1
|
Mustafa A, ArumughamIndiran M, Perumal E, Ponnala A, Rasheed DA, Ramalingam K, Shanmugham R, Karobari MI. Chemopreventive effects of chitosan nanogel with thiocolchicoside and lauric acid in chemically induced oral carcinogenesis, in a rodent model. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04185-w. [PMID: 40314765 DOI: 10.1007/s00210-025-04185-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 04/13/2025] [Indexed: 05/03/2025]
Abstract
There is always a quest for newer, more effective chemopreventive agents to prevent and manage oral cancer. A novel nanogel prepared using thiocolchicoside, lauric acid, and chitosan showed promising anticancer activity in KB-1 cell lines. The current manuscript aims to investigate the chemopreventive activity of chitosan nanogel with thiocolchicoside and lauric acid in chemically induced oral carcinogenesis using Wistar rats. Forty-six male Wistar rats were divided into three different groups: group I (control group), group II (cancer induction group), and group III (cancer induction with a chemopreventive agent). Male Wistar rats were given 20 μl/ml 4NQO solutions daily in their drinking water. One group received a daily oral application of CTL nanogel and carcinogen in drinking water. After the 23-week carcinogen treatment, the rats were euthanized; the tongues of the rats were dissected and histopathologically examined. Additionally, RT-PCR was employed to assess the gene expression of various signaling molecules involved in cancer progression like Erk1/2, β-catenin, Ki-67, Cyclin D1, TNF-α, NFκB, COX-2, and RAC1. Wistar rats developed white lesions and growth in the tongue in the cancer induction group. At the same time, the incidence and size of tumors were significantly less in the CTL nanogel-treated group. There was a significant increase in p53, Caspase-3, and Bax expression levels, while Bcl-2 showed a decreased expression in the CTL nanogel-treated group. There was also a significant decrease in the expression of EGFR and VEGFR signaling molecules in the CTL nanogel-treated group (p < 0.05 level). CTL nanogel shows potent chemopreventive efficiency in reducing the occurrence and severity of 4NQO-induced oral cancer in Wistar rats, marking it a promising candidate for further investigation in cancer prevention strategies.
Collapse
Affiliation(s)
- Ameena Mustafa
- Department of Oral Pathology and Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
- Department of Oral Pathology and Microbiology, Azeezia College of Dental Sciences & Research, Kollam, Kerala, India
| | - Meignana ArumughamIndiran
- Department of Public Health Dentistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Elumalai Perumal
- Department of Biochemistry, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Anandakumar Ponnala
- Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Dinsha Abdul Rasheed
- Department of Pediatric Dentistry, City University College of Ajman, Ajman, United Arab Emirates
| | - Karthikeyan Ramalingam
- Department of Oral Pathology and Microbiology, Malla Reddy Institute of Dental Sciences, Malla Reddy Vishwa Vidyapeeth, Suraram, Hyderabad, Telangana, India
| | - Rajeshkumar Shanmugham
- Department of Biochemistry, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Mohmed Isaqali Karobari
- Department of Conservative Dentistry and Endodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India.
- Department of Restorative Dentistry & Endodontics, Faculty of Dentistry, University of Puthisastra, Phnom Penh, 12211, Cambodia.
| |
Collapse
|
2
|
Kadry MO, Ali H. Fischer's ratio and DNA damage in hypoxemia-induced brain injury in rat model: prophylactic role of quercetin and mexamine supplementation. PLoS One 2025; 20:e0319898. [PMID: 40100888 PMCID: PMC11918368 DOI: 10.1371/journal.pone.0319898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 02/03/2025] [Indexed: 03/20/2025] Open
Abstract
Hypoxemia brain injuries arise when the brain's oxygen supply is restricted. Brain cells gradually die and become impaired as a result of the restricted oxygen flow a diversity of signaling pathways are involved in the pathophysiology of brain damage. One of the main concerns when examining the rate of protein breakdown is the measurement of the serum amino acid ratio. Valine, leucine, and isoleucine make up branched-chain amino acids, while phenylalanine and tyrosine make up aromatic amino acids. A vital tool for assessing the severity of hypoxemia is Fischer's ratio. The goal of this article is to determine how quercetin (QUR) and/or mexamine (MEX) prevented synfat (SN)-induced brain damage in a rat models. It also aimed to elucidate the various cross-linked inflammatory pathways, DNA damage, and Fischer's ratio. Following QUR and MEX therapy, synfat-induced hypoxemia. Hemoglobin (Hb) levels were markedly reduced by synfat-intoxication, and oxidative stress and inflammatory biomarkers, including TNF-??, MDA, interleukin-6 (IL-6), and C -reactive protein (CRP), were elevated. Hemoglobin levels, oxidative stress biomarkers, and the aberrant expression of pro-inflammatory cytokines were all altered by QUR and/or MEX therapy. Similarly, the concentration of γ-aminobutyric acid, serotonine, noradrenaline, and intropin in cerebral tissue is restricted. Similarly, the COMET assay and 8-oxo-7,8-dihydro-2'-deoxyguanosine analysis (8-oxodG) demonstrated that QUR and MEX potentially altered synfat-induced brain DNA damage. The results confirmed the potential impact of this combined strategy as a powerful therapy for brain hypoxemia, concluding that treatment via QUR with MEX was superior therapy in modulating synfat-triggered cerebral injury.
Collapse
Affiliation(s)
- Mai O. Kadry
- Therapeutic Chemistry Department, National Research Centre, El Buhouth St., Dokki, Egypt
| | - Hanaa Ali
- Department of Genetics and Cytology, National Research Centre, Dokki, Egypt
| |
Collapse
|
3
|
Bai H, Liu T, Wang H, Li Y, Wang Z. Metabolic reprogramming of corn oligopeptide in regulating sodium nitrite-induced canine hepatocyte injury via TGF/NF-κB signaling pathways and aminoacyl-tRNA biosynthesis. Food Chem Toxicol 2024; 192:114935. [PMID: 39151875 DOI: 10.1016/j.fct.2024.114935] [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: 06/21/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Sodium nitrite (SN), a prevalent food preservative, is known to precipitate hepatotoxicity upon exposure. This study elucidates the hepatoprotective effects of corn oligopeptide (COP) and vitamin E (VE) against SN-induced hepatic injury in canine hepatocytes. Canine liver cells were subjected to SN to induce hepatotoxicity, followed by treatment with COP and VE. Evaluations included assays for cell viability, oxidative stress markers, apoptosis, and inflammatory cytokines. Additionally, transcriptomic and metabolomic analyses were performed to delineate the underlying molecular mechanisms. The findings demonstrated that COP and VE significantly ameliorated SN-induced cytotoxicity, oxidative stress, and apoptosis. It was evidenced by restored cell viability, enhanced antioxidant enzyme activity, reduced cytoplasmic enzyme leakage, and decreased levels of malondialdehyde and inflammatory cytokines, with COP showing superior efficacy. The RNA sequencing revealed that COP treatment suppressed the SN-activated aminoacyl-tRNA biosynthesis pathway and TGF-β/NF-κB signaling pathways, thereby mitigating amino acid depletion, apoptosis, and inflammation. Moreover, COP treatment upregulated genes associated with protein folding, bile acid synthesis, and DNA repair. Metabolomic analysis corroborated these results, showing that COP restored amino acid levels and enhanced bile acid metabolism, alleviating SN-induced metabolic disruptions. These findings offered significant insights into the protective mechanisms of COP underscoring its prospective application in treating liver injuries.
Collapse
Affiliation(s)
- Huasong Bai
- Nourse Science Centre for Pet Nutrition, Wuhu, 241200, PR China
| | - Tong Liu
- Nourse Science Centre for Pet Nutrition, Wuhu, 241200, PR China
| | - Hengyan Wang
- Nourse Science Centre for Pet Nutrition, Wuhu, 241200, PR China
| | - Yunliang Li
- School of Food and Biological Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, PR China
| | - Zhanzhong Wang
- Nourse Science Centre for Pet Nutrition, Wuhu, 241200, PR China.
| |
Collapse
|
4
|
Elshaer SE, Hamad GM, Sobhy SE, Darwish AMG, Baghdadi HH, H Abo Nahas H, El-Demerdash FM, Kabeil SSA, Altamimi AS, Al-Olayan E, Alsunbul M, Docmac OK, Jaremko M, Hafez EE, Saied EM. Supplementation of Saussurea costus root alleviates sodium nitrite-induced hepatorenal toxicity by modulating metabolic profile, inflammation, and apoptosis. Front Pharmacol 2024; 15:1378249. [PMID: 38881874 PMCID: PMC11177093 DOI: 10.3389/fphar.2024.1378249] [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: 01/29/2024] [Accepted: 05/06/2024] [Indexed: 06/18/2024] Open
Abstract
Sodium nitrite (NaNO2) is a widely used food ingredient, although excessive concentrations can pose potential health risks. In the present study, we evaluated the deterioration effects of NaNO2 additives on hematology, metabolic profile, liver function, and kidney function of male Wistar rats. We further explored the therapeutic potential of supplementation with S. costus root ethanolic extract (SCREE) to improve NaNO2-induced hepatorenal toxicity. In this regard, 65 adult male rats were divided into eight groups; Group 1: control, Groups 2, 3, and 4 received SCREE in 200, 400, and 600 mg/kg body weight, respectively, Group 5: NaNO2 (6.5 mg/kg body weight), Groups 6, 7 and 8 received NaNO2 (6.5 mg/kg body weight) in combination with SCREE (200, 400, and 600 mg/kg body weight), respectively. Our results revealed that the NaNO2-treated group shows a significant change in deterioration in body and organ weights, hematological parameters, lipid profile, and hepatorenal dysfunction, as well as immunohistochemical and histopathological alterations. Furthermore, the NaNO2-treated group demonstrated a considerable increase in the expression of TNF-α cytokine and tumor suppressor gene P53 in the kidney and liver, while a significant reduction was detected in the anti-inflammatory cytokine IL-4 and the apoptosis suppressor gene BCL-2, compared to the control group. Interestingly, SCREE administration demonstrated the ability to significantly alleviate the toxic effects of NaNO2 and improve liver function in a dose-dependent manner, including hematological parameters, lipid profile, and modulation of histopathological architecture. Additionally, SCREE exhibited the ability to modulate the expression levels of inflammatory cytokines and apoptotic genes in the liver and kidney. The phytochemical analysis revealed a wide set of primary metabolites in SCREE, including phenolics, flavonoids, vitamins, alkaloids, saponins and tannins, while the untargeted UPLC/T-TOF-MS/MS analysis identified 183 metabolites in both positive and negative ionization modes. Together, our findings establish the potential of SCREE in mitigating the toxic effects of NaNO2 by modulating metabolic, inflammatory, and apoptosis. Together, this study underscores the promise of SCREE as a potential natural food detoxifying additive to counteract the harmful impacts of sodium nitrite.
Collapse
Affiliation(s)
- Samy E Elshaer
- Department of Environmental Studies, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Gamal M Hamad
- Department of Food Technology, Arid Lands Cultivation Research Institute (ALCRI), City of Scientific Research and Technological Applications (SRTA-City), Alexandria, Egypt
| | - Sherien E Sobhy
- Department of Plant Protection and Biomolecular Diagnosis, Arid Lands Cultivation Research Institute (ALCRI), City of Scientific Research and Technological Applications (SRTA-City), Alexandria, Egypt
| | - Amira M Galal Darwish
- Department of Food Technology, Arid Lands Cultivation Research Institute (ALCRI), City of Scientific Research and Technological Applications (SRTA-City), Alexandria, Egypt
- Food Industry Technology Program, Faculty of Industrial and Energy Technology, Borg Al Arab Technological University (BATU), Alexandria, Egypt
| | - Hoda H Baghdadi
- Department of Environmental Studies, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | | | - Fatma M El-Demerdash
- Department of Environmental Studies, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Sanaa S A Kabeil
- Department of Protein Research, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), Alexandria, Egypt
| | - Abdulmalik S Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Ebtesam Al-Olayan
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Maha Alsunbul
- Department of Pharmaceutical Sciences., College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Omaima Kamel Docmac
- Anatomy and Embryology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Mariusz Jaremko
- Smart-Health Initiative and Red Sea Research Center, Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Elsayed E Hafez
- Department of Plant Protection and Biomolecular Diagnosis, Arid Lands Cultivation Research Institute (ALCRI), City of Scientific Research and Technological Applications (SRTA-City), Alexandria, Egypt
| | - Essa M Saied
- Chemistry Department (Biochemistry Division), Faculty of Science, Suez Canal University, Ismailia, Egypt
- Institute for Chemistry, Humboldt Universität zu Berlin, Berlin, Germany
| |
Collapse
|
5
|
O Kadry M, Ali HM. Fischer's oligopeptide ratio in ischemic hypoxia: prophylactic amendment of sophoretin and melatonin supplementation. Future Sci OA 2024; 10:FSO911. [PMID: 38827802 PMCID: PMC11140683 DOI: 10.2144/fsoa-2023-0117] [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: 06/21/2023] [Accepted: 10/02/2023] [Indexed: 06/05/2024] Open
Abstract
Aim: The fundamental pathophysiology of ischemic-hypoxia is oxygen depletion. Fischer's ratio is essential for monitoring hypoxia intensity. Methods: the current study highlighted the prophylactic role of sophoretin (QRC) and/or melatonin (MLN) versus sodium nitrite (SN) brain hypoxia. Results: Prophylactic treatment with sophoretin and MLN, was preceded with hypoxia-induction via sodium nitrite (60 mg/kg, S.C.). SN decreased hemoglobin (Hb), elevated HIF-α, HSP-70, IL-6 and TNF-α. Sophoretin and/or MLN restored the ameliorated inflammatory biomarkers, modulated norepinephrine, dopamine, serotonin and gamma-aminobutyric acid (GABA). Similarly, single-cell gel electrophoresis (SCGE or COMET) DNA damage assay confirmed this finding. Conclusion: Treatment via sophoretin and MLN was the most effective therapy for improving sodium nitrite-induced brain injury.
Collapse
Affiliation(s)
- Mai O Kadry
- Therapeutic Chemistry Department, National Research Centre, El Buhouth St., Dokki, 12622, Egypt
| | - Hanaa Mahmoud Ali
- Department of Genetics & Cytology, National Research Centre, Dokki, 12622, Egypt
| |
Collapse
|
6
|
Uzun-Goren D, Uz YH. Preventive effects of quercetin against inflammation and apoptosis in cyclophosphamide-induced testicular damage. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:647-656. [PMID: 38629094 PMCID: PMC11017851 DOI: 10.22038/ijbms.2024.74458.16177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 12/03/2023] [Indexed: 04/19/2024]
Abstract
Objectives We aimed to investigate the effects of quercetin (QRC) against cyclophosphamide (CP)-induced testicular damage and how it interacts with apoptotic and inflammatory signaling pathways. Materials and Methods Forty male Wistar rats were randomly divided into four groups, 10 in each group; Control group (corn oil, intragastrically, 14 days), QRC group (100 mg/kg QRC, dissolved in corn oil, 14 days), CP group (200 mg/kg CP, intraperitoneally, single dose on the 7th day), and CP+QRC group (100 mg/kg QRC, intragastrically, 14 days and 200 mg/kg CP, intraperitoneally, single dose on the 7th day). Animals were sacrificed one day after the last QRC application and the effects of quercetin were evaluated by histological, morphometrical, and hormonal parameters. Also, nuclear factor kappa B (NFkB), nuclear factor erythroid 2 related factor 2 (Nrf2), Bcl-2 associated X protein (Bax), and B-cell lymphoma-2 (Bcl-2) immunoreactivities were evaluated immunohistochemically. Results CP increased the testicular weight/body weight ratio, significantly decreasing body weights and testicular weights. All hormone levels were also reduced significantly. Morphometrically, seminiferous tubules diameter and germinal epithelial thickness decreased, while a significant increase was determined in interstitial field width in addition to histological damage. Furthermore, immunohistochemical findings also indicated that NFkB and Bax immunoreactivity were increased in the CP group, whereas significant decrease was seen in Nrf2 and Bcl-2 immunoreactivity. Apoptotic cell and tubule index were reduced in CP. QRC ensured improvement in all findings. Conclusion Data showed us, that QRC may have preventive effects in CP-induced testicular damage by acting on NFkB, Nrf2, Bax, and Bcl-2 pathways.
Collapse
Affiliation(s)
- Duygu Uzun-Goren
- Department of Histology and Embryology, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Yesim Hulya Uz
- Department of Histology and Embryology, Faculty of Medicine, Trakya University, Edirne, Turkey
| |
Collapse
|
7
|
Lokman MS, Althagafi HA, Alharthi F, Habotta OA, Hassan AA, Elhefny MA, Al Sberi H, Theyab A, Mufti AH, Alhazmi A, Hawsawi YM, Khafaga AF, Gewaily MS, Alsharif KF, Albrakati A, Kassab RB. Protective effect of quercetin against 5-fluorouracil-induced cardiac impairments through activating Nrf2 and inhibiting NF-κB and caspase-3 activities. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:17657-17669. [PMID: 36197616 DOI: 10.1007/s11356-022-23314-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 09/24/2022] [Indexed: 06/16/2023]
Abstract
5-Fluorouracil (5-FU) is a chemotherapy used to treat many types of cancer. Cardiotoxicity is one of the common drawbacks of 5-FU therapy. Quercetin (Qu) is a bioflavonoid with striking biological activities. This research aimed to assess the ameliorative effect of Qu against 5-FU-mediated cardiotoxicity. Thirty-five rats were allocated into five groups: control group (normal saline), 5-FU group (30 mg/kg, intraperitoneally), Qu group (50 mg/kg, oral), 25 mg/kg Qu+5-FU group, and 50 mg/kg Qu+5-FU. The experimental animals were received the above-mentioned drugs for 21 days. Results showed that 5-FU significantly elevated creatine kinase, lactate dehydrogenase, serum cholesterol and triglyceride, and upregulated troponin and renin mRNA expression. Additionally, cardiac oxidant/antioxidant imbalance was evident in elevated oxidants (malondialdehyde and nitric oxide) and depleted antioxidants (superoxide dismutase, catalase, glutathione peroxidase, and glutathione). 5-FU also downregulated the gene expression of nuclear factor erythroid 2-related factor 2. Furthermore, 5-FU significantly increased cardiac pro-inflammatory cytokines (tumor necrosis factor-alpha and interleukin-1 beta) and upregulated gene expression of nuclear factor kappa-B. 5-FU significantly enhanced cardiac apoptosis through upregulating caspase-3 expression and downregulating B-cell lymphoma 2. Immunohistochemical and histopathological examinations verified the above-mentioned findings. However, all these changes were significantly ameliorated in Qu pre-administered rats. Conclusively, Qu counteracted 5-FU-mediated cardiotoxicity through potent antioxidant, anti-inflammatory, and anti-apoptotic effects.
Collapse
Affiliation(s)
- Maha S Lokman
- Department of Biology, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-kharj, 11942, Saudi Arabia.
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, 11795, Egypt.
| | - Hussam A Althagafi
- Department of Biology, Faculty of Science and Arts, Al-Baha University, Almakhwah, Al-Baha, Saudi Arabia
| | - Fahad Alharthi
- Department of Biology, College of Science, Taif University, Taif, Saudi Arabia
| | - Ola A Habotta
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Arwa A Hassan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Pharmaceutical Industries, Sinai University, El Arish, Egypt
| | - Mohamed A Elhefny
- Department of Cancer and Molecular Biology, National Cancer Institute, Cairo University, Cairo, Egypt
- Department of Medical Genetics, Faculty of Medicine, Umm Al-Qura University, Alqunfudah, Saudi Arabia
| | - Hassan Al Sberi
- Basic Medical Science, Histopathology Department, National Organization for Drug Control and Research, Giza, Egypt
- Department of Biology, Faculty of Science, Taif University, Taif, Saudi Arabia
| | - Abdulrahman Theyab
- Department of Laboratory & Blood Bank, Security Forces Hospital, P.O. Box 14799, Mecca, 21955, Saudi Arabia
- College of Medicine, Al-Faisal University, P.O. Box 50927, Riyadh, 11533, Saudi Arabia
| | - Ahmad Hasan Mufti
- Medical Genetics Department, Faculty of Medicine, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Alaa Alhazmi
- Medical Laboratory Technology Department, Jazan University, Jazan, Saudi Arabia
- SMIRES for Consultation in Specialized Medical Laboratories, Jazan University, Jazan, Saudi Arabia
| | - Yousef M Hawsawi
- Research Center, King Faisal Specialist Hospital and Research Center, MBC-J04, PO Box 40047, Jeddah, 21499, Saudi Arabia
| | - Asmaa F Khafaga
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Edfina, 22758, Egypt
| | - Mahmoud S Gewaily
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Khalaf F Alsharif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, PO Box 11099, Taif, 21944, Saudi Arabia
| | - Ashraf Albrakati
- Department of Human Anatomy, College of Medicine, Taif University, PO Box 11099, Taif, 21944, Saudi Arabia
| | - Rami B Kassab
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, 11795, Egypt
- Department of Biology, Faculty of Science and Arts, Al-Baha University, Almakhwah, Al-Baha, Saudi Arabia
| |
Collapse
|
8
|
Ahmadi F, Louei Monfared A, Shakarami N. Protective effect of Zataria multiflora Boiss against sodium nitrite-induced hepatotoxicity in rats. AVICENNA JOURNAL OF PHYTOMEDICINE 2022; 12:213-223. [PMID: 36186930 PMCID: PMC9482715 DOI: 10.22038/ajp.2021.18781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 11/13/2022]
Abstract
Objective Sodium nitrite (NaNO2) is used as a color stabilizer and antimicrobial agent in preservation of cured meat and fish. However, extensive use of this agent in the meat industries increased worries about its detrimental effects on human health. Zataria multiflora (Z. multiflora) is a well-known plant with therapeutic properties in the traditional medicine. Therefore, the present study was conducted to investigate the protective effect of this plant against sodium nitrite-induced hepatotoxicity. Materials and methods Thirty-two male Wistar rats were divided into 4 groups: Control (without any treatment), nitrite (350 mg/kg by gavage for 60 days), NaNO2 plus Z. multiflora (rats treated with NaNO2 350 mg/kg gavage for 60 days and simultaneously received Z. multiflora extract at 200 mg/kg, ip) and Z. multiflora group (rats treated with Z. multiflora extract at 200 mg/kg, ip). At the end of the study, rats were euthanized and liver tissue samples were taken and studied under microscopy. Also, serum levels of liver function enzymes and antioxidant defense systems were measured. The results were analyzed using SPSS software and a p<0.0.5 was considered significant. Results Results showed that NaNO2 induces liver injuries and altered hepatic histo-architecture. Also, NaNO2 significantly altered the biochemical profiles and antioxidant defense parameters of the liver. However, treatment with Z. multiflora improved tissue integrity as well as antioxidant defense status and biochemical conditions of the liver. Conclusion Administration of Z. multiflora extract has beneficial effects on the NaNO2-induced histological and functional toxicity in the liver.
Collapse
Affiliation(s)
- Fatemeh Ahmadi
- Department of Histology and Bacteriology, Faculty of Para Veterinary, Ilam University, Ilam, Iran
| | - Ali Louei Monfared
- Department of Histology and Bacteriology, Faculty of Para Veterinary, Ilam University, Ilam, Iran,Corresponding Author: Tel: +98-8432224308, Fax: +98-8432224308,
| | - Neamatollah Shakarami
- Department of Histology and Bacteriology, Faculty of Para Veterinary, Ilam University, Ilam, Iran
| |
Collapse
|
9
|
Albadrani GM, BinMowyna MN, Bin-Jumah MN, El–Akabawy G, Aldera H, AL-Farga AM. Quercetin prevents myocardial infarction adverse remodeling in rats by attenuating TGF-β1/Smad3 signaling: Different mechanisms of action. Saudi J Biol Sci 2021; 28:2772-2782. [PMID: 34012318 PMCID: PMC8116976 DOI: 10.1016/j.sjbs.2021.02.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/19/2021] [Accepted: 02/01/2021] [Indexed: 12/16/2022] Open
Abstract
This study investigated the anti-remodeling and anti-fibrotic and effect of quercetin (QUR) in the remote non-infarcted of rats after myocardial infarction (MI). Rats were divided as control, control + QUR, MI, and MI + QUR. MI was introduced to the rats by ligating the eft anterior descending (LAD) coronary artery. All treatments were given for 30 days, daily. QUR persevered the LV hemodynamic parameters and prevented remote myocardium damage and fibrosis. Also, QUR supressed the generation of ROS, increased the nuclear levels of Nrf2, and enhanced SOD and GSH levels in the LVs of the control and MI model rats. It also reduced angiotensin II, nuclear level/activity of the nuclear factor NF-κβ p65, and protein expression of TGF-β1, α-SMA, and total/phospho-smad3 in the LVs of both groups. Concomitantly, QUR upregulated LV smad7 and BMP7. In conclusion, QUR prevents MI-induced LV remodeling by antioxidant, anti-inflammatory, and anti-fibroticα effects mediated by ROS scavenging, suppressing NF-κβ, and stimulating Nrf-2, Smad7, and BMP7.
Collapse
Affiliation(s)
- Ghadeer M. Albadrani
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mona N. BinMowyna
- College of Applied Medical Sciences, Shaqra University, Shaqra, Saudi Arabia
| | - May N. Bin-Jumah
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Gehan El–Akabawy
- Department of Basic Sciences, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
- Department of Anatomy and Embryology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Hussain Aldera
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
| | - Ammar M. AL-Farga
- Biochemistry Department, College of Sciences, University of Jeddah, Jeddah, Saudi Arabia
| |
Collapse
|
10
|
Eissa MM, Ahmed MM, Abd Eldaim MA, Mousa AA, Elkirdasy AF, Mohamed MA, Orabi SH. Chlorella vulgaris ameliorates sodium nitrite-induced hepatotoxicity in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:9731-9741. [PMID: 33151487 DOI: 10.1007/s11356-020-11474-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/28/2020] [Indexed: 06/11/2023]
Abstract
The current was conducted to evaluate the ameliorating effect of Chlorella vulgaris (CV) extract against sodium nitrite-induced hepatotoxicity in rats. Forty-five rats were allocated randomly into 5 groups (n = 9). Group I (GI), control group: orally gavaged with normal saline daily. Group II (GII): orally gavaged with CV extract (70 mg/kg BW) for 3 months. Group III (GIII): orally gavaged with sodium nitrite (80 mg/kg BW) for 3 months. Group IV (GIV): received sodium nitrite as GIII and CV extract as GII simultaneously for 3 months. Group V (GV): received CV extract as GII and then, sodium nitrite as in GIII from the end of first month until the end of the experiment. Sodium nitrite significantly increased the activities of serum alanine aminotransferase, aspartate aminotransferase, and serum concentrations of tumor interleukin 1-β and necrosis factor α. In addition, it increased concentrations of malondialdehyde and nitric oxide and expression level of caspase-3 in the hepatic tissue. However, it decreased activities of hepatic glutathione peroxidase, catalase, and superoxide dismutase and induced degenerative and necrotic changes in hepatic tissues. In contrast, CV extract administration modulated sodium nitrite-induced inflammation, oxidative stress, and alteration in hepatic tissue function and architecture. This study indicated that CV extract modulated sodium nitrite-induced hepatic toxicity through decreasing oxidative stress and inflammation and enhancing antioxidant enzyme activities in hepatic tissue of rats.
Collapse
Affiliation(s)
- Mai M Eissa
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, 32897, Egypt
| | - Mohamed M Ahmed
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, 32897, Egypt.
| | - Mabrouk A Abd Eldaim
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Menoufia University, Sheben Elkom, 32511, Egypt
| | - Ahmed A Mousa
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, 32897, Egypt
| | - Ahmed F Elkirdasy
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, 32897, Egypt
| | - Mostafa A Mohamed
- Department of Pathology, Faculty of Veterinary Medicine, Menoufia University, Sheben Elkom, Egypt
| | - Sahar H Orabi
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, 32897, Egypt
| |
Collapse
|
11
|
Eissa MM, Ahmed MM, Abd Eldaim MA, Orabi SH, Elbaz HT, Mohamed MA, Elweza AE, Mousa AA. Methanolic extract of Chlorella vulgaris protects against sodium nitrite-induced reproductive toxicity in male rats. Andrologia 2020; 52:e13811. [PMID: 32897594 DOI: 10.1111/and.13811] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/13/2020] [Accepted: 07/26/2020] [Indexed: 11/29/2022] Open
Abstract
The current study aimed to investigate the protective potential of Chlorella Vulgaris (CV) extract against the reproductive dysfunction induced by sodium nitrite toxicity. Forty-five male Wistar albino rats were assigned into five groups (n = 9). Control group received normal saline orally for 3 months, CV-treated: administered CV extract (70 mg/kg.BW) orally for 3 months, sodium nitrite-treated: received sodium nitrite (80 mg/kg.BW) orally for 3 months, co-treated: simultaneously received CV along with sodium nitrite treatment, orally, daily for 3 months, and CV-pre-treated: pre-treated with CV extract for 4 weeks followed by simultaneous treatment with sodium nitrite and CV extract for additional 8 weeks. Treatment with sodium nitrite significantly decreased serum testosterone and follicle-stimulating hormone concentrations, sperm count, motility, and viability. Besides, it decreased testicular superoxide dismutase and glutathione peroxidase activities while increased malondialdehyde concentration. This effect of sodium nitrite was associated with degenerative, necrotic, vascular, and inflammatory changes in testicular tissues. Treatment of sodium nitrite-intoxicated rats with CV in co-treated and pre-treated groups significantly prevented sodium nitrite-induced alterations of sperm parameters, hormonal concentrations, testicular oxidative-antioxidant status, and histological architecture. This study indicates that CV extract ameliorates the reproductive dysfunction induced by sodium nitrite toxicity via improving reproductive hormonal levels and testicular antioxidant activities.
Collapse
Affiliation(s)
- Mai M Eissa
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Mohamed M Ahmed
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Mabrouk A Abd Eldaim
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Menoufia University, Sheben Elkom, Egypt
| | - Sahar H Orabi
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Hamed T Elbaz
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Mostafa A Mohamed
- Department of Pathology, Faculty of Veterinary Medicine, Menoufia University, Sheben Elkom, Egypt
| | - Ahmed E Elweza
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Ahmed A Mousa
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| |
Collapse
|
12
|
El-Nabarawy NA, Gouda AS, Khattab MA, Rashed LA. Effects of nitrite graded doses on hepatotoxicity and nephrotoxicity, histopathological alterations, and activation of apoptosis in adult rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:14019-14032. [PMID: 32036525 DOI: 10.1007/s11356-020-07901-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 01/27/2020] [Indexed: 06/10/2023]
Abstract
Nitrites are found in several forms; they are widely found in water resources and used as additives and preservatives for food and as a color source. We investigated the hazardous effects of exposing rats to different doses of nitrites. Moreover, we examined such impacts, after acute ingestion, on liver and renal tissues in rats and to what extent this affects the organs' functions. Animals were divided into five groups: one control group 1 (group C) and four sodium nitrite (NaNO2)-treated group (8 rats per group). The four NaNO2-treated groups include group 2 (N20), group 3 (N40), group 4 (N60), and group 5 (N75). NaNO2 was dissolved in distilled water, and single acute dose was orally given by gavage at 20, 40, 60, and 75 mg/kg body weight, respectively. Our results revealed significant increase of liver enzymes activity-aspartate transaminase (AST), alanine aminotransferase (ALT), and creatinine between different groups with increasing doses of nitrite ingestion. The results of hepatic and renal oxidative stress showed significant increase in the malondialdehyde (MDA) levels and significant decrease in the antioxidant parameters, such as reduced glutathione (GSH), superoxide dismutase (SOD), and catalase (CAT), as the dose of nitrite increases. Further, the methemoglobin percent showed significant increase with increasing nitrite doses. Abnormal morphological alterations in the liver and kidney tissues were obviously proportional to the administered nitrite doses. The expression of caspase 3 and Bax level showed enhanced induction of immunoexpression, especially in the high doses of nitrites. On the other hand, the maximal immunoexpression level of anti-apoptotic marker Bcl2 was found in lower doses of nitrites, whereas marked decrease of Bcl2 levels was observed in the higher doses. In conclusion, administration of sodium nitrite in a dose-dependent manner is capable of inducing cellular and genetic toxicities and causes disturbance in biochemical analysis, oxidative and anti-oxidative balance, and methemoglobinemia. It also makes histopathological alterations and leads to the activation of apoptosis-related Bax, Bcl2, and caspase 3 genes of liver and kidney tissues in rats.
Collapse
Affiliation(s)
- Nagla A El-Nabarawy
- National Egyptian Center of Environmental and Toxicological Research (NECTR), Faculty of Medicine, Cairo University, Cairo, Egypt.
| | - Ahmed S Gouda
- National Egyptian Center of Environmental and Toxicological Research (NECTR), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed A Khattab
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Laila A Rashed
- Department of Biochemistry, Faculty of Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
13
|
Ferenczyova K, Kalocayova B, Bartekova M. Potential Implications of Quercetin and its Derivatives in Cardioprotection. Int J Mol Sci 2020; 21:E1585. [PMID: 32111033 PMCID: PMC7084176 DOI: 10.3390/ijms21051585] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 02/20/2020] [Accepted: 02/25/2020] [Indexed: 12/24/2022] Open
Abstract
Quercetin (QCT) is a natural polyphenolic compound enriched in human food, mainly in vegetables, fruits and berries. QCT and its main derivatives, such as rhamnetin, rutin, hyperoside, etc., have been documented to possess many beneficial effects in the human body including their positive effects in the cardiovascular system. However, clinical implications of QCT and its derivatives are still rare. In the current paper we provide a complex picture of the most recent knowledge on the effects of QCT and its derivatives in different types of cardiac injury, mainly in ischemia-reperfusion (I/R) injury of the heart, but also in other pathologies such as anthracycline-induced cardiotoxicity or oxidative stress-induced cardiac injury, documented in in vitro and ex vivo, as well as in in vivo experimental models of cardiac injury. Moreover, we focus on cardiac effects of QCT in presence of metabolic comorbidities in addition to cardiovascular disease (CVD). Finally, we provide a short summary of clinical studies focused on cardiac effects of QCT. In general, it seems that QCT and its metabolites exert strong cardioprotective effects in a wide range of experimental models of cardiac injury, likely via their antioxidant, anti-inflammatory and molecular pathways-modulating properties; however, ageing and presence of lifestyle-related comorbidities may confound their beneficial effects in heart disease. On the other hand, due to very limited number of clinical trials focused on cardiac effects of QCT and its derivatives, clinical data are inconclusive. Thus, additional well-designed human studies including a high enough number of patients testing different concentrations of QCT are needed to reveal real therapeutic potential of QCT in CVD. Finally, several negative or controversial effects of QCT in the heart have been reported, and this should be also taken into consideration in QCT-based approaches aimed to treat CVD in humans.
Collapse
Affiliation(s)
- Kristina Ferenczyova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia; (K.F.); (B.K.)
| | - Barbora Kalocayova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia; (K.F.); (B.K.)
| | - Monika Bartekova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia; (K.F.); (B.K.)
- Institute of Physiology, Comenius University in Bratislava, 81372 Bratislava, Slovakia
| |
Collapse
|
14
|
Alhusaini AM, Faddah LM, Hasan IH, Jarallah SJ, Alghamdi SH, Alhadab NM, Badr A, Elorabi N, Zakaria E, Al-anazi A. Vitamin C and Turmeric Attenuate Bax and Bcl-2 Proteins' Expressions and DNA Damage in Lead Acetate-Induced Liver Injury. Dose Response 2019; 17:1559325819885782. [PMID: 31798354 PMCID: PMC6864043 DOI: 10.1177/1559325819885782] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/29/2019] [Accepted: 10/08/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Lead is a common environmental and occupational pollutant which induced multiorgans dysfunction. The present study was designed to investigate the hepatoprotective effects of turmeric (TUR) and/or vitamin C (Vit-C) alone or together against lead acetate toxicity and to explore novel molecular pathways. METHOD Acute hepatotoxicity was induced by lead acetate (100 mg/kg/day, i.p.) in male rats, and the effect of TUR (200 mg/kg/day, orally) and/or Vit-C (250 mg/kg/day, orally) along with lead acetate for 7 days was studied. RESULTS Lead acetate increased serum alanine transaminase, aspartate transaminase, lactate dehydrogenase, hepatic lipid peroxidation and nitric oxide; while, hepatic superoxide dismutase and glutathione activities were downregulated. Hepatic Bcl-2-associated X (Bax) and B-cell lymphoma-2 (Bcl-2) proteins expressions were altered and hepatic DNA damaged was increased as well. Liver/body weight ratio was decreased. Hematoxylin and eosin demonstrated that lead acetate induced focal areas of massive hepatic degeneration of the hepatocytes. Treatment with both antioxidants ameliorated all the altered parameters and induced marked improvement of liver architecture. CONCLUSION The combination of TUR and Vit-C has shown the most protective effects against lead acetate-induced hepatotoxicity.
Collapse
Affiliation(s)
- Ahlam M. Alhusaini
- Department of Pharmacology and Toxicology, College of Pharmacy, King
Saud University, Riyadh, Saudi Arabia
| | - Laila M. Faddah
- Department of Pharmacology and Toxicology, College of Pharmacy, King
Saud University, Riyadh, Saudi Arabia
| | - Iman H. Hasan
- Department of Pharmacology and Toxicology, College of Pharmacy, King
Saud University, Riyadh, Saudi Arabia
| | - Somayah J. Jarallah
- Department of Pharmacology and Toxicology, College of Pharmacy, King
Saud University, Riyadh, Saudi Arabia
| | - Shrouq H. Alghamdi
- Department of Pharmacology and Toxicology, College of Pharmacy, King
Saud University, Riyadh, Saudi Arabia
| | - Norah M. Alhadab
- Department of Pharmacology and Toxicology, College of Pharmacy, King
Saud University, Riyadh, Saudi Arabia
| | - Amira Badr
- Department of Pharmacology and Toxicology, College of Pharmacy, King
Saud University, Riyadh, Saudi Arabia
- Department of Pharmacology and Toxicology, College of Pharmacy, AIN
Shams University, Egypt
| | - Najlaa Elorabi
- Department of Pharmacology and Toxicology, College of Pharmacy, King
Saud University, Riyadh, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez
Canal University, Ismailia, Egypt
| | - Enas Zakaria
- Department of Pharmaceutics Department, College of Pharmacy, King
Saud University, Riyadh, Saudi Arabia
| | - Abeer Al-anazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King
Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
15
|
Xiao G, Lyu M, Wang Y, He S, Liu X, Ni J, Li L, Fan G, Han J, Gao X, Wang X, Zhu Y. Ginkgo Flavonol Glycosides or Ginkgolides Tend to Differentially Protect Myocardial or Cerebral Ischemia-Reperfusion Injury via Regulation of TWEAK-Fn14 Signaling in Heart and Brain. Front Pharmacol 2019; 10:735. [PMID: 31333457 PMCID: PMC6624656 DOI: 10.3389/fphar.2019.00735] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 06/07/2019] [Indexed: 12/26/2022] Open
Abstract
Shuxuening injection (SXNI), one of the pharmaceutical preparations of Ginkgo biloba extract, has significant effects on both ischemic stroke and heart diseases from bench to bedside. Its major active ingredients are ginkgo flavonol glycosides (GFGs) and ginkgolides (GGs). We have previously reported that SXNI as a whole protected ischemic brain and heart, but the active ingredients and their contribution to the therapeutic effects remain unclear. Therefore, we combined experimental and network analysis approach to further explore the specific effects and underlying mechanisms of GFGs and GGs of SXNI on ischemia–reperfusion injury in mouse brain and heart. In the myocardial ischemia–reperfusion injury (MIRI) model, pretreatment with GFGs at 2.5 ml/kg was superior to the same dose of GGs in improving cardiac function and coronary blood flow and reducing the levels of lactate dehydrogenase and aspartate aminotransferase in serum, with an effect similar to that achieved by SXNI. In contrast, pretreatment with GGs at 2.5 ml/kg reduced cerebral infarction area and cerebral edema similarly to that of SXNI but more significantly compared with GFGs in cerebral ischemia–reperfusion injury (CIRI) model. Network pharmacology analysis of GFGs and GGs revealed that tumor necrosis factor-related weak inducer of apoptosis (TWEAK)–fibroblast growth factor-inducible 14 (Fn14) signaling pathway as an important common mechanism but with differential targets in MIRI and CIRI. In addition, immunohistochemistry and enzyme linked immunosorbent assay (ELISA) assays were performed to evaluate the regulatory roles of GFGs and GGs on the common TWEAK–Fn14 signaling pathway to protect the heart and brain. Experimental results confirmed that TWEAK ligand and Fn14 receptor were downregulated by GFGs to mitigate MIRI in the heart while upregulated by GGs to improve CIRI in the brain. In conclusion, our study showed that GFGs and GGs of SXNI tend to differentially protect brain and heart from ischemia–reperfusion injuries at least in part by regulating a common TWEAK–Fn14 signaling pathway.
Collapse
Affiliation(s)
- Guangxu Xiao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, Tianjin, China
| | - Ming Lyu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, Tianjin, China.,Institute of Chinese Materia Medica, China Academy of Chinese Medicial Sciences, Beijing, China
| | - Yule Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, Tianjin, China
| | - Shuang He
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, Tianjin, China
| | - Xinyan Liu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, Tianjin, China
| | - Jingyu Ni
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lan Li
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guanwei Fan
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jihong Han
- College of Life Sciences, Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China; College of Biomedical Engineering, Hefei University of Technology, Hefei, China
| | - Xiumei Gao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoying Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Yan Zhu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, Tianjin, China
| |
Collapse
|
16
|
Wu J, Yi J, Wu Y, Chen X, Zeng J, Wu J, Peng W. 3, 3-Dimethylquercetin Inhibits the Proliferation of Human Colon Cancer RKO Cells through Inducing G2/M Cell Cycle Arrest and Apoptosis. Anticancer Agents Med Chem 2019; 19:402-409. [PMID: 30398122 DOI: 10.2174/1871520618666181106120718] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 10/10/2018] [Accepted: 10/20/2018] [Indexed: 01/24/2023]
Abstract
Background:
Our previous study successfully identified that 3,3-Dimethylquercetin (DMQ) acted
as a potent anticancer agent against human colon cancer cell lines RKO. Thus, this study was conducted to investigate
the underlying mechanism by which DMQ displayed inhibitory activity in RKO cells.
Methods:
Flow cytometry was used to evaluate the effect of DMQ on the cell cycle arrest, as well as the mitochondrial
membrane potential in RKO cells. DAPI staining and DNA fragmentation ladder assays were performed
to assess the apoptosis inducing activity of DMQ. Furthermore, western blot analysis was conducted to
examine the expression of related proteins responsible for the cell cycle arrest and apoptosis.
Results:
Treatment with DMQ caused a significant increase in the fraction of G2/M cells, and induced remarkable
apoptosis. Furthermore, western blot analysis showed that DMQ arrested cells at G2/M checkpoint by
down-regulation of cyclin B1, cdc2 and cdc25c and up-regulation of p21, and induced cell apoptosis via affecting
the ratio of Bax/Bcl-2, causing loss of the mitochondrial membrane potential and enhancing the expression
of cleaved caspase-9 (C-caspase-9) and cleaved caspase-3 (C-caspase-3).
Conclusion:
These data showed that DMQ could suppress RKO cell growth by arresting RKO cells at G2/M
checkpoint and inducing mitochondria-dependent cell apoptosis. Our findings shed light on the potential use of
DMQ as a chemotherapeutic agent for CRC.
Collapse
Affiliation(s)
- Jianguo Wu
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Jun Yi
- Department of Chemistry and Life Science, Fujian Institute of Education, Fuzhou, 350025, China
| | - Yanbin Wu
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Xuzheng Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Jianwei Zeng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Jinzhong Wu
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Wei Peng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| |
Collapse
|