1,196 results on '"Meclofenamic acid"'
Search Results
2. Detouring NSAID into Mitochondria to Induce Apoptosis in Cancer Cells.
- Author
-
Mishra, Tripti, Goswami, Antara, Bajpai, Aman, and Basu, Sudipta
- Abstract
In recent years, impairing mitochondria in cancer cells gained attention as alternative cancer therapy. In this context, non‐steroidal anti‐inflammatory (NSAID) drugs are interesting candidates to damage mitochondria in cancer cells. However, routing NSAIDs specifically into the mitochondria remained a major challenge and less explored. Herein, we have synthesized a small library of Meclofenamic acid and Naproxen derivatives having ester and amide linkage with substituted triphenylphosphonium cations for mitochondria targeting. Screening in cervical cancer (HeLa), breast cancer (MCF7) and colon cancer (HCT‐116) cells revealed a Meclofenamic acid derivative having ester linkage with tri (4‐methoxyphenyl) phosphonium cation (8A3) which induced mitochondrial damage through mitochondrial outer membrane permeabilization (MOMP) followed by generation of reactive oxygen species (ROS) in the HCT‐116 cells. This 8A3‐mediated mitochondrial impairment triggered apoptosis by inhibiting Cox‐2, reduction in Bcl‐2/Bcl‐xl expression and Caspase‐3/9 cleavage leading to remarkable HCT‐116 cell death. This novel mitochondrion targeted Meclofenamic acid derivative has the potential to be used as a chemical biology tool to understand the role of NSAIDs in mitochondria towards cancer therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. The Impact of Simultaneous Epigenetic and Epitranscriptomic Intervention in Breast Cancer Cells.
- Author
-
YANAR, Sevinç, DEVECİ ÖZKAN, Asuman, BAL ALBAYRAK, Merve Gülşen, and BETTS, Zeynep
- Subjects
SODIUM butyrate ,HISTONE deacetylase inhibitors ,BREAST cancer ,STAINS & staining (Microscopy) ,ACRIDINE orange - Abstract
Copyright of Istanbul Gelisim University Journal of Health Sciences / İstanbul Gelişim Üniversitesi Sağlık Bilimleri Dergisi is the property of Istanbul Gelisim Universitesi Saglik Bilimleri Yuksekokulu and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2024
- Full Text
- View/download PDF
4. Investigation of the effect of meclofenamic acid on the proteome of LNCaP cells reveals changes in alternative polyadenylation and splicing machinery.
- Author
-
Saglam, Busra Sahinoz, Kanli, Aylin, Yanar, Sevinc, Kasap, Murat, and Akpinar, Gurler
- 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. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
5. Pentosan Polysulfate Sodium and Meclofenamic Acid as Treatments in Patients With Psychotic Disorders
- Published
- 2019
6. Recent Findings from National Autonomous University of Mexico (UNAM) Provides New Insights into Cancer (Repurposing Auranofin and Meclofenamic Acid As Energy-metabolism Inhibitors and Anti-cancer Drugs).
- Abstract
Recent research from the National Autonomous University of Mexico (UNAM) explores the potential of repurposing auranofin and meclofenamic acid as energy-metabolism inhibitors and anti-cancer drugs. The study found that auranofin and meclofenamic acid exhibited cytotoxic effects on cancer cell lines and mitochondria, impacting cell proliferation, energy metabolism, and metastasis. The researchers suggest that auranofin shows promise as an anticancer drug and may warrant further investigation for clinical treatment protocols. [Extracted from the article]
- Published
- 2024
7. Preparation of N-Aryl Anthranilic Acid Drugs by Modified Ullmann Coupling Reaction in Ionic Liquids.
- Author
-
Gu, Zhengyu, Xue, Feng, Yu, Jiale, and Ju, Shengui
- Subjects
- *
AMINOBENZOIC acids , *IONIC liquids , *PROTON magnetic resonance , *NUCLEAR magnetic resonance , *X-ray crystallography , *MASS spectrometry - Abstract
N-Aryl anthranilic acid drugs have been synthesized by a simple, environmentally friendly, low-cost, and high-yielding modified Ullmann coupling reaction protocol using potassium 2-bromobenzoate, substituted anilines, and copper acetate in tetrabutylphosphonium chloride ([TBP]Cl) ionic liquid. The optimal conditions have been found and applied to the synthesis of N-aryl anthranilic acid drugs at 170°C. Mass spectrometry, X-ray crystallography, and proton nuclear magnetic resonance were used to describe the structure of the products. Copper(I) complex catalyst was used as a starting catalyst for the Ullmann reaction because of the good fluidity and homogeneity in [TBP]Cl ionic liquid. Meclofenamic, mefenamic, clofenamic, and flufenamic acids were synthesized efficiently using the proposed general procedure. The distinct advantages of the described protocol are operational simplicity, cleaner reaction, high selectivity, excellent yield, rapid conversion, easy preparation, and the use of a low-cost catalyst. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
8. Meclofenamic Acid Restores Gefinitib Sensitivity by Downregulating Breast Cancer Resistance Protein and Multidrug Resistance Protein 7 via FTO/m6A-Demethylation/c-Myc in Non-Small Cell Lung Cancer.
- Author
-
Chen, Hui, Jia, Bin, Zhang, Qiang, and Zhang, Yu
- Subjects
NON-small-cell lung carcinoma ,MULTIDRUG resistance ,EPIDERMAL growth factor receptors ,BREAST cancer ,PROTEIN-tyrosine kinase inhibitors - Abstract
Background and Objective: Gefitinib (GE) is a first-line epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) for patients with advanced non-small cell lung cancer (NSCLC) carrying EGFR activating mutations. However, drug resistance limits the clinical efficacy of gefitinib and ultimately leads to extremely poor clinical benefit. Meclofenamic acid (MA) is a non-steroidal anti-inflammatory drug (NSAID) that relieves moderate and severe pain. In the present study, we aim to determine the MA sensibilization of GE in NSCLC. Methods: MTT assay was conducted to determine the synergistic effect of MA with GE in GE-sensitive and -resistant cell lines based on the Chou–Talalay method. The Annexin V-PI flow cytometry analysis was conducted to evaluate apoptosis. Western blot assay was used to detect alterations of EGFR downstream molecules. Tritium-labeled GE accumulation analysis was used to determine the efflux activity of GE. Dot blot assays were conducted to determine m6A levels after the MA and GE co-administration. Western blot evaluated the expression of FTO, c-Myc, MRP7, BCRP, and apoptotic proteins. Results: MA showed a significant synergistic effect with GE in GE-resistant NSCLC cells; co-administration of MA with GE induced caspase-related apoptosis in resistant NSCLC cells. Moreover, EGFR downstream molecules, including Akt and MAPKs pathways, were significantly inhibited by the MA-GE combination. Short-term incubation of MA did not alter the efflux of GE; however, after incubation for 24 h, the accumulation of tritium-labeled GE significantly increased. A mechanism study showed that co-administration of MA and GE significantly downregulated BCRP and MRP7 expression in GE-resistant cells; increased N6-methylation was also observed after co-administration. The FTO/c-Myc was determined as target pathways on MA and GE co-administration mechanisms. Conclusion: Our findings provide novel therapeutic approaches for GE-resistant NSCLC by combination use with MA through FTO-mediated N6-demethylation. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
9. Meclofenamic Acid Restores Gefinitib Sensitivity by Downregulating Breast Cancer Resistance Protein and Multidrug Resistance Protein 7 via FTO/m6A-Demethylation/c-Myc in Non-Small Cell Lung Cancer
- Author
-
Hui Chen, Bin Jia, Qiang Zhang, and Yu Zhang
- Subjects
meclofenamic acid ,gefitinib ,M6A ,non-small cell lung cancer (NSCLC) ,drug resistance ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background and ObjectiveGefitinib (GE) is a first-line epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) for patients with advanced non-small cell lung cancer (NSCLC) carrying EGFR activating mutations. However, drug resistance limits the clinical efficacy of gefitinib and ultimately leads to extremely poor clinical benefit. Meclofenamic acid (MA) is a non-steroidal anti-inflammatory drug (NSAID) that relieves moderate and severe pain. In the present study, we aim to determine the MA sensibilization of GE in NSCLC.MethodsMTT assay was conducted to determine the synergistic effect of MA with GE in GE-sensitive and -resistant cell lines based on the Chou–Talalay method. The Annexin V-PI flow cytometry analysis was conducted to evaluate apoptosis. Western blot assay was used to detect alterations of EGFR downstream molecules. Tritium-labeled GE accumulation analysis was used to determine the efflux activity of GE. Dot blot assays were conducted to determine m6A levels after the MA and GE co-administration. Western blot evaluated the expression of FTO, c-Myc, MRP7, BCRP, and apoptotic proteins.ResultsMA showed a significant synergistic effect with GE in GE-resistant NSCLC cells; co-administration of MA with GE induced caspase-related apoptosis in resistant NSCLC cells. Moreover, EGFR downstream molecules, including Akt and MAPKs pathways, were significantly inhibited by the MA-GE combination. Short-term incubation of MA did not alter the efflux of GE; however, after incubation for 24 h, the accumulation of tritium-labeled GE significantly increased. A mechanism study showed that co-administration of MA and GE significantly downregulated BCRP and MRP7 expression in GE-resistant cells; increased N6-methylation was also observed after co-administration. The FTO/c-Myc was determined as target pathways on MA and GE co-administration mechanisms.ConclusionOur findings provide novel therapeutic approaches for GE-resistant NSCLC by combination use with MA through FTO-mediated N6-demethylation.
- Published
- 2022
- Full Text
- View/download PDF
10. Down-regulation of FTO promotes proliferation and migration, and protects bladder cancer cells from cisplatin-induced cytotoxicity
- Author
-
Lijie Wen, Xianwei Pan, Yang Yu, and Bo Yang
- Subjects
FTO ,Bladder urothelial carcinoma ,Meclofenamic acid ,Cisplatin ,Diseases of the genitourinary system. Urology ,RC870-923 - Abstract
Abstract Background FTO is known to be associated with body mass and obesity in humans and its over-expression affects the energy metabolism of cancer cells. The aim of the present study is to investigate the biological role of FTO in human bladder urothelial carcinoma. Methods PCR and western blotting are used to measure the levels of FTO in both tissues and cell lines (5637, T24, TCCSUP) of human bladder urothelial carcinoma. Raw RNA-Sequencing reads and the corresponding clinical information for bladder urothelial carcinoma are downloaded from TCGA. Cell Counting Kit-8 and wound healing assays are used to explore the effect of FTO on proliferation and migration of bladder cancer cells. Results The expression of FTO mRNA in bladder urothelial carcinoma decreases significantly compared with the normal controls from both the data of real-time PCR (p
- Published
- 2020
- Full Text
- View/download PDF
11. Effect of Pharmacological Inhibition of Fat-Mass and Obesity-Associated Protein on Nerve Trauma-Induced Pain Hypersensitivities.
- Author
-
Zheng, Bi-Xin, Guo, Xinying, Albik, Sfian, Eloy, Jean, and Tao, Yuan-Xiang
- Abstract
Genetic knockout or knockdown of fat-mass and obesity-associated protein (FTO), a demethylase that participates in RNA N
6 -methyladenosine modification in injured dorsal root ganglion (DRG), has been demonstrated to alleviate nerve trauma-induced nociceptive hypersensitivities. However, these genetic strategies are still impractical in clinical neuropathic pain management. The present study sought to examine the effect of intrathecal administration of two specific FTO inhibitors, meclofenamic acid (MA) and N-CDPCB, on the development and maintenance of nociceptive hypersensitivities caused by unilateral L5 spinal nerve ligation (SNL) in rats. Intrathecal injection of either MA or N-CDPCB diminished dose-dependently the SNL-induced mechanical allodynia, heat hyperalgesia, cold hyperalgesia, and spontaneous ongoing nociceptive responses in both development and maintenance periods, without altering acute/basal pain and locomotor function. Intrathecal MA also reduced the SNL-induced neuronal and astrocyte hyperactivities in the ipsilateral L5 dorsal horn. Mechanistically, intrathecal injection of these two inhibitors blocked the SNL-induced increase in the histone methyltransferase G9a expression and rescued the G9a-controlled downregulation of mu opioid receptor and Kv1.2 proteins in the ipsilateral L5 DRG. These findings further indicate the role of DRG FTO in neuropathic pain and suggest potential clinical application of the FTO inhibitors for management of this disorder. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
12. Meclofenamic acid represses spermatogonial proliferation through modulating m6A RNA modification
- Author
-
Tao Huang, Jiayin Guo, Yinghua Lv, Yi Zheng, Tongying Feng, Qiang Gao, and Wenxian Zeng
- Subjects
Cell cycle ,FTO ,Meclofenamic acid ,N6-methyladenosine ,Spermatogonial proliferation ,Animal culture ,SF1-1100 ,Veterinary medicine ,SF600-1100 - Abstract
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.
- Published
- 2019
- Full Text
- View/download PDF
13. Nonsteroidal Anti-Inflammatory Drugs
- Author
-
Graeme, Kimberlie A., Brent, Jeffrey, editor, Burkhart, Keith, editor, Dargan, Paul, editor, Hatten, Benjamin, editor, Megarbane, Bruno, editor, Palmer, Robert, editor, and White, Julian, editor
- Published
- 2017
- Full Text
- View/download PDF
14. Down-regulation of FTO promotes proliferation and migration, and protects bladder cancer cells from cisplatin-induced cytotoxicity.
- Author
-
Wen, Lijie, Pan, Xianwei, Yu, Yang, and Yang, Bo
- Subjects
BLADDER cancer ,CANCER cells ,TRANSITIONAL cell carcinoma ,CANCER cell migration ,OBESITY - Abstract
Background: FTO is known to be associated with body mass and obesity in humans and its over-expression affects the energy metabolism of cancer cells. The aim of the present study is to investigate the biological role of FTO in human bladder urothelial carcinoma.Methods: PCR and western blotting are used to measure the levels of FTO in both tissues and cell lines (5637, T24, TCCSUP) of human bladder urothelial carcinoma. Raw RNA-Sequencing reads and the corresponding clinical information for bladder urothelial carcinoma are downloaded from TCGA. Cell Counting Kit-8 and wound healing assays are used to explore the effect of FTO on proliferation and migration of bladder cancer cells.Results: The expression of FTO mRNA in bladder urothelial carcinoma decreases significantly compared with the normal controls from both the data of real-time PCR (p < 0.05) and TCGA (p < 0.01). Loss-of-function assays revealed that knockdown of FTO significantly promotes proliferation and migration of 5637 and T24 cells. Consistently, we found that the cisplatin-induced cytotoxicity of bladder cancer cell could be rescued by co-treatment with MA2, which was previously reported as a highly selective inhibitor of FTO, compared with the cisplatin-control group.Conclusions: These findings suggest that down-regulation of FTO plays an oncogenic role in bladder cancer. The further exploration of regulation of FTO expression may provide us a potential therapeutic target for the treatment of bladder cancer. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
15. Combinations of classical and non-classical voltage dependent potassium channel openers suppress nociceptor discharge and reverse chronic pain signs in a rat model of Gulf War illness
- Author
-
B Y, Cooper, T J, Nutter, L D, Flunker, and C M, Bowers
- Subjects
Dacarbazine ,Meclofenamic Acid ,Diclofenac ,Potassium Channels ,Ganglia, Spinal ,General Neuroscience ,Animals ,Nociceptors ,Persian Gulf Syndrome ,Chronic Pain ,Toxicology ,Rats - Abstract
In a companion paper we examined whether combinations of K
- Published
- 2022
- Full Text
- View/download PDF
16. Synergistic effect of a nonsteroidal anti-inflammatory drug in combination with topotecan on small cell lung cancer cells.
- Author
-
Yanar S, Kanli A, Kasap M, Bal Albayrak MG, Eskiler GG, and Ozkan AD
- Subjects
- Humans, Topotecan pharmacology, Neoplasm Recurrence, Local, Anti-Inflammatory Agents, Non-Steroidal, Meclofenamic Acid, Small Cell Lung Carcinoma drug therapy, Lung Neoplasms drug therapy
- Abstract
Background: The topoisomerase I inhibitor topotecan (TPT) is used in the treatment of recurrent small cell lung cancer (SCLC). However, the drug has a limited success rate and causes distress to patients due to its side effects, such as hematologic toxicities, including anemia and thrombocytopenia. Due to these pharmacokinetic limitations and undesirable side effects of chemotherapeutic drugs, the development of combination therapies has gained popularity in SCLC. Meclofenamic acid (MA), a nonsteroidal anti-inflammatory drug, has demonstrated anticancer effects on various types of cancers through different mechanisms. This study aims to investigate the potential synergistic effects of MA and TPT on the small cell lung cancer cell line DMS114., Methods and Results: To assess the cytotoxic and apoptotic effects of the combined treatment of MA and TPT, trypan blue exclusion assay, Annexin V, acridine orange/propidium iodide staining, western blot, and cell cycle analysis were conducted. The results demonstrated that the combination of MA and TPT elicited synergistic effects by enhancing toxicity in DMS114 cells (P < 0.01) without causing toxicity in healthy epithelial lung cells MRC5. The strongest synergistic effect was observed when the cells were treated with 60 µM MA and 10 nM TPT for 48 h (CI = 0,751; DRI = 10,871)., Conclusion: This study, for the first time, furnishes compelling evidence that MA and TPT synergistically reduce cellular proliferation and induce apoptosis in SCLC cells. Combinations of these drugs holds promise as a potential therapeutic strategy to improve efficacy and reduce the side effects associated with TPT., (© 2024. The Author(s), under exclusive licence to Springer Nature B.V.)
- Published
- 2024
- Full Text
- View/download PDF
17. Fenamates as Potential Therapeutics for Neurodegenerative Disorders
- Author
-
Jaunetta Hill and Nasser H. Zawia
- Subjects
tolfenamic acid ,mefenamic acid ,flufenamic acid ,meclofenamic acid ,fenamate ,NSAID ,Cytology ,QH573-671 - Abstract
Neurodegenerative disorders are desperately lacking treatment options. It is imperative that drug repurposing be considered in the fight against neurodegenerative diseases. Fenamates have been studied for efficacy in treating several neurodegenerative diseases. The purpose of this review is to comprehensively present the past and current research on fenamates in the context of neurodegenerative diseases with a special emphasis on tolfenamic acid and Alzheimer’s disease. Furthermore, this review discusses the major molecular pathways modulated by fenamates.
- Published
- 2021
- Full Text
- View/download PDF
18. Meclofenamic Acid Reduces Reactive Oxygen Species Accumulation and Apoptosis, Inhibits Excessive Autophagy, and Protects Hair Cell-Like HEI-OC1 Cells From Cisplatin-Induced Damage
- Author
-
He Li, Yongdong Song, Zuhong He, Xiaoyun Chen, Xianmin Wu, Xiaofei Li, Xiaohui Bai, Wenwen Liu, Boqin Li, Shanshan Wang, Yuechen Han, Lei Xu, Daogong Zhang, Jianfeng Li, Renjie Chai, Haibo Wang, and Zhaomin Fan
- Subjects
meclofenamic acid ,HEI-OC1 cells ,reactive oxygen species ,autophagy ,cisplatin ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Hearing loss is the most common sensory disorder in humans, and a significant number of cases is due to the ototoxicity of drugs such as cisplatin that cause hair cell (HC) damage. Thus, there is great interest in finding agents and mechanisms that protect HCs from ototoxic drug damage. It has been proposed that epigenetic modifications are related to inner ear development and play a significant role in HC protection and HC regeneration; however, whether the m6A modification and the ethyl ester form of meclofenamic acid (MA2), which is a highly selective inhibitor of FTO (fatmass and obesity-associated enzyme, one of the primary human demethylases), can affect the process of HC apoptosis induced by ototoxic drugs remains largely unexplored. In this study, we took advantage of the HEI-OC1 cell line, which is a cochlear HC-like cell line, to investigate the role of epigenetic modifications in cisplatin-induced cell death. We found that cisplatin injury caused reactive oxygen species accumulation and increased apoptosis in HEI-OC1 cells, and the cisplatin injury was reduced by co-treatment with MA2 compared to the cisplatin-only group. Further investigation showed that MA2 attenuated cisplatin-induced oxidative stress and apoptosis in HEI-OC1 cells. We next found that the cisplatin-induced upregulation of autophagy was significantly inhibited after MA2 treatment, indicating that MA2 inhibited the cisplatin-induced excessive autophagy. Our findings show that MA2 has a protective effect and improves the viability of HEI-OC1 cells after cisplatin treatment, and they provide new insights into potential therapeutic targets for the amelioration of cisplatin-induced ototoxicity.
- Published
- 2018
- Full Text
- View/download PDF
19. Histological changes caused by meclofenamic acid in androgen independent prostate cancer tumors: evaluation in a mouse model
- Author
-
Iván Delgado-Enciso, Alejandro D. Soriano-Hernández, Alejandrina Rodriguez-Hernandez, Héctor R. Galvan-Salazar, Daniel A. Montes-Galindo, Rafael Martinez-Martinez, Laura L. Valdez-Velazquez, Rafael Gonzalez-Alvarez, Francisco Espinoza-Gómez, Oscar A. Newton-Sanchez, Agustín Lara-Esqueda, and Jose Guzman-Esquivel
- Subjects
Prostatic Neoplasms ,Meclofenamic Acid ,Therapeutics ,Anti-Inflammatory Agents ,Diseases of the genitourinary system. Urology ,RC870-923 - Abstract
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.
- Published
- 2015
- Full Text
- View/download PDF
20. Two Binding Sites of SARS-CoV-2 Macrodomain 3 Probed by Oxaprozin and Meclomen
- Author
-
Jiao Li, Fumei Zhong, Mingwei Li, Yaqian Liu, Lei Wang, Mingqing Liu, Fudong Li, Jiahai Zhang, Jihui Wu, Yunyu Shi, Zhiyong Zhang, Xiaoming Tu, Ke Ruan, and Jia Gao
- Subjects
Meclofenamic Acid ,Binding Sites ,SARS-CoV-2 ,Oxaprozin ,Drug Discovery ,Molecular Medicine ,Humans ,Coronavirus Papain-Like Proteases ,Viral Nonstructural Proteins ,COVID-19 Drug Treatment - Abstract
Severe acute respiratory syndrome-coronavirus-1/2 (SARS-CoV-1/2) macrodomain 3 (Mac3) is critical for replication and transcription of the viral genome and is therefore a potential therapeutic target. Here, we solved the crystal structure of SARS-CoV-2 Mac3, which reveals a small-molecule binding pocket. Two low-molecular-weight drugs, oxaprozin and meclomen, induced different patterns of nuclear magnetic resonance (NMR) chemical shift perturbations (CSPs). Meclomen binds to site I of SARS-CoV-2 Mac3 with binding pose determined by NMR CSP and transferred paramagnetic relaxation enhancement, while oxaprozin binds to site II as revealed by the crystal structure. Interestingly, oxaprozin and meclomen both perturb residues in site I of SARS-CoV Mac3. Fluorescence polarization experiments further demonstrated that oxaprozin and meclomen inhibited the binding of DNA-G4s to SARS-CoV-2 Mac3. Our work identified two adjacent ligand-binding sites of SARS-CoV-2 Mac3 that shall facilitate structure-guided fragment linking of these compounds for more potent inhibitors.
- Published
- 2022
21. Simvastatin in combination with meclofenamic acid inhibits the proliferation and migration of human prostate cancer PC‑3 cells via an AKR1C3 mechanism.
- Author
-
Sekine, Yoshitaka, Nakayama, Hiroshi, Miyazawa, Yoshiyuki, Kato, Haruo, Furuya, Yosuke, Arai, Seiji, Koike, Hidekazu, Matsui, Hiroshi, Shibata, Yasuhiro, Ito, Kazuto, and Suzuki, Kazuhiro
- Subjects
- *
PROSTATE cancer & genetics , *SIMVASTATIN , *STATINS (Cardiovascular agents) , *PROSTATE cancer treatment , *COMBINATION drug therapy , *CANCER cell migration , *CANCER cell proliferation , *THERAPEUTICS - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
22. FTO (Fat-Mass and Obesity-Associated Protein) Participates in Hemorrhage-Induced Thalamic Pain by Stabilizing Toll-Like Receptor 4 Expression in Thalamic Neurons
- Author
-
Xiaozhou Feng, Sfian Albik, Minghui Cao, Alex Bekker, Yuan Xiang Tao, Tianfeng Huang, Shaogen Wu, Shibin Du, and Ganglan Fu
- Subjects
Advanced and Specialized Nursing ,0303 health sciences ,medicine.medical_specialty ,Toll-like receptor ,business.industry ,Thalamus ,nutritional and metabolic diseases ,medicine.disease ,Obesity ,Fat mass ,Meclofenamic acid ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Internal medicine ,Thalamic pain ,Microinjections ,medicine ,Neurology (clinical) ,Cardiology and Cardiovascular Medicine ,business ,030217 neurology & neurosurgery ,030304 developmental biology ,medicine.drug - Abstract
Background and Purpose: Hemorrhage-caused gene changes in the thalamus likely contribute to thalamic pain genesis. RNA N 6 -methyladenosine modification is an additional layer of gene regulation. Whether FTO (fat-mass and obesity-associated protein), an N 6 -methyladenosine demethylase, participates in hemorrhage-induced thalamic pain is unknown. Methods: Expression of Fto mRNA and protein was assessed in mouse thalamus after hemorrhage caused by microinjection of Coll IV (type IV collagenase) into unilateral thalamus. Effect of intraperitoneal administration of meclofenamic acid (a FTO inhibitor) or microinjection of adeno-associated virus 5 (AAV5) expressing Cre into the thalamus of Fto fl/fl mice on the Coll IV microinjection–induced TLR4 (Toll-like receptor 4) upregulation and nociceptive hypersensitivity was examined. Effect of thalamic microinjection of AAV5 expressing Fto (AAV5- Fto ) on basal thalamic TLR4 expression and nociceptive thresholds was also analyzed. Additionally, level of N 6 -methyladenosine in Tlr4 mRNA and its binding to FTO or YTHDF2 (YTH N 6 -methyladenosine RNA binding protein 2) were observed. Results: FTO was detected in neuronal nuclei of thalamus. Level of FTO protein, but not mRNA, was time-dependently increased in the ipsilateral thalamus on days 1 to 14 after Coll IV microinjection. Intraperitoneal injection of meclofenamic acid or adeno-associated virus-5 expressing Cre microinjection into Fto fl/fl mouse thalamus attenuated the Coll IV microinjection–induced TLR4 upregulation and tissue damage in the ipsilateral thalamus and development and maintenance of nociceptive hypersensitivities on the contralateral side. Thalamic microinjection of AAV5- Fto increased TLR4 expression and elicited hypersensitivities to mechanical, heat and cold stimuli. Mechanistically, Coll IV microinjection produced an increase in FTO binding to Tlr4 mRNA, an FTO-dependent loss of N 6 -methyladenosine sites in Tlr4 mRNA and a reduction in the binding of YTHDF2 to Tlr4 mRNA in the ipsilateral thalamus. Conclusions: Our findings suggest that FTO participates in hemorrhage-induced thalamic pain by stabilizing TLR4 upregulation in thalamic neurons. FTO may be a potential target for the treatment of this disorder.
- Published
- 2021
- Full Text
- View/download PDF
23. Investigation of the effect of meclofenamic acid on the proteome of LNCaP cells reveals changes in alternative polyadenylation and splicing machinery
- Author
-
Busra Sahinoz Saglam, Aylin Kanli, Sevinc Yanar, Murat Kasap, and Gurler Akpinar
- Subjects
Male ,Meclofenamic Acid ,Proteomics ,Cancer Research ,Proteome ,Oncology ,Humans ,Prostatic Neoplasms ,Hematology ,General Medicine ,Polyadenylation - 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.
- Published
- 2022
- Full Text
- View/download PDF
24. Meklofenamik asit vimentin ifadesini düzenleyerek LNCaP prostat kanseri hücrelerinin invazyon ve migrasyonunu inhibe eder
- Author
-
KANLI, Aylin and YANAR, Sevinc
- Subjects
Meclofenamic acid ,prostate cancer ,invasion ,migration ,vimentin ,Hücre Biyolojisi ,Biochemistry and Molecular Biology ,Medicine ,Biyokimya ve Moleküler Biyoloji ,Cell Biology ,Meklofenamik Asit ,prostat kanseri ,invazyon ,migrasyon ,Tıp - Abstract
Objectives: Despite various studies to improve the early detection and development of novel treatment agents, no significant progress has been made in the treatment of prostate cancer. Moreover, the metastasis of prostate carcinoma is a major challenge in treatment modalities. Therefore, in the present study, it was aimed to investigate the effect of meclofenamic acid (MA), a nonsteroidal anti-inflammatory drug, on the migration and invasion of LNCaP prostate carcinoma cells.Methods: Firstly, the non-toxic concentrations of MA on LNCaP cells were determined by trypan blue exclusion assay. After that, the effect of MA on migration and invasion was assessed by wound healing assay and matrigel invasion assay, respectively. Finally, the expression level of vimentin, which is a marker for epithelial-to-mesenchymal cell transition, was assessed by western blotting.Results: The results of trypan blue exclusion assay showed that 60 and 80 µM concentrations of MA were non-toxic to the cells. The migration rate of non-treated control cells was 44.4%, while it was significantly reduced to 12.9% and 2.9% in 60 and 80 µM MA-treated groups, respectively. When the cells were treated with 80 µM MA, the drug significantly reduced the invasion of LNCaP cells from 52% to 30.3%. Western blot results showed that the level of vimentin expression was significantly decreased in MA-treated cells.Conclusion: The study shows for the first time that MA inhibits the invasion and migration of prostate cancer LNCaP cells by decreasing the expression level of vimentin., Amaç: Prostat kanserinde erken teşhise ve yeni tedavi ajanlarının geliştirilmesine yönelik çeşitli çalışmalara rağmen, tedavide önemli bir ilerleme kaydedilememiştir. Ayrıca, prostat kanserinde metastaz tedavide büyük zorluk oluşturmaktadır. Bu nedenle, bu çalışmada nonsteroid antiinflamatuar bir ilaç olan meclofenamik asidin (MA) LNCaP prostat kanseri hücrelerinin migrasyonu ve invazyonu üzerine etkisinin araştırılması amaçlanmıştır.Yöntem: İlk olarak, MA'nın LNCaP hücrelerine toksik olmayan konsantrasyonları, tripan mavisi analizi ile belirlendi. Daha sonra, MA'nın migrasyon ve invazyon üzerindeki etkisi, sırasıyla yara iyileştirme analizi ve matrigel invazyon analizi ile değerlendirildi. Son olarak, epitelyal-mezenkimal hücre geçişi için bir belirteç olan vimentinin ifade düzeyi western blotlama ile tespit edildi.Bulgular: Tripan mavisi analizinin sonuçları, 60 ve 80 µM MA konsantrasyonlarının hücreler için toksik olmadığını göstermiştir. İlaç uygulanmamış kontrol hücrelerinin migrasyon oranı %44,4 iken, bu oran 60 ve 80 µM MA uygulanmış gruplarda istatistiksel olarak önemli ölçüde, sırasıyla, %12,9 ve %2,9'a düşmüştür. 80 µM MA uygulanan hücrelerde invazyon oranının %52'den %30.3'e düştüğü görülmüştür. Western blotlama sonuçları, MA verilen hücrelerde vimentin ifade seviyesinin önemli ölçüde azaldığını göstermiştir. Sonuç: Çalışma sonucunda, MA'nın vimentin ekspresyon seviyesini azaltarak LNCaP prostat kanseri hücrelerinin migrasyonu ve invazyonunu azalttığı ilk kez gösterilmiştir.
- Published
- 2022
25. The molecular and biological basis for COX-2 selectivity
- Author
-
Trummlitz, Guenter, van Ryn, Joanne, Warner, Timothy D., Parnham, Michael J., editor, Bruinvels, J., editor, Pairet, Michel, editor, and van Ryn, Joanne, editor
- Published
- 2004
- Full Text
- View/download PDF
26. Structural diversity of selective COX-2 inhibitors
- Author
-
Marnett, Lawrence J., Kalgutkar, Amit S., Parnham, Michael J., editor, Bruinvels, J., editor, Pairet, Michel, editor, and van Ryn, Joanne, editor
- Published
- 2004
- Full Text
- View/download PDF
27. Quantitative Automated Assays in Living Cells to Screen for Inhibitors of Hemichannel Function
- Author
-
Anaelle da Costa, Magda Mortier, Marie-Odile Fauvarque, Marjorie Comte, Franck Mouthon, Caroline Barette, Emmanuelle Soleilhac, Mathieu Charvériat, Laurence Aubry, Christèle Picoli, Genetics and Chemogenomics (GenChem ), Laboratoire de Biologie à Grande Échelle (BGE - UMR S1038), Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut de Recherche Interdisciplinaire de Grenoble (IRIG), Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Grenoble Alpes (UGA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut de Recherche Interdisciplinaire de Grenoble (IRIG), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Grenoble Alpes (UGA), Theranexus [Lyon], and Soleilhac, Emmanuelle
- Subjects
0301 basic medicine ,[SDV.BIO]Life Sciences [q-bio]/Biotechnology ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,Cell ,Gene Expression ,Biochemistry ,Analytical Chemistry ,0302 clinical medicine ,RNA, Small Interfering ,Benzoxazoles ,Chemistry ,Drug discovery ,Quinolinium Compounds ,Gap junction ,Transmembrane protein ,3. Good health ,Cell biology ,medicine.anatomical_structure ,[SDV.SP.PHARMA] Life Sciences [q-bio]/Pharmaceutical sciences/Pharmacology ,Carbenoxolone ,Molecular Medicine ,Biological Assay ,Neuroglia ,Intracellular ,Biotechnology ,Prescription Drugs ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Time-Lapse Imaging ,drug discovery ,high-content screening (HCS) ,central nervous system (CNS) ,03 medical and health sciences ,Bacterial Proteins ,Downregulation and upregulation ,Cell Line, Tumor ,Extracellular ,medicine ,Humans ,[SDV.BC] Life Sciences [q-bio]/Cellular Biology ,Fluorescent Dyes ,Automation, Laboratory ,Meclofenamic Acid ,hemichannel (HC) ,Cell Membrane ,[SDV.NEU.NB] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,Drugs, Investigational ,Iodides ,[SDV.BIO] Life Sciences [q-bio]/Biotechnology ,Luminescent Proteins ,030104 developmental biology ,Connexin 43 ,connexin (Cx) ,[SDV.SP.PHARMA]Life Sciences [q-bio]/Pharmaceutical sciences/Pharmacology ,Calcium ,sense organs ,030217 neurology & neurosurgery ,Function (biology) - Abstract
International audience; In vertebrates, intercellular communication is largely mediated by connexins (Cx), a family of structurally related transmembrane proteins that assemble to form hemichannels (HCs) at the plasma membrane. HCs are upregulated in different brain disorders and represent innovative therapeutic targets. Identifying modulators of Cx-based HCs is of great interest to better understand their function and define new treatments. In this study, we developed automated versions of two different cell-based assays to identify new pharmacological modulators of Cx43-HCs. As HCs remain mostly closed under physiological conditions in cell culture, depletion of extracellular Ca 2+ was used to increase the probability of opening of HCs. The first assay follows the incorporation of a fluorescent dye, Yo-Pro, by real-time imaging, while the second is based on the quenching of a fluorescent protein, YFP QL , by iodide after iodide uptake. These assays were then used to screen a collection of 2242 approved drugs and compounds under development. This study led to the identification of 11 candidate hits blocking Cx43-HC, active in the two assays, with 5 drugs active on HC but not on gap junction (GJ) activities. To our knowledge, this is the first screening on HC activity and our results suggest the potential of a new use of already approved drugs in central nervous system disorders with HC impairments.
- Published
- 2021
- Full Text
- View/download PDF
28. Anchoring Drugs to a Zinc(II) Coordination Polymer Network: Exploiting Structural Rationale toward the Design of Metallogels for Drug-Delivery Applications
- Author
-
Parthasarathi Dastidar and Protap Biswas
- Subjects
Naproxen ,Silver ,Mefenamic acid ,Polymers ,Coordination polymer ,Anti-Inflammatory Agents ,Metal Nanoparticles ,Microbial Sensitivity Tests ,010402 general chemistry ,01 natural sciences ,Inorganic Chemistry ,Mice ,chemistry.chemical_compound ,Coordination Complexes ,Cell Line, Tumor ,Klebsiella ,medicine ,Animals ,Molecule ,Carboxylate ,Physical and Theoretical Chemistry ,Drug Carriers ,Fenoprofen ,010405 organic chemistry ,Combinatorial chemistry ,Anti-Bacterial Agents ,0104 chemical sciences ,Meclofenamic acid ,Drug Liberation ,Zinc ,chemistry ,Drug delivery ,Drug Screening Assays, Antitumor ,Gels ,medicine.drug - Abstract
A new series of coordination polymers (CPs) were synthesized and crystallographically characterized by single-crystal X-ray diffraction with the aim of developing drug-delivery systems via metallogel formation. Structural rationale was employed to design such coordination-polymer-based metallogels. As many as nine CPs were obtained by reacting two bis(pyridyl)urea ligands, namely, 1,3-dipyridin-3-ylurea (3U) and 1,3-dipyridin-4-ylurea (4U), and the sodium salt of various nonsteroidal antiinflammatory drugs, namely, ibuprofen (IBU), naproxen (NAP), fenoprofen (FEN), diclofenac (DIC), meclofenamic acid (MEC), mefenamic acid (MEF), and Zn(NO3)2. All of the CPs displayed 1D polymeric chains that were self-assembled through various hydrogen-bonding interactions involving the urea N-H and carboxylate O atoms and, in a few cases, lattice-occluded water molecules. The reacting components of the CPs produced five metallogels in dimethyl sulfoxide/water. The gels were characterized by rheology and transmission electron microscopy. Three selected metallogelators, namely, 3UMEFg, 3UNAPg, and 3UMECg, showed in vitro anticancer, cell imaging, and multidrug delivery for antibacterial applications, respectively. The shear-thinning properties of 3UMECg (rheoreversibility and injectability) make it a potential candidate for plausible topical application.
- Published
- 2021
- Full Text
- View/download PDF
29. Soy Protein-Based Systems for Different Tissue Regeneration Applications
- Author
-
Vaz, C. M., de Graaf, L. A., Reis, R. L., Cunha, A. M., Reis, Rui L., editor, and Cohn, Daniel, editor
- Published
- 2002
- Full Text
- View/download PDF
30. Synthesis, anti-inflammatory activities and docking studies of amide derivatives of meclofenamic acid.
- Author
-
Narsinghani, Tamanna and Sharma, Rajesh
- Abstract
NSAIDs constitute a heterogeneous class of pharmacological agents widely prescribed for the treatment of inflammation, pain and edema, as well as osteoarthritis, rheumatoid arthritis and musculoskeletal disorders. This class of drugs has proved efficacious on account of their analgesic, anti-pyretic and anti-inflammatory activities, but gastrointestinal toxicity exists as the biggest problem associated with their chronic use. Many attempts have been made to structurally modify conventional NSAIDs as selective COX-2 inhibitors based on the old and still prevalent common belief that selective inhibition of COX-2 would provide safer NSAIDs. The present work thus focused on the synthesis of amide derivatives of one of the conventional non-selective NSAID, meclofenamic acid utilizing the one pot procedure involving a selective agent, bis (2-oxo-3-oxazolidinyl) phosphonic chloride. The synthesized compounds were tested for their in vivo inflammatory activity using carrageenan rat paw edema assay, and were subsequently docked on COX-2 PDB code 4COX to have better insights into their mechanism of action. The amide derivative with N-4-methoxybenzyl moiety (TSN4) proved to have anti-inflammatory potential (72.8%) better than meclofenamic acid (56.75%). This compound also docked with the highest dock score among the synthesized compounds and was found to have both hydrogen bonding with Arg120 and Tyr355 and hydrophobic interactions with Val349, Leu352, Ser353, Tyr385, Trp387, Met522, Val523, Ala527 and Ser530. N-4-methoxybenzyl amide derivative (TSN4) followed by benzyl amide derivative (TSN1) of meclofenamic acid were identified as potential anti-inflammatory compounds in both in vivo and in silico studies. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
31. Structure, Function and Inhibition of Cyclo-oxygenases
- Author
-
Marnett, L. J., Stock, G., editor, Lessl, M., editor, Serhan, C. N., editor, and Perez, H. D., editor
- Published
- 2000
- Full Text
- View/download PDF
32. Drug Photosensitivity
- Author
-
Santoianni, P., Procaccini, E. M., Katsambas, Andreas D., editor, and Lotti, Torello M., editor
- Published
- 2000
- Full Text
- View/download PDF
33. Significance of Multiple Bioactivation Pathways for Meclofenamate as Revealed through Modeling and Reaction Kinetics
- Author
-
Benjamin Mark Schleiff, Grover P. Miller, Sasin Payakachat, Dennis W. Province, Noah R. Flynn, Gunnar Boysen, S. Joshua Swamidass, Anna O. Pinson, and Mary A. Schleiff
- Subjects
Pharmaceutical Science ,Pharmacology ,030226 pharmacology & pharmacy ,Mass Spectrometry ,Activation, Metabolic ,Chemical kinetics ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Benzoquinones ,Humans ,Enzyme kinetics ,Meclofenamic Acid ,Nonsteroidal ,Human liver ,Chemistry ,Mechanism (biology) ,Anti-Inflammatory Agents, Non-Steroidal ,Articles ,Glutathione ,Spectrometry, Fluorescence ,Models, Chemical ,030220 oncology & carcinogenesis ,Reactive metabolite ,Microsomes, Liver ,Total metabolism ,Chromatography, Liquid - Abstract
Meclofenamate is a nonsteroidal anti-inflammatory drug used in the treatment of mild-to-moderate pain yet poses a rare risk of hepatotoxicity through an unknown mechanism. Nonsteroidal anti-inflammatory drug (NSAID) bioactivation is a common molecular initiating event for hepatotoxicity. Thus, we hypothesized a similar mechanism for meclofenamate and leveraged computational and experimental approaches to identify and characterize its bioactivation. Analyses employing our XenoNet model indicated possible pathways to meclofenamate bioactivation into 19 reactive metabolites subsequently trapped into glutathione adducts. We describe the first reported bioactivation kinetics for meclofenamate and relative importance of those pathways using human liver microsomes. The findings validated only four of the many bioactivation pathways predicted by modeling. For experimental studies, dansyl glutathione was a critical trap for reactive quinone metabolites and provided a way to characterize adduct structures by mass spectrometry and quantitate yields during reactions. Of the four quinone adducts, we were able to characterize structures for three of them. Based on kinetics, the most efficient bioactivation pathway led to the monohydroxy para-quinone-imine followed by the dechloro-ortho-quinone-imine. Two very inefficient pathways led to the dihydroxy ortho-quinone and a likely multiply adducted quinone. When taken together, bioactivation pathways for meclofenamate accounted for approximately 13% of total metabolism. In sum, XenoNet facilitated prediction of reactive metabolite structures, whereas quantitative experimental studies provided a tractable approach to validate actual bioactivation pathways for meclofenamate. Our results provide a foundation for assessing reactive metabolite load more accurately for future comparative studies with other NSAIDs and drugs in general. Significance Statement Meclofenamate bioactivation may initiate hepatotoxicity, yet common risk assessment approaches are often cumbersome and inefficient and yield qualitative insights that do not scale relative bioactivation risks. We developed and applied innovative computational modeling and quantitative kinetics to identify and validate meclofenamate bioactivation pathways and relevance as a function of time and concentration. This strategy yielded novel insights on meclofenamate bioactivation and provides a tractable approach to more accurately and efficiently assess other drug bioactivations and correlate risks to toxicological outcomes.
- Published
- 2020
- Full Text
- View/download PDF
34. Designing Metallogelators Derived from NSAID‐based Zn(II) Coordination Complexes for Drug‐Delivery Applications
- Author
-
Swapneswar Mondal and Parthasarathi Dastidar
- Subjects
Models, Molecular ,Naproxen ,Surface Properties ,Supramolecular chemistry ,Antineoplastic Agents ,Crystallography, X-Ray ,010402 general chemistry ,Crystal engineering ,01 natural sciences ,Biochemistry ,Drug Delivery Systems ,Cell Movement ,Coordination Complexes ,Tumor Cells, Cultured ,medicine ,Humans ,Particle Size ,Cell Proliferation ,Molecular Structure ,010405 organic chemistry ,Chemistry ,Anti-Inflammatory Agents, Non-Steroidal ,Organic Chemistry ,Synthon ,General Chemistry ,Ibuprofen ,Combinatorial chemistry ,0104 chemical sciences ,Meclofenamic acid ,Zinc ,Flufenamic acid ,Drug Design ,Drug delivery ,Drug Screening Assays, Antitumor ,Gels ,medicine.drug - Abstract
A crystal engineering approach has been invoked to design a new series of eight Zn(II) coordination complexes derived from various non-steroidal anti-inflammatory drugs (NSAIDs), namely diclofenac (DIC), ibuprofen (IBU), naproxen (NAP), flufenamic acid (FLU) and meclofenamic acid (MEC), and two co-ligands, namely N-phenyl-3-pyridylamide (3-Py) and N-phenyl-4-pyridylamide (4-Py), and Zn(NO3 )2 as potential supramolecular gelators. Half of the coordination complexes thus synthesized were able to form aqueous gels (MG-3-PyMEC, MG-3-PyDIC, MG-4-PyNAP and MG-4-PyMEC). Single-crystal structures of all eight complexes revealed that they possessed a gelation-inducing 1D hydrogen-bonded network including amide…amide synthon in some cases, which supported strongly the design principles based on which these complexes were synthesized. Interestingly, one such metallogelator complex, namely 3-PyMEC, showed an intriguing anticancer property against a human breast cancer cell line (MDA-MB-231), as revealed by both MTT and cell migration assays.
- Published
- 2020
- Full Text
- View/download PDF
35. FTO Inhibition Enhances the Antitumor Effect of Temozolomide by Targeting MYC-miR-155/23a Cluster-MXI1 Feedback Circuit in Glioma
- Author
-
Xiaodi Li, Peng Zhou, Chen Xie, Songshan Jiang, Jianwen Zhou, Zekun Mu, and Li Xiao
- Subjects
0301 basic medicine ,Cancer Research ,Alpha-Ketoglutarate-Dependent Dioxygenase FTO ,Mice, Nude ,medicine.disease_cause ,Proto-Oncogene Proteins c-myc ,miR-155 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Glioma ,Basic Helix-Loop-Helix Transcription Factors ,Temozolomide ,medicine ,Animals ,Humans ,Cyclooxygenase Inhibitors ,Antineoplastic Agents, Alkylating ,Cell Proliferation ,Feedback, Physiological ,Meclofenamic Acid ,biology ,Brain Neoplasms ,Chemistry ,Tumor Suppressor Proteins ,RNA ,Drug Synergism ,medicine.disease ,Meclofenamic acid ,MicroRNAs ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Demethylase ,Female ,Signal transduction ,Carcinogenesis ,Neoplasm Transplantation ,medicine.drug - Abstract
Malignant glioma constitutes one of the fatal primary brain tumors in adults. Such poor prognosis calls for a better understanding of cancer-related signaling pathways of this disease. Here we elucidate a MYC-miRNA-MXI1 feedback loop that regulates proliferation and tumorigenesis in glioma. MYC suppressed MXI1 expression via microRNA-155 (miR-155) and the microRNA-23a∼27a∼24-2 cluster (miR-23a cluster), whereas MXI1, in turn, inhibited MYC expression by binding to its promoter. Overexpression of miR-155 and the miR-23a cluster promoted tumorigenesis in U87 glioma cells. Furthermore, fat mass and obesity-associated protein (FTO), an N6-methyladenosine (m6A) RNA demethylase, regulated the loop by targeting MYC. The ethyl ester form of meclofenamic acid (MA2) inhibited FTO and enhanced the effect of the chemotherapy drug temozolomide on suppressing proliferation of glioma cells and negatively regulated the loop. These data collectively highlight a key regulatory circuit in glioma and provide potential targets for clinical treatment. Significance: These findings elucidate a novel feedback loop that regulates proliferation in glioma and can be targeted via inhibition of FTO to enhance the efficacy of temozolomide.
- Published
- 2020
- Full Text
- View/download PDF
36. Molecular determinants and therapeutic potential of focal ectopic activity: more than meets the Iti
- Author
-
Jordi Heijman, Dobromir Dobrev, Cardiologie, RS: Carim - H01 Clinical atrial fibrillation, and RS: Carim - H04 Arrhythmogenesis and cardiogenetics
- Subjects
Meclofenamic Acid ,Medizin ,Humans ,TRPM Cation Channels ,DELAYED AFTERDEPOLARIZATIONS ,Calcium ,Arrhythmias, Cardiac ,Cardiology and Cardiovascular Medicine - Published
- 2022
- Full Text
- View/download PDF
37. Phase I/II trial of meclofenamate in progressive MGMT-methylated glioblastoma under temozolomide second-line therapy-the MecMeth/NOA-24 trial
- Author
-
Zeyen, Thomas, Potthoff, Anna-Laura, Nemeth, Robert, Heiland, Dieter H., Burger, Michael C., Steinbach, Joachim P., Hau, Peter, Tabatabai, Ghazaleh, Glas, Martin, Schlegel, Uwe, Grauer, Oliver, Krex, Dietmar, Schnell, Oliver, Goldbrunner, Roland, Sabel, Michael, Thon, Niklas, Delev, Daniel, Clusmann, Hans, Seidel, Clemens, Güresir, Erdem, Schmid, Matthias, Schuss, Patrick, Giordano, Frank A., Radbruch, Alexander, Becker, Albert, Weller, Johannes, Schaub, Christina, Vatter, Hartmut, Schilling, Judith, Winkler, Frank, Herrlinger, Ulrich, and Schneider, Matthias
- Subjects
Meclofenamic Acid ,Medicine (General) ,Tumor Suppressor Proteins ,Medizin ,Study Protocol ,R5-920 ,DNA Repair Enzymes ,Temozolomide ,Humans ,Relapse ,Neoplasm Recurrence, Local ,Glioblastoma ,Second-line therapy ,Antineoplastic Agents, Alkylating ,DNA Modification Methylases ,Meclofenamate - Abstract
Trials 23, 57 (2022). doi:10.1186/s13063-021-05977-0, Published by BioMed Central, London
- Published
- 2022
- Full Text
- View/download PDF
38. Thyroid Hormone Transport
- Author
-
Stockigt, J. R., Lim, C.-F., Barlow, J. W., Topliss, D. J., Weetman, A. P., editor, and Grossman, A., editor
- Published
- 1997
- Full Text
- View/download PDF
39. X-Ray Structures and Thermal Analyses of New CD/Drug Inclusion Compounds
- Author
-
Caira, M. R., Griffith, V. J., Brown, G. R., Nassimbeni, L. R., Szejtli, J., editor, and Szente, L., editor
- Published
- 1996
- Full Text
- View/download PDF
40. New highly selective cyclooxygenase-2 inhibitors
- Author
-
Ford-Hutchinson, A. W., Bazan, Nicolas, editor, Botting, Jack, editor, and Vane, John, editor
- Published
- 1996
- Full Text
- View/download PDF
41. Selectively attacking tumor cells of Ru/Ir-arene complexes based on meclofenamic acid via cyclooxygenase-2 inhibition.
- Author
-
Huang Y, Lv M, Guo B, Hu G, Qian Y, Su Z, Xue X, and Liu HK
- Subjects
- Anti-Inflammatory Agents, Non-Steroidal pharmacology, Cyclooxygenase 2, Meclofenamic Acid, Humans, Antineoplastic Agents pharmacology, Coordination Complexes pharmacology, Ruthenium pharmacology
- Abstract
Breast cancer (BC) is one of the most common malignant tumors and often accompanied by inflammatory processes. Inflammation is an essential component of the tumor microenvironment, which might influence tumor proliferation and metastasis. Herein, three metal-arene complexes MA-bip-Ru, MA-bpy-Ir, and MA-bpy-Ru were prepared by tethering the non-steroidal anti-inflammatory drug meclofenamic acid (MA). Among them, MA-bip-Ru and MA-bpy-Ir showed lower cytotoxicity towards cancer cells, but MA-bpy-Ru showed significantly high selectivity and cytotoxicity towards MCF-7 cells through the autophagic pathway and exhibited no toxicity against normal HLF cells, showing potential for selective treatment of tumor cells. MA-bpy-Ru could also effectively destroy the 3D multicellular tumor spheroids, demonstrating its potential for clinical application. Besides, MA-bip-Ru, MA-bpy-Ir, and MA-bpy-Ru exhibited anti-inflammatory properties superior to MA, notably downregulating the expression of cyclooxygenase-2 (COX-2) and inhibiting the secretion of prostaglandin E2 in vitro . These findings demonstrated that MA-bpy-Ru was capable of intervening in inflammatory processes and showed the potential of MA-bpy-Ru to act as a selective anticancer agent, thus presenting a new mechanism of action for metal-arene complexes.
- Published
- 2023
- Full Text
- View/download PDF
42. Blockade of pathological retinal ganglion cell hyperactivity improves optogenetically evoked light responses in rd1 mice
- Author
-
John Martin Barrett, Patrick eDegenaar, and Evelyne eSernagor
- Subjects
Meclofenamic Acid ,Retinal Degeneration ,spontaneous activity ,optogenetics ,Retinal prosthesis ,Flupirtine ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Retinitis pigmentosa (RP) is a progressive retinal dystrophy that causes visual impairment and eventual blindness. Retinal prostheses are the best currently available vision-restoring treatment for RP, but only restore crude vision. One possible contributing factor to the poor quality of vision achieved with prosthetic devices is the pathological retinal ganglion cell (RGC) hyperactivity that occurs in photoreceptor dystrophic disorders. Gap junction blockade with meclofenamic acid (MFA) was recently shown to diminish RGC hyperactivity and improve the signal-to-noise ratio (SNR) of RGC responses to light flashes and electrical stimulation in the rd10 mouse model of RP. We sought to extend these results to spatiotemporally patterned optogenetic stimulation in the faster-degenerating rd1 model and compare the effectiveness of a number of drugs known to disrupt rd1 hyperactivity.We crossed rd1 mice with a transgenic mouse line expressing the light-sensitive cation channel channelrhodopsin2 (ChR2) in RGCs, allowing them to be stimulated directly using high-intensity blue light. We used 60-channel ITO multielectrode arrays to record ChR2-mediated RGC responses from wholemount, ex-vivo retinas to full-field and patterned stimuli before and after application of MFA, 18-ß-glycyrrhetinic acid (18BGA, another gap junction blocker) or flupirtine (Flu, a Kv7 potassium channel opener). All three drugs decreased spontaneous RGC firing, but 18BGA and Flu also decreased the sensitivity of RGCs to optogenetic stimulation. Nevertheless, all three drugs improved the SNR of ChR2-mediated responses. MFA also made it easier to discern motion direction of a moving bar from RGC population responses.Our results support the hypothesis that reduction of pathological RGC spontaneous activity characteristic in retinal degenerative disorders may improve the quality of visual responses in retinal prostheses and they provide insights into how best to achieve this for optogenetic prostheses.
- Published
- 2015
- Full Text
- View/download PDF
43. Prostanoids, Prostanoid Receptors and Tumor Progression
- Author
-
Coleman, R. A., Nigam, Santosh, editor, Honn, Kenneth V., editor, Marnett, Lawrence J., editor, and Walden, Thomas L., Jr., editor
- Published
- 1993
- Full Text
- View/download PDF
44. Mefenamic acid in combination with ribavirin shows significant effects in reducing chikungunya virus infection in vitro and in vivo.
- Author
-
Rothan, Hussin A., Bahrani, Hirbod, Abdulrahman, Ammar Y., Mohamed, Zulqarnain, Teoh, Teow Chong, Othman, Shatrah, Rashid, Nurshamimi Nor, Rahman, Noorsaadah A., and Yusof, Rohana
- Subjects
- *
MEFENAMIC acid , *RIBAVIRIN , *CHIKUNGUNYA , *AEDES aegypti , *AEDES albopictus , *IN vitro studies - Abstract
Chikungunya virus (CHIKV) infection is a persistent problem worldwide due to efficient adaptation of the viral vectors, Aedes aegypti and Aedes albopictus mosquitoes. Therefore, the absence of effective anti-CHIKV drugs to combat chikungunya outbreaks often leads to a significant impact on public health care. In this study, we investigated the antiviral activity of drugs that are used to alleviate infection symptoms, namely, the non-steroidal anti-inflammatory drugs (NSAIDs), on the premise that active compounds with potential antiviral and anti-inflammatory activities could be directly subjected for human use to treat CHIKV infections. Amongst the various NSAID compounds, Mefenamic acid (MEFE) and Meclofenamic acid (MECLO) showed considerable antiviral activity against viral replication individually or in combination with the common antiviral drug, Ribavirin (RIBA). The 50% effective concentration (EC 50 ) was estimated to be 13 μM for MEFE, 18 μM for MECLO and 10 μM for RIBA, while MEFE + RIBA (1:1) exhibited an EC 50 of 3 μM, and MECLO + RIBA (1:1) was 5 μM. Because MEFE is commercially available and its synthesis is easier compared with MECLO, MEFE was selected for further in vivo antiviral activity analysis. Treatment with MEFE + RIBA resulted in a significant reduction of hypertrophic effects by CHIKV on the mouse liver and spleen. Viral titre quantification in the blood of CHIKV-infected mice through the plaque formation assay revealed that treatment with MEFE + RIBA exhibited a 6.5-fold reduction compared with untreated controls. In conclusion, our study demonstrated that MEFE in combination with RIBA exhibited significant anti-CHIKV activity by impairing viral replication in vitro and in vivo . Indeed, this finding may lead to an even broader application of these combinatorial treatments against other viral infections. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
45. Metabolism of non-steroidal anti-inflammatory drugs (NSAIDs) by Streptomyces griseolus CYP105A1 and its variants
- Author
-
Yuya Yogo, Kaori Yasuda, Teisuke Takita, Kiyoshi Yasukawa, Yuichiro Iwai, Miyu Nishikawa, Hiroshi Sugimoto, Shinichi Ikushiro, and Toshiyuki Sakaki
- Subjects
Pharmacology ,Meclofenamic Acid ,Mefenamic Acid ,Diclofenac ,Bacterial Proteins ,Cytochrome P-450 Enzyme System ,Anti-Inflammatory Agents, Non-Steroidal ,Pharmaceutical Science ,Humans ,Pharmacology (medical) ,Ibuprofen ,Streptomyces ,Flufenamic Acid - Abstract
In the field of drug development, technology for producing human metabolites at a low cost is required. In this study, we explored the possibility of using prokaryotic water-soluble cytochrome P450 (CYP) to produce human metabolites. Streptomyces griseolus CYP105A1 metabolizes various non-steroidal anti-inflammatory drugs (NSAIDs), including diclofenac, mefenamic acid, flufenamic acid, tolfenamic acid, meclofenamic acid, and ibuprofen. CYP105A1 showed 4'-hydroxylation activity towards diclofenac, mefenamic acid, flufenamic acid, tolfenamic acid, and meclofenamic acid. It should be noted that this reaction specificity was similar to that of human CYP2C9. In the case of mefenamic acid, another metabolite, 3'-hydroxymethyl mefenamic acid, was detected as a major metabolite. Substitution of Arg at position 73 with Ala in CYP105A1 dramatically reduced the hydroxylation activity toward diclofenac, flufenamic acid, and ibuprofen, indicating that Arg73 is essential for the hydroxylation of these substrates. In contrast, substitution of Arg84 with Ala remarkably increased the hydroxylation activity towards diclofenac, mefenamic acid, and flufenamic acid. Recombinant Rhodococcus erythrocyte cells expressing the CYP105A1 variant R84A/M239A showed complete conversion of diclofenac into 4'-hydroxydiclofenac. These results suggest the usefulness of recombinant R. erythropolis cells expressing actinomycete CYP, such as CYP105A1, for the production of human drug metabolites.
- Published
- 2021
46. TRPM4 inhibition by meclofenamate suppresses Ca2+-dependent triggered arrhythmias
- Author
-
Frone Vandewiele, Andy Pironet, Griet Jacobs, Miklos Kecskés, Jörg Wegener, Sara Kerselaers, Lio Hendrikx, Joren Verelst, Koenraad Philippaert, Wouter Oosterlinck, Andrei Segal, Evy Van Den Broeck, Silvia Pinto, Silvia G Priori, Stephan E Lehnart, Bernd Nilius, Thomas Voets, and Rudi Vennekens
- Subjects
Meclofenamic Acid ,Mice ,Tachycardia, Ventricular ,Animals ,TRPM Cation Channels ,Calcium ,Arrhythmias, Cardiac ,Myocytes, Cardiac ,Cardiology and Cardiovascular Medicine - Abstract
Aims Cardiac arrhythmias are a major factor in the occurrence of morbidity and sudden death in patients with cardiovascular disease. Disturbances of Ca2+ homeostasis in the heart contribute to the initiation and maintenance of cardiac arrhythmias. Extrasystolic increases in intracellular Ca2+ lead to delayed afterdepolarizations and triggered activity, which can result in heart rhythm abnormalities. It is being suggested that the Ca2+-activated nonselective cation channel TRPM4 is involved in the aetiology of triggered activity, but the exact contribution and in vivo significance are still unclear. Methods and results In vitro electrophysiological and calcium imaging technique as well as in vivo intracardiac and telemetric electrocardiogram measurements in physiological and pathophysiological conditions were performed. In two distinct Ca2+-dependent proarrhythmic models, freely moving Trpm4−/− mice displayed a reduced burden of cardiac arrhythmias. Looking further into the specific contribution of TRPM4 to the cellular mechanism of arrhythmias, TRPM4 was found to contribute to a long-lasting Ca2+ overload-induced background current, thereby regulating cell excitability in Ca2+ overload conditions. To expand these results, a compound screening revealed meclofenamate as a potent antagonist of TRPM4. In line with the findings from Trpm4−/− mice, 10 µM meclofenamate inhibited the Ca2+ overload-induced background current in ventricular cardiomyocytes and 15 mg/kg meclofenamate suppressed catecholaminergic polymorphic ventricular tachycardia-associated arrhythmias in a TRPM4-dependent manner. Conclusion The presented data establish that TRPM4 represents a novel target in the prevention and treatment of Ca2+-dependent triggered arrhythmias.
- Published
- 2021
47. Silencing glioblastoma networks to make temozolomide more effective
- Author
-
Frank Winkler
- Subjects
Meclofenamic Acid ,Cancer Research ,Temozolomide ,business.industry ,Brain Neoplasms ,Editorials ,In Vitro Techniques ,medicine.disease ,Dacarbazine ,Oncology ,Cell Line, Tumor ,Basic and Translational Investigations ,Cancer research ,Medicine ,Gene silencing ,Humans ,Neurology (clinical) ,business ,Glioblastoma ,Antineoplastic Agents, Alkylating ,medicine.drug ,Cell Proliferation - Abstract
BACKGROUND: Glioblastoma cells assemble to a syncytial communicating network based on tumor microtubes (TMs) as ultra-long membrane protrusions. The relationship between network architecture and transcriptional profile remains poorly investigated. Drugs that interfere with this syncytial connectivity such as meclofenamate (MFA) may be highly attractive for glioblastoma therapy. METHODS: In a human neocortical slice model using glioblastoma cell populations of different transcriptional signatures, three-dimensional tumor networks were reconstructed, and TM-based intercellular connectivity was mapped on the basis of two-photon imaging data. MFA was used to modulate morphological and functional connectivity; downstream effects of MFA treatment were investigated by RNA sequencing and fluorescence-activated cell sorting (FACS) analysis. RESULTS: TM-based network morphology strongly differed between the transcriptional cellular subtypes of glioblastoma and was dependent on axon guidance molecule expression. MFA revealed both a functional and morphological demolishment of glioblastoma network architectures which was reflected by a reduction of TM-mediated intercellular cytosolic traffic as well as a breakdown of TM length. RNA sequencing confirmed a downregulation of NCAM and axon guidance molecule signaling upon MFA treatment. Loss of glioblastoma communicating networks was accompanied by a failure in the upregulation of genes that are required for DNA repair in response to temozolomide (TMZ) treatment and culminated in profound treatment response to TMZ-mediated toxicity. CONCLUSION: The capacity of TM formation reflects transcriptional cellular heterogeneity. MFA effectively demolishes functional and morphological TM-based syncytial network architectures. These findings might pave the way to a clinical implementation of MFA as a TM-targeted therapeutic approach.
- Published
- 2021
48. Biochemistry and Pharmacology of Prostacyclin
- Author
-
Bönner, Gerd, Rahn, Karl-Heinz, Bönner, Gerd, and Rahn, Karl-Heinz
- Published
- 1990
- Full Text
- View/download PDF
49. Meclofenamic acid promotes cisplatin-induced acute kidney injury by inhibiting fat mass and obesity-associated protein-mediated m6A abrogation in RNA
- Author
-
Qingqing Xu, Ming Wu, Peihui Zhou, Li Wang, and Chaoyang Ye
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Alpha-Ketoglutarate-Dependent Dioxygenase FTO ,Apoptosis ,urologic and male genital diseases ,Biochemistry ,Cell Line ,Mice ,03 medical and health sciences ,In vivo ,Internal medicine ,medicine ,Animals ,Humans ,Molecular Biology ,Meclofenamic Acid ,Cisplatin ,Messenger RNA ,Gene knockdown ,Kidney ,030102 biochemistry & molecular biology ,Chemistry ,Acute kidney injury ,nutritional and metabolic diseases ,Molecular Bases of Disease ,Drug Synergism ,Cell Biology ,Acute Kidney Injury ,medicine.disease ,Mice, Inbred C57BL ,Meclofenamic acid ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Adipose Tissue ,Gene Expression Regulation ,Gene Knockdown Techniques ,RNA ,Tumor Suppressor Protein p53 ,medicine.drug - Abstract
The role of RNA methylation on the sixth N atom of adenylate (m(6)A) in acute kidney injury (AKI) is unknown. FTO (fat mass and obesity-associated protein) reverses the m(6)A modification in cisplatin-induced AKI. Here, we aimed to determine FTO's role in AKI. We induced AKI in c57BL/6 mice by intraperitoneal cisplatin injection and treated the animal with vehicle or an FTO inhibitor meclofenamic acid (MA) for 3 days. Moreover, as an in vitro model, human kidney proximal tubular cells (HK2 cells) were treated with cisplatin. We found that the cisplatin treatment reduces FTO expression and increases m(6)A levels in vivo and in vitro. MA aggravated renal damage and increased apoptosis in cisplatin-treated kidneys, phenotypes that were correlated with reduced FTO expression and increased m(6)A levels. Moreover, MA promoted apoptosis in cisplatin-treated HK2 cells, which was correlated with the reduced FTO expression and increased m(6)A in HK2 cells. FTO protein overexpression reduced m(6)A levels and inhibited apoptosis in cisplatin-treated HK2 cells and also blocked the MA-induced increase in m(6)A levels and apoptosis rates. In agreement, overexpression of the m(6)A-generating methyltransferase-like 3 and 14 (METTL3 and METTL14) or siRNA-mediated FTO knockdown promoted apoptosis and enhanced m(6)A levels in cisplatin-treated HK2 cells. MA increased p53 mRNA and protein levels in AKI both in vitro and in vivo, and FTO overexpression reduced p53 expression and reversed the MA-induced p53 increase in AKI. In conclusion, reduced renal FTO expression in cisplatin-induced AKI increases RNA m(6)A levels and aggravates renal damages.
- Published
- 2019
- Full Text
- View/download PDF
50. Multiple actions of fenamates and other nonsteroidal anti-inflammatory drugs on GABAA receptors
- Author
-
Salla Mansikkamäki, Saku T. Sinkkonen, Esa R. Korpi, Hartmut Lüddens, Department of Pharmacology, University of Helsinki, Faculty of Medicine, Korva-, nenä- ja kurkkutautien klinikka, HUS Head and Neck Center, Clinicum, Department of Ophthalmology and Otorhinolaryngology, and Esa Risto Korpi / Principal Investigator
- Subjects
MECHANISM ,0301 basic medicine ,NSAID drugs ,Mefenamic acid ,Allosteric regulation ,Pharmacology ,BINDING-SITES ,GABA ,03 medical and health sciences ,0302 clinical medicine ,Tolfenamic acid ,Niflumic acid ,medicine ,SHIFT ,MODULATION ,Receptor ,Xenopus oocytes ,AGENT ,Chemistry ,GABAA receptor ,ANION GRADIENT ,A RECEPTORS ,SUBUNITS ,3. Good health ,Meclofenamic acid ,Fenamates ,030104 developmental biology ,Flufenamic acid ,317 Pharmacy ,ACID ,Autoradiography ,030217 neurology & neurosurgery ,Recombinant GABA(A) receptors ,RESPONSES ,medicine.drug - Abstract
The nonsteroidal anti-inflammatory drug (NSAID) niflumic acid, a fenamate in structure, has many molecular targets, one of them being specific subtypes of the main inhibitory ligand-gated anion channel, the GABA(A) receptor. Here, we report on the effects of other fenamates and other classes of NSAIDs on brain picrotoxinin-sensitive GABA A receptors, using an autoradiographic assay with [S-35]TBPS as a ligand on mouse brain sections. We found that the other fenamates studied (flufenamic acid, meclofenamic acid, mefenamic acid and tolfenamic acid) affected the autoradiographic signal at low micromolar concentrations in a facilitatory-like allosteric fashion, i.e., without having affinity to the [S-35]TBPS binding site. Unlike niflumic acid that shows clear preference for inhibiting cerebellar granule cell layer GABA(A) receptors, the other fenamates showed little brain regional selectivity, indicating that their actions are not receptor-subtype selective. Of the non-fenamate NSAIDs studied at 100 mu M concentration, diclofenac induced the greatest inhibition of the binding, which is not surprising as it has close structural similarity with the potent fenamate meclofenamic acid. Using two-electrode voltage-clamp assays on Xenopus oocytes, the effect of niflumic acid was found to be dependent on the beta subunit variant and the presence of gamma 2 subunit in rat recombinant alpha 1 beta and alpha 1 beta gamma 2 GABA(A) receptors, with the beta 1 allowing the niflumic acid inhibition and beta 3 the stimulation of the receptor-mediated currents. In summary, the fenamate NSAID5 constitute an interesting class of compounds that could be used for development of potent GABA(A) receptor allosteric agonists with other targets to moderate inflammation, pain and associated anxiety/depression.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.