1
|
El-Gendy ZA, Ammar NM, Kassem AM, Attia MS, Afifi SM, Ibrahim AH, Emam SE, Ms Korany R, El-Nasser G El-Gendy A, Elshamy AI. Myricetin-loaded SBA-15 silica nanoparticles for enhanced management of pyrexia, pain, and inflammation through modulation of MAPK/NF-κB and COX-2/PGE-2 pathways: Evidence from the biochemical, histological, and metabolomic analysis. Int J Pharm 2024; 666:124775. [PMID: 39353498 DOI: 10.1016/j.ijpharm.2024.124775] [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: 08/08/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
Myricetin (MYR) is a natural flavonoid that has several biological functions. However, some of its beneficial effects are diminished due to low water solubility, stability, and bioavailability. Herein, several kinds of silica nanoparticles (MCM-41 and SBA-15) were loaded with MYR to improve its biological activity as an analgesic, antipyretic, and anti-inflammatory component, thereby overcoming its drawbacks. The nanoparticles (MYR@SBA-15) were formulated optimally, transforming MYR into an amorphous state. This transformation was confirmed via several strategies, including differential scanning calorimetry, Fourier transform infrared spectroscopy, and powder x-ray diffraction. As a result, there was a significant enhancement in the solubility and rate of dissolution in water. The anti-inflammatory benefits as an innovative strategy and the underlying mechanism of action of MYR and its SBA-15 silica nanoparticles (MYR@SBA-15) were investigated based on the biochemical, histological, immunohistochemical, and metabolomic assays alongside their antipyretic and analgesic characteristics. Compared to the usage of raw MYR, the administration of MYR@SBA-15 at doses of 25, 50, and 100 mg/kg significantly decreases pain perception by inhibiting the body's writhing motions induced by acetic acid. Furthermore, it helps regulate increased body temperature caused by baking yeast and effectively stabilizes it. It reduces the release of NO and PGE-2 in a concentration-dependent manner by down-regulating iNOS and COX-2 expression in the inflammatory model. MYR and MYR@SBA-15 also inhibit the nuclear translocation of NF-κB, downregulate the expression of mitogen-activated protein kinases (MAPKs), such as p38, ERK1/2, and JNK protein, and reduce the generation of proinflammatory cytokines, such as TNF-α. In addition, inflammatory cardinal signs like paw edema caused by carrageenan in rats are greatly suppressed by MYR and MYR@SBA-15 treatment when compared to the untreated group. More noteworthy outcomes are shown in the MYR@SBA-15, particularly at a dose of 100 mg/kg. These results of biochemical and immuno-histochemistry suggest that MYR@SBA-15 may be a useful analgesic antipyretic and may also help reduce inflammation by altering MAPKs/NF-κB and COX-2/PGE-2 signaling cascades. Serum metabolomics study demonstrated modifications in various low molecular weight metabolites with arthritis development. These metabolite levels were restored to normal when MYR@SBA-15 was administered via modulating several metabolic pathways, i.e., pyrimidine, energy metabolism, and proteins. Overall, MYR-loaded SBA-15 silica nanoparticles have demonstrated significant promise in enhancing the disturbed metaboloic pathways and providing a substantial capacity to regulate several oxidative stress and inflammatory mediators.
Collapse
Affiliation(s)
- Zeinab A El-Gendy
- Pharmacology Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Naglaa M Ammar
- Therapeutic Chemistry Department, National Research Centre, Dokki, Giza 12622, Egypt
| | - Abdulsalam M Kassem
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11751, Cairo, Egypt
| | - Mohamed S Attia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Sherif M Afifi
- Department for Life Quality Studies, Rimini Campus, University of Bologna, Corso d'Augusto 237, 47921 Rimini, Italy
| | - Ahmed H Ibrahim
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11751, Cairo, Egypt
| | - Sherif E Emam
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Reda Ms Korany
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Abd El-Nasser G El-Gendy
- Medicinal and Aromatic Plants Research Department, National Research Centre, Dokki, Giza 12622, Egypt
| | - Abdelsamed I Elshamy
- Department of Natural Compounds Chemistry, National Research Centre, Dokki, Giza 12622, Egypt.
| |
Collapse
|
2
|
Famurewa AC, George MY, Ukwubile CA, Kumar S, Kamal MV, Belle VS, Othman EM, Pai SRK. Trace elements and metal nanoparticles: mechanistic approaches to mitigating chemotherapy-induced toxicity-a review of literature evidence. Biometals 2024; 37:1325-1378. [PMID: 39347848 DOI: 10.1007/s10534-024-00637-7] [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: 04/15/2024] [Accepted: 08/30/2024] [Indexed: 10/01/2024]
Abstract
Anticancer chemotherapy (ACT) remains a cornerstone in cancer treatment, despite significant advances in pharmacology over recent decades. However, its associated side effect toxicity continues to pose a major concern for both oncology clinicians and patients, significantly impacting treatment protocols and patient quality of life. Current clinical strategies to mitigate ACT-induced toxicity have proven largely unsatisfactory, leaving a critical unmet need to block toxicity mechanisms without diminishing ACT's therapeutic efficacy. This review aims to document the molecular mechanisms underlying ACT toxicity and highlight research efforts exploring the protective effects of trace elements (TEs) and their nanoparticles (NPs) against these mechanisms. Our literature review reveals that the primary driver of ACT toxicity is redox imbalance, which triggers oxidative inflammation, apoptosis, endoplasmic reticulum stress, mitochondrial dysfunction, autophagy, and dysregulation of signaling pathways such as PI3K/mTOR/Akt. Studies suggest that TEs, including zinc, selenium, boron, manganese, and molybdenum, and their NPs, can potentially counteract ACT-induced toxicity by inhibiting oxidative stress-mediated pathways, including NF-κB/TLR4/MAPK/NLRP3, STAT-3/NLRP3, Bcl-2/Bid/p53/caspases, and LC3/Beclin-1/CHOP/ATG6, while also upregulating protective signaling pathways like Sirt1/PPAR-γ/PGC-1α/FOXO-3 and Nrf2/HO-1/ARE. However, evidence regarding the roles of lncRNA and the Wnt/β-catenin pathway in ACT toxicity remains inconsistent, and the impact of TEs and NPs on ACT efficacy is not fully understood. Further research is needed to confirm the protective effects of TEs and their NPs against ACT toxicity in cancer patients. In summary, TEs and their NPs present a promising avenue as adjuvant agents for preventing non-target organ toxicity induced by ACT.
Collapse
Affiliation(s)
- Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex Ekwueme Federal University Ndufu-Alike Ikwo, Abakaliki, Ebonyi, Nigeria.
- Centre for Natural Products Discovery, School of P harmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK.
- Department of Pharmacology, Manipal College of Pharmaceutical Science, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Cletus A Ukwubile
- Department of Pharmacognosy, Faculty of Pharmacy, University of Maiduguri, Bama Road, Maiduguri, Borno, Nigeria
| | - Sachindra Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Science, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Mehta V Kamal
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Vijetha S Belle
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Eman M Othman
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
- Cancer Therapy Research Center, Department of Biochemistry-I, Biocenter, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
- Department of Bioinformatics, University of Würzburg, Am Hubland, 97074, BiocenterWürzburg, Germany
| | - Sreedhara Ranganath K Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Science, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| |
Collapse
|
3
|
Li J, Luo T, Zhao Y, Wang D, Jin Y, Wu Z, Yang G, Qi X. Cardioprotective potentials of myricetin on doxorubicin-induced cardiotoxicity based on biochemical and transcriptomic analysis. Biomed Pharmacother 2024; 175:116748. [PMID: 38776683 DOI: 10.1016/j.biopha.2024.116748] [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: 02/05/2024] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Doxorubicin (DOX) is a commonly used anthracycline in cancer chemotherapy. The clinical application of DOX is constrained by its cardiotoxicity. Myricetin (MYR) is a natural flavonoid widely present in many plants with antioxidant and anti-inflammatory properties. However, MYR's beneficial effects and mechanisms in alleviating DOX-induced cardiotoxicity (DIC) remain unknown. C57BL/6 mice were injected with 15 mg/kg of DOX to establish the DIC, and MYR solutions were administrated by gavage to investigate its cardioprotective potentials. Histopathological analysis, physiological indicators assessment, transcriptomics analysis, and RT-qPCR were used to elucidate the potential mechanism of MYR in DIC treatment. MYR reduced cardiac injury produced by DOX, decreased levels of cTnI, AST, LDH, and BNP, and improved myocardial injury and fibrosis. MYR effectively prevented DOX-induced oxidative stress, such as lowered MDA levels and elevated SOD, CAT, and GSH activities. MYR effectively suppressed NLRP3 and ASC gene expression levels to inhibit pyroptosis while regulating Caspase1 and Bax levels to reduce cardiac cell apoptosis. According to the transcriptomic analysis, glucose and fatty acid metabolism were associated with differential gene expression. KEGG pathway analysis revealed differential gene enrichment in PPAR and AMPK pathways, among others. Following validation, MYR was found to alleviate DIC by regulating glycolipid metabolism and AMPK pathway-related genes. Our findings demonstrated that MYR could mitigate DIC by regulating the processes of oxidative stress, apoptosis, and pyroptosis. MYR is critical in improving DOX-induced myocardial energy metabolism abnormalities mediated by the AMPK signaling pathway. In conclusion, MYR holds promise as a therapeutic strategy for DIC.
Collapse
Affiliation(s)
- Jaili Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, Zhejiang Province 315832, China; State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang Province 310021, China
| | - Ting Luo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, Zhejiang Province 315832, China; State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang Province 310021, China.
| | - Yao Zhao
- Xianghu Laboratory, Hangzhou, Zhejiang Province 311231, China
| | - Dou Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, Zhejiang Province 315832, China; State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang Province 310021, China; Key Laboratory of Traceability for Agricultural Genetically Modified Organisms, Ministry of Agriculture and Rural Affairs, China
| | - Yuanxiang Jin
- Xianghu Laboratory, Hangzhou, Zhejiang Province 311231, China; College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang Province 310032, China
| | - Zufang Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, Zhejiang Province 315832, China.
| | - Guiling Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, Zhejiang Province 315832, China; State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang Province 310021, China; Xianghu Laboratory, Hangzhou, Zhejiang Province 311231, China.
| | - Xingjiang Qi
- Xianghu Laboratory, Hangzhou, Zhejiang Province 311231, China.
| |
Collapse
|
4
|
Wang J, Hu Y, Li Q, Liu YN, Lin J, Xu RA. Effects of myricetin and quercetin on ticagrelor metabolism and the underlying mechanism. Chem Biol Interact 2024; 392:110924. [PMID: 38401715 DOI: 10.1016/j.cbi.2024.110924] [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: 07/21/2023] [Revised: 01/25/2024] [Accepted: 02/18/2024] [Indexed: 02/26/2024]
Abstract
The aim of this study was to investigate the potential drug-drug interactions (DDIs) between ticagrelor and other drugs as well as their underlying mechanisms. Rat liver microsome (RLM) reaction system was used to screen potential DDIs in vitro, and ultra performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) was applied to detect the levels of ticagrelor and AR-C124910XX, the main metabolite of ticagrelor. A total of 68 drugs were screened, 11 of which inhibited the production of AR-C124910XX to 20% or less, especially two flavonoids (myricetin and quercetin). The half-maximal inhibitory concentration (IC50) of myricetin on ticagrelor was 11.51 ± 0.28 μM in RLM and 17.96 ± 0.54 μM in human liver microsome (HLM). The IC50 of quercetin in inhibiting ticagrelor in RLM and HLM was 16.92 ± 0.49 μM and 60.15 ± 0.43 μM, respectively. They all inhibited the metabolism of ticagrelor through a mixed mechanism. In addition, Sprague-Dawley (SD) rats were used to study the interactions of ticagrelor with selected drugs in vivo. We found that the main pharmacokinetic parameters including AUC (0-t), AUC (0-∞) and Cmax of ticagrelor were significantly increased when ticagrelor was combined with these two flavonoids. Our results suggested that myricetin and quercetin of flavonoids both had significant effects on the metabolism of ticagrelor, providing reference data for the clinical individualized medication of ticagrelor.
Collapse
Affiliation(s)
- Jing Wang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yingying Hu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qingqing Li
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ya-Nan Liu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingjing Lin
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Ren-Ai Xu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
5
|
Nagy A, Börzsei D, Hoffmann A, Török S, Veszelka M, Almási N, Varga C, Szabó R. A Comprehensive Overview on Chemotherapy-Induced Cardiotoxicity: Insights into the Underlying Inflammatory and Oxidative Mechanisms. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07574-0. [PMID: 38492161 DOI: 10.1007/s10557-024-07574-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
While oncotherapy has made rapid progress in recent years, side effects of anti-cancer drugs and treatments have also come to the fore. These side effects include cardiotoxicity, which can cause irreversible cardiac damages with long-term morbidity and mortality. Despite the continuous in-depth research on anti-cancer drugs, an improved knowledge of the underlying mechanisms of cardiotoxicity are necessary for early detection and management of cardiac risk. Although most reviews focus on the cardiotoxic effect of a specific individual chemotherapeutic agent, the aim of our review is to provide comprehensive insight into various agents that induced cardiotoxicity and their underlying mechanisms. Characterization of these mechanisms are underpinned by research on animal models and clinical studies. In order to gain insight into these complex mechanisms, we emphasize the role of inflammatory processes and oxidative stress on chemotherapy-induced cardiac changes. A better understanding and identification of the interplay between chemotherapy and inflammatory/oxidative processes hold some promise to prevent or at least mitigate cardiotoxicity-associated morbidity and mortality among cancer survivors.
Collapse
Affiliation(s)
- András Nagy
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Denise Börzsei
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Alexandra Hoffmann
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Szilvia Török
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Médea Veszelka
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Nikoletta Almási
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Csaba Varga
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Renáta Szabó
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary.
| |
Collapse
|
6
|
Refaie MMM, Shehata S, El-Hussieny M, Fawzy MA, Ahmed NZM, Marey H, Hishmat AM, Alkully T, Rahman ESMAE. Molecular mechanism of empagliflozin cardioprotection in 5-fluorouracil (5-FU)-induced cardiotoxicity via modulation of SGLT2 and TNFα/TLR/NF-κB signaling pathway in rats. Toxicol Res 2024; 40:139-151. [PMID: 38223670 PMCID: PMC10786789 DOI: 10.1007/s43188-023-00204-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 07/18/2023] [Accepted: 07/26/2023] [Indexed: 01/16/2024] Open
Abstract
One of the commoly used chemotherapeutic agents is 5-Fluorouracil (5-FU). Unfortunately, the clinical administration of 5-FU is complicated with serious cardiotoxic effects and the safe use becomes an urgent task in cardio-oncology. Till now, there are no studies discussed the role of empagliflozin (EMP) against 5-FU cardiotoxicity. Thus, we investigated this effect and the involved mechanisms in 5-FU induced heart injury. Forty male rats of Wistar albino species were used and divided randomly into four groups. Group I is the control group, group II is EMP given group, group III is 5-FU cardiotoxic group and group IV is 5-FU plus EMP group. 5-FU (150 mg/kg) was administered as a single intraperitoneal (i.p.) dose on 1st day to induce cardiotoxicity with or without EMP (30 mg/kg/d) orally for 5 days. The dose of 5-FU is relevant to the human toxic dose. Our data showed that 5-FU given group caused cardiotoxicity with significant increase of serum cardiac enzymes, toll like receptors, enhancement of nuclear factor kappa B (NF-κB), interleukin1β (IL1β), IL6, myeloid-differentiation-factor 88 (MYD88), heart weight, malondialdehyde (MDA), tumor-necrosis-factor-alpha (TNFα), sodium glucose co-transporter 2 (SGLT2), P53 and caspase3 expression with clear histopathological features of cardiotoxicity. Moreover, there is a significant decrease in reduced glutathione (GSH) and total antioxidant capacity (TAC). Interestingly, co-administration of EMP could ameliorate 5-FU induced biochemical and histopathological changes. This effect may be due to modulation of SGLT2, decreasing inflammation, oxidative stress and apoptosis with downregulation of an essential inflammatory cascade that mediates 5-FU cardiotoxicity; TNFα/TLR/NF-κB. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-023-00204-1.
Collapse
Affiliation(s)
| | - Sayed Shehata
- Department of Cardiology, Faculty of Medicine, Minia University, El-Minia, 61511 Egypt
| | - Maram El-Hussieny
- Department of Pathology, Faculty of Medicine, Minia University, El-Minia, 61511 Egypt
| | - Michael Atef Fawzy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, El-Minia, 61511 Egypt
| | - Nagwa Zenhom Mustafa Ahmed
- Department of Biochemistry, Faculty of Medicine, Minia University, El-Minia, 61511 Egypt
- Department of Biochemistry, Faculty of Medicine, Al-Baha University, 65525 Albaha, Saudi Arabia
| | - Heba Marey
- Department of Biochemistry, Faculty of Medicine, Minia University, El-Minia, 61511 Egypt
| | - Asmaa Mohammed Hishmat
- Department of Forensic Medicine & Clinical Toxicology, Faculty of Medicine, Minia University, El-Minia, 61511 Egypt
| | - Turki Alkully
- Department of Internal Medicine, Faculty of Medicine, Al-Baha University, 65525 Albaha, Saudi Arabia
| | | |
Collapse
|
7
|
Banu Bal N, Güney C, Gökhan Yıldırım O, Akar F, Demirel-Yılmaz E. Myricetin May Improve Cardiac Dysfunction Possibly Through Regulating Blood Pressure and Cellular Stress Molecules in High-Fructose-Fed Rats. Anatol J Cardiol 2024; 28:55-64. [PMID: 38167793 PMCID: PMC10796237 DOI: 10.14744/anatoljcardiol.2023.3866] [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: 09/19/2023] [Accepted: 10/02/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND The aim of this study was to examine the effect of myricetin on cardiac dysfunction caused by high fructose intake. METHODS Fructose was given to the rats as a 20% solution in drinking water for 15 weeks. Myricetin was administered by oral gavage for the last 6 weeks. Systolic blood pressure was measured by tail-cuff method. The effects of isoprenaline, phenylephrine, and acetylcholine on cardiac contractility and rhythmicity were recorded in the isolated right atrium and left ventricular papillary muscles. In addition to biochemical measurements, the cardiac expressions of cellular stress-related proteins were determined by western blotting. RESULTS Myricetin improved systolic blood pressure but did not affect body weight, plasma glucose, and triglyceride levels in fructose-fed rats. The impairment of isoprenaline- and phenylephrine-mediated increases in atrial contraction and sinus rate in fructose-fed rats was restored by myricetin treatment. Isoprenaline, phenylephrine, and acetylcholine-mediated papillary muscle contractions were not changed by fructose or myricetin administration. The expression of the mitochondrial fission marker dynamin-related protein 1 and the mitophagic marker PTEN-induced kinase 1 (PINK1) was enhanced in the fructose-fed rat, and myricetin treatment markedly attenuated PINK1 expression. High-fructose intake augmented phosphorylation of the proinflammatory molecule Nuclear factor kappa B (NF-κB) and the stress-regulated kinase JNK1, but myricetin only reduced NF-κB expression. Moreover, myricetin diminished the elevation in the expression of the pro-apoptotic Bax. CONCLUSION Our results imply that myricetin has a protective role in cardiac irregularities induced by a high-fructose diet through reducing systolic blood pressure, improving cardiac adrenergic responses, suppressing PINK1, NF-κB, and Bax expression, and thus reflecting a potential therapeutic value.
Collapse
Affiliation(s)
- Nur Banu Bal
- Department of Pharmacology, Faculty of Pharmacy, Gazi University, Ankara, Türkiye
| | - Ceren Güney
- Department of Pharmacology, Faculty of Pharmacy, Gazi University, Ankara, Türkiye
| | - Onur Gökhan Yıldırım
- Department of Pharmacology, Faculty of Pharmacy, Düzce University, Düzce, Türkiye
| | - Fatma Akar
- Department of Pharmacology, Faculty of Pharmacy, Gazi University, Ankara, Türkiye
| | - Emine Demirel-Yılmaz
- Department of Pharmacy Services, Vocational School of Health Services, Artvin Çoruh University, Artvin, Türkiye
| |
Collapse
|
8
|
Nadalin P, Kim JK, Park SU. Recent studies on myricetin and its biological and pharmacological activities. EXCLI JOURNAL 2023; 22:1223-1231. [PMID: 38317860 PMCID: PMC10839238 DOI: 10.17179/excli2023-6571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/14/2023] [Indexed: 02/07/2024]
Affiliation(s)
- Priscilla Nadalin
- Department of Crop Science, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Korea
| | - Jae Kwang Kim
- Division of Life Sciences and Convergence Research Center for Insect Vectors, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea
| | - Sang Un Park
- Department of Crop Science, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Korea
| |
Collapse
|
9
|
Kumar S, Swamy N, Tuli HS, Rani S, Garg A, Mishra D, Abdulabbas HS, Sandhu SS. Myricetin: a potential plant-derived anticancer bioactive compound-an updated overview. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2179-2196. [PMID: 37083713 DOI: 10.1007/s00210-023-02479-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/28/2023] [Indexed: 04/22/2023]
Abstract
The globe is currently confronting a global fight against the deadliest cancer sickness. Chemotherapy, hormonal therapy, surgery, and radiation therapy are among cancer treatment options. Still, these treatments can induce patient side effects, including recurrence, multidrug resistance, fever, and weakness. As a result, the scientific community is always working on natural phytochemical substances. Numerous phytochemical compounds, including taxol analogues, vinca alkaloids such as vincristine and vinblastine, and podophyllotoxin analogues, are currently undergoing testing and have shown promising results against a number of the deadliest diseases, as well as considerable advantages due to their safety and low cost. According to research, secondary plant metabolites such as myricetin, a flavonoid in berries, herbs, and walnuts, have emerged as valuable bio-agents for cancer prevention. Myricetin and its derivatives have antiinflammatory, anticancer, apoptosis-inducing, and anticarcinogenic properties and can prevent cancer cell proliferation. Multiple studies have found that myricetin has anticancer characteristics in various malignancies, including colon, breast, prostate, bladder, and pancreatic cancers. Current knowledge of the anticancer effects of myricetin reveals its promise as a potentially bioactive chemical produced from plants for the prevention and treatment of cancer. This review aimed to study the numerous bioactivities, mode of action, and modification of several cellular processes that myricetin possesses to impede the spread of cancer cells. This review also addresses the challenges and future prospects of using myricetin as a anticancer drug.
Collapse
Affiliation(s)
- Suneel Kumar
- Department of Botany, Government Girls College Khargone, 451001, Khargone, Madhya Pradesh, India
| | - Nitin Swamy
- Fungal Biotechnology and Invertebrate Pathology Laboratory, Department of Biological Sciences, Rani Durgavati University, Jabalpur, 482001, Madhya Pradesh, India
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, Haryana, India
| | - Seema Rani
- Department of Chemistry, Government M. H. College of Home Science & Science for Women, Autonomous, Jabalpur, 482002, Madhya Pradesh, India
| | - Abhijeet Garg
- Fungal Biotechnology and Invertebrate Pathology Laboratory, Department of Biological Sciences, Rani Durgavati University, Jabalpur, 482001, Madhya Pradesh, India
| | - Deepa Mishra
- Department of Biotechnology, Mata Gujri Mahila Mahavidyalaya Jabalpur, 482001, Jabalpur, Madhya Pradesh, India
| | - Hadi Sajid Abdulabbas
- Continuous Education Department, Faculty of Dentistry, University of Al-Ameed, Karbala, 56001, Iraq
| | - Sardul Singh Sandhu
- Bio-Design Innovation Centre, Rani Durgavati University, Jabalpur, 482001, Madhya Pradesh, India.
| |
Collapse
|
10
|
Wendlocha D, Krzykawski K, Mielczarek-Palacz A, Kubina R. Selected Flavonols in Breast and Gynecological Cancer: A Systematic Review. Nutrients 2023; 15:2938. [PMID: 37447264 DOI: 10.3390/nu15132938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/16/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
The consumption of foods that are rich in phenolic compounds has chemopreventive effects on many cancers, including breast cancer, ovarian cancer, and endometrial cancer. A wide spectrum of their health-promoting properties such as antioxidant, anti-inflammatory, and anticancer activities, has been demonstrated. This paper analyzes the mechanisms of the anticancer action of selected common flavonols, including kemferol, myricetin, quercetin, fisetin, galangin, isorhamnetin, and morin, in preclinical studies, with particular emphasis on in vitro studies in gynecological cancers and breast cancer. In the future, these compounds may find applications in the prevention and treatment of gynecological cancers and breast cancer, but this requires further, more advanced research.
Collapse
Affiliation(s)
- Dominika Wendlocha
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland
| | - Kamil Krzykawski
- Silesia LabMed: Centre for Research and Implementation, Medical University of Silesia in Katowice, 40-752 Katowice, Poland
| | - Aleksandra Mielczarek-Palacz
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland
| | - Robert Kubina
- Silesia LabMed: Centre for Research and Implementation, Medical University of Silesia in Katowice, 40-752 Katowice, Poland
- Department of Pathology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland
| |
Collapse
|
11
|
da Silva MC, Fabiano LC, da Costa Salomão KC, de Freitas PLZ, Neves CQ, Borges SC, de Souza Carvalho MDG, Breithaupt-Faloppa AC, de Thomaz AA, Dos Santos AM, Buttow NC. A Rodent Model of Human-Dose-Equivalent 5-Fluorouracil: Toxicity in the Liver, Kidneys, and Lungs. Antioxidants (Basel) 2023; 12:antiox12051005. [PMID: 37237871 DOI: 10.3390/antiox12051005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
5-Fluorouracil (5-FU) is a chemotherapy drug widely used to treat a range of cancer types, despite the recurrence of adverse reactions. Therefore, information on its side effects when administered at a clinically recommended dose is relevant. On this basis, we examined the effects of the 5-FU clinical treatment on the integrity of the liver, kidneys, and lungs of rats. For this purpose, 14 male Wistar rats were divided into treated and control groups and 5-FU was administered at 15 mg/kg (4 consecutive days), 6 mg/kg (4 alternate days), and 15 mg/kg on the 14th day. On the 15th day, blood, liver, kidney, and lung samples were collected for histological, oxidative stress, and inflammatory evaluations. We observed a reduction in the antioxidant markers and an increase in lipid hydroperoxides (LOOH) in the liver of treated animals. We also detected elevated levels of inflammatory markers, histological lesions, apoptotic cells, and aspartate aminotransferase. Clinical treatment with 5-FU did not promote inflammatory or oxidative alterations in the kidney samples; however, histological and biochemical changes were observed, including increased serum urea and uric acid. 5-FU reduces endogenous antioxidant defenses and increases LOOH levels in the lungs, suggesting oxidative stress. Inflammation and histopathological alterations were also detected. The clinical protocol of 5-FU promotes toxicity in the liver, kidneys, and lungs of healthy rats, resulting in different levels of histological and biochemical alterations. These results will be useful in the search for new adjuvants to attenuate the adverse effects of 5-FU in such organs.
Collapse
Affiliation(s)
- Mariana Conceição da Silva
- Biological Physics and Cell Signaling Laboratory, Institute of Biology, Department of Structural and Functional Biology, State University of Campinas, Campinas 13083-970, SP, Brazil
| | - Lilian Catarim Fabiano
- Department of Morphological Science, State University of Maringá, Maringá 87020-900, PR, Brazil
| | | | | | - Camila Quaglio Neves
- Department of Morphological Science, State University of Maringá, Maringá 87020-900, PR, Brazil
| | | | - Maria das Graças de Souza Carvalho
- Biological Physics and Cell Signaling Laboratory, Institute of Biology, Department of Structural and Functional Biology, State University of Campinas, Campinas 13083-970, SP, Brazil
| | - Ana Cristina Breithaupt-Faloppa
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-904, SP, Brasil
| | - André Alexandre de Thomaz
- Quantum Electronic Department, Institute of Physics Gleb Wataghin, State University of Campinas, Campinas 13083-872, SP, Brazil
| | - Aline Mara Dos Santos
- Biological Physics and Cell Signaling Laboratory, Institute of Biology, Department of Structural and Functional Biology, State University of Campinas, Campinas 13083-970, SP, Brazil
| | - Nilza Cristina Buttow
- Department of Morphological Science, State University of Maringá, Maringá 87020-900, PR, Brazil
| |
Collapse
|
12
|
Gui Y, Famurewa AC, Olatunji OJ. Naringin ameliorates 5-fluorouracil induced cardiotoxicity: An insight into its modulatory impact on oxidative stress, inflammatory and apoptotic parameters. Tissue Cell 2023; 81:102035. [PMID: 36753813 DOI: 10.1016/j.tice.2023.102035] [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: 10/17/2022] [Revised: 02/03/2023] [Accepted: 02/03/2023] [Indexed: 02/07/2023]
Abstract
5-fluorouracil (5-FU) is an efficacious fluoropyrimidine antimetabolite anticancer drug, however, its clinical utility is constrained due to side effect toxicity on delicate organs, including the heart. This study thus aimed at exploring the cardioprotective potentials of naringin (NRG) against 5-FU-induced cardiotoxicity in rats. We divided Wistar rats into four experimental groups (n = 6) for the administration of NRG (100 mg/kg bw, orally) and/or 5-FU (150 mg/kg bw, intraperitoneal). NRG was administered for 10 days, while 5-FU was injected on the 8th day only. Serum troponin-I (cTn-I) and creatine kinase (CK) were estimated. Cardiac activities/level of catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPx), reduced glutathione (GSH), malondialdehyde (MDA), interleukin-1β (IL-1β), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), inducible nitric oxide synthase (iNOS) and nuclear factor-ĸB (NF-κB) and caspase-3 were determined. 5-FU markedly increased cTn-I, CK, cardiac inflammatory mediators and caspase-3 expressions, whereas antioxidant mediators decreased appreciably when compared to the control groups. Interestingly, the prophylactic administration of NRG prominently inhibited the 5-FU-provoked oxidative stress, pro-inflammation and apoptosis in the heart of rats. Histopathology confirmed the biochemical results of the heart. Therefore, NRG is a potential natural flavonoid for mitigation of 5-FU cardiotoxicity in rats.
Collapse
Affiliation(s)
- Yang Gui
- Department of General Surgery, The Second Peoples Hospital of Wuhu, Wuhu 241001, Anhui, China
| | - Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex-Ekwueme Federal University, Ndufu-Alike, Ikwo, Ebonyi, Nigeria; Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal University, Karnataka State, India
| | - Opeyemi Joshua Olatunji
- Traditional Thai Medical Research and Innovation Center, Faculty of Traditional Thai Medicine, Prince of Songkla University, Hat Yai 90110, Thailand; African Genome Center, Mohammed VI Polytechnic University, Ben Guerir 43150, Morocco.
| |
Collapse
|
13
|
Tambe PK, Mathew AJ, Bharati S. Cardioprotective potential of mitochondria-targeted antioxidant, mito-TEMPO, in 5-fluorouracil-induced cardiotoxicity. Cancer Chemother Pharmacol 2023; 91:389-400. [PMID: 36997656 PMCID: PMC10156775 DOI: 10.1007/s00280-023-04529-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/21/2023] [Indexed: 04/03/2023]
Abstract
Abstract
Purpose
The mitochondria-targeted antioxidants (MTAs) are known to offer protection against mitochondrial oxidative stress. The recent evidences support their role in mitigating oxidative stress-induced diseases, including cancer. Therefore, this study investigated cardioprotective potential of mito-TEMPO against 5-FU-induced cardiotoxicity.
Methods
Mito-TEMPO was administered to male BALB/C mice (intraperitoneally, 0.1 mg/kg b.w. for 7 days) followed by intraperitoneal administration of 5- FU (12 mg/kg b.w. for 4 days). During this period, mito-TEMPO treatment was also continued. The cardioprotective potential of mito-TEMPO was assessed by evaluating cardiac injury markers, extent of non-viable myocardium and histopathological alterations. Mitochondrial functional status and mitochondrial oxidative stress were assessed in cardiac tissue. 8-OHdG expression and apoptotic cell death were assessed using immunohistochemical techniques.
Results
The level of cardiac injury markers CK-MB and AST were significantly (P ≤ 0.05) decreased in mito-TEMPO pre-protected group which was further reflected in histopathology as decrease in the percentage of non-viable myocardial tissue, disorganization, and loss of myofibrils. Mito-TEMPO ameliorated mtROS, mtLPO and conserved mitochondrial membrane potential. Further, it had significantly (P ≤ 0.05) improved the activity of mitochondrial complexes and mitochondrial enzymes. A significant (P ≤ 0.05) increase in the level of mtGSH, activity of mitochondrial glutathione reductase, glutathione peroxidase, and mitochondrial superoxide dismutase was observed. A decreased expression of 8-OHdG and reduced apoptotic cell death were observed in mito-TEMPO pre-protected group.
Conclusion
Mito-TEMPO effectively mitigated 5-FU-induced cardiotoxicity by modulating mitochondrial oxidative stress, hence may serve as a protective agent/adjuvant in 5-FU-based combinatorial chemotherapy.
Collapse
|
14
|
Aminzadeh A, Darijani MH, Bashiri H. Investigating the effect of myricetin against arsenic-induced cardiac toxicity in rats. Toxicol Res (Camb) 2023; 12:117-123. [PMID: 36866219 PMCID: PMC9972817 DOI: 10.1093/toxres/tfad003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 02/05/2023] Open
Abstract
Arsenic intoxication is a serious health hazard worldwide. Its toxicity is associated with several disorders and health problems in humans. Recent studies revealed that myricetin has various biological effects, including anti-oxidation. The aim of this study is to investigate the protective effect of myricetin against arsenic-induced cardiotoxicity in rats. Rats were randomized to one of the following groups: control, myricetin (2 mg/kg), arsenic (5 mg/kg), myricetin (1 mg/kg) + arsenic, and myricetin (2 mg/kg) + arsenic. Myricetin was given intraperitoneally 30 min before arsenic administration (5 mg/kg for 10 days). After treatments, the activity of lactate dehydrogenase (LDH) and the levels of aspartate aminotransferase (AST), creatine kinase myocardial band (CK-MB), lipid peroxidation (LPO), total antioxidant capacity (TAC), and total thiol molecules (TTM) were determined in serum samples and cardiac tissues. Also, histological changes in cardiac tissue were evaluated. Myricetin pretreatment inhibited arsenic-induced increase in LDH, AST, CK-MB, and LPO levels. Pretreatment with myricetin also enhanced the decreased TAC and TTM levels. In addition, myricetin improved histopathological alterations in arsenic-treated rats. In conclusion, the results of the present study demonstrated that treatment with myricetin prevented arsenic-induced cardiac toxicity at least in part by decreasing oxidative stress and restoring the antioxidant system.
Collapse
Affiliation(s)
- Azadeh Aminzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman 7616911319, Iran
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7616911319, Iran
| | - Mohammad Hossein Darijani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman 7616911319, Iran
| | - Hamideh Bashiri
- Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman 7616914115, Iran
| |
Collapse
|
15
|
Choudhary R, Singh A, Upadhyay A, Singh R, Thangalakshmi S, Dar AH, Bajpai VK, Shukla S. Exotic god fruit, persimmon (
Diospyros kaki
): Pharmacological importance and human health aspects. EFOOD 2023. [DOI: 10.1002/efd2.52] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Rita Choudhary
- TERI‐Deakin Nanobiotechnology Centre, Division of Sustainable Agriculture The Energy and Resources Institute, Gwal Pahari Haryana Gurugram India
| | - Anurag Singh
- Department of Food Science and Technology National Institute of Food Technology Entrepreneurship and Management, Kundli Sonipat Haryana India
| | - Ashutosh Upadhyay
- Department of Food Science and Technology National Institute of Food Technology Entrepreneurship and Management, Kundli Sonipat Haryana India
| | - Rakhi Singh
- Department of Food Science and Technology National Institute of Food Technology Entrepreneurship and Management, Kundli Sonipat Haryana India
| | - S. Thangalakshmi
- Department of Food Engineering National Institute of Food Technology Entrepreneurship and Management, Kundli Sonipat Haryana India
| | - Aamir H. Dar
- Department of Food Technology Islamic University of Sciences and Technology Awantipora Kashmir India
| | - Vivek K. Bajpai
- Department of Energy and Materials Engineering Dongguk University Seoul Republic of Korea
| | - Shruti Shukla
- TERI‐Deakin Nanobiotechnology Centre, Division of Sustainable Agriculture The Energy and Resources Institute, Gwal Pahari Haryana Gurugram India
| |
Collapse
|
16
|
Alsaffar RM, Ali A, Rashid SM, Ahmad SB, Alkholifi FK, Kawoosa MS, Ahmad SP, Rehman MU. Zerumbone Protects Rats from Collagen-Induced Arthritis by Inhibiting Oxidative Outbursts and Inflammatory Cytokine Levels. ACS OMEGA 2023; 8:2982-2991. [PMID: 36713739 PMCID: PMC9878628 DOI: 10.1021/acsomega.2c05749] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 12/22/2022] [Indexed: 05/25/2023]
Abstract
Rheumatoid arthritis (RA) is an immunocompromised disorder characterized by a marked increase in the synthesis of inflammatory molecules that stimulates the destruction of bones and cartilage. The conventional treatment modalities for RA are associated with adverse side effects and lack sensitivity, suggesting an immediate demand for alternate beneficial therapeutic remedies. The current study sought to understand more about zerumbone's anti-inflammatory properties in diagnosing collagen-induced arthritis (CIA) in experimental animals. The current study observed that zerumbone reduced clinical severity in CIA-induced animals compared to healthy animals. Zerumbone administration significantly decreased (p < 0.001) the concentration of SOD, CAT, GR, and GSH in treatment groups. Zerumbone administration drove down significantly (p < 0.001) the concentration of inflammatory cytokine molecules. Zerumbone was effective in bringing significant changes in levels of MPO, NO, LDH, MMP-8, and ELA. The therapeutic potential of zerumbone was found to be associated with reduced joint destruction and restored normal histology in the cartilage and tissue. Adsorption, distribution, metabolism, excretion, and toxicity studies were used to determine the druglike properties of zerumbone. ProTox-II studies revealed that zerumbone did not possess toxic properties like hepatotoxicity, immunotoxicity, carcinogenicity, mutagenicity, and cytotoxicity. Therefore, the present study evaluated the therapeutic properties of zerumbone in CIA animal models.
Collapse
Affiliation(s)
- Rana M Alsaffar
- Department
of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi
Arabia
| | - Aarif Ali
- Division
of Veterinary Biochemistry, Faculty of Veterinary
Science and Animal Husbandry, SKUAST-Kashmir, Shuhama, Alusteng, Srinagar, Jammu and Kashmir 190006, India
| | - Shahzada Mudasir Rashid
- Division
of Veterinary Biochemistry, Faculty of Veterinary
Science and Animal Husbandry, SKUAST-Kashmir, Shuhama, Alusteng, Srinagar, Jammu and Kashmir 190006, India
| | - Sheikh Bilal Ahmad
- Division
of Veterinary Biochemistry, Faculty of Veterinary
Science and Animal Husbandry, SKUAST-Kashmir, Shuhama, Alusteng, Srinagar, Jammu and Kashmir 190006, India
| | - Faisal K Alkholifi
- Department
of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi
Arabia
| | - Majid Shafi Kawoosa
- Mycoplasma
Laboratory, Faculty of Veterinary Sciences
and Animal Husbandry, Shuhama, Alusteng, Srinagar, Jammu and Kashmir 190006, India
| | - Sheikh Parvaiz Ahmad
- Department
of Statistics, University of Kashmir, Srinagar, Jammu and Kashmir 190006, India
| | - Muneeb U Rehman
- Department
of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
17
|
Jiang C, Xie L, Wang Y, Liang J, Li H, Luo L, Li T, Liang Z, Tang L, Ning D, Ya Y, Yan F. Highly sensitive electrochemical detection of myricetin in food samples based on the enhancement effect of Al-MOFs. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2022; 14:3521-3528. [PMID: 36018228 DOI: 10.1039/d2ay00957a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Microporous aluminum-based metal-organic frameworks (CAU-1) are used to develop a simple and sensitive electrochemical sensor for myricetin (MYR) based on a modified carbon paste electrode (CPE) for the first time. The morphologies and electrochemical properties of the as-synthesized CAU-1 are studied utilizing various analytical methods including scanning electron microscopy, transmission electron microscopy, X-ray diffraction, Fourier transform infrared spectroscopy, N2 adsorption-desorption, and electrochemical impedance spectroscopy. In terms of electrochemical oxidation of MYR, CAU-1/CPE with its large number of active micropores and rapid electron transfer demonstrates superior performance compared to the bare CPE. Under optimized conditions, the calibration curve for MYR exhibits a linear range of 1.0-10 μg L-1 and 10-1000 μg L-1 with a detection limit of 0.50 μg L-1. The developed CAU-1/CPE exhibits superior analytical characteristics, compared to previously reported electrochemical sensors for MYR detection. Furthermore, CAU-1/CPE is employed to determine MYR in Myrica bark samples, and the results are consistent with those obtained by high-performance liquid chromatography, demonstrating the excellent potential of CAU-1/CPE for the rapid analysis of MYR in complicated real samples.
Collapse
Affiliation(s)
- Cuiwen Jiang
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Liping Xie
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Yanli Wang
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Jing Liang
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Huiling Li
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Lihong Luo
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Tao Li
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Zhongdan Liang
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Li Tang
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Dejiao Ning
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Yu Ya
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Feiyan Yan
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| |
Collapse
|
18
|
AlAsmari AF, Ali N, Alharbi M, Alqahtani F, Alasmari F, Almoqbel D, AlSwayyed M, Alshammari A, Alanazi MM, Alhoshani A, Al-Harbi NO. Geraniol Ameliorates Doxorubicin-Mediated Kidney Injury through Alteration of Antioxidant Status, Inflammation, and Apoptosis: Potential Roles of NF-κB and Nrf2/Ho-1. Nutrients 2022; 14:nu14081620. [PMID: 35458182 PMCID: PMC9031157 DOI: 10.3390/nu14081620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 12/10/2022] Open
Abstract
Doxorubicin-mediated kidney impairment is a serious problem in cancer treatment. Accordingly, this work investigated the ability of geraniol to modulate doxorubicin-induced kidney damage using a rat model. Rats were randomly assigned to four groups: control, doxorubicin (20 mg/kg, intraperitoneal, i.p.), doxorubicin plus 100 mg/kg of geraniol, and doxorubicin plus 200 mg/kg of geraniol. A single doxorubicin injection triggered kidney impairment, as evidenced by the altered serum creatinine, blood urea nitrogen, and albumin values; it also caused histological changes in the kidney architecture. Additionally, doxorubicin enhanced lipid peroxidation while lowering reduced glutathione, catalase activity, and the expression of glutathione peroxidase and superoxide dismutase. Interestingly, pre-treatment with geraniol rescued doxorubicin-induced alterations in kidney antioxidant parameters, enzymatic activity, and the expression of inflammatory and apoptosis-mediating gene and proteins. Moreover, prophylactic treatment with geraniol preserved most kidney histological characteristics in a dose-dependent manner. These findings support that geraniol could protect against doxorubicin-mediated kidney dysfunction. However, further research is needed to clarify the mechanisms of geraniol’s protective effects against doxorubicin-mediated kidney dysfunction.
Collapse
Affiliation(s)
- Abdullah F. AlAsmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (M.A.); (F.A.); (F.A.); (D.A.); (A.A.); (M.M.A.); (A.A.); (N.O.A.-H.)
- Correspondence: ; Tel.: +966-114677180
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (M.A.); (F.A.); (F.A.); (D.A.); (A.A.); (M.M.A.); (A.A.); (N.O.A.-H.)
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (M.A.); (F.A.); (F.A.); (D.A.); (A.A.); (M.M.A.); (A.A.); (N.O.A.-H.)
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (M.A.); (F.A.); (F.A.); (D.A.); (A.A.); (M.M.A.); (A.A.); (N.O.A.-H.)
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (M.A.); (F.A.); (F.A.); (D.A.); (A.A.); (M.M.A.); (A.A.); (N.O.A.-H.)
| | - Daad Almoqbel
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (M.A.); (F.A.); (F.A.); (D.A.); (A.A.); (M.M.A.); (A.A.); (N.O.A.-H.)
| | - Mohammed AlSwayyed
- Department of Pathology, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (M.A.); (F.A.); (F.A.); (D.A.); (A.A.); (M.M.A.); (A.A.); (N.O.A.-H.)
| | - Mohammed M. Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (M.A.); (F.A.); (F.A.); (D.A.); (A.A.); (M.M.A.); (A.A.); (N.O.A.-H.)
| | - Ali Alhoshani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (M.A.); (F.A.); (F.A.); (D.A.); (A.A.); (M.M.A.); (A.A.); (N.O.A.-H.)
| | - Naif O. Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (M.A.); (F.A.); (F.A.); (D.A.); (A.A.); (M.M.A.); (A.A.); (N.O.A.-H.)
| |
Collapse
|