inhibited hippocampal neuronal ferroptosis, and mitigated synaptic damage. Cellular assays demonstrated that STL alleviated APPswe overexpression-induced ferroptosis and synaptic damage by activating the NADK/NADPH/GSH pathway, with NADK knockdown abolishing this neuroprotective effect of STL. Computational analysis and cellular thermal shift assay identified Gomisin D as the key STL component with strong affinity for NADK, driving its neuroprotective effects. CONCLUSION NADK emerges as a novel potential therapeutic target for AD, with STL activating NADK, promoting NADPH and GSH production, thereby mitigating neuronal ferroptosis in AD.
Collapse
Affiliation(s)
- Yuying Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Mengying Wei
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Mengyao Wang
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Minsong Guo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hengyuan Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yong Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Tengfei Xu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Yuan Zhou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China.
| |
Collapse
|
3
|
Gujar V, Li H, Paull TT, Neumann CA, Weyemi U. Unraveling the nexus: Genomic instability and metabolism in cancer. Cell Rep 2025; 44:115540. [PMID: 40208791 PMCID: PMC12043202 DOI: 10.1016/j.celrep.2025.115540] [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: 11/25/2024] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/12/2025] Open
Abstract
The DNA-damage response (DDR) is a signaling network that enables cells to detect and repair genomic damage. Over the past three decades, inhibiting DDR has proven to be an effective cancer therapeutic strategy. Although cancer drugs targeting DDR have received approval for treating various cancers, tumor cells often develop resistance to these therapies, owing to their ability to undergo energetic metabolic reprogramming. Metabolic intermediates also influence tumor cells' ability to sense oxidative stress, leading to impaired redox metabolism, thus creating redox vulnerabilities. In this review, we summarize recent advances in understanding the crosstalk between DDR and metabolism. We discuss combination therapies that target DDR, metabolism, and redox vulnerabilities in cancer. We also outline potential obstacles in targeting metabolism and propose strategies to overcome these challenges.
Collapse
Affiliation(s)
- Vaibhavi Gujar
- NCI Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Haojian Li
- NCI Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tanya T Paull
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Carola A Neumann
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Urbain Weyemi
- NCI Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
4
|
Li G, Cheng M, Lin Y, Ho Y, Lin G, Chiu C, Ho H. Downregulation of NAD Kinase Expression in β-Cells Contributes to the Aging-Associated Decline in Glucose-Stimulated Insulin Secretion. Aging Cell 2025; 24:e70037. [PMID: 40045495 PMCID: PMC11984695 DOI: 10.1111/acel.70037] [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: 03/20/2024] [Revised: 02/13/2025] [Accepted: 02/20/2025] [Indexed: 04/12/2025] Open
Abstract
Nicotinamide adenine dinucleotide kinase (NADK) is essential to the generation of nicotinamide adenine dinucleotide phosphate (NADP(H)), an important metabolic coupling factor involved in glucose-stimulated insulin secretion. In the present study, we showed that the expression of Nadk and Nadk2 transcripts and NADP(H) content were lower in islets of 80-week-old (aged) mice than those of 8-week-old (young) mice. This was associated with diminished oral glucose tolerance of old mice and the glucose-stimulated insulin secretion (GSIS) response of islets. Knockdown (KD) of Nadk or Nadk2 gene expression in NIT-1 cells impaired glucose-stimulated insulin secretion. Metabolomic analysis revealed that Nadk KD specifically affected purine metabolism in glucose-stimulated cells. The levels of 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) were higher in KD cells than in the non-targeting control (NTC) cells. Phosphorylation of AMP-activated protein kinase (AMPK) was elevated in glucose-treated KD cells compared to that of NTC cells. Increased AICAR level and AMPKα phosphorylation were observed in the glucose-stimulated islets of the aged mice. Genetic and pharmacological inhibition of AMPK promoted glucose-stimulated insulin release by KD cells and the aged mouse islets. It is likely that NADK is modulatory to AMPK activation in pancreatic β-cells and to their GSIS response. Enhanced AICAR formation in KD cells was accompanied by significantly increased conversion from inosine monophosphate (IMP) in a tetrahydrofolate (THF)-dependent manner. Folate supplementation augmented the GSIS response of KD cells and aged mouse islets. Taken together, these findings suggest that the aging-associated decline in NADK expression may underlie the reduced insulin secretory capacity of pancreatic β-cells.
Collapse
Grants
- MOST 111-2634-F-182-001 Ministry of Education in Taiwan and the National Science and Technology Council, Taiwan
- EMRPD1K0441 Ministry of Education in Taiwan
- EMRPD1K0481 Ministry of Education in Taiwan
- EMRPD1L0421 Ministry of Education in Taiwan
- 110-2320-B-182-017-MY3 National Science and Technology Council
- 111-2320-B-182-011 National Science and Technology Council
- 112-2320-B-182-020-MY3 National Science and Technology Council
- 113-2320-B-182-018-MY3 National Science and Technology Council
- BMRP564 Chang Gung Memorial Hospital, Linkou
- BMRP819 Chang Gung Memorial Hospital, Linkou
- CLRPG3K0023 Chang Gung Memorial Hospital, Linkou
- CMRPD1J0263 Chang Gung Memorial Hospital, Linkou
- CMRPD1L0161 Chang Gung Memorial Hospital, Linkou
- CMRPD1L0162 Chang Gung Memorial Hospital, Linkou
- CMRPD1M0341 Chang Gung Memorial Hospital, Linkou
- CMRPD1M0342 Chang Gung Memorial Hospital, Linkou
- CMRPD1M0351 Chang Gung Memorial Hospital, Linkou
- CMRPD1M0352 Chang Gung Memorial Hospital, Linkou
- CMRPD1N0071 Chang Gung Memorial Hospital, Linkou
- CMRPD1N0151 Chang Gung Memorial Hospital, Linkou
- CMRPD1P0171 Chang Gung Memorial Hospital, Linkou
- CORPD1P0011 Chang Gung Memorial Hospital, Linkou
- CORPD1P0021 Chang Gung Memorial Hospital, Linkou
- National Science and Technology Council
- Chang Gung Memorial Hospital, Linkou
Collapse
Affiliation(s)
- Guan‐Jie Li
- Graduate Institute of Biomedical Sciences, College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Mei‐Ling Cheng
- Metabolomics Core Laboratory, Healthy Aging Research CenterChang Gung UniversityTaoyuanTaiwan
- Clinical Metabolomics Core LaboratoryChang Gung Memorial Hospital at LinkouTaoyuanTaiwan
- Department of Biomedical Sciences, College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Yu‐Ting Lin
- School of Physical Therapy, College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Yu‐Hsuan Ho
- Department of Biomedical Sciences, College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Gigin Lin
- Clinical Metabolomics Core LaboratoryChang Gung Memorial Hospital at LinkouTaoyuanTaiwan
- Department of Medical Imaging and InterventionChang Gung Memorial Hospital at Linkou and Chang Gung UniversityTaoyuanTaiwan
- Department of Medical Imaging and Radiological SciencesChang Gung UniversityTaoyuanTaiwan
| | - Chih‐Yung Chiu
- Clinical Metabolomics Core LaboratoryChang Gung Memorial Hospital at LinkouTaoyuanTaiwan
- Department of PediatricsChang Gung Memorial Hospital at Linkou, and Chang Gung UniversityTaoyuanTaiwan
- Department of PediatricsChang Gung Memorial Hospital at Keelung, and Chang Gung UniversityTaoyuanTaiwan
| | - Hung‐Yao Ho
- Graduate Institute of Biomedical Sciences, College of MedicineChang Gung UniversityTaoyuanTaiwan
- Metabolomics Core Laboratory, Healthy Aging Research CenterChang Gung UniversityTaoyuanTaiwan
- Clinical Metabolomics Core LaboratoryChang Gung Memorial Hospital at LinkouTaoyuanTaiwan
- Department of Medical Biotechnology and Laboratory Science, College of MedicineChang Gung UniversityTaoyuanTaiwan
- Research Center for Emerging Viral InfectionsChang Gung UniversityTaoyuanTaiwan
| |
Collapse
|
5
|
Mi T, Kong X, Chen M, Guo P, He D. Inducing disulfidptosis in tumors:potential pathways and significance. MedComm (Beijing) 2024; 5:e791. [PMID: 39415848 PMCID: PMC11480524 DOI: 10.1002/mco2.791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/28/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
Regulated cell death (RCD) is crucial for the elimination of abnormal cells. In recent years, strategies aimed at inducing RCD, particularly apoptosis, have become increasingly important in cancer therapy. However, the ability of tumor cells to evade apoptosis has led to treatment resistance and relapse, prompting extensive research into alternative death processes in cancer cells. A recent study identified a novel form of RCD known as disulfidptosis, which is linked to disulfide stress. Cancer cells import cystine from the extracellular environment via solute carrier family 7 member 11 (SLC7A11) and convert it to cysteine using nicotinamide adenine dinucleotide phosphate (NADPH). When NADPH is deficient or its utilization is impaired, cystine accumulates, leading to the formation of disulfide bonds in the actin cytoskeleton, triggering disulfidptosis. Disulfidptosis reveals a metabolic vulnerability in tumors, offering new insights into cancer therapy strategies. This review provides a detailed overview of the mechanisms underlying disulfidptosis, the current research progress, and limitations. It also highlights innovative strategies for inducing disulfidptosis and explores the potential of combining these approaches with traditional cancer therapies, particularly immunotherapy, to expedite clinical translation.
Collapse
Affiliation(s)
- Tao Mi
- Department of UrologyChildren's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and DisordersChongqingP.R. China
- Chongqing Key Laboratory of Structural Birth Defect and ReconstructionChongqingP.R. China
| | - Xiangpan Kong
- Department of UrologyChildren's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and DisordersChongqingP.R. China
- Chongqing Key Laboratory of Structural Birth Defect and ReconstructionChongqingP.R. China
| | - Meiling Chen
- Department of UrologyChildren's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and DisordersChongqingP.R. China
- Chongqing Key Laboratory of Structural Birth Defect and ReconstructionChongqingP.R. China
| | - Peng Guo
- Department of UrologyChildren's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and DisordersChongqingP.R. China
- Chongqing Key Laboratory of Structural Birth Defect and ReconstructionChongqingP.R. China
- Institute of Basic Medicine and Cancer (IBMC)Chinese Academy of SciencesHangzhouP.R. China
| | - Dawei He
- Department of UrologyChildren's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and DisordersChongqingP.R. China
- Chongqing Key Laboratory of Structural Birth Defect and ReconstructionChongqingP.R. China
| |
Collapse
|
6
|
Huang Y, Zhao H, Zhang Y, Zhao C, Ren J, Qu X. Bioorthogonal Regulated Metabolic Balance for Promotion of Ferroptosis and Mild Photothermal Therapy. ACS NANO 2024; 18:28104-28114. [PMID: 39373015 DOI: 10.1021/acsnano.4c07558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The nanozyme with NADPH oxidase (NOX)-like activity can promote the consumption of NADPH and the generation of free radicals. In consideration of that the upregulation of glucose-6-phosphate dehydrogenase (G6PD) would accelerate the compensation production of NADPH, for inhibition of G6PD activity, our designed bioorthogonal nanozyme can in situ catalyze pro-DHEA to produce G6PD inhibitor and dehydroepiandrosterone (DHEA) drugs to inhibit G6PD activity. Therefore, the well-defined platform can disrupt NADPH homeostasis, leading to the collapse of the antioxidant defense system in the tumor cells. The enzyme-like activity of PdCuFe is further enhanced when irradiated by NIR-II light. The destruction of NADPH homeostasis can promote ferroptosis and, in turn, facilitate mild photothermal therapy. Our design can realize NADPH depletion and greatly improve the therapeutic effect through metabolic regulation, which may provide inspiration for the design of bioorthogonal catalysis.
Collapse
Affiliation(s)
- Ying Huang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Huisi Zhao
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Yu Zhang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Chuanqi Zhao
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| |
Collapse
|
7
|
Chen L, Xing X, Zhang P, Chen L, Pei H. Homeostatic regulation of NAD(H) and NADP(H) in cells. Genes Dis 2024; 11:101146. [PMID: 38988322 PMCID: PMC11233901 DOI: 10.1016/j.gendis.2023.101146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/31/2023] [Accepted: 09/11/2023] [Indexed: 07/12/2024] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+)/reduced NAD+ (NADH) and nicotinamide adenine dinucleotide phosphate (NADP+)/reduced NADP+ (NADPH) are essential metabolites involved in multiple metabolic pathways and cellular processes. NAD+ and NADH redox couple plays a vital role in catabolic redox reactions, while NADPH is crucial for cellular anabolism and antioxidant responses. Maintaining NAD(H) and NADP(H) homeostasis is crucial for normal physiological activity and is tightly regulated through various mechanisms, such as biosynthesis, consumption, recycling, and conversion between NAD(H) and NADP(H). The conversions between NAD(H) and NADP(H) are controlled by NAD kinases (NADKs) and NADP(H) phosphatases [specifically, metazoan SpoT homolog-1 (MESH1) and nocturnin (NOCT)]. NADKs facilitate the synthesis of NADP+ from NAD+, while MESH1 and NOCT convert NADP(H) into NAD(H). In this review, we summarize the physiological roles of NAD(H) and NADP(H) and discuss the regulatory mechanisms governing NAD(H) and NADP(H) homeostasis in three key aspects: the transcriptional and posttranslational regulation of NADKs, the role of MESH1 and NOCT in maintaining NAD(H) and NADP(H) homeostasis, and the influence of the circadian clock on NAD(H) and NADP(H) homeostasis. In conclusion, NADKs, MESH1, and NOCT are integral to various cellular processes, regulating NAD(H) and NADP(H) homeostasis. Dysregulation of these enzymes results in various human diseases, such as cancers and metabolic disorders. Hence, strategies aiming to restore NAD(H) and NADP(H) homeostasis hold promise as novel therapeutic approaches for these diseases.
Collapse
Affiliation(s)
- Luojun Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430062, China
| | - Xiaoke Xing
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430062, China
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Pingfeng Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430062, China
| | - Lulu Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430062, China
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Huadong Pei
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
8
|
Zhang Y, Qiu S, Shao S, Cao Y, Hong Y, Xu X, Fang X, Di C, Yang J, Tan X. NMN partially rescues cuproptosis by upregulating sirt2 to increase intracellular NADPH. Sci Rep 2024; 14:19392. [PMID: 39169144 PMCID: PMC11339376 DOI: 10.1038/s41598-024-70245-5] [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: 03/16/2024] [Accepted: 08/14/2024] [Indexed: 08/23/2024] Open
Abstract
Cuproptosis is characterized by lipoylated protein aggregation and loss of iron-sulfur (Fe-S) proteins, which are crucial for a wide range of important cellular functions, including DNA replication and damage repair. Sirt2 and sirt4 are lipoamidases that remove the lipoyl moiety from lipoylated proteins using nicotinamide adenine dinucleotide (NAD+) as a cofactor. However, to date, it is not clear whether nicotinamide mononucleotide (NMN), a precursor of NAD+, affects cellular sensitivity to cuproptosis. Therefore, in the current study, cuproptosis was induced by the copper (Cu) ionophore elesclomol (Es) in HeLa cells. It was also found that Es/Cu treatment increased cellular DNA damage level. On the other hand, NMN treatment partially rescued cuproptosis in a dose-dependent manner, as well as reduced cellular DNA damage level. In addition, NMN upregulated the expression of Fe-S protein POLD1, without affecting the aggregation of lipoylated proteins. Mechanistic study revealed that NMN increased the expression of sirt2 and cellular reduced nicotinamide adenine dinucleotide phosphate (NADPH) level. Overexpression of sirt2 and sirt4 did not change the aggregation of lipoylated proteins, however, sirt2, but not sirt4, increased cellular NADPH levels and partially rescued cuproptosis. Inhibition of NAD+ kinase (NADK), which is responsible for generating NADPH, abolished the rescuing function of NMN and sirt2 for Es/Cu induced cell death. Taken together, our results suggested that DNA damage is a characteristic feature of cuproptosis. NMN can partially rescue cuproptosis by upregulating sirt2, increase intracellular NADPH content and maintain the level of Fe-S proteins, independent of the lipoamidase activity of sirt2.
Collapse
Affiliation(s)
- Yingying Zhang
- School of Public Health, Hangzhou Normal University, Hangzhou, China
- The Affiliated Hospital, Hangzhou Normal University, Hangzhou, China
| | - Shuting Qiu
- School of Public Health, Hangzhou Normal University, Hangzhou, China
- The Affiliated Hospital, Hangzhou Normal University, Hangzhou, China
| | - Shihan Shao
- School of Public Health, Hangzhou Normal University, Hangzhou, China
- The Affiliated Hospital, Hangzhou Normal University, Hangzhou, China
| | - Yuejia Cao
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Yu Hong
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Xianrong Xu
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Xuexian Fang
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Chunhong Di
- The Affiliated Hospital, Hangzhou Normal University, Hangzhou, China.
| | - Jun Yang
- School of Public Health, Hangzhou Normal University, Hangzhou, China.
| | - Xiaohua Tan
- School of Public Health, Hangzhou Normal University, Hangzhou, China.
| |
Collapse
|
9
|
Chang H, Clemens S, Gao P, Li Q, Zhao H, Wang L, Zhang J, Zhou P, Johnsson K, Wang L. Fluorogenic Rhodamine-Based Chemigenetic Biosensor for Monitoring Cellular NADPH Dynamics. J Am Chem Soc 2024. [PMID: 39037873 DOI: 10.1021/jacs.3c13137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Ratiometric biosensors employing Förster Resonance Energy Transfer (FRET) enable the real-time tracking of metabolite dynamics. Here, we introduce an approach for generating a FRET-based biosensor in which changes in apparent FRET efficiency rely on the analyte-controlled fluorogenicity of a rhodamine rather than the commonly used distance change between donor-acceptor fluorophores. Our fluorogenic, rhodamine-based, chemigenetic biosensor (FOCS) relies on a synthetic, protein-tethered FRET probe, in which the rhodamine acting as the FRET acceptor switches in an analyte-dependent manner from a dark to a fluorescent state. This allows ratiometric sensing of the analyte concentration. We use this approach to generate a chemigenetic biosensor for nicotinamide adenine dinucleotide phosphate (NADPH). FOCS-NADPH exhibits a rapid and reversible response toward NAPDH with a good dynamic range, selectivity, and pH insensitivity. FOCS-NADPH allows real-time monitoring of cytosolic NADPH fluctuations in live cells during oxidative stress or after drug exposure. We furthermore used FOCS-NADPH to investigate NADPH homeostasis regulation through the pentose phosphate pathway of glucose metabolism. FOCS-NADPH is a powerful tool for studying NADPH metabolism and serves as a blueprint for the development of future fluorescent biosensors.
Collapse
Affiliation(s)
- Huimin Chang
- Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Zhangheng Road 826, Shanghai 201203, China
| | - Simon Clemens
- Department of Chemical Biology, Max Planck Institute for Medical Research, Jahnstrasse 29, Heidelberg D-69120, Germany
| | - Pingting Gao
- Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Zhangheng Road 826, Shanghai 201203, China
| | - Quanlin Li
- Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Zhangheng Road 826, Shanghai 201203, China
| | - Hanqing Zhao
- Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Zhangheng Road 826, Shanghai 201203, China
| | - Lehua Wang
- Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Zhangheng Road 826, Shanghai 201203, China
| | - Jingye Zhang
- Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Zhangheng Road 826, Shanghai 201203, China
| | - Pinghong Zhou
- Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Zhangheng Road 826, Shanghai 201203, China
| | - Kai Johnsson
- Department of Chemical Biology, Max Planck Institute for Medical Research, Jahnstrasse 29, Heidelberg D-69120, Germany
- Institute of Chemical Sciences and Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Lu Wang
- Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Zhangheng Road 826, Shanghai 201203, China
| |
Collapse
|
10
|
Jiang H, Zuo J, Li B, Chen R, Luo K, Xiang X, Lu S, Huang C, Liu L, Tang J, Gao F. Drug-induced oxidative stress in cancer treatments: Angel or devil? Redox Biol 2023; 63:102754. [PMID: 37224697 DOI: 10.1016/j.redox.2023.102754] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/03/2023] [Accepted: 05/17/2023] [Indexed: 05/26/2023] Open
Abstract
Oxidative stress (OS), defined as redox imbalance in favor of oxidant burden, is one of the most significant biological events in cancer progression. Cancer cells generally represent a higher oxidant level, which suggests a dual therapeutic strategy by regulating redox status (i.e., pro-oxidant therapy and/or antioxidant therapy). Indeed, pro-oxidant therapy exhibits a great anti-cancer capability, attributing to a higher oxidant accumulation within cancer cells, whereas antioxidant therapy to restore redox homeostasis has been claimed to fail in several clinical practices. Targeting the redox vulnerability of cancer cells by pro-oxidants capable of generating excessive reactive oxygen species (ROS) has surfaced as an important anti-cancer strategy. However, multiple adverse effects caused by the indiscriminate attacks of uncontrolled drug-induced OS on normal tissues and the drug-tolerant capacity of some certain cancer cells greatly limit their further applications. Herein, we review several representative oxidative anti-cancer drugs and summarize their side effects on normal tissues and organs, emphasizing that seeking a balance between pro-oxidant therapy and oxidative damage is of great value in exploiting next-generation OS-based anti-cancer chemotherapeutics.
Collapse
Affiliation(s)
- Hao Jiang
- The First Hospital of Ningbo University, Ningbo, 315020, China
| | - Jing Zuo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bowen Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rui Chen
- The First Hospital of Ningbo University, Ningbo, 315020, China
| | - Kangjia Luo
- The First Hospital of Ningbo University, Ningbo, 315020, China
| | - Xionghua Xiang
- The First Hospital of Ningbo University, Ningbo, 315020, China
| | - Shuaijun Lu
- The First Hospital of Ningbo University, Ningbo, 315020, China
| | - Canhua Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lin Liu
- Ningbo Women & Children's Hospital, Ningbo, 315012, China.
| | - Jing Tang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.
| | - Feng Gao
- The First Hospital of Ningbo University, Ningbo, 315020, China.
| |
Collapse
|
11
|
Ahmed AF, Dai CF, Kuo YH, Sheu JH. The Invasive Anemone Condylactis sp. of the Coral Reef as a Source of Sulfur- and Nitrogen-Containing Metabolites and Cytotoxic 5,8-Epidioxy Steroids. Metabolites 2023; 13:metabo13030392. [PMID: 36984832 PMCID: PMC10056678 DOI: 10.3390/metabo13030392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/10/2023] Open
Abstract
The Condylactis-genus anemones were examined for their proteinaceous poisons over 50 years ago. On the other hand, the current research focuses on isolating and describing the non-proteinaceous secondary metabolites from the invasive Condylactis anemones, which help take advantage of their population outbreak as a new source of chemical candidates and potential drug leads. From an organic extract of Condylactis sp., a 1,2,4-thiadiazole-based alkaloid, identified as 3,5-bis(3-pyridinyl)-1,2,4-thiadiazole (1), was found to be a new natural alkaloid despite being previously synthesized. The full assignment of NMR data of compound 1, based on the analysis of 2D NMR correlations, is reported herein for the first time. The proposed biosynthetic precursor thionicotinamide (2) was also isolated for the first time from nature along with nicotinamide (3), uridine (5), hypoxanthine (6), and four 5,8-epidioxysteroids (7–10). A major secondary metabolite (−)-betonicine (4) was isolated from Condylactis sp. and found for the first time in marine invertebrates. The four 5,8-epidioxysteroids, among other metabolites, exhibited cytotoxicity (IC50 3.5–9.0 μg/mL) toward five cancer cell lines.
Collapse
Affiliation(s)
- Atallah F. Ahmed
- Department of Marine Resources, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Chang-Feng Dai
- Institute of Oceanography, National Taiwan University, Taipei 106, Taiwan
| | - Yao-Haur Kuo
- Division of Herbal Drugs and Natural Products, National Research Institute of Chinese Medicine, Taipei 112, Taiwan
| | - Jyh-Horng Sheu
- Department of Marine Resources, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
- Frontier Center for Ocean Science and Technology, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Correspondence: ; Tel.: +88-(67)-5252000 (ext. 5030)
| |
Collapse
|
12
|
lncRNA ELFN1-AS1 promotes proliferation, migration and invasion and suppresses apoptosis in colorectal cancer cells by enhancing G6PD activity. Acta Biochim Biophys Sin (Shanghai) 2023; 55:649-660. [PMID: 36786074 DOI: 10.3724/abbs.2023010] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Tumour cells change their metabolic patterns to support high proliferation rates and cope with oxidative stress. The lncRNA ELFN1-AS1 is highly expressed in a wide range of cancers and is essential to the proliferation and apoptosis of tumour cells. Nevertheless, its function in the metabolic reprogramming of tumour cells is unclear. Here we show that ELFN1-AS1 promotes glucose consumption as well as lactate and NADPH production. Database searching, bioinformatics analysis, RNA immunoprecipitation (RIP) and RNA pull-down assays show that ELFN1-AS1 enhances glucose-6-phosphate dehydrogenase ( G6PD) expression and activates the pentose phosphate pathway (PPP) by promoting TP53 degradation. In addition, luciferase reporter assay and chromatin immunoprecipitation (ChIP) show that YY1 binds to the ELFN1-AS1 promoter to promote transcriptional activation of ELFN1-AS1. Consistent with the in vitro experiments, knockdown of ELFN1-AS1 impedes the growth of tumours transplanted into mice by inhibiting the expression of G6PD. In conclusion, this study reveals that ELFN1-AS1 activates the PPP, and validates the regulatory role of the YY1/ ELFN1-AS1/ TP53/ G6PD axis in colorectal cancer.
Collapse
|
13
|
Oka SI, Titus AS, Zablocki D, Sadoshima J. Molecular properties and regulation of NAD + kinase (NADK). Redox Biol 2022; 59:102561. [PMID: 36512915 PMCID: PMC9763689 DOI: 10.1016/j.redox.2022.102561] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 11/27/2022] [Indexed: 12/11/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) kinase (NADK) phosphorylates NAD+, thereby producing nicotinamide adenine dinucleotide phosphate (NADP). Both NADK genes and the NADP(H)-producing mechanism are evolutionarily conserved among archaea, bacteria, plants and mammals. In mammals, NADK is activated by phosphorylation and protein-protein interaction. Recent studies conducted using genetically altered models validate the essential role of NADK in cellular redox homeostasis and metabolism in multicellular organisms. Here, we describe the evolutionary conservation, molecular properties, and signaling mechanisms and discuss the pathophysiological significance of NADK.
Collapse
Affiliation(s)
| | | | | | - Junichi Sadoshima
- Rutgers New Jersey Medical School Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, Newark, NJ, 07101, USA.
| |
Collapse
|
14
|
Liu T, Shi W, Ding Y, Wu Q, Zhang B, Zhang N, Wang M, Du D, Zhang H, Han B, Guo D, Zheng J, Li Q, Luo C. (-)-Epigallocatechin Gallate is a Noncompetitive Inhibitor of NAD Kinase. ACS Med Chem Lett 2022; 13:1699-1706. [PMID: 36385933 PMCID: PMC9661698 DOI: 10.1021/acsmedchemlett.2c00163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 10/26/2022] [Indexed: 11/29/2022] Open
Abstract
Nicotinamide adenine dinucleotide kinase (NADK) controls the intracellular NADPH content and provides reducing power for the synthesis of macromolecules and anti-ROS. Moreover, NADK is considered to be a synthetic lethal gene for KRAS mutations. To discover NADK-targeted probes, a high-throughput screening assay was established and optimized with a Z factor of 0.71. The natural product (-)-epigallocatechin gallate (EGCG) was found to be a noncompetitive inhibitor of NADK with K i = 3.28 ± 0.32 μΜ. The direct binding of EGCG to NADK was determined by several biophysical methods, including NMR spectroscopy, surface plasmon resonance (SPR) assay, and hydrogen-deuterium exchange mass spectrometry (HDX-MS). The SPR assay showed a K d of 1.78 ± 1.15 μΜ. The HDX-MS experiment showed that EGCG was bound at the non-substrate-binding sites of NADK. Besides, binding mode prediction and derivative activity analysis revealed a potential structure-activity relationship between EGCG and NADK. Furthermore, EGCG can specifically inhibit the proliferation of KRAS-mutated lung cancer cell lines without affecting KRAS wild-type lung cancer cell lines.
Collapse
Affiliation(s)
- Tonghai Liu
- School
of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
- State
Key Laboratory of Drug Research, Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenjia Shi
- School
of Chinese Materia Medica, Nanjing University
of Chinese Medicine, Nanjing 210023, China
| | - Yiluan Ding
- State
Key Laboratory of Drug Research, Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Qiqi Wu
- School
of Chinese Materia Medica, Nanjing University
of Chinese Medicine, Nanjing 210023, China
| | - Bei Zhang
- State
Key Laboratory of Drug Research, Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Naixia Zhang
- State
Key Laboratory of Drug Research, Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Mingliang Wang
- Zhongshan
Institute for Drug Discovery, Shanghai Institute
of Materia Medica, Chinese Academy of Sciences, Zhongshan 528437, China
| | - Daohai Du
- State
Key Laboratory of Drug Research, Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hao Zhang
- State
Key Laboratory of Drug Research, Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Bo Han
- School
of Pharmacy/Key Laboratory of Xinjiang Phytomedicine Resource and
Utilization, Ministry of Education, Shihezi
University, Shihezi 832003, China
| | - Dean Guo
- State
Key Laboratory of Drug Research, Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jie Zheng
- State
Key Laboratory of Drug Research, Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi Li
- State
Key Laboratory of Drug Research, Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Cheng Luo
- School
of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
- State
Key Laboratory of Drug Research, Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
- School
of Chinese Materia Medica, Nanjing University
of Chinese Medicine, Nanjing 210023, China
- Zhongshan
Institute for Drug Discovery, Shanghai Institute
of Materia Medica, Chinese Academy of Sciences, Zhongshan 528437, China
| |
Collapse
|
15
|
Inhibition of NAD kinase elevates the hepatic NAD+ pool and alleviates acetaminophen-induced acute liver injury in mice. Biochem Biophys Res Commun 2022; 612:70-76. [DOI: 10.1016/j.bbrc.2022.04.079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/14/2022] [Accepted: 04/18/2022] [Indexed: 11/22/2022]
|
16
|
Gupta A, Vankar JK, Jadav JP, Gururaja GN. Water Mediated Direct Thioamidation of Aldehydes at Room Temperature. J Org Chem 2022; 87:2410-2420. [PMID: 35133151 DOI: 10.1021/acs.joc.1c02307] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A mild, greener approach toward thioamide synthesis has been developed. Its unique features include water-mediated reaction with no input energy, additives, or catalysts as well. The presented protocol is attractive with readily available starting materials and the use of different array amines, along with a scaled-up method. Biologically active molecules such as thionicotinamide and thioisonicotinamide can be synthesized from this procedure.
Collapse
Affiliation(s)
- Ankush Gupta
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar 382030, India
| | - Jigarkumar K Vankar
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar 382030, India
| | - Jaydeepbhai P Jadav
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar 382030, India
| | | |
Collapse
|
17
|
Colombo G, Gelardi ELM, Balestrero FC, Moro M, Travelli C, Genazzani AA. Insight Into Nicotinamide Adenine Dinucleotide Homeostasis as a Targetable Metabolic Pathway in Colorectal Cancer. Front Pharmacol 2021; 12:758320. [PMID: 34880756 PMCID: PMC8645963 DOI: 10.3389/fphar.2021.758320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 11/02/2021] [Indexed: 11/13/2022] Open
Abstract
Tumour cells modify their cellular metabolism with the aim to sustain uncontrolled proliferation. Cancer cells necessitate adequate amounts of NAD and NADPH to support several enzymes that are usually overexpressed and/or overactivated. Nicotinamide adenine dinucleotide (NAD) is an essential cofactor and substrate of several NAD-consuming enzymes, such as PARPs and sirtuins, while NADPH is important in the regulation of the redox status in cells. The present review explores the rationale for targeting the key enzymes that maintain the cellular NAD/NADPH pool in colorectal cancer and the enzymes that consume or use NADP(H).
Collapse
Affiliation(s)
- Giorgia Colombo
- Department of Pharmaceutical Sciences, Università Del Piemonte Orientale, Novara, Italy
| | | | | | - Marianna Moro
- Department of Pharmaceutical Sciences, Università Del Piemonte Orientale, Novara, Italy
| | - Cristina Travelli
- Department of Drug Sciences, Università Degli Studi di Pavia, Pavia, Italy
| | - Armando A. Genazzani
- Department of Pharmaceutical Sciences, Università Del Piemonte Orientale, Novara, Italy
| |
Collapse
|
18
|
Oka SI, Byun J, Huang CY, Imai N, Ralda G, Zhai P, Xu X, Kashyap S, Warren JS, Alan Maschek J, Tippetts TS, Tong M, Venkatesh S, Ikeda Y, Mizushima W, Kashihara T, Sadoshima J. Nampt Potentiates Antioxidant Defense in Diabetic Cardiomyopathy. Circ Res 2021; 129:114-130. [PMID: 33928788 PMCID: PMC8513534 DOI: 10.1161/circresaha.120.317943] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
MESH Headings
- Animals
- Antioxidants/metabolism
- Apoptosis
- Autophagy
- Cells, Cultured
- Cytokines/genetics
- Cytokines/metabolism
- Diabetic Cardiomyopathies/enzymology
- Diabetic Cardiomyopathies/genetics
- Diabetic Cardiomyopathies/pathology
- Diet, High-Fat
- Disease Models, Animal
- Fibrosis
- Glutathione/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria, Heart/enzymology
- Mitochondria, Heart/genetics
- Mitochondria, Heart/pathology
- Mitophagy
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- NAD/metabolism
- NADP/metabolism
- Nicotinamide Phosphoribosyltransferase/genetics
- Nicotinamide Phosphoribosyltransferase/metabolism
- Oxidative Stress
- Rats, Wistar
- Sirtuins/genetics
- Sirtuins/metabolism
- Thioredoxins/metabolism
- Mice
- Rats
Collapse
Affiliation(s)
- Shin-Ichi Oka
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Jaemin Byun
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Chun-Yang Huang
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taiwan (C.-y.H.)
- Institute of Clinical Medicine, School of Medicine National Yang-Ming University, Taipei, Taiwan (C.-y.H.)
| | - Nobushige Imai
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Guersom Ralda
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Peiyong Zhai
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Xiaoyong Xu
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
- Department of Cardiology, Ningbo Medical Center Lihuili Hospital, Zhejiang, China (X.X.)
| | - Sanchita Kashyap
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Junco S Warren
- Nora Eccles Harrison Cardiovascular Research and Training Institute (J.S.W.), University of Utah, Salt Lake City
| | - John Alan Maschek
- Metabolomics, Proteomics, and Mass Spectrometry Cores (J.A.M.), University of Utah, Salt Lake City
- Department of Nutrition and Integrative Physiology (J.A.M.), University of Utah, Salt Lake City
| | - Trevor S Tippetts
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center (T.S.T., S.V.), University of Utah, Salt Lake City
| | - Mingming Tong
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Sundararajan Venkatesh
- Department of Microbiology, Biochemistry, and Molecular Genetics (S.V.), Rutgers New Jersey Medical School, Newark
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center (T.S.T., S.V.), University of Utah, Salt Lake City
| | - Yoshiyuki Ikeda
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Wataru Mizushima
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Toshihide Kashihara
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| |
Collapse
|
19
|
Yang H, Xie S, Liang B, Tang Q, Liu H, Wang D, Huang G. Exosomal IDH1 increases the resistance of colorectal cancer cells to 5-Fluorouracil. J Cancer 2021; 12:4862-4872. [PMID: 34234856 PMCID: PMC8247374 DOI: 10.7150/jca.58846] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/30/2021] [Indexed: 11/05/2022] Open
Abstract
Chemoresistance challenges the clinical treatment of colorectal cancer and requires an urgent solution. Isocitrate dehydrogenase 1 (IDH1) is a key enzyme involved in glucose metabolism that mediates the malignant transformation of tumors. However, the mechanisms by which IDH1 is involved in colorectal cancer cell proliferation and drug resistance induction remain unclear. In this study, we found that IDH1 was highly expressed in human colorectal cancer tissues and could be used to indicate a high-grade tumor. In vitro gene overexpression and knockdown were used to determine whether IDH1 promoted the proliferation of the colorectal cancer cell line HCT8 and resistance to 5-Fluorouracil (5FU). Further studies have shown that the 5FU-resistant cell line, HCT8FU, secreted exosomes that contained a high level of IDH1 protein. The exosomal IDH1 derived from 5FU-resistant cells enhanced the resistance of 5FU-sensitive cells. Metabolic assays revealed that exosomes derived from 5FU-resistant cells promoted a decrease in the level of IDH1-mediated NADPH, which is associated with the development of 5FU resistance in colorectal cancer cells. Therefore, exosomal IDH1 may be the transmitter and driver of chemoresistance in colorectal cancer and a potential chemotherapy target.
Collapse
Affiliation(s)
- Hao Yang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.,Department of Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Sha Xie
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Beibei Liang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Qiqi Tang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Huanchen Liu
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Dongliang Wang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.,Department of Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.,Department of Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| |
Collapse
|
20
|
Rather GM, Pramono AA, Szekely Z, Bertino JR, Tedeschi PM. In cancer, all roads lead to NADPH. Pharmacol Ther 2021; 226:107864. [PMID: 33894275 DOI: 10.1016/j.pharmthera.2021.107864] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 02/06/2023]
Abstract
Cancer cells require increased levels of NADPH for increased nucleotide synthesis and for protection from ROS. Recent studies show that increased NADPH is generated in several ways. Activated AKT phosphorylates NAD kinase (NADK), increasing its activity. NADP formed, is rapidly converted to NADPH by glucose 6-phosphate dehydrogenase and malic enzymes, overexpressed in tumor cells with mutant p53. Calmodulin, overexpressed in some cancers, also increases NADK activity. Also, in IDH1/2 mutant cancer, NADPH serves as the cofactor to generate D-2 hydroxyglutarate, an oncometabolite. The requirement of cancer cells for elevated levels of NADPH provides an opportunity to target its synthesis for cancer treatment.
Collapse
Affiliation(s)
- Gulam Mohmad Rather
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Alvinsyah Adhityo Pramono
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA; Research Center of Medical Genetics, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Zoltan Szekely
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA; Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA; Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Joseph R Bertino
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA; Department of Medicine and Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.
| | - Philip Michael Tedeschi
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
21
|
Rabani R, Cossette C, Graham F, Powell WS. Protein kinase C activates NAD kinase in human neutrophils. Free Radic Biol Med 2020; 161:50-59. [PMID: 33011272 DOI: 10.1016/j.freeradbiomed.2020.09.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 08/14/2020] [Accepted: 09/26/2020] [Indexed: 12/23/2022]
Abstract
NAD kinase (NADK) is required for the de novo synthesis of NADP+ from NAD+. In neutrophils, NADK plays an essential role by providing sufficient levels of NADPH to support a robust oxidative burst. Activation of NADPH oxidase-2 (NOX-2) in neutrophils by stimulators of protein kinase C (PKC), such as phorbol myristate acetate (PMA), results in the rapid generation of superoxide at the expense of oxidation of NADPH to NADP+. In this study, we measured the levels of pyridine nucleotides following the addition of PMA to neutrophils. PMA elicited a rapid increase in NADP+ in neutrophils, which was not due to oxidation of NADPH, the levels of which also rose. This was mirrored by a rapid reduction in NAD+ levels, suggesting that NADK had been activated. PMA-induced depletion of NAD+ in neutrophils was blocked by PKC inhibitors, but was not dependent on NOX-2, as it was not blocked by the NOX inhibitor, diphenyleneiodonium. PMA also increased NADK activity in neutrophil lysates as well as NADK phosphorylation, as revealed by a monoclonal antibody selective for phospho-NADK. Human recombinant NADK was phosphorylated by PKCδ, resulting in increased immunoreactivity, but unchanged enzyme activity, suggesting that PKC-induced phosphorylation alone is insufficient to increase NADK activity in neutrophils. This leads us to speculate that phosphorylation of NADK promotes the dissociation of an inhibitory molecule from a complex, thereby increasing enzyme activity. Activation of NADK by PKC in phagocytic cells could be critical for the rapid provision of sufficient levels of superoxide for host defence against invading microorganisms.
Collapse
Affiliation(s)
- Razieh Rabani
- Meakins-Christie Laboratories, Centre for Translational Biology, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC H4A 3J1, Canada
| | - Chantal Cossette
- Meakins-Christie Laboratories, Centre for Translational Biology, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC H4A 3J1, Canada
| | - François Graham
- Meakins-Christie Laboratories, Centre for Translational Biology, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC H4A 3J1, Canada
| | - William S Powell
- Meakins-Christie Laboratories, Centre for Translational Biology, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC H4A 3J1, Canada.
| |
Collapse
|
22
|
NADPH homeostasis in cancer: functions, mechanisms and therapeutic implications. Signal Transduct Target Ther 2020; 5:231. [PMID: 33028807 PMCID: PMC7542157 DOI: 10.1038/s41392-020-00326-0] [Citation(s) in RCA: 285] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 08/09/2020] [Accepted: 09/14/2020] [Indexed: 02/08/2023] Open
Abstract
Nicotinamide adenine dinucleotide phosphate (NADPH) is an essential electron donor in all organisms, and provides the reducing power for anabolic reactions and redox balance. NADPH homeostasis is regulated by varied signaling pathways and several metabolic enzymes that undergo adaptive alteration in cancer cells. The metabolic reprogramming of NADPH renders cancer cells both highly dependent on this metabolic network for antioxidant capacity and more susceptible to oxidative stress. Modulating the unique NADPH homeostasis of cancer cells might be an effective strategy to eliminate these cells. In this review, we summarize the current existing literatures on NADPH homeostasis, including its biological functions, regulatory mechanisms and the corresponding therapeutic interventions in human cancers, providing insights into therapeutic implications of targeting NADPH metabolism and the associated mechanism for cancer therapy.
Collapse
|
23
|
Pramono AA, Rather GM, Herman H, Lestari K, Bertino JR. NAD- and NADPH-Contributing Enzymes as Therapeutic Targets in Cancer: An Overview. Biomolecules 2020; 10:biom10030358. [PMID: 32111066 PMCID: PMC7175141 DOI: 10.3390/biom10030358] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/19/2020] [Accepted: 02/21/2020] [Indexed: 12/14/2022] Open
Abstract
Actively proliferating cancer cells require sufficient amount of NADH and NADPH for biogenesis and to protect cells from the detrimental effect of reactive oxygen species. As both normal and cancer cells share the same NAD biosynthetic and metabolic pathways, selectively lowering levels of NAD(H) and NADPH would be a promising strategy for cancer treatment. Targeting nicotinamide phosphoribosyltransferase (NAMPT), a rate limiting enzyme of the NAD salvage pathway, affects the NAD and NADPH pool. Similarly, lowering NADPH by mutant isocitrate dehydrogenase 1/2 (IDH1/2) which produces D-2-hydroxyglutarate (D-2HG), an oncometabolite that downregulates nicotinate phosphoribosyltransferase (NAPRT) via hypermethylation on the promoter region, results in epigenetic regulation. NADPH is used to generate D-2HG, and is also needed to protect dihydrofolate reductase, the target for methotrexate, from degradation. NAD and NADPH pools in various cancer types are regulated by several metabolic enzymes, including methylenetetrahydrofolate dehydrogenase, serine hydroxymethyltransferase, and aldehyde dehydrogenase. Thus, targeting NAD and NADPH synthesis under special circumstances is a novel approach to treat some cancers. This article provides the rationale for targeting the key enzymes that maintain the NAD/NADPH pool, and reviews preclinical studies of targeting these enzymes in cancers.
Collapse
Affiliation(s)
- Alvinsyah Adhityo Pramono
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA; (A.A.P.); (G.M.R.)
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Gulam M. Rather
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA; (A.A.P.); (G.M.R.)
| | - Herry Herman
- Division of Oncology, Department of Orthopaedic Surgery, Faculty of Medicine, Universitas Padjadjaran, Bandung 40161, Indonesia;
| | - Keri Lestari
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Joseph R. Bertino
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA; (A.A.P.); (G.M.R.)
- Department of Pharmacology and Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Correspondence: ; Tel.: +1-(732)-235-8510
| |
Collapse
|
24
|
Depaix A, Kowalska J. NAD Analogs in Aid of Chemical Biology and Medicinal Chemistry. Molecules 2019; 24:molecules24224187. [PMID: 31752261 PMCID: PMC6891637 DOI: 10.3390/molecules24224187] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/06/2019] [Accepted: 11/12/2019] [Indexed: 12/21/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) serves as an essential redox co-factor and mediator of multiple biological processes. Besides its well-established role in electron transfer reactions, NAD serves as a substrate for other biotransformations, which, at the molecular level, can be classified as protein post-translational modifications (protein deacylation, mono-, and polyADP-ribosylation) and formation of signaling molecules (e.g., cyclic ADP ribose). These biochemical reactions control many crucial biological processes, such as cellular signaling and recognition, DNA repair and epigenetic modifications, stress response, immune response, aging and senescence, and many others. However, the links between the biological effects and underlying molecular processes are often poorly understood. Moreover, NAD has recently been found to tag the 5′-ends of some cellular RNAs, but the function of these NAD-capped RNAs remains largely unrevealed. Synthetic NAD analogs are invaluable molecular tools to detect, monitor, structurally investigate, and modulate activity of NAD-related enzymes and biological processes in order to aid their deeper understanding. Here, we review the recent advances in the design and development of NAD analogs as probes for various cellular NAD-related enzymes, enzymatic inhibitors with anticancer or antimicrobial therapeutic potential, and other NAD-related chemical biology tools. We focus on research papers published within the last 10 years.
Collapse
|
25
|
Zhu Y, Liu J, Park J, Rai P, Zhai RG. Subcellular compartmentalization of NAD + and its role in cancer: A sereNADe of metabolic melodies. Pharmacol Ther 2019; 200:27-41. [PMID: 30974124 PMCID: PMC7010080 DOI: 10.1016/j.pharmthera.2019.04.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/02/2019] [Indexed: 02/07/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential biomolecule involved in many critical processes. Its role as both a driver of energy production and a signaling molecule underscores its importance in health and disease. NAD+ signaling impacts multiple processes that are dysregulated in cancer, including DNA repair, cell proliferation, differentiation, redox regulation, and oxidative stress. Distribution of NAD+ is highly compartmentalized, with each subcellular NAD+ pool differentially regulated and preferentially involved in distinct NAD+-dependent signaling or metabolic events. Emerging evidence suggests that targeting NAD+ metabolism is likely to repress many specific mechanisms underlying tumor development and progression, including proliferation, survival, metabolic adaptations, invasive capabilities, heterotypic interactions with the tumor microenvironment, and stress response including notably DNA maintenance and repair. Here we provide a comprehensive overview of how compartmentalized NAD+ metabolism in mitochondria, nucleus, cytosol, and extracellular space impacts cancer formation and progression, along with a discussion of the therapeutic potential of NAD+-targeting drugs in cancer.
Collapse
Affiliation(s)
- Yi Zhu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, China; Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Jiaqi Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, China
| | - Joun Park
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Priyamvada Rai
- Department of Medicine/Medical Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Rong G Zhai
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, China.
| |
Collapse
|
26
|
Zaman S, Yu X, Bencivenga AF, Blanden AR, Liu Y, Withers T, Na B, Blayney AJ, Gilleran J, Boothman DA, Loh SN, Kimball SD, Carpizo DR. Combinatorial Therapy of Zinc Metallochaperones with Mutant p53 Reactivation and Diminished Copper Binding. Mol Cancer Ther 2019; 18:1355-1365. [PMID: 31196889 DOI: 10.1158/1535-7163.mct-18-1080] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 04/08/2019] [Accepted: 06/03/2019] [Indexed: 12/19/2022]
Abstract
Chemotherapy and radiation are more effective in wild-type (WT) p53 tumors due to p53 activation. This is one rationale for developing drugs that reactivate mutant p53 to synergize with chemotherapy and radiation. Zinc metallochaperones (ZMC) are a new class of mutant p53 reactivators that restore WT structure and function to zinc-deficient p53 mutants. We hypothesized that the thiosemicarbazone, ZMC1, would synergize with chemotherapy and radiation. Surprisingly, this was not found. We explored the mechanism of this and found the reactive oxygen species (ROS) activity of ZMC1 negates the signal on p53 that is generated with chemotherapy and radiation. We hypothesized that a zinc scaffold generating less ROS would synergize with chemotherapy and radiation. The ROS effect of ZMC1 is generated by its chelation of redox active copper. ZMC1 copper binding (K Cu) studies reveal its affinity for copper is approximately 108 greater than Zn2+ We identified an alternative zinc scaffold (nitrilotriacetic acid) and synthesized derivatives to improve cell permeability. These compounds bind zinc in the same range as ZMC1 but bound copper much less avidly (106- to 107-fold lower) and induced less ROS. These compounds were synergistic with chemotherapy and radiation by inducing p53 signaling events on mutant p53. We explored other combinations with ZMC1 based on its mechanism of action and demonstrate that ZMC1 is synergistic with MDM2 antagonists, BCL2 antagonists, and molecules that deplete cellular reducing agents. We have identified an optimal Cu2+:Zn2+ binding ratio to facilitate development of ZMCs as chemotherapy and radiation sensitizers. Although ZMC1 is not synergistic with chemotherapy and radiation, it is synergistic with a number of other targeted agents.
Collapse
Affiliation(s)
- Saif Zaman
- Department of Molecular Biology, Rutgers University, Piscataway, New Jersey
| | - Xin Yu
- Program of Surgical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, New Jersey.,Department of Surgery, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Anthony F Bencivenga
- Department of Medicinal Chemistry, Rutgers Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey
| | - Adam R Blanden
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York
| | - Yue Liu
- Program of Surgical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, New Jersey.,Department of Surgery, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Tracy Withers
- Program of Surgical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, New Jersey.,Department of Surgery, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Bing Na
- Program of Surgical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, New Jersey.,Department of Surgery, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Alan J Blayney
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York
| | - John Gilleran
- Department of Medicinal Chemistry, Rutgers Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey
| | - David A Boothman
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas.,Departments of Pharmacology and Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Stewart N Loh
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York
| | - S David Kimball
- Department of Medicinal Chemistry, Rutgers Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey.,Rutgers Translational Sciences, Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, New Jersey.,Z53 Therapeutics, Inc., Holmdel, New Jersey
| | - Darren R Carpizo
- Program of Surgical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, New Jersey. .,Department of Surgery, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey.,Z53 Therapeutics, Inc., Holmdel, New Jersey
| |
Collapse
|
27
|
Mehrzadi S, Mehrabani M, Malayeri AR, Bakhshayesh M, Kalantari H, Goudarzi M. Ellagic acid as a potential antioxidant, alleviates methotrexate-induced hepatotoxicity in male rats. Acta Chir Belg 2019; 119:69-77. [PMID: 29587597 DOI: 10.1080/00015458.2018.1455419] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Hepatotoxicity is one of the most life-threatening side-effects of Methotrexate therapy. Former studies highlighted the significance of oxidative stress in promoting Methotrexate-induced hepatotoxicity (MIH). Hence, the current study investigated the protective effect of Ellagic acid (EA), a poly-phenolic antioxidant, against MIH. METHODS Twenty-eight male Wistar rats were grouped into four sets: group 1 (control), group 2 (injected intraperitoneally with 20 mg/kg of Methotrexate on the 9th day), group 3 (treated orally with 10 mg/kg/day of EA for 10 days and injected with Methotrexate on the 9th day) and group 4 (treated with EA for 10 days). Subsequently, biochemical and histopathological parameters were evaluated in serum samples and liver tissues. RESULTS Methotrexate significantly increased activities of aminotransferases and ALP enzymes as well as levels of oxidative stress parameters in liver tissue. Likewise, Methotrexate decreased hepatic reduced glutathione level and activities of antioxidant enzymes. EA pre-treatment markedly attenuated the activities of aminotransferases and ALP, levels of oxidative stress parameters and augmented activities of antioxidant enzymes. Similarly, the remarkable protective effect of EA on liver has been confirmed by histological examination. CONCLUSION In sum, the current study supports the hypothesis that EA may be used as a promising pre-therapy to prevent the MIH.
Collapse
Affiliation(s)
- Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mehrnaz Mehrabani
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Reza Malayeri
- Medicinal Plant Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masoumeh Bakhshayesh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Heibatullah Kalantari
- Medicinal Plant Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mehdi Goudarzi
- Medicinal Plant Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
28
|
Li BB, Wang X, Tai L, Ma TT, Shalmani A, Liu WT, Li WQ, Chen KM. NAD Kinases: Metabolic Targets Controlling Redox Co-enzymes and Reducing Power Partitioning in Plant Stress and Development. FRONTIERS IN PLANT SCIENCE 2018; 9:379. [PMID: 29662499 PMCID: PMC5890153 DOI: 10.3389/fpls.2018.00379] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 03/07/2018] [Indexed: 05/03/2023]
Abstract
NAD(H) and NADP(H) are essential co-enzymes which dominantly control a number of fundamental biological processes by acting as reducing power and maintaining the intracellular redox balance of all life kingdoms. As the only enzymes that catalyze NAD(H) and ATP to synthesize NADP(H), NAD Kinases (NADKs) participate in many essential metabolic reactions, redox sensitive regulation, photosynthetic performance and also reactive oxygen species (ROS) homeostasis of cells and therefore, play crucial roles in both development and stress responses of plants. NADKs are highly conserved enzymes in amino acid sequences but have multiple subcellular localization and diverse functions. They may function as monomers, dimers or multimers in cells but the enzymatic properties in plants are not well elucidated yet. The activity of plant NADK is regulated by calcium/calmodulin and plays crucial roles in photosynthesis and redox co-enzyme control. NADK genes are expressed in almost all tissues and developmental stages of plants with specificity for different members. Their transcripts can be greatly stimulated by a number of environmental factors such as pathogenic attack, irritant applications and abiotic stress treatments. Using transgenic approaches, several studies have shown that NADKs are involved in chlorophyll synthesis, photosynthetic efficiency, oxidative stress protection, hormone metabolism and signaling regulation, and therefore contribute to the growth regulation and stress tolerance of plants. In this review, the enzymatic properties and functional mechanisms of plant NADKs are thoroughly investigated based on literature and databases. The results obtained here are greatly advantageous for further exploration of NADK function in plants.
Collapse
|
29
|
Zhang K, Kim H, Fu Z, Qiu Y, Yang Z, Wang J, Zhang D, Tong X, Yin L, Li J, Wu J, Qi NR, Houten SM, Zhang R. Deficiency of the Mitochondrial NAD Kinase Causes Stress-Induced Hepatic Steatosis in Mice. Gastroenterology 2018; 154:224-237. [PMID: 28923496 PMCID: PMC5742027 DOI: 10.1053/j.gastro.2017.09.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 08/31/2017] [Accepted: 09/10/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS The mitochondrial nicotinamide adenine dinucleotide (NAD) kinase (NADK2, also called MNADK) catalyzes phosphorylation of NAD to yield NADP. Little is known about the functions of mitochondrial NADP and MNADK in liver physiology and pathology. We investigated the effects of reduced mitochondrial NADP by deleting MNADK in mice. METHODS We generated MNADK knockout (KO) mice on a C57BL/6NTac background; mice with a wild-type Mnadk gene were used as controls. Some mice were placed on an atherogenic high-fat diet (16% fat, 41% carbohydrate, and 1.25% cholesterol supplemented with 0.5% sodium cholate) or given methotrexate intraperitoneally. We measured rates of fatty acid oxidation in primary hepatocytes using radiolabeled palmitate and in mice using indirect calorimetry. We measured levels of reactive oxygen species in mouse livers and primary hepatocytes. Metabolomic analyses were used to quantify serum metabolites, such as amino acids and acylcarnitines. RESULTS The KO mice had metabolic features of MNADK-deficient patients, such as increased serum concentrations of lysine and C10:2 carnitine. When placed on the atherogenic high-fat diet, the KO mice developed features of nonalcoholic fatty liver disease and had increased levels of reactive oxygen species in livers and primary hepatocytes, compared with control mice. During fasting, the KO mice had a defect in fatty acid oxidation. MNADK deficiency reduced the activation of cAMP-responsive element binding protein-hepatocyte specific and peroxisome proliferator-activated receptor alpha, which are transcriptional activators that mediate the fasting response. The activity of mitochondrial sirtuins was reduced in livers of the KO mice. Methotrexate inhibited the catalytic activity of MNADK in hepatocytes and in livers in mice with methotrexate injection. In mice given injections of methotrexate, supplementation of a diet with nicotinamide riboside, an NAD precursor, replenished hepatic NADP and protected the mice from hepatotoxicity, based on markers such as increased level of serum alanine aminotransferase. CONCLUSION MNADK facilitates fatty acid oxidation, counteracts oxidative damage, maintains mitochondrial sirtuin activity, and prevents metabolic stress-induced non-alcoholic fatty liver disease in mice.
Collapse
Affiliation(s)
- Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan; Department of Microbiology, Immunology and Biochemistry, Wayne State University School of Medicine, Detroit, Michigan.
| | - Hyunbae Kim
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Zhiyao Fu
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yining Qiu
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Zhao Yang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Jiemei Wang
- College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Deqiang Zhang
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Xin Tong
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Lei Yin
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jing Li
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Jianmei Wu
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Nathan R. Qi
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sander M. Houten
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ren Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan.
| |
Collapse
|
30
|
Fe-S Clusters Emerging as Targets of Therapeutic Drugs. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3647657. [PMID: 29445445 PMCID: PMC5763138 DOI: 10.1155/2017/3647657] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/27/2017] [Accepted: 12/06/2017] [Indexed: 01/11/2023]
Abstract
Fe-S centers exhibit strong electronic plasticity, which is of importance for insuring fine redox tuning of protein biological properties. In accordance, Fe-S clusters are also highly sensitive to oxidation and can be very easily altered in vivo by different drugs, either directly or indirectly due to catabolic by-products, such as nitric oxide species (NOS) or reactive oxygen species (ROS). In case of metal ions, Fe-S cluster alteration might be the result of metal liganding to the coordinating sulfur atoms, as suggested for copper. Several drugs presented through this review are either capable of direct interaction with Fe-S clusters or of secondary Fe-S clusters alteration following ROS or NOS production. Reactions leading to Fe-S cluster disruption are also reported. Due to the recent interest and progress in Fe-S biology, it is very likely that an increasing number of drugs already used in clinics will emerge as molecules interfering with Fe-S centers in the near future. Targeting Fe-S centers could also become a promising strategy for drug development.
Collapse
|
31
|
Yau EH, Kummetha IR, Lichinchi G, Tang R, Zhang Y, Rana TM. Genome-Wide CRISPR Screen for Essential Cell Growth Mediators in Mutant KRAS Colorectal Cancers. Cancer Res 2017; 77:6330-6339. [PMID: 28954733 DOI: 10.1158/0008-5472.can-17-2043] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 09/15/2017] [Accepted: 09/15/2017] [Indexed: 12/22/2022]
Abstract
Targeting mutant KRAS signaling pathways continues to attract attention as a therapeutic strategy for KRAS-driven tumors. In this study, we exploited the power of the CRISPR-Cas9 system to identify genes affecting the tumor xenograft growth of human mutant KRAS (KRASMUT) colorectal cancers. Using pooled lentiviral single-guide RNA libraries, we conducted a genome-wide loss-of-function genetic screen in an isogenic pair of human colorectal cancer cell lines harboring mutant or wild-type KRAS. The screen identified novel and established synthetic enhancers or synthetic lethals for KRASMUT colorectal cancer, including targetable metabolic genes. Notably, genetic disruption or pharmacologic inhibition of the metabolic enzymes NAD kinase or ketohexokinase was growth inhibitory in vivo In addition, the chromatin remodeling protein INO80C was identified as a novel tumor suppressor in KRASMUT colorectal and pancreatic tumor xenografts. Our findings define a novel targetable set of therapeutic targets for KRASMUT tumors. Cancer Res; 77(22); 6330-9. ©2017 AACR.
Collapse
Affiliation(s)
- Edwin H Yau
- Department of Pediatrics and Institute for Genomic Medicine, University of California San Diego School of Medicine, La Jolla, California.,Division of Hematology-Oncology, Department of Internal Medicine, University of California San Diego School of Medicine, La Jolla, California.,Solid Tumor Therapeutics Program, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Indrasena Reddy Kummetha
- Department of Pediatrics and Institute for Genomic Medicine, University of California San Diego School of Medicine, La Jolla, California
| | - Gianluigi Lichinchi
- Department of Pediatrics and Institute for Genomic Medicine, University of California San Diego School of Medicine, La Jolla, California
| | - Rachel Tang
- Department of Pediatrics and Institute for Genomic Medicine, University of California San Diego School of Medicine, La Jolla, California
| | - Yunlin Zhang
- Department of Pediatrics and Institute for Genomic Medicine, University of California San Diego School of Medicine, La Jolla, California
| | - Tariq M Rana
- Department of Pediatrics and Institute for Genomic Medicine, University of California San Diego School of Medicine, La Jolla, California. .,Solid Tumor Therapeutics Program, Moores Cancer Center, University of California, San Diego, La Jolla, California
| |
Collapse
|
32
|
Abstract
Reactive oxygen species (ROS) are important signaling molecules that act through the oxidation of nucleic acids, proteins, and lipids. Several hallmarks of cancer, including uncontrolled proliferation, angiogenesis, and genomic instability, are promoted by the increased ROS levels commonly found in tumor cells. To counteract excessive ROS accumulation, oxidative stress, and death, cancer cells tightly regulate ROS levels by enhancing scavenging enzymes, which are dependent on the reducing cofactor nicotinamide adenine dinucleotide phosphate (NADPH). This review focuses on mitochondrial ROS homeostasis with a description of six pathways of NADPH production in mitochondria and a discussion of the possible strategies of pharmacological intervention to selectively eliminate cancer cells by increasing their ROS levels.
Collapse
Affiliation(s)
- Francesco Ciccarese
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Vincenzo Ciminale
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy.,Veneto Institute of Oncology - IRCCS, Padua, Italy
| |
Collapse
|
33
|
Tedeschi PM, Bansal N, Kerrigan JE, Abali EE, Scotto KW, Bertino JR. NAD+ Kinase as a Therapeutic Target in Cancer. Clin Cancer Res 2016; 22:5189-5195. [PMID: 27582489 DOI: 10.1158/1078-0432.ccr-16-1129] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 06/20/2016] [Indexed: 11/16/2022]
Abstract
NAD+ kinase (NADK) catalyzes the phosphorylation of nicotinamide adenine dinucleotide (NAD+) to nicotinamide adenine dinucleotide phosphate (NADP+) using ATP as the phosphate donor. NADP+ is then reduced to NADPH by dehydrogenases, in particular glucose-6-phosphate dehydrogenase and the malic enzymes. NADPH functions as an important cofactor in a variety of metabolic and biosynthetic pathways. The demand for NADPH is particularly high in proliferating cancer cells, where it acts as a cofactor for the synthesis of nucleotides, proteins, and fatty acids. Moreover, NADPH is essential for the neutralization of the dangerously high levels of reactive oxygen species (ROS) generated by increased metabolic activity. Given its key role in metabolism and regulation of ROS, it is not surprising that several recent studies, including in vitro and in vivo assays of tumor growth and querying of patient samples, have identified NADK as a potential therapeutic target for the treatment of cancer. In this review, we will discuss the experimental evidence justifying further exploration of NADK as a clinically relevant drug target and describe our studies with a lead compound, thionicotinamide, an NADK inhibitor prodrug. Clin Cancer Res; 22(21); 5189-95. ©2016 AACR.
Collapse
Affiliation(s)
- Philip M Tedeschi
- Departments of Medicine and Pharmacology, Rutgers Robert Wood Johnson Medical School, the Rutgers Cancer Institute of New Jersey, and the Rutgers Graduate School of Biomedical Sciences, New Brunswick, New Jersey
| | - Nitu Bansal
- Departments of Medicine and Pharmacology, Rutgers Robert Wood Johnson Medical School, the Rutgers Cancer Institute of New Jersey, and the Rutgers Graduate School of Biomedical Sciences, New Brunswick, New Jersey
| | - John E Kerrigan
- Departments of Medicine and Pharmacology, Rutgers Robert Wood Johnson Medical School, the Rutgers Cancer Institute of New Jersey, and the Rutgers Graduate School of Biomedical Sciences, New Brunswick, New Jersey
| | - Emine E Abali
- Departments of Medicine and Pharmacology, Rutgers Robert Wood Johnson Medical School, the Rutgers Cancer Institute of New Jersey, and the Rutgers Graduate School of Biomedical Sciences, New Brunswick, New Jersey
| | - Kathleen W Scotto
- Departments of Medicine and Pharmacology, Rutgers Robert Wood Johnson Medical School, the Rutgers Cancer Institute of New Jersey, and the Rutgers Graduate School of Biomedical Sciences, New Brunswick, New Jersey.
| | - Joseph R Bertino
- Departments of Medicine and Pharmacology, Rutgers Robert Wood Johnson Medical School, the Rutgers Cancer Institute of New Jersey, and the Rutgers Graduate School of Biomedical Sciences, New Brunswick, New Jersey.
| |
Collapse
|