1
|
Li J, Wang X, Zhang H, Hu X, Peng X, Jiang W, Zhuo L, Peng Y, Zeng G, Wang Z. Fenamates: Forgotten treasure for cancer treatment and prevention: Mechanisms of action, structural modification, and bright future. Med Res Rev 2024. [PMID: 39171404 DOI: 10.1002/med.22079] [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: 12/13/2022] [Revised: 08/03/2024] [Accepted: 08/08/2024] [Indexed: 08/23/2024]
Abstract
Fenamates as classical nonsteroidal anti-inflammatory agents are widely used for relieving pain. Preclinical studies and epidemiological data highlight their chemo-preventive and chemotherapeutic potential for cancer. However, comprehensive reviews of fenamates in cancer are limited. To accelerate the repurposing of fenamates, this review summarizes the results of fenamates alone or in combination with existing chemotherapeutic agents. This paper also explores targets of fenamates in cancer therapy, including COX, AKR family, AR, gap junction, FTO, TEAD, DHODH, TAS2R14, ion channels, and DNA. Besides, this paper discusses other mechanisms, such as regulating Wnt/β-catenin, TGF-β, p38 MAPK, and NF-κB pathway, and the regulation of the expressions of Sp, EGR-1, NAG-1, ATF-3, ErbB2, AR, as well as the modulation of the tumor immune microenvironment. Furthermore, this paper outlined the structural modifications of fenamates, highlighting their potential as promising leads for anticancer drugs.
Collapse
Affiliation(s)
- Junfang Li
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaodong Wang
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Honghua Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaoling Hu
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Xue Peng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weifan Jiang
- Postdoctoral Station for Basic Medicine, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Linsheng Zhuo
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Postdoctoral Station for Basic Medicine, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yan Peng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Guo Zeng
- Postdoctoral Station for Basic Medicine, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhen Wang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Postdoctoral Station for Basic Medicine, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
2
|
Kofman K, Levin M. Bioelectric pharmacology of cancer: A systematic review of ion channel drugs affecting the cancer phenotype. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 191:25-39. [PMID: 38971325 DOI: 10.1016/j.pbiomolbio.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/21/2024] [Accepted: 07/04/2024] [Indexed: 07/08/2024]
Abstract
Cancer is a pernicious and pressing medical problem; moreover, it is a failure of multicellular morphogenesis that sheds much light on evolutionary developmental biology. Numerous classes of pharmacological agents have been considered as cancer therapeutics and evaluated as potential carcinogenic agents; however, these are spread throughout the primary literature. Here, we briefly review recent work on ion channel drugs as promising anti-cancer treatments and present a systematic review of the known cancer-relevant effects of 109 drugs targeting ion channels. The roles of ion channels in cancer are consistent with the importance of bioelectrical parameters in cell regulation and with the functions of bioelectric signaling in morphogenetic signals that act as cancer suppressors. We find that compounds that are well-known for having targets in the nervous system, such as voltage-gated ion channels, ligand-gated ion channels, proton pumps, and gap junctions are especially relevant to cancer. Our review suggests further opportunities for the repurposing of numerous promising candidates in the field of cancer electroceuticals.
Collapse
Affiliation(s)
- Karina Kofman
- Faculty of Dentistry, University of Toronto, Toronto, Canada
| | - Michael Levin
- Allen Discovery Center at Tufts University, USA; Wyss Institute for Biologically Inspired Engineering at Harvard University, USA.
| |
Collapse
|
3
|
Añazco-Guenkova AM, Miguel-López B, Monteagudo-García Ó, García-Vílchez R, Blanco S. The impact of tRNA modifications on translation in cancer: identifying novel therapeutic avenues. NAR Cancer 2024; 6:zcae012. [PMID: 38476632 PMCID: PMC10928989 DOI: 10.1093/narcan/zcae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/16/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Recent advancements have illuminated the critical role of RNA modifications in post-transcriptional regulation, shaping the landscape of gene expression. This review explores how tRNA modifications emerge as critical players, fine-tuning functionalities that not only maintain the fidelity of protein synthesis but also dictate gene expression and translation profiles. Highlighting their dysregulation as a common denominator in various cancers, we systematically investigate the intersection of both cytosolic and mitochondrial tRNA modifications with cancer biology. These modifications impact key processes such as cell proliferation, tumorigenesis, migration, metastasis, bioenergetics and the modulation of the tumor immune microenvironment. The recurrence of altered tRNA modification patterns across different cancer types underscores their significance in cancer development, proposing them as potential biomarkers and as actionable targets to disrupt tumorigenic processes, offering new avenues for precision medicine in the battle against cancer.
Collapse
Affiliation(s)
- Ana M Añazco-Guenkova
- Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) - University of Salamanca, 37007 Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| | - Borja Miguel-López
- Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) - University of Salamanca, 37007 Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| | - Óscar Monteagudo-García
- Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) - University of Salamanca, 37007 Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| | - Raquel García-Vílchez
- Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) - University of Salamanca, 37007 Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| | - Sandra Blanco
- Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) - University of Salamanca, 37007 Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
4
|
Ringuette D, EbrahimAmini A, Sangphosuk W, Aquilino MS, Carroll G, Ashley M, Bazzigaluppi P, Dufour S, Droguerre M, Stefanovic B, Levi O, Charveriat M, Monnier PP, Carlen PL. Spreading depolarization suppression from inter-astrocytic gap junction blockade assessed with multimodal imaging and a novel wavefront detection scheme. Neurotherapeutics 2024; 21:e00298. [PMID: 38241157 PMCID: PMC10903093 DOI: 10.1016/j.neurot.2023.10.008] [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: 10/03/2023] [Accepted: 10/07/2023] [Indexed: 01/21/2024] Open
Abstract
Spreading depolarizations (SDs) are an enigmatic and ubiquitous co-morbidity of neural dysfunction. SDs are propagating waves of local field depolarization and increased extracellular potassium. They increase the metabolic demand on brain tissue, resulting in changes in tissue blood flow, and are associated with adverse neurological consequences including stroke, epilepsy, neurotrauma, and migraine. Their occurrence is associated with poor patient prognosis through mechanisms which are only partially understood. Here we show in vivo that two (structurally dissimilar) drugs, which suppress astroglial gap junctional communication, can acutely suppress SDs. We found that mefloquine hydrochloride (MQH), administered IP, slowed the propagation of the SD potassium waveform and intermittently led to its suppression. The hemodynamic response was similarly delayed and intermittently suppressed. Furthermore, in instances where SD led to transient tissue swelling, MQH reduced observable tissue displacement. Administration of meclofenamic acid (MFA) IP was found to reduce blood flow, both proximal and distal, to the site of SD induction, preceding a large reduction in the amplitude of the SD-associated potassium wave. We introduce a novel image processing scheme for SD wavefront localization under low-contrast imaging conditions permitting full-field wavefront velocity mapping and wavefront parametrization. We found that MQH administration delayed SD wavefront's optical correlates. These two clinically used drugs, both gap junctional blockers found to distinctly suppress SDs, may be of therapeutic benefit in the various brain disorders associated with recurrent SDs.
Collapse
Affiliation(s)
- Dene Ringuette
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada; Division of Genetics and Development, Krembil Research Institute, 60 Leonard Ave., Toronto, Ontario M5T 2S8, Canada; Krembil Neuroscience, Krembil Research Institute, 60 Leonard Ave., Toronto, Ontario M5T 2S8, Canada.
| | - Azin EbrahimAmini
- Krembil Neuroscience, Krembil Research Institute, 60 Leonard Ave., Toronto, Ontario M5T 2S8, Canada; The Institute Biomedical Engineering, University of Toronto, 164 College St., Toronto, Ontario M5S 3G9, Canada
| | - Weerawong Sangphosuk
- Krembil Neuroscience, Krembil Research Institute, 60 Leonard Ave., Toronto, Ontario M5T 2S8, Canada
| | - Mark S Aquilino
- The Institute Biomedical Engineering, University of Toronto, 164 College St., Toronto, Ontario M5S 3G9, Canada
| | - Gwennyth Carroll
- The Institute Biomedical Engineering, University of Toronto, 164 College St., Toronto, Ontario M5S 3G9, Canada
| | - Max Ashley
- Krembil Neuroscience, Krembil Research Institute, 60 Leonard Ave., Toronto, Ontario M5T 2S8, Canada
| | - Paolo Bazzigaluppi
- Sunnybrook Health Sciences Center, 2075 Bayview Ave., Toronto, Ontario M4N 3M5, Canada
| | - Suzie Dufour
- The Institute Biomedical Engineering, University of Toronto, 164 College St., Toronto, Ontario M5S 3G9, Canada
| | | | - Bojana Stefanovic
- Department of Medical Biophysics, University of Toronto, 610 University Ave., Toronto, Ontario M5G 2M9, Canada; Sunnybrook Health Sciences Center, 2075 Bayview Ave., Toronto, Ontario M4N 3M5, Canada
| | - Ofer Levi
- The Institute Biomedical Engineering, University of Toronto, 164 College St., Toronto, Ontario M5S 3G9, Canada; The Edward S. Rogers Sr. Department of Electrical and Computer Engineering, University of Toronto, 10 King's College Rd., Toronto, Ontario M5S 3G4, Canada
| | | | - Philippe P Monnier
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada; Division of Genetics and Development, Krembil Research Institute, 60 Leonard Ave., Toronto, Ontario M5T 2S8, Canada; Department of Ophthalmology & Vision Science, Faculty of Medicine, University of Toronto, 340 College St., Toronto, Ontario M5T 3A9, Canada
| | - Peter L Carlen
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada; Division of Genetics and Development, Krembil Research Institute, 60 Leonard Ave., Toronto, Ontario M5T 2S8, Canada; Krembil Neuroscience, Krembil Research Institute, 60 Leonard Ave., Toronto, Ontario M5T 2S8, Canada; The Institute Biomedical Engineering, University of Toronto, 164 College St., Toronto, Ontario M5S 3G9, Canada
| |
Collapse
|
5
|
Sun Y, Jin D, Zhang Z, Ji H, An X, Zhang Y, Yang C, Sun W, Zhang Y, Duan Y, Kang X, Jiang L, Zhao X, Lian F. N6-methyladenosine (m6A) methylation in kidney diseases: Mechanisms and therapeutic potential. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194967. [PMID: 37553065 DOI: 10.1016/j.bbagrm.2023.194967] [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: 03/27/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/10/2023]
Abstract
The N6-methyladenosine (m6A) modification is regulated by methylases, commonly referred to as "writers," and demethylases, known as "erasers," leading to a dynamic and reversible process. Changes in m6A levels have been implicated in a wide range of cellular processes, including nuclear RNA export, mRNA metabolism, protein translation, and RNA splicing, establishing a strong correlation with various diseases. Both physiologically and pathologically, m6A methylation plays a critical role in the initiation and progression of kidney disease. The methylation of m6A may also facilitate the early diagnosis and treatment of kidney diseases, according to accumulating research. This review aims to provide a comprehensive overview of the potential role and mechanism of m6A methylation in kidney diseases, as well as its potential application in the treatment of such diseases. There will be a thorough examination of m6A methylation mechanisms, paying particular attention to the interplay between m6A writers, m6A erasers, and m6A readers. Furthermore, this paper will elucidate the interplay between various kidney diseases and m6A methylation, summarize the expression patterns of m6A in pathological kidney tissues, and discuss the potential therapeutic benefits of targeting m6A in the context of kidney diseases.
Collapse
Affiliation(s)
- Yuting Sun
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - De Jin
- Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Ziwei Zhang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Hangyu Ji
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuedong An
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuehong Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Cunqing Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenjie Sun
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuqing Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yingying Duan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaomin Kang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Linlin Jiang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuefei Zhao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengmei Lian
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
6
|
Shewaiter MA, Selim AA, Rashed HM, Moustafa YM, Gad S. Niosomal formulation of mefenamic acid for enhanced cancer targeting; preparation, characterization and biodistribution study using radiolabeling technique. J Cancer Res Clin Oncol 2023; 149:18065-18080. [PMID: 37982828 PMCID: PMC10725351 DOI: 10.1007/s00432-023-05482-8] [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: 09/08/2023] [Accepted: 10/18/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND This work aimed to prepare niosomal formulations of an anticancer agent [mefenamic acid (MEF)] to enhance its cancer targeting. 131I was utilized as a radiolabeling isotope to study the radio-kinetics of MEF niosomes. METHODS niosomal formulations were prepared by the ether injection method and assessed for entrapment efficiency (EE%), zeta potential (ZP), polydispersity index (PDI) and particle size (PS). MEF was labeled with 131I by direct electrophilic substitution reaction through optimization of radiolabeling-related parameters. In the radio-kinetic study, the optimal 131I-MEF niosomal formula was administered intravenously (I.V.) to solid tumor-bearing mice and compared to I.V. 131I-MEF solution as a control. RESULTS the average PS and ZP values of the optimal formulation were 247.23 ± 2.32 nm and - 28.3 ± 1.21, respectively. The highest 131I-MEF labeling yield was 98.7 ± 0.8%. The biodistribution study revealed that the highest tumor uptake of 131I-MEF niosomal formula and 131I-MEF solution at 60 min post-injection were 2.73 and 1.94% ID/g, respectively. CONCLUSION MEF-loaded niosomes could be a hopeful candidate in cancer treatment due to their potent tumor uptake. Such high targeting was attributed to passive targeting of the nanosized niosomes and confirmed by radiokinetic evaluation.
Collapse
Affiliation(s)
- Mona A Shewaiter
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Sinai University, Kantara, Egypt
| | - Adli A Selim
- Labeled Compounds Department, Hot Laboratories Center, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Hassan M Rashed
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Sinai University, Kantara, Egypt.
- Labeled Compounds Department, Hot Laboratories Center, Egyptian Atomic Energy Authority, Cairo, Egypt.
| | - Yasser M Moustafa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Shadeed Gad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt.
| |
Collapse
|
7
|
Maghsoudi H, Sheikhnia F, Sitarek P, Hajmalek N, Hassani S, Rashidi V, Khodagholi S, Mir SM, Malekinejad F, Kheradmand F, Ghorbanpour M, Ghasemzadeh N, Kowalczyk T. The Potential Preventive and Therapeutic Roles of NSAIDs in Prostate Cancer. Cancers (Basel) 2023; 15:5435. [PMID: 38001694 PMCID: PMC10670652 DOI: 10.3390/cancers15225435] [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: 10/20/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Prostate cancer (PC) is the second most common type of cancer and the leading cause of death among men worldwide. Preventing the progression of cancer after treatments such as radical prostatectomy, radiation therapy, and hormone therapy is a major concern faced by prostate cancer patients. Inflammation, which can be caused by various factors such as infections, the microbiome, obesity and a high-fat diet, is considered to be the main cause of PC. Inflammatory cells are believed to play a crucial role in tumor progression. Therefore, nonsteroidal anti-inflammatory drugs along with their effects on the treatment of inflammation-related diseases, can prevent cancer and its progression by suppressing various inflammatory pathways. Recent evidence shows that nonsteroidal anti-inflammatory drugs are effective in the prevention and treatment of prostate cancer. In this review, we discuss the different pathways through which these drugs exert their potential preventive and therapeutic effects on prostate cancer.
Collapse
Affiliation(s)
- Hossein Maghsoudi
- Student Research Committee, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (H.M.); (F.S.); (V.R.); (F.M.)
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (S.H.); (F.K.); (N.G.)
| | - Farhad Sheikhnia
- Student Research Committee, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (H.M.); (F.S.); (V.R.); (F.M.)
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (S.H.); (F.K.); (N.G.)
| | - Przemysław Sitarek
- Department of Medical Biology, Medical University of Lodz, 90-151 Lodz, Poland
| | - Nooshin Hajmalek
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol 47176-47754, Iran;
| | - Sepideh Hassani
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (S.H.); (F.K.); (N.G.)
| | - Vahid Rashidi
- Student Research Committee, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (H.M.); (F.S.); (V.R.); (F.M.)
| | - Sadaf Khodagholi
- School of Kinesiology and Health Science, York University, Toronto, ON M3J 1P3, Canada;
| | - Seyed Mostafa Mir
- Metabolic Disorders Research Center, Department of Biochemistry and Biophysics, Gorgan Faculty of Medicine, Golestan University of Medical Sciences, Gorgan 49189-36316, Iran;
| | - Faezeh Malekinejad
- Student Research Committee, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (H.M.); (F.S.); (V.R.); (F.M.)
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (S.H.); (F.K.); (N.G.)
| | - Fatemeh Kheradmand
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (S.H.); (F.K.); (N.G.)
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia 57147-83734, Iran
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia 57147-83734, Iran
| | - Mansour Ghorbanpour
- Department of Medicinal Plants, Faculty of Agriculture and Natural Resources, Arak University, Arak 38156-88349, Iran;
| | - Navid Ghasemzadeh
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (S.H.); (F.K.); (N.G.)
| | - Tomasz Kowalczyk
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland;
| |
Collapse
|
8
|
Chang R, Zhu S, Peng J, Lang Z, Zhou X, Liao H, Zou J, Zeng P, Tan S. The hippocampal FTO-BDNF-TrkB pathway is required for novel object recognition memory reconsolidation in mice. Transl Psychiatry 2023; 13:349. [PMID: 37963912 PMCID: PMC10645923 DOI: 10.1038/s41398-023-02647-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 10/24/2023] [Accepted: 11/01/2023] [Indexed: 11/16/2023] Open
Abstract
Memory reconsolidation refers to the process by which the consolidated memory was restored after reactivation (RA). Memory trace becomes labile after reactivation and inhibition of memory reconsolidation may disrupt or update the original memory trace, which provided a new strategy for the treatment of several psychiatric diseases, such as drug addiction and post-traumatic stress disorder. Fat mass and obesity-associated gene (FTO) is a novel demethylase of N6-methyladenosine (m6A) and it has been intensively involved in learning and memory. However, the role of FTO in memory reconsolidation has not been determined. In the present study, the function of FTO in memory reconsolidation was investigated in the novel object recognition (NOR) model in mice. The results showed that RA of NOR memory increased hippocampal FTO expression in a time-dependent manner, while FTO inhibitor meclofenamic acid (MA) injected immediately, but not 6 h after RA disrupted NOR memory reconsolidation. MA downregulated BDNF expression during NOR memory reconsolidation in the hippocampus, while the TrkB agonist 7,8-Dihydroxyflavone (7,8-DHF) reversed the disruptive effects of MA on NOR memory reconsolidation. Furthermore, overexpression of FTO increased BDNF expression via decreasing mRNA m6A in HT22 cells. Taken together, these results indicate that FTO may up-regulate the BDNF-TrkB pathway to promote NOR memory reconsolidation through m6A modification.
Collapse
Affiliation(s)
- Rui Chang
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Shanshan Zhu
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Jionghong Peng
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Zhenyi Lang
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Xinyu Zhou
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Hailin Liao
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Ju Zou
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Peng Zeng
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China.
| | - Sijie Tan
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China.
| |
Collapse
|
9
|
Zhou X, Li C, Chen T, Li W, Wang X, Yang Q. Targeting RNA N6-methyladenosine to synergize with immune checkpoint therapy. Mol Cancer 2023; 22:36. [PMID: 36810108 PMCID: PMC9942356 DOI: 10.1186/s12943-023-01746-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 02/11/2023] [Indexed: 02/24/2023] Open
Abstract
Cancer immunotherapy, especially immune checkpoint therapy, has revolutionized therapeutic options by reactivating the host immune system. However, the efficacy varies, and only a small portion of patients develop sustained antitumor responses. Hence, illustrating novel strategies that improve the clinical outcome of immune checkpoint therapy is urgently needed. N6-methyladenosine (m6A) has been proved to be an efficient and dynamic posttranscriptional modification process. It is involved in numerous RNA processing, such as splicing, trafficking, translation and degradation. Compelling evidence emphasizes the paramount role of m6A modification in the regulation of immune response. These findings may provide a foundation for the rational combination of targeting m6A modification and immune checkpoints in cancer treatment. In the present review, we summarize the current landscape of m6A modification in RNA biology, and highlight the latest findings on the complex mechanisms by which m6A modification governs immune checkpoint molecules. Furthermore, given the critical role of m6A modification in antitumor immunity, we discuss the clinical significance of targeting m6A modification to improve the efficacy of immune checkpoint therapy for cancer control.
Collapse
Affiliation(s)
- Xianyong Zhou
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Wenhua Xi Road No. 107, Jinan, Shandong China ,grid.476866.dDepartment of Breast Surgery, Binzhou People’s Hospital, Binzhou, Shandong China
| | - Chen Li
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Wenhua Xi Road No. 107, Jinan, Shandong China
| | - Tong Chen
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Wenhua Xi Road No. 107, Jinan, Shandong China
| | - Wenhao Li
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Wenhua Xi Road No. 107, Jinan, Shandong China
| | - Xiaolong Wang
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Wenhua Xi Road No. 107, Jinan, Shandong, China.
| | - Qifeng Yang
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Wenhua Xi Road No. 107, Jinan, Shandong, China. .,Department of Pathology Tissue Bank, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Xi Road No. 107, Shandong, Jinan, China. .,Research Institute of Breast Cancer, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
10
|
Saglam BS, Kanli A, Yanar S, Kasap M, Akpinar G. Investigation of the effect of meclofenamic acid on the proteome of LNCaP cells reveals changes in alternative polyadenylation and splicing machinery. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:190. [PMID: 36071279 DOI: 10.1007/s12032-022-01795-9] [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: 06/21/2022] [Accepted: 07/09/2022] [Indexed: 12/24/2022]
Abstract
Prostate cancer is the most common type of cancer among men, and there is still no definitively effective drug treatment. Thus, the search for novel drug agents that may be used for the effective treatment continues. Meclofenamic acid (MA), a non-steroidal anti-inflammatory drug, with anti-tumor effects in various types of cancers was used to investigate its effects on LNCaP cells, a prostate cancer cell line, at the proteome level. The cells were treated with 80 µM MA for 24 h and a comparative proteomic analysis was performed with their untreated control cells. Proteins were extracted from the cells and then were subjected to two-dimensional gel electrophoresis. Protein spots displaying changes in their regulation ratios for more than two-fold were excised from the gels and identified with MALDI-TOF/TOF mass spectrometry. Bioinformatics analysis of the differentially regulated proteins that we identified showed that they were all associated with and took part in related pathways. Glycolytic pathway, cytoskeletal formation, transport activity, protein metabolism, and most notably an mRNA processing pathway were affected by the MA treatment. In addition to presenting a detailed information for what is happening inside the cells upon MA treatment, the proteins affected by MA treatment hold the potential to be novel targets for prostate cancer treatment provided that further in vivo experiments are carried out.
Collapse
Affiliation(s)
- Busra Sahinoz Saglam
- Department of Medical Biology, Faculty of Medicine, Kocaeli University, İzmit, Kocaeli, Turkey
| | - Aylin Kanli
- Department of Medical Biology, Faculty of Medicine, Kocaeli University, İzmit, Kocaeli, Turkey.
| | - Sevinc Yanar
- Department of Medical Biology, Faculty of Medicine, Kocaeli University, İzmit, Kocaeli, Turkey
- Department of Histology and Embryology, Faculty of Medicine, Sakarya University, Serdivan, Sakarya, Turkey
| | - Murat Kasap
- Department of Medical Biology, Faculty of Medicine, Kocaeli University, İzmit, Kocaeli, Turkey
| | - Gurler Akpinar
- Department of Medical Biology, Faculty of Medicine, Kocaeli University, İzmit, Kocaeli, Turkey
| |
Collapse
|
11
|
Mahboubi-Rabbani M, Zarghi A. Lipoxygenase Inhibitors as Cancer Chemopreventives: Discovery, Recent Developments and Future Perspectives. Curr Med Chem 2021; 28:1143-1175. [PMID: 31820690 DOI: 10.2174/0929867326666191210104820] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/31/2019] [Accepted: 11/10/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Leukotrienes (LTs) constitute a bioactive group of Polyunsaturated Fatty Acid (PUFA) metabolites molded by the enzymatic activity of lipoxygenase (LO) and have a pivotal role in inflammation and allergy. Evidence is accumulating both by in vitro cell culture experiments and animal tumor model studies in support of the direct involvement of aberrant metabolism of arachidonic acid (ACD) in the development of several types of human cancers such as lung, prostate, pancreatic and colorectal malignancies. Several independent experimental data suggest a correlation between tumoral cells viability and LO gene expression, especially, 5-lipoxygenase (5-LO). Overexpressed 5-LO cells live longer, proliferate faster, invade more effectively through extracellular matrix destruction and activate the anti-apoptotic signaling mechanisms more intensively compared to the normal counterparts. Thus, some groups of lipoxygenase inhibitors may be effective as promising chemopreventive agents. METHODS A structured search of bibliographic databases for peer-reviewed research literature regarding the role of LO in the pathogenesis of cancer was performed. The characteristics of screened papers were summarized and the latest advances focused on the discovery of new LO inhibitors as anticancer agents were discussed. RESULTS More than 180 papers were included and summarized in this review; the majority was about the newly designed and synthesized 5-LO inhibitors as anti-inflammatory and anticancer agents. The enzyme's structure, 5-LO pathway, 5-LO inhibitors structure-activity relationships as well as the correlation between these drugs and a number of most prevalent human cancers were described. In most cases, it has been emphasized that dual cyclooxygenase-2/5-lipoxygenase (COX-2/5-LO) or dual 5-lipoxygenase/microsomal prostaglandin E synthase-1 (5-LO/mPGES-1) inhibitors possess considerable inhibitory activities against their target enzymes as well as potent antiproliferative effects. Several papers disclosing 5-lipoxygenase activating protein (FLAP) antagonists as a new group of 5-LO activity regulators are also subject to this review. Also, the potential of 12-lipoxygenase (12- LO) and 15-lipoxygenase (15-LO) inhibitors as chemopreventive agents was outlined to expand the scope of new anticancer agents discovery. Some peptides and peptidomimetics with anti-LT activities were described as well. In addition, the cytotoxic effects of lipoxygenase inhibitors and their adverse effects were discussed and some novel series of natural-product-derived inhibitors of LO was also discussed in this review. CONCLUSION This review gives insights into the novel lipoxygenase inhibitors with anticancer activity as well as the different molecular pharmacological strategies to inhibit the enzyme effectively. The findings confirm that certain groups of LO inhibitors could act as promising chemopreventive agents.
Collapse
Affiliation(s)
- Mohammad Mahboubi-Rabbani
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afshin Zarghi
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Kowalski-Chauvel A, Lacore MG, Arnauduc F, Delmas C, Toulas C, Cohen-Jonathan-Moyal E, Seva C. The m6A RNA Demethylase ALKBH5 Promotes Radioresistance and Invasion Capability of Glioma Stem Cells. Cancers (Basel) 2020; 13:cancers13010040. [PMID: 33375621 PMCID: PMC7795604 DOI: 10.3390/cancers13010040] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/17/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Glioblastoma stem cells (GBMSCs), which are particularly radio-resistant and invasive, are responsible for the high recurrence of glioblastoma (GBM). Therefore, there is a real need for a better understanding of the mechanisms involved in these processes and to identify new factors that might be targeted to radiosensitize GBMSC and decrease their invasive capability. Here, we report that the m6A RNA demethylase ALKBH5, which is overexpressed in GBMSCs, promotes their radioresistance by controlling the homologous repair. ALKBH5 was also involved in GBMSC invasion. These data suggest that ALKBH5 inhibition might be a novel approach to radiosensitize GBMSCs and to overcome their invasiveness. Abstract Recurrence of GBM is thought to be due to GBMSCs, which are particularly chemo-radioresistant and characterized by a high capacity to invade normal brain. Evidence is emerging that modulation of m6A RNA methylation plays an important role in tumor progression. However, the impact of this mRNA modification in GBM is poorly studied. We used patient-derived GBMSCs to demonstrate that high expression of the RNA demethylase, ALKBH5, increases radioresistance by regulating homologous recombination (HR). In cells downregulated for ALKBH5, we observed a decrease in GBMSC survival after irradiation likely due to a defect in DNA-damage repair. Indeed, we observed a decrease in the expression of several genes involved in the HR, including CHK1 and RAD51, as well as a persistence of γ-H2AX staining after IR. We also demonstrated in this study that ALKBH5 contributes to the aggressiveness of GBM by favoring the invasion of GBMSCs. Indeed, GBMSCs deficient for ALKBH5 exhibited a significant reduced invasion capability relative to control cells. Our data suggest that ALKBH5 is an attractive therapeutic target to overcome radioresistance and invasiveness of GBMSCs.
Collapse
Affiliation(s)
- Aline Kowalski-Chauvel
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier Toulouse III, 31100 Toulouse, France; (A.K.-C.); (M.G.L.); (F.A.); (C.D.); (C.T.); (E.C.-J.-M.)
| | - Marie Géraldine Lacore
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier Toulouse III, 31100 Toulouse, France; (A.K.-C.); (M.G.L.); (F.A.); (C.D.); (C.T.); (E.C.-J.-M.)
| | - Florent Arnauduc
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier Toulouse III, 31100 Toulouse, France; (A.K.-C.); (M.G.L.); (F.A.); (C.D.); (C.T.); (E.C.-J.-M.)
| | - Caroline Delmas
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier Toulouse III, 31100 Toulouse, France; (A.K.-C.); (M.G.L.); (F.A.); (C.D.); (C.T.); (E.C.-J.-M.)
- IUCT-Oncopole Toulouse, 31000 Tolouse, France
| | - Christine Toulas
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier Toulouse III, 31100 Toulouse, France; (A.K.-C.); (M.G.L.); (F.A.); (C.D.); (C.T.); (E.C.-J.-M.)
- IUCT-Oncopole Toulouse, 31000 Tolouse, France
| | - Elizabeth Cohen-Jonathan-Moyal
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier Toulouse III, 31100 Toulouse, France; (A.K.-C.); (M.G.L.); (F.A.); (C.D.); (C.T.); (E.C.-J.-M.)
- IUCT-Oncopole Toulouse, 31000 Tolouse, France
| | - Catherine Seva
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier Toulouse III, 31100 Toulouse, France; (A.K.-C.); (M.G.L.); (F.A.); (C.D.); (C.T.); (E.C.-J.-M.)
- Correspondence: ; Tel.: +33-(5)82741604
| |
Collapse
|
13
|
Surmounting cancer drug resistance: New insights from the perspective of N6-methyladenosine RNA modification. Drug Resist Updat 2020; 53:100720. [DOI: 10.1016/j.drup.2020.100720] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 08/10/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023]
|
14
|
Lei Y, Chen Y, Lin Z, Tian D, Han P. Comprehensive analysis of key biomarkers, immune infiltration and potential therapeutic agents for ulcerative colitis. Life Sci 2020; 260:118437. [PMID: 32950577 DOI: 10.1016/j.lfs.2020.118437] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023]
Abstract
AIMS There has been an increasing trend towards the ulcerative colitis (UC) incidence worldwide. The present study aimed to explore novel biomarkers and potential therapeutic agents for UC. MATERIALS AND METHODS Differentially expressed genes (DEGs) among UC and healthy control samples were identified by GEO2R online tool. Functional analysis was performed and protein-protein interaction networks were constructed. The hub genes were explored by Cytoscape, and quantitative real-time-PCR (qRT-PCR) was used to valid their expression in clinical samples. ImmuCellAI was utilized to analyze the fraction of 24 types of immune cells. The L1000 platform was applied to determine potential agents for UC treatment. The dextran sulfate sodium (DSS)-induced colitis model was used to identify the therapeutic effect of meclofenamic acid. KEY FINDINGS A total of 270 DEGs were identified among UC and healthy control samples. Functional analysis indicated that the DEGs were primarily enriched in several immune response and digestion pathways. A proportion of 18 immune-cell types was found to be significantly altered between UC and healthy control samples. 10 compounds were predicted to have therapeutic potentials for treating UC. Among them, we selected meclofenamic acid to identify its therapeutic effect on UC treatment by animal experiments. SIGNIFICANCE The current study comprehensively analyzed the DEGs and immune infiltration in UC, as well as screened for potential agents for UC treatment.
Collapse
Affiliation(s)
- Yu Lei
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Yu Chen
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Zhuoying Lin
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Dean Tian
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China.
| | - Ping Han
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China.
| |
Collapse
|
15
|
Guzman-Esquivel J, Mendoza-Hernandez MA, Tiburcio-Jimenez D, Avila-Zamora ON, Delgado-Enciso J, De-Leon-Zaragoza L, Casarez-Price JC, Rodriguez-Sanchez IP, Martinez-Fierro ML, Meza-Robles C, Barocio-Acosta A, Baltazar-Rodriguez LM, Zaizar-Fregoso SA, Plata-Florenzano JE, Delgado-Enciso I. Decreased biochemical progression in patients with castration-resistant prostate cancer using a novel mefenamic acid anti-inflammatory therapy: A randomized controlled trial. Oncol Lett 2020; 19:4151-4160. [PMID: 32391109 DOI: 10.3892/ol.2020.11509] [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: 07/19/2019] [Accepted: 11/13/2019] [Indexed: 12/19/2022] Open
Abstract
Prostate cancer (PCa) is the second most common non-dermatological cancer in men and is a growing public health problem. Castration-resistant disease (CRD) is the most advanced stage of the disease and is difficult to control. Patients with CRD may no longer accept conventional therapies as they are not in appropriate clinical conditions or they refuse to receive it. Given that inflammation is an essential component of CRD origin and progression, anti-inflammatory agents could be a therapeutic option with fenamates as one of the proposed choices. A prospective, randomized, double-blinded, 2-arm, parallel group, phase II-III clinical trial was performed involving 20 patients with CRD-PCa (with a prostate specific antigen level <100 ng/ml) that were undergoing androgen deprivation therapy (ADT) and did not accept any established treatment for that disease stage. In addition to ADT, 10 patients received placebo and 10 received mefenamic acid (500 mg orally every 12 h) for 6 months. The primary endpoint was the change in serum prostate-specific antigen (PSA) at 6 months. The PSA levels decreased significantly with mefenamic acid (an average 42% decrease), whereas there was an average 55% increase in the placebo group (P=0.024). In the patients treated with the placebo, 70% had biochemical disease progression (an increase of ≥25% in PSA levels), which did not occur in any of the patients treated with mefenamic acid (relative risk=0.12; 95% confidence interval, 0.01-0.85; P=0.033). There was a significant increase in quality of life (EQ-5D-5L score) and body mass index (BMI) with the experimental treatment. In conclusion, mefenamic acid administration decreased biochemical progression in patients with castration resistant PCa, improved their quality of life and increased their BMI. Future studies are required in order to strengthen the findings of the present clinical trial. Trial registration, Cuban Public Registry of Clinical Trials Database RPCEC00000248, August 2017.
Collapse
Affiliation(s)
- José Guzman-Esquivel
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico.,Department of Research, General Hospital of Zone No. 1 IMSS, Villa de Alvarez, Colima 28983, Mexico
| | | | - Daniel Tiburcio-Jimenez
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico
| | - Oscar N Avila-Zamora
- Department of Research, Cancerology State Institute, Colima State Health Services, Colima 28085, Mexico
| | - Josuel Delgado-Enciso
- Department of Research, Foundation for Cancer Ethics, Education and Research of The Cancerology State Institute, Colima 28085, Mexico
| | - Luis De-Leon-Zaragoza
- Department of Research, General Hospital of Zone No. 1 IMSS, Villa de Alvarez, Colima 28983, Mexico.,Department of Research, Cancerology State Institute, Colima State Health Services, Colima 28085, Mexico
| | - Juan C Casarez-Price
- Department of Research, General Hospital of Zone No. 1 IMSS, Villa de Alvarez, Colima 28983, Mexico.,Department of Research, Cancerology State Institute, Colima State Health Services, Colima 28085, Mexico
| | - Iram P Rodriguez-Sanchez
- Molecular and Structural Physiology Laboratory, School of Biological Sciences, Autonomous University of Nuevo León, Monterrey, Nuevo León 64460, Mexico
| | - Margarita L Martinez-Fierro
- Molecular Medicine Laboratory, Academic Unit of Human Medicine and Health Sciences, Autonomous University of Zacatecas, Zacatecas, Zacatecas 98160, Mexico
| | - Carmen Meza-Robles
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico.,Department of Research, Cancerology State Institute, Colima State Health Services, Colima 28085, Mexico
| | - Alejandro Barocio-Acosta
- Department of Research, Cancerology State Institute, Colima State Health Services, Colima 28085, Mexico
| | - Luz M Baltazar-Rodriguez
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico
| | - Sergio A Zaizar-Fregoso
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico
| | - Jorge E Plata-Florenzano
- Department of Research, General Hospital of Zone No. 1 IMSS, Villa de Alvarez, Colima 28983, Mexico.,Department of Research, Cancerology State Institute, Colima State Health Services, Colima 28085, Mexico
| | - Iván Delgado-Enciso
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico.,Department of Research, Cancerology State Institute, Colima State Health Services, Colima 28085, Mexico
| |
Collapse
|
16
|
Torabizadeh SA, Rezaeifar M, Jomehzadeh A, Nabizadeh Haghighi F, Ansari M. Radioprotective Potential of Sulindac Sulfide to Prevent DNA Damage Due to Ionizing Radiation. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:4127-4134. [PMID: 31827319 PMCID: PMC6902880 DOI: 10.2147/dddt.s218022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 11/15/2019] [Indexed: 11/23/2022]
Abstract
Introduction: The ionizing radiation exposure of the normal cell causes damage to DNA, which leads to cell dysfunction or even cell death. However, it is necessary to identify new radio protectives in order to protect normal cells. Sulindac sulfide (SS) is a metabolite of sulindac (a non-steroidal anti-inflammatory drug) known as a cyclooxygenase inhibitor. Free radicals and reactive oxygen species are generated in the IR-exposed cells. Also, the induced inflammation process causes damage in DNA. Purpose In this research, the radioprotective effect of SS was investigated against genotoxicity and lipid peroxidation induced by ionizing radiation in the human blood lymphocytes. Methods In this study, the human blood samples were pretreated with SS at different concentrations (10, 25, 50, 100 and 250 μM) and then were exposed to IR at a dose of 1.5 Gy. The micronucleus (MN) assay was used to indicate the radioprotective effects of SS on exposed cells. Total antioxidant activity of the SS was measured by using FRAP and DPPH assay. Also, the malondialdehyde (MDA) levels and the activity of superoxide dismutase (SOD) on the exposed cells were evaluated. Results It was found that SS decreased the percentage of MN induced by IR in exposed cells. Maximum reduction in the frequency of MN was observed at 250 μM of SS (87%) that provides the highest degree of protection against IR. On the other hand, pretreatment at 250 μM of SS inhibited IR-induced oxidative stress, which led to a decrease in the MN frequencies and MDA levels, while SOD activity showed an increase in the exposed cells. Conclusion It could be concluded that SS as a good radioprotective agent protects the human normal cells against the oxidative stress and genetic damage induced by IR.
Collapse
Affiliation(s)
- Seyedeh Atekeh Torabizadeh
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehdi Rezaeifar
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Jomehzadeh
- Department of Medical Physics, Faculty of Medicine, Medical Physics Department, Radiotherapy & Oncology Unit, Shafa Kerman Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Farzaneh Nabizadeh Haghighi
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehdi Ansari
- Drug and Food Control Department, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
17
|
Huang T, Guo J, Lv Y, Zheng Y, Feng T, Gao Q, Zeng W. Meclofenamic acid represses spermatogonial proliferation through modulating m 6A RNA modification. J Anim Sci Biotechnol 2019; 10:63. [PMID: 31333841 PMCID: PMC6621992 DOI: 10.1186/s40104-019-0361-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 05/10/2019] [Indexed: 12/12/2022] Open
Abstract
Background N6-Methyladenosine (m6A), the most prevalent modification in mammalian mRNA, plays important roles in numerous biological processes. Several m6A associated proteins such as methyltransferase like 3 (METTL3), methyltransferase like 14 (METTL14), α-ketoglutarate-dependent dioxygenase AlkB homolog 5 (ALKBH5) and YTH domain containing 2 (YTHDC2) are involved in the regulation of spermatogenesis and oogenesis. However, the role of the first detected m6A demethylase, fat mass and obesity associate protein (FTO), in germ cells remains elusive. Elucidation of FTO roles in the regulation of germ cell fate will provide novel insights into the mammalian reproduction. Methods Mouse GC-1 spg cells were treated with the ester form of meclofenamic acid (MA2) to inhibit the demethylase activity of FTO. The cellular m6A and m6Am level were analyzed through high performance liquid chromatography combined with tandem mass spectrometry (HPLC/MS-MS). The cell apoptosis was detected via TUNEL and flow cytometry. The cell proliferation was detected through EdU and western blot. The mRNA level of core cyclin dependent kinases (CDKs) was quantified via q-PCR. RNA decay assay were performed to detect RNA stability. Dual fluorescence assay was conducted to study whether MA2 affects the expression of CDK2 dependent on the m6A modification at 3’UTR. Results MA2 significantly increased the cellular m6A level and down-regulated the expression of CDK1, CDK2, CDK6 and CdC25a, resulting in arrest of G1/S transition and decrease of cell proliferation. MA2 downregulated CDK2 mRNA stability. Additionally, mutation of the predicted m6A sites in the Cdk2–3’UTR could mitigated the degradation of CDK2 mRNA after MA2 treatment. Conclusion MA2 affected CDKs expression through the m6A-dependent mRNA degradation pathway, and thus repressed spermatogonial proliferation. Electronic supplementary material The online version of this article (10.1186/s40104-019-0361-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tao Huang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 Shaanxi China
| | - Jiayin Guo
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 Shaanxi China
| | - Yinghua Lv
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 Shaanxi China
| | - Yi Zheng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 Shaanxi China
| | - Tongying Feng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 Shaanxi China
| | - Qiang Gao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 Shaanxi China
| | - Wenxian Zeng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 Shaanxi China
| |
Collapse
|
18
|
Sekine Y, Nakayama H, Miyazawa Y, Kato H, Furuya Y, Arai S, Koike H, Matsui H, Shibata Y, Ito K, Suzuki K. Simvastatin in combination with meclofenamic acid inhibits the proliferation and migration of human prostate cancer PC-3 cells via an AKR1C3 mechanism. Oncol Lett 2017; 15:3167-3172. [PMID: 29435052 DOI: 10.3892/ol.2017.7721] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 12/11/2017] [Indexed: 01/24/2023] Open
Abstract
Statins have become of interest in research due to their anticancer effects. However, the exact mechanism of their anticancer properties remains unclear. The authors previously reported that statins decrease intracellular cholesterol levels in androgen-independent prostate cancer cells. In de novo androgen synthesis, cholesterol is the primary material and certain enzymes have important roles. The present study aimed to determine whether simvastatin alters the expression of androgen synthesis-associated enzymes in androgen-independent prostate cancer cells. A novel combination therapy of statins and other drugs that inhibit the overexpression of enzymes involved in androgen synthesis was explored. The cytotoxicity of simvastatin and meclofenamic acid was assessed in prostate cancer cells using MTS and migration assays. Testosterone and dihydrotestosterone concentrations in the culture medium were measured using liquid chromatography-tandem mass spectrometry. RAC-α-serine/threonine-protein kinase (Akt) phosphorylation was detected by western blot analysis. Following treatment with simvastatin, aldo-keto reductase family 1 member C3 (AKR1C3) expression increased in PC-3 (>60-fold) and LNCaP-LA cells, however not in 22Rv1 cells. Small interfering (si)RNA was used to clarify the effects of AKR1C3 expression. The reduction in AKR1C3 expression in PC-3 cells following siRNA transfection was not associated with basal cell proliferation and migration; however, treatment with simvastatin decreased cell proliferation and migration. The combination of simvastatin and meclofenamic acid, an AKR1C3 inhibitor, further enhanced the inhibition of cell proliferation and migration compared with treatment with either drug alone. Furthermore, treatment with simvastatin attenuated insulin-like growth factor 1-induced Akt activation; however, the combination of simvastatin and meclofenamic acid further inhibited Akt activation. These results suggest that the combination of simvastatin and meclofenamic acid may be an effective strategy for the treatment of castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Yoshitaka Sekine
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Hiroshi Nakayama
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Yoshiyuki Miyazawa
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Haruo Kato
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Yosuke Furuya
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Seiji Arai
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Hidekazu Koike
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Hiroshi Matsui
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Yasuhiro Shibata
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Kazuto Ito
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Kazuhiro Suzuki
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
19
|
Wu Z, Cheng F, Li J, Li W, Liu G, Tang Y. SDTNBI: an integrated network and chemoinformatics tool for systematic prediction of drug-target interactions and drug repositioning. Brief Bioinform 2017; 18:333-347. [PMID: 26944082 DOI: 10.1093/bib/bbw012] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Indexed: 01/11/2023] Open
Abstract
Computational prediction of drug-target interactions (DTIs) and drug repositioning provides a low-cost and high-efficiency approach for drug discovery and development. The traditional social network-derived methods based on the naïve DTI topology information cannot predict potential targets for new chemical entities or failed drugs in clinical trials. There are currently millions of commercially available molecules with biologically relevant representations in chemical databases. It is urgent to develop novel computational approaches to predict targets for new chemical entities and failed drugs on a large scale. In this study, we developed a useful tool, namely substructure-drug-target network-based inference (SDTNBI), to prioritize potential targets for old drugs, failed drugs and new chemical entities. SDTNBI incorporates network and chemoinformatics to bridge the gap between new chemical entities and known DTI network. High performance was yielded in 10-fold and leave-one-out cross validations using four benchmark data sets, covering G protein-coupled receptors, kinases, ion channels and nuclear receptors. Furthermore, the highest areas under the receiver operating characteristic curve were 0.797 and 0.863 for two external validation sets, respectively. Finally, we identified thousands of new potential DTIs via implementing SDTNBI on a global network. As a proof-of-principle, we showcased the use of SDTNBI to identify novel anticancer indications for nonsteroidal anti-inflammatory drugs by inhibiting AKR1C3, CA9 or CA12. In summary, SDTNBI is a powerful network-based approach that predicts potential targets for new chemical entities on a large scale and will provide a new tool for DTI prediction and drug repositioning. The program and predicted DTIs are available on request.
Collapse
Affiliation(s)
- Zengrui Wu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, China
| | - Feixiong Cheng
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jie Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, China
| | - Weihua Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, China
| | - Guixia Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yun Tang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, China
| |
Collapse
|
20
|
Galván-Salazar HR, Soriano-Hernández AD, Montes-Galindo DA, Espíritu GC, Guzman-Esquivel J, Rodríguez-Sánchez IP, Newton-Sánchez OA, Martinez-Fierro ML, Gómez XGB, Rojas-Martínez A, Delgado-Enciso I. Preclinical trial on the use of doxycycline for the treatment of adenocarcinoma of the duodenum. Mol Clin Oncol 2016; 5:657-659. [PMID: 27900107 DOI: 10.3892/mco.2016.1013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 03/24/2016] [Indexed: 01/08/2023] Open
Abstract
Adenocarcinoma of the duodenum comprises 50-70% of duodenal tumors. There is an increase in extracellular matrix metalloproteinases in this disease and it has been suggested that they play an important role in the development and pathology. Therefore, new therapeutic recommendations based on inhibitors of these enzymes, such as doxycycline, are under investigation. The cytotoxic effect of doxycycline was evaluated in the HuTu-80 duodenal adenocarcinoma cell line and its antitumor effect was determined in an immunodeficient murine model. A 10-µM (4.4 µg/ml) concentration of doxycycline was capable of causing apoptosis in 90% of the culture cells. Doxycycline was also responsible for a decrease in tumor growth and an increase in the survival of the mice with HuTu-80-cell tumors. These results suggest that doxycycline is a potential cytotoxic and antitumor agent effective in the treatment of adenocarcinoma of the duodenum.
Collapse
Affiliation(s)
- Hector R Galván-Salazar
- School of Medicine, University of Colima, Colima 28040, Mexico; State Cancer Institute, State Ministry of Health of Colima, Colima 28000, Mexico; Zone No. 1 General Hospital General of the Mexican Social Security Institute (IMSS), Colima 28000, Mexico
| | - Alejandro D Soriano-Hernández
- School of Medicine, University of Colima, Colima 28040, Mexico; State Cancer Institute, State Ministry of Health of Colima, Colima 28000, Mexico
| | | | - Gabriel Ceja Espíritu
- School of Medicine, University of Colima, Colima 28040, Mexico; Zone No. 1 General Hospital General of the Mexican Social Security Institute (IMSS), Colima 28000, Mexico
| | - José Guzman-Esquivel
- Zone No. 1 General Hospital General of the Mexican Social Security Institute (IMSS), Colima 28000, Mexico
| | - Iram P Rodríguez-Sánchez
- Department of Genetics, School of Medicine, Autonomous University of Nuevo León (UANL), Monterrey, Nuevo León 64460, Mexico
| | | | - Margarita L Martinez-Fierro
- Molecular Medicine Laboratory, Academic Unit of Human Medicine and Health Sciences, Autonomous University of Zacatecas, Zacatecas 98160, Mexico
| | | | - Augusto Rojas-Martínez
- Center of Research and Development in Health Sciences and School of Medicine, UANL, Monterrey, Nuevo León 64460, Mexico
| | - Iván Delgado-Enciso
- School of Medicine, University of Colima, Colima 28040, Mexico; State Cancer Institute, State Ministry of Health of Colima, Colima 28000, Mexico
| |
Collapse
|
21
|
Delgado-Enciso I, Soriano-Hernández AD, Rodriguez-Hernandez A, Galvan-Salazar HR, Montes-Galindo DA, Martinez-Martinez R, Valdez-Velazquez LL, Gonzalez-Alvarez R, Espinoza-Gómez F, Newton-Sanchez OA, Lara-Esqueda A, Guzman-Esquivel J. Histological changes caused by meclofenamic acid in androgen-independent prostate cancer tumors: evaluation in a mouse model. Int Braz J Urol 2016; 41:1002-7. [PMID: 26689527 PMCID: PMC4756978 DOI: 10.1590/s1677-5538.ibju.2013.00186] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Accepted: 06/14/2014] [Indexed: 11/22/2022] Open
Abstract
Meclofenamic acid is a nonsteroidal anti-inflammatory drug that has shown therapeutic potential for different types of cancers, including androgen-independent prostate neoplasms. The antitumor effect of diverse nonsteroidal anti-inflammatory drugs has been shown to be accompanied by histological and molecular changes that are responsible for this beneficial effect. The objective of the present work was to analyze the histological changes caused by meclofenamic acid in androgen-independent prostate cancer. Tumors were created in a nude mouse model using PC3 cancerous human cells. Meclofenamic acid (10 mg/kg/day; experimental group, n=5) or saline solution (control group, n=5) was administered intraperitoneally for twenty days. Histological analysis was then carried out on the tumors, describing changes in the cellular architecture, fibrosis, and quantification of cellular proliferation and tumor vasculature. Meclofenamic acid causes histological changes that indicate less tumor aggression (less hypercellularity, fewer atypical mitoses, and fewer nuclear polymorphisms), an increase in fibrosis, and reduced cellular proliferation and tumor vascularity. Further studies are needed to evaluate the molecular changes that cause the beneficial and therapeutic effects of meclofenamic acid in androgen-independent prostate cancer.
Collapse
Affiliation(s)
- Iván Delgado-Enciso
- School of Medicine, University of Colima, Colima, México.,Instituto Estatal de Cancerología, Servicios de Salud del Estado de Colima, Colima, México
| | | | | | - Héctor R Galvan-Salazar
- School of Medicine, University of Colima, Colima, México.,Instituto Estatal de Cancerología, Servicios de Salud del Estado de Colima, Colima, México
| | | | | | | | | | | | | | | | - Jose Guzman-Esquivel
- School of Medicine, University of Colima, Colima, México.,Hospital General de Zona Nº1 del IMSS, Colima, México
| |
Collapse
|
22
|
Che JB, Liu ZH, Ma HB, Li Y, Zhao H, Li XH, Liu WC, Shi GN. Influence of As2O3 combined with ginsenosides Rg3 on inhibition of lung cancer NCI-H1299 cells and on subsistence of nude mice bearing hepatoma. ASIAN PAC J TROP MED 2015; 7:772-5. [PMID: 25129458 DOI: 10.1016/s1995-7645(14)60134-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 08/15/2014] [Accepted: 09/15/2014] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVES To study the effect of arsenic trioxide (As2O3) combined with ginsenosides Rg3 on inhibiting the NCI-H1299 lung cancer cells and subsistence in nude mice bearing hepatoma. METHODS MTT method was used to measure the inhibition effect of As2O3 combined Rg3 on NCI-H1299 cells, and the proliferation inhibiting effect was observed via establishing the transplanted tumor model in vitro. A total of 40 tumor-bearing nude mice were randomly divided into normal saline group, As2O3, Rg3 and As2O3+Rg3 group. Transplantation tumor model of lung cancer in nude mice was constructed, followed by injection of certain concentrations of normal saline, As2O3, ginseng saponin Rg3 and As2O3+Rg3 every day. The survival duration and the tumors size of the mice were recorded and the Kaplan-Meier curve was made; microscopic observation of apoptosis of tumor cells in vivo was done using TUNEL staining. RESULTS After 72 h of injection, inhibition rate of tumor cell in normal saline group, As2O3 group, Rg3 group and As2O3+Rg3 group was (5.66±0.31)%, (65.58±4.75)%, (44.69±3.32)% and (82.67±5.43)%, respectively. Inhibition rate of tumor cell in As2O3 group, Rg3 group and As2O3+Rg3 group was significantly higher than that of normal saline group (P<0.01); inhibition rate of tumor cells of As2O3+Rg3 group was significantly higher than that of the two groups given As2O3 or Rg3 alone (P<0.01). The tumor volume of As2O3 group, Rg3 group and As2O3+Rg3 group shrank to (65.38±3.25)%, (77.68±3.43)% and (42.65±3.55)% of the original, tumor volume of saline group was 1.21 times of the original size (P<0.01); Median survival of saline group, Rg3 group, As2O3 group were significantly shorter than that of As2O3+Rg3 group (P<0.01); co-ordinated intervention ability of As2O3+Rg3 on NCI-H1299 cell was significantly higher than that of As2O3 or Rg3, separately. CONCLUSIONS As2O3 combined with Rg3 can significantly inhibit proliferation of NCI-H1299 cells in lung cancer, prolong survival of tumor-bearing nude mice, and promote tumor cell apoptosis, and have significant effect on lung cancer treatment.
Collapse
Affiliation(s)
- Jian-Bo Che
- Huaihe River Hospital, Henan University, Kaifeng 475000, Henan, China
| | - Zhong-Hua Liu
- Huaihe River Hospital, Henan University, Kaifeng 475000, Henan, China
| | - Hong-Bing Ma
- Huaihe River Hospital, Henan University, Kaifeng 475000, Henan, China
| | - Yong Li
- Huaihe River Hospital, Henan University, Kaifeng 475000, Henan, China
| | - Hui Zhao
- Huaihe River Hospital, Henan University, Kaifeng 475000, Henan, China
| | - Xiao-Hui Li
- Huaihe River Hospital, Henan University, Kaifeng 475000, Henan, China
| | - Wei-Chao Liu
- Huaihe River Hospital, Henan University, Kaifeng 475000, Henan, China
| | - Gong-Ning Shi
- Huaihe River Hospital, Henan University, Kaifeng 475000, Henan, China.
| |
Collapse
|
23
|
Soriano-Hernandez AD, Madrigal-Pérez D, Galvan-Salazar HR, Martinez-Fierro ML, Valdez-Velazquez LL, Espinoza-Gómez F, Vazquez-Vuelvas OF, Olmedo-Buenrostro BA, Guzman-Esquivel J, Rodriguez-Sanchez IP, Lara-Esqueda A, Montes-Galindo DA, Delgado-Enciso I. Anti-inflammatory drugs and uterine cervical cancer cells: Antineoplastic effect of meclofenamic acid. Oncol Lett 2015; 10:2574-2578. [PMID: 26622892 DOI: 10.3892/ol.2015.3580] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 05/22/2015] [Indexed: 01/26/2023] Open
Abstract
Uterine cervical cancer (UCC) is one of the main causes of cancer-associated mortality in women. Inflammation has been identified as an important component of this neoplasia; in this context, anti-inflammatory drugs represent possible prophylactic and/or therapeutic alternatives that require further investigation. Anti-inflammatory drugs are common and each one may exhibit a different antineoplastic effect. As a result, the present study investigated different anti-inflammatory models of UCC in vitro and in vivo. Celecoxib, sulindac, nimesulide, dexamethasone, meclofenamic acid, flufenamic acid and mefenamic acid were tested in UCC HeLa, VIPA, INBL and SiHa cell lines. The cytotoxicity of the drugs was evaluated in vitro. Celecoxib, sulindac, nimesulide, mefenamic acid and flufenamic acid presented with slight to moderate toxicity (10-40% of cell death corresponding to 100 µM) in certain cell lines, while meclofenamic acid exhibited significant cytotoxicity in all essayed cell lines (50-90% of cell death corresponding to 100 µM). The meclofenamic acid was tested in murine models (immunodeficient and immunocompetent) of UCC, which manifested a significant reduction in tumor growth and increased mouse survival. It was demonstrated that of the evaluated anti-inflammatory drugs, meclofenamic acid was the most cytotoxic, with a significant antitumor effect in murine models. Subsequent studies are necessary to evaluate the clinical utility of this drug.
Collapse
Affiliation(s)
- Alejandro D Soriano-Hernandez
- School of Medicine, University of Colima, Colima, Colima 28030, Mexico ; Cancerology State Institute, Colima State Health Services, Colima, Colima 28000, Mexico
| | | | - Hector R Galvan-Salazar
- School of Medicine, University of Colima, Colima, Colima 28030, Mexico ; Cancerology State Institute, Colima State Health Services, Colima, Colima 28000, Mexico
| | - Margarita L Martinez-Fierro
- Molecular Medicine Laboratory, Academic Unit of Human Medicine and Health Sciences, Autononous University of Zacatecas, Zacatecas, Zacatecas 98160, Mexico
| | | | | | | | | | - Jose Guzman-Esquivel
- General Hospital of Zone No. 1, Mexican Institute of Social Security, Colima, Colima 28000, Mexico
| | - Iram P Rodriguez-Sanchez
- Department of Genetics, School of Medicine, Nuevo Leon Autonomous University, Monterrey, Nuevo León 64460, Mexico
| | - Agustin Lara-Esqueda
- Cancerology State Institute, Colima State Health Services, Colima, Colima 28000, Mexico
| | - Daniel A Montes-Galindo
- School of Medicine, University of Colima, Colima, Colima 28030, Mexico ; School of Chemistry, University of Colima, Coquimatlán, Colima 28400, Mexico
| | - Ivan Delgado-Enciso
- School of Medicine, University of Colima, Colima, Colima 28030, Mexico ; Cancerology State Institute, Colima State Health Services, Colima, Colima 28000, Mexico
| |
Collapse
|
24
|
Cunha VRR, Izumi CMS, Petersen PAD, Magalhães A, Temperini MLA, Petrilli HM, Constantino VRL. Mefenamic Acid Anti-Inflammatory Drug: Probing Its Polymorphs by Vibrational (IR and Raman) and Solid-State NMR Spectroscopies. J Phys Chem B 2014; 118:4333-44. [DOI: 10.1021/jp500988k] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Vanessa R. R. Cunha
- Departamento
de Química Fundamental, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, CEP 05508-000, São Paulo-SP, Brazil
| | - Celly M. S. Izumi
- Departamento
de Química Fundamental, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, CEP 05508-000, São Paulo-SP, Brazil
| | - Philippe A. D. Petersen
- Departamento
de Física dos Materiais e Mecânica, Instituto de Física, Universidade de São Paulo, CEP 05315-970, São Paulo-SP, Brazil
| | - Alviclér Magalhães
- Departamento
de Química Inorgânica, Instituto de Química, Universidade Estadual de Campinas, CEP 13083-970, Campinas-SP, Brazil
| | - Marcia L. A. Temperini
- Departamento
de Química Fundamental, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, CEP 05508-000, São Paulo-SP, Brazil
| | - Helena M. Petrilli
- Departamento
de Física dos Materiais e Mecânica, Instituto de Física, Universidade de São Paulo, CEP 05315-970, São Paulo-SP, Brazil
| | - Vera R. L. Constantino
- Departamento
de Química Fundamental, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, CEP 05508-000, São Paulo-SP, Brazil
| |
Collapse
|
25
|
Bishayee K, Khuda-Bukhsh AR. 5-lipoxygenase antagonist therapy: a new approach towards targeted cancer chemotherapy. Acta Biochim Biophys Sin (Shanghai) 2013; 45:709-19. [PMID: 23752617 DOI: 10.1093/abbs/gmt064] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Leukotrienes are the bioactive group of fatty acids and major constituents of arachidonic acid metabolism molded by the catalytic activity of 5-lipoxygenase (5-LOX). Evidence is accumulating in support of the direct involvement of 5-LOX in the progression of different types of cancer including prostate, lung, colon, and colorectal cancers. Several independent studies now support the correlation between the 5-LOX expression and cancer cell viability, proliferation, cell migration, invasion through extracellular matrix destruction, metastasis, and activation of anti-apoptotic signaling cascades. The involvement of epidermal growth factor receptor and 5-oxo-ETE receptor (OXER1) is the major talking point in the downstream of the 5-LOX pathway, which relates the cancer cells to the proliferative pathways. Antisense technology approaches and use of different kinds of blocker targeted to 5-LOX, FLAP (5-LOX-activating protein), and OXER1 have shown a greater efficiency in combating different cancer cell types. Lastly, suppression of 5-LOX activity that reduces the cell proliferation activity also induces intrinsic mitochondrial apoptotic pathway in either p53-dependent or independent manner. Pharmacological agents that specifically inhibit the LOX-mediated signaling pathways have been used during last few years to treat inflammatory diseases such as asthma and arthritis. Studies of these well-characterized agents are therefore warranted for their use as possible candidates for chemotherapeutic studies against the killer disease cancer.
Collapse
Affiliation(s)
- Kausik Bishayee
- Cytogenetics and Molecular Biology Laboratory, Department of Zoology, University of Kalyani, Kalyani, India
| | | |
Collapse
|
26
|
Choi ES, Han G, Park SK, Lee K, Kim HJ, Cho SD, Kim HM. A248, a novel synthetic HDAC inhibitor, induces apoptosis through the inhibition of specificity protein 1 and its downstream proteins in human prostate cancer cells. Mol Med Rep 2013; 8:195-200. [PMID: 23685644 DOI: 10.3892/mmr.2013.1481] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 04/17/2013] [Indexed: 11/05/2022] Open
Abstract
Histone deacetylase (HDAC) inhibitors are emerging as potent anticancer agents due to their ability to induce apoptosis in various cancer cells, including prostate cancer cells. In the present study, we synthesized a novel HDAC inhibitor, A248, and investigated its apoptotic activity and molecular target in the DU145 and PC3 human prostate cancer cell lines. A248 inhibited the growth of DU145 and PC3 cells and induced apoptosis, as demonstrated by nuclear fragmentation and the accumulation of cells at subG1 phase of cell cycle. The treatment of DU145 and PC3 prostate cancer cells with A248 resulted in the downregulation of specificity protein 1 (Sp1) expression. Since the expression levels of survivin and Mcl-1 depend on Sp1, we also investigated the effects of A248 on survivin and Mcl-1 expression using western blot analysis and immunocytochemistry. The results showed that A248 markedly decreased the expression of survivin and Mcl-1. These data suggest that A248 has apoptotic activity in human prostate cancer cells and that Sp1 may be the molecular target of A248 treatment for inducing apoptosis in prostate cancer cells.
Collapse
Affiliation(s)
- Eun-Sun Choi
- Department of Oral Pathology, School of Dentistry and Institute of Oral Bioscience, Brain Korea 21 Project, Chonbuk National University, Jeonju 561-756, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|