1
|
Ramasamy S, Jeyaram K, Narayanan A, Arunachalam S, Ethiraj S, Sankar M, Pandian B. Repurposing fluvoxamine as an inhibitor for NUDT5 in breast cancer cell: an in silico and in vitro study. In Silico Pharmacol 2024; 13:5. [PMID: 39726906 PMCID: PMC11668718 DOI: 10.1007/s40203-024-00293-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
Drug repurposing is necessary to accelerate drug discovery and meet the drug needs. This study investigated the possibility of using fluvoxamine to inhibit the cellular metabolizing enzyme NUDT5 in breast cancer. Computational and experimental techniques were used to evaluate the structural flexibility, binding stability, and chemical reactivity of the drugs. These findings indicated that fluvoxamine effectively suppressed the activity of NUDT5, as evidenced by a binding score of - 8.514 kcal/mol. Furthermore, the binding positions of fluvoxamine and NUDT5 were optimized. Fluvoxamine attachment to the active sites of Trp28, Trp46, Glu47, Arg51, Arg84, and Leu98 in NUDT5 has been shown to alter the metabolism of ADPr. These alterations play a role in ATP production in the breast cancer cells. In addition, an MTT assay conducted on the MCF-7 cell line using fluvoxamine revealed an IC50 value of 53.86 ± 0.05 µM. Fluvoxamine-induced apoptosis was confirmed as evidenced by AO/EtBr and DAPI staining. Graphical abstract Effect of fluvoxamine on breast cancer cells.
Collapse
Affiliation(s)
- Sumathi Ramasamy
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, 626126 Tamil Nadu India
| | - Kanimozhi Jeyaram
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, 626126 Tamil Nadu India
| | - Aathimoolam Narayanan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, 626126 Tamil Nadu India
| | | | - Selvarajan Ethiraj
- Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603203 Tamil Nadu India
| | - Muthumanickam Sankar
- Department of Bioinformatics, Alagappa University, Karaikudi, 630003 Tamil Nadu India
| | - Boomi Pandian
- Department of Bioinformatics, Alagappa University, Karaikudi, 630003 Tamil Nadu India
| |
Collapse
|
2
|
Dhanasekaran S, Selvadoss PP, Manoharan SS, Jeyabalan S, Muthu Laxmi V, Choudhury AA, Rajeswari VD, Ramanathan G, Thamaraikani T, Subramaniyan V, Sekar M, Shing WL. Targeting Nudix Hydrolase 5 with Bioactive Flavonoids: Molecular Dynamics and Docking Studies for Breast Cancer Therapy. Cell Biochem Biophys 2024:10.1007/s12013-024-01609-x. [PMID: 39638981 DOI: 10.1007/s12013-024-01609-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2024] [Indexed: 12/07/2024]
Abstract
Breast cancer (BC) is the most prevalent malignancy among women globally and the leading cause of cancer-related mortality. Consequently, there is an urgent need for new, effective treatment strategies for breast cancer. Research has shown that the enzyme nudix hydrolase 5 (NUDT5) plays a critical role in promoting breast cancer aggressiveness and serves as a key regulator of oncogenic pathways. The development of NUDT5 inhibitors presents a viable strategy for enhancing treatment results in managing BC. The ability of the flavonoids to modulate key biochemical pathways and improve therapeutic outcomes highlights their promise in developing novel breast cancer treatments. Hence, the main objective of the present investigation is to identify the potential interaction of structurally diverse bioactive flavonoids with the active site of the target NUDT5. Our docking analysis revealed that the flavonoids such as naringin and genistein have shown a significant binding association with residues Arg51, Asp60, Gln82, Arg84, Ala96, Leu98, Glu112, Glu116, Met132, Cys139, Ile141, and Glu166 of NUDT5, suggesting its potential as a potent inhibitor. The stabilizing effects of these leads (naringin and genistein) were further validated using molecular dynamics investigations, including RMSD, RMRF, Rg, SASA, PCA, and FEL. The results of the MD simulation studies evidenced a more significant interaction between genistein and NUDT5, indicating a steady and robust affinity, making genistein a more promising inhibitor. In conclusion, the flavonoid genistein has a strong potential as a therapeutic agent for targeting NUDT5 in breast cancer treatment making it viable candidates for further preclinical and clinical investigations.
Collapse
Affiliation(s)
- Sivaraman Dhanasekaran
- Department of Biotechnology, School of Energy Technology, Pandit Deendayal Energy University, Knowledge Corridor, Gandhinagar, Gujarat, India.
| | - Pradeep Pushparaj Selvadoss
- Department of Biotechnology, School of Energy Technology, Pandit Deendayal Energy University, Knowledge Corridor, Gandhinagar, Gujarat, India
| | - Solomon Sundar Manoharan
- Department of Biotechnology, School of Energy Technology, Pandit Deendayal Energy University, Knowledge Corridor, Gandhinagar, Gujarat, India
| | - Srikanth Jeyabalan
- Department of Pharmacology, Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - V Muthu Laxmi
- Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | | | | | | | - Tamilanban Thamaraikani
- Department of Pharmacology, Faculty of Medicine, MAHSA University, Jenjarom, Selangor, Malaysia
| | - Vetriselvan Subramaniyan
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Jalan University, Petaling Jaya, Malaysia
| | | | - Wong Ling Shing
- INTI International University, Nilai, Negeri Sembilan, Malaysia
| |
Collapse
|
3
|
He L, Meng F, Chen R, Qin J, Sun M, Fan Z, Du J. Precise Regulations at the Subcellular Level through Intracellular Polymerization, Assembly, and Transformation. JACS AU 2024; 4:4162-4186. [PMID: 39610726 PMCID: PMC11600172 DOI: 10.1021/jacsau.4c00849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/30/2024]
Abstract
A living cell is an intricate machine that creates subregions to operate cell functions effectively. Subcellular dysfunction has been identified as a potential druggable target for successful drug design and therapy. The treatments based on intracellular polymerization, self-assembly, or transformation offer various advantages, including enhanced blood circulation of monomers, long-term drug delivery pharmacokinetics, low drug resistance, and the ability to target deep tissues and organelles. In this review, we discuss the latest developments of intracellular synthesis applied to precisely control cellular functions. First, we discuss the design and applications of endogenous and exogenous stimuli-triggered intracellular polymerization, self-assembly, and dynamic morphology transformation of biomolecules at the subcellular level. Second, we highlight the benefits of these strategies applied in cancer diagnosis and treatment and modulating cellular states or cell metabolism of living systems. Finally, we conclude the recent progress in this field, discuss future perspectives, analyze the challenges of the intracellular functional reactions for regulation, and find future opportunities.
Collapse
Affiliation(s)
- Le He
- School
of Materials Science and Engineering, East
China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
- Department
of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology
and Brain Functional Modulation, Clinical Research Center for Anesthesiology
and Perioperative Medicine, Translational Research Institute of Brain
and Brain-Like Intelligence, Shanghai Fourth People’s Hospital,
School of Medicine, Tongji University, Shanghai 200434, China
| | - Fanying Meng
- Department
of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Ran Chen
- Department
of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Jinlong Qin
- Department
of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology
and Brain Functional Modulation, Clinical Research Center for Anesthesiology
and Perioperative Medicine, Translational Research Institute of Brain
and Brain-Like Intelligence, Shanghai Fourth People’s Hospital,
School of Medicine, Tongji University, Shanghai 200434, China
| | - Min Sun
- Department
of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology
and Brain Functional Modulation, Clinical Research Center for Anesthesiology
and Perioperative Medicine, Translational Research Institute of Brain
and Brain-Like Intelligence, Shanghai Fourth People’s Hospital,
School of Medicine, Tongji University, Shanghai 200434, China
| | - Zhen Fan
- Department
of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology
and Brain Functional Modulation, Clinical Research Center for Anesthesiology
and Perioperative Medicine, Translational Research Institute of Brain
and Brain-Like Intelligence, Shanghai Fourth People’s Hospital,
School of Medicine, Tongji University, Shanghai 200434, China
- Department
of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Jianzhong Du
- School
of Materials Science and Engineering, East
China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
- Department
of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology
and Brain Functional Modulation, Clinical Research Center for Anesthesiology
and Perioperative Medicine, Translational Research Institute of Brain
and Brain-Like Intelligence, Shanghai Fourth People’s Hospital,
School of Medicine, Tongji University, Shanghai 200434, China
- Department
of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| |
Collapse
|
4
|
Yi Q, Zhu G, Zhu W, Wang J, Ouyang X, Yang K, Fan Y, Zhong J. LINC01094: A key long non-coding RNA in the regulation of cancer progression and therapeutic targets. Heliyon 2024; 10:e37527. [PMID: 39309878 PMCID: PMC11415682 DOI: 10.1016/j.heliyon.2024.e37527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/25/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
LINC01094 is a long non-coding RNA that plays a crucial role in cancer progression by modulating key signaling pathways, such as PI3K/AKT, Wnt/β-catenin and TGF-β Signaling Pathway Feedback Loop. In this review we summarize the recent research on the functional mechanisms of LINC01094 in various cancers, including its impact on tumor growth, metastasis, and resistance to therapy. We also discuss the therapeutic potential of targeting LINC01094 and highlight the current strategies and challenges in this area. Perspectives on future development of LINC01094-based therapies are also provided.
Collapse
Affiliation(s)
- Qiang Yi
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Gangfeng Zhu
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Weijian Zhu
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Jiaqi Wang
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Xinting Ouyang
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Kuan Yang
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Yu Fan
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Jinghua Zhong
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| |
Collapse
|
5
|
Dubey A, Alanazi AM, Bhardwaj R, Ragusa A. Identification of potential NUDT5 inhibitors from marine bacterial natural compounds via molecular dynamics and free energy landscape analysis. Mol Divers 2024:10.1007/s11030-024-10950-5. [PMID: 39225905 DOI: 10.1007/s11030-024-10950-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024]
Abstract
NUDIX hydrolase 5 (NUDT5) is an enzyme involved in the hydrolysis of nucleoside diphosphates linked to other moieties, such as ADP-ribose. This cofactor is vital in redox reactions and is essential for the activity of sirtuins and poly(ADP-ribose) polymerases, which are involved in DNA repair and genomic stability. It has been shown that NUDT5 activity can also influence NAD+ homeostasis, thereby affecting cancer cell metabolism and survival. In this regard, the discovery of NUDT5 inhibitors has emerged as a potential therapeutic approach in cancer treatment. In this study, we conducted a high-throughput virtual screening of marine bacterial compounds against the NUDT5 enzyme and four molecules were selected based on their docking scores. These compounds established strong interactions within the NUDT5 active site, with molecular analysis highlighting the key role of Trp28A and Trp46B residues. Molecular dynamics simulations over 200 ns indicated a stable behavior, in association with root mean square deviation values always below 3 Å, suggesting conformational stability. Free energy landscape analysis further supported their potential as NUDT5 inhibitors, offering avenues for novel therapeutic strategies against NUDT5-associated breast cancer.
Collapse
Affiliation(s)
- Amit Dubey
- Department of Pharmacology, Saveetha Dental College, and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nādu, 600077, India
- Department of Computational Chemistry and Drug Discovery Division, Quanta Calculus, Greater Noida, 201310, India
| | - Amer M Alanazi
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Rima Bhardwaj
- Department of Chemistry, Poona College, Savitribai Phule Pune University, Pune, India.
| | - Andrea Ragusa
- Institute of Nanotechnology, CNR-Nanotec, Via Monteroni, 73100, Lecce, Italy.
- Department of Life Sciences, Health and Health Professions, Link Campus University, Via del Casale Di San Pio V 44, 00165, Rome, Italy.
| |
Collapse
|
6
|
Kumar RMR, Joghee S. Enhancing breast cancer treatment through pharmacogenomics: A narrative review. Clin Chim Acta 2024; 562:119893. [PMID: 39068964 DOI: 10.1016/j.cca.2024.119893] [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: 06/27/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024]
Abstract
Pharmacogenomics has become integral to personalised medicine in breast cancer, utilising genetic insights to customize treatment strategies and enhance patient outcomes. Understanding how genetic variations influence drug metabolism, response, and toxicity is crucial for guiding treatment selection and dosing regimens. Genetic polymorphisms in drug-metabolizing enzymes and transporters significantly impact pharmacokinetic variability, influencing the efficacy and safety of chemotherapy agents and targeted therapies. Biomarkers associated with the hormone receptor status of breast cancer and mutations serve as key determinants of treatment response, aiding in the selection of therapies. Despite substantial progress in understanding the pharmacogenomic landscape of breast cancer, efforts to identify novel genetic markers and refine treatment optimisation strategies are required. Genome-wide association studies and advanced sequencing technologies hold promise for uncovering genetic determinants of drug response variability and elucidating complex pharmacogenomic interactions. The future of pharmacogenomics in breast cancer lies in real-time treatment monitoring, the discovery of additional predictive markers, and the seamless integration of pharmacogenomic data into clinical decision-making processes. However, translating pharmacogenomic discoveries into routine clinical practice requires collaborative efforts among stakeholders to address implementation challenges and ensure equitable access to genetic testing. By embracing pharmacogenomics, clinicians can tailor treatment approaches to individual patients, maximizing therapeutic benefits while minimizing adverse effects. This review discusses the integration of pharmacogenomics in breast cancer treatment, highlighting the significance of understanding genetic influences on treatment response and toxicity, and the potential of advanced technologies in refining treatment strategies.
Collapse
Affiliation(s)
- Ram Mohan Ram Kumar
- Department of Pharmaceutical Biotechnology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, Karnataka, India.
| | - Suresh Joghee
- Department of Pharmacognosy, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, Karnataka, India
| |
Collapse
|
7
|
Woo SW, Kim M, Kang D, Choe YH, Oh SJ, You AS, Lee SL, Kim J. Genome-Wide Characterization of Somatic Mutation Patterns in Cloned Dogs Reveals Implications for Neuronal Function, Tumorigenesis, and Aging. Genes (Basel) 2024; 15:801. [PMID: 38927737 PMCID: PMC11202621 DOI: 10.3390/genes15060801] [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: 04/25/2024] [Revised: 05/27/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
Studies on somatic mutations in cloned animals have revealed slight genetic variances between clones and their originals, but have yet to identify the precise effects of these differences within the organism. Somatic mutations contribute to aging and are implicated in tumor development and other age-related diseases. Thus, we compared whole genome sequencing data from an original dog with that of cloned dogs, identifying candidate somatic mutations that were disproportionately located within genes previously implicated in aging. The substitutional signature of cloning-specific somatic mutations mirrored the uniform distribution characteristic of the signature associated with human aging. Further analysis of genes revealed significant enrichment of traits associated with body size as well as the molecular mechanisms underlying neuronal function and tumorigenesis. Overall, the somatic mutations found in cloned dogs may indicate a conserved mechanism driving aging across species and a broad spectrum of pathway alterations.
Collapse
Affiliation(s)
- Seung-Wan Woo
- Division of Applied Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea; (S.-W.W.); (D.K.)
- Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea;
| | - Miju Kim
- Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea;
| | - Dayeon Kang
- Division of Applied Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea; (S.-W.W.); (D.K.)
- Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea;
| | - Yong-ho Choe
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (Y.-h.C.); (S.-J.O.); (S.-L.L.)
| | - Seong-Ju Oh
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (Y.-h.C.); (S.-J.O.); (S.-L.L.)
| | - Are-Sun You
- Division of Animal Diseases & Health, National Institute of Animal Science, RDA, Wanju 55365, Republic of Korea;
| | - Sung-Lim Lee
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (Y.-h.C.); (S.-J.O.); (S.-L.L.)
| | - Jaemin Kim
- Division of Applied Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea; (S.-W.W.); (D.K.)
- Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea;
| |
Collapse
|
8
|
Preethi V, Vijukumar V, AnilaRaj S, Vidya V. Synthesis, characterization, DFT studies and evaluation of the potential anti-tumour activity of nicotinic hydrazide based Schiff base using in vitro and molecular docking techniques. Heliyon 2024; 10:e29689. [PMID: 38720735 PMCID: PMC11076660 DOI: 10.1016/j.heliyon.2024.e29689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 03/29/2024] [Accepted: 04/12/2024] [Indexed: 05/12/2024] Open
Abstract
Breast cancer, one of the most serious issues worldwide, has been raising day by day. It has now become a necessary to develop a suitable drug to combat this terrible illness. Schiff bases are increasingly being used as powerful medications for a number of illnesses. BNH has now synthesized from Benzil and Nicotinic hydrazide and characterized experimentally by FT-IR, UV, 1H NMR,13CNMR and Mass analysis. DFT calculations were done using Gaussian 16 W with B3LYP/6-311 + G (d,p) and geometry of the compound is optimized. Frontier Molecular orbit (FMO), Mullikan atomic charges and Molecular Electrostatic Potential (MEP) were studied. Invitro antimicrobial studies were done using various bacteria and fungi. The synthesized compound is appropriate against bacterial and fungal actions. Invitro study were done using MCF-7 cell lines to analyze the anticancer property of the ligand. The outcome suggests that BNH may be employed in the future as a novel anticancer medication.
Collapse
Affiliation(s)
- V. Preethi
- Department of Chemistry, University College, Paalayam, Thiruvananthapuram, 695034, Kerala, India
| | - V.G. Vijukumar
- Department of Chemistry, University College, Paalayam, Thiruvananthapuram, 695034, Kerala, India
| | - S. AnilaRaj
- Department of Chemistry, University College, Paalayam, Thiruvananthapuram, 695034, Kerala, India
| | - V.G. Vidya
- Department of Chemistry, University College, Paalayam, Thiruvananthapuram, 695034, Kerala, India
| |
Collapse
|
9
|
Han J, Liang J, Zhou W, Zhang M, Jin T. Association between NUDT17 polymorphisms and breast cancer risk. Expert Rev Mol Diagn 2024; 24:459-466. [PMID: 38756100 DOI: 10.1080/14737159.2024.2353700] [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: 10/19/2023] [Accepted: 03/18/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND Breast cancer (BC) is the leading cause of cancer death among women worldwide. The nudix hydrolase 17 (NUDT17) may play notable roles in cancer growth and metastasis. In this study, we explored the importance of NUDT17 gene polymorphism in patients with BC. METHODS In our study, 563 BC patients and 552 healthy controls participated. We used logistic regression analysis to calculate odds ratios (OR) and 95% confidence intervals (CI), and multifactor dimension reduction (MDR) analysis of SNP-SNP interactions. Finally, UALCAN and THPA databases were used for bioinformatics analysis. RESULTS The rs9286836 G allele was associated with a decreased the BC risk (p = 0.022), and the carriers of rs2004659 G allele had a 32% decreased risk of BC than individuals with allele A (p = 0.004). In the four genetic models, rs9286836 and rs2004659 reduced the risk of BC. Additionally, we found that the NUDT17 SNPs were associated with BC risk under age, tumor size, and clinical stage stratification. The MDR analysis showed that the five-locus interaction model was the best in the multi-locus model. CONCLUSION Our study found that NUDT17 single nucleotide polymorphisms are associated with BC susceptibility in Chinese Han population.
Collapse
Affiliation(s)
- Junhui Han
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, Shaanxi, China
- College of Life Science, Northwest University, Xi'an, Shaanxi, China
- Shaanxi Provincial Key Laboratory of Biotechnology, Northwest University, Xi'an, Shaanxi, China
| | - Jing Liang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, Shaanxi, China
- College of Life Science, Northwest University, Xi'an, Shaanxi, China
- Shaanxi Provincial Key Laboratory of Biotechnology, Northwest University, Xi'an, Shaanxi, China
| | - Wenqian Zhou
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, Shaanxi, China
- College of Life Science, Northwest University, Xi'an, Shaanxi, China
- Shaanxi Provincial Key Laboratory of Biotechnology, Northwest University, Xi'an, Shaanxi, China
| | - Man Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, Shaanxi, China
- College of Life Science, Northwest University, Xi'an, Shaanxi, China
- Shaanxi Provincial Key Laboratory of Biotechnology, Northwest University, Xi'an, Shaanxi, China
| | - Tianbo Jin
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, Shaanxi, China
- College of Life Science, Northwest University, Xi'an, Shaanxi, China
- Shaanxi Provincial Key Laboratory of Biotechnology, Northwest University, Xi'an, Shaanxi, China
| |
Collapse
|
10
|
Taiyab A, Choudhury A, Haidar S, Yousuf M, Rathi A, Koul P, Chakrabarty A, Islam A, Shamsi A, Hassan MI. Exploring MTH1 inhibitory potential of Thymoquinone and Baicalin for therapeutic targeting of breast cancer. Biomed Pharmacother 2024; 173:116332. [PMID: 38430630 DOI: 10.1016/j.biopha.2024.116332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/05/2024] Open
Abstract
Cancers frequently have increased ROS levels due to disrupted redox balance, leading to oxidative DNA and protein damage, mutations, and apoptosis. The MTH1 protein plays a crucial role by sanitizing the oxidized dNTP pools. Hence, cancer cells rely on MTH1 to prevent the integration of oxidized dNTPs into DNA, preventing DNA damage and allowing cancer cell proliferation. We have discovered Thymoquinone (TQ) and Baicalin (BC) as inhibitors of MTH1 using combined docking and MD simulation approaches complemented by experimental validations via assessing binding affinity and enzyme inhibition. Docking and MD simulations studies revealed an efficient binding of TQ and BC to the active site pocket of the MTH1, and the resultant complexes are appreciably stable. Fluorescence measurements estimated a strong binding affinity of TQ and BC with Ka 3.4 ×106 and 1.0 ×105, respectively. Treating breast cancer cells with TQ and BC significantly inhibited the growth and proliferation (IC50 values 28.3 µM and 34.8 µM) and induced apoptosis. TQ and BC increased the ROS production in MCF7 cells, imposing substantial oxidative stress on cancer cells and leading to cell death. Finally, TQ and BC are proven strong MTH1 inhibitors, offering promising prospects for anti-cancer therapy.
Collapse
Affiliation(s)
- Aaliya Taiyab
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Arunabh Choudhury
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Shaista Haidar
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University, NH91, Tehsil Dadri, Gautam Buddha Nagar, Uttar Pradesh 201314, India
| | - Mohd Yousuf
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Aanchal Rathi
- Department of Bioscience, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Priyanka Koul
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Anindita Chakrabarty
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University, NH91, Tehsil Dadri, Gautam Buddha Nagar, Uttar Pradesh 201314, India
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Anas Shamsi
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 364, United Arab Emirates.
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India.
| |
Collapse
|
11
|
Qian J, Ma Y, Tahaney WM, Moyer CL, Lanier A, Hill J, Coleman D, Koupaei N, Hilsenbeck SG, Savage MI, Page BDG, Mazumdar A, Brown PH. The novel phosphatase NUDT5 is a critical regulator of triple-negative breast cancer growth. Breast Cancer Res 2024; 26:23. [PMID: 38317231 PMCID: PMC10845800 DOI: 10.1186/s13058-024-01778-w] [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: 02/28/2023] [Accepted: 01/22/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND The most aggressive form of breast cancer is triple-negative breast cancer (TNBC), which lacks expression of the estrogen receptor (ER) and progesterone receptor (PR), and does not have overexpression of the human epidermal growth factor receptor 2 (HER2). Treatment options for women with TNBC tumors are limited, unlike those with ER-positive tumors that can be treated with hormone therapy, or those with HER2-positive tumors that can be treated with anti-HER2 therapy. Therefore, we have sought to identify novel targeted therapies for TNBC. In this study, we investigated the potential of a novel phosphatase, NUDT5, as a potential therapeutic target for TNBC. METHODS The mRNA expression levels of NUDT5 in breast cancers were investigated using TCGA and METABRIC (Curtis) datasets. NUDT5 ablation was achieved through siRNA targeting and NUDT5 inhibition with the small molecule inhibitor TH5427. Xenograft TNBC animal models were employed to assess the effect of NUDT5 inhibition on in vivo tumor growth. Proliferation, death, and DNA replication assays were conducted to investigate the cellular biological effects of NUDT5 loss or inhibition. The accumulation of 8-oxo-guanine (8-oxoG) and the induction of γH2AX after NUDT5 loss was determined by immunofluorescence staining. The impact of NUDT5 loss on replication fork was assessed by measuring DNA fiber length. RESULTS In this study, we demonstrated the significant role of an overexpressed phosphatase, NUDT5, in regulating oxidative DNA damage in TNBCs. Our findings indicate that loss of NUDT5 results in suppressed growth of TNBC both in vitro and in vivo. This growth inhibition is not attributed to cell death, but rather to the suppression of proliferation. The loss or inhibition of NUDT5 led to an increase in the oxidative DNA lesion 8-oxoG, and triggered the DNA damage response in the nucleus. The interference with DNA replication ultimately inhibited proliferation. CONCLUSIONS NUDT5 plays a crucial role in preventing oxidative DNA damage in TNBC cells. The loss or inhibition of NUDT5 significantly suppresses the growth of TNBCs. These biological and mechanistic studies provide the groundwork for future research and the potential development of NUDT5 inhibitors as a promising therapeutic approach for TNBC patients.
Collapse
Affiliation(s)
- Jing Qian
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Yanxia Ma
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William M Tahaney
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Monte Rosa Therapeutics, Boston, USA
| | - Cassandra L Moyer
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amanda Lanier
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jamal Hill
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Darian Coleman
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Negar Koupaei
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Susan G Hilsenbeck
- Lester and Sue Smith Breast Center and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Michelle I Savage
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brent D G Page
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
| | - Abhijit Mazumdar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Powel H Brown
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
12
|
Raafat SN, El Wahed SA, Badawi NM, Saber MM, Abdollah MR. Enhancing the anticancer potential of metformin: fabrication of efficient nanospanlastics, in vitro cytotoxic studies on HEP-2 cells and reactome enhanced pathway analysis. Int J Pharm X 2023; 6:100215. [PMID: 38024451 PMCID: PMC10630776 DOI: 10.1016/j.ijpx.2023.100215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 10/17/2023] [Accepted: 10/21/2023] [Indexed: 12/01/2023] Open
Abstract
Metformin (MET), an oral antidiabetic drug, was reported to possess promising anticancer effects. We hypothesized that MET encapsulation in unique nanospanlastics would enhance its anticancer potential against HEP-2 cells. Our results showed the successful fabrication of Nano-MET spanlastics (d = 232.10 ± 0.20 nm; PDI = 0.25 ± 0.11; zeta potential = (-) 44.50 ± 0.96; drug content = 99.90 ± 0.11 and entrapment efficiency = 88.01 ± 2.50%). MTT assay revealed the enhanced Nano-MET cytotoxicity over MET with a calculated IC50 of 50 μg/mL and > 500 μg/mL, respectively. Annexin V/PI apoptosis assay showed that Nano-MET significantly decreased the percentage of live cells from 95.49 to 93.70 compared to MET and increased the percentage of cells arrested in the G0/G1 phase by 8.38%. Moreover, Nano-MET downregulated BCL-2 and upregulated BAX protein levels by 1.57 and 1.88 folds, respectively. RT-qPCR revealed that Nano-MET caused a significant 13.75, 4.15, and 2.23-fold increase in caspase-3, -8, and - 9 levels as well as a 100 and 43.47-fold decrease in cyclin D1 and mTOR levels, respectively. The proliferation marker Ki67 immunofluorescent staining revealed a 3-fold decrease in positive cells in Nano-MET compared to the control. Utilizing the combined Pathway-Enrichment Analysis (PEA) and Reactome analysis indicated high enrichment of certain pathways including nucleotides metabolism, Nudix-type hydrolase enzymes, carbon dioxide hydration, hemostasis, and the innate immune system. In summary, our results confirm MET cytotoxicity enhancement by its encapsulation in nanospanlastics. We also highlight, using PEA, that MET can modulate multiple pathways implicated in carcinogenesis.
Collapse
Affiliation(s)
- Shereen Nader Raafat
- Department of Pharmacology, Faculty of Dentistry, The British University in Egypt, Cairo, Egypt
- Stem Cells and Tissue Culture Hub (CIDS), Faculty of Dentistry, The British University in Egypt, Cairo, Egypt
| | - Sara Abd El Wahed
- Department of Oral Pathology, Faculty of Dentistry, The British University in Egypt, Cairo, Egypt
| | - Noha M. Badawi
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
- Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El Sherouk City, Egypt
| | - Mona M. Saber
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Giza, Egypt
| | - Maha R.A. Abdollah
- Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El Sherouk City, Egypt
- Department of Pharmacology, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| |
Collapse
|
13
|
Walter M, Mayr F, Hanna BMF, Cookson V, Mortusewicz O, Helleday T, Herr P. NUDT22 promotes cancer growth through pyrimidine salvage. Oncogene 2023; 42:1282-1293. [PMID: 36871087 PMCID: PMC10101856 DOI: 10.1038/s41388-023-02643-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 03/06/2023]
Abstract
The NUDIX hydrolase NUDT22 converts UDP-glucose into glucose-1-phosphate and the pyrimidine nucleotide uridine monophosphate but a biological significance for this biochemical reaction has not yet been established. Glucose-1-phosphate is an important metabolite for energy and biomass production through glycolysis and nucleotides required for DNA replication are produced through energetically expensive de novo or energy-efficient salvage pathways. Here, we describe p53-regulated pyrimidine salvage through NUDT22-dependent hydrolysis of UDP-glucose to maintain cancer cell growth and to prevent replication stress. NUDT22 expression is consistently elevated in cancer tissues and high NUDT22 expression correlates with worse survival outcomes in patients indicating an increased dependency of cancer cells to NUDT22. Furthermore, we show that NUDT22 transcription is induced after inhibition of glycolysis, MYC-mediated oncogenic stress, and DNA damage directly through p53. NUDT22-deficient cancer cells suffer from growth retardation, S-phase delay, and slower DNA replication fork speed. Uridine supplementation rescues replication fork progression and alleviates replication stress and DNA damage. Conversely, NUDT22 deficiency sensitizes cells to de novo pyrimidine synthesis inhibition in vitro and reduces cancer growth in vivo. In conclusion, NUDT22 maintains pyrimidine supply in cancer cells and depletion of NUDT22 leads to genome instability. Targeting NUDT22 therefore has high potential for therapeutic applications in cancer therapy.
Collapse
Affiliation(s)
- Melanie Walter
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2RX, UK
| | - Florian Mayr
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institute, 171 76, Stockholm, Sweden
| | - Bishoy M F Hanna
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institute, 171 76, Stockholm, Sweden
| | - Victoria Cookson
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2RX, UK
| | - Oliver Mortusewicz
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institute, 171 76, Stockholm, Sweden
| | - Thomas Helleday
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institute, 171 76, Stockholm, Sweden
| | - Patrick Herr
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2RX, UK.
| |
Collapse
|
14
|
Liu G, Guo Z, Zhao X, Sun J, Yue S, Li M, Chen Z, Ma Z, Zhao H. Whole Genome Resequencing Identifies Single-Nucleotide Polymorphism Markers of Growth and Reproduction Traits in Zhedong and Zi Crossbred Geese. Genes (Basel) 2023; 14:487. [PMID: 36833414 PMCID: PMC9956059 DOI: 10.3390/genes14020487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
The broodiness traits of domestic geese are a bottleneck that prevents the rapid development of the goose industry. To reduce the broodiness of the Zhedong goose and thus improve it, this study hybridized it with the Zi goose, which has almost no broody behavior. Genome resequencing was performed for the purebred Zhedong goose, as well as the F2 and F3 hybrids. The results showed that the F1 hybrids displayed significant heterosis in growth traits, and their body weight was significantly greater than those of the other groups. The F2 hybrids showed significant heterosis in egg-laying traits, and the number of eggs laid was significantly greater than those of the other groups. A total of 7,979,421 single-nucleotide polymorphisms (SNPs) were obtained, and three SNPs were screened. Molecular docking results showed that SNP11 located in the gene NUDT9 altered the structure and affinity of the binding pocket. The results suggested that SNP11 is an SNP related to goose broodiness. In the future, we will use the cage breeding method to sample the same half-sib families to accurately identify SNP markers of growth and reproductive traits.
Collapse
Affiliation(s)
- Guojun Liu
- Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, No. 368 Xuefu Road, Harbin 150086, China
| | - Zhenhua Guo
- Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, No. 368 Xuefu Road, Harbin 150086, China
| | - Xiuhua Zhao
- Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, No. 368 Xuefu Road, Harbin 150086, China
| | - Jinyan Sun
- Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, No. 368 Xuefu Road, Harbin 150086, China
| | - Shan Yue
- Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, No. 368 Xuefu Road, Harbin 150086, China
| | - Manyu Li
- Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, No. 368 Xuefu Road, Harbin 150086, China
| | - Zhifeng Chen
- Heilongjiang Academy of Agricultural Sciences, Qiqihare Branch Academy, No. 2 Heyi Road, Qiqihare 161005, China
| | - Zhigang Ma
- Heilongjiang Academy of Agricultural Sciences, Qiqihare Branch Academy, No. 2 Heyi Road, Qiqihare 161005, China
| | - Hui Zhao
- Liaoning Academy of Agricultural Sciences, No. 84 Dongling Road, Shenyang 110161, China
| |
Collapse
|
15
|
Liu Y, Jiang B, Lin C, Zhu W, Chen D, Sheng Y, Lou Z, Ji Z, Wu C, Wu M. m7G-related gene NUDT4 as a novel biomarker promoting cancer cell proliferation in lung adenocarcinoma. Front Oncol 2023; 12:1055605. [PMID: 36761423 PMCID: PMC9902657 DOI: 10.3389/fonc.2022.1055605] [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: 09/28/2022] [Accepted: 12/29/2022] [Indexed: 01/25/2023] Open
Abstract
Background Lung cancer is the leading cause of mortality in cancer patients. N7-methylguanosine (m7G) modification as a translational regulation pattern has been reported to participate in multiple types of cancer progression, but little is known in lung cancer. This study attempts to explore the role of m7G-related proteins in genetic and epigenetic variations in lung adenocarcinoma, and its relationship with clinical prognosis, immune infiltration, and immunotherapy. Methods Sequencing data were obtained from the Genomic Data Commons (GDC) Data Portal and Gene Expression Omnibus (GEO) databases. Consensus clustering was utilized to distinguish m7G clusters, and responses to immunotherapy were also evaluated. Moreover, univariate and multivariate Cox and Least absolute shrinkage and selection operator LASSO Cox regression analyses were used to screen independent prognostic factors and generated risk scores for constructing a survival prediction model. Multiple cell types such as epithelial cells and immune cells were identified to verify the bulk RNA results. Short hairpin RNA (shRNA) Tet-on plasmids, Clustered Regularly Interspaced Short Palindromic Repeats CRISPR/Cas9 for knockout plasmids, and nucleoside diphosphate linked to moiety X-type motif 4 (NUDT4) overexpression plasmids were constructed to inhibit or promote tumor cell NUDT4 expression, then RT-qPCR, Cell Counting Kit-8 CCK8 proliferation assay, and Transwell assay were used to observe tumor cell biological functions. Results Fifteen m7G-related genes were highly expressed in tumor samples, and 12 genes were associated with poor prognosis. m7G cluster-B had lower immune infiltration level, worse survival, and samples that predicted poor responses to immunotherapy. The multivariate Cox model showed that NUDT4 and WDR4 (WD repeat domain 4) were independent risk factors. Single-cell m7G gene set variation analysis (GSVA) scores also had a negative correlation tendency with immune infiltration level and T-cell Programmed Death-1 PD-1 expression, but the statistics were not significant. Knocking down and knocking out the NUDT4 expression significantly inhibited cell proliferation capability in A549 and H1299 cells. In contrast, overexpressing NUDT4 promoted tumor cell proliferation. However, there was no difference in migration capability in the knockdown, knockout, or overexpression groups. Conclusions Our study revealed that m7G modification-related proteins are closely related to the tumor microenvironment, immune cell infiltration, responses to immunotherapy, and patients' prognosis in lung adenocarcinoma and could be useful biomarkers for the identification of patients who could benefit from immunotherapy. The m7G modification protein NUDT4 may be a novel biomarker in promoting the progression of lung cancer.
Collapse
Affiliation(s)
- Yafei Liu
- Department of Thoracic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hang Zhou, China
| | - Bin Jiang
- Department of Thoracic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hang Zhou, China
| | - Chunjie Lin
- Life Sciences Institute, Zhejiang University, Hang Zhou, China
| | - Wanyinhui Zhu
- Life Sciences Institute, Zhejiang University, Hang Zhou, China
| | - Dingrui Chen
- Life Sciences Institute, Zhejiang University, Hang Zhou, China
| | - Yinuo Sheng
- Life Sciences Institute, Zhejiang University, Hang Zhou, China
| | - Zhiling Lou
- Department of Thoracic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hang Zhou, China
| | - Zhiheng Ji
- Department of Thoracic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hang Zhou, China
| | - Chuanqiang Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hang Zhou, China
| | - Ming Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hang Zhou, China,*Correspondence: Ming Wu,
| |
Collapse
|
16
|
Design of Novel Coumarin Derivatives as NUDT5 Antagonists That Act by Restricting ATP Synthesis in Breast Cancer Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010089. [PMID: 36615284 PMCID: PMC9822328 DOI: 10.3390/molecules28010089] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/06/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
Breast cancer, a heterogeneous disease, is among the most frequently diagnosed diseases and is the second leading cause of death due to cancer among women after lung cancer. Phytoactives (plant-based derivatives) and their derivatives are safer than synthetic compounds in combating chemoresistance. In the current work, a template-based design of the coumarin derivative was designed to target the ADP-sugar pyrophosphatase protein. The novel coumarin derivative (2R)-2-((S)-sec-butyl)-5-oxo-4-(2-oxochroman-4-yl)-2,5-dihydro-1H-pyrrol-3-olate was designed. Molecular docking studies provided a docking score of -6.574 kcal/mol and an MM-GBSA value of -29.15 kcal/mol. Molecular dynamics simulation studies were carried out for 500 ns, providing better insights into the interaction. An RMSD change of 2.4 Å proved that there was a stable interaction and that there was no conformational change induced to the receptor. Metadynamics studies were performed to calculate the unbinding energy of the principal compound with NUDT5, which was found to be -75.171 kcal/mol. In vitro validation via a cytotoxicity assay (MTT assay) of the principal compound was carried out with quercetin as a positive control in the MCF7 cell line and with an IC50 value of 55.57 (+/-) 0.7 μg/mL. This work promoted the research of novel natural derivatives to discover their anticancer activity.
Collapse
|
17
|
Shaikh MAJ, Alharbi KS, Almalki WH, Imam SS, Albratty M, Meraya AM, Alzarea SI, Kazmi I, Al-Abbasi FA, Afzal O, Altamimi ASA, Singh Y, Singh SK, Dua K, Gupta G. Sodium alginate based drug delivery in management of breast cancer. Carbohydr Polym 2022; 292:119689. [PMID: 35725179 DOI: 10.1016/j.carbpol.2022.119689] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 05/14/2022] [Accepted: 05/31/2022] [Indexed: 11/19/2022]
Abstract
Among women, breast cancer (B·C.) is a common form of cancer that can strike either developed or developing countries. In addition to pregnancy-related variables, hormone therapy lifestyle factors (e.g., physical inactivity, smoking, and alcohol use) may all influence the progression of B·C. The creation of anti-B·C. medication carriers with better stability, controlled and targeted administration, and the goal of minimizing unwanted effects has taken a lot of time and effort. Naturally generated biopolymers-based pharmaceutical delivery techniques have attracted attention for their potential use in treating B·C. It's been shown that natural polymers can deliver high medication concentrations to the desired place and provide prolonged release of pharmaceuticals useful in treating B.C. Alginate is one of the most commonly used drug carriers for delayed and targeted release. In present review will discuss the utilization of sodium alginate as an carrier of anticancer drug, such as paclitaxel, doxorubicin, tamoxifen, curcumin, and others.
Collapse
Affiliation(s)
- Mohammad Arshad Javed Shaikh
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura 302017, Mahal Road, Jaipur, India; Department of Pharmacy, TPCT's College of Engineering, Osmanabad, Maharashtra, India
| | - Khalid Saad Alharbi
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Syed Sarim Imam
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Abdulkarim M Meraya
- Pharmacy Practice Research Unit, Department of Clinical Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fahad A Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | | | - Yogendra Singh
- Department of Pharmacology, Maharishi Arvind College of Pharmacy, Ambabari Circle, Ambabari, Jaipur 302023, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura 302017, Mahal Road, Jaipur, India; Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India.
| |
Collapse
|
18
|
Design, molecular docking, and molecular dynamics of thiourea-iron (III) metal complexes as NUDT5 inhibitors for breast cancer treatment. Heliyon 2022; 8:e10694. [PMID: 36177227 PMCID: PMC9513778 DOI: 10.1016/j.heliyon.2022.e10694] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/02/2022] [Accepted: 09/14/2022] [Indexed: 11/29/2022] Open
Abstract
In research, anticancer agents, such as thiourea derivative compounds, and metal complexes, such as those complexed with iron (III) metal, are often studied. The metal complexes are presumably more active than thiourea derivatives as free ligands; some negative effects may be reduced. The computational studies used in this study involved molecular docking with AutoDock and molecular dynamics (MD) simulations using Desmond to evaluate the stability of the interactions. The docking and MD analysis results showed that compounds 2 and 6 had stable interactions with NUDIX hydrolase type 5 (NUDT5)—one of the therapeutic targets for breast cancer—where they had the lowest root mean square deviation (RMSD) and root mean square fluctuation (RMSF) values compared to the other compounds. Together, these compounds are anti-breast cancer drug candidates.
Collapse
|
19
|
Li J, Wang ZH, Dang YM, Li DN, Liu Z, Dai DP, Cai JP. MTH1 suppression enhances the stemness of MCF7 through upregulation of STAT3. Free Radic Biol Med 2022; 188:447-458. [PMID: 35809767 DOI: 10.1016/j.freeradbiomed.2022.06.240] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 11/22/2022]
Abstract
MTH1 protein can sanitize the damaged (d)NTP pool and MTH1 inhibitors have been developed to impede the growth of rapidly proliferating tumor cells; however, the effect of MTH1 inhibition on breast cancer stemness has not been reported yet. Here, we constructed breast cancer cell lines with the stable depletion of MTH1. MTH1 suppression clearly increased the ratio of CD44+CD24-/low subpopulations and promoted the formation of tumorspheres in MCF7 and T47D cells. RNA expression profiling, RT-qPCR and Western blotting showed the upregulation of master stem cell transcription factors Sox2, Oct4 and Nanog in MTH1 knockdown cells. GSEA suggested and Western blotting verified that MTH1 knockdown increased the expression of phosphorylated STAT3 (Tyr705). Furthermore, we indirectly demonstrated that the increased concentration of 8-oxo-dGTP and 8-oxo-GTP in MTH1-knockdown cells and exogenous 8-oxoGTP, rather than 8-oxo-dGTP, could significantly increase the phosphorylation of STAT3. In conclusion, this work indicates that MTH1 inhibition increased the proportion of breast cancer stem cells (BCSCs) and promoted stemness properties in MCF7 cells.
Collapse
Affiliation(s)
- Jin Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China
| | - Zi-Hui Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China; Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, PR China
| | - Ya-Min Dang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China; Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, PR China
| | - Dan-Ni Li
- The Clinical Laboratory of Beijing Hospital, Ministry of Health, Beijing, PR China
| | - Zhen Liu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China; Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, PR China
| | - Da-Peng Dai
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China
| | - Jian-Ping Cai
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China.
| |
Collapse
|
20
|
Wang X, Song K, Fan Y, Du J, Liu J, Xu J, Zheng L, Ouyang R, Li Y, Miao Y, Zhang D. Ultrasound-triggered reactive oxygen species effector nanoamplifier for enhanced combination therapy of mutant p53 tumors. Colloids Surf B Biointerfaces 2022; 215:112489. [PMID: 35395477 DOI: 10.1016/j.colsurfb.2022.112489] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 10/18/2022]
Abstract
Reactive oxygen species (ROS) damage is a crucial method with which to inhibit tumor cell proliferation; however, tumor cells can reduce ROS damage by modulating multiple repair mechanisms, thus, reducing the efficacy of ROS damage in tumor therapy. In this study, we built an ultrasound-triggered ROS damage nanoamplifier using a synergistic strategy consisting of ROS damage and decreased tumor self-protection capability to enhance the treatment efficacy of mutant p53 tumors. A ROS damage nanoamplifier (PT@PTGA) was fabricated using amphiphilic polyglutamic acid (PTGA) to load with a sonosensitizer (protoporphyrin IX, PpIX) and an MTH1 inhibitor (TH287). Under ultrasonic excitation, PpIX catalyzes oxygen to produce singlet oxygen and release TH287 to inhibit MTH1 activity, thereby causing the accumulation of 8-oxo-dGTP, which enhances DNA damage and further induces cell apoptosis. In addition, TH287 allies with ROS to eliminate the mutated p53 protein in tumor cells, thus reducing the self-protective capacity of tumor cells. As a result, the "internal and external" aspects were combined to enhance sensitization for mutant p53 tumor therapy. The construction of a ROS nanoamplifier not only provides an effective strategy for the treatment of mutant p53 tumors but also supplies an integrated platform for tumor diagnosis and therapy.
Collapse
Affiliation(s)
- Xiang Wang
- Institute of Bismuth and Rhenium Science & School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Kang Song
- Institute of Bismuth and Rhenium Science & School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yan Fan
- Engineering Research Center of Optical Instrument and System, the Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Jun Du
- Institute of Bismuth and Rhenium Science & School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Jinliang Liu
- Institute of Bismuth and Rhenium Science & School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Jiayu Xu
- Institute of Bismuth and Rhenium Science & School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Lulu Zheng
- Engineering Research Center of Optical Instrument and System, the Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Ruizhuo Ouyang
- Institute of Bismuth and Rhenium Science & School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China; Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yuhao Li
- Institute of Bismuth and Rhenium Science & School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China; Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Yuqing Miao
- Institute of Bismuth and Rhenium Science & School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China; Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Dawei Zhang
- Engineering Research Center of Optical Instrument and System, the Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| |
Collapse
|
21
|
Tan D, Zhang Y. Silencing of Nudix type 5 represses proliferation and invasion and enhances chemosensitivity of gastric carcinoma cells by affecting the AKT/GSK-3β/β-catenin pathway. Toxicol Appl Pharmacol 2022; 441:115968. [PMID: 35247377 DOI: 10.1016/j.taap.2022.115968] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/22/2022] [Accepted: 02/27/2022] [Indexed: 01/01/2023]
Abstract
Nudix type 5 (NUDT5) has been recently identified as a new cancer-associated protein that is involved in numerous cancers. To date, the relationship between NUDT5 and gastric carcinoma has not been addressed. In the current research, we focused on exploring the potential relevance of NUDT5 in gastric carcinoma. The initial analysis of NUDT5 expression in gastric carcinoma by TCGA data revealed a clear increase in NUDT5 expression in tumor versus normal tissue. The increased expression of NUDT5 was also validated in the clinical specimens of gastric carcinoma by immunoblotting detection. Moreover, high NUDT5 levels predicted a poorer overall survival in gastric carcinoma patients. A series of cellular functional assays demonstrated that gastric carcinoma cells with silenced NUDT5 exhibited decreased proliferative and invasive ability, increased cell cycle arrest at the G0/G1 phase, and enhanced chemosensitivity. In-depth research showed that the silencing of NUDT5 led to a reduction in the activation of AKT and β-catenin. The reactivation of AKT blocked the repressive effect of NUDT5 silencing on β-catenin activation. The forced expression of β-catenin also reversed NUDT5-silencing-mediated anticancer effects. A Xenograft tumor assay confirmed the anticancer role of NUDT5 in gastric carcinoma in vivo. In short, these findings reveal elevated NUDT5 levels in gastric carcinoma and demonstrate that the inhibition of NUDT5 displays promising anticancer effects by affecting the AKT/β-catenin pathway. Thus, our work unveils a vital role of NUDT5 in gastric carcinoma and indicates it as a viable candidate target for anticancer drug discovery.
Collapse
Affiliation(s)
- Dong Tan
- Department of General Surgery, No. 215 Hospital of Shaanxi Nuclear Industry, No. 52 Weiyang West Road, Xianyang, Shaanxi 712000, PR China
| | - Yafei Zhang
- Department of Endoscopy, No. 215 Hospital of Shaanxi Nuclear Industry, No. 52 Weiyang West Road, Xianyang, Shaanxi 712000, China.
| |
Collapse
|
22
|
Bialkowski K, Szpila A. Specific 8-oxo-dGTPase activity of MTH1 (NUDT1) protein as a quantitative marker and prognostic factor in human colorectal cancer. Free Radic Biol Med 2021; 176:257-264. [PMID: 34624481 DOI: 10.1016/j.freeradbiomed.2021.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/23/2021] [Accepted: 10/03/2021] [Indexed: 11/27/2022]
Abstract
The MTH1 (NUDT1) gene, because it is frequently upregulated in many types of human cancers, has been considered a general marker of carcinogenesis for over two decades. The MTH1 protein hydrolyzes the oxidized mutagenic DNA precursor, 8-oxo-7,8-dihydro-2'-deoxyguanosine 5'-triphosphate (8-oxo-dGTP), to the corresponding 5'-monophosphate and inorganic pyrophosphate. This prevents its incorporation into DNA by DNA polymerases and protects cells from the accumulation of 8-oxo-dGTP-induced point mutations. Elevated MTH1 mRNA and protein in many types of human cancer indicate a worse prognosis. However, the enzymatic activity of MTH1 has remained largely uninvestigated in this context. Therefore, we have set out to determine the specific 8-oxo-dGTPase activity of MTH1 in 57 pairs of human colorectal cancers (CRC) and adjacent cancer-free tissues (CFCF). The goal was to ascertain the potential for measuring this enzymatic activity as a way to differentiate cancerous from non-cancerous specimens of the intestine, as well as defining its capabilities as a prognostic value for disease-free survival. We found that 79% of CRC tumors exhibited a higher MTH1 activity than did CFCF, with a significant 1.6-fold increase in overall median value (p < 1E-6). The 8-oxo-dGTPase in both tissues was proportional to the corresponding levels of MTH1 protein, as assayed by Western blotting. Activity higher than the ROC-optimized threshold (AUC = 0.71) indicated cancerous tissue, with a 54% sensitivity and an 83% specificity. Postoperative fate followed for up to 100 months showed that higher 8-oxo-dGTPase, in either the CFCF or the CRC tumor, clearly lowered the probability of a relapse-free survival, although borderline statistical significance (p < 0.05) was crossed only for the CFCF.
Collapse
Affiliation(s)
- Karol Bialkowski
- Department of Clinical Biochemistry, L. Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Bydgoszcz, Poland.
| | - Anna Szpila
- Department of Clinical Biochemistry, L. Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Bydgoszcz, Poland
| |
Collapse
|
23
|
Poltronieri P, Miwa M, Masutani M. ADP-Ribosylation as Post-Translational Modification of Proteins: Use of Inhibitors in Cancer Control. Int J Mol Sci 2021; 22:10829. [PMID: 34639169 PMCID: PMC8509805 DOI: 10.3390/ijms221910829] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/02/2021] [Accepted: 10/05/2021] [Indexed: 12/14/2022] Open
Abstract
Among the post-translational modifications of proteins, ADP-ribosylation has been studied for over fifty years, and a large set of functions, including DNA repair, transcription, and cell signaling, have been assigned to this post-translational modification (PTM). This review presents an update on the function of a large set of enzyme writers, the readers that are recruited by the modified targets, and the erasers that reverse the modification to the original amino acid residue, removing the covalent bonds formed. In particular, the review provides details on the involvement of the enzymes performing monoADP-ribosylation/polyADP-ribosylation (MAR/PAR) cycling in cancers. Of note, there is potential for the application of the inhibitors developed for cancer also in the therapy of non-oncological diseases such as the protection against oxidative stress, the suppression of inflammatory responses, and the treatment of neurodegenerative diseases. This field of studies is not concluded, since novel enzymes are being discovered at a rapid pace.
Collapse
Affiliation(s)
- Palmiro Poltronieri
- Institute of Sciences of Food Productions, National Research Council of Italy, CNR-ISPA, Via Monteroni, 73100 Lecce, Italy
| | - Masanao Miwa
- Nagahama Institute of Bio-Science and Technology, Nagahama 526-0829, Japan;
| | - Mitsuko Masutani
- Department of Molecular and Genomic Biomedicine, CBMM, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan
| |
Collapse
|