101
|
Ngatuni D, Wairagu P, Jillani N, Isaac AO, Nyariki JN. A glyphosate-based herbicide disrupted hematopoiesis and induced organ toxicities, ameliorated by vitamin B12 in a mouse model. Saudi J Biol Sci 2022; 29:103278. [PMID: 35401022 PMCID: PMC8987997 DOI: 10.1016/j.sjbs.2022.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 03/25/2022] [Indexed: 11/30/2022] Open
Abstract
Glyphosate-based herbicides (GBH) are widely used worldwide. Their negative impact on human health is a matter of debate by regulatory bodies and the public. The present study sought to determine the impact of a GBH on the vital organs; and the potential protective effects of vitamin B12 (cyanocobalamin) supplementation. Sixty white Swiss mice were randomly assigned to five treatment groups, each containing twelve mice. Group one represented the normal control; Group two mice were treated with 375 mg/kg of GBH for 56 days; Group three mice received 10 mg/kg of cyanocobalamin for 56 days; Group four mice were administered with 375 mg/kg of GBH and 10 mg/kg cyanocobalamin for 56 days and Group five received 10 mg/kg cyanocobalamin first for 7 days, then continued thereafter co-administered together with 375 mg/kg of GBH for 56 days). Oral administration of GBH induced severe anemia in mice, which was attenuated by cyanocobalamin. Moreover, GBH resulted in a very significant alteration of platelets, WBCs, and its sub-types. Once again, cyanocobalamin stabilized the levels of platelets and WBCs in the presence of GBH. GBH-induced elevation of triglycerides and HDL was nullified by the administration of cyanocobalamin. Further studies showed evidence for GBH-induced inflammation represented by an imbalance in serum levels of the TNF-α: IL-10 and IFN-γ ratios. The GBH severely depleted GSH levels in the liver. A GBH-induced rise in GSH in the kidney, lungs and brain was noted; and is an indicator of antioxidant capacity enhancement in response to a GBH-induced oxidant challenge. Moreover, cyanocobalamin supplementation abrogated GBH-induced oxidative stress as depicted by stabilized GSH levels in the liver, kidney, lungs, and brain. In the presence of cyanocobalamin, the GBH-induced liver injury depicted by elevation of AST, ALT, and bilirubin, was attenuated. From the results, we conclude that the capacity of cyanocobalamin to assuage GBH-induced inflammatory responses, hepatotoxicity, and hematological alteration as well as oxidative stress may be attributable to its antioxidant and anti-inflammatory properties. The current findings provide a solid foundation for further scrutiny of this phenomenon, with vital implications in GBH exposure and the role of potent antioxidant supplementation in the management of GBH-induced toxicity.
Collapse
|
102
|
Zhou Q, Zhang N, Hu T, Xu H, Duan X, Liu B, Chen F, Wang M. Dietary phenolic-type Nrf2-activators: implications in the control of toxin-induced hepatic disorders. Food Funct 2022; 13:5480-5497. [PMID: 35411358 DOI: 10.1039/d1fo04237h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Numerous studies have exemplified the importance of nuclear factor erythroid 2-related factor 2 (Nrf2) activation in the alleviation of toxin-induced hepatic disorders primarily through eliminating oxidative stress. Whereafter, increasingly more efforts have been contributed to finding Nrf2-activators, especially from dietary polyphenols. The present review summarized the phenolic-type Nrf2-activators published in the past few decades, analyzed their effectiveness based on their structural characteristics and outlined their related mechanisms. It turns out that flavonoids are the largest group of phenolic-type Nrf2-activators, followed by nonflavonoids and phenolic acids. When counting on subgroups, the top three types are flavonols, flavones, and hydroxycinnamic acids, with curcuminoids having the highest effective doses. Moreover, most polyphenols work through the phosphorylation of Nrf2. Besides, mitogen-activated protein kinases (MAPKs) and protein kinase B (Akt) are the frequent targets of these Nrf2-activators, which indirectly mediate the behavior of Nrf2. However, current data are not sufficient to conclude any structure-activity relationship.
Collapse
Affiliation(s)
- Qian Zhou
- Institute for Advanced Study, Shenzhen University, Shenzhen, China. .,Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China.
| | - Nana Zhang
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Tingyan Hu
- Institute for Advanced Study, Shenzhen University, Shenzhen, China. .,Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China.
| | - Hui Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen, China. .,Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China.
| | - Xinxing Duan
- Schlegel Research Institute for Aging & Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Canada
| | - Bin Liu
- Institute for Advanced Study, Shenzhen University, Shenzhen, China. .,Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China.
| | - Feng Chen
- Institute for Advanced Study, Shenzhen University, Shenzhen, China. .,Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China.
| | - Mingfu Wang
- Institute for Advanced Study, Shenzhen University, Shenzhen, China. .,Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China.
| |
Collapse
|
103
|
Ma W, Ao S, Zhou J, Li J, Liang X, Yang X, Zhang H, Liu B, Tang W, Liu H, Xiao H, Liang H, Yang X. Methylsulfonylmethane protects against lethal dose MRSA-induced sepsis through promoting M2 macrophage polarization. Mol Immunol 2022; 146:69-77. [PMID: 35461144 DOI: 10.1016/j.molimm.2022.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 02/25/2022] [Accepted: 04/04/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Multi-drug-resistant bacterial infections, which have become a global threat, lack effective treatments. The discoveries of non-antibiotics with different modes of antibacterial action, such as methylsulfonylmethane (MSM), are a promising new treatment for multi-drug-resistant pathogens. METHODS We constructed a mouse peritonitis infection model to evaluate the effects of MSM against methicillin-resistant Staphylococcus aureus (MRSA) infection. The time-kill kinetics of MSM against MRSA and the effect of MSM on the integrity of bacterial cell membrane were measured. Viability effects of MSM on THP1 cells were performed by CCK-8 cytotoxicity assay. Systematic inflammatory factor levels of mice were detected using ELISA. The immune response of peritoneal macrophages during MRSA-infection was evaluated using RNA sequencing. Gene Ontology function, Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses, and correlation analyses were applied to analysis RNA sequencing data. RT-qPCR, western blotting and flow cytometry were performed to analysis the gene and protein expression levels of macrophages. RESULTS In in vitro experiments, MSM did not show significant killing effects on the growth of MRSA directly and did not destroy bacterial membrane integrity. MSM also displayed no significant effects on the proliferative capacity of THP1 cells. However, MSM treatment protected mice against a lethal dose MRSA-infection and decreased systemic inflammation. MSM upregulated metabolic pathway in peritoneal macrophages, especial glycolysis, during MRSA infection. MSM increased the expression of M2 markers (such as Arg1), promoted phosphorylation of STAT3 (which regulates M2 polarization), and decreased the expression of M1 markers in peritoneal macrophages. Additionally, MSM treatment increased the expression of H3K18 lactylation specific target genes, including Arg1. GNE-140, the LDHA-specific inhibitor of glycolysis, blocked the MSM-induced Arg1 expression in this disease model. CONCLUSIONS MSM protects against MRSA infection through immunomodulation. MSM promotes the expression of Arg1 by lactate-H3K18la pathway to control macrophage to M2 polarization; it firstly provides therapeutic potential for drug-resistant infections and sepsis.
Collapse
Affiliation(s)
- Wei Ma
- Department of Wound Infection and Drug, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, PR China
| | - Shengxiang Ao
- Department of Wound Infection and Drug, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, PR China
| | - Jianping Zhou
- College of Basic Medical Sciences, Panzihua University, Panzihua 617000, PR China
| | - Jiaxin Li
- Department of Wound Infection and Drug, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, PR China
| | - Xin Liang
- Department of Wound Infection and Drug, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, PR China
| | - Xue Yang
- Department of Wound Infection and Drug, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, PR China
| | - Hao Zhang
- Deparment of Critical Care Medicine, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, PR China
| | - Boyang Liu
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Wanqi Tang
- Department of Wound Infection and Drug, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, PR China
| | - Haoru Liu
- Department of Wound Infection and Drug, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, PR China
| | - Hongyan Xiao
- Department of Wound Infection and Drug, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, PR China
| | - Huaping Liang
- Department of Wound Infection and Drug, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, PR China.
| | - Xia Yang
- Department of Wound Infection and Drug, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, PR China.
| |
Collapse
|
104
|
Tan S, Bai J, Xu M, Zhang L, Wang Y. Thioredoxin-1 Activation by Pterostilbene Protects Against Doxorubicin-Induced Hepatotoxicity via Inhibiting the NLRP3 Inflammasome. Front Pharmacol 2022; 13:841330. [PMID: 35496300 PMCID: PMC9043100 DOI: 10.3389/fphar.2022.841330] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/28/2022] [Indexed: 11/21/2022] Open
Abstract
Background: Doxorubicin (DOX) has been widely used in cancer treatment. However, DOX can cause a range of significant side effects, of which hepatotoxicity is a common one, and therefore limits its clinical use. Pterostilbene (PTS) has been shown to exhibit anti-oxidant and anti-inflammatory effects in the treatment of liver diseases but whether PTS could protect against hepatotoxicity in DOX-treated mice is unknown. Methods: In our study, we use C57/BL6J mice and the HepG2 cell line. We divided the mice in 4 groups: the control, the PTS treatment, the DOX treatment, and the DOX + PTS treatment group. Liver histopathology was judged by performing hematoxylin–eosin and Masson staining. Immunohistochemistry was used to perform the expression of NLRP3. The levels of serum alanine transaminase (ALT) and aspartate transaminase (AST) were evaluated. Levels of malondialdehyde (MDA), superoxide dismutase (SOD), glutathione (GSH), and DCFH-DA staining were used to evaluate the oxidative injury. Western blot and real-time PCR were applied to evaluate the expressions of proteins and mRNA. MTT was used to evaluate DOX-induced cell injury and the protective effects of PTS. Recombinant Trx-1 was used to analyze the mechanism of PTS. A TUNEL assay was used to detect apoptosis in DOX-induced HepG2 cells and the protective effects of PTS. Results: PTS ameliorated DOX-induced liver pathological changes and the levels of AST and ALT. PTS also decreased the level of MDA, increased the level of SOD, GSH, and the expression of Trx-1 in DOX-treated mice. PTS decreased the levels of NLRP3 and IL-1β mRNA and the expressions of their proteins in DOX-treated mice. In addition, PTS also decreased the expression of Cleaved Caspase-3 and BAX and increased the expression of BCL-2. In vitro, after treatment with recombinant Trx-1, ROS and NLRP3 inflammasome were both decreased. Treatment with PTS could rescue the downregulation of Trx-1, decreased the ROS level and the NLRP3 inflammasome, and protected HepG2 cells against DOX-induced apoptosis. Conclusion: The results show that PTS exhibits protective effects against DOX-induced liver injuries via suppression of oxidative stress, fibrosis, NLRP3 inflammasome stimulation, and cell apoptosis which might lead to a new approach of preventing DOX-induced hepatotoxicity.
Collapse
Affiliation(s)
- Shiqing Tan
- The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jie Bai
- Nutrition and Food Hygiene, Dalian Medical University, Dalian, China
| | - Mingxi Xu
- The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Longying Zhang
- The Second Affiliated Hospital, Dalian Medical University, Dalian, China
- *Correspondence: Longying Zhang, ; Ying Wang,
| | - Ying Wang
- The Second Affiliated Hospital, Dalian Medical University, Dalian, China
- *Correspondence: Longying Zhang, ; Ying Wang,
| |
Collapse
|
105
|
Sun T, Zhang L, Feng J, Bao L, Wang J, Song Z, Mao Z, Li J, Hu Z. Characterization of cellular senescence in doxorubicin-induced aging mice. Exp Gerontol 2022; 163:111800. [DOI: 10.1016/j.exger.2022.111800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 03/31/2022] [Accepted: 04/02/2022] [Indexed: 11/04/2022]
|
106
|
Xing W, Wen C, Wang D, Shao H, Liu C, He C, Olatunji OJ. Cardiorenal Protective Effect of Costunolide against Doxorubicin-Induced Toxicity in Rats by Modulating Oxidative Stress, Inflammation and Apoptosis. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27072122. [PMID: 35408518 PMCID: PMC9000510 DOI: 10.3390/molecules27072122] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 11/16/2022]
Abstract
Doxorubicin (DXB) is one of the most commonly used anticancer agents for treating solid and hematological malignancies; however, DXB-induced cardiorenal toxicity presents a limiting factor to its clinical usefulness in cancer patients. Costunolide (COST) is a naturally occurring sesquiterpene lactone with excellent anti-inflammatory, antioxidant and antiapoptotic properties. This study evaluated the effect of COST on DXB-induced cardiorenal toxicity in rats. Rats were orally treated with COST for 4 weeks and received weekly 5 mg/kg doses of DXB for three weeks. Cardiorenal biochemical biomarkers, lipid profile, oxidative stress, inflammatory cytokines, histological and immunohistochemical analyses were evaluated. DXB-treated rats displayed significantly increased levels of lipid profiles, markers of cardiorenal dysfunction (aspartate aminotransferase, creatine kinase, lactate dehydrogenase, troponin T, blood urea nitrogen, uric acid and creatinine). In addition, DXB markedly upregulated cardiorenal malondialdehyde, tumor necrosis factor-α, interleukin-1β, interleukin-6 levels and decreased glutathione, superoxide dismutase and catalase activities. COST treatment significantly attenuated the aforementioned alterations induced by DXB. Furthermore, histopathological and immunohistochemical analyses revealed that COST ameliorated the histopathological features and reduced p53 and myeloperoxidase expression in the treated rats. These results suggest that COST exhibits cardiorenal protective effects against DXB-induced injury presumably via suppression of oxidative stress, inflammation and apoptosis.
Collapse
Affiliation(s)
- Wen Xing
- Department of Gerontology, Wannan Medical College Affiliated Yijishan Hospital, Wuhu 241001, China; (W.X.); (D.W.)
| | - Chaoling Wen
- Anhui Traditional Chinese Medicine College, Wuhu 241001, China;
| | - Deguo Wang
- Department of Gerontology, Wannan Medical College Affiliated Yijishan Hospital, Wuhu 241001, China; (W.X.); (D.W.)
| | - Hui Shao
- Department of Clinical Laboratory, East China Normal University Affiliated Wuhu Hospital, Wuhu 241001, China;
| | - Chunhong Liu
- The Second Peoples Hospital of Wuhu City, Wuhu 241001, China;
| | - Chunling He
- Department of Endocrinology, Wannan Medical College Affiliated Yijishan Hospital, Wuhu 241001, China
- Correspondence: (C.H.); (O.J.O.)
| | - Opeyemi Joshua Olatunji
- Traditional Thai Medical Research and Innovation Center, Faculty of Traditional Thai Medicine, Prince of Songkla University, Hat Yai 90110, Thailand
- Correspondence: (C.H.); (O.J.O.)
| |
Collapse
|
107
|
Alov P, Al Sharif M, Aluani D, Chegaev K, Dinic J, Divac Rankov A, Fernandes MX, Fusi F, García-Sosa AT, Juvonen R, Kondeva-Burdina M, Padrón JM, Pajeva I, Pencheva T, Puerta A, Raunio H, Riganti C, Tsakovska I, Tzankova V, Yordanov Y, Saponara S. A Comprehensive Evaluation of Sdox, a Promising H2S-Releasing Doxorubicin for the Treatment of Chemoresistant Tumors. Front Pharmacol 2022; 13:831791. [PMID: 35321325 PMCID: PMC8936434 DOI: 10.3389/fphar.2022.831791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/25/2022] [Indexed: 12/11/2022] Open
Abstract
Sdox is a hydrogen sulfide (H2S)-releasing doxorubicin effective in P-glycoprotein-overexpressing/doxorubicin-resistant tumor models and not cytotoxic, as the parental drug, in H9c2 cardiomyocytes. The aim of this study was the assessment of Sdox drug-like features and its absorption, distribution, metabolism, and excretion (ADME)/toxicity properties, by a multi- and transdisciplinary in silico, in vitro, and in vivo approach. Doxorubicin was used as the reference compound. The in silico profiling suggested that Sdox possesses higher lipophilicity and lower solubility compared to doxorubicin, and the off-targets prediction revealed relevant differences between Dox and Sdox towards several cancer targets, suggesting different toxicological profiles. In vitro data showed that Sdox is a substrate with lower affinity for P-glycoprotein, less hepatotoxic, and causes less oxidative damage than doxorubicin. Both anthracyclines inhibited CYP3A4, but not hERG currents. Unlike doxorubicin, the percentage of zebrafish live embryos at 72 hpf was not affected by Sdox treatment. In conclusion, these findings demonstrate that Sdox displays a more favorable drug-like ADME/toxicity profile than doxorubicin, different selectivity towards cancer targets, along with a greater preclinical efficacy in resistant tumors. Therefore, Sdox represents a prototype of innovative anthracyclines, worthy of further investigations in clinical settings.
Collapse
Affiliation(s)
- Petko Alov
- Department of QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Merilin Al Sharif
- Department of QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Denitsa Aluani
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Konstantin Chegaev
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Jelena Dinic
- Department of Neurobiology, Institute for Biological Research Siniša Stanković, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Divac Rankov
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Miguel X. Fernandes
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, Spain
| | - Fabio Fusi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | | | - Risto Juvonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Magdalena Kondeva-Burdina
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - José M. Padrón
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, Spain
| | - Ilza Pajeva
- Department of QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Tania Pencheva
- Department of QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Adrián Puerta
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, Spain
| | - Hannu Raunio
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Chiara Riganti
- Department of Oncology, University of Torino, Torino, Italy
| | - Ivanka Tsakovska
- Department of QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Virginia Tzankova
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Yordan Yordanov
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Simona Saponara
- Department of Life Sciences, University of Siena, Siena, Italy
- *Correspondence: Simona Saponara,
| |
Collapse
|
108
|
Effects of bone marrow‐derived mesenchymal stem cells on doxorubicin‐induced liver injury in rats. J Biochem Mol Toxicol 2022; 36:e22985. [DOI: 10.1002/jbt.22985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 11/25/2021] [Accepted: 01/04/2022] [Indexed: 11/07/2022]
|
109
|
ORAK N, ÇAKMAK ARSLAN G, KAYA ST. An Evaluation of Damages Caused by Doxorubicin in Liver Tissue and Potential Protective Effect of Propolis on These Damages. KONURALP TIP DERGISI 2022. [DOI: 10.18521/ktd.1049919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
110
|
Alagal RI, AlFaris NA, Alshammari GM, ALTamimi JZ, AlMousa LA, Yahya MA. Kaempferol attenuates doxorubicin-mediated nephropathy in rats by activating SIRT1 signaling. J Funct Foods 2022. [DOI: 10.1016/j.jff.2021.104918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
111
|
Al-Qahtani WH, Alshammari GM, Ajarem JS, Al-Zahrani AY, Alzuwaydi A, Eid R, Yahya MA. Isoliquiritigenin prevents Doxorubicin-induced hepatic damage in rats by upregulating and activating SIRT1. Biomed Pharmacother 2022; 146:112594. [DOI: 10.1016/j.biopha.2021.112594] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/14/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023] Open
|
112
|
Wu L, Shi Y, Ni Z, Yu T, Chen Z. Preparation of a Self-Assembled Rhein-Doxorubicin Nanogel Targeting Mitochondria and Investigation on Its Antihepatoma Activity. Mol Pharm 2022; 19:35-50. [PMID: 34890210 DOI: 10.1021/acs.molpharmaceut.1c00565] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mitochondria are involved in the regulation of apoptosis, making them a promising target for the development of new anticancer drugs. Doxorubicin (DOX), a chemotherapeutic drug, can induce reactive oxygen species (ROS)-mediated apoptosis, improving its anticancer effects. Herein, Rhein, an active ingredient in rhubarb, with the capability of self-assembly and mitochondrial targeting, was used in conjunction with DOX to form efficient nanomaterials (Rhein-DOX nanogel) capable of sustained drug release. It was self-assembled with a hydrogen bond, π-π stacking interactions, and hydrophobic interactions as the main driving force, and its loading efficiency was up to 100%. Based on its self-assembly characteristics, we evaluated the mechanism of this material to target mitochondria, induce ROS production, and promote apoptosis. The IC50 of the Rhein-DOX nanogel (3.74 μM) was only 46.3% of that of DOX (11.89 μM), and the tumor inhibition rate of the Rhein-DOX nanogel was 79.4% in vivo, 2.3 times that of DOX. This study not only addresses the disadvantages of high toxicity of DOX and low bioavailability of Rhein, when DOX and Rhein are combined for the treatment of hepatoma, but it also significantly improved the synergistic antihepatoma efficacy of Rhein and DOX, which provides a new idea for the development of long-term antihepatoma agents with low toxicity.
Collapse
Affiliation(s)
- Li Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yan Shi
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zihui Ni
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tao Yu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhipeng Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
113
|
Abo-Golayel M, Fatani A, Baothman O, Shash L, Abuaraki H, Zeyadi M, Hosawi S, Altayb H. Hepatoprotective effect of date palm fruit extract against doxorubicin intoxication in Wistar rats: In vivo and in silico studies. Asian Pac J Trop Biomed 2022. [DOI: 10.4103/2221-1691.350184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
114
|
Han X, Long Y, Duan X, Liu Z, Hu X, Zhou J, Li N, Wang Y, Qin J. ZEB1 induces ROS generation through directly promoting MCT4 transcription to facilitate breast cancer. Exp Cell Res 2022; 412:113044. [DOI: 10.1016/j.yexcr.2022.113044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/31/2021] [Accepted: 01/22/2022] [Indexed: 12/17/2022]
|
115
|
Lu J, Shi TT, Yuan SS, Xie RR, Zhao RX, Zhu JJ, Yang JK. Cisapride induced hypoglycemia via the KCNH6 potassium channel. Front Endocrinol (Lausanne) 2022; 13:1011238. [PMID: 36325440 PMCID: PMC9618959 DOI: 10.3389/fendo.2022.1011238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Mutations in KCNH6 has been proved to cause hypoinsulinemia and diabetes in human and mice. Cisapride is a stomach-intestinal motility drug used to treat gastrointestinal dysfunction. Cisapride has been reported to be a potential inhibitor of the KCNH family, but it remained unclear whether cisapride inhibited KCNH6. Here, we discovered the role of cisapride on glucose metabolism, focusing on the KCNH6 potassium channel protein. Cisapride reduced blood glucose level and increased serum insulin secretion in wild-type (WT) mice fed standard normal chow/a high-fat diet or in db/db mice, especially when combined with tolbutamide. This effect was much stronger after 4 weeks of intraperitoneal injection. Whole-cell patch-clamp showed that cisapride inhibited KCNH6 currents in transfected HEK293 cells in a concentration-dependent manner. Cisapride induced an increased insulin secretion through the disruption of intracellular calcium homeostasis in a rat pancreatic β-cell line, INS-1E. Further experiments revealed that cisapride did not decrease blood glucose or increase serum insulin in KCNH6 β-cell knockout (Kcnh6-β-KO) mice when compared with WT mice. Cisapride also ameliorated glucose-stimulated insulin secretion (GSIS) in response to high glucose in WT but not Kcnh6-β-KO mice. Thus, our data reveal a novel way for the effect of KCNH6 in cisapride-induced hypoglycemia.
Collapse
Affiliation(s)
- Jing Lu
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ting-Ting Shi
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Sha-Sha Yuan
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Department of Endocrinology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Rong-Rong Xie
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ru-Xuan Zhao
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Juan-Juan Zhu
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jin-Kui Yang
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- *Correspondence: Jin-Kui Yang,
| |
Collapse
|
116
|
Mustafa S, Ijaz MU, ul Ain Q, Afsar T, Almajwal A, Shafique H, Razak S. OUP accepted manuscript. Toxicol Res (Camb) 2022; 11:475-485. [PMID: 35782651 PMCID: PMC9244725 DOI: 10.1093/toxres/tfac024] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/07/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Shama Mustafa
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad 38040, Pakistan
| | - Muhammad Umar Ijaz
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad 38040, Pakistan
| | - Qurat ul Ain
- Department of Zoology, Government College Women University, Sialkot 51310, Pakistan
| | - Tayyaba Afsar
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ali Almajwal
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Huma Shafique
- Institute of cellular medicine, Newcastle University Medical School, Newcastle University, Newcastle NE17RU, United Kingdom
| | - Suhail Razak
- Corresponding author: Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
117
|
Diosmin Alleviates Doxorubicin-Induced Liver Injury via Modulation of Oxidative Stress-Mediated Hepatic Inflammation and Apoptosis via NfkB and MAPK Pathway: A Preclinical Study. Antioxidants (Basel) 2021; 10:antiox10121998. [PMID: 34943101 PMCID: PMC8698866 DOI: 10.3390/antiox10121998] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022] Open
Abstract
Hepatotoxicity caused by chemotherapeutic drugs (e.g., doxorubicin) is of critical concern in cancer therapy. This study focused on investigating the modulatory effects of diosmin against doxorubicin-induced hepatotoxicity in Male Wistar rats. Male Wistar rats were randomly divided into four groups: Group I was served as control, Group II was treated with doxorubicin (20 mg/kg, intraperitoneal, i.p.), Group III was treated with a combination of doxorubicin and low-dose diosmin (100 mg/kg orally), and Group IV was treated with a combination of doxorubicin and high-dose diosmin (200 mg/kg orally) supplementation. A single dose of doxorubicin (i.p.) caused hepatic impairment, as shown by increases in the concentrations of serum alanine aminotransferase, aspartate aminotransferase, and alkaline phosphatase. Doxorubicin produced histological abnormalities in the liver. In addition, a single injection of doxorubicin increased lipid peroxidation and reduced glutathione, catalase, and superoxide dismutase (SOD) levels. Importantly, pre-treatment with diosmin restored hepatic antioxidant factors and serum enzymatic activities and reduced the inflammatory and apoptotic-mediated proteins and genes. These findings demonstrate that diosmin has a protective effect against doxorubicin-induced hepatotoxicity.
Collapse
|
118
|
Yu XF, Zhou ZL, Lu XD, Long SQ. Hepatoprotective effect of naringenin in rats with alcoholic liver disease. Shijie Huaren Xiaohua Zazhi 2021; 29:1334-1340. [DOI: 10.11569/wcjd.v29.i23.1334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Previous studies have shown that naringenin (Nar) can play a protective role in animal models with acute liver injury, but its role in alcoholic liver disease (ALD) remains unclear.
AIM To explore the effect of Nar on ALD rats and the possible mechanism involved.
METHODS Forty Sprague-Dawley rats were randomly divided into a control group, model group, low-dose Nar group, and high-dose Nar group, with 10 rats in each group. A rat model of ALD was generated by alcohol induction. Blood samples and liver tissues were collected at the end of the regimen. The levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) in serum were determined with an automatic biochemical analyzer. The morphology of the liver was observed by HE staining. Glutathione peroxidase (GPX) and superoxide dismutase (SOD) activities, malondialdehyde (MDA) content, and reactive oxygen species (ROS) production level in the liver were determined with commercial kits. The levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in liver tissues were detected by immunohistochemical staining. The expression levels of Nrf-2, HO-1, and p-NF-κB P65 in liver tissues were detected by Western blot.
RESULTS Compared with the model group, the levels of AST and AST in serum of rats in the low- and high-dose Nar groups were significantly decreased, the activities of SOD and GPx and the expression levels of Nrf-2 and HO-1 in liver tissue were significantly increased, and MDA content, ROS level, and TNF-α, IL-6, and p-NF-κB P65 expression levels were significantly decreased, especially in the high-dose group.
CONCLUSION Nar can alleviate liver injury in ALD rats, and this effect may be related to the reduction of oxidative stress and inflammatory response in liver tissue.
Collapse
Affiliation(s)
- Xiu-Feng Yu
- Zhejiang Chinese Medical University, Hangzhou 310051, Zhejiang Province, China,Department of Emergency Medicine, Lishui City People's Hospital, Lishui 323000, Zhejiang Province, China
| | - Zeng-Li Zhou
- Department of Gastroenterology, Lishui City People's Hospital, Lishui 323000, Zhejiang Province, China
| | - Xu-Dong Lu
- Department of Emergency Medicine, Lishui City People's Hospital, Lishui 323000, Zhejiang Province, China
| | - Si-Qin Long
- Department of Infectious Diseases, Lishui City People's Hospital, Lishui 323000, Zhejiang Province, China
| |
Collapse
|
119
|
Materón EM, Shimizu FM, Figueiredo Dos Santos K, Nascimento GF, Geraldo VPN, Oliveira ON, Faria RC. Membrane model as key tool in the study of glutathione-s-transferase mediated anticancer drug resistance. Biomed Pharmacother 2021; 145:112426. [PMID: 34861633 DOI: 10.1016/j.biopha.2021.112426] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/05/2021] [Accepted: 11/12/2021] [Indexed: 11/02/2022] Open
Abstract
Glutathione-s-transferase is believed to be involved in the resistance to chemotherapeutic drugs, which depends on the interaction with the cell membranes. In this study, we employed Langmuir monolayers of a mixture of phospholipids and cholesterol (MIX) as models for tumor cell membranes and investigated their interaction with the anticancer drugs cisplatin (CDDP) and doxorubicin (DOX). We found that both DOX and CDDP expand and affect the elasticity of MIX monolayers, but these effects are hindered when glutathione-s-transferase (GST) and its cofactor glutathione (GSH) are incorporated. Changes are induced by DOX or CDDP on the polarization-modulated infrared reflection absorption spectroscopy (PM-IRRAS) data for MIX/GST/GSH monolayers, thus denoting some degree of interaction that is not sufficient to alter the monolayer mechanical properties. Overall, the results presented here give support to the hypothesis of the inactivation of DOX and CDDP by GST and point to possible directions to detect and fight drug resistance.
Collapse
Affiliation(s)
- Elsa M Materón
- Chemistry Department, Federal University of São Carlos, CP 676, São Carlos 13565-905, São Paulo, Brazil; São Carlos Institute of Physics, University of São Paulo, P.O Box 369, 13560-970 São Carlos, SP, Brazil.
| | - Flavio M Shimizu
- São Carlos Institute of Physics, University of São Paulo, P.O Box 369, 13560-970 São Carlos, SP, Brazil; Department of Applied Physics, "Gleb Wataghin" Institute of Physics (IFGW), University of Campinas (UNICAMP), Campinas, SP 13083-859, Brazil.
| | | | - Gustavo F Nascimento
- São Carlos Institute of Physics, University of São Paulo, P.O Box 369, 13560-970 São Carlos, SP, Brazil
| | - Vananélia P N Geraldo
- São Carlos Institute of Physics, University of São Paulo, P.O Box 369, 13560-970 São Carlos, SP, Brazil
| | - Osvaldo N Oliveira
- São Carlos Institute of Physics, University of São Paulo, P.O Box 369, 13560-970 São Carlos, SP, Brazil.
| | - Ronaldo C Faria
- Chemistry Department, Federal University of São Carlos, CP 676, São Carlos 13565-905, São Paulo, Brazil.
| |
Collapse
|
120
|
Farag MR, Moselhy AAA, El-Mleeh A, Aljuaydi SH, Ismail TA, Di Cerbo A, Crescenzo G, Abou-Zeid SM. Quercetin Alleviates the Immunotoxic Impact Mediated by Oxidative Stress and Inflammation Induced by Doxorubicin Exposure in Rats. Antioxidants (Basel) 2021; 10:antiox10121906. [PMID: 34943009 PMCID: PMC8750303 DOI: 10.3390/antiox10121906] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 11/16/2022] Open
Abstract
Doxorubicin (DOX) is a chemotherapeutic agent against hematogenous and solid tumors with undesirable side effects including immunosuppression. Quercetin (QUR), a natural flavonoid abundant in fruits and vegetables, has a potent antioxidant activity. The aim of the current study was to assess the impact of QUR on DOX-induced hematological and immunological dysfunctions in a rodent model. Randomly grouped rats were treated as follows: control, QUR alone (50 mg/kg for 15 days per os), DOX alone (2.5 mg/kg I/P, three times a week, for two weeks), and co-treated rats with QUR for 15 days prior to and concomitantly with DOX (for two weeks), at the doses intended for groups two and three. DOX alone significantly disrupted the erythrogram and leukogram variables. Serum immunoglobulin (IgG, IgM, and IgE) levels and the activities of catalase (CAT) and superoxide dismutase (SOD) in spleen were declined. The DNA damage traits in spleen were elevated with an upregulation of the expression of the apoptotic markers (p53 and Caspase-3 genes) and the proinflammatory cytokines (IL-6 and TNF-α genes), while the expression of CAT gene was downregulated. These biochemical changes were accompanied by morphological changes in the spleen of DOX-treated rats. Co-treatment with QUR abated most of the DOX-mediated alterations in hematological variables, serum immunoglobulins, and spleen antioxidant status, pro-inflammatory and apoptotic responses, and histopathological alterations. In essence, these data suggest that QUR alleviated DOX-induced toxicities on the bone marrow, spleen, and antibody-producing cells. Supplementation of chemotherapy patients with QUR could circumvent the DOX-induced inflammation and immunotoxicity, and thus prevent chemotherapy failure.
Collapse
Affiliation(s)
- Mayada R. Farag
- Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
- Correspondence: (M.R.F.); (A.D.C.)
| | - Attia A. A. Moselhy
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt;
| | - Amany El-Mleeh
- Department of Pharmacology, Faculty of Veterinary Medicine, Menoufia University, Shebin Elkoum 32511, Egypt;
| | - Samira H. Aljuaydi
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt;
| | - Tamer Ahmed Ismail
- Department of Clinical Laboratory Sciences, Turabah University College, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Alessandro Di Cerbo
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Matelica, Italy
- Correspondence: (M.R.F.); (A.D.C.)
| | - Giuseppe Crescenzo
- Department of Veterinary Medicine, University of Bari ‘Aldo Moro’, 70121 Bari, Italy;
| | - Shimaa M. Abou-Zeid
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City 6012201, Egypt;
| |
Collapse
|
121
|
Qi X, Li S, Song X, Gong Y, Guo Z, Cui C, Wang X, Tan Z. An Fe‐MIL100 Based Drug Delivery System for pH and Glutathione Dual‐Responsive Drug Release. ChemistrySelect 2021. [DOI: 10.1002/slct.202103551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Xiuyu Qi
- State Key Laboratory of Fine Chemicals School of Chemical Engineering Dalian University of Technology Panjin Liaoning 124221 P. R. China
| | - Shanshan Li
- School of Life Science and Medicine Dalian University of Technology Panjin Liaoning 124221 P. R. China
| | - Xue‐Zhi Song
- State Key Laboratory of Fine Chemicals School of Chemical Engineering Dalian University of Technology Panjin Liaoning 124221 P. R. China
| | - Yishu Gong
- State Key Laboratory of Fine Chemicals School of Chemical Engineering Dalian University of Technology Panjin Liaoning 124221 P. R. China
| | - Zhaoming Guo
- School of Life Science and Medicine Dalian University of Technology Panjin Liaoning 124221 P. R. China
| | - Changhao Cui
- School of Life Science and Medicine Dalian University of Technology Panjin Liaoning 124221 P. R. China
| | - Xiaofeng Wang
- School of Mathematics and Physics Science Dalian University of Technology Panjin Liaoning 124221 P. R. China
| | - Zhenquan Tan
- State Key Laboratory of Fine Chemicals School of Chemical Engineering Dalian University of Technology Panjin Liaoning 124221 P. R. China
| |
Collapse
|
122
|
Lages EB, Fernandes RS, Andrade MMS, Paiyabhroma N, de Oliveira RB, Fernandes C, Cassali GD, Sicard P, Richard S, Branco de Barros AL, Ferreira LAM. pH-sensitive doxorubicin-tocopherol succinate prodrug encapsulated in docosahexaenoic acid-based nanostructured lipid carriers: An effective strategy to improve pharmacokinetics and reduce toxic effects. Biomed Pharmacother 2021; 144:112373. [PMID: 34794238 DOI: 10.1016/j.biopha.2021.112373] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/16/2021] [Accepted: 10/25/2021] [Indexed: 12/13/2022] Open
Abstract
Side effects often limit the use of doxorubicin (DOX) in cancer treatment. We have recently developed a nanostructured lipid carrier (NLC) formulation for synergistic chemotherapy, encapsulating DOX and the anticancer adjuvants docosahexaenoic acid (DHA) and α-tocopherol succinate (TS). Hydrophobic ion-pairing with TS allowed a high DOX entrapment in the nanocarrier. In this work, we investigated the pharmacokinetics of this formulation after intravenous administration in mice. The first data obtained led us to propose synthesizing covalent DOX-TS conjugates to increase DOX retention in the NLC. We successfully conjugated DOX to TS via an amide or hydrazone bond. In vitro studies in 4T1 tumor cells indicated low cytotoxicity of the amide derivative, while the hydrazone conjugate was effective in killing cancer cells. We encapsulated the hydrazone derivative in a DHA-based nanocarrier (DOX-hyd-TS/NLC), which had reduced particle size and high drug encapsulation efficiency. The pH-sensitive hydrazone bond allowed controlled DOX release from the NLC, with increased drug release at acidic conditions. In vivo studies revealed that DOX-hyd-TS/NLC had a better pharmacokinetic profile than free DOX and attenuated the short-term cardiotoxic effects caused by DOX, such as QT prolongation and impaired left ventricular systolic function. Moreover, this formulation showed excellent therapeutic performance by reducing tumor growth in 4T1 tumor-bearing mice and decreasing DOX-induced toxicity to the heart and liver, demonstrated by hematologic, biochemical, and histologic analyses. These results indicate that DOX-hyd-TS/NLC may be a promising nanocarrier for breast cancer treatment.
Collapse
Affiliation(s)
- Eduardo Burgarelli Lages
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Renata Salgado Fernandes
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marina Mol Sena Andrade
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Renata Barbosa de Oliveira
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Christian Fernandes
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Geovanni Dantas Cassali
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pierre Sicard
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France; IPAM, BioCampus Montpellier, INSERM, CNRS, Université de Montpellier, Montpellier, France
| | - Sylvain Richard
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France; IPAM, BioCampus Montpellier, INSERM, CNRS, Université de Montpellier, Montpellier, France
| | - André Luís Branco de Barros
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lucas Antônio Miranda Ferreira
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
123
|
Sirwi A, Shaik RA, Alamoudi AJ, Eid BG, Kammoun AK, Ibrahim SRM, Mohamed GA, Abdallah HM, Abdel-Naim AB. Mokko Lactone Attenuates Doxorubicin-Induced Hepatotoxicity in Rats: Emphasis on Sirt-1/FOXO1/NF-κB Axis. Nutrients 2021; 13:nu13114142. [PMID: 34836397 PMCID: PMC8621765 DOI: 10.3390/nu13114142] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 12/28/2022] Open
Abstract
Doxorubicin (DOX), a common chemotherapeutic agent, suffers serious adverse effects including hepatotoxicity. Mokko lactone (ML) is a guainolide sesquiterpene with promising biological activities. The study aimed to evaluate the protection offered by ML against hepatotoxicity induced by DOX in rats. Our data indicated ML exhibited protective effects as evidenced by ameliorating the rise in serum activities of alanine transaminase, aspartate transaminase and alkaline phosphatase. This was confirmed histologically as ML prevented DOX-induced pathological alteration in liver architecture. Further, ML administration significantly prevented malondialdehyde accumulation, glutathione depletion and superoxide dismutase and catalase exhaustion. Antioxidant action of ML was associated with enhanced expression of the nuclear translocation of NF-E2-related factor 2 (Nrf2) and a lower expression of forkhead box protein O1 (FOXO1). Also, ML showed potent anti-inflammatory activities highlighted by decreased expression of interleukin 6, tumor necrosis factor α and nuclear factor κB (NF-κB). The anti-apoptotic effects of ML were associated with decreased Bax and enhanced Bcl-2 mRNA expression in liver tissues. ML caused a significant up-regulation in the expression of silent information regulator 1 (Sirt-1). Therefore, it can be concluded that ML prevents liver injury caused by DOX. This could partially be due to the ML regulatory activities on Sirt-1/FOXO1/NF-κB axis.
Collapse
Affiliation(s)
- Alaa Sirwi
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.S.); (G.A.M.); (H.M.A.)
| | - Rasheed A. Shaik
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (R.A.S.); (A.J.A.); (B.G.E.)
| | - Abdulmohsin J. Alamoudi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (R.A.S.); (A.J.A.); (B.G.E.)
| | - Basma G. Eid
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (R.A.S.); (A.J.A.); (B.G.E.)
| | - Ahmed K. Kammoun
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Sabrin R. M. Ibrahim
- Batterjee Medical College, Preparatory Year Program, Jeddah 21442, Saudi Arabia;
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Gamal A. Mohamed
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.S.); (G.A.M.); (H.M.A.)
- Department of Pharmacognosy, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Hossam M. Abdallah
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.S.); (G.A.M.); (H.M.A.)
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Ashraf B. Abdel-Naim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (R.A.S.); (A.J.A.); (B.G.E.)
- Correspondence:
| |
Collapse
|
124
|
Kara H, Orem A, Yulug E, Balaban Yucesan F, Kerimoglu G, Vanizor Kural B, Ozer Yaman S, Bodur A, Turedi S, Alasalvar C. Effects of hazelnut supplemented diet on doxorubicin-induced damage of reproductive system in male rats. J Food Biochem 2021; 45:e13973. [PMID: 34664725 DOI: 10.1111/jfbc.13973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/26/2021] [Accepted: 09/28/2021] [Indexed: 11/27/2022]
Abstract
The present study was objected to investigate the effect of hazelnut supplemented diet on the levels of oxidative stress and fertility parameters against doxorubicin-induced testicular and epididymal tissue damage of male rats. Rats were randomly divided into four groups (each n = 8), namely control group (CG), doxorubicin group (DG), doxorubicin + hazelnut group (DHG), and doxorubicin + vitamin E group (DEG). This is the first study designed using DHG. Doxorubicin was intraperitoneally injected into all diet groups except CG at a dose of 3 mg/kg body weight on days 1, 7, 14, 21, and 28. In addition, DHG was supplemented with a hazelnut diet at a dose of 3 g/kg body weight/day and vitamin E was added to the drinking water of DEG at a dose of 50 mg/kg body weight/day. DHG reversed the side effects of doxorubicin and positively improved the epididymis sperm quality, testicular and epididymal tissue injury, testosterone level, epididymis oxidative stress index, and lipid peroxidation in male rats. These findings suggest that hazelnut has positive effects against doxorubicin dependent damage on male rats and it may be a promising supplement for amelioration of testicular toxicity. PRACTICAL APPLICATIONS: Hazelnut has numerous positive health effects due to its macronutrients, micronutrients, lipid-soluble compounds and bioactive phenolics. Studies have shown that regular consumption of hazelnut may have a positive effect on lipid parameters, oxidative stress, inflammation markers, and endothelial dysfunction in both healthy people and patients with chronic diseases. Although doxorubicin (Adriamycin, DOX) is an antibiotic that has been widely used in cancer treatment for nearly 30 years, it causes organ toxicity including testicular tissue. Hazelnut may have positive effects on the damage caused by DOX in the reproductive system. However, studies on the effect of hazelnut on male reproductive health are scarce. Therefore, this study provided a basis for the clinical evaluation of the effects of hazelnut on the reproductive system.
Collapse
Affiliation(s)
- Hanife Kara
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Amasya University, Amasya, Turkey.,Graduate School of Health Sciences, Department of Medical Biochemistry, Karadeniz Technical University, Trabzon, Turkey
| | - Asım Orem
- Faculty of Medicine, Department of Medical Biochemistry, Karadeniz Technical University, Trabzon, Turkey
| | - Esin Yulug
- Faculty of Medicine, Department of Histology and Embryology, Karadeniz Technical University, Trabzon, Turkey
| | - Fulya Balaban Yucesan
- Faculty of Medicine, Department of Medical Biochemistry, Karadeniz Technical University, Trabzon, Turkey
| | - Gokcen Kerimoglu
- Faculty of Medicine, Department of Histology and Embryology, Karadeniz Technical University, Trabzon, Turkey
| | - Birgul Vanizor Kural
- Faculty of Medicine, Department of Medical Biochemistry, Karadeniz Technical University, Trabzon, Turkey
| | - Serap Ozer Yaman
- Graduate School of Health Sciences, Department of Medical Biochemistry, Karadeniz Technical University, Trabzon, Turkey
| | - Akın Bodur
- Graduate School of Health Sciences, Department of Medical Biochemistry, Karadeniz Technical University, Trabzon, Turkey
| | - Sibel Turedi
- Faculty of Medicine, Department of Histology and Embryology, Harran University, Sanlıurfa, Turkey
| | | |
Collapse
|
125
|
Mirzaei S, Abadi AJ, Gholami MH, Hashemi F, Zabolian A, Hushmandi K, Zarrabi A, Entezari M, Aref AR, Khan H, Ashrafizadeh M, Samarghandian S. The involvement of epithelial-to-mesenchymal transition in doxorubicin resistance: Possible molecular targets. Eur J Pharmacol 2021; 908:174344. [PMID: 34270987 DOI: 10.1016/j.ejphar.2021.174344] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 06/30/2021] [Accepted: 07/11/2021] [Indexed: 12/14/2022]
Abstract
Considering the fact that cancer cells can switch among various molecular pathways and mechanisms to ensure their progression, chemotherapy is no longer effective enough in cancer therapy. As an anti-tumor agent, doxorubicin (DOX) is derived from Streptomyces peucetius and can induce cytotoxicity by binding to topoisomerase enzymes to suppress DNA replication, leading to apoptosis and cell cycle arrest. However, efficacy of DOX in suppressing cancer progression is restricted by development of drug resistance. Cancer cells elevate their metastasis in triggering DOX resistance. The epithelial-to-mesenchymal transition (EMT) mechanism participates in transforming epithelial cells into mesenchymal cells that have fibroblast-like features. The EMT diminishes intercellular adhesion and enhances migration of cells that are necessary for carcinogenesis. Various oncogenic molecular pathways stimulate EMT in cancer. EMT can induce DOX resistance, and in this way, upstream mediators such as ZEB proteins, microRNAs, Twist1 and TGF-β play a significant role. Identification of molecular pathways involved in EMT regulation and DOX resistance has resulted in using gene therapy such as microRNA transfection and siRNA in overcoming chemoresistance. Furthermore, curcumin and formononetin, owing to their cytotoxicity against cancer cells, can suppress EMT in mediating DOX sensitivity. For promoting efficacy in DOX sensitivity, nanoparticles have been developed for boosting ability in EMT inhibition.
Collapse
Affiliation(s)
- Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Asal Jalal Abadi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Farid Hashemi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Translational Sciences, Xsphera Biosciences Inc. 6 Tide Street, Boston, MA, 02210, USA
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, 23200, Pakistan.
| | - Milad Ashrafizadeh
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey; Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956, Istanbul, Turkey.
| | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
126
|
Sahlan M, Rizka Alia Hapsari N, Diah Pratami K, Cahya Khayrani A, Lischer K, Alhazmi A, Mohammedsaleh ZM, Shater AF, Saleh FM, Alsanie WF, Sayed S, Gaber A. Potential hepatoprotective effects of flavonoids contained in propolis from South Sulawesi against chemotherapy agents. Saudi J Biol Sci 2021; 28:5461-5468. [PMID: 34588856 PMCID: PMC8459154 DOI: 10.1016/j.sjbs.2021.08.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/03/2021] [Accepted: 08/08/2021] [Indexed: 02/01/2023] Open
Abstract
The use of doxorubicin and epirubicin as chemotherapy agent causes side effects such as liver damage due to oxidative stress by reactive oxygen species (ROS) that cause increased of ALT and AST level as liver parameter. One source of natural antioxidants as ROS neutralizer comes from flavonoid that contain in propolis. Most researchers claim that flavonoid can be used to protect the liver. The aim of this study was to test the hepatoprotective effect of flavonoid in propolis from South Sulawesi against doxorubicin and epirubicin. The experiment included male Sprague dawley rats divided into nine groups. The rats received the microcapsule propolis or the quercetin orally for 15 days. The hepatotoxicity was promoted by injection epirubicin and doxorubicin (i.v.) with a cumulative dose of 9 mg/kg. In this study, total polyphenol and flavonoid tests of propolis have been carried out, there were 1.1% polyphenols and 2.7% flavonoids, the antioxidant activity tests showed IC50 value of 9849 ppm and LCMS/MS tests supported the presence of phenolic compounds in propolis from South Sulawesi. Liver parameter was measured and the results showed that the propolis 200 mg/kg group produced the lowest ALT and had potential protective effect against doxorubicin and epirubicin-induced hepatotoxicity.
Collapse
Affiliation(s)
- Muhamad Sahlan
- Department of Chemical Engineering, Faculty of Engineering, Universitas Indonesia, West Java, 16424, Depok, Indonesia.,Research Center for Biomedical Engineering, Faculty of Engineering, Universitas Indonesia, West Java, 16424, Depok, Indonesia
| | - Nur Rizka Alia Hapsari
- Department of Chemical Engineering, Faculty of Engineering, Universitas Indonesia, West Java, 16424, Depok, Indonesia
| | | | - Apriliana Cahya Khayrani
- Department of Chemical Engineering, Faculty of Engineering, Universitas Indonesia, West Java, 16424, Depok, Indonesia
| | - Kenny Lischer
- Department of Chemical Engineering, Faculty of Engineering, Universitas Indonesia, West Java, 16424, Depok, Indonesia.,Research Center for Biomedical Engineering, Faculty of Engineering, Universitas Indonesia, West Java, 16424, Depok, Indonesia
| | - Alaa Alhazmi
- Medical Laboratory Technology Department, Jazan University, Jazan, Saudi Arabia.,SMIRES for Consultation in Specialized Medical Laboratories, Jazan University, Jazan, Saudi Arabia
| | - Zuhair M Mohammedsaleh
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Abdullah F Shater
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Fayez M Saleh
- Department of Medical Microbiology, Faculty of Medicine, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Walaa F Alsanie
- Center of Biomedical Sciences Research (CBSR), Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.,Department of Clinical Laboratories Sciences, The Faculty of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Samy Sayed
- Department of Science and Technology, University College-Ranyah, Taif University, B.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ahmed Gaber
- Center of Biomedical Sciences Research (CBSR), Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.,Department of Biology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
127
|
Bai H, Kong F, Feng K, Zhang X, Dong H, Liu D, Ma M, Liu F, Gu N, Zhang Y. Prussian Blue Nanozymes Prevent Anthracycline-Induced Liver Injury by Attenuating Oxidative Stress and Regulating Inflammation. ACS APPLIED MATERIALS & INTERFACES 2021; 13:42382-42395. [PMID: 34473471 DOI: 10.1021/acsami.1c09838] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Anthracycline-induced liver injury (AILI) is becoming an increasingly serious and potential clinical complication and is linked to reactive oxygen species (ROS) production and subsequent inflammatory response. Herein, we demonstrated that artificial Prussian blue nanozymes (PBZs) prevented daunorubicin-induced liver injury, a prototype of AILI, by attenuating ROS production and regulating inflammation. PBZs exhibited multienzyme activity and could scavenge ROS and free radicals. At the cellular level, PBZs could effectively eliminate ROS, suppress hepatocyte apoptosis, reduce deoxyribonucleic acid damage, and decrease the levels of inflammatory cytokines and chemokines. According to the results of the in vivo study, pretreatment with PBZs also resulted in a desirable protective effect against AILI, as indicated by both a decrease in biochemical indicator levels and hepatocyte necrosis. PBZs upregulated antioxidative genes by activating the Nrf2 pathway to reduce oxidative stress. Meanwhile, PBZs counteracted the inflammatory response based on the decreased expression levels of myeloperoxidase and F4/80 in the liver. Collectively, our findings indicate that PBZ-based nanotherapy is a novel strategy for protecting against AILI.
Collapse
Affiliation(s)
- Huiyuan Bai
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing 210096, P. R. China
| | - Fei Kong
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing 210096, P. R. China
| | - Kaizheng Feng
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing 210096, P. R. China
| | - Xuan Zhang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing 210096, P. R. China
| | - Haijiao Dong
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing 210096, P. R. China
| | - Di Liu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing 210096, P. R. China
| | - Ming Ma
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing 210096, P. R. China
| | - Fangzhou Liu
- Department of Head & Neck Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing 210096, P. R. China
| | - Yu Zhang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing 210096, P. R. China
| |
Collapse
|
128
|
Dehpour AR, Yousefi-Manesh H, Sheibani M, Sadeghi MA, Hemmati S, Noori T, Shirooie S. Evaluation of Anti-inflammatory and Antioxidant Effects of Sumatriptan on Carbon Tetrachloride-induced Hepatotoxicity in Rats. Drug Res (Stuttg) 2021; 72:41-46. [PMID: 34500479 DOI: 10.1055/a-1589-5395] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The liver detoxifies and metabolizes many drugs and xenobiotics which may cause hepatotoxicity due to some toxic agents. Carbon tetrachloride (CCl4) is metabolized in cytochrome P450 and its reactive radical metabolites cause lipid peroxidation, cellular injury, and apoptosis. Sumatriptan (SUM), 5-HT1B/1D receptor agonist, had anti-inflammatory and anti-oxidant effects. In this research the effect of SUM pre-treatment against CCl4-induced hepatotoxicity was examined. Adult rats received SUM (0.1, 0.3 and 1 mg/kg; i.p.) for 3 consecutive days before CCl4 (2 ml/kg; i.p. on the 3rd day). The aminotransferases serum levels, tissue levels of anti-oxidant and pro-inflammatory markers and histopathological examination were evaluated. SUM (0.3 mg/kg) prevented significantly the elevation of aminotransferases versus the control group (CCl4 group) (P<0.0001) and also, reversed meaningfully the changes of the MPO, MDA, SOD and CAT, IL-1β and TNF-α levels. Additionally, CCl4-intoxication resulted to the disruption of lobular and cellular structures and inflammation in histopathological evaluation which is prevented by SUM (0.3 mg/kg). These data revealed that SUM (0.3 mg/kg), but no at doses 0.1 and 1 mg/kg, decreases the hepatotoxicity of induced by CCl4 in rats.
Collapse
Affiliation(s)
- Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hasan Yousefi-Manesh
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sheibani
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Sadeghi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Hemmati
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Tayebeh Noori
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Samira Shirooie
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
129
|
Zhao W, Chen L, Zhou H, Deng C, Han Q, Chen Y, Wu Q, Li S. Protective effect of carvacrol on liver injury in type 2 diabetic db/db mice. Mol Med Rep 2021; 24:741. [PMID: 34435648 PMCID: PMC8430346 DOI: 10.3892/mmr.2021.12381] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 02/24/2021] [Indexed: 12/11/2022] Open
Abstract
The present study aimed to investigate the protective effect of carvacrol on liver injury in mice with type 2 diabetes mellitus (T2DM) and to assess its potential molecular mechanism. Mice were divided into three groups (n=15/group): Non-diabetic db/m+ mice group, db/db mice group and db/db mice + carvacrol group. In the db/db mice + carvacrol group, db/db mice were administered 10 mg/kg carvacrol daily by gavage for 6 weeks. Fasting blood glucose and insulin levels were separately examined. Pathological changes were observed using hematoxylin and eosin, Masson's trichrome, periodic acid Schiff and reticular fiber staining. In addition, immunohistochemistry, immunofluorescence and western blotting were used to examine the expression levels of Toll-like receptor 4 (TLR4), NF-κB, NALP3, AKT1, phosphorylated (p)-AKT1, insulin receptor (INSR), p-INSR, mTOR, p-mTOR, insulin receptor substrate 1 (IRS1) and p-IRS1 in the liver tissues. The results revealed that carvacrol improved blood glucose and insulin resistance of T2DM db/db mice. After treatment with carvacrol for 6 weeks, the serum levels of TC, TG and LDL-C were markedly reduced, whereas HDL-C levels were significantly increased in db/db mice. Furthermore, carvacrol administration significantly decreased serum ALT and AST levels in db/db mice. Serum BUN, Cre and UA levels were markedly higher in db/db mice compared with those in the control group; however, carvacrol treatment markedly reduced their serum levels in db/db mice. Furthermore, histological examinations confirmed that carvacrol could protect the liver of db/db mice. Carvacrol could ameliorate liver injury induced by T2DM via mediating insulin, TLR4/NF-κB and AKT1/mTOR signaling pathways. The present findings suggested that carvacrol exerted protective effects on the liver in T2DM db/db mice, which could be related to insulin, TLR4/NF-κB and AKT1/mTOR signaling pathways.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Endocrinology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550003, P.R. China
| | - Li Chen
- Department of Prevention and Health Care, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550003, P.R. China
| | - Heng Zhou
- Department of Endocrinology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550003, P.R. China
| | - Chunyan Deng
- Department of Endocrinology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550003, P.R. China
| | - Qizhen Han
- Department of Endocrinology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550003, P.R. China
| | - Yonghua Chen
- Department of Endocrinology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550003, P.R. China
| | - Qing Wu
- Department of Endocrinology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550003, P.R. China
| | - Shanshan Li
- Department of Endocrinology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550003, P.R. China
| |
Collapse
|
130
|
Hsu CM, Yang MY, Tsai MS, Chang GH, Yang YH, Tsai YT, Wu CY, Chang SF. Dihydroisotanshinone I as a Treatment Option for Head and Neck Squamous Cell Carcinomas. Int J Mol Sci 2021; 22:ijms22168881. [PMID: 34445585 PMCID: PMC8396193 DOI: 10.3390/ijms22168881] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/15/2021] [Accepted: 08/16/2021] [Indexed: 11/17/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are the most common cancers of the head and neck, and their prevalence is rapidly increasing. HNSCCs present a clinical challenge because of their high recurrence rate, therapeutic resistance to radiation and chemotherapy drugs, and adverse effects. Hence, traditional Chinese herbal treatment may be advantageous to therapeutic strategies for HNSCCs. Danshen (Salvia miltiorrhiza), a well-known Chinese herb, has been extensively applied in treatments for various diseases, including cancer, because of its high degree of safety and low rate of adverse effects despite its unclear mechanism. Thus, we aimed to explore the possible anticancer effects and mechanisms of dihydroisotanshinone I (DT), a compound in danshen (extract from danshen), on HNSCCs. Three HNSCCs cell lines were used for in vitro studies, and a Detroit 562 xenograft mouse model was chosen for in vivo studies. Our in vitro results showed that DT could initiate apoptosis, resulting in cell death, and the p38 signaling partially regulated DT-initiated cell apoptosis in the Detroit 562 model. In the xenograft mouse model, DT reduced tumor size with no obvious adverse effect of hepatotoxicity. The present study suggests that DT is a promising novel candidate for anti-HNSCCs therapy.
Collapse
Affiliation(s)
- Cheng-Ming Hsu
- Department of Otolaryngology-Head and Neck Surgery, Chiayi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (C.-M.H.); (M.-S.T.); (G.-H.C.); (Y.-T.T.)
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ming-Yu Yang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Ming-Shao Tsai
- Department of Otolaryngology-Head and Neck Surgery, Chiayi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (C.-M.H.); (M.-S.T.); (G.-H.C.); (Y.-T.T.)
| | - Geng-He Chang
- Department of Otolaryngology-Head and Neck Surgery, Chiayi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (C.-M.H.); (M.-S.T.); (G.-H.C.); (Y.-T.T.)
| | - Yao-Hsu Yang
- Department of Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan;
| | - Yao-Te Tsai
- Department of Otolaryngology-Head and Neck Surgery, Chiayi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (C.-M.H.); (M.-S.T.); (G.-H.C.); (Y.-T.T.)
| | - Ching-Yuan Wu
- Department of Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan;
- School of Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Correspondence: (C.-Y.W.); (S.-F.C.)
| | - Shun-Fu Chang
- Department of Medical Research and Development, Chiayi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
- Correspondence: (C.-Y.W.); (S.-F.C.)
| |
Collapse
|
131
|
Goda AE, Elenany AM, Elsisi AE. Novel in vivo potential of trifluoperazine to ameliorate doxorubicin-induced cardiotoxicity involves suppression of NF-κB and apoptosis. Life Sci 2021; 283:119849. [PMID: 34343539 DOI: 10.1016/j.lfs.2021.119849] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 07/13/2021] [Accepted: 07/17/2021] [Indexed: 11/25/2022]
Abstract
AIMS Cardiotoxicity of doxorubicin frequently complicates treatment outcome. Aberrantly activated calcium/calmodulin pathway can eventually trigger signaling cascades that mediate cardiotoxicity. Therefore, we tested the hypothesis that trifluoperazine, a strong calmodulin antagonist, may alleviate this morbidity. MATERIALS AND METHODS Heart failure and cardiotoxicity were assessed via echocardiography, PCR, immunohistochemistry, histopathology, Masson's trichrome staining and transmission electron microscopy. Whereas liver and kidney structural and functional alterations were evaluated histopathologically and biochemically. KEY FINDINGS Results revealed that combination treatment with trifluoperazine could overcome doxorubicin-induced heart failure with reduced ejection fraction. Moreover, heart weight/body weight ratio and histopathological examination showed that trifluoperazine mitigated doxorubicin-induced cardiac atrophy, inflammation and myofibril degeneration. Transmission electron microscopy further confirmed the marked restoration of the left ventricular ultrastructures by trifluoperazine pretreatment. In addition, Masson's trichrome staining revealed that trifluoperazine could significantly inhibit doxorubicin-induced left ventricular remodeling by fibrosis. Of note, doxorubicin induced the expression of myocardial nuclear NF-κB-p65 and caspase-3 which were markedly inhibited by trifluoperazine, suggesting that cardioprotection conferred by trifluoperazine involved, at least in part, suppression of NF-κB and apoptosis. Furthermore, biochemical and histopathological examinations showed that trifluoperazine improved doxorubicin-induced renal and hepatic impairments both functionally and structurally. SIGNIFICANCE In conclusion, the present in vivo study is the first to provide evidences underscoring the protective effects of trifluoperazine that may pave the way for repurposing this calmodulin antagonist in ameliorating organ toxicity by doxorubicin.
Collapse
Affiliation(s)
- Ahmed E Goda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Egypt.
| | - Amr M Elenany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Egypt
| | - Alaa E Elsisi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Egypt
| |
Collapse
|
132
|
Alsherbiny MA, Bhuyan DJ, Radwan I, Chang D, Li CG. Metabolomic Identification of Anticancer Metabolites of Australian Propolis and Proteomic Elucidation of Its Synergistic Mechanisms with Doxorubicin in the MCF7 Cells. Int J Mol Sci 2021; 22:ijms22157840. [PMID: 34360606 PMCID: PMC8346082 DOI: 10.3390/ijms22157840] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 12/11/2022] Open
Abstract
The combination of natural products with standard chemotherapeutic agents offers a promising strategy to enhance the efficacy or reduce the side effects of standard chemotherapy. Doxorubicin (DOX), a standard drug for breast cancer, has several disadvantages, including severe side effects and the development of drug resistance. Recently, we reported the potential bioactive markers of Australian propolis extract (AP-1) and their broad spectrum of pharmacological activities. In the present study, we explored the synergistic interactions between AP-1 and DOX in the MCF7 breast adenocarcinoma cells using different synergy quantitation models. Biochemometric and metabolomics-driven analysis was performed to identify the potential anticancer metabolites in AP-1. The molecular mechanisms of synergy were studied by analysing the apoptotic profile via flow cytometry, apoptotic proteome array and measuring the oxidative status of the MCF7 cells treated with the most synergistic combination. Furthermore, label-free quantification proteomics analysis was performed to decipher the underlying synergistic mechanisms. Five prenylated stilbenes were identified as the key metabolites in the most active AP-1 fraction. Strong synergy was observed when AP-1 was combined with DOX in the ratio of 100:0.29 (w/w) as validated by different synergy quantitation models implemented. AP-1 significantly enhanced the inhibitory effect of DOX against MCF7 cell proliferation in a dose-dependent manner with significant inhibition of the reactive oxygen species (p < 0.0001) compared to DOX alone. AP-1 enabled the reversal of DOX-mediated necrosis to programmed cell death, which may be advantageous to decline DOX-related side effects. AP-1 also significantly enhanced the apoptotic effect of DOX after 24 h of treatment with significant upregulation of catalase, HTRA2/Omi, FADD together with DR5 and DR4 TRAIL-mediated apoptosis (p < 0.05), contributing to the antiproliferative activity of AP-1. Significant upregulation of pro-apoptotic p27, PON2 and catalase with downregulated anti-apoptotic XIAP, HSP60 and HIF-1α, and increased antioxidant proteins (catalase and PON2) may be associated with the improved apoptosis and oxidative status of the synergistic combination-treated MCF7 cells compared to the mono treatments. Shotgun proteomics identified 21 significantly dysregulated proteins in the synergistic combination-treated cells versus the mono treatments. These proteins were involved in the TP53/ATM-regulated non-homologous end-joining pathway and double-strand breaks repairs, recruiting the overexpressed BRCA1 and suppressed RIF1 encoded proteins. The overexpression of UPF2 was noticed in the synergistic combination treatment, which could assist in overcoming doxorubicin resistance-associated long non-coding RNA and metastasis of the MCF7 cells. In conclusion, we identified the significant synergy and highlighted the key molecular pathways in the interaction between AP-1 and DOX in the MCF7 cells together with the AP-1 anticancer metabolites. Further in vivo and clinical studies are warranted on this synergistic combination.
Collapse
Affiliation(s)
- Muhammad A. Alsherbiny
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia;
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
- Correspondence: (M.A.A.); (D.J.B.); (C.-G.L.)
| | - Deep J. Bhuyan
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia;
- Correspondence: (M.A.A.); (D.J.B.); (C.-G.L.)
| | - Ibrahim Radwan
- Faculty of Science and Technology, University of Canberra, Canberra, ACT 2617, Australia;
| | - Dennis Chang
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia;
| | - Chun-Guang Li
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia;
- Correspondence: (M.A.A.); (D.J.B.); (C.-G.L.)
| |
Collapse
|
133
|
Doxorubicin and doxorubicin-loaded nanoliposome induce senescence by enhancing oxidative stress, hepatotoxicity, and in vivo genotoxicity in male Wistar rats. Naunyn Schmiedebergs Arch Pharmacol 2021; 394:1803-1813. [PMID: 34219194 DOI: 10.1007/s00210-021-02119-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 06/24/2021] [Indexed: 10/20/2022]
Abstract
The senescence phenomenon is historically considered as a tumor-suppressing mechanism that can permanently arrest the proliferation of damaged cells, and prevent tumor eradication by activating cell cycle regulatory pathways. Doxorubicin (DX) as an antineoplastic agent has been used for the treatment of solid and hematological malignancies for a long time, but its clinical use is limited due to irreversible toxicity on off-target tissues. Thereby, the encapsulation of plain drugs in a vehicle may decrease the side effects while increasing their permeability and availability in target cells. Here, we aimed to investigate and compare the effects of DX and DX-loaded nanoliposome (NLDX) on the induction of senescence via assessment of the occurrence of apoptosis/necrosis, genomic damage, oxidative stress, and liver pathologies. The study groups included DX (0.75, 0.5, 0.1 mg/kg/BW), NLDX groups (0.1, 0.05, 0.025 mg/kg/BW), and an untreated control group. The liver tissues were used to investigate the oxidative stress parameters and probable biochemical and histopathological alterations. Annexin V/PI staining was carried out to find the type of cellular death in the liver tissue of healthy rats exposed to different concentrations of DOX and LDOX. Data revealed that the highest dose of NLDX (0.1 mg/kg/BW) could significantly induce cellular senescence throughout significant increasing the level of genotoxic damage (p < 0.0001) and the oxidative stress (p < 0.001) compared with a similar dose of DX, in which the obtained results were further confirmed by flow cytometry and histopathological assessments of the liver tissue. This investigation provides sufficient evidence of improved therapeutic efficacy of NLDX compared with plain DX in male Wistar rats.
Collapse
|
134
|
Chen J, Qian C, Ren P, Yu H, Kong X, Huang C, Luo H, Chen G. Light-Responsive Micelles Loaded With Doxorubicin for Osteosarcoma Suppression. Front Pharmacol 2021; 12:679610. [PMID: 34220512 PMCID: PMC8249570 DOI: 10.3389/fphar.2021.679610] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/19/2021] [Indexed: 01/14/2023] Open
Abstract
The enhancement of tumor targeting and cellular uptake of drugs are significant factors in maximizing anticancer therapy and minimizing the side effects of chemotherapeutic drugs. A key challenge remains to explore stimulus-responsive polymeric nanoparticles to achieve efficient drug delivery. In this study, doxorubicin conjugated polymer (Poly-Dox) with light-responsiveness was synthesized, which can self-assemble to form polymeric micelles (Poly-Dox-M) in water. As an inert structure, the polyethylene glycol (PEG) can shield the adsorption of protein and avoid becoming a protein crown in the blood circulation, improving the tumor targeting of drugs and reducing the cardiotoxicity of doxorubicin (Dox). Besides, after ultraviolet irradiation, the amide bond connecting Dox with PEG can be broken, which induced the responsive detachment of PEG and enhanced cellular uptake of Dox. Notably, the results of immunohistochemistry in vivo showed that Poly-Dox-M had no significant damage to normal organs. Meanwhile, they showed efficient tumor-suppressive effects. This nano-delivery system with the light-responsive feature might hold great promises for the targeted therapy for osteosarcoma.
Collapse
Affiliation(s)
- Jiayi Chen
- Bengbu Medical College, Bengbu, China.,Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | | | - Peng Ren
- Bengbu Medical College, Bengbu, China.,Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Han Yu
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Xiangjia Kong
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Chenglong Huang
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Huanhuan Luo
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Gang Chen
- Bengbu Medical College, Bengbu, China.,Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
135
|
Renu K, Pureti LP, Vellingiri B, Valsala Gopalakrishnan A. Toxic effects and molecular mechanism of doxorubicin on different organs – an update. TOXIN REV 2021. [DOI: 10.1080/15569543.2021.1912099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Kaviyarasi Renu
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, India
| | - Lakshmi Prasanna Pureti
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, India
| | - Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, India
| |
Collapse
|
136
|
Moezian GSA, Javadinia SA, Sales SS, Fanipakdel A, Elyasi S, Karimi G. Oral silymarin formulation efficacy in management of AC-T protocol induced hepatotoxicity in breast cancer patients: A randomized, triple blind, placebo-controlled clinical trial. J Oncol Pharm Pract 2021; 28:827-835. [PMID: 33861657 DOI: 10.1177/10781552211006182] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Chemotherapeutic agents, with or without other drugs and radiation, may cause indirect or direct hepatotoxicity. Doxorubicin-induced hepatotoxicity (DIH) is a major health concern in cancer patients receiving this cytotoxic drug that is mostly resulted from the production of reactive oxygen species leading to transient or permanent liver damages. Silymarin, a flavonoid extracted from the Silybum marianum, exhibits antioxidant and anti-inflammatory activities. PURPOSE This study aimed to investigate the clinical efficacy of systemic administration of silymarin in management of chemotherapy induced hepatotoxicity in patients with non-metastatic breast cancer who received doxorubicin/cyclophosphamide-paclitaxel (AC-T) regimen.Material: In this randomized, triple blind, placebo-controlled clinical trial, 30 patients who received AC-T who fulfilled the inclusion criteria were randomly allocated to silymarin (n = 15) or placebo (n = 15) groups to receive oral silymarin 140 mg three times a day or placebo tablets, respectively. Fatty liver severity was assessed by liver ultrasound imaging and FibroScan® and also measurement of liver function tests before and after the intervention. RESULTS There was a non-significant trend toward more severe liver involvement in placebo group comparing to the silymarin group after intervention based on ultrasonography (p = 0.083). Besides, in silymarin group, hepatic involvement grade based on ultrasonography considerably reduced after intervention (p = 0.012). However, no difference was found between two groups based on FibroScan and liver function tests. CONCLUSION Oral administration of silymarin could significantly reduce hepatotoxicity severity after 1 month of treatment in non-metastatic breast cancer patients treated with AC-T regimen.
Collapse
Affiliation(s)
| | - Seyed Alireza Javadinia
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | | | - Azar Fanipakdel
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sepideh Elyasi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmaceutical Research Center, Faculty of Pharmacy, Mashhad University of Medical Sciences
| |
Collapse
|
137
|
Sonowal H, Saxena A, Qiu S, Srivastava S, Ramana KV. Aldose reductase regulates doxorubicin-induced immune and inflammatory responses by activating mitochondrial biogenesis. Eur J Pharmacol 2021; 895:173884. [PMID: 33482179 DOI: 10.1016/j.ejphar.2021.173884] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 01/19/2023]
Abstract
We have recently demonstrated that aldose reductase (AR) inhibitor; fidarestat prevents doxorubicin (Dox)-induced cardiotoxic side effects and inflammation in vitro and in vivo. However, the effect of fidarestat and its combination with Dox on immune cell activation and the immunomodulatory effects are not known. In this study, we examined the immunomodulatory effects of fidarestat in combination with Dox in vivo and in vitro. We observed that fidarestat decreased Dox-induced upregulation of CD11b in THP-1 monocytes. Fidarestat further attenuated Dox-induced upregulation of IL-6, IL-1β, and Nos2 in murine BMDM. Fidarestat also attenuated Dox-induced activation and infiltration of multiple subsets of inflammatory immune cells identified by expression of markers CD11b+, CD11b+F4/80+, Ly6C+CCR2high, and Ly6C+CD11b+ in the mouse spleen and liver. Furthermore, significant upregulation of markers of mitochondrial biogenesis PGC-1α, COX IV, TFAM, and phosphorylation of AMPKα1 (Ser485) was observed in THP-1 cells and livers of mice treated with Dox in combination with fidarestat. Our results suggest that fidarestat by up-regulating mitochondrial biogenesis exerts protection against Dox-induced immune and inflammatory responses in vitro and in vivo, providing further evidence for developing fidarestat as a combination agent with anthracycline drugs to prevent chemotherapy-induced inflammation and toxicity.
Collapse
Affiliation(s)
- Himangshu Sonowal
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| | - Ashish Saxena
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Sumin Qiu
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Sanjay Srivastava
- Department of Environmental Cardiology, University of Louisville, KY, USA
| | - Kota V Ramana
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
138
|
Shati AA, El-Kott AF. Acylated ghrelin protects against doxorubicin-induced nephropathy by activating silent information regulator 1. Basic Clin Pharmacol Toxicol 2021; 128:805-821. [PMID: 33547742 DOI: 10.1111/bcpt.13569] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 01/01/2021] [Accepted: 02/03/2021] [Indexed: 02/06/2023]
Abstract
This study investigated the nephroprotective role of acylated ghrelin (AG) against DOX-induced nephropathy and examined whether the protection involves silent information regulator 1 (SIRT1). Rats were divided into control, control + AG, DOX, DOX + AG, DOX + AG + [D-Lys3]-GHRP-6 (a ghrelin receptor antagonist), and DOX + AG + EX-527 (a sirt1 inhibitor). DOX was given over the first 2 weeks. AG (10 ng/kg) and both inhibitors were given as 3 doses/wk for 5 weeks. AG improved the structure and the function of the kidneys; down-regulated the renal expression of TGF-β1, collagen 1A1 and α-SMA; and inhibited the renal collagen deposition in the kidneys of DOX-treated rats. Concomitantly, it reduced the renal levels of ROS, MDA, TNF-α, and IL-6 and protein levels of cytochrome-c, TGF-β1, Smad3 and α-SMA in these rats. In both the control and DOX-treated rats, AG significantly increased the renal levels of SOD and GSH, decreased the expression of cleaved caspase-3 and Bax, increased the total levels and the nuclear activity of SIRT1 and reduced the deacetylation of p53, NF-κB and FOXO-31. All the effects were abolished by the concurrent administration of EX-527 and [D-Lys3]-GHRP-6. In conclusion, AG prevents DOX-induced nephropathy in SIRT1 and GSHRa1-dependent mechanism.
Collapse
Affiliation(s)
- Ali A Shati
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Attalla F El-Kott
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia.,Department of Zoology, Faculty of Science, Damanhour University, Damanhour, Egypt
| |
Collapse
|
139
|
Nicolella HD, Fernandes G, Ozelin SD, Rinaldi-Neto F, Ribeiro AB, Furtado RA, Senedese JM, Esperandim TR, Veneziani RCS, Tavares DC. Manool, a diterpene from Salvia officinalis, exerts preventive effects on chromosomal damage and preneoplastic lesions. Mutagenesis 2021; 36:177-185. [PMID: 33512444 DOI: 10.1093/mutage/geab001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 01/12/2021] [Indexed: 11/14/2022] Open
Abstract
The present study aimed to evaluate the effect of the manool diterpene on genomic integrity. For this purpose, we evaluated the influence of manool on genotoxicity induced by mutagens with different mechanisms of action, as well as on colon carcinogenesis. The results showed that manool (0.5 and 1.0 µg/ml) significantly reduced the frequency of micronuclei induced by doxorubicin (DXR) and hydrogen peroxide in V79 cells but did not influence genotoxicity induced by etoposide. Mice receiving manool (1.25 mg/kg) exhibited a significant reduction (79.5%) in DXR-induced chromosomal damage. The higher doses of manool (5.0 and 20 mg/kg) did not influence the genotoxicity induced by DXR. The anticarcinogenic effect of manool (0.3125, 1.25 and 5.0 mg/kg) was also observed against preneoplastic lesions chemically induced in rat colon. A gradual increase in manool doses did not cause a proportional reduction of preneoplastic lesions, thus demonstrating the absence of a dose-response relationship. The analysis of serum biochemical indicators revealed the absence of hepatotoxicity and nephrotoxicity of treatments. To explore the chemopreventive mechanisms of manool via anti-inflammatory pathways, we evaluated its effect on nitric oxide (NO) production and on the expression of the NF-kB gene. At the highest concentration tested (4 μg/ml), manool significantly increased NO production when compared to the negative control. On the other hand, in the prophylactic treatment model, manool (0.5 and 1.0 μg/ml) was able to significantly reduce NO levels produced by macrophages stimulated with lipopolysaccharide. Analysis of NF-kB in hepatic and renal tissues of mice treated with manool and DXR revealed that the mutagen was unable to stimulate expression of the gene. In conclusion, manool possesses antigenotoxic and anticarcinogenic effects and its anti-inflammatory potential might be related, at least in part, to its chemopreventive activity.
Collapse
Affiliation(s)
- Heloiza Diniz Nicolella
- Mutagenesis Laboratory, Universidade de Franca, Avenida Dr. Armando Salles de Oliveira, 201 - Parque Universitário, 14404-600 Franca, São Paulo, Brazil
| | - Gabriela Fernandes
- Mutagenesis Laboratory, Universidade de Franca, Avenida Dr. Armando Salles de Oliveira, 201 - Parque Universitário, 14404-600 Franca, São Paulo, Brazil
| | - Saulo Duarte Ozelin
- Mutagenesis Laboratory, Universidade de Franca, Avenida Dr. Armando Salles de Oliveira, 201 - Parque Universitário, 14404-600 Franca, São Paulo, Brazil
| | - Francisco Rinaldi-Neto
- Mutagenesis Laboratory, Universidade de Franca, Avenida Dr. Armando Salles de Oliveira, 201 - Parque Universitário, 14404-600 Franca, São Paulo, Brazil
| | - Arthur Barcelos Ribeiro
- Mutagenesis Laboratory, Universidade de Franca, Avenida Dr. Armando Salles de Oliveira, 201 - Parque Universitário, 14404-600 Franca, São Paulo, Brazil
| | - Ricardo Andrade Furtado
- Mutagenesis Laboratory, Universidade de Franca, Avenida Dr. Armando Salles de Oliveira, 201 - Parque Universitário, 14404-600 Franca, São Paulo, Brazil
| | - Juliana Marques Senedese
- Mutagenesis Laboratory, Universidade de Franca, Avenida Dr. Armando Salles de Oliveira, 201 - Parque Universitário, 14404-600 Franca, São Paulo, Brazil
| | - Tábata Rodrigues Esperandim
- Mutagenesis Laboratory, Universidade de Franca, Avenida Dr. Armando Salles de Oliveira, 201 - Parque Universitário, 14404-600 Franca, São Paulo, Brazil
| | - Rodrigo Cassio Sola Veneziani
- Mutagenesis Laboratory, Universidade de Franca, Avenida Dr. Armando Salles de Oliveira, 201 - Parque Universitário, 14404-600 Franca, São Paulo, Brazil
| | - Denise Crispim Tavares
- Mutagenesis Laboratory, Universidade de Franca, Avenida Dr. Armando Salles de Oliveira, 201 - Parque Universitário, 14404-600 Franca, São Paulo, Brazil
| |
Collapse
|
140
|
Timm KN, Ball V, Miller JJ, Savic D, West JA, Griffin JL, Tyler DJ. Metabolic Effects of Doxorubicin on the Rat Liver Assessed With Hyperpolarized MRI and Metabolomics. Front Physiol 2021; 12:782745. [PMID: 35069242 PMCID: PMC8766499 DOI: 10.3389/fphys.2021.782745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/13/2021] [Indexed: 11/30/2022] Open
Abstract
Doxorubicin (DOX) is a successful chemotherapeutic widely used for the treatment of a range of cancers. However, DOX can have serious side-effects, with cardiotoxicity and hepatotoxicity being the most common events. Oxidative stress and changes in metabolism and bioenergetics are thought to be at the core of these toxicities. We have previously shown in a clinically-relevant rat model that a low DOX dose of 2 mg kg-1 week-1 for 6 weeks does not lead to cardiac functional decline or changes in cardiac carbohydrate metabolism, assessed with hyperpolarized [1-13C]pyruvate magnetic resonance spectroscopy (MRS). We now set out to assess whether there are any signs of liver damage or altered liver metabolism using this subclinical model. We found no increase in plasma alanine aminotransferase (ALT) activity, a measure of liver damage, following DOX treatment in rats at any time point. We also saw no changes in liver carbohydrate metabolism, using hyperpolarized [1-13C]pyruvate MRS. However, using metabolomic analysis of liver metabolite extracts at the final time point, we found an increase in most acyl-carnitine species as well as increases in high energy phosphates, citrate and markers of oxidative stress. This may indicate early signs of steatohepatitis, with increased and decompensated fatty acid uptake and oxidation, leading to oxidative stress.
Collapse
Affiliation(s)
- Kerstin N. Timm
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
- *Correspondence: Kerstin N. Timm,
| | - Vicky Ball
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Jack J. Miller
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
- Department of Physics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Clinical Magnetic Resonance Research, John Radcliffe Hospital, Oxford, United Kingdom
- The MR Research Center, The PET Centre, Aarhus University Hospital, Aarhus University, Aarhus, Denmark
| | - Dragana Savic
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Clinical Magnetic Resonance Research, John Radcliffe Hospital, Oxford, United Kingdom
| | - James A. West
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Julian L. Griffin
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Damian J. Tyler
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Clinical Magnetic Resonance Research, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
141
|
Ashrafizadeh M, Zarrabi A, Hushmandi K, Hashemi F, Rahmani Moghadam E, Raei M, Kalantari M, Tavakol S, Mohammadinejad R, Najafi M, Tay FR, Makvandi P. Progress in Natural Compounds/siRNA Co-delivery Employing Nanovehicles for Cancer Therapy. ACS COMBINATORIAL SCIENCE 2020; 22:669-700. [PMID: 33095554 PMCID: PMC8015217 DOI: 10.1021/acscombsci.0c00099] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 10/05/2020] [Indexed: 02/06/2023]
Abstract
Chemotherapy using natural compounds, such as resveratrol, curcumin, paclitaxel, docetaxel, etoposide, doxorubicin, and camptothecin, is of importance in cancer therapy because of the outstanding therapeutic activity and multitargeting capability of these compounds. However, poor solubility and bioavailability of natural compounds have limited their efficacy in cancer therapy. To circumvent this hurdle, nanocarriers have been designed to improve the antitumor activity of the aforementioned compounds. Nevertheless, cancer treatment is still a challenge, demanding novel strategies. It is well-known that a combination of natural products and gene therapy is advantageous over monotherapy. Delivery of multiple therapeutic agents/small interfering RNA (siRNA) as a potent gene-editing tool in cancer therapy can maximize the synergistic effects against tumor cells. In the present review, co-delivery of natural compounds/siRNA using nanovehicles are highlighted to provide a backdrop for future research.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty
of Engineering and Natural Sciences, Sabanci
University, Orta Mahalle,
Üniversite Caddesi No. 27, Orhanlı,
Tuzla, 34956 Istanbul, Turkey
- Sabanci
University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul Turkey
| | - Ali Zarrabi
- Sabanci
University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul Turkey
| | - Kiavash Hushmandi
- Department
of Food Hygiene and Quality Control, Division of Epidemiology &
Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran 1419963114, Iran
| | - Farid Hashemi
- Department
of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ebrahim Rahmani Moghadam
- Department
of Anatomical Sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran
| | - Mehdi Raei
- Health Research
Center, Life Style Institute, Baqiyatallah
University of Medical Sciences, Tehran 1435916471, Iran
| | - Mahshad Kalantari
- Department
of Genetics, Tehran Medical Sciences Branch, Azad University, Tehran 19168931813, Iran
| | - Shima Tavakol
- Cellular
and Molecular Research Center, Iran University
of Medical Sciences, Tehran 1449614525, Iran
| | - Reza Mohammadinejad
- Pharmaceutics
Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7616911319, Iran
| | - Masoud Najafi
- Medical
Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
- Radiology
and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
| | - Franklin R. Tay
- College
of Graduate Studies, Augusta University, Augusta, Georgia 30912, United States
| | - Pooyan Makvandi
- Istituto
Italiano di Tecnologia, Centre for Micro-BioRobotics, viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa Italy
- Department
of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, 14496-14535 Tehran, Iran
| |
Collapse
|
142
|
Abbas NAT, Awad MM, Nafea OE. Silymarin in combination with chlorogenic acid protects against hepatotoxicity induced by doxorubicin in rats: possible role of adenosine monophosphate-activated protein kinase pathway. Toxicol Res (Camb) 2020; 9:771-777. [PMID: 33447361 DOI: 10.1093/toxres/tfaa080] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/25/2020] [Accepted: 10/05/2020] [Indexed: 12/29/2022] Open
Abstract
Many xenobiotics are known to cause hepatic damage with subsequent significant morbidity and mortality. Doxorubicin (DOX) is a broad-spectrum antineoplastic agent. DOX is reported to cause hepatocellular damage. Previous studies verified the promising role of many natural antioxidant products against various models of hepatic dysfunction. We conducted this study to evaluate the possible hepatoprotective effect of silymarin (SILY) and/or chlorogenic acid (CGA) in a rat model of DOX-induced hepatotoxicity. For this purpose, we randomly divided 30 adult male rats into five equal groups as control, DOX, co-treated DOX with SILY, co-treated DOX with GCA and co-treated DOX with SILY and CGA groups. All treatments were administered every second day for 4 weeks. Our results showed that simultaneous SILY and CGA administration caused a significant decrease in hepatic apoptosis biomarkers (hepatic caspase-3 and nuclear factor-κB levels), a significant improvement in hepatic oxidant/antioxidant status (malondialdehyde and superoxide dismutase) and significant decrease in hepatic pro-inflammatory biomarkers (tumor necrosis factor-alpha and interlukin-1β) compared with DOX treatment. We concluded that adding CGA to SILY acts as a hepatoprotective agent against DOX-induced liver injury through inhibiting apoptosis biomarkers, maintaining antioxidant enzyme levels, decreasing pro-inflammatory cytokines as well as regulating liver adenosine monophosphate-activated protein kinase signaling.
Collapse
Affiliation(s)
- Noha A T Abbas
- Faculty of Medicine, Department of Clinical Pharmacology, Zagazig University, Zagazig 44519, Egypt
| | - Mohammed M Awad
- Endocrinology Division, Faculty of Medicine, Department of Internal Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Ola E Nafea
- Faculty of Medicine, Department of Forensic Medicine and Clinical Toxicology, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
143
|
Shen W, Zheng J, Zhou Z, Zhang D. Approaches for the synthesis of o-nitrobenzyl and coumarin linkers for use in photocleavable biomaterials and bioconjugates and their biomedical applications. Acta Biomater 2020; 115:75-91. [PMID: 32853806 DOI: 10.1016/j.actbio.2020.08.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/18/2020] [Accepted: 08/18/2020] [Indexed: 12/20/2022]
Abstract
Photocleavable biomaterials and bioconjugates are particularly interesting because light sources are easy to obtain and the responsiveness of materials is convenient to control. In recent years, various photocleavable biomaterials and bioconjugates have been synthesized for the control of payload release, regulation of biomolecule activity, 3D cell culture, and investigation of molecular mechanisms. Photocleavable linkers are crucial components of photocleavable biomaterials, which significantly influence the photoresponsive capabilities of materials. Photosensitive molecules, such as o-nitrobenzyls and coumarins, have been extensively developed as photocleavable linkers. In the present review, we provide comprehensive knowledge regarding the synthetic strategies of o-nitrobenzyl and coumarin derived linkers with various functional groups and their applications for the construction of photocleavable biomaterials and bioconjugates. Finally, the biomedical applications of o-nitrobenzyl and coumarin-based photocleavable biomaterials and bioconjugates will be summarized and discussed.
Collapse
|
144
|
Hepatotoxicity of nutmeg: A pilot study based on metabolomics. Biomed Pharmacother 2020; 131:110780. [PMID: 33152938 DOI: 10.1016/j.biopha.2020.110780] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/08/2020] [Accepted: 09/17/2020] [Indexed: 12/22/2022] Open
Abstract
Incidences of abuse and poisoning have been reported for nutmeg, a household spice made from grinding the seed of Myristica fragrans, owing to its hallucinogenic properties. However, there have been no reports on nutmeg hepatotoxicity in relation to dose and duration of exposure. To investigate the hepatotoxicity of different nutmeg exposure durations and doses, male mice were administered daily with normal saline, 1.0 g/kg nutmeg, or 4.0 g/kg nutmeg by intragastrical gavage for either 7 or 14 days (for a total of six treatment groups, n = 6). Body weight of each mouse was monitored daily. Histological analysis of liver tissues was performed using hematoxylin and eosin (H&E) staining to investigate the morphological changes in hepatocytes. Serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels were determined using enzyme-linked immunosorbent assay (ELISA) to investigate liver function. Metabolomics and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed between treatment groups for identifying differential metabolites. Mice in the nutmeg exposure groups exhibited slow growth trends, hepatocyte damage, and significantly elevated serum AST and ALT levels associated with nutmeg dose and exposure duration. Metabolomics and KEGG enrichment pathway analyses also revealed differential levels of some metabolites related to liver function upon nutmeg exposure. Therefore, the present study reasonably speculates that nutmeg exposure may cause liver damage and affect liver function depending on the dose and duration.
Collapse
|
145
|
MORPHOLOGICAL FEATURES OF DOXORUBICIN-INDUCED LIVER DAMAGE ASSOCIATED WITH NONALCOHOLIC STEATOHEPATITIS. WORLD OF MEDICINE AND BIOLOGY 2020. [DOI: 10.26724/2079-8334-2020-3-73-189-194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
146
|
THE ROLE OF ARGININE/CITRULLINE CYCLE DISORDERS IN THE PATHOGENESIS OF DOXORUBICIN-INDUCED LIVER INJURY ASSOCIATED WITH NONALCOHOLIC STEATOHEPATITIS IN RATS. WORLD OF MEDICINE AND BIOLOGY 2020. [DOI: 10.26724/2079-8334-2020-2-72-188-192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|