1
|
Albaqami A, Alosaimi ME, Jafri I, Mohamed AAR, Abd El-Hakim YM, Khamis T, Elazab ST, Noreldin AE, Elhamouly M, El-Far AH, Eskandrani AA, Alotaibi BS, M Abdelnour H, Saleh AA. Pulmonary damage induction upon Acrylic amide exposure via activating miRNA-223-3p and miRNA-325-3p inflammasome/pyroptosis and fibrosis signaling pathway: New mechanistic approaches of A green-synthesized extract. Toxicology 2024; 506:153869. [PMID: 38909937 DOI: 10.1016/j.tox.2024.153869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/01/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024]
Abstract
Exposure to acrylic amide (AD) has garnered worldwide attention due to its potential adverse health effects, prompting calls from the World Health Organization for intensified research into associated risks. Despite this, the relationship between oral acrylic amide (acrylamide) (AD) exposure and pulmonary dysfunction remains poorly understood. Our study aimed to investigate the correlation between internal oral exposure to AD and the decline in lung function, while exploring potential mediating factors such as tissue inflammation, oxidative stress, pyroptosis, and apoptosis. Additionally, we aimed to evaluate the potential protective effect of zinc oxide nanoparticles green-synthesized moringa extract (ZNO-MONPs) (10 mg/kg b.wt) against ACR toxicity and conducted comprehensive miRNA expression profiling to uncover novel targets and mechanisms of AD toxicity (miRNA 223-3 P and miRNA 325-3 P). Furthermore, we employed computational techniques to predict the interactions between acrylic amide and/or MO-extract components and tissue proteins. Using a rat model, we exposed animals to oral acrylamide (20 mg/kg b.wt for 2 months). Our findings revealed that AD significantly downregulated the expression of miRNA 223-3 P and miRNA 325-3 P, targeting NLRP-3 & GSDMD, respectively, indicating the induction of pyroptosis in pulmonary tissue via an inflammasome activating pathway. Moreover, AD exposure resulted in lipid peroxidative damage and reduced levels of GPX, CAT, GSH, and GSSG. Notably, AD exposure upregulated apoptotic, pyroptotic, and inflammatory genes, accompanied by histopathological damage in lung tissue. Immunohistochemical and immunofluorescence techniques detected elevated levels of indicative harmful proteins including vimentin and 4HNE. Conversely, concurrent administration of ZNO-MONPs with AD significantly elevated the expression of miRNA 223-3 P and miRNA 325-3 P, protecting against oxidative stress, apoptosis, pyroptosis, inflammation, and fibrosis in rat lungs. In conclusion, our study highlights the efficacy of ZNO-MONPs NPs in protecting pulmonary tissue against the detrimental impacts of foodborne toxin AD.
Collapse
Affiliation(s)
- Amirah Albaqami
- Department of Clinical Laboratory Sciences, Turabah University College, Taif University, Taif 21944, Saudi Arabia
| | - Manal E Alosaimi
- Department of Basic Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, P.O Box 84428, Riyadh 11671, Saudi Arabia.
| | - Ibrahim Jafri
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Amany Abdel-Rahman Mohamed
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt.
| | - Yasmina M Abd El-Hakim
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Tarek Khamis
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, 44511 Zagazig, Egypt; Laboratory of Biotechnology, Faculty of Veterinary Medicine, Zagazig University, 44519 Zagazig, Egypt
| | - Sara T Elazab
- Department of Pharmacology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed E Noreldin
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt
| | - Moustafa Elhamouly
- Department of Histology and Cytology Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Ali H El-Far
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, China; Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt
| | - Areej A Eskandrani
- Chemistry Department, College of Science, Taibah University, P.O. Box 344, Medina 30002, Saudi Arabia
| | - Badriyah S Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Hanim M Abdelnour
- Department of Biochemistry, Faculty of Medicine, Zagazig University, Egypt
| | - Ayman A Saleh
- Department of Pathology, College of Medicine, University of Hail, Hail, Kingdom of Saudi Arabia
| |
Collapse
|
2
|
Alqahtani LS, Alosaimi ME, Abdel-Rahman Mohamed A, Abd-Elhakim YM, Khamis T, Noreldin AE, El-Far AH, Alotaibi BS, Hakami MA, Dahran N, Babteen NA. Acrylamide-targeting renal miR-21a-5p/Fibrotic and miR122-5p/ inflammatory signaling pathways and the role of a green approach for nano-zinc detected via in silico and in vivo approaches. Front Pharmacol 2024; 15:1413844. [PMID: 39086388 PMCID: PMC11289894 DOI: 10.3389/fphar.2024.1413844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/26/2024] [Indexed: 08/02/2024] Open
Abstract
Introduction: Any disruption in renal function can have cascading effects on overall health. Understanding how a heat-born toxicant like acrylamide (ACR) affects kidney tissue is vital for realizing its broader implications for systemic health. Methods: This study investigated the ACR-induced renal damage mechanisms, particularly focusing on the regulating role of miR-21a-5p/fibrotic and miR-122-5p/inflammatory signaling pathways via targeting Timp-3 and TP53 proteins in an In silico preliminary study. Besides, renal function assessment, oxidative status, protein profile, and the expression of renal biomarkers (Timp-1, Keap-1, Kim-1, P53, TNF-α, Bax, and Caspase3) were assessed in a 60-day experiment. The examination was additionally extended to explore the potential protective effects of green-synthesized zinc oxide nanoparticles (ZNO-MONPs). A four-group experiment including control, ZNO-MONPs (10 mg/kg b.wt.), ACR (20 mg/kg b.wt.), and ZNO-MONPs + ACR was established encompassing biochemical, histological, and molecular levels. The study further investigated the protein-binding ability of ZNO and MONPs to inactivate caspase-3, Keap-1, Kim-1, and TNFRS-1A. Results: ZNO-MONPs significantly reduced ACR-induced renal tissue damage as evidenced by increased serum creatinine, uric acid, albumin, and oxidative stress markers. ACR-induced oxidative stress, apoptosis, and inflammationare revealed by biochemical tests, gene expression, and the presence of apoptotic nuclei microscopically. Also, molecular docking revealed binding affinity between ACR-BCL-2 and glutathione-synthetase, elucidating the potential mechanisms through which ACR induces renal damage. Notably, ZNO-MONPs revealed a protective potential against ACR-induced damage. Zn levels in the renal tissues of ACR-exposed rats were significantly restored in those treated with ACR + ZNO-MONPs. In conclusion, this study establishes the efficacy of ZNO-MONPs in mitigating ACR-induced disturbances in renal tissue functions, oxidative stress, inflammation, and apoptosis. The findings shed light on the potential renoprotective activity of green-synthesized nanomaterials, offering insights into novel therapeutic approaches for countering ACR-induced renal damage.
Collapse
Affiliation(s)
- Leena S. Alqahtani
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Manal E. Alosaimi
- Department of Basic Sciences, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Amany Abdel-Rahman Mohamed
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Yasmina M. Abd-Elhakim
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Tarek Khamis
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
- Laboratory of Biotechnology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Ahmed E. Noreldin
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Ali H. El-Far
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Badriyah S. Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mohammed Ageeli Hakami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Naief Dahran
- Department of Anatomy, Faculty of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Nouf A. Babteen
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| |
Collapse
|
3
|
Qu J, Pei H, Li XZ, Li Y, Chen JM, Zhang M, Lu ZQ. Erythrocyte membrane biomimetic EGCG nanoparticles attenuate renal injury induced by diquat through the NF-κB/NLRP3 inflammasome pathway. Front Pharmacol 2024; 15:1414918. [PMID: 39045044 PMCID: PMC11263105 DOI: 10.3389/fphar.2024.1414918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/14/2024] [Indexed: 07/25/2024] Open
Abstract
Diquat (DQ) poisoning can cause multiple organ damage, and the kidney is considered to be the main target organ. Increasing evidence shows that alleviating oxidative stress and inflammatory response has promising application prospects. Epigallocatechin gallate (EGCG) has potent antioxidant and anti-inflammatory effects. In this study, red blood cell membrane (RBCm)-camouflaged polylactic-co-glycolic acid (PLGA) nanoparticles (NPs) were synthesized to deliver EGCG (EGCG-RBCm/NPs) for renal injury induced by DQ. Human renal tubular epithelial cells (HK-2 cells) were stimulated with 600 μM DQ for 12 h and mice were intraperitoneally injected with 50 mg/kg b.w. DQ, followed by 20 mg/kg b.w./day EGCG or EGCG-RBCM/NPs for 3 days. The assessment of cellular vitality was carried out using the CCK-8 assay, while the quantification of reactive oxygen species (ROS) was performed through ROS specific probes. Apoptosis analysis was conducted by both flow cytometry and TUNEL staining methods. Pathological changes in renal tissue were observed. The expressions of NLRP3, IL-1β, IL-18, NFκB and Caspase1 were detected by quantitative reverse transcription polymerase chain reaction (qRT-PCR), immunohistochemistry, immunofluorescence, and Western blot. The results showed that the DQ group had increased ROS expression, increased the level of oxidative stress, and increased apoptosis rate compared with the control group. Histopathological analysis of mice in the DQ group showed renal tubular injury and elevated levels of blood urea nitrogen (BUN), serum creatinine (SCr), kidney injury molecule-1 (KIM-1), and cystatin C (Cys C). Furthermore, the DQ group exhibited heightened expression of NLRP3, p-NFκB p65, Caspase1 p20, IL-1β, and IL-18. However, EGCG-RBCm/NPs treatment mitigated DQ-induced increases in ROS, apoptosis, and oxidative stress, as well as renal toxicity and decreases in renal biomarker levels. Meanwhile, the expression of the above proteins were significantly decreased, and the survival rate of mice was ultimately improved, with an effect better than that of the EGCG treatment group. In conclusion, EGCG-RBCm/NPs can improve oxidative stress, inflammation, and apoptosis induced by DQ. This effect is related to the NF-κB/NLRP3 inflammasome pathway. Overall, this study provides a new approach for treating renal injury induced by DQ.
Collapse
Affiliation(s)
- Jie Qu
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Hui Pei
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Xin-Ze Li
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Yan Li
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Jian-Ming Chen
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Min Zhang
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Zhong-Qiu Lu
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| |
Collapse
|
4
|
Sarkar KK, Mitra T, Aktaruzzaman M, Abid MA, Rahman MA, Debnath P, Sadhu SK. Exploring antioxidative, cytotoxic and neuropharmacological insights into Bixa orellana leaves: Experimental and in silico approaches. Heliyon 2024; 10:e27001. [PMID: 38468966 PMCID: PMC10926075 DOI: 10.1016/j.heliyon.2024.e27001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/13/2024] Open
Abstract
Background Study The aim of this research was to examine possible antioxidant, cytotoxic and neurological activity of methanol and n-hexane extracts of Bixa orellana leaves. Additionally, we aimed to identify potential lead compounds through in-silico analysis. Methods In-vitro antioxidative properties were investigated through different assays, including: total phenolic content assay (TPC), total flavonoid content assay (TFC), DPPH free radical scavenging assay and reducing power assay. Also, the cytotoxic effect of the samples was assessed using the brine shrimp lethality test. In addition, anxiolytic, locomotor, and CNS depressant activities were assessed utilizing various established methods. Moreover, reported compounds were used in the in silico study to explore the best-fit phytoconstituents against gamma-aminobutyric acid (GABAA) receptor. Results MBOL displayed substantial antioxidative activities in various established assays compared to NBOL. In brine shrimp lethality bioassay, both MBOL and NBOL revealed cytotoxic activity in a concentration-dependent approach. Again, in Elevated Plus Maze test, 200 and 400 mg/kg of NBOL and MBOL demonstrated significant anxiolytic activities evident from time spent in open arms. In addition, maximum number of head dipping was demonstrated by MBOL at 400 mg/kg (53.90 ± 1.16) in Hole Board test. NBOL and MBOL at both doses significantly diminished the magnitude of movements from the 2nd to 5th observation periods in Open Field test. Furthermore, in Hole Cross test, MBOL remarkably dwindled the locomotor activity at 120 min and 180 min (3.60 ± 0.40 and 2.40 ± 0.51) at 400 mg/kg. Finally, in silico analysis revealed 13 compounds as promising leads with strong binding affinity to GABAA receptor along with good pharmacokinetics and toxicity profiles. Conclusion Therefore, the present study's findings advocate the traditional usage of this plant and recommend both MBOL and NBOL as as a potential source of therapeutic candidate for the management of neurological disorders.
Collapse
Affiliation(s)
- Kishore Kumar Sarkar
- Department of Pharmacy, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Trina Mitra
- Department of Pharmacy, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Md Aktaruzzaman
- Department of Pharmacy, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Md Ahsan Abid
- Department of Pharmacy, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Md Asibur Rahman
- Department of Pharmacy, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Pradip Debnath
- Department of Pharmacy, Comilla University, Cumilla, Bangladesh
| | - Samir Kumar Sadhu
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, 9208, Bangladesh
| |
Collapse
|
5
|
Eissa MA, Gohar EY. Aromatase enzyme: Paving the way for exploring aromatization for cardio-renal protection. Biomed Pharmacother 2023; 168:115832. [PMID: 37931519 PMCID: PMC10843764 DOI: 10.1016/j.biopha.2023.115832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 10/15/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023] Open
Abstract
Documented male-female differences in the risk of cardiovascular and chronic kidney diseases have been largely attributed to estrogens. The cardiovascular and renal protective effects of estrogens are mediated via the activation of estrogen receptors (ERα and ERβ) and G protein-coupled estrogen receptor, and involve interactions with the renin-angiotensin-aldosterone system. Aromatase, also called estrogen synthase, is a cytochrome P-450 enzyme that plays a pivotal role in the conversion of androgens into estrogens. Estrogens are biosynthesized in gonadal and extra-gonadal sites by the action of aromatase. Evidence suggests that aromatase inhibitors, which are used to treat high estrogen-related pathologies, are associated with the development of cardiovascular events. We review the potential role of aromatization in providing cardio-renal protection and highlight several meta-analysis studies on cardiovascular events associated with aromatase inhibitors. Overall, we present the potential of aromatase enzyme as a fundamental contributor to cardio-renal protection.
Collapse
Affiliation(s)
- Manar A Eissa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Merit University, New Sohag, Sohag, Egypt
| | - Eman Y Gohar
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States.
| |
Collapse
|
6
|
Yousefzadeh N, Jeddi S, Zarkesh M, Norouzirad R, Kashfi K, Ghasemi A. Protective effects of long-term nitrate administration against ovariectomy-induced kidney dysfunction in rats. Pharmacol Rep 2023:10.1007/s43440-023-00499-9. [PMID: 37258800 DOI: 10.1007/s43440-023-00499-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 05/15/2023] [Accepted: 05/19/2023] [Indexed: 06/02/2023]
Abstract
BACKGROUND Menopause is associated with higher risks of chronic kidney disease. We determined the effect of nitrate on ovariectomy-induced kidney dysfunction METHODS: Control, ovariectomized (OVX), control + nitrate, and OVX + nitrate female Wistar rats (n = 10/group); sodium nitrate (100 mg/L) administered in drinking water for 9 months. Glomerular filtration rate (GFR) and albumin excretion rate (AER) were calculated from serum and urine parameters. At month 9, serum and kidney levels of nitric oxide (NO) metabolites (NOx), oxidative stress indices, and mRNA expression of endothelial NO synthase (eNOS) were measured; with histological analyses of the kidney. RESULTS Compared to controls, OVX rats had lower GFR (31%, p = 0.0079), higher glomerular tuft volume (30%, p = 0.0402), and Bowman's capsule space (39%, p = 0.0224). OVX rats had lower serum NOx (33%, p = 0.0061) and kidney eNOS mRNA expression (34%, p = 0.0368). Nitrate administration to: (i) control rats increased serum NOx (59%, p < 0.0001), with no effect on other parameters; (ii) OVX rats increased serum (85%, p < 0.0001) and kidney (106%, p = 0.0008) NOx values, and restored kidney eNOS expression to normal value. Nitrate administration to OVX rats increased GFR (36%, p = 0.0361) and restored glomerular tuft volume and Bowman's capsule space to normal values. In OVX rats, it also increased serum catalase (CAT) activity, serum and kidney total antioxidant capacity (TAC), and decreased serum malondialdehyde (MDA). CONCLUSIONS Low-dose long-term nitrate administration protects against ovariectomy-induced kidney dysfunction in rats. This effect is associated with reducing ovariectomy-induced oxidative stress and restoring eNOS-derived NO deficiency in systemic circulation and the kidney.
Collapse
Affiliation(s)
- Nasibeh Yousefzadeh
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Zarkesh
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Norouzirad
- Department of Biochemistry, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, Newyork, NY, USA
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Ibrahim HA, Abd El-Alim AEAF, El-Hafeez MA, Metwally MMM, Khamis T, Galal AAA. Baicalein prevents capecitabine-induced heart damage in female Wistar rats and enhances its anticancer potential in MCF-7 breast cancer cells. Life Sci 2023; 319:121523. [PMID: 36842762 DOI: 10.1016/j.lfs.2023.121523] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/18/2023] [Accepted: 02/19/2023] [Indexed: 02/26/2023]
Abstract
AIM We investigated the ability of baicalein (BAI) to enhance the anticancer potential of capecitabine (CAP) in the MCF-7 cell line and its protective effect on CAP-induced cardiotoxicity in female Wistar rats. METHODS AND KEY FINDINGS In vitro study involved evaluating the effect of BAI and/or CAP on cell viability, cell cycle progression, and BAX and Bcl2 gene expression in MCF-7 cells. Co-treatment of BAI with CAP significantly reduced the viability of MCF-7 cells, improved their cytotoxic effect, markedly elevated the percentage of the sub-G1 population, drastically reduced the G2/M population, and significantly altered the mRNA expression of BAX and Bcl2 genes compared with each treatment alone. In vivo study revealed that the oral administration of CAP (140 mg/kg BW) to adult female rats significantly elevated the levels of serum creatine kinase-myocardial band (CK-MB), lactate dehydrogenase (LDH), tumor necrosis factor (TNF)-α, and interleukin (IL)-1β and cardiac TNF-α, IL-1β malondialdehyde (MDA) concentration, whereas it reduced the serum and cardiac total antioxidant capacity (TAC), level of cardiac glutathione (GSH) and activity of glutathione peroxidase (GPx) with a vast array of circulatory, inflammatory, degenerative, and necrotic alterations in the cardiac tissue. Furthermore, CAP administration significantly upregulated the mRNA expression of NF-κB, TLR4, MyD88, ATF6, CHOP, and JNK genes. Concurrent administration of BAI (200 mg/kg BW) and CAP significantly improved the biochemical alterations and cardiac oxidant/antioxidant status and architecture. In addition, it modulated the TLR4/MyD88/NF-κB pathway and endoplasmic reticulum stress. SIGNIFICANCE Altogether, BAI can augment the anticancer potential of CAP and alleviate its cardiotoxic effects during cancer treatment.
Collapse
Affiliation(s)
- Hosny A Ibrahim
- Pharmacology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | | | - Mai Abd El-Hafeez
- Pharmacology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Mohamed M M Metwally
- Pathology department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Tarek Khamis
- Pharmacology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Azza A A Galal
- Pharmacology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt.
| |
Collapse
|
8
|
Hassan N, Rashad M, Elleithy E, Sabry Z, Ali G, Elmosalamy S. L-Carnitine alleviates hepatic and renal mitochondrial-dependent apoptotic progression induced by letrozole in female rats through modulation of Nrf-2, Cyt c and CASP- 3 signaling. Drug Chem Toxicol 2023; 46:357-368. [PMID: 35176959 DOI: 10.1080/01480545.2022.2039180] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Letrozole (LTZ) is a non-steroidal aromatase inhibitor that is commonly used in breast cancer therapy. It has several side effects that might lead to the drug's cessation and data of LTZ's potential adverse effects on the hepatorenal microenvironment was conflicting. In addition, searching for therapeutic interventions that could modulate its adverse effects will be very beneficial. So, this study aims to determine the impact of LTZ on the hepatorenal microenvironment in cyclic female rats with a proposed regulatory role of L-Carnitine (LC) supplementation giving molecular insights into its possible mechanism of action. LTZ (1 mg/kg using 0.5% carboxy methyl cellulose as a vehicle for 21 consecutive days orally) to assess its impact on hepatorenal microenvironment. After treatment with LC (100 mg/kg orally) for 14 days, hepatorenal redox state (lipid peroxides (MDA), reduced glutathione (GSH) and catalase enzyme (CAT)), as well as relative gene expression of nuclear factor erythroid 2-related factor 2 (Nrf-2), cytochrome-c (Cyt c) and caspase-3 (CASP-3) were evaluated. Histopathological examination and immunohistochemical staining of CASP-3 in both liver and kidney were done. LTZ altered hepatic and renal functions. Relative gene expression of hepatorenal Nrf-2, Cyt c and CASP-3 as well as redox state revealed significant deterioration. Also, the liver and kidney tissues showed several micromorphological changes and intense reaction to CASP-3 upon immunohistochemical staining. It can be concluded that LC alleviates LTZ induced hepatorenal oxidative stress (OS) and mitochondrial-dependent apoptotic progression through modulation of Nrf-2, Cyt c, and CASP-3 signaling in female rats.
Collapse
Affiliation(s)
- Neven Hassan
- Department of Physiology, Faculty of Veterinary Medicine, Cairo University, Cario, Egypt
| | - Maha Rashad
- Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Cairo University, Cario, Egypt
| | - Ebtihal Elleithy
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Cario, Egypt
| | - Zainab Sabry
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Cario, Egypt
| | - Ghada Ali
- Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Cairo University, Cario, Egypt
| | - Sherif Elmosalamy
- Department of Physiology, Faculty of Veterinary Medicine, Cairo University, Cario, Egypt
| |
Collapse
|
9
|
Xu Q, Li Y, Du W, Zheng N, Wang J, Zhao S. Effect of dietary biochanin A on lactation performance, antioxidant capacity, rumen fermentation and rumen microbiome of dairy goat. Front Microbiol 2023; 14:1101849. [PMID: 36814572 PMCID: PMC9939525 DOI: 10.3389/fmicb.2023.1101849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/23/2023] [Indexed: 02/08/2023] Open
Abstract
Biochanin A (BCA), an isoflavone phytoestrogen, is a secondary metabolite produced mainly in leguminous plants. The objective of this study was to evaluate the effect of BCA on lactation performance, nitrogen metabolism, and the health of dairy goat. Thirty mid-lactation Saanen dairy goats were divided into three groups randomly: control, 2 g/d BCA group, and 6 g/d BCA group. After 36 days of feeding, 30 dairy goats were transferred to individual metabolic cages. Subsequently, milk yield, feed intake, total feces, and urine excretion were recorded and samples were collected continuously for 3 days. Blood and ruminal fluid samples were collected over the subsequent 4 days. Milk yield, milk protein, fat content, and the feed conversion ratio of dairy goat were significantly increased by the BCA treatment. The levels of serum 17β-estradiol, growth hormone, insulin-like growth factor 1, glutathione peroxidase activity, and total antioxidant capacity were also increased significantly by BCA, indicating that BCA enhanced the antioxidant capacity of dairy goat. Amino acid degradation was significantly inhibited, while the ammonia nitrogen content was reduced significantly by BCA. Total volatile fatty acids was significantly increased by BCA supplementation. In addition, the relative abundance of Verrucomicrobiota was decreased significantly. However, the growth of nitrogen metabolism and cellulolytic bacteria was significantly increased under BCA treatment, including Prevotella sp., Treponema sp., Ruminococcus flavefaciens, and Ruminobacter amylophilus. In conclusion, supplementation with BCA improved the milk production performance, nitrogen metabolism, rumen fermentation and antioxidant capacity, and regulated the rumen microbiome of dairy goat.
Collapse
Affiliation(s)
- Qingbiao Xu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China,College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China,MoE Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yanjun Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Wenjuan Du
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Nan Zheng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jiaqi Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China,*Correspondence: Jiaqi Wang,
| | - Shengguo Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China,Shengguo Zhao,
| |
Collapse
|
10
|
Almutlaq RN, Newell-Fugate AE, Evans LC, Fatima H, Gohar EY. Aromatase inhibition increases blood pressure and markers of renal injury in female rats. Am J Physiol Renal Physiol 2022; 323:F349-F360. [PMID: 35900340 PMCID: PMC9423724 DOI: 10.1152/ajprenal.00055.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/22/2022] [Accepted: 07/13/2022] [Indexed: 11/22/2022] Open
Abstract
Aromatase is a monooxygenase that catalyzes the rate-limiting step of estrogen biosynthesis from androgens. Aromatase inhibitors are widely used for the treatment of patients with hormone receptor-positive breast cancer. However, the effects of aromatase inhibitors on cardiovascular and renal health in females are understudied. Given that estrogen is protective against cardiovascular and kidney diseases, we hypothesized that aromatase inhibition elevates blood pressure and induces kidney injury in female Sprague-Dawley rats. Twelve-week-old female rats were implanted with radiotelemetry transmitters to continuously monitor blood pressure. After baseline blood pressure recording, rats were randomly assigned to treatment with the aromatase inhibitor anastrozole (ASZ) or vehicle (Veh) in drinking water. Twenty days after treatment initiation, rats were shifted from a normal-salt (NS) diet to a high-salt (HS) diet for an additional 40 days. Rats were euthanized 60 days after treatment initiation. Body weight increased in both groups over the study period, but the increase was greater in the ASZ-treated group than in the Veh-treated group. Mean arterial pressure increased in ASZ-treated rats during the NS and HS diet phases but remained unchanged in Veh-treated rats. In addition, urinary excretion of albumin and kidney injury marker-1 and plasma urea were increased in response to aromatase inhibition. Furthermore, histological assessment revealed that ASZ treatment increased morphological evidence of renal tubular injury and proximal tubular brush border loss. In conclusion, chronic aromatase inhibition in vivo with ASZ increases blood pressure and markers of renal proximal tubular injury in female Sprague-Dawley rats, suggesting an important role for aromatization in the maintenance cardiovascular and renal health in females.NEW & NOTEWORTHY Aromatase enzyme catalyzes the rate-limiting step in estrogen biosynthesis. Aromatase inhibitors are clinically used for the treatment of patients with breast cancer; however, the impact of inhibiting aromatization on blood pressure and renal function is incompletely understood. The present findings demonstrate that systemic anastrozole treatment increases blood pressure and renal tubular injury markers in female rats fed a high-salt diet, suggesting an important role for aromatization in preserving cardiovascular and renal health in females.
Collapse
Affiliation(s)
- Rawan N Almutlaq
- Cardiorenal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Annie E Newell-Fugate
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Louise C Evans
- Department of Surgery, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Huma Fatima
- Division of Anatomic and Clinical Pathology, Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Eman Y Gohar
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
11
|
Ding S, Liu S, Chen Y, Peng Y, Zheng J. Anastrozole and Related Glucuronic Acid Conjugate are Electrophilic Species. Xenobiotica 2022; 52:380-388. [PMID: 35656966 DOI: 10.1080/00498254.2022.2086503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Anastrozole (ANA), is an inhibitor of non-steroidal aromatase, widely employed for the treatment of breast cancer. However, ANA-associated liver injury cases have been documented in the application of the drug.The major purposes of the present study were to identify the structure of reactive metabolites derived from ANA and to study related metabolic pathways of ANA.We found ANA itself is an electrophilic species reactive to GSH. ANA can be metabolized to ANA-N+-glucuronide (1) catalyzed by UGT1A4. An ANA GSH conjugate (2) was detected in bile and livers of rats treated with ANA. UGT1A4 participated in the phase II metabolic pathway.This work allowed us to better understand the mechanisms of the hepatotoxicity of ANA and provided new avenue to define the possible role of metabolic activation in hepatotoxicity.
Collapse
Affiliation(s)
- Siyu Ding
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Siyu Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Yaxuan Chen
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Ying Peng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Jiang Zheng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China.,State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou 550025, P. R. China.,Key Laboratory of Environmental Pollution, Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, P. R. China
| |
Collapse
|
12
|
Hou Y, Zhao W, Yu H, Zhang F, Zhang HT, Zhou Y. Biochanin A alleviates cognitive impairment and hippocampal mitochondrial damage in ovariectomized APP/PS1 mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 100:154056. [PMID: 35338989 DOI: 10.1016/j.phymed.2022.154056] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/27/2022] [Accepted: 03/13/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Estrogen deficiency leads to mitochondrial defects that precede Alzheimer's disease (AD)-associated pathological changes in a postmenopausal mouse model. Biochanin A (BCA) is a phytoestrogen isolated from Trifolium pratense L. used to relieve postmenopausal problems in women. In previous work, we observed that oral BCA treatment led to neuroprotection in an ovariectomized rat model. The objective of this study was to investigate whether and how BCA protects against hippocampal mitochondrial damage in a postmenopausal model of AD. METHOD APP/PS1 mice underwent bilateral ovariectomy and then, seven days later, received oral BCA at 20 or 40 mg/kg, or oral estradiol at 0.5 mg/kg, daily for 90 days. Sham animals were not ovariectomized and received no additional treatments. Cognitive function was examined using the passive avoidance task, novel object recognition test, and Morris water maze test. The level of circulating estrogen in vivo was assessed indirectly by measuring the wet weight of the uterus. We detected Aβ deposition and PGC-1α in brain by immunohistochemistry; p62, by immunofluorescence; and ERα, ERβ, PGC-1α, NRF1, mtTFA, Drp1, OPA1, Mfn2, Beclin1, LC3B, Pink1, and Parkin by immunoblotting. RESULTS BCA treatment rescued cognitive decline and reduced Aβ deposition and BACE1 expression in the hippocampus of ovariectomized APP/PS1 mice. BCA reversed the imbalance of mitochondrial dynamics caused by ovariectomy by increasing the expression of phospho-Drp1 (ser637), OPA1, and Mfn2. BCA reversed abnormal mitophagy induced by ovariectomy by increasing the expression of Beclin1, LC3B, Pink1, and Parkin, as well as by reducing the expression of p62. CONCLUSIONS BCA treatment enhances learning and memory abilities and alleviates AD symptoms in a postmenopausal model of AD. A possible mechanism is that BCA rescues the reduction of mitochondrial biogenesis, imbalance of mitochondrial dynamics, and abnormal mitophagy caused by ovariectomy. This study supports further research on BCA to develop treatments for postmenopausal women with AD.
Collapse
Affiliation(s)
- Yue Hou
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, No. 619 Changcheng Street, Daiyue district, Taian 271021, China
| | - Wei Zhao
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, No. 619 Changcheng Street, Daiyue district, Taian 271021, China
| | - Haiyang Yu
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, No. 619 Changcheng Street, Daiyue district, Taian 271021, China
| | - Fangfang Zhang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, No. 619 Changcheng Street, Daiyue district, Taian 271021, China
| | - Han-Ting Zhang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, No. 619 Changcheng Street, Daiyue district, Taian 271021, China; Department of Pharmacology, School of Pharmacy, Qingdao University, No. 16, Jiangsu Road, Shinan District, Qingdao 266011, China.
| | - Yanmeng Zhou
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, No. 619 Changcheng Street, Daiyue district, Taian 271021, China.
| |
Collapse
|
13
|
Ren L, Li F, Di Z, Xiong Y, Zhang S, Ma Q, Bian X, Lang Z, Ye Q, Wang Y. Estradiol Ameliorates Acute Kidney Ischemia-Reperfusion Injury by Inhibiting the TGF-βRI-SMAD Pathway. Front Immunol 2022; 13:822604. [PMID: 35281024 PMCID: PMC8907449 DOI: 10.3389/fimmu.2022.822604] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/17/2022] [Indexed: 12/16/2022] Open
Abstract
Renal ischemia–reperfusion injury (IRI) is less extensive in females than males in both animals and humans; however, this protection diminishes after menopause, suggesting that estrogen plays a pivotal role in IRI, but the underlying mechanism remains largely unknown. Our study found that 45 min of warm ischemia was sufficient to induce significant pathological changes without causing death in model animals. Compared with male rats, female rats exhibited less extensive apoptosis, kidney injury, and fibrosis; these effects were worsened in ovariectomized (OVX) rats and ameliorated upon estradiol (E2) supplementation. Furthermore, the levels of TGF-βRI, but not TGF-βRII or TGF-β1, were significantly increased in OVX rats, accompanied by phosphorylated SMAD2/3 activation. Interestingly, the alteration trend of the nuclear ERα level was opposite that of TGF-βRI. Furthermore, dual luciferase reporter and chromatin immunoprecipitation assays showed that ERα could bind to the promoter region of TGF-βRI and negatively regulate its mRNA expression. Moreover, an in vitro study using NRK-52E cells showed that ERα knockdown blocked E2-mediated protection, while TGF-βRI knockdown protected cells against hypoxic insult. The findings of this study suggest that renal IRI is closely related to the TGF-βRI-SMAD pathway in females and that E2 exert its protective effect via the ERα-mediated transcriptional inhibition of TGF-βRI expression.
Collapse
Affiliation(s)
- Lian Ren
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, China.,Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Fang Li
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ziyang Di
- Department of Gastrointestinal Surgery & Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yan Xiong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, China
| | - Shichen Zhang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, China
| | - Qing Ma
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, China
| | - Xiaoen Bian
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, China
| | - Zhiquan Lang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, China
| | - Qifa Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, China
| | - Yanfeng Wang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, China
| |
Collapse
|
14
|
Yangzom P, Amruthanand S, Sharma M, Mahajan S, Lingaraju MC, Parida S, Sahoo M, Kumar D, Singh TU. Subacute 28 days oral toxicity study of kaempferol and biochanin-A in the mouse model. J Biochem Mol Toxicol 2022; 36:e23090. [PMID: 35502512 DOI: 10.1002/jbt.23090] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 03/10/2022] [Accepted: 04/19/2022] [Indexed: 11/06/2022]
Abstract
The present study was undertaken to investigate the safety of kaempferol (KEM) and biochanin-A (BCA) following subacute exposure in mice. KEM and BCA were administered in three different doses by oral administration for 28 days. Evaluation of general toxicity parameters by examining the clinical signs, body weight, organ weights, haematological, biochemical, oxidative stress parameters, and histopathology was done. Administration of KEM and BCA for 28 days did not show any clinical signs of toxicity, nor any treatment-related changes in body weight and organ weights in comparison to control. The haematological parameters such as red blood cell, white blood cell, platelets count, haemoglobin (Hb) level, haematocrit, mean corpuscular haemoglobin concentration, red cell distribution width, and platelet distribution width did not show any change in the treated groups and control. Furthermore, different biochemical parameters like markers of the liver (alanine aminotransferase and aspartate aminotransferase), kidney (creatinine and urea), and heart (creatinine kinase-myocardial band and lactate dehydrogenase) injury along with other biochemical parameters showed nonsignificant differences between treated groups and control. Results of oxidative stress parameters in treated groups showed insignificant variations with control. The level of antioxidant enzymes such as superoxide dismutase and catalase were markedly increased in the treated groups; however, these were nonsignificant in comparison to control. In histopathology, evaluation of all vital organs, such as liver, kidney, heart, and lungs, did not show any morphological abnormalities and lesions in treated groups and control. The present study suggests that KEM and BCA have no adverse effects on the general physiology in mice.
Collapse
Affiliation(s)
- Padma Yangzom
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - S Amruthanand
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Meemansha Sharma
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Sumit Mahajan
- Division of Medicine, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Madhu Cholenahalli Lingaraju
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Subhashree Parida
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Monalisa Sahoo
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Dinesh Kumar
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Thakur Uttam Singh
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| |
Collapse
|
15
|
Sarfraz A, Javeed M, Shah MA, Hussain G, Shafiq N, Sarfraz I, Riaz A, Sadiqa A, Zara R, Zafar S, Kanwal L, Sarker SD, Rasul A. Biochanin A: A novel bioactive multifunctional compound from nature. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 722:137907. [PMID: 32208265 DOI: 10.1016/j.scitotenv.2020.137907] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/25/2020] [Accepted: 03/11/2020] [Indexed: 06/10/2023]
Abstract
Natural products (NPs) will continue to serve humans as matchless source of novel drug leads and an inspiration for the synthesis of non-natural drugs. As our scientific understanding of 'nature' is rapidly expanding, it would be worthwhile to illuminate the pharmacological distinctions of NPs to the scientific community and the public. Flavonoids have long fascinated scientists with their remarkable structural diversity as well as biological functions. Consequently, this review aims to shed light on the sources and pharmacological significance of a dietary isoflavone, biochanin A, which has been recently emerged as a multitargeted and multifunctional guardian of human health. Biochanin A possesses anti-inflammatory, anticancer, neuroprotective, antioxidant, anti-microbial, and hepatoprotective properties. It combats cancer development by inducing apoptosis, inhibition of metastasis and arresting cell cycle via targeting several deregulated signaling pathways of cancer. It fights inflammation by blocking the expression and activity of pro-inflammatory cytokines via modulation of NF-κB and MAPKs. Biochanin A acts as a neuroprotective agent by inhibiting microglial activation and apoptosis of neurons. As biochanin A has potential to modulate several biological networks, thus, it can be anticipated that this therapeutically potent compound might serve as a novel lead for drug development in the near future.
Collapse
Affiliation(s)
- Ayesha Sarfraz
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Maria Javeed
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Muhammad Ajmal Shah
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan.
| | - Ghulam Hussain
- Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Nusrat Shafiq
- Department of Chemistry, Government College Woman University Faisalabad (GCWUF), 38000 Faisalabad, Pakistan
| | - Iqra Sarfraz
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Ammara Riaz
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Ayesha Sadiqa
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Rabia Zara
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Saba Zafar
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Lubna Kanwal
- Institute of Pure and Applied Zoology, University of Okara, Okara, Pakistan
| | - Satyajit D Sarker
- Centre for Natural Products Discovery (CNPD), School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, England, UK
| | - Azhar Rasul
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan.
| |
Collapse
|
16
|
Yu C, Zhang P, Lou L, Wang Y. Perspectives Regarding the Role of Biochanin A in Humans. Front Pharmacol 2019; 10:793. [PMID: 31354500 PMCID: PMC6639423 DOI: 10.3389/fphar.2019.00793] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/18/2019] [Indexed: 12/15/2022] Open
Abstract
Biochanin A (BCA) is an isoflavone mainly found in red clover with poor solubility and oral absorption that is known to have various effects, including anti-inflammatory, estrogen-like, and glucose and lipid metabolism modulatory activity, as well as cancer preventive, neuroprotective, and drug interaction effects. BCA is already commercially available and is among the main ingredients in many types of supplements used to alleviate postmenopausal symptoms in women. The activity of BCA has not been adequately evaluated in humans. However, the results of many in vitro and in vivo studies investigating the potential health benefits of BCA are available, and the complex mechanisms by which BCA modulates transcription, apoptosis, metabolism, and immune responses have been revealed. Many efforts have been exerted to improve the poor bioavailability of BCA, and very promising results have been reported. This review focuses on the major effects of BCA and its possible molecular targets, potential uses, and limitations in health maintenance and treatment.
Collapse
Affiliation(s)
- Chen Yu
- Department of Infectious Diseases, First Hospital of Jilin University, Changchun, China
| | - Peng Zhang
- Department of Infectious Diseases, First Hospital of Jilin University, Changchun, China.,Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Lixin Lou
- Department of Infectious Diseases, First Hospital of Jilin University, Changchun, China
| | - Yang Wang
- Department of Infectious Diseases, First Hospital of Jilin University, Changchun, China.,Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
17
|
Mohamed AAR, Ahmed MM, Gomaa M, Ebraheim LLM. Bone health consequence of adjuvant Anastrozole in monotherapy or associated with biochanin-A in ovariectomized rat model. Life Sci 2018; 212:159-167. [PMID: 30290186 DOI: 10.1016/j.lfs.2018.09.059] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 09/01/2018] [Accepted: 09/28/2018] [Indexed: 02/07/2023]
Abstract
AIMS We investigate the consequence of adjuvant anastrozole (ANA) in monotherapy or associated with biochanin A (BCA) in ovariectomized (OVX) rat model and the degree of developing bone loss in both conditions. MATERIALS AND METHODS Sixty female rats were assigned to six groups. Five groups were bilaterally OVX, and one was sham operated. The five groups were; ANA group (0.5 mg/kg b.wt orally), BCA (5 mg/kg b.wt intraperitoneally (I/P), co-treated group (BCA + ANA), two control groups receiving even distilled water orally or DMSO I/P for twenty weeks. Bone turnover biomarkers BALP, OC, PTH, TRAP and TNFα were determined in serum. Bone mineral content, histological and morphometric measurements on rat femurs were performed. BMD by X-ray technique on tibias of rats and CT analysis of lumbar vertebrae of all treated and sham groups were applied. KEY FINDINGS There was marked elevation in bone turnover biomarkers with high serum Ca and P content in the ANA-treated rats. Moreover marked elevation of TNFα, PTH, TC and TG, ANA caused severe changes in the BMD detected by X-ray in tibial bones and CT analysis of lumbar vertebrae of OVX rats. While I/P injection of BCA ameliorated the adverse bone health decrements caused by ANA. SIGNIFICANCE The study highlights the importance of the BCA supplementation in accordance with the ANA therapy in case of ovariectomized rat model of osteoporosis which is clinically presented in Postmenopausal women with breast cancer during which considerable risk of developing osteoporosis is predicted during treatment.
Collapse
Affiliation(s)
| | - Mona M Ahmed
- Dept. of Forensic Medicine and Toxicology, Zagazig University, Zagazig, Egypt
| | - Mohamed Gomaa
- Dept. of Surgery, Zagazig University, Zagazig, Egypt
| | | |
Collapse
|
18
|
Edrees NE, Galal AA, Abdel Monaem AR, Beheiry RR, Metwally MM. Curcumin alleviates colistin-induced nephrotoxicity and neurotoxicity in rats via attenuation of oxidative stress, inflammation and apoptosis. Chem Biol Interact 2018; 294:56-64. [DOI: 10.1016/j.cbi.2018.08.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/13/2018] [Accepted: 08/15/2018] [Indexed: 12/29/2022]
|
19
|
|